NZ738963B2 - Apoa-1 fusion polypeptides and related compositions and methods - Google Patents
Apoa-1 fusion polypeptides and related compositions and methods Download PDFInfo
- Publication number
- NZ738963B2 NZ738963B2 NZ738963A NZ73896316A NZ738963B2 NZ 738963 B2 NZ738963 B2 NZ 738963B2 NZ 738963 A NZ738963 A NZ 738963A NZ 73896316 A NZ73896316 A NZ 73896316A NZ 738963 B2 NZ738963 B2 NZ 738963B2
- Authority
- NZ
- New Zealand
- Prior art keywords
- amino acid
- seq
- polypeptide
- residues
- fusion
- Prior art date
Links
- 229920001184 polypeptide Polymers 0.000 title claims abstract description 525
- 230000004927 fusion Effects 0.000 title claims abstract description 339
- 239000000203 mixture Substances 0.000 title claims abstract description 53
- 102000004169 proteins and genes Human genes 0.000 claims abstract description 172
- 108090000623 proteins and genes Proteins 0.000 claims abstract description 172
- 230000000694 effects Effects 0.000 claims abstract description 73
- 108010061846 Cholesterol Ester Transfer Proteins Proteins 0.000 claims abstract description 65
- 102000012336 Cholesterol Ester Transfer Proteins Human genes 0.000 claims abstract description 65
- 102000006382 Ribonucleases Human genes 0.000 claims abstract description 64
- 108010083644 Ribonucleases Proteins 0.000 claims abstract description 64
- 238000002560 therapeutic procedure Methods 0.000 claims abstract description 58
- 101710038787 PLA2G7 Proteins 0.000 claims abstract description 51
- 101700047632 ASPG Proteins 0.000 claims abstract description 45
- 101700074758 LCAT Proteins 0.000 claims abstract description 45
- 102000006996 Aryldialkylphosphatase Human genes 0.000 claims abstract description 44
- 108010008184 Aryldialkylphosphatase Proteins 0.000 claims abstract description 44
- 102100005974 LCAT Human genes 0.000 claims abstract description 42
- 125000000539 amino acid group Chemical group 0.000 claims description 156
- 125000003275 alpha amino acid group Chemical group 0.000 claims description 151
- 210000004027 cells Anatomy 0.000 claims description 146
- HVYWMOMLDIMFJA-DPAQBDIFSA-N (3β)-Cholest-5-en-3-ol Chemical compound C1C=C2C[C@@H](O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H]([C@H](C)CCCC(C)C)[C@@]1(C)CC2 HVYWMOMLDIMFJA-DPAQBDIFSA-N 0.000 claims description 128
- 241000282414 Homo sapiens Species 0.000 claims description 102
- 201000010099 disease Diseases 0.000 claims description 72
- 229940107161 Cholesterol Drugs 0.000 claims description 64
- 235000012000 cholesterol Nutrition 0.000 claims description 62
- 230000014509 gene expression Effects 0.000 claims description 59
- 230000035695 Efflux Effects 0.000 claims description 55
- 229920003013 deoxyribonucleic acid Polymers 0.000 claims description 46
- 201000011510 cancer Diseases 0.000 claims description 42
- 201000000596 systemic lupus erythematosus Diseases 0.000 claims description 32
- 201000006417 multiple sclerosis Diseases 0.000 claims description 30
- 238000006467 substitution reaction Methods 0.000 claims description 30
- 238000004519 manufacturing process Methods 0.000 claims description 28
- 230000001965 increased Effects 0.000 claims description 27
- 200000000018 inflammatory disease Diseases 0.000 claims description 25
- 239000003814 drug Substances 0.000 claims description 23
- 206010003210 Arteriosclerosis Diseases 0.000 claims description 22
- 206010053643 Neurodegenerative disease Diseases 0.000 claims description 20
- 229920000023 polynucleotide Polymers 0.000 claims description 20
- 239000002157 polynucleotide Substances 0.000 claims description 20
- 206010003816 Autoimmune disease Diseases 0.000 claims description 19
- 238000002648 combination therapy Methods 0.000 claims description 19
- 230000002757 inflammatory Effects 0.000 claims description 19
- 230000001404 mediated Effects 0.000 claims description 19
- 208000008787 Cardiovascular Disease Diseases 0.000 claims description 18
- 206010039073 Rheumatoid arthritis Diseases 0.000 claims description 18
- 201000001320 atherosclerosis Diseases 0.000 claims description 17
- 102000018358 Immunoglobulins Human genes 0.000 claims description 16
- 108060003951 Immunoglobulins Proteins 0.000 claims description 16
- 201000009596 autoimmune hypersensitivity disease Diseases 0.000 claims description 16
- 201000009910 diseases by infectious agent Diseases 0.000 claims description 16
- 206010001897 Alzheimer's disease Diseases 0.000 claims description 15
- 208000009856 Lung Disease Diseases 0.000 claims description 15
- 201000008739 coronary artery disease Diseases 0.000 claims description 14
- 208000004981 Coronary Disease Diseases 0.000 claims description 13
- QKSKPIVNLNLAAV-UHFFFAOYSA-N Sulfur mustard Chemical compound ClCCSCCCl QKSKPIVNLNLAAV-UHFFFAOYSA-N 0.000 claims description 13
- 206010025650 Malignant melanoma Diseases 0.000 claims description 11
- 201000001441 melanoma Diseases 0.000 claims description 11
- 238000002626 targeted therapy Methods 0.000 claims description 10
- 206010067584 Type 1 diabetes mellitus Diseases 0.000 claims description 9
- 239000002253 acid Substances 0.000 claims description 9
- 230000035897 transcription Effects 0.000 claims description 9
- 101700077954 APOA1 Proteins 0.000 claims description 8
- 230000002519 immonomodulatory Effects 0.000 claims description 8
- 208000004476 Acute Coronary Syndrome Diseases 0.000 claims description 7
- 208000008589 Obesity Diseases 0.000 claims description 7
- 201000009794 idiopathic pulmonary fibrosis Diseases 0.000 claims description 7
- 235000020824 obesity Nutrition 0.000 claims description 7
- 206010001052 Acute respiratory distress syndrome Diseases 0.000 claims description 6
- 208000006673 Asthma Diseases 0.000 claims description 6
- 206010060945 Bacterial infection Diseases 0.000 claims description 6
- 206010012289 Dementia Diseases 0.000 claims description 6
- 206010021143 Hypoxia Diseases 0.000 claims description 6
- 208000001072 Type 2 Diabetes Mellitus Diseases 0.000 claims description 6
- 239000003937 drug carrier Substances 0.000 claims description 6
- 230000001146 hypoxic Effects 0.000 claims description 6
- 206010053219 Non-alcoholic steatohepatitis Diseases 0.000 claims description 5
- 230000000222 hyperoxic Effects 0.000 claims description 5
- 206010005003 Bladder cancer Diseases 0.000 claims description 4
- 208000006545 Chronic Obstructive Pulmonary Disease Diseases 0.000 claims description 4
- 206010058490 Hyperoxia Diseases 0.000 claims description 4
- 208000002154 Non-Small-Cell Lung Carcinoma Diseases 0.000 claims description 4
- 108009000071 Non-small cell lung cancer Proteins 0.000 claims description 4
- 208000006265 Renal Cell Carcinoma Diseases 0.000 claims description 4
- 201000009267 bronchiectasis Diseases 0.000 claims description 4
- 238000002512 chemotherapy Methods 0.000 claims description 4
- 238000001959 radiotherapy Methods 0.000 claims description 4
- 201000005112 urinary bladder cancer Diseases 0.000 claims description 4
- 201000000028 adult respiratory distress syndrome Diseases 0.000 claims description 3
- 210000004748 cultured cells Anatomy 0.000 claims description 3
- 201000010536 head and neck cancer Diseases 0.000 claims description 3
- 208000006011 Stroke Diseases 0.000 claims description 2
- MTCFGRXMJLQNBG-UWTATZPHSA-N D-serine Chemical compound OC[C@@H](N)C(O)=O MTCFGRXMJLQNBG-UWTATZPHSA-N 0.000 claims 4
- 102100007290 CD274 Human genes 0.000 claims 1
- 101710012053 CD274 Proteins 0.000 claims 1
- 108010059886 Apolipoprotein A-I Proteins 0.000 abstract description 244
- 102000005666 Apolipoprotein A-I Human genes 0.000 abstract description 244
- 230000000447 dimerizing Effects 0.000 abstract description 53
- 150000001413 amino acids Chemical class 0.000 abstract description 47
- 108010011964 Phosphatidylcholine-Sterol O-Acyltransferase Proteins 0.000 abstract description 39
- 102000013455 Amyloid beta-Peptides Human genes 0.000 abstract description 16
- 108010090849 Amyloid beta-Peptides Proteins 0.000 abstract description 16
- 230000000875 corresponding Effects 0.000 abstract description 14
- 235000018102 proteins Nutrition 0.000 description 162
- 125000005647 linker group Chemical group 0.000 description 106
- 108020001507 fusion proteins Proteins 0.000 description 103
- 102000037240 fusion proteins Human genes 0.000 description 103
- 235000001014 amino acid Nutrition 0.000 description 84
- 230000027455 binding Effects 0.000 description 61
- 102000015779 HDL Lipoproteins Human genes 0.000 description 58
- 108060003412 GRP Proteins 0.000 description 57
- 102100004065 PLA2G7 Human genes 0.000 description 47
- 239000002609 media Substances 0.000 description 37
- 238000004166 bioassay Methods 0.000 description 35
- 150000002632 lipids Chemical class 0.000 description 35
- 102000004965 antibodies Human genes 0.000 description 34
- 108090001123 antibodies Proteins 0.000 description 34
- 125000003729 nucleotide group Chemical group 0.000 description 34
- 230000035492 administration Effects 0.000 description 33
- 239000002773 nucleotide Substances 0.000 description 33
- 206010028980 Neoplasm Diseases 0.000 description 28
- 230000002401 inhibitory effect Effects 0.000 description 28
- 102000004190 Enzymes Human genes 0.000 description 24
- 108090000790 Enzymes Proteins 0.000 description 24
- 239000002245 particle Substances 0.000 description 24
- 229940088598 Enzyme Drugs 0.000 description 23
- 101700009910 PON1 Proteins 0.000 description 23
- 102100019848 PON1 Human genes 0.000 description 23
- 230000002829 reduced Effects 0.000 description 23
- 230000001603 reducing Effects 0.000 description 23
- 230000001225 therapeutic Effects 0.000 description 23
- 230000036499 Half live Effects 0.000 description 21
- 239000000499 gel Substances 0.000 description 21
- 238000000034 method Methods 0.000 description 21
- 108020004707 nucleic acids Proteins 0.000 description 21
- 150000007523 nucleic acids Chemical class 0.000 description 21
- 229920000160 (ribonucleotides)n+m Polymers 0.000 description 20
- 102000003896 Myeloperoxidases Human genes 0.000 description 20
- 108090000235 Myeloperoxidases Proteins 0.000 description 20
- 239000000758 substrate Substances 0.000 description 20
- 239000003112 inhibitor Substances 0.000 description 19
- 150000003904 phospholipids Chemical class 0.000 description 19
- 101710005523 RUVBL1 Proteins 0.000 description 18
- 230000004044 response Effects 0.000 description 18
- 229920001850 Nucleic acid sequence Polymers 0.000 description 17
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 17
- 230000004141 reverse cholesterol transport Effects 0.000 description 17
- 238000001890 transfection Methods 0.000 description 17
- 239000003795 chemical substances by application Substances 0.000 description 15
- 230000003247 decreasing Effects 0.000 description 15
- 238000006471 dimerization reaction Methods 0.000 description 15
- 229940079593 drugs Drugs 0.000 description 15
- 238000000746 purification Methods 0.000 description 15
- 230000003248 secreting Effects 0.000 description 15
- 230000004913 activation Effects 0.000 description 14
- 238000004458 analytical method Methods 0.000 description 14
- 239000012228 culture supernatant Substances 0.000 description 14
- 230000035772 mutation Effects 0.000 description 14
- 102100014838 FCGRT Human genes 0.000 description 13
- MTCFGRXMJLQNBG-REOHCLBHSA-N L-serine Chemical compound OC[C@H](N)C(O)=O MTCFGRXMJLQNBG-REOHCLBHSA-N 0.000 description 13
- 230000015572 biosynthetic process Effects 0.000 description 13
- UIIMBOGNXHQVGW-UHFFFAOYSA-M buffer Substances [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 13
- 238000000338 in vitro Methods 0.000 description 13
- 230000032258 transport Effects 0.000 description 13
- 101710003435 FCGRT Proteins 0.000 description 12
- 210000002540 Macrophages Anatomy 0.000 description 12
- 235000004400 serine Nutrition 0.000 description 12
- 210000002966 Serum Anatomy 0.000 description 11
- 239000000427 antigen Substances 0.000 description 11
- 108091007172 antigens Proteins 0.000 description 11
- 102000038129 antigens Human genes 0.000 description 11
- 230000024881 catalytic activity Effects 0.000 description 11
- 235000018417 cysteine Nutrition 0.000 description 11
- 230000001976 improved Effects 0.000 description 11
- 238000006722 reduction reaction Methods 0.000 description 11
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 10
- 239000000370 acceptor Substances 0.000 description 10
- 125000000151 cysteine group Chemical group N[C@@H](CS)C(=O)* 0.000 description 10
- 230000003899 glycosylation Effects 0.000 description 10
- 238000006206 glycosylation reaction Methods 0.000 description 10
- 210000004962 mammalian cells Anatomy 0.000 description 10
- 230000003647 oxidation Effects 0.000 description 10
- 238000007254 oxidation reaction Methods 0.000 description 10
- 239000008194 pharmaceutical composition Substances 0.000 description 10
- 102000006496 Immunoglobulin Heavy Chains Human genes 0.000 description 9
- 108010019476 Immunoglobulin Heavy Chains Proteins 0.000 description 9
- 229940067631 Phospholipids Drugs 0.000 description 9
- 238000011161 development Methods 0.000 description 9
- 230000018109 developmental process Effects 0.000 description 9
- 239000000539 dimer Substances 0.000 description 9
- 238000001802 infusion Methods 0.000 description 9
- 102000015219 ATP Binding Cassette Transporter 1 Human genes 0.000 description 8
- 108010064466 ATP Binding Cassette Transporter 1 Proteins 0.000 description 8
- 206010003246 Arthritis Diseases 0.000 description 8
- 206010061218 Inflammation Diseases 0.000 description 8
- 210000004185 Liver Anatomy 0.000 description 8
- 206010029164 Nephrotic syndrome Diseases 0.000 description 8
- 108009000551 Nephrotic syndrome Proteins 0.000 description 8
- 101710002310 RNASE1 Proteins 0.000 description 8
- 102100001970 RNASE1 Human genes 0.000 description 8
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 8
- 238000010171 animal model Methods 0.000 description 8
- -1 cholesteryl ester Chemical class 0.000 description 8
- 238000003776 cleavage reaction Methods 0.000 description 8
- 238000005755 formation reaction Methods 0.000 description 8
- 230000004054 inflammatory process Effects 0.000 description 8
- 230000002018 overexpression Effects 0.000 description 8
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 8
- 241000701447 unidentified baculovirus Species 0.000 description 8
- 101710027066 ALB Proteins 0.000 description 7
- 210000003169 Central Nervous System Anatomy 0.000 description 7
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 7
- 241001465754 Metazoa Species 0.000 description 7
- 235000014680 Saccharomyces cerevisiae Nutrition 0.000 description 7
- 230000003110 anti-inflammatory Effects 0.000 description 7
- 102000030962 arylesterase Human genes 0.000 description 7
- 108010009043 arylesterase Proteins 0.000 description 7
- 230000001588 bifunctional Effects 0.000 description 7
- 230000002596 correlated Effects 0.000 description 7
- 230000002950 deficient Effects 0.000 description 7
- 239000001963 growth media Substances 0.000 description 7
- 230000003993 interaction Effects 0.000 description 7
- 239000003550 marker Substances 0.000 description 7
- 229920000642 polymer Polymers 0.000 description 7
- 238000003118 sandwich ELISA Methods 0.000 description 7
- 239000000243 solution Substances 0.000 description 7
- 210000001519 tissues Anatomy 0.000 description 7
- 229920000936 Agarose Polymers 0.000 description 6
- 241000894006 Bacteria Species 0.000 description 6
- 229920001405 Coding region Polymers 0.000 description 6
- 229920002676 Complementary DNA Polymers 0.000 description 6
- 241000282412 Homo Species 0.000 description 6
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 6
- 102000000853 LDL receptors Human genes 0.000 description 6
- 108010001831 LDL receptors Proteins 0.000 description 6
- 239000006146 Roswell Park Memorial Institute medium Substances 0.000 description 6
- 238000007792 addition Methods 0.000 description 6
- 238000001042 affinity chromatography Methods 0.000 description 6
- 230000002708 enhancing Effects 0.000 description 6
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 6
- 230000002068 genetic Effects 0.000 description 6
- 125000003630 glycyl group Chemical group [H]N([H])C([H])([H])C(*)=O 0.000 description 6
- 230000012010 growth Effects 0.000 description 6
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 description 6
- 239000011159 matrix material Substances 0.000 description 6
- 230000037361 pathway Effects 0.000 description 6
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N phenol Chemical compound OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 6
- 102000004196 processed proteins & peptides Human genes 0.000 description 6
- 108090000765 processed proteins & peptides Proteins 0.000 description 6
- 230000001681 protective Effects 0.000 description 6
- 241000894007 species Species 0.000 description 6
- 230000001131 transforming Effects 0.000 description 6
- 102000018619 Apolipoproteins A Human genes 0.000 description 5
- 108010027004 Apolipoproteins A Proteins 0.000 description 5
- 108020004635 Complementary DNA Proteins 0.000 description 5
- 229920001272 Exogenous DNA Polymers 0.000 description 5
- 229920003004 Extracellular RNA Polymers 0.000 description 5
- 108091007152 Extracellular RNA Proteins 0.000 description 5
- KDXKERNSBIXSRK-YFKPBYRVSA-N L-lysine Chemical compound NCCCC[C@H](N)C(O)=O KDXKERNSBIXSRK-YFKPBYRVSA-N 0.000 description 5
- FBOZXECLQNJBKD-ZDUSSCGKSA-N L-methotrexate Chemical compound C=1N=C2N=C(N)N=C(N)C2=NC=1CN(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FBOZXECLQNJBKD-ZDUSSCGKSA-N 0.000 description 5
- 102000004895 Lipoproteins Human genes 0.000 description 5
- 108090001030 Lipoproteins Proteins 0.000 description 5
- 239000004472 Lysine Substances 0.000 description 5
- 241000124008 Mammalia Species 0.000 description 5
- 206010061536 Parkinson's disease Diseases 0.000 description 5
- 210000002381 Plasma Anatomy 0.000 description 5
- 241000589517 Pseudomonas aeruginosa Species 0.000 description 5
- 229940055023 Pseudomonas aeruginosa Drugs 0.000 description 5
- 241000700159 Rattus Species 0.000 description 5
- 230000001154 acute Effects 0.000 description 5
- 230000003321 amplification Effects 0.000 description 5
- 230000003078 antioxidant Effects 0.000 description 5
- 230000001580 bacterial Effects 0.000 description 5
- 238000010804 cDNA synthesis Methods 0.000 description 5
- 239000000969 carrier Substances 0.000 description 5
- 239000003153 chemical reaction reagent Substances 0.000 description 5
- 239000003354 cholesterol ester transfer protein inhibitor Substances 0.000 description 5
- 230000000295 complement Effects 0.000 description 5
- 239000002299 complementary DNA Substances 0.000 description 5
- 230000001419 dependent Effects 0.000 description 5
- 238000000502 dialysis Methods 0.000 description 5
- 238000004520 electroporation Methods 0.000 description 5
- 238000000605 extraction Methods 0.000 description 5
- DHMQDGOQFOQNFH-UHFFFAOYSA-N glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 5
- 239000006166 lysate Substances 0.000 description 5
- 229960000485 methotrexate Drugs 0.000 description 5
- 238000010172 mouse model Methods 0.000 description 5
- 238000002703 mutagenesis Methods 0.000 description 5
- 231100000350 mutagenesis Toxicity 0.000 description 5
- 238000003199 nucleic acid amplification method Methods 0.000 description 5
- 230000000770 pro-inflamatory Effects 0.000 description 5
- 230000002633 protecting Effects 0.000 description 5
- 125000003616 serine group Chemical group [H]N([H])[C@]([H])(C(=O)[*])C(O[H])([H])[H] 0.000 description 5
- 239000007787 solid Substances 0.000 description 5
- 230000002194 synthesizing Effects 0.000 description 5
- 231100000419 toxicity Toxicity 0.000 description 5
- 230000001988 toxicity Effects 0.000 description 5
- 230000004614 tumor growth Effects 0.000 description 5
- 230000003442 weekly Effects 0.000 description 5
- 239000004475 Arginine Substances 0.000 description 4
- OWMVSZAMULFTJU-UHFFFAOYSA-N Bis-tris methane Chemical compound OCCN(CCO)C(CO)(CO)CO OWMVSZAMULFTJU-UHFFFAOYSA-N 0.000 description 4
- 210000004556 Brain Anatomy 0.000 description 4
- 241000283707 Capra Species 0.000 description 4
- 108090000695 Cytokines Proteins 0.000 description 4
- 102000004127 Cytokines Human genes 0.000 description 4
- 238000002965 ELISA Methods 0.000 description 4
- 241000588724 Escherichia coli Species 0.000 description 4
- 108010001336 Horseradish Peroxidase Proteins 0.000 description 4
- XUJNEKJLAYXESH-REOHCLBHSA-N L-cysteine Chemical compound SC[C@H](N)C(O)=O XUJNEKJLAYXESH-REOHCLBHSA-N 0.000 description 4
- AGPKZVBTJJNPAG-WHFBIAKZSA-N L-isoleucine Chemical compound CC[C@H](C)[C@H](N)C(O)=O AGPKZVBTJJNPAG-WHFBIAKZSA-N 0.000 description 4
- AYFVYJQAPQTCCC-GBXIJSLDSA-N L-threonine Chemical compound C[C@@H](O)[C@H](N)C(O)=O AYFVYJQAPQTCCC-GBXIJSLDSA-N 0.000 description 4
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 4
- 210000001616 Monocytes Anatomy 0.000 description 4
- 241000699666 Mus <mouse, genus> Species 0.000 description 4
- 208000010125 Myocardial Infarction Diseases 0.000 description 4
- 108091005503 Nucleic proteins Proteins 0.000 description 4
- 101000031113 PON1 Proteins 0.000 description 4
- 210000001744 T-Lymphocytes Anatomy 0.000 description 4
- 102100012088 TLR3 Human genes 0.000 description 4
- 101700023131 TLR3 Proteins 0.000 description 4
- 102100006355 TLR7 Human genes 0.000 description 4
- 101700075266 TLR7 Proteins 0.000 description 4
- 150000007513 acids Chemical class 0.000 description 4
- 230000003143 atherosclerotic Effects 0.000 description 4
- 230000001363 autoimmune Effects 0.000 description 4
- 239000011324 bead Substances 0.000 description 4
- 230000000271 cardiovascular Effects 0.000 description 4
- 230000001413 cellular Effects 0.000 description 4
- 238000006243 chemical reaction Methods 0.000 description 4
- 238000004587 chromatography analysis Methods 0.000 description 4
- 230000001684 chronic Effects 0.000 description 4
- 150000001875 compounds Chemical class 0.000 description 4
- 230000029087 digestion Effects 0.000 description 4
- 201000002491 encephalomyelitis Diseases 0.000 description 4
- 239000002158 endotoxin Substances 0.000 description 4
- 238000009472 formulation Methods 0.000 description 4
- 102000026159 human PON1 protein Human genes 0.000 description 4
- 239000012274 immune-checkpoint protein inhibitor Substances 0.000 description 4
- 229940121650 immune-checkpoint protein inhibitors Drugs 0.000 description 4
- 238000002347 injection Methods 0.000 description 4
- 239000007924 injection Substances 0.000 description 4
- 230000003834 intracellular Effects 0.000 description 4
- 238000002372 labelling Methods 0.000 description 4
- 230000000051 modifying Effects 0.000 description 4
- 108010045030 monoclonal antibodies Proteins 0.000 description 4
- 102000005614 monoclonal antibodies Human genes 0.000 description 4
- 230000004770 neurodegeneration Effects 0.000 description 4
- 230000003389 potentiating Effects 0.000 description 4
- 238000002360 preparation method Methods 0.000 description 4
- 238000001742 protein purification Methods 0.000 description 4
- 102000005962 receptors Human genes 0.000 description 4
- 108020003175 receptors Proteins 0.000 description 4
- 238000011160 research Methods 0.000 description 4
- 229920005989 resin Polymers 0.000 description 4
- 239000011347 resin Substances 0.000 description 4
- 150000003384 small molecules Chemical class 0.000 description 4
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 4
- 238000003786 synthesis reaction Methods 0.000 description 4
- 241000701161 unidentified adenovirus Species 0.000 description 4
- XSQUKJJJFZCRTK-UHFFFAOYSA-N urea Chemical compound NC(N)=O XSQUKJJJFZCRTK-UHFFFAOYSA-N 0.000 description 4
- 102000013918 Apolipoproteins E Human genes 0.000 description 3
- 108010025628 Apolipoproteins E Proteins 0.000 description 3
- 101700004551 BRAF Proteins 0.000 description 3
- 210000001218 Blood-Brain Barrier Anatomy 0.000 description 3
- 108060006634 CAMP Proteins 0.000 description 3
- 108010002025 CER-001 Proteins 0.000 description 3
- 208000003398 Cerebral Arterial Disease Diseases 0.000 description 3
- 108020004705 Codon Proteins 0.000 description 3
- 102000008186 Collagen Human genes 0.000 description 3
- 108010035532 Collagen Proteins 0.000 description 3
- 241000699802 Cricetulus griseus Species 0.000 description 3
- 206010012601 Diabetes mellitus Diseases 0.000 description 3
- 208000000999 Encephalomyelitis, Autoimmune, Experimental Diseases 0.000 description 3
- 108010087819 Fc receptors Proteins 0.000 description 3
- 102000009109 Fc receptors Human genes 0.000 description 3
- 239000004471 Glycine Substances 0.000 description 3
- 241000238631 Hexapoda Species 0.000 description 3
- 229960000310 ISOLEUCINE Drugs 0.000 description 3
- 102000004877 Insulin Human genes 0.000 description 3
- 108090001061 Insulin Proteins 0.000 description 3
- 210000003734 Kidney Anatomy 0.000 description 3
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 3
- COLNVLDHVKWLRT-QMMMGPOBSA-N L-phenylalanine Chemical compound OC(=O)[C@@H](N)CC1=CC=CC=C1 COLNVLDHVKWLRT-QMMMGPOBSA-N 0.000 description 3
- KZSNJWFQEVHDMF-BYPYZUCNSA-N L-valine Chemical compound CC(C)[C@H](N)C(O)=O KZSNJWFQEVHDMF-BYPYZUCNSA-N 0.000 description 3
- 108010007622 LDL Lipoproteins Proteins 0.000 description 3
- 241001452677 Ogataea methanolica Species 0.000 description 3
- 241000283973 Oryctolagus cuniculus Species 0.000 description 3
- 108020002230 Pancreatic ribonucleases Proteins 0.000 description 3
- 102000005891 Pancreatic ribonucleases Human genes 0.000 description 3
- 108010003541 Platelet Activating Factor Proteins 0.000 description 3
- 229920002319 Poly(methyl acrylate) Polymers 0.000 description 3
- 229920001213 Polysorbate 20 Polymers 0.000 description 3
- 206010063401 Primary progressive multiple sclerosis Diseases 0.000 description 3
- 206010037162 Psoriatic arthropathy Diseases 0.000 description 3
- 101710005015 RNASEL Proteins 0.000 description 3
- 241000219061 Rheum Species 0.000 description 3
- 241000785686 Sander Species 0.000 description 3
- 206010039491 Sarcoma Diseases 0.000 description 3
- 241000256251 Spodoptera frugiperda Species 0.000 description 3
- 102100015249 VEGFA Human genes 0.000 description 3
- 108010073929 Vascular Endothelial Growth Factor A Proteins 0.000 description 3
- 241000700605 Viruses Species 0.000 description 3
- 238000009825 accumulation Methods 0.000 description 3
- 239000004480 active ingredient Substances 0.000 description 3
- 235000004279 alanine Nutrition 0.000 description 3
- 230000001093 anti-cancer Effects 0.000 description 3
- 230000000778 atheroprotective Effects 0.000 description 3
- 101700027839 bah Proteins 0.000 description 3
- 239000012501 chromatography media Substances 0.000 description 3
- 239000007979 citrate buffer Substances 0.000 description 3
- 229920001436 collagen Polymers 0.000 description 3
- 229960005188 collagen Drugs 0.000 description 3
- 230000001808 coupling Effects 0.000 description 3
- 238000010168 coupling process Methods 0.000 description 3
- 238000005859 coupling reaction Methods 0.000 description 3
- SWYGLBJVTQMYSE-UHFFFAOYSA-N cyclohexyloxy(methyl)phosphinic acid Chemical compound CP(O)(=O)OC1CCCCC1 SWYGLBJVTQMYSE-UHFFFAOYSA-N 0.000 description 3
- 230000002354 daily Effects 0.000 description 3
- 238000010790 dilution Methods 0.000 description 3
- 239000012153 distilled water Substances 0.000 description 3
- 150000002019 disulfides Chemical class 0.000 description 3
- 238000001962 electrophoresis Methods 0.000 description 3
- 239000003797 essential amino acid Substances 0.000 description 3
- 235000020776 essential amino acid Nutrition 0.000 description 3
- 210000003527 eukaryotic cell Anatomy 0.000 description 3
- 230000005714 functional activity Effects 0.000 description 3
- 230000002440 hepatic Effects 0.000 description 3
- 235000009200 high fat diet Nutrition 0.000 description 3
- 230000002209 hydrophobic Effects 0.000 description 3
- 230000002163 immunogen Effects 0.000 description 3
- 239000007943 implant Substances 0.000 description 3
- 230000002779 inactivation Effects 0.000 description 3
- 238000003780 insertion Methods 0.000 description 3
- 238000007917 intracranial administration Methods 0.000 description 3
- 238000001990 intravenous administration Methods 0.000 description 3
- 150000002500 ions Chemical class 0.000 description 3
- 239000003446 ligand Substances 0.000 description 3
- 230000035800 maturation Effects 0.000 description 3
- 238000005259 measurement Methods 0.000 description 3
- 229910052751 metal Inorganic materials 0.000 description 3
- 239000002184 metal Substances 0.000 description 3
- 230000004048 modification Effects 0.000 description 3
- 238000006011 modification reaction Methods 0.000 description 3
- 239000000178 monomer Substances 0.000 description 3
- 230000000324 neuroprotective Effects 0.000 description 3
- WLJVXDMOQOGPHL-UHFFFAOYSA-M phenylacetate Chemical compound [O-]C(=O)CC1=CC=CC=C1 WLJVXDMOQOGPHL-UHFFFAOYSA-M 0.000 description 3
- 229940049953 phenylacetate Drugs 0.000 description 3
- 229920002401 polyacrylamide Polymers 0.000 description 3
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 3
- 239000000843 powder Substances 0.000 description 3
- 230000002797 proteolythic Effects 0.000 description 3
- 201000001263 psoriatic arthritis Diseases 0.000 description 3
- 238000011084 recovery Methods 0.000 description 3
- 230000028327 secretion Effects 0.000 description 3
- 238000001998 small-angle neutron scattering Methods 0.000 description 3
- HEMHJVSKTPXQMS-UHFFFAOYSA-M sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 3
- 230000000638 stimulation Effects 0.000 description 3
- 239000006228 supernatant Substances 0.000 description 3
- 229960005486 vaccines Drugs 0.000 description 3
- 239000004474 valine Substances 0.000 description 3
- 230000002792 vascular Effects 0.000 description 3
- 238000001262 western blot Methods 0.000 description 3
- SWEICGMKXPNXNU-UHFFFAOYSA-N 1,2-dihydroindazol-3-one Chemical class C1=CC=C2C(O)=NNC2=C1 SWEICGMKXPNXNU-UHFFFAOYSA-N 0.000 description 2
- BAXOFTOLAUCFNW-UHFFFAOYSA-N 1H-indazole Chemical class C1=CC=C2C=NNC2=C1 BAXOFTOLAUCFNW-UHFFFAOYSA-N 0.000 description 2
- LENZDBCJOHFCAS-UHFFFAOYSA-N 2-amino-2-(hydroxymethyl)propane-1,3-diol Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 2
- ZWEHNKRNPOVVGH-UHFFFAOYSA-N 2-butanone Chemical compound CCC(C)=O ZWEHNKRNPOVVGH-UHFFFAOYSA-N 0.000 description 2
- 102100001249 ALB Human genes 0.000 description 2
- 101000447387 APOA1 Proteins 0.000 description 2
- 108010061171 Abatacept Proteins 0.000 description 2
- 210000000612 Antigen-Presenting Cells Anatomy 0.000 description 2
- 208000003343 Antiphospholipid Syndrome Diseases 0.000 description 2
- 206010003645 Atopy Diseases 0.000 description 2
- 108090000206 Autoantibodies Proteins 0.000 description 2
- 102000003852 Autoantibodies Human genes 0.000 description 2
- 241000201370 Autographa californica nucleopolyhedrovirus Species 0.000 description 2
- 241000271566 Aves Species 0.000 description 2
- 210000004369 Blood Anatomy 0.000 description 2
- 206010005949 Bone cancer Diseases 0.000 description 2
- 229940077737 Brain-Derived Neurotrophic Factor Drugs 0.000 description 2
- 102000004219 Brain-Derived Neurotrophic Factor Human genes 0.000 description 2
- 108090000715 Brain-Derived Neurotrophic Factor Proteins 0.000 description 2
- 102100008428 CCL2 Human genes 0.000 description 2
- 101700006000 CCL2 Proteins 0.000 description 2
- 102100013137 CD40 Human genes 0.000 description 2
- 101710040446 CD40 Proteins 0.000 description 2
- AIYUHDOJVYHVIT-UHFFFAOYSA-M Caesium chloride Chemical compound [Cl-].[Cs+] AIYUHDOJVYHVIT-UHFFFAOYSA-M 0.000 description 2
- 210000000170 Cell Membrane Anatomy 0.000 description 2
- 206010009900 Colitis ulcerative Diseases 0.000 description 2
- 206010011401 Crohn's disease Diseases 0.000 description 2
- 210000000805 Cytoplasm Anatomy 0.000 description 2
- WQZGKKKJIJFFOK-GASJEMHNSA-N D-Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 2
- QIVBCDIJIAJPQS-SECBINFHSA-N D-tryptophane Chemical compound C1=CC=C2C(C[C@@H](N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-SECBINFHSA-N 0.000 description 2
- IQFYYKKMVGJFEH-XLPZGREQSA-N DEOXYTHYMIDINE Chemical compound O=C1NC(=O)C(C)=CN1[C@@H]1O[C@H](CO)[C@@H](O)C1 IQFYYKKMVGJFEH-XLPZGREQSA-N 0.000 description 2
- 101710007887 DHFR Proteins 0.000 description 2
- 102100005838 DHFR Human genes 0.000 description 2
- 229920002307 Dextran Polymers 0.000 description 2
- 206010059866 Drug resistance Diseases 0.000 description 2
- 241000196324 Embryophyta Species 0.000 description 2
- 241000282326 Felis catus Species 0.000 description 2
- PNNNRSAQSRJVSB-SLPGGIOYSA-N Fucose Natural products C[C@H](O)[C@@H](O)[C@H](O)[C@H](O)C=O PNNNRSAQSRJVSB-SLPGGIOYSA-N 0.000 description 2
- 206010051066 Gastrointestinal stromal tumour Diseases 0.000 description 2
- 206010018364 Glomerulonephritis Diseases 0.000 description 2
- 229960004198 Guanidine Drugs 0.000 description 2
- PJJJBBJSCAKJQF-UHFFFAOYSA-N Guanidinium chloride Chemical compound [Cl-].NC(N)=[NH2+] PJJJBBJSCAKJQF-UHFFFAOYSA-N 0.000 description 2
- VEXZGXHMUGYJMC-UHFFFAOYSA-N HCl Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 2
- 208000006454 Hepatitis Diseases 0.000 description 2
- FDGQSTZJBFJUBT-UHFFFAOYSA-N Hypoxanthine Chemical compound O=C1NC=NC2=C1NC=N2 FDGQSTZJBFJUBT-UHFFFAOYSA-N 0.000 description 2
- 101000187184 INSR Proteins 0.000 description 2
- 101710006572 ITGAM Proteins 0.000 description 2
- 102100019441 ITGAM Human genes 0.000 description 2
- 108010073807 IgG Receptors Proteins 0.000 description 2
- 102000009490 IgG Receptors Human genes 0.000 description 2
- 108010058683 Immobilized Proteins Proteins 0.000 description 2
- 210000000987 Immune System Anatomy 0.000 description 2
- 102000018071 Immunoglobulin Fc Fragments Human genes 0.000 description 2
- 108010091135 Immunoglobulin Fc Fragments Proteins 0.000 description 2
- 102000013463 Immunoglobulin Light Chains Human genes 0.000 description 2
- 108010065825 Immunoglobulin Light Chains Proteins 0.000 description 2
- XKTZWUACRZHVAN-VADRZIEHSA-N Interleukin-8 Chemical compound C([C@H](NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@@H](NC(C)=O)CCSC)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H]([C@@H](C)O)C(=O)NCC(=O)N[C@@H](CCSC)C(=O)N1[C@H](CCC1)C(=O)N1[C@H](CCC1)C(=O)N[C@@H](C)C(=O)N[C@H](CC(O)=O)C(=O)N[C@H](CCC(O)=O)C(=O)N[C@H](CC(O)=O)C(=O)N[C@H](CC=1C=CC(O)=CC=1)C(=O)N[C@H](CO)C(=O)N1[C@H](CCC1)C(N)=O)C1=CC=CC=C1 XKTZWUACRZHVAN-VADRZIEHSA-N 0.000 description 2
- 229940096397 Interleukin-8 Drugs 0.000 description 2
- 102000004890 Interleukin-8 Human genes 0.000 description 2
- 108090001007 Interleukin-8 Proteins 0.000 description 2
- 208000002551 Irritable Bowel Syndrome Diseases 0.000 description 2
- 208000003456 Juvenile Arthritis Diseases 0.000 description 2
- 206010059176 Juvenile idiopathic arthritis Diseases 0.000 description 2
- 208000007766 Kaposi Sarcoma Diseases 0.000 description 2
- 208000001083 Kidney Disease Diseases 0.000 description 2
- 241000235058 Komagataella pastoris Species 0.000 description 2
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-N 0.000 description 2
- 125000000174 L-prolyl group Chemical group [H]N1C([H])([H])C([H])([H])C([H])([H])[C@@]1([H])C(*)=O 0.000 description 2
- 102000007330 LDL Lipoproteins Human genes 0.000 description 2
- 210000004072 Lung Anatomy 0.000 description 2
- 208000005777 Lupus Nephritis Diseases 0.000 description 2
- 229920002521 Macromolecule Polymers 0.000 description 2
- 206010027191 Meningioma Diseases 0.000 description 2
- 208000001145 Metabolic Syndrome Diseases 0.000 description 2
- 101710017500 MitHPPK/DHPS Proteins 0.000 description 2
- 241000699660 Mus musculus Species 0.000 description 2
- 229940053128 Nerve Growth Factor Drugs 0.000 description 2
- 102000015336 Nerve Growth Factor Human genes 0.000 description 2
- 108010025020 Nerve Growth Factor Proteins 0.000 description 2
- 208000007538 Neurilemmoma Diseases 0.000 description 2
- 229940032018 Neurotrophin 3 Drugs 0.000 description 2
- 102000004230 Neurotrophin-3 Human genes 0.000 description 2
- 108090000742 Neurotrophin-3 Proteins 0.000 description 2
- 229940053207 Niacin Drugs 0.000 description 2
- 229940035567 Orencia Drugs 0.000 description 2
- 206010033128 Ovarian cancer Diseases 0.000 description 2
- 210000001672 Ovary Anatomy 0.000 description 2
- 108009000578 Oxidative Stress Proteins 0.000 description 2
- 101710043203 P23p89 Proteins 0.000 description 2
- 101700048149 PON3 Proteins 0.000 description 2
- 102100019180 PON3 Human genes 0.000 description 2
- 208000006551 Parasitic Disease Diseases 0.000 description 2
- 210000001322 Periplasm Anatomy 0.000 description 2
- 102000003728 Peroxisome Proliferator-Activated Receptors Human genes 0.000 description 2
- 108090000029 Peroxisome Proliferator-Activated Receptors Proteins 0.000 description 2
- 229960005190 Phenylalanine Drugs 0.000 description 2
- 241000235648 Pichia Species 0.000 description 2
- 241000288906 Primates Species 0.000 description 2
- 229920000320 RNA (poly(A)) Polymers 0.000 description 2
- 108020004412 RNA 3' Polyadenylation Signals Proteins 0.000 description 2
- 101700016196 RNP30 Proteins 0.000 description 2
- 108010033725 Recombinant Proteins Proteins 0.000 description 2
- 102000007312 Recombinant Proteins Human genes 0.000 description 2
- 208000007400 Relapsing-Remitting Multiple Sclerosis Diseases 0.000 description 2
- 235000003534 Saccharomyces carlsbergensis Nutrition 0.000 description 2
- 229940081969 Saccharomyces cerevisiae Drugs 0.000 description 2
- 206010039667 Schwannoma Diseases 0.000 description 2
- 206010039710 Scleroderma Diseases 0.000 description 2
- 229920002684 Sepharose Polymers 0.000 description 2
- 206010040767 Sjogren's syndrome Diseases 0.000 description 2
- 239000004473 Threonine Substances 0.000 description 2
- 208000007536 Thrombosis Diseases 0.000 description 2
- 102000004338 Transferrin Human genes 0.000 description 2
- 108090000901 Transferrin Proteins 0.000 description 2
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 2
- 229960003697 abatacept Drugs 0.000 description 2
- 125000002252 acyl group Chemical group 0.000 description 2
- 239000000443 aerosol Substances 0.000 description 2
- 125000003295 alanine group Chemical group N[C@@H](C)C(=O)* 0.000 description 2
- 230000004075 alteration Effects 0.000 description 2
- 230000003466 anti-cipated Effects 0.000 description 2
- 230000000259 anti-tumor Effects 0.000 description 2
- 239000003963 antioxidant agent Substances 0.000 description 2
- 235000006708 antioxidants Nutrition 0.000 description 2
- 125000000511 arginine group Chemical group N[C@@H](CCCNC(N)=N)C(=O)* 0.000 description 2
- 230000002238 attenuated Effects 0.000 description 2
- 201000004988 autoimmune vasculitis Diseases 0.000 description 2
- WARCRYXKINZHGQ-UHFFFAOYSA-N benzohydrazide Chemical compound NNC(=O)C1=CC=CC=C1 WARCRYXKINZHGQ-UHFFFAOYSA-N 0.000 description 2
- 239000008280 blood Substances 0.000 description 2
- 238000000055 blue native polyacrylamide gel electrophoresis Methods 0.000 description 2
- 125000000484 butyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 2
- 239000004202 carbamide Substances 0.000 description 2
- 125000000837 carbohydrate group Chemical group 0.000 description 2
- 150000001720 carbohydrates Chemical class 0.000 description 2
- 230000003197 catalytic Effects 0.000 description 2
- 238000004113 cell culture Methods 0.000 description 2
- 230000003931 cognitive performance Effects 0.000 description 2
- 230000021615 conjugation Effects 0.000 description 2
- 238000010276 construction Methods 0.000 description 2
- 230000001276 controlling effect Effects 0.000 description 2
- 238000007796 conventional method Methods 0.000 description 2
- 230000001086 cytosolic Effects 0.000 description 2
- 238000007405 data analysis Methods 0.000 description 2
- 239000003398 denaturant Substances 0.000 description 2
- 239000003599 detergent Substances 0.000 description 2
- 230000004069 differentiation Effects 0.000 description 2
- 239000002552 dosage form Substances 0.000 description 2
- 238000010828 elution Methods 0.000 description 2
- 201000011523 endocrine gland cancer Diseases 0.000 description 2
- 238000011067 equilibration Methods 0.000 description 2
- 230000005284 excitation Effects 0.000 description 2
- 239000000284 extract Substances 0.000 description 2
- 239000012530 fluid Substances 0.000 description 2
- 238000005194 fractionation Methods 0.000 description 2
- 230000002538 fungal Effects 0.000 description 2
- 201000011243 gastrointestinal stromal tumor Diseases 0.000 description 2
- 238000001502 gel electrophoresis Methods 0.000 description 2
- 239000008103 glucose Substances 0.000 description 2
- 125000000404 glutamine group Chemical group N[C@@H](CCC(N)=O)C(=O)* 0.000 description 2
- 150000004676 glycans Chemical class 0.000 description 2
- 239000008187 granular material Substances 0.000 description 2
- 230000005484 gravity Effects 0.000 description 2
- 108010044853 histidine-rich proteins Proteins 0.000 description 2
- 102000019638 human APOA1 protein Human genes 0.000 description 2
- 102000034561 human INSR protein Human genes 0.000 description 2
- 229910000041 hydrogen chloride Inorganic materials 0.000 description 2
- 238000006460 hydrolysis reaction Methods 0.000 description 2
- 230000001900 immune effect Effects 0.000 description 2
- 230000002458 infectious Effects 0.000 description 2
- 230000028709 inflammatory response Effects 0.000 description 2
- 102000010681 interleukin-8 receptors Human genes 0.000 description 2
- 108010038415 interleukin-8 receptors Proteins 0.000 description 2
- 238000007913 intrathecal administration Methods 0.000 description 2
- 238000002955 isolation Methods 0.000 description 2
- 201000002215 juvenile rheumatoid arthritis Diseases 0.000 description 2
- 238000003367 kinetic assay Methods 0.000 description 2
- 230000004576 lipid-binding Effects 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- 238000005567 liquid scintillation counting Methods 0.000 description 2
- 239000012160 loading buffer Substances 0.000 description 2
- 125000003588 lysine group Chemical group [H]N([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])(N([H])[H])C(*)=O 0.000 description 2
- 239000000463 material Substances 0.000 description 2
- 125000001360 methionine group Chemical group N[C@@H](CCSC)C(=O)* 0.000 description 2
- 239000004005 microsphere Substances 0.000 description 2
- 238000001426 native polyacrylamide gel electrophoresis Methods 0.000 description 2
- 230000001537 neural Effects 0.000 description 2
- 230000000926 neurological Effects 0.000 description 2
- DFPAKSUCGFBDDF-UHFFFAOYSA-N nicotinamide Chemical compound NC(=O)C1=CC=CN=C1 DFPAKSUCGFBDDF-UHFFFAOYSA-N 0.000 description 2
- 229960003512 nicotinic acid Drugs 0.000 description 2
- 235000001968 nicotinic acid Nutrition 0.000 description 2
- 239000011664 nicotinic acid Substances 0.000 description 2
- IJGRMHOSHXDMSA-UHFFFAOYSA-N nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 2
- 230000003000 nontoxic Effects 0.000 description 2
- 231100000252 nontoxic Toxicity 0.000 description 2
- 235000015097 nutrients Nutrition 0.000 description 2
- 210000000056 organs Anatomy 0.000 description 2
- 230000003204 osmotic Effects 0.000 description 2
- 230000036542 oxidative stress Effects 0.000 description 2
- 238000002640 oxygen therapy Methods 0.000 description 2
- RZVAJINKPMORJF-UHFFFAOYSA-N p-acetaminophenol Chemical compound CC(=O)NC1=CC=C(O)C=C1 RZVAJINKPMORJF-UHFFFAOYSA-N 0.000 description 2
- 229960005489 paracetamol Drugs 0.000 description 2
- 230000008506 pathogenesis Effects 0.000 description 2
- 239000002953 phosphate buffered saline Substances 0.000 description 2
- 229940096700 plain lipid modifying drugs Fibrates Drugs 0.000 description 2
- 238000007747 plating Methods 0.000 description 2
- 229920001606 poly(lactic acid-co-glycolic acid) Polymers 0.000 description 2
- 229920002704 polyhistidine Polymers 0.000 description 2
- 238000003752 polymerase chain reaction Methods 0.000 description 2
- 238000001556 precipitation Methods 0.000 description 2
- 230000002265 prevention Effects 0.000 description 2
- 239000000047 product Substances 0.000 description 2
- 230000000750 progressive Effects 0.000 description 2
- 108020001580 protein domains Proteins 0.000 description 2
- 238000000575 proteomic Methods 0.000 description 2
- 201000004681 psoriasis Diseases 0.000 description 2
- 230000018612 quorum sensing Effects 0.000 description 2
- 230000002285 radioactive Effects 0.000 description 2
- 108091007521 restriction endonucleases Proteins 0.000 description 2
- 230000000717 retained Effects 0.000 description 2
- 230000035945 sensitivity Effects 0.000 description 2
- 239000004017 serum-free culture media Substances 0.000 description 2
- 230000011664 signaling Effects 0.000 description 2
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 2
- PXIPVTKHYLBLMZ-UHFFFAOYSA-N sodium azide Chemical compound [Na+].[N-]=[N+]=[N-] PXIPVTKHYLBLMZ-UHFFFAOYSA-N 0.000 description 2
- 239000011780 sodium chloride Substances 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- 238000001228 spectrum Methods 0.000 description 2
- 238000011105 stabilization Methods 0.000 description 2
- 230000000087 stabilizing Effects 0.000 description 2
- 238000007920 subcutaneous administration Methods 0.000 description 2
- 239000000126 substance Substances 0.000 description 2
- 125000001424 substituent group Chemical group 0.000 description 2
- 230000001629 suppression Effects 0.000 description 2
- 230000002459 sustained Effects 0.000 description 2
- 125000003396 thiol group Chemical group [H]S* 0.000 description 2
- 201000002510 thyroid cancer Diseases 0.000 description 2
- 238000010361 transduction Methods 0.000 description 2
- 230000026683 transduction Effects 0.000 description 2
- 239000012581 transferrin Substances 0.000 description 2
- 230000001052 transient Effects 0.000 description 2
- 238000003146 transient transfection Methods 0.000 description 2
- 238000004450 types of analysis Methods 0.000 description 2
- 201000006704 ulcerative colitis Diseases 0.000 description 2
- 230000035899 viability Effects 0.000 description 2
- 230000003612 virological Effects 0.000 description 2
- DSNRZTVNAUGXKR-UHFFFAOYSA-N (2,3-diethyl-4-nitrophenyl) dihydrogen phosphate Chemical compound CCC1=C(OP(O)(O)=O)C=CC([N+]([O-])=O)=C1CC DSNRZTVNAUGXKR-UHFFFAOYSA-N 0.000 description 1
- XIFDBACLRPLVEQ-BXRBKJIMSA-L (2S)-2-aminobutanedioate;(2S)-2,4-diamino-4-oxobutanoic acid Chemical compound OC(=O)[C@@H](N)CC(N)=O.[O-]C(=O)[C@@H](N)CC([O-])=O XIFDBACLRPLVEQ-BXRBKJIMSA-L 0.000 description 1
- BKKWZCSSYWYNDS-JEDNCBNOSA-N 2-aminoacetic acid;(2S)-2,6-diaminohexanoic acid Chemical compound NCC(O)=O.NCCCC[C@H](N)C(O)=O BKKWZCSSYWYNDS-JEDNCBNOSA-N 0.000 description 1
- GUPXYSSGJWIURR-UHFFFAOYSA-N 3-octoxypropane-1,2-diol Chemical compound CCCCCCCCOCC(O)CO GUPXYSSGJWIURR-UHFFFAOYSA-N 0.000 description 1
- 102100007787 APOA1 Human genes 0.000 description 1
- 229940022659 Acetaminophen Drugs 0.000 description 1
- 241000228431 Acremonium chrysogenum Species 0.000 description 1
- 102000019632 Acyl transferases Human genes 0.000 description 1
- 108091022082 Acyl transferases Proteins 0.000 description 1
- 201000004304 Addison's disease Diseases 0.000 description 1
- 206010052747 Adenocarcinoma pancreas Diseases 0.000 description 1
- 241000589156 Agrobacterium rhizogenes Species 0.000 description 1
- BFNBIHQBYMNNAN-UHFFFAOYSA-N Ammonium sulfate Chemical compound N.N.OS(O)(=O)=O BFNBIHQBYMNNAN-UHFFFAOYSA-N 0.000 description 1
- AVKUERGKIZMTKX-NJBDSQKTSA-N Ampicillin Chemical compound C1([C@@H](N)C(=O)N[C@H]2[C@H]3SC([C@@H](N3C2=O)C(O)=O)(C)C)=CC=CC=C1 AVKUERGKIZMTKX-NJBDSQKTSA-N 0.000 description 1
- 206010002022 Amyloidosis Diseases 0.000 description 1
- 206010002023 Amyloidosis Diseases 0.000 description 1
- 206010061424 Anal cancer Diseases 0.000 description 1
- 206010002224 Anaplastic astrocytoma Diseases 0.000 description 1
- 206010073128 Anaplastic oligodendroglioma Diseases 0.000 description 1
- 208000009094 Anemia, Hemolytic, Autoimmune Diseases 0.000 description 1
- 206010002556 Ankylosing spondylitis Diseases 0.000 description 1
- 108010047814 Antigen-Antibody Complex Proteins 0.000 description 1
- 208000009400 Aortic Disease Diseases 0.000 description 1
- RVEWUBJVAHOGKA-WOYAITHZSA-N Arginine glutamate Chemical compound OC(=O)[C@@H](N)CCC(O)=O.OC(=O)[C@@H](N)CCCNC(N)=N RVEWUBJVAHOGKA-WOYAITHZSA-N 0.000 description 1
- 229960001230 Asparagine Drugs 0.000 description 1
- 229940009098 Aspartate Drugs 0.000 description 1
- 241000228212 Aspergillus Species 0.000 description 1
- 206010003571 Astrocytoma Diseases 0.000 description 1
- 241000972773 Aulopiformes Species 0.000 description 1
- 208000005783 Autoimmune Thyroiditis Diseases 0.000 description 1
- 206010069002 Autoimmune pancreatitis Diseases 0.000 description 1
- 102100004328 BRAF Human genes 0.000 description 1
- 241000193830 Bacillus <bacterium> Species 0.000 description 1
- 206010004146 Basal cell carcinoma Diseases 0.000 description 1
- 108010005144 Bevacizumab Proteins 0.000 description 1
- 241000212384 Bifora Species 0.000 description 1
- 210000003445 Biliary Tract Anatomy 0.000 description 1
- 206010004661 Biliary cirrhosis primary Diseases 0.000 description 1
- 108010006654 Bleomycin Proteins 0.000 description 1
- 229960001561 Bleomycin Drugs 0.000 description 1
- 206010006007 Bone sarcoma Diseases 0.000 description 1
- GXJABQQUPOEUTA-RDJZCZTQSA-N Bortezomib Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)B(O)O)NC(=O)C=1N=CC=NC=1)C1=CC=CC=C1 GXJABQQUPOEUTA-RDJZCZTQSA-N 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 206010048962 Brain oedema Diseases 0.000 description 1
- 206010006187 Breast cancer Diseases 0.000 description 1
- 206010055113 Breast cancer metastatic Diseases 0.000 description 1
- 102000025380 C-Reactive Protein Human genes 0.000 description 1
- 108010074051 C-Reactive Protein Proteins 0.000 description 1
- 102100008431 CCL26 Human genes 0.000 description 1
- 101700040437 CCL26 Proteins 0.000 description 1
- 102100016449 CCL5 Human genes 0.000 description 1
- 125000006414 CCl Chemical group ClC* 0.000 description 1
- 102100013077 CD4 Human genes 0.000 description 1
- 101700022938 CD4 Proteins 0.000 description 1
- 102100008191 CD8A Human genes 0.000 description 1
- 101700054655 CD8A Proteins 0.000 description 1
- 101000006026 CETP Proteins 0.000 description 1
- 102100008895 CLEC3B Human genes 0.000 description 1
- 101700066277 COS-1 Proteins 0.000 description 1
- 101700060761 COS7 Proteins 0.000 description 1
- 102100006400 CSF2 Human genes 0.000 description 1
- 102100005310 CTLA4 Human genes 0.000 description 1
- 101700054183 CTLA4 Proteins 0.000 description 1
- 241000222128 Candida maltosa Species 0.000 description 1
- 241000282465 Canis Species 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- 229920002134 Carboxymethyl cellulose Polymers 0.000 description 1
- 208000002458 Carcinoid Tumor Diseases 0.000 description 1
- 206010007275 Carcinoid tumour Diseases 0.000 description 1
- 210000001715 Carotid Arteries Anatomy 0.000 description 1
- 206010007953 Central nervous system lymphoma Diseases 0.000 description 1
- 206010008342 Cervix carcinoma Diseases 0.000 description 1
- 108010022830 Cetuximab Proteins 0.000 description 1
- 108010055166 Chemokine CCL5 Proteins 0.000 description 1
- 206010008583 Chloroma Diseases 0.000 description 1
- 208000006990 Cholangiocarcinoma Diseases 0.000 description 1
- 210000002987 Choroid Plexus Anatomy 0.000 description 1
- 206010008909 Chronic hepatitis Diseases 0.000 description 1
- 230000037250 Clearance Effects 0.000 description 1
- 206010009839 Coeliac disease Diseases 0.000 description 1
- 206010057668 Cognitive disease Diseases 0.000 description 1
- 108009000280 Complement Activation Proteins 0.000 description 1
- 208000009798 Craniopharyngioma Diseases 0.000 description 1
- ATDGTVJJHBUTRL-UHFFFAOYSA-N Cyanogen bromide Chemical compound BrC#N ATDGTVJJHBUTRL-UHFFFAOYSA-N 0.000 description 1
- 108090000266 Cyclin-Dependent Kinases Proteins 0.000 description 1
- 102000003903 Cyclin-Dependent Kinases Human genes 0.000 description 1
- 229920000858 Cyclodextrin Polymers 0.000 description 1
- 241000701022 Cytomegalovirus Species 0.000 description 1
- 108010064945 D-4F peptide Proteins 0.000 description 1
- ZKKBZMXTFBAQLP-YNPPSTMPSA-N D-4F peptide Chemical compound C([C@H](C(=O)N[C@H](CC(O)=O)C(=O)N[C@H](CCCCN)C(=O)N[C@H](C(C)C)C(=O)N[C@H](C)C(=O)N[C@H](CCC(O)=O)C(=O)N[C@H](CCCCN)C(=O)N[C@H](CC=1C=CC=CC=1)C(=O)N[C@H](CCCCN)C(=O)N[C@H](CCC(O)=O)C(=O)N[C@H](C)C(=O)N[C@H](CC=1C=CC=CC=1)C(N)=O)NC(=O)[C@@H](CC=1C=CC=CC=1)NC(=O)[C@@H](C)NC(=O)[C@@H](CCCCN)NC(=O)[C@@H](CC=1C=CC=CC=1)NC(=O)[C@@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC(O)=O)NC(C)=O)C1=CC=C(O)C=C1 ZKKBZMXTFBAQLP-YNPPSTMPSA-N 0.000 description 1
- CKLJMWTZIZZHCS-UHFFFAOYSA-N DL-aspartic acid Chemical compound OC(=O)C(N)CC(O)=O CKLJMWTZIZZHCS-UHFFFAOYSA-N 0.000 description 1
- 238000001712 DNA sequencing Methods 0.000 description 1
- 102000004163 DNA-directed RNA polymerases Human genes 0.000 description 1
- 108090000626 DNA-directed RNA polymerases Proteins 0.000 description 1
- BFSMGDJOXZAERB-UHFFFAOYSA-N Dabrafenib Chemical compound S1C(C(C)(C)C)=NC(C=2C(=C(NS(=O)(=O)C=3C(=CC=CC=3F)F)C=CC=2)F)=C1C1=CC=NC(N)=N1 BFSMGDJOXZAERB-UHFFFAOYSA-N 0.000 description 1
- 206010012305 Demyelination Diseases 0.000 description 1
- 210000004443 Dendritic Cells Anatomy 0.000 description 1
- 208000007784 Diverticulitis Diseases 0.000 description 1
- 108020004461 Double-Stranded RNA Proteins 0.000 description 1
- 241000255601 Drosophila melanogaster Species 0.000 description 1
- 102000007698 EC 1.1.1.1 Human genes 0.000 description 1
- 108010021809 EC 1.1.1.1 Proteins 0.000 description 1
- 101700008821 EXO Proteins 0.000 description 1
- 101700083023 EXRN Proteins 0.000 description 1
- 206010014714 Endocrine neoplasms Diseases 0.000 description 1
- 206010014733 Endometrial cancer Diseases 0.000 description 1
- 102000004533 Endonucleases Human genes 0.000 description 1
- 108010042407 Endonucleases Proteins 0.000 description 1
- 206010067410 Endotoxaemia Diseases 0.000 description 1
- 206010014967 Ependymoma Diseases 0.000 description 1
- 210000000981 Epithelium Anatomy 0.000 description 1
- 241000283086 Equidae Species 0.000 description 1
- 241000283073 Equus caballus Species 0.000 description 1
- 208000006168 Ewing Sarcoma Diseases 0.000 description 1
- 102100015541 FCGR3A Human genes 0.000 description 1
- 101710044656 FCGR3A Proteins 0.000 description 1
- 101710044657 FCGR3B Proteins 0.000 description 1
- 108091006004 Fc-tagged proteins Proteins 0.000 description 1
- 241000282324 Felis Species 0.000 description 1
- 208000001640 Fibromyalgia Diseases 0.000 description 1
- 239000004606 Fillers/Extenders Substances 0.000 description 1
- 210000002683 Foot Anatomy 0.000 description 1
- 108060001039 GCN4 Proteins 0.000 description 1
- 101700014779 GLB1 Proteins 0.000 description 1
- 241000276438 Gadus morhua Species 0.000 description 1
- 206010017758 Gastric cancer Diseases 0.000 description 1
- 206010064571 Gene mutation Diseases 0.000 description 1
- 208000005017 Glioblastoma Diseases 0.000 description 1
- 206010018372 Glomerulonephritis membranous Diseases 0.000 description 1
- 206010061989 Glomerulosclerosis Diseases 0.000 description 1
- 206010018404 Glucagonoma Diseases 0.000 description 1
- 108010053070 Glutathione Disulfide Proteins 0.000 description 1
- 102000005720 Glutathione Transferase family Human genes 0.000 description 1
- 108010070675 Glutathione Transferase family Proteins 0.000 description 1
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 description 1
- 206010072579 Granulomatosis with polyangiitis Diseases 0.000 description 1
- 208000003807 Graves Disease Diseases 0.000 description 1
- 201000004779 Graves' disease Diseases 0.000 description 1
- 102000004144 Green Fluorescent Proteins Human genes 0.000 description 1
- 108010043121 Green Fluorescent Proteins Proteins 0.000 description 1
- 108010023302 HDL Cholesterol Proteins 0.000 description 1
- 108010010234 HDL Lipoproteins Proteins 0.000 description 1
- 239000007995 HEPES buffer Substances 0.000 description 1
- 101710015954 HVA1 Proteins 0.000 description 1
- 210000003128 Head Anatomy 0.000 description 1
- 208000001258 Hemangiosarcoma Diseases 0.000 description 1
- 229960002897 Heparin Drugs 0.000 description 1
- ZFGMDIBRIDKWMY-PASTXAENSA-N Heparin Chemical compound CC(O)=N[C@@H]1[C@@H](O)[C@H](O)[C@@H](COS(O)(=O)=O)O[C@@H]1O[C@@H]1[C@@H](C(O)=O)O[C@@H](O[C@H]2[C@@H]([C@@H](OS(O)(=O)=O)[C@@H](O[C@@H]3[C@@H](OC(O)[C@H](OS(O)(=O)=O)[C@H]3O)C(O)=O)O[C@@H]2O)CS(O)(=O)=O)[C@H](O)[C@H]1O ZFGMDIBRIDKWMY-PASTXAENSA-N 0.000 description 1
- 102000019267 Hepatic lipase Human genes 0.000 description 1
- 108050006747 Hepatic lipase Proteins 0.000 description 1
- 206010073071 Hepatocellular carcinoma Diseases 0.000 description 1
- 102000003839 Human Proteins Human genes 0.000 description 1
- 108090000144 Human Proteins Proteins 0.000 description 1
- 201000001971 Huntington's disease Diseases 0.000 description 1
- 208000006575 Hypertriglyceridemia Diseases 0.000 description 1
- 210000003026 Hypopharynx Anatomy 0.000 description 1
- 208000010159 IGA Glomerulonephritis Diseases 0.000 description 1
- 229940072221 IMMUNOGLOBULINS Drugs 0.000 description 1
- 101710044247 ITGA2B Proteins 0.000 description 1
- 102100019332 ITGA2B Human genes 0.000 description 1
- 102100019442 ITGAL Human genes 0.000 description 1
- 206010021263 IgA nephropathy Diseases 0.000 description 1
- KTUFNOKKBVMGRW-UHFFFAOYSA-N Imatinib Chemical compound C1CN(C)CCN1CC1=CC=C(C(=O)NC=2C=C(NC=3N=C(C=CN=3)C=3C=NC=CC=3)C(C)=CC=2)C=C1 KTUFNOKKBVMGRW-UHFFFAOYSA-N 0.000 description 1
- 206010061598 Immunodeficiency Diseases 0.000 description 1
- 102000012745 Immunoglobulin Subunits Human genes 0.000 description 1
- 108010079585 Immunoglobulin Subunits Proteins 0.000 description 1
- 206010061216 Infarction Diseases 0.000 description 1
- 208000005726 Inflammatory Breast Neoplasms Diseases 0.000 description 1
- 210000004969 Inflammatory Cells Anatomy 0.000 description 1
- 206010021972 Inflammatory bowel disease Diseases 0.000 description 1
- 206010021980 Inflammatory carcinoma of the breast Diseases 0.000 description 1
- 229940102223 Injectable Solution Drugs 0.000 description 1
- 102000003746 Insulin Receptor Human genes 0.000 description 1
- 108010001127 Insulin Receptor Proteins 0.000 description 1
- 206010022489 Insulin resistance Diseases 0.000 description 1
- 206010022498 Insulinoma Diseases 0.000 description 1
- 108090001005 Interleukin-6 Proteins 0.000 description 1
- 231100000601 Intoxication Toxicity 0.000 description 1
- 206010061252 Intraocular melanoma Diseases 0.000 description 1
- 108010089187 Ipilimumab Proteins 0.000 description 1
- QRXWMOHMRWLFEY-UHFFFAOYSA-N Isoniazid Chemical compound NNC(=O)C1=CC=NC=C1 QRXWMOHMRWLFEY-UHFFFAOYSA-N 0.000 description 1
- 210000000244 Kidney Pelvis Anatomy 0.000 description 1
- 241000235649 Kluyveromyces Species 0.000 description 1
- 244000285963 Kluyveromyces fragilis Species 0.000 description 1
- 235000014663 Kluyveromyces fragilis Nutrition 0.000 description 1
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 1
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 1
- ONIBWKKTOPOVIA-BYPYZUCNSA-N L-proline Chemical compound OC(=O)[C@@H]1CCCN1 ONIBWKKTOPOVIA-BYPYZUCNSA-N 0.000 description 1
- 125000000510 L-tryptophano group Chemical group [H]C1=C([H])C([H])=C2N([H])C([H])=C(C([H])([H])[C@@]([H])(C(O[H])=O)N([H])[*])C2=C1[H] 0.000 description 1
- 239000005517 L01XE01 - Imatinib Substances 0.000 description 1
- 238000008214 LDL Cholesterol Methods 0.000 description 1
- 101700065814 LEA2 Proteins 0.000 description 1
- 101700021338 LEC Proteins 0.000 description 1
- 101700077545 LECC Proteins 0.000 description 1
- 101700028499 LECG Proteins 0.000 description 1
- 101700063913 LECT Proteins 0.000 description 1
- 241000283953 Lagomorpha Species 0.000 description 1
- 210000000867 Larynx Anatomy 0.000 description 1
- 206010057168 Leishmania infection Diseases 0.000 description 1
- 208000004554 Leishmaniasis Diseases 0.000 description 1
- 208000002364 Leriche Syndrome Diseases 0.000 description 1
- 210000000265 Leukocytes Anatomy 0.000 description 1
- 208000006552 Lewis Lung Carcinoma Diseases 0.000 description 1
- 210000000088 Lip Anatomy 0.000 description 1
- 208000004883 Lipoid Nephrosis Diseases 0.000 description 1
- 208000002404 Liver Cell Adenoma Diseases 0.000 description 1
- 206010067125 Liver injury Diseases 0.000 description 1
- 108010015372 Low Density Lipoprotein Receptor-Related Protein-2 Proteins 0.000 description 1
- 208000004852 Lung Injury Diseases 0.000 description 1
- 206010058467 Lung neoplasm malignant Diseases 0.000 description 1
- 206010025135 Lupus erythematosus Diseases 0.000 description 1
- 239000006137 Luria-Bertani broth Substances 0.000 description 1
- 210000002751 Lymph Anatomy 0.000 description 1
- 108010064548 Lymphocyte Function-Associated Antigen-1 Proteins 0.000 description 1
- 210000004698 Lymphocytes Anatomy 0.000 description 1
- 101700067851 MMP9 Proteins 0.000 description 1
- 239000007993 MOPS buffer Substances 0.000 description 1
- 241000282553 Macaca Species 0.000 description 1
- 241000282560 Macaca mulatta Species 0.000 description 1
- 206010061269 Malignant peritoneal neoplasm Diseases 0.000 description 1
- 208000000172 Medulloblastoma Diseases 0.000 description 1
- 210000003975 Mesenteric Arteries Anatomy 0.000 description 1
- 230000036740 Metabolism Effects 0.000 description 1
- 108090000157 Metallothionein Proteins 0.000 description 1
- 102000003792 Metallothionein Human genes 0.000 description 1
- 206010027476 Metastasis Diseases 0.000 description 1
- VKHAHZOOUSRJNA-GCNJZUOMSA-N Mifepristone Chemical compound C1([C@@H]2C3=C4CCC(=O)C=C4CC[C@H]3[C@@H]3CC[C@@]([C@]3(C2)C)(O)C#CC)=CC=C(N(C)C)C=C1 VKHAHZOOUSRJNA-GCNJZUOMSA-N 0.000 description 1
- 210000004080 Milk Anatomy 0.000 description 1
- 210000000214 Mouth Anatomy 0.000 description 1
- 210000002027 Muscle, Skeletal Anatomy 0.000 description 1
- 210000002464 Muscle, Smooth, Vascular Anatomy 0.000 description 1
- 206010028417 Myasthenia gravis Diseases 0.000 description 1
- 108010079364 N-glycylalanine Proteins 0.000 description 1
- NQTADLQHYWFPDB-UHFFFAOYSA-N N-hydroxy-Succinimide Chemical compound ON1C(=O)CCC1=O NQTADLQHYWFPDB-UHFFFAOYSA-N 0.000 description 1
- 102000003945 NF-kappa B Human genes 0.000 description 1
- 108010057466 NF-kappa B Proteins 0.000 description 1
- 238000005481 NMR spectroscopy Methods 0.000 description 1
- 101700080605 NUC1 Proteins 0.000 description 1
- 229920002274 Nalgene Polymers 0.000 description 1
- 210000003928 Nasal Cavity Anatomy 0.000 description 1
- 229940097496 Nasal Spray Drugs 0.000 description 1
- 210000001989 Nasopharynx Anatomy 0.000 description 1
- 102000007339 Nerve Growth Factor Receptors Human genes 0.000 description 1
- 108010032605 Nerve Growth Factor Receptors Proteins 0.000 description 1
- 206010029240 Neuritis Diseases 0.000 description 1
- 241000221960 Neurospora Species 0.000 description 1
- 206010029350 Neurotoxicity Diseases 0.000 description 1
- 231100000618 Neurotoxin Toxicity 0.000 description 1
- 108010061543 Neutralizing Antibodies Proteins 0.000 description 1
- 210000000440 Neutrophils Anatomy 0.000 description 1
- 239000000020 Nitrocellulose Substances 0.000 description 1
- 108010019706 Nivolumab Proteins 0.000 description 1
- 206010054107 Nodule Diseases 0.000 description 1
- 229920002332 Noncoding DNA Polymers 0.000 description 1
- 206010030155 Oesophageal carcinoma Diseases 0.000 description 1
- 229920000272 Oligonucleotide Polymers 0.000 description 1
- 101710034340 Os04g0173800 Proteins 0.000 description 1
- 208000000035 Osteochondroma Diseases 0.000 description 1
- 241000283898 Ovis Species 0.000 description 1
- HVAUUPRFYPCOCA-AREMUKBSSA-O PAF Chemical compound CCCCCCCCCCCCCCCCOC[C@@H](OC(C)=O)COP(O)(=O)OCC[N+](C)(C)C HVAUUPRFYPCOCA-AREMUKBSSA-O 0.000 description 1
- 101000363300 PLA2G7 Proteins 0.000 description 1
- RXWNCPJZOCPEPQ-NVWDDTSBSA-N PUROMYCIN Chemical compound C1=CC(OC)=CC=C1C[C@H](N)C(=O)N[C@H]1[C@@H](O)[C@H](N2C3=NC=NC(=C3N=C2)N(C)C)O[C@@H]1CO RXWNCPJZOCPEPQ-NVWDDTSBSA-N 0.000 description 1
- 229950010131 PUROMYCIN Drugs 0.000 description 1
- 210000003695 Paranasal Sinuses Anatomy 0.000 description 1
- WYMSBXTXOHUIGT-UHFFFAOYSA-N Paraoxon Chemical compound CCOP(=O)(OCC)OC1=CC=C([N+]([O-])=O)C=C1 WYMSBXTXOHUIGT-UHFFFAOYSA-N 0.000 description 1
- 206010034299 Penile cancer Diseases 0.000 description 1
- 208000005764 Peripheral Arterial Disease Diseases 0.000 description 1
- 206010034695 Pernicious anaemia Diseases 0.000 description 1
- 206010034800 Phaeochromocytoma Diseases 0.000 description 1
- 206010057249 Phagocytosis Diseases 0.000 description 1
- BELBBZDIHDAJOR-UHFFFAOYSA-N Phenolsulfonephthalein Chemical compound C1=CC(O)=CC=C1C1(C=2C=CC(O)=CC=2)C2=CC=CC=C2S(=O)(=O)O1 BELBBZDIHDAJOR-UHFFFAOYSA-N 0.000 description 1
- 229960003531 Phenolsulfonphthalein Drugs 0.000 description 1
- 102000004203 Phosphoric Triester Hydrolases Human genes 0.000 description 1
- 108090000754 Phosphoric Triester Hydrolases Proteins 0.000 description 1
- 108091000081 Phosphotransferases Proteins 0.000 description 1
- 210000004560 Pineal Gland Anatomy 0.000 description 1
- 208000007913 Pituitary Neoplasms Diseases 0.000 description 1
- 210000002826 Placenta Anatomy 0.000 description 1
- 206010035226 Plasma cell myeloma Diseases 0.000 description 1
- 208000005374 Poisoning Diseases 0.000 description 1
- 241000276498 Pollachius virens Species 0.000 description 1
- 229920002732 Polyanhydride Polymers 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 229920002873 Polyethylenimine Polymers 0.000 description 1
- 208000005987 Polymyositis Diseases 0.000 description 1
- 229920001710 Polyorthoester Polymers 0.000 description 1
- 239000004793 Polystyrene Substances 0.000 description 1
- TUZYXOIXSAXUGO-PZAWKZKUSA-N Pravastatin Chemical compound C1=C[C@H](C)[C@H](CC[C@@H](O)C[C@@H](O)CC(O)=O)[C@H]2[C@@H](OC(=O)[C@@H](C)CC)C[C@H](O)C=C21 TUZYXOIXSAXUGO-PZAWKZKUSA-N 0.000 description 1
- 229960002965 Pravastatin Drugs 0.000 description 1
- 102000012412 Presenilin-1 Human genes 0.000 description 1
- 108010036933 Presenilin-1 Proteins 0.000 description 1
- 206010060862 Prostate cancer Diseases 0.000 description 1
- 102000004245 Proteasome Endopeptidase Complex Human genes 0.000 description 1
- 108090000708 Proteasome Endopeptidase Complex Proteins 0.000 description 1
- 208000008425 Protein Deficiency Diseases 0.000 description 1
- 102000001253 Protein Kinases Human genes 0.000 description 1
- 102000004022 Protein-Tyrosine Kinases Human genes 0.000 description 1
- 108090000412 Protein-Tyrosine Kinases Proteins 0.000 description 1
- 208000005069 Pulmonary Fibrosis Diseases 0.000 description 1
- 229940076788 Pyruvate Drugs 0.000 description 1
- 102100013504 RPL17 Human genes 0.000 description 1
- 108020004511 Recombinant DNA Proteins 0.000 description 1
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 1
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 1
- 206010038389 Renal cancer Diseases 0.000 description 1
- 229920001914 Ribonucleotide Polymers 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- 101700061430 SAT19 Proteins 0.000 description 1
- 108060007796 SPATA2 Proteins 0.000 description 1
- 210000003079 Salivary Glands Anatomy 0.000 description 1
- 241000235347 Schizosaccharomyces pombe Species 0.000 description 1
- BTIHMVBBUGXLCJ-OAHLLOKOSA-N Seliciclib Chemical compound C=12N=CN(C(C)C)C2=NC(N[C@@H](CO)CC)=NC=1NCC1=CC=CC=C1 BTIHMVBBUGXLCJ-OAHLLOKOSA-N 0.000 description 1
- 229950000055 Seliciclib Drugs 0.000 description 1
- 108010071390 Serum Albumin Proteins 0.000 description 1
- 230000037165 Serum Concentration Effects 0.000 description 1
- 102000005632 Single-Chain Antibodies Human genes 0.000 description 1
- 108010070144 Single-Chain Antibodies Proteins 0.000 description 1
- 206010041823 Squamous cell carcinoma Diseases 0.000 description 1
- 241000282887 Suidae Species 0.000 description 1
- 102100008904 TFRC Human genes 0.000 description 1
- 102100012096 TLR2 Human genes 0.000 description 1
- 101700064022 TLR2 Proteins 0.000 description 1
- 101710040537 TNF Proteins 0.000 description 1
- 102000003714 TNF receptor-associated factor 6 Human genes 0.000 description 1
- 108090000009 TNF receptor-associated factor 6 Proteins 0.000 description 1
- 101700081234 TTR Proteins 0.000 description 1
- 108020003835 TX1 Proteins 0.000 description 1
- 206010057644 Testis cancer Diseases 0.000 description 1
- 229940104230 Thymidine Drugs 0.000 description 1
- 206010043709 Thyroid disease Diseases 0.000 description 1
- 206010044221 Toxic encephalopathy Diseases 0.000 description 1
- 108010033576 Transferrin Receptors Proteins 0.000 description 1
- 206010044412 Transitional cell carcinoma Diseases 0.000 description 1
- 206010069363 Traumatic lung injury Diseases 0.000 description 1
- QORWJWZARLRLPR-UHFFFAOYSA-H Tricalcium phosphate Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 1
- 241000255993 Trichoplusia ni Species 0.000 description 1
- DSDAICPXUXPBCC-MWDJDSKUSA-N Trimethyl-β-cyclodextrin Chemical compound COC[C@H]([C@H]([C@@H]([C@H]1OC)OC)O[C@H]2O[C@@H]([C@@H](O[C@H]3O[C@H](COC)[C@H]([C@@H]([C@H]3OC)OC)O[C@H]3O[C@H](COC)[C@H]([C@@H]([C@H]3OC)OC)O[C@H]3O[C@H](COC)[C@H]([C@@H]([C@H]3OC)OC)O[C@H]3O[C@H](COC)[C@H]([C@@H]([C@H]3OC)OC)O3)[C@H](OC)[C@H]2OC)COC)O[C@@H]1O[C@H]1[C@H](OC)[C@@H](OC)[C@@H]3O[C@@H]1COC DSDAICPXUXPBCC-MWDJDSKUSA-N 0.000 description 1
- 241000223105 Trypanosoma brucei Species 0.000 description 1
- 206010045261 Type IIa hyperlipidaemia Diseases 0.000 description 1
- 210000000626 Ureter Anatomy 0.000 description 1
- 206010046431 Urethral cancer Diseases 0.000 description 1
- 210000001635 Urinary Tract Anatomy 0.000 description 1
- 241000221566 Ustilago Species 0.000 description 1
- 206010046766 Uterine cancer Diseases 0.000 description 1
- 206010046851 Uveitis Diseases 0.000 description 1
- 102100019577 VCAM1 Human genes 0.000 description 1
- 229940029983 VITAMINS Drugs 0.000 description 1
- 108010062497 VLDL Lipoproteins Proteins 0.000 description 1
- 206010046885 Vaginal cancer Diseases 0.000 description 1
- 108010000134 Vascular Cell Adhesion Molecule-1 Proteins 0.000 description 1
- 208000010019 Vascular System Injury Diseases 0.000 description 1
- 206010027701 Vascular injury Diseases 0.000 description 1
- 108020005202 Viral DNA Proteins 0.000 description 1
- 108020000999 Viral RNA Proteins 0.000 description 1
- 206010047461 Viral infection Diseases 0.000 description 1
- 208000001756 Virus Disease Diseases 0.000 description 1
- 229940021016 Vitamin IV solution additives Drugs 0.000 description 1
- 206010047642 Vitiligo Diseases 0.000 description 1
- 206010047741 Vulval cancer Diseases 0.000 description 1
- OKRCXSCDWKJWSW-UHFFFAOYSA-N [4-(chloromethyl)phenyl] acetate Chemical compound CC(=O)OC1=CC=C(CCl)C=C1 OKRCXSCDWKJWSW-UHFFFAOYSA-N 0.000 description 1
- 238000002835 absorbance Methods 0.000 description 1
- 230000002378 acidificating Effects 0.000 description 1
- 230000003213 activating Effects 0.000 description 1
- 230000000996 additive Effects 0.000 description 1
- 201000005179 adrenal carcinoma Diseases 0.000 description 1
- 201000005188 adrenal gland cancer Diseases 0.000 description 1
- 239000000556 agonist Substances 0.000 description 1
- 229940050528 albumin Drugs 0.000 description 1
- 150000001412 amines Chemical class 0.000 description 1
- 125000003277 amino group Chemical group 0.000 description 1
- 229910052921 ammonium sulfate Inorganic materials 0.000 description 1
- 235000011130 ammonium sulphate Nutrition 0.000 description 1
- 229960000723 ampicillin Drugs 0.000 description 1
- 201000000315 ampulla of Vater cancer Diseases 0.000 description 1
- 230000033115 angiogenesis Effects 0.000 description 1
- 201000003076 angiosarcoma Diseases 0.000 description 1
- 238000000137 annealing Methods 0.000 description 1
- 108010093001 anti-IgG Proteins 0.000 description 1
- 230000000489 anti-atherogenic Effects 0.000 description 1
- 230000000845 anti-microbial Effects 0.000 description 1
- 230000000890 antigenic Effects 0.000 description 1
- 201000011165 anus cancer Diseases 0.000 description 1
- 108010063201 apolipoprotein A-I mimetic peptide 4F Proteins 0.000 description 1
- 239000006286 aqueous extract Substances 0.000 description 1
- 108010013835 arginine glutamate Proteins 0.000 description 1
- 229960004246 arginine glutamate Drugs 0.000 description 1
- 201000002936 artery disease Diseases 0.000 description 1
- 125000003118 aryl group Chemical group 0.000 description 1
- 235000009582 asparagine Nutrition 0.000 description 1
- 201000000448 autoimmune hemolytic anemia Diseases 0.000 description 1
- 229910052790 beryllium Inorganic materials 0.000 description 1
- ATBAMAFKBVZNFJ-UHFFFAOYSA-N beryllium(0) Chemical compound [Be] ATBAMAFKBVZNFJ-UHFFFAOYSA-N 0.000 description 1
- 229960000397 bevacizumab Drugs 0.000 description 1
- 102000024070 binding proteins Human genes 0.000 description 1
- 108091007650 binding proteins Proteins 0.000 description 1
- 230000003115 biocidal Effects 0.000 description 1
- 239000000560 biocompatible material Substances 0.000 description 1
- 239000000090 biomarker Substances 0.000 description 1
- 230000037348 biosynthesis Effects 0.000 description 1
- 125000006367 bivalent amino carbonyl group Chemical group [H]N([*:1])C([*:2])=O 0.000 description 1
- OYVAGSVQBOHSSS-UAPAGMARSA-O bleomycin A2 Chemical compound N([C@H](C(=O)N[C@H](C)[C@@H](O)[C@H](C)C(=O)N[C@@H]([C@H](O)C)C(=O)NCCC=1SC=C(N=1)C=1SC=C(N=1)C(=O)NCCC[S+](C)C)[C@@H](O[C@H]1[C@H]([C@@H](O)[C@H](O)[C@H](CO)O1)O[C@@H]1[C@H]([C@@H](OC(N)=O)[C@H](O)[C@@H](CO)O1)O)C=1N=CNC=1)C(=O)C1=NC([C@H](CC(N)=O)NC[C@H](N)C(N)=O)=NC(N)=C1C OYVAGSVQBOHSSS-UAPAGMARSA-O 0.000 description 1
- 239000001045 blue dye Substances 0.000 description 1
- 230000037396 body weight Effects 0.000 description 1
- 238000006664 bond formation reaction Methods 0.000 description 1
- ZOXJGFHDIHLPTG-UHFFFAOYSA-N boron Chemical compound [B] ZOXJGFHDIHLPTG-UHFFFAOYSA-N 0.000 description 1
- 229910052796 boron Inorganic materials 0.000 description 1
- 229960001467 bortezomib Drugs 0.000 description 1
- 239000007975 buffered saline Substances 0.000 description 1
- 239000006172 buffering agent Substances 0.000 description 1
- UXVMQQNJUSDDNG-UHFFFAOYSA-L cacl2 Chemical compound [Cl-].[Cl-].[Ca+2] UXVMQQNJUSDDNG-UHFFFAOYSA-L 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 229910000389 calcium phosphate Inorganic materials 0.000 description 1
- 235000011010 calcium phosphates Nutrition 0.000 description 1
- 238000007816 calorimetric assay Methods 0.000 description 1
- 238000002619 cancer immunotherapy Methods 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 150000001718 carbodiimides Chemical class 0.000 description 1
- 235000014633 carbohydrates Nutrition 0.000 description 1
- OKTJSMMVPCPJKN-UHFFFAOYSA-N carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 1
- 229910052799 carbon Inorganic materials 0.000 description 1
- 235000010948 carboxy methyl cellulose Nutrition 0.000 description 1
- 201000009030 carcinoma Diseases 0.000 description 1
- 230000020411 cell activation Effects 0.000 description 1
- 238000010370 cell cloning Methods 0.000 description 1
- 230000010261 cell growth Effects 0.000 description 1
- 230000022534 cell killing Effects 0.000 description 1
- 230000003833 cell viability Effects 0.000 description 1
- 229920002678 cellulose Polymers 0.000 description 1
- 239000001913 cellulose Substances 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 201000010881 cervical cancer Diseases 0.000 description 1
- 229960005395 cetuximab Drugs 0.000 description 1
- 239000002738 chelating agent Substances 0.000 description 1
- 239000007795 chemical reaction product Substances 0.000 description 1
- 238000005660 chlorination reaction Methods 0.000 description 1
- 150000001841 cholesterols Chemical class 0.000 description 1
- 201000009047 chordoma Diseases 0.000 description 1
- 230000002759 chromosomal Effects 0.000 description 1
- 230000035512 clearance Effects 0.000 description 1
- 235000019516 cod Nutrition 0.000 description 1
- 201000011231 colorectal cancer Diseases 0.000 description 1
- 230000002860 competitive Effects 0.000 description 1
- 230000024203 complement activation Effects 0.000 description 1
- 230000004540 complement-dependent cytotoxicity Effects 0.000 description 1
- 238000003271 compound fluorescence assay Methods 0.000 description 1
- 230000001143 conditioned Effects 0.000 description 1
- 238000011109 contamination Methods 0.000 description 1
- 239000006071 cream Substances 0.000 description 1
- 238000004132 cross linking Methods 0.000 description 1
- 238000010192 crystallographic characterization Methods 0.000 description 1
- 201000010305 cutaneous fibrous histiocytoma Diseases 0.000 description 1
- 239000002875 cyclin dependent kinase inhibitor Substances 0.000 description 1
- 230000009089 cytolysis Effects 0.000 description 1
- 230000001472 cytotoxic Effects 0.000 description 1
- 231100000433 cytotoxic Toxicity 0.000 description 1
- 230000003013 cytotoxicity Effects 0.000 description 1
- 231100000135 cytotoxicity Toxicity 0.000 description 1
- 229960002465 dabrafenib Drugs 0.000 description 1
- 230000034994 death Effects 0.000 description 1
- 231100000517 death Toxicity 0.000 description 1
- 230000003210 demyelinating Effects 0.000 description 1
- 238000004925 denaturation Methods 0.000 description 1
- 230000036425 denaturation Effects 0.000 description 1
- 239000005547 deoxyribonucleotide Substances 0.000 description 1
- 125000002637 deoxyribonucleotide group Chemical group 0.000 description 1
- 201000004624 dermatitis Diseases 0.000 description 1
- 231100000406 dermatitis Toxicity 0.000 description 1
- 238000011033 desalting Methods 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 230000001627 detrimental Effects 0.000 description 1
- 238000010586 diagram Methods 0.000 description 1
- 238000002050 diffraction method Methods 0.000 description 1
- 102000004419 dihydrofolate reductase family Human genes 0.000 description 1
- 108020001096 dihydrofolate reductase family Proteins 0.000 description 1
- 238000007865 diluting Methods 0.000 description 1
- 230000003292 diminished Effects 0.000 description 1
- OVTCUIZCVUGJHS-UHFFFAOYSA-N dipyrrin Chemical compound C=1C=CNC=1C=C1C=CC=N1 OVTCUIZCVUGJHS-UHFFFAOYSA-N 0.000 description 1
- 201000008325 diseases of cellular proliferation Diseases 0.000 description 1
- 230000003291 dopaminomimetic Effects 0.000 description 1
- 231100000673 dose–response relationship Toxicity 0.000 description 1
- 238000009509 drug development Methods 0.000 description 1
- 238000007876 drug discovery Methods 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 239000003623 enhancer Substances 0.000 description 1
- 230000002255 enzymatic Effects 0.000 description 1
- 238000001952 enzyme assay Methods 0.000 description 1
- 238000001976 enzyme digestion Methods 0.000 description 1
- 102000017256 epidermal growth factor-activated receptor activity proteins Human genes 0.000 description 1
- 108040009258 epidermal growth factor-activated receptor activity proteins Proteins 0.000 description 1
- 210000002919 epithelial cells Anatomy 0.000 description 1
- 150000002118 epoxides Chemical class 0.000 description 1
- 201000004101 esophageal cancer Diseases 0.000 description 1
- 235000020774 essential nutrients Nutrition 0.000 description 1
- 238000005886 esterification reaction Methods 0.000 description 1
- BFMKFCLXZSUVPI-UHFFFAOYSA-N ethyl but-3-enoate Chemical compound CCOC(=O)CC=C BFMKFCLXZSUVPI-UHFFFAOYSA-N 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 201000001342 fallopian tube cancer Diseases 0.000 description 1
- 235000013861 fat-free Nutrition 0.000 description 1
- 230000035611 feeding Effects 0.000 description 1
- GNBHRKFJIUUOQI-UHFFFAOYSA-N fluorescein Chemical compound O1C(=O)C2=CC=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 GNBHRKFJIUUOQI-UHFFFAOYSA-N 0.000 description 1
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 1
- 239000007850 fluorescent dye Substances 0.000 description 1
- 201000005206 focal segmental glomerulosclerosis Diseases 0.000 description 1
- 238000006062 fragmentation reaction Methods 0.000 description 1
- 238000007710 freezing Methods 0.000 description 1
- 201000010175 gallbladder cancer Diseases 0.000 description 1
- 201000010231 gastrointestinal system cancer Diseases 0.000 description 1
- 238000002523 gelfiltration Methods 0.000 description 1
- 238000001476 gene delivery Methods 0.000 description 1
- 238000010353 genetic engineering Methods 0.000 description 1
- 210000004602 germ cell Anatomy 0.000 description 1
- 201000007116 gestational trophoblastic neoplasm Diseases 0.000 description 1
- 239000011521 glass Substances 0.000 description 1
- 230000002518 glial Effects 0.000 description 1
- 230000004190 glucose uptake Effects 0.000 description 1
- 239000004220 glutamic acid Substances 0.000 description 1
- YPZRWBKMTBYPTK-BJDJZHNGSA-N glutathione disulfide Chemical compound OC(=O)[C@@H](N)CCC(=O)N[C@H](C(=O)NCC(O)=O)CSSC[C@@H](C(=O)NCC(O)=O)NC(=O)CC[C@H](N)C(O)=O YPZRWBKMTBYPTK-BJDJZHNGSA-N 0.000 description 1
- VPZXBVLAVMBEQI-VKHMYHEASA-N gly ala Chemical compound OC(=O)[C@H](C)NC(=O)CN VPZXBVLAVMBEQI-VKHMYHEASA-N 0.000 description 1
- 230000036252 glycation Effects 0.000 description 1
- 150000002332 glycine derivatives Chemical class 0.000 description 1
- LYCAIKOWRPUZTN-UHFFFAOYSA-N glycol Chemical compound OCCO LYCAIKOWRPUZTN-UHFFFAOYSA-N 0.000 description 1
- 230000002414 glycolytic Effects 0.000 description 1
- 102000035362 glycosylated proteins Human genes 0.000 description 1
- 108091005600 glycosylated proteins Proteins 0.000 description 1
- YMAWOPBAYDPSLA-UHFFFAOYSA-N glycylglycine zwitterion Chemical compound [NH3+]CC(=O)NCC([O-])=O YMAWOPBAYDPSLA-UHFFFAOYSA-N 0.000 description 1
- PCHJSUWPFVWCPO-UHFFFAOYSA-N gold Chemical compound [Au] PCHJSUWPFVWCPO-UHFFFAOYSA-N 0.000 description 1
- 239000010931 gold Substances 0.000 description 1
- 229910052737 gold Inorganic materials 0.000 description 1
- 201000011626 grade III astrocytoma Diseases 0.000 description 1
- 239000005090 green fluorescent protein Substances 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- 238000003306 harvesting Methods 0.000 description 1
- 238000010438 heat treatment Methods 0.000 description 1
- 229920000669 heparin Polymers 0.000 description 1
- 231100000753 hepatic injury Toxicity 0.000 description 1
- 231100000283 hepatitis Toxicity 0.000 description 1
- 231100000844 hepatocellular carcinoma Toxicity 0.000 description 1
- 231100000304 hepatotoxicity Toxicity 0.000 description 1
- 239000000833 heterodimer Substances 0.000 description 1
- 230000000971 hippocampal Effects 0.000 description 1
- 238000002744 homologous recombination Methods 0.000 description 1
- 102000023239 human CETP protein Human genes 0.000 description 1
- 238000009396 hybridization Methods 0.000 description 1
- 239000001257 hydrogen Substances 0.000 description 1
- 229910052739 hydrogen Inorganic materials 0.000 description 1
- 230000003301 hydrolyzing Effects 0.000 description 1
- 125000001165 hydrophobic group Chemical group 0.000 description 1
- 125000002887 hydroxy group Chemical group [H]O* 0.000 description 1
- 229910052588 hydroxylapatite Inorganic materials 0.000 description 1
- 229960002411 imatinib Drugs 0.000 description 1
- 230000028993 immune response Effects 0.000 description 1
- 230000003053 immunization Effects 0.000 description 1
- 238000002649 immunization Methods 0.000 description 1
- 230000002998 immunogenetic Effects 0.000 description 1
- 230000004957 immunoregulator effect Effects 0.000 description 1
- 238000009169 immunotherapy Methods 0.000 description 1
- 230000001771 impaired Effects 0.000 description 1
- 238000000099 in vitro assay Methods 0.000 description 1
- 230000001939 inductive effect Effects 0.000 description 1
- 238000001764 infiltration Methods 0.000 description 1
- 230000000977 initiatory Effects 0.000 description 1
- 238000011081 inoculation Methods 0.000 description 1
- 239000002054 inoculum Substances 0.000 description 1
- 229910052500 inorganic mineral Inorganic materials 0.000 description 1
- 230000000749 insecticidal Effects 0.000 description 1
- 239000002917 insecticide Substances 0.000 description 1
- 230000003914 insulin secretion Effects 0.000 description 1
- 102000006495 integrins Human genes 0.000 description 1
- 108010044426 integrins Proteins 0.000 description 1
- 230000035987 intoxication Effects 0.000 description 1
- 231100000566 intoxication Toxicity 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000002608 intravascular ultrasound Methods 0.000 description 1
- 108010003082 intrinsic factor-cobalamin receptor Proteins 0.000 description 1
- 238000004255 ion exchange chromatography Methods 0.000 description 1
- 229960005386 ipilimumab Drugs 0.000 description 1
- 230000001788 irregular Effects 0.000 description 1
- KFZMGEQAYNKOFK-UHFFFAOYSA-N iso-propanol Chemical compound CC(C)O KFZMGEQAYNKOFK-UHFFFAOYSA-N 0.000 description 1
- 201000010982 kidney cancer Diseases 0.000 description 1
- 201000010901 lateral sclerosis Diseases 0.000 description 1
- 101700036391 lecA Proteins 0.000 description 1
- 239000002523 lectin Substances 0.000 description 1
- 102000005861 leptin receptors Human genes 0.000 description 1
- 108010019813 leptin receptors Proteins 0.000 description 1
- 230000003902 lesions Effects 0.000 description 1
- 231100000225 lethality Toxicity 0.000 description 1
- 230000000670 limiting Effects 0.000 description 1
- 230000037356 lipid metabolism Effects 0.000 description 1
- 239000002502 liposome Substances 0.000 description 1
- 239000008263 liquid aerosol Substances 0.000 description 1
- 238000011068 load Methods 0.000 description 1
- 230000004807 localization Effects 0.000 description 1
- 231100000053 low toxicity Toxicity 0.000 description 1
- 201000005202 lung cancer Diseases 0.000 description 1
- 231100000516 lung damage Toxicity 0.000 description 1
- 231100000515 lung injury Toxicity 0.000 description 1
- 201000001142 lung small cell carcinoma Diseases 0.000 description 1
- 108091022076 maltose binding proteins Proteins 0.000 description 1
- 101700001016 mbhA Proteins 0.000 description 1
- 239000012577 media supplement Substances 0.000 description 1
- 102000006240 membrane receptors Human genes 0.000 description 1
- 108020004084 membrane receptors Proteins 0.000 description 1
- 201000008350 membranous glomerulonephritis Diseases 0.000 description 1
- 231100000855 membranous nephropathy Toxicity 0.000 description 1
- 201000008806 mesenchymal cell neoplasm Diseases 0.000 description 1
- 238000010197 meta-analysis Methods 0.000 description 1
- 230000037353 metabolic pathway Effects 0.000 description 1
- 230000004060 metabolic process Effects 0.000 description 1
- 230000035786 metabolism Effects 0.000 description 1
- LUBKKVGXMXTXOZ-UHFFFAOYSA-N methyl 5,7-dichloro-4-hydroxy-5'-methoxy-6-methyl-3,3'-dioxospiro[1-benzofuran-2,6'-cyclohexa-1,4-diene]-1'-carboxylate Chemical compound COC(=O)C1=CC(=O)C=C(OC)C11C(=O)C(C(O)=C(Cl)C(C)=C2Cl)=C2O1 LUBKKVGXMXTXOZ-UHFFFAOYSA-N 0.000 description 1
- 230000002025 microglial Effects 0.000 description 1
- 229960003248 mifepristone Drugs 0.000 description 1
- 235000013336 milk Nutrition 0.000 description 1
- 239000008267 milk Substances 0.000 description 1
- 239000011707 mineral Substances 0.000 description 1
- 235000010755 mineral Nutrition 0.000 description 1
- 238000001565 modulated differential scanning calorimetry Methods 0.000 description 1
- 238000010369 molecular cloning Methods 0.000 description 1
- 238000009126 molecular therapy Methods 0.000 description 1
- 230000036457 multidrug resistance Effects 0.000 description 1
- 201000009251 multiple myeloma Diseases 0.000 description 1
- 231100000483 muscle toxicity Toxicity 0.000 description 1
- 201000005987 myeloid sarcoma Diseases 0.000 description 1
- 230000002071 myeloproliferative Effects 0.000 description 1
- 239000002077 nanosphere Substances 0.000 description 1
- 239000007922 nasal spray Substances 0.000 description 1
- 239000006199 nebulizer Substances 0.000 description 1
- 230000017074 necrotic cell death Effects 0.000 description 1
- 229960004927 neomycin Drugs 0.000 description 1
- 108010068617 neonatal Fc receptor Proteins 0.000 description 1
- 201000008383 nephritis Diseases 0.000 description 1
- 201000011682 nervous system cancer Diseases 0.000 description 1
- 230000000626 neurodegenerative Effects 0.000 description 1
- 230000016273 neuron death Effects 0.000 description 1
- 210000002569 neurons Anatomy 0.000 description 1
- 230000002887 neurotoxic Effects 0.000 description 1
- 231100000228 neurotoxicity Toxicity 0.000 description 1
- 239000002581 neurotoxin Substances 0.000 description 1
- 230000001264 neutralization Effects 0.000 description 1
- 229920001220 nitrocellulos Polymers 0.000 description 1
- 229910052757 nitrogen Inorganic materials 0.000 description 1
- 229960003301 nivolumab Drugs 0.000 description 1
- 101700006494 nucA Proteins 0.000 description 1
- 101700050775 oct-1 Proteins 0.000 description 1
- 125000002347 octyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 201000002575 ocular melanoma Diseases 0.000 description 1
- 201000010133 oligodendroglioma Diseases 0.000 description 1
- 230000014207 opsonization Effects 0.000 description 1
- 230000003571 opsonizing Effects 0.000 description 1
- 201000008482 osteoarthritis Diseases 0.000 description 1
- 201000008968 osteosarcoma Diseases 0.000 description 1
- 201000001539 ovarian carcinoma Diseases 0.000 description 1
- 239000001301 oxygen Substances 0.000 description 1
- 229910052760 oxygen Inorganic materials 0.000 description 1
- MYMOFIZGZYHOMD-UHFFFAOYSA-N oxygen Chemical compound O=O MYMOFIZGZYHOMD-UHFFFAOYSA-N 0.000 description 1
- 201000010287 pancreatic acinar cell adenocarcinoma Diseases 0.000 description 1
- 201000002094 pancreatic adenocarcinoma Diseases 0.000 description 1
- 229960004623 paraoxon Drugs 0.000 description 1
- 201000003913 parathyroid carcinoma Diseases 0.000 description 1
- 230000001575 pathological Effects 0.000 description 1
- 108010026276 pembrolizumab Proteins 0.000 description 1
- 229960002621 pembrolizumab Drugs 0.000 description 1
- 201000011152 pemphigus Diseases 0.000 description 1
- 201000001976 pemphigus vulgaris Diseases 0.000 description 1
- 201000002911 peripheral artery disease Diseases 0.000 description 1
- 201000002524 peritoneal carcinoma Diseases 0.000 description 1
- 201000008944 perivascular tumor Diseases 0.000 description 1
- 239000000575 pesticide Substances 0.000 description 1
- 238000002823 phage display Methods 0.000 description 1
- 230000008782 phagocytosis Effects 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- 238000009522 phase III clinical trial Methods 0.000 description 1
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 description 1
- WTJKGGKOPKCXLL-RRHRGVEJSA-N phosphatidylcholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCCC=CCCCCCCCC WTJKGGKOPKCXLL-RRHRGVEJSA-N 0.000 description 1
- 239000003757 phosphotransferase inhibitor Substances 0.000 description 1
- 230000004962 physiological condition Effects 0.000 description 1
- 230000035790 physiological processes and functions Effects 0.000 description 1
- 239000000902 placebo Substances 0.000 description 1
- 229940068196 placebo Drugs 0.000 description 1
- 229920003023 plastic Polymers 0.000 description 1
- 239000004033 plastic Substances 0.000 description 1
- 229920002496 poly(ether sulfone) Polymers 0.000 description 1
- 229920003208 poly(ethylene sulfide) Polymers 0.000 description 1
- 230000001402 polyadenylating Effects 0.000 description 1
- 229920001601 polyetherimide Polymers 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 229920001282 polysaccharide Polymers 0.000 description 1
- 239000005017 polysaccharide Substances 0.000 description 1
- 150000004804 polysaccharides Polymers 0.000 description 1
- 229920002223 polystyrene Polymers 0.000 description 1
- OFPTZWGZSRJCOT-MSPNRCMCSA-M potassium;2-[(1S,2S,3R,4S,5S,6R)-3-(diaminomethylideneamino)-4-[(2R,3R,4R,5S)-3-[(2S,3S,4S,5R,6S)-4,5-dihydroxy-6-(hydroxymethyl)-3-(methylamino)oxan-2-yl]oxy-4-formyl-4-hydroxy-5-methyloxolan-2-yl]oxy-2,5,6-trihydroxycyclohexyl]guanidine;(2S,5R,6R)-3,3-d Chemical compound [K+].N([C@H]1[C@H]2SC([C@@H](N2C1=O)C([O-])=O)(C)C)C(=O)CC1=CC=CC=C1.CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@H]1[C@H](N=C(N)N)[C@@H](O)[C@H](N=C(N)N)[C@@H](O)[C@@H]1O OFPTZWGZSRJCOT-MSPNRCMCSA-M 0.000 description 1
- OZAIFHULBGXAKX-UHFFFAOYSA-N precursor Substances N#CC(C)(C)N=NC(C)(C)C#N OZAIFHULBGXAKX-UHFFFAOYSA-N 0.000 description 1
- 230000002028 premature Effects 0.000 description 1
- 230000002335 preservative Effects 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 201000002728 primary biliary cirrhosis Diseases 0.000 description 1
- 230000000207 pro-atherogenic Effects 0.000 description 1
- 230000001737 promoting Effects 0.000 description 1
- 239000003207 proteasome inhibitor Substances 0.000 description 1
- 230000036678 protein binding Effects 0.000 description 1
- 235000004252 protein component Nutrition 0.000 description 1
- 230000004853 protein function Effects 0.000 description 1
- 238000000164 protein isolation Methods 0.000 description 1
- 201000001474 proteinuria Diseases 0.000 description 1
- 108010030416 proteoliposomes Proteins 0.000 description 1
- 230000017854 proteolysis Effects 0.000 description 1
- 239000002510 pyrogen Substances 0.000 description 1
- LCTONWCANYUPML-UHFFFAOYSA-M pyruvate Chemical compound CC(=O)C([O-])=O LCTONWCANYUPML-UHFFFAOYSA-M 0.000 description 1
- 238000010188 recombinant method Methods 0.000 description 1
- 239000003638 reducing agent Substances 0.000 description 1
- 201000006351 renal artery disease Diseases 0.000 description 1
- 230000000268 renotropic Effects 0.000 description 1
- 238000010405 reoxidation reaction Methods 0.000 description 1
- 238000004007 reversed phase HPLC Methods 0.000 description 1
- 239000002336 ribonucleotide Substances 0.000 description 1
- 125000002652 ribonucleotide group Chemical group 0.000 description 1
- 239000012146 running buffer Substances 0.000 description 1
- 235000019515 salmon Nutrition 0.000 description 1
- 150000003839 salts Chemical class 0.000 description 1
- 201000000306 sarcoidosis Diseases 0.000 description 1
- 238000007423 screening assay Methods 0.000 description 1
- 238000010845 search algorithm Methods 0.000 description 1
- 239000006152 selective media Substances 0.000 description 1
- 230000035939 shock Effects 0.000 description 1
- 230000019491 signal transduction Effects 0.000 description 1
- 239000000377 silicon dioxide Substances 0.000 description 1
- 238000002741 site-directed mutagenesis Methods 0.000 description 1
- 201000000849 skin cancer Diseases 0.000 description 1
- 210000000813 small intestine Anatomy 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 210000001082 somatic cell Anatomy 0.000 description 1
- 238000000527 sonication Methods 0.000 description 1
- 201000011096 spinal cancer Diseases 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 238000007619 statistical method Methods 0.000 description 1
- 150000003432 sterols Chemical class 0.000 description 1
- 235000003702 sterols Nutrition 0.000 description 1
- 230000004936 stimulating Effects 0.000 description 1
- 201000011549 stomach cancer Diseases 0.000 description 1
- NINIDFKCEFEMDL-UHFFFAOYSA-N sulfur Chemical compound [S] NINIDFKCEFEMDL-UHFFFAOYSA-N 0.000 description 1
- 229910052717 sulfur Inorganic materials 0.000 description 1
- 239000011593 sulfur Substances 0.000 description 1
- 238000002198 surface plasmon resonance spectroscopy Methods 0.000 description 1
- 230000004083 survival Effects 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 230000002889 sympathetic Effects 0.000 description 1
- 230000002195 synergetic Effects 0.000 description 1
- 239000003826 tablet Substances 0.000 description 1
- 101710032063 tcf7l1a Proteins 0.000 description 1
- 201000003120 testicular cancer Diseases 0.000 description 1
- 108010013645 tetranectin Proteins 0.000 description 1
- 230000004797 therapeutic response Effects 0.000 description 1
- 230000001732 thrombotic Effects 0.000 description 1
- 201000005161 thyroid carcinoma Diseases 0.000 description 1
- 201000000170 thyroid lymphoma Diseases 0.000 description 1
- 239000003104 tissue culture media Substances 0.000 description 1
- 102000002689 toll-like receptors Human genes 0.000 description 1
- 108020000411 toll-like receptors Proteins 0.000 description 1
- 231100000167 toxic agent Toxicity 0.000 description 1
- 231100000224 toxic side effect Toxicity 0.000 description 1
- 239000003440 toxic substance Substances 0.000 description 1
- 230000037317 transdermal delivery Effects 0.000 description 1
- 238000005829 trimerization reaction Methods 0.000 description 1
- 210000004881 tumor cells Anatomy 0.000 description 1
- 239000005483 tyrosine kinase inhibitor Substances 0.000 description 1
- 229940121358 tyrosine kinase inhibitors Drugs 0.000 description 1
- 230000002485 urinary Effects 0.000 description 1
- 201000011528 vascular disease Diseases 0.000 description 1
- 229960003862 vemurafenib Drugs 0.000 description 1
- GPXBXXGIAQBQNI-UHFFFAOYSA-N vemurafenib Chemical compound CCCS(=O)(=O)NC1=CC=C(F)C(C(=O)C=2C3=CC(=CN=C3NC=2)C=2C=CC(Cl)=CC=2)=C1F GPXBXXGIAQBQNI-UHFFFAOYSA-N 0.000 description 1
- 244000052613 viral pathogens Species 0.000 description 1
- 230000017613 viral reproduction Effects 0.000 description 1
- 239000000304 virulence factor Substances 0.000 description 1
- 239000011782 vitamin Substances 0.000 description 1
- 235000013343 vitamin Nutrition 0.000 description 1
- 229930003231 vitamins Natural products 0.000 description 1
- 239000011534 wash buffer Substances 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P1/00—Drugs for disorders of the alimentary tract or the digestive system
- A61P1/16—Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P11/00—Drugs for disorders of the respiratory system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P11/00—Drugs for disorders of the respiratory system
- A61P11/06—Antiasthmatics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P19/00—Drugs for skeletal disorders
- A61P19/02—Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
- A61P25/28—Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P29/00—Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P3/00—Drugs for disorders of the metabolism
- A61P3/04—Anorexiants; Antiobesity agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/04—Antibacterial agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P39/00—General protective or antinoxious agents
- A61P39/02—Antidotes
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P43/00—Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P9/00—Drugs for disorders of the cardiovascular system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P9/00—Drugs for disorders of the cardiovascular system
- A61P9/10—Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/775—Apolipopeptides
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/30—Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/62—DNA sequences coding for fusion proteins
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N9/00—Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
- C12N9/14—Hydrolases (3)
- C12N9/16—Hydrolases (3) acting on ester bonds (3.1)
- C12N9/18—Carboxylic ester hydrolases (3.1.1)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N9/00—Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
- C12N9/14—Hydrolases (3)
- C12N9/16—Hydrolases (3) acting on ester bonds (3.1)
- C12N9/22—Ribonucleases RNAses, DNAses
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Y—ENZYMES
- C12Y301/00—Hydrolases acting on ester bonds (3.1)
- C12Y301/01—Carboxylic ester hydrolases (3.1.1)
- C12Y301/01002—Arylesterase (3.1.1.2)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Y—ENZYMES
- C12Y301/00—Hydrolases acting on ester bonds (3.1)
- C12Y301/01—Carboxylic ester hydrolases (3.1.1)
- C12Y301/01047—1-Alkyl-2-acetylglycerophosphocholine esterase (3.1.1.47), i.e. platelet-activating factor acetylhydrolase
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Y—ENZYMES
- C12Y301/00—Hydrolases acting on ester bonds (3.1)
- C12Y301/08—Phosphoric triester hydrolases (3.1.8)
- C12Y301/08001—Aryldialkylphosphatase (3.1.8.1), i.e. paraoxonase
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Y—ENZYMES
- C12Y301/00—Hydrolases acting on ester bonds (3.1)
- C12Y301/27—Endoribonucleases producing 3'-phosphomonoesters (3.1.27)
- C12Y301/27005—Pancreatic ribonuclease (3.1.27.5)
Abstract
Compositions and methods relating to ApoA-1 fusion polypeptides are disclosed. The fusion polypeptides include a first polypeptide segment corresponding to an ApoA-1 polypeptide or ApoA-1 mimetic, and may also include a dimerizing domain such as, e.g., an Fc region, which is typically linked carboxyl-terminal to the first polypeptide segment via a flexible linker that is at least 5 amino acids in length. In some embodiments, the fusion polypeptide further includes a second polypeptide segment located carboxyl-terminal to the first polypeptide segment and which confers a second biological activity (e.g., an RNase, paraoxonase, platelet-activating factor acetylhydrolase, cholesterol ester transfer protein, lecithin-cholesterol acyltransferase, or polypeptide that specifically binds to amyloid beta). Also disclosed are dimeric proteins comprising first and second ApoA-1 fusion polypeptides as disclosed herein. The fusion polypeptides and dimeric proteins are useful in methods for therapy. l-terminal to the first polypeptide segment via a flexible linker that is at least 5 amino acids in length. In some embodiments, the fusion polypeptide further includes a second polypeptide segment located carboxyl-terminal to the first polypeptide segment and which confers a second biological activity (e.g., an RNase, paraoxonase, platelet-activating factor acetylhydrolase, cholesterol ester transfer protein, lecithin-cholesterol acyltransferase, or polypeptide that specifically binds to amyloid beta). Also disclosed are dimeric proteins comprising first and second ApoA-1 fusion polypeptides as disclosed herein. The fusion polypeptides and dimeric proteins are useful in methods for therapy.
Description
(12) Granted patent specificaon (19) NZ (11) 738963 (13) B2
(47) Publicaon date: 2021.12.24
(54) APOA-1 FUSION POLYPEPTIDES AND RELATED COMPOSITIONS AND METHODS
(51) Internaonal Patent Classificaon(s):
C07K 14/775 C12N 9/18 C12N 9/22
(22) Filing date: (73) Owner(s):
2016.09.06 THERIPION, INC.
(23) Complete specificaon filing date: (74) Contact:
2016.09.06 FB Rice Pty Ltd
(30) Internaonal Priority Data: (72) Inventor(s):
US 62/215,256 2015.09.08 HAYDEN-LEDBETTER, Martha S.
LEDBETTER, Jeffrey A.
(86) Internaonal Applicaon No.: MONTES, Vince
(87) Internaonal Publicaon number:
WO/2017/044424
(57) Abstract:
Composions and methods relang to ApoA-1 fusion polypepdes are disclosed. The fusion
polypepdes include a first polypepde segment corresponding to an ApoA-1 polypepde or
ApoA-1 mimec, and may also include a dimerizing domain such as, e.g., an Fc region, which
is typically linked carboxyl-terminal to the first polypepde segment via a flexible linker that is
at least 5 amino acids in length. In some embodiments, the fusion polypepde further includes
a second polypepde segment located carboxyl-terminal to the first polypepde segment and
which confers a second biological acvity (e.g., an RNase, paraoxonase, platelet-acvang
factor acetylhydrolase, cholesterol ester transfer protein, lecithin-cholesterol acyltransferase, or
polypepde that specifically binds to amyloid beta). Also disclosed are dimeric proteins comprising
first and second ApoA-1 fusion polypepdes as disclosed herein. The fusion polypepdes and
dimeric proteins are useful in methods for therapy.
NZ 738963 B2
APOA-1 FUSION POLYPEPTIDES AND RELATED COMPOSITIONS AND METHODS
TO RELATED APPLICATIONS
CROSS-REFERENCE
This application claims the benefit of U.S. Provisional Application No. 62/215,256,
filed September 8, 2015, which is incorporated by reference herein in its entirety.
REFERENCE TO SEQUENCE LISTING
The instant application contains
a Sequence Listing which has been submitted in
ASCII
format via EFS-Web and is hereby incorporated by reference in its entirety.
Said ASCII Copy,
created on August 31, 2016, is named "TRP_0110PC_20160831 SeqListingST25" and is 161,132
bytes in size.
BACKGROUND OF THE INVENTION
Apolipoprotein A-I (ApoA-1) and High Density Lipoprotein (HDL)
Cardiovascular disease is the leading cause of mortality in many nations, accounting
for approximately 16.7 million deaths each year world-wide. The most common consequences of
cardiovascular disease are myocardial infarction and stroke, which have a common underlying
etiology of atherosclerosis.
Epidemiological studies since the 1970's have shown that low levels of high density
lipoprotein (HDL) is associated with increased risk for myocardial infarction. This has led to multiple
approaches to new therapies targeting HDL (see Kingwell et al., Nature Reviews Drug Discovery
13:445-64, 2014) and to a consensus view that the process of reverse cholesterol transport (RCT) is
central to beneficial
HDL activity rather than simply an increase in HDL without RCT.
For example,
in clinical trials so far, drugs that increase HDL by inhibition of RCT with inhibitors of cholesterol
ester transfer protein (CETP) have not been efficacious. Further, it has more recently been realized
that measuring levels of HDL is not sufficient to determine its function in patients because HDL is
damaged by oxidation and glycation, including during chemotherapy and in patients with
neurodegenerative disorders. See Keeney et al., Proteomics Clin Appl. 7:109-122, 2013.
Apolipoprotein A-i
(ApoA-1) is the principal protein component of HDL. Phillips,
Journal of Lipid Research 54:2034-2048, 2013. Human ApoA-1 is a 243 amino acid protein, with a
series of eight 22-mer and two 11-mer amphipathic a-helices spanning residues 44-243. Lund-Katz
and Phillips, Subcell Biochem. 51:183-227, 2010. The helices in the amino-terminal two-thirds of the
molecule form a
helix bundle structure, whereas the carboxyl-terminal region forms
a separate,
relatively disorganized domain important for lipid binding. The interaction of the C-terminal segment
with lipids induces conformational changes in the ApoA-1 structure, increasing the a-helix content of
the molecule and allowing subsequent opening of the N-terminal helix bundle. See id. The lipid
affinity of ApoA-1 confers detergent-like properties, and it can solubilize phospholipids to form
discoidal HDL particles containing a segment of phospholipid bilayer and two ApoA-1 molecules
arranged in an anti-parallel, double-belt conformation around the edge of the disc. Phillips, supra.
The conformational adaptability ApoA-1 also confers stability to HDL particles, including discoidal
particles of different sizes as well as spherical HDL particles. See id. These characteristics allow
ApoA-1 to partner with ABCA1 in mediating efflux of cellular phospholipid and cholesterol and the
production of stable HDL particles. See Phillips, supra; Lund-Katz and Phillips, supra.
Due to its important role in HDL particle formation and function, ApoA-1 has
become the focus for several HDL-targeted therapeutic strategies. Drugs including niacin and fibrates
that increase synthesis of ApoA-1, however, also decrease the concentration of VLDL and are thus
not specific for HDL. Clinical trials of niacin were halted due to lack of efficacy, whereas fibrates
that activate peroxisome proliferator activated receptors (PPARs) were found to cause a 10%
reduction in major cardiovascular events (p<0.05) and a 13% reduction in coronary events (p<0.0001)
in a meta-analysis. See Jun et al., Lancet 375:1875, 2010. Because more effective therapies are
needed, there are several other orally active drugs that increase ApoA-1 in preclinical development.
See Kingwell et al., supra.
An alternative approach to increasing ApoA-1 is by direct injection of the purified
protein. See, e.g., Kingwell et al., Circulation 128:1112, 2013. ApoA-1 has been purified from
human plasma (reconstituted HDL) and tested in clinical trials. Recombinant ApoA-1 has also been
expressed in both bacterial and mammalian expression systems and tested in clinical trials. These
studies have shown that infusion of ApoA-1, reconstituted with phospholipids into pre- HDL, causes
reduction of plaque volume and improvement in plaque morphology as measured by intravascular
ultrasound (IVUS) after small (47-60 patients) clinical trials. While promising, use of natural or
recombinant ApoA-1 has several limitations, including a requirement for weekly administration due
to a short ApoA- 1 half-life and a high cost of manufacturing.
Recombinant ApoA-1 Milano, a highly active ApoA-1 mutant, was expressed in
bacterial
cells and tested in clinical trials in patients with acute coronary syndromes
(see Nissen et al.,
JAMA 290:2292, 2003), where reduction in plaque volume was seen. While this study is considered
the first to directly test and confirm the HDL hypothesis, ApoA-1 produced in bacterial systems has
due to low expression levels and high manufacturing costs. Recombinant ApoA-1
not progressed
produced in mammalian cells has progressed further in clinical trials, including recently completed
phase II studies. CER-001, in development by Cerenis Therapeutics, is a recombinant ApoA-1
produced by mammalian cells and formulated with specific lipids to form pre-p-like HDL particles.
According to Cerenis, CER-001 met its primary end point of a reduction in carotid plaque volume
measured by MRI in patients with familial hypercholesterolaemia in the MODE trial (NCT01412034).
In the CHI-SQUARE trial (NCT01201837), Cerenis announced that CER-001 reduced plaque volume
versus baseline in patients with acute coronary syndrome, but the reduction was not significant versus
placebo.
In another study in macaques, ApoA-1 Milano, reconstituted with lipids (POPC), was
infused at relatively high doses (30, 100, and 300 mg/kg) given every second day for 21 infusions.
Kempen et al., J. Lipid. Res. 54:2341-2353, 2013. Drug infusion quickly decreased the endogenous
cholesterol esterification rate, increased the formation of large ApoE-rich particles due to lack of
LCAT activation, and caused a large increase in free cholesterol due to sustained stimulation of
ABCA1-mediated efflux. See id. These results show that infusion of large amounts of reconstituted
ApoA-1 Milano disrupt HDL metabolism by enhancing cholesterol efflux without the ability to
process it through the normal metabolic pathways.
While the prospects for HDL infusion therapy are very promising, there is a need for
improved recombinant ApoA-1 molecules that overcome some of the limitations of current
approaches. Several recombinant ApoA-1 fusion proteins have been produced, including ApoA-1
produced in bacteria with a His tag to simplify purification. See, e.g., Prieto et al., Protein J. 31:681
688, 2012; Ryan et al., Protein Expr. Purif 27:98-103, 2003. In another example, IFNa was attached
to the amino terminus of ApoA-1 through a 3aa (Gly Ala Pro) linker. See Fioravanti et al., J.
Immunol. 188:3988-3992, 2012. The linker in this construct was created by the choice of restriction
enzymes, and the fusion protein was tested by adenovirus delivery to target to the liver and reduce the
toxicity of IFNa therapy. ApoA-1 has also been fused with an Fc domain (ApoAIg) and is
available commercially from Creative Biomart (cat. No. APOA33H) and from Life Technologies
(Cat # 10686-HO2H-5). However, this ApoAIg molecule has very low functional activity (see
Example 1).
Additional recombinant ApoA-1 fusion proteins include anti-CD20 scFv-ApoA-1
(Crosby et al., Biochem. Cell Biol. 10:1139/bcb, 2015), ILApoA-1 (Ochoa et al., Cancer Res.
73:139-149, 2013), and a trimeric ApoA-1 fusion protein made by the addition of the trimerization
domain of human tetranectin (Graversen et al., J. Cardiovascular Pharmacol. 51:170-77, 2008). In
these examples, the fusion was at the N-terminus of ApoA-1.
trimeric tetranectin-ApoA-1 (TN-ApoA-1) was effective in reverse cholesterol
efflux and its half-life in mice was increased to 12 hours versus three hours for monomeric ApoA-1.
See Gaversen et al., supra. In an aggressive model of atherosclerosis (LDLR -/- mice fed a high-fat
diet), trimeric TN-ApoA-1 slowed progression of lesions in the aortic roots. See id. Recent studies in
nonhuman primates, however, showed that multiple infusions of lipidated TN-ApoA-1 were not well
tolerated and resulted in high immunogenicity and
lipid accumulation. See Regeness-Lechner et al.,
Toxilogical Sciences 150:378-89, 2016. The trimer fusion protein was complexed with phospholipids
and injected at concentrations of 100 mg/kg and 400 mg/kg every four days for three weeks, followed
by a six week recovery period. After multiple infusions of lipidated TN-ApoA-1, clinical condition
deteriorated
and was accompanied by changes indicative of a progressive inflammatory
response,
increased levels of cytokines, C-reactive protein and vascular/perivascular infiltrates in multiple
tissues. Rapid formation of antidrug antibodies occurred in all animals receiving lipidated TN-ApoA
1. See id. The accumulation of trimeric TN-ApoA-1 in tissues of the treated animals resembles fibril
formation and deposition of ApoA-1 in patients who have mutations near the N-terminus. See
Mizuguchi et al., J. Biol. Chem. 290:20947-20959, 2015; Das et al., J. Mol. Biol. 2015.10.029; Obici
et al., Amyloid 13:191-205, 2006.
Current forms of ApoA- 1 in clinical development require formulation with specific
lipids into prep-like HDL particles prior to infusion, because the half-life of ApoA- 1 in the absence of
lipids is very short. See Nanjee et al, Arterioscler Thromb Vasc Biol 16:1203-1214, 1996 (showing
that lipid-free ApoA-1 has a half-life of only 2-2.3 hours after either bolus or slow infusion in
humans). After lipid formulation, half-life increases to about 48 hours, so frequent (weekly)
administration is still required.
ApoA-1 therapy has also shown significant benefit in improving insulin sensitivity
and glucose uptake (see Drew et al., Nature Reviews Endocrinology 8:237, 2012), and may be useful
in patients with diabetes and with NASH (non-alcoholic steatohepatitis). In addition, ApoA-1 binds
amyloid-beta and prevents neurotoxicity in cultured hippocampal neuronal cells. See Koldamova et
al., Biochemistry 40:3553, 2001; Paula-Lima et al., Int. J. Biochem. Cell Biol. 41:1361, 2009.
Further, ApoA-1 polymorphisms are linked to risk for Alzheimer's disease and ApoA-1 is found at
decreased levels in patients with neurodegenerative disorders. See Keeney et al., Proteomics Clin.
Appl. 7: 109-122, 2013).
Efficacy of ApoA-1 therapy has also been demonstrated in animal models of cancer.
One study examined the effect of ApoA-1 infusion on growth of tumors in mice. See Zamanian
Daryoush et al., J. Biol. Chem. 288:21237-21252, 2013. Zamanian-Daryoush et al. found that ApoA
1 potently suppresses tumor growth and metastasis in multiple syngeneic tumor models, including
B16F1OL malignant melanoma and Lewis Lung carcinoma. The effect of ApoA-1 was due to
the immune response. Recruitment and expansion of myeloid-derived suppressor cells
modulation of
(MDSC) in the tumors was inhibited. There was also inhibition of tumor angiogenesis and the matrix
degrading protease MMP-9. In contrast, ApoA-l therapy increased CD11b macrophages and
increased amounts of IFNy, IL-12b, and CXCL1O, markers of a Thl response supporting T cell
activation. The authors showed that T cells were required for the potent suppressive effect of ApoA- 1
on tumor growth, and ApoA-l therapy caused a specific increase in CD8' T cells in the tumors. See
id. While the results of Zamanian-Daryoush et al. are promising, the study used high doses of lipid
free ApoA-l to achieve the observed effects (15 mg every second day per mouse), see id., which was
likely required because of the short half-life of ApoA- 1.
Another cancer study showed that ApoA-1 and mimetic peptides (L-4F, D-4F, L-5F)
inhibit tumor development in a murine model of ovarian carcinoma. See Su et al., Proc. Natl. Acad.
Sci. USA 107:19997-20002, 2010. Su et al. found that ApoA-1 overexpression in transgenic mice, or
peptide mimetic administration, reduced stimulatory phospholipids, implicating an additional
mechanism for inhibition of tumor growth. See id.
Studies have also suggested a role for ApoA-1 in the pathogenesis of multiple
sclerosis (MS). In particular, ApoA-1 expression was shown to be lower in MS patients compared to
healthy controls, and primary progressive MS patients had less plasma ApoA-1 than patients with
other forms of MS. See Meyers et al., J. Neuroimmunol. 277:176-185, 2014. Using experimental
allergic encephalomyelitis (EAE) as a model for MS, mice deficient in ApoA-1 exhibited worse
clinical disease and more neurodegeneration compared to wild-type animals. The authors suggest that
agents that increase ApoA-1 levels are possible therapies for MS. See id. Another MS study found
that the ApoA-1 promoter polymorphism A-allele, associated with elevated ApoA-1 levels, is
correlated with improved cognitive performance in patients with MS; A-allele carriers displayed
overall superior cognitive performance and had a three-fold decreased overall risk of cognitive
impairment. See Koutsis et al., Mult. Scler. 15:174-179, 2009.
Peptide Mimetics
ApoA-1 mimetic peptides have shown efficacy in a number of animal models of
disease and have properties that make them attractive as potential therapeutic agents. See, e.g., Reddy
et al., Curr. Opin. Lipidol. 25:304-308, 2014 and White et al., J. Lipid. Res. 55:2007-2021, 2014.
Peptide 4F has been tested in high risk patients with coronary artery disease. Several ApoA-1
mimetic peptides that are resistant to oxidation have been described in the past several years. While
these a-helical peptides show activity in animal models, they require daily dosing because of their
short half-life. In addition, toxicity, including muscle toxicity and hypertriglyceridemia, have been
seen in peptide-treated animals (these toxicities have been seen in mice treated with ApoA-1).
Advances to reduce toxicity by sequence design and to reduce cost of peptide production were
e.g., Bielicki, Curr. Opin. Lipidol. 27:40-46, 2016. Another approach has been to
described. See,
synthesize D-peptides, including the highly studied D-4F peptide. These have a longer half-life and
can be given orally, but the high cost of manufacturing and accumulation of D-peptides in tissues may
be preventing these peptides from moving past initial clinical testing.
RNase
RNase has been studied as a therapy for cancer and autoimmune disease. For cancer
therapy, both natural (onconase, frog RNase), and recombinant human RNasel resistant to inhibition
by cytoplasmic inhibitor (see US Patent No. 8,569,457) have been reported. In addition, targeting of
RNase to tumor cells by conjugation of cytotoxic RNase (onconase) to anti-tumor antibodies has been
reported. See Lui et al., Mol. Cancer 13:1186, 2014; Newton et al., Blood 97:528-535, 2001.
RNase therapy has also been studied in a mouse model of cardiovascular disease. See
Simsekyilmaz et al., Circulation 129:598-606, 2014. They and others show that extracellular RNA
accumulates at sites of vascular injury and that extracellular RNA causes production of inflammatory
cytokines. See Fischer et al., Thromb. Haemost. 108:730-741, 2012. RNase therapy reduced
neointima formation in a mouse model of accelerated cardiovascular disease, reduced plaque
macrophage content, and inhibited leukocyte recruitment to injured carotid arteries in vivo. See
Simsekyilmaz et al., supra.
RNase therapy has also been studied in models of acute stroke, where it was found to
reduce infarction size. See Walberer et al., Curr. Neurovasc. Res. 6:12-19, 2009. Thus systemic
treatment with RNase 1 rescued mice from arterial thrombotic occlusion to limit cerebral edema and
to serve as a potent anti-inflammatory regimen in vivo. In these RNase therapy studies, the RNase
was given by continuous infusion using osmotic minipumps implanted subcutaneously because the
half-life of RNase 1 is very short.
RNase therapy has also been studied in a mouse model of systemic lupus
erythematosus (SLE). See Sun et al., J. Immunol. 190:2536-2543, 2013. Overexpression of TLR7, an
RNA sensor, causes a lupus-like disease with autoantibodies, kidney disease, and early mortality.
Crossing these mice with mice that overexpress RNase A as a transgene resulted in progeny with
increased survival, reduced lymphocyte activation, reduced kidney deposits of IgG and C3, and
reduced hepatic inflammation and necrosis.
Extracellular single stranded viral RNA caused widespread neurodegeneration after
intrathecal administration to mice, and the neuronal damage was mediated by TLR7. See Lehmann et
al., J. Immunol. 189: 1448-58, 2012.
RNase-Ig wherein human RNase 1 is fused to a mutated human IgG1 Fc domain
comprising p238s and p331s mutations (see US Patent No. 8,937,157) is in clinical development by
Resolve Therapeutics in patients with systemic lupus erythematosus (SLE).
Paraoxonase
Human Paraoxonase 1 (PON1) is a lipolactonase with efficient esterase activity and
capable of hydrolyzing organophosphates. PONi prevents LDL and cell membrane oxidation and is
considered to be atheroprotective. PONi is exclusively associated with HDL and contributes to the
antioxidative function of HDL. See, e.g., Mackness et al., Gene 567:12-21, 2015. Reductions in
HDL-PON1 activity are present in a wide variety of inflammatory diseases where loss of PONi
activity leads to dysfunctional HDL which can promote inflammation
and atherosclerosis. See, e.g.,
Eren et al., Cholesterol. 792090 doi 10.1155/2013/792090, 2013. PONi activity is also decreased in
patients with Alzheimer's disease and other dementias, suggesting a possible neuroprotective role of
PON1. See Menini et al., Redox Rep. 19:49-58, 2014.
PON1 has shown protective activity in multiple animal models. Overexpression of
human PON1 inhibited the development of atherosclerosis in mice with combined leptin and LDL
receptor deficiency, a model of metabolic syndrome. See Mackness et al., Arterioscler. Thromb.
Vasc. Biol. 26:1545-50, 2006.
In another study, injection of recombinant PONi to mice prior to STZ-induced
diabetes resulted in reduced incidence of diabetes and higher serum insulin levels. Addition of HDL
simultaneously
with PONi had an additive effect on insulin secretion.
See Koren-Gluser et al.,
Atherosclerosis 219:510-518, 2011.
In another study, a PONi fusion protein containing a protein transduction domain
(PTD) was used to transduce PONi into cells and tissues. PONi transduction protected microglial
cells in vitro from oxidative stress-induced inflammatory responses and protected against
dopaminergic neuronal cell death in a Parkinsons disease model. See Kim et al., Biomaterials 64:45
56, 2015.
In another study, recombinant PONi was administered to mice where there was a
significant reduction in cholesterol mass and an inhibition in the cholesterol biosynthesis rate, effects
that could probably lead to attenuation of atherosclerosis. See Rosenblat et al., Biofactors 37:462
467, 2011.
In another study, mice were given recombinant adenovirus PONt or PON3 and either
was shown to protect against CCl(4)-induced liver injury. Overexpression of either human PONt or
human PON3 reduced hepatic oxidative stress and strengthened the antioxidant capabilities in the
liver. See Peng et al., Toxicol. Lett. 193:159-166, 2010.
In another study, PONI was fused to the C-terminus of an Fc domain, and expressed
as a bispecific molecule using an antibody to human insulin receptor (HIR). This molecule, termed
HIRMAb-PONI, was stable after expression in CHO cells, and was shown in Rhesus monkeys to
have a high blood brain barrier
permeation but was rapidly cleared by the liver. See Boado et al.,
Biotechnol. Bioeng. 108: 186-196, 2011.
Platelet-activating factor acetylhydrolase
Platelet-activating factor acetylhydrolase (PAF-AH) is an LDL and HDL-associated
enzyme that hydrolyzes short chain acyl groups of phospholipids such as platelet-activating factor and
oxidized phospholipids to reduce their inflammatory properties. See Watson et al., J. Clin. Invest.
95:774-782, 1995; Stafforini, Cardiovasc. Drugs Ther. 23:73-83, 2009. Therapy with PAF-AH
through adenovirus-mediated gene delivery has been reported to ameliorate proteinuria and
glomerulosclerosis in a rat model. See Iso-O et al., Molecular Therapy 13:118-126, 2006. PAF-AH
also enhanced liver recovery after paracetamol intoxication in the rat, and PAF is associated with liver
toxicity from high doses of acetaminophen. See Grypioti et al., Dig. Dis. Sci. 52:2580-2590, 2007;
Grypioti et al., Dig. Dis. Sci. 53:1054-1062, 2008. A mutation in PAF-AH that causes a loss of
function is present in 4% of Japanese, and PAF-AH was found to be an independent risk factor for
cardiovascular disease and stroke in these individuals. See Blankenberg et al., J. Lipid Res. 44:1381
1386, 2003. Recombinant PAF-AH was tested in phase III clinical trials in patients with acute
respiratory distress syndrome (ARDS) and in patients with sepsis. See Karabina et al., Biochim.
Biophys. Acta 1761:1351-1358, 2006.
Cholesteryl ester transfer protein
Cholesteryl ester transfer protein (CETP) transports cholesteryl ester from high
density lipoproteins (HDL) to low density and very low density lipoproteins (LDL and VLDL). Many
CETP inhibitors have been developed and tested in clinical trials. Torecetrapib was the first CETP
inhibitor to advance to late stage clinical trials, and showed a significant effect on plasma lipoprotein
levels, raising antiatherogenic HDL cholesterol levels while lowering proatherogenic LDL cholesterol
levels. Torecetrapib binds deeply within CETP and shifts the bound cholesteryl ester in the N
terminal
pocket of the hydrophobic tunnel and displaces phospholipid from
the pocket. See Liu et al.,
J. Biol. Chem. 287:37321-37329, 2012. Initial hopes that CETP inhibitors would be useful for
therapy of cardiovascular disease have not been fulfilled; four inhibitors have reached late stage
clinical trials but have failed to show a reduction in cardiovascular events. See Kosmas et al., Clinical
Medical Insights: Cardiology 2016: 10 37-42 doi: 10.4137/CMC.S32667).
Alternative views of CETP inhibitors and cardiovascular disease have emerged. See,
e.g., Miller, F100Research 3:124, 2014. There is mounting evidence for a protective role of CETP.
multiple studies in man now show that cardiovascular disease is related inversely to
For example,
CETP levels. In addition, CETP alleles that have reduced hepatic secretion are associated with
increased risk of myocardial infarction. See Miller, supra. The original idea, that CETP inhibitors
increase HDL cholesterol levels
and would therefore be beneficial in reducing cardiovascular disease,
may not be correct. It is likely that HDL cholesterol is beneficial because of its lipid transport
function, and the CETP-mediated transfer of cholesteryl ester from HDL to LDL and VLDL is an
important component of this function.
SUMMARY OF THE INVENTION
In one aspect, the present invention provides a fusion polypeptide comprising, from
an amino-terminal position to a carboxyl-terminal position, ApoAl-L1-D, where ApoAl is a first
polypeptide segment having cholesterol efflux activity and which is selected from (i) a polypeptide
comprising an amino acid sequence having at least 90% or at least 95% identity with amino acid
residues 19-267, 25-267, or 1-267 of SEQ ID NO:2 and (ii) an ApoA-i mimetic; LI is a first
polypeptide linker; and D is a dimerizing domain. In certain embodiments, LI comprises at least two
amino acid residues, at least three amino acid residues, or at least 16 amino acid residues. For
example, in particular variations, LI consists of from two to 60 amino acid residues, from three to 60
amino acid residues, from five to 40 amino acid residues, from 15 to 40 amino acid residues, or from
16 to 36 amino acid residues. In a more specific variation, LI consists of 16 amino acid residues, 21
amino acid residues, 26 amino acid residues, 31 amino acid residues, or 36 amino acid residues; in
some such embodiments, LI has the amino acid sequence shown in residues 268-283 of SEQ ID
NO:22, residues 268-288 of SEQ ID NO:26, residues 268-293 of SEQ ID NO:2, SEQ ID NO:54, or
residues 268-303 of SEQ ID NO:24. In certain embodiments, the first polypeptide segment comprises
the amino acid sequence shown in residues 19-267 or 25-267 of SEQ ID NO:2.
In some embodiments of a fusion polypeptide as above, D is an immunoglobulin
heavy chain constant region such as, for example, an immunoglobulin Fc region. In certain
embodiments where the dimerizing domain is an immunoglobulin Fc region, the Fc region is a human
Fc region such as, e.g., a human Fc variant comprising one or more amino acid substitutions relative
to the wild-type human sequence. Particularly suitable Fc regions include human 71 and 73 Fc
regions. In some variations, the Fc region is a human 71 Fc variant in which Eu residue C220 is
replaced by serine; in some such embodiments Eu residues C226 and C229 are each replaced by
serine, and/or Eu residue P238 is replaced by serine. In further variations comprising an Fc region as
above, the Fc region is a human 71 Fc variant in which Eu residue P331 is replaced by serine. Fc
variants may include an amino acid substitution that reduces glycosylation relative to the wild-type
human sequence; in some such embodiments, Eu residue N297 is replaced with another amino acid.
comprising an Fc region as above, the Fc region is an Fc variant comprising an
In further variations
amino acid substitution that increases or reduces binding affinity for an Fc receptor (e.g., an amino
acid substitution that increases or reduces binding affinity for at least one of FcyRI, FcyRII, and
FcyRIII). In certain embodiments, an Fc variant includes an amino acid substitution that increases or
reduces binding affinity for the neonatal Fc receptor (FcRn). Suitable Fc regions include (i) an Fc
region comprising the amino acid sequence shown in residues 294-525 or 294-524 of SEQ ID NO:2
and (ii) an Fc region comprising the amino acid sequence shown in residues 294-525 or 294-524 of
SEQ ID NO:13.
In certain embodiments of a fusion polypeptide as above, the fusion polypeptide
comprises an amino acid sequence having at least 90% or at least 95% identity with (i) residues 19
525, 19-524, 25-525, or 25-524 of SEQ ID NO:2, (ii) residues 19-525, 19-524, 25-525, or 25-524 of
SEQ ID NO:13, (iii) residues 19-501, 19-500, 25-501, or 25-501 of SEQ ID NO:20, (iv) residues 19
515, 19-514, 25-515, or 25-514 of SEQ ID NO:22, (v) residues 19-520, 19-519, 25-520, or 25-519 of
SEQ ID NO:26, or (vi) residues 19-535, 19-534, 25-535, or 25-534 of SEQ ID NO:24. In more
specific variations, the fusion polypeptide comprises the amino acid sequence shown in (i) residues
19-525, 19-524, 25-525, or 25-524 of SEQ ID NO:2, (ii) residues 19-525, 19-524, 25-525, or 25-524
of SEQ ID NO:13, (iii) residues 19-501, 19-500, 25-501, or 25-501 of SEQ ID NO:20, (iv) residues
19-515, 19-514, 25-515, or 25-514 of SEQ ID NO:22, (v) residues 19-520, 19-519, 25-520, or 25-519
of SEQ ID NO:26, or (vi) residues 19-535, 19-534, 25-535, or 25-534 of SEQ ID NO:24.
In some embodiments of a fusion polypeptide as above, the fusion polypeptide further
includes a second polypeptide segment located carboxyl-terminal to the dimerizing domain. In
particular variations, the second polypeptide segment is an RNase, a paraoxonase, a platelet-activating
factor acetylhydrolase (PAF-AH), a cholesterol ester transfer protein (CETP), a lecithin-cholesterol
acyltransferase (LCAT), or a polypeptide that specifically binds to amyloid beta (AP) such as, e.g., an
AP-specific scFv. A fusion polypeptide comprising a second polypeptide segment as above may be
represented by the formula ApoAl-L1-D-L2-P (from an amino-terminal position to a carboxyl
terminal position), where ApoAl, LI, and D are each defined as above, where L2 is a second
polypeptide linker and is optionally present, and where P is the second polypeptide segment. In some
embodiments of a fusion polypeptide where L2 is present, L2 has the amino acid sequence shown in
residues 526-541 of SEQ ID NO:4.
In another aspect, the present invention provides a fusion polypeptide comprising a
first polypeptide segment having cholesterol efflux activity and which is selected from (i) a
polypeptide comprising an amino acid sequence having at least 90% or at least 95% identity with
amino acid residues 19-267 or 25-267 of SEQ ID NO:2 and (ii) an ApoA-1 mimetic, and a second
polypeptide segment located carboxyl-terminal to the first polypeptide segment, where the second
segment is selected from an RNase, a paraoxonase, a platelet-activating factor
polypeptide
acetylhydrolase (PAF-AH), a cholesterol ester transfer protein (CETP), a lecithin-cholesterol
acyltransferase (LCAT), and a polypeptide that specifically binds to amyloid beta such as, e.g., an Ap
specific scFv. In some embodiments, the first polypeptide segment has the amino acid sequence
shown in residues 19-267 or 25-267 of SEQ ID NO:2. In some variations, the fusion polypeptide
further includes a linker polypeptide located carboxyl-terminal to the first polypeptide segment and
amino-terminal to the second polypeptide segment. In some embodiments, the fusion polypeptide
further includes a dimerizing domain.
In some embodiments of a fusion polypeptide as above comprising an RNase as a
second polypeptide segment, the RNase is human RNAse 1 or a functional variant or fragment
thereof. In certain embodiments, the RNase has at least 90% or at least 95% identity with amino acid
residues 542-675 of SEQ ID NO:4. In a specific variation, the RNase has the amino acid sequence
shown in residues 542-675 of SEQ ID NO:4. In particular embodiments of a fusion polypeptide
comprising an RNase and having the formula ApoAl-L1-D-L2-P as above, the fusion polypeptide
comprises an amino acid sequence having at least 90% or at least 95% identity with (i) residues 19
675 or 25-675 of SEQ ID NO:4 or (ii) residues 19-675 or 25-675 of SEQ ID NO:14; in some such
embodiments, the fusion polypeptide comprises the amino acid sequence shown in (i) residues 19
675or 25-675 of SEQ ID NO:4 or (ii) residues 19-675 or 25-675 of SEQ ID NO: 14.
In some embodiments of a fusion polypeptide as above comprising a paraoxonase as
a second polypeptide segment, the paraoxonase is human paraoxonase 1 (PONI) or a functional
variant thereof. In certain embodiments, the paraoxonase has at least 90% or at least 95% identity
with amino acid residues 16-355 of SEQ ID NO: 12, amino acid residues 16-355 of SEQ ID NO:42, or
amino acid residues 16-355 of SEQ ID NO:44. In specific variations, the paraoxonase comprises the
amino acid sequence shown in residues 16-355
of SEQ ID NO: 12, residues 16-355 of SEQ ID NO:42,
or residues 16-355 of SEQ ID NO:44. In particular embodiments of a fusion polypeptide comprising
an paraoxonase and having the formula ApoAl-L1-D-L2-P as above, the fusion polypeptide
comprises an amino acid sequence having at least 90% or at least 95% identity with (i) residues 19
883 or 25-883 of SEQ ID NO:28, (ii) residues 19-873 or 25-873 of SEQ ID NO:38, (iii) residues 19
883 or 25-883 of SEQ ID NO:46, or (iv) residues 19-883 or 25-883 of SEQ ID NO:48; in some such
embodiments, the fusion polypeptide comprises the amino acid sequence shown in (i) residues 19-883
or 25-883 of SEQ ID NO:28, (ii) residues 19-873 or 25-873 of SEQ ID NO:38, (iii) residues 19-883
or 25-883 of SEQ ID NO:46, or (iv) residues 19-883 or 25-883 of SEQ ID NO:48.
In some embodiments of a fusion polypeptide as above comprising a platelet
activating factor acetylhydrolase (PAF-AH) as a second polypeptide segment, the platelet-activating
factor acetylhydrolase is a human PAF-AH
or a functional variant thereof. In certain embodiments,
factor acetyihydrolase has at least 90% or at least 95% identity with amino acid
the platelet-activating
residues 22-441 of SEQ ID NO:32. In a specific variation, the paraoxonase comprises the amino acid
sequence shown in residues 22-441 of SEQ ID NO:32. In particular embodiments of a fusion
polypeptide comprising an platelet-activating factor acetylhydrolase and having the formula ApoAl
L1-D-L2-P as above, the fusion polypeptide comprises an amino acid sequence having at least 90% or
at least 95% identity with residues 19-963 or 25-963
of SEQ ID NO:34; in some such embodiments,
the fusion polypeptide comprises the amino acid sequence shown in residues 19-963 or 25-963 of
SEQ ID NO:34.
In some embodiments of a fusion polypeptide as above comprising a cholesterol ester
transfer protein (CETP) as a second polypeptide segment, the cholesterol ester transfer protein is
human CETP or a functional variant thereof. In certain embodiments, the cholesterol ester transfer
protein has at least 90% or at least 95% identity with amino acid residues 18-493 of SEQ ID NO:30.
In a specific variation, the cholesterol ester transfer protein comprises the amino acid sequence shown
in residues 18-493 of SEQ ID NO:30. In particular embodiments of a fusion polypeptide comprising
an platelet-activating factor acetylhydrolase and having the formula ApoAl-L1-D-L2-P as above, the
fusion polypeptide comprises an amino acid sequence having at least 90% or at least 95% identity
with residues 19-1019 or 25-1019 of SEQ ID NO:40; in some such embodiments, the fusion
polypeptide comprises the amino acid sequence shown in residues 19-1019 or 25-1019 of SEQ ID
NO:40.
In certain embodiments of a fusion polypeptide as above, the fusion polypeptide is
linked to a myeloperoxidase (MPO) inhibitor.
In another aspect, the present invention provides a dimeric protein comprising a first
fusion polypeptide and a second fusion polypeptide, where each of said first and second fusion
polypeptides is a fusion polypeptide comprising a dimerizing domain, as described above.
In another aspect, the present invention provides a polynucleotide encoding a fusion
polypeptide as described above.
In still another aspect, the present invention provides an expression vector comprising
the following operably linked elements: a transcription promoter, a DNA segment encoding a fusion
polypeptide as described above, and a transcription terminator. Also provided is a cultured cell into
which has been introduced an expression vector as above, wherein the cell expresses the DNA
segment.
In another aspect, the present invention provides a method of making a fusion
polypeptide. The method generally includes culturing a cell into which has been introduced an
as described above, where the cell expresses the DNA segment and the encoded
expression vector
fusion polypeptide is produced, and recovering the fusion polypeptide.
In yet another aspect, the present invention provides a method of making a dimeric
protein. The method generally includes culturing a cell into which has been introduced an expression
vector as described above, where the cell expresses the DNA segment and the encoded fusion
polypeptide is produced as a dimeric protein, and recovering the dimeric protein.
In another aspect, the present invention provides a composition comprising a fusion
polypeptide as described above and a pharmaceutically acceptable carrier.
In another aspect, the present invention provides a composition comprising a dimeric
protein as described above and a pharmaceutically acceptable carrier.
In still another aspect, the present invention provides a method for treating a
cardiovascular disease characterized by atherosclerosis. The method generally includes administering
to a subject having the cardiovascular disease an effective amount of a fusion polypeptide or dimeric
fusion protein as described above. In some embodiments, the cardiovascular disease is selected from
the group consisting of coronary heart disease and stroke. In certain variations, the coronary heart
disease is characterized by acute coronary syndrome.
In another aspect, the present invention provides a method for treating a
neurodegenerative disease. The method generally includes administering to a subject having the
neurodegenerative disease an effective amount of a fusion polypeptide or dimeric fusion protein as
described above. In some embodiments, the neurodegenerative disease is selected from the group
consisting of Alzheimer's disease and multiple sclerosis. In certain embodiments, the
neurodegenerative disease is characterized by dementia; in some such variations, the
neurodegenerative disease is Alzheimer's disease.
In another aspect, the present invention provides a method for treating a disease
characterized by amyloid deposit. The method generally includes administering to a subject having
the disease characterized by amyloid deposit an effective amount of a fusion polypeptide or dimeric
fusion protein as described above. In some embodiments, the disease is Alzheimer's disease.
In another aspect, the present invention provides a method for treating an
autoimmune disease. The method generally includes administering to a subject having the
autoimmune disease an effective amount of a fusion polypeptide or dimeric fusion protein as
described above. In some embodiments, the autoimmune disease is selected from the group
consisting of rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, and type 1
diabetes.
In yet another aspect, the present invention provides a method for treating an
inflammatory disease. The method generally includes administering to a subject having the
inflammatory disease an effective amount of a fusion polypeptide or dimeric fusion protein as
described above. In some embodiments, the inflammatory disease is selected from the group
consisting of rheumatoid arthritis,
systemic lupus erythematosus, multiple sclerosis, type 1 diabetes,
type 2 diabetes, obesity, non-alcoholic steatohepatitis, coronary heart disease, and stroke. In other
embodiments, the inflammatory disease is an inflammatory lung disease
such as, for example, asthma,
chronic obstructive pulmonary disease (COPD), bronchiectasis,
idiopathic pulmonary fibrosis,
hyperoxia, hypoxia, or acute respiratory distress syndrome.
In still another aspect, the present invention provides a method for treating an
infectious disease. The method generally includes administering to a subject having the infectious
disease an effective amount of a fusion polypeptide or dimeric fusion protein as described above. In
certain embodiments, the infectious disease is characterized by a bacterial infection; in some such
embodiments, the bacterial infection is a Pseudomonas aeruginosa infection.
In another aspect, the present invention provides a method for treating nephrotic
syndrome (NS). The method generally includes administering to a subject having nephrotic syndrome
an effective amount of a fusion polypeptide or dimeric fusion protein as described above. In specific
variations, the subject's nephrotic syndrome is associated with a disease selected from the group
consisting of a primary kidney disease (e.g., minimal-change
nephropathy, focal glomerulosclerosis,
membranous nephropathy, or IgA nephropathy), amyloidosis, systemic lupus erythematosus, type 1
diabetes, and type 2 diabetes.
In yet another aspect, the present invention provides a method for treating exposure to
sulfur mustard gas or to an organophosphate. The method generally includes administering to a
subject exposed to the sulfur mustard gas or to the organophosphate an effective amount of a fusion
polypeptide or dimeric fusion protein as described above.
In still another aspect, the present invention provides a method for treating cancer.
The method generally includes administering to a subject having cancer an effective amount of a
fusion polypeptide or dimeric fusion protein as described above. In some embodiments, the cancer is
selected from the group consisting of malignant melanoma, renal cell carcinoma, non-small cell lung
cancer, bladder cancer, and head and neck cancer. In certain variations, the cancer treatment is a
combination therapy. In some combination therapy embodiments, the combination therapy includes a
non-ApoAl -mediated immunomodulatory therapy such as, e.g., an immunomodulatory therapy
comprising an anti-PD-i/PD-Li therapy, an anti-CTLA-4 therapy, or both. In other combination
therapy embodiments, the combination therapy includes radiation therapy or chemotherapy. In some
combination therapy embodiments, the combination therapy includes a targeted therapy; in some such
the targeted therapy includes (i) a therapeutic monoclonal antibody targeting a specific
embodiments,
cell-surface or extracellular antigen (e.g., VEGF, EGFR, CTLA-4, PD-1, or PD-Li) or (ii) a small
molecule targeting an intracellular protein such as, for example, an intracellular enzyme (e.g., a
proteasome, a tyrosine kinase, a cyclin-dependent kinase, serine/threonine-protein kinase B-Raf
(BRAF), or a MEK kinase).
These and other aspects of the invention will become evident upon reference to the
following detailed description of the invention.
DEFINITIONS
Unless defined otherwise, all technical and scientific terms used herein have the same
meaning as commonly understood by one of ordinary skill in the art pertinent to the methods and
compositions described. As used herein, the following terms and phrases have the meanings ascribed
to them unless specified otherwise.
The terms "a," "an," and "the" include plural referents, unless the context clearly
indicates otherwise.
A "polypeptide" is a polymer of amino acid residues joined
by peptide bonds,
whether produced naturally or synthetically. Polypeptides of less than about 10 amino acid residues
are commonly referred to as "peptides."
A "protein" is a macromolecule comprising one or more polypeptide chains. A
protein may also comprise non-peptidic components, such as carbohydrate groups. Carbohydrates
and other non-peptidic substituents may be added to a protein by the cell in which the protein is
produced, and will vary with the type of cell. Proteins are defined herein in terms of their amino acid
backbone structures; substituents such as carbohydrate groups are generally not specified, but may be
present nonetheless.
The terms "amino-terminal" (or "N-terminal") and "carboxyl-terminal" (or "C
terminal") are used herein to denote positions within polypeptides. Where the context allows, these
terms are used with reference to a particular sequence or portion of a polypeptide to denote proximity
or relative position. For example, a certain sequence positioned carboxyl-terminal to a reference
sequence within a polypeptide is
located proximal to the carboxyl terminus of the reference sequence,
but is not necessarily at the carboxyl terminus of the complete polypeptide.
The terms "polynucleotide" and "nucleic acid" are used synonymously herein and
refer to a single- or double-stranded polymer of deoxyribonucleotide or ribonucleotide bases read
from the 5' to the 3' end. Polynucleotides include RNA and DNA, and may be isolated from natural
in vitro, or prepared from a combination of natural and synthetic molecules.
sources, synthesized
Sizes of polynucleotides are expressed as base pairs (abbreviated "bp"), nucleotides ("nt"), or
kilobases ("kb"). Where the context allows, the latter two terms may describe polynucleotides that are
single-stranded or double-stranded. It will be recognized by those skilled in the art that the two
strands of a double-stranded polynucleotide may differ slightly in length and that the ends thereof may
be staggered as a result of enzymatic cleavage; thus all nucleotides within a double-stranded
polynucleotide molecule may not be paired. Such unpaired ends will in general not exceed 20 nt in
length.
A "segment" is a portion of a larger molecule (e.g., polynucleotide or polypeptide)
having specified attributes. For example, a DNA segment encoding a specified polypeptide is a
portion of a longer DNA molecule, such as a plasmid or plasmid fragment that, when read from the 5'
to the 3' direction, encodes the sequence of amino acids of the specified polypeptide. Also, in the
context of a fusion polypeptide in accordance with the present invention, a polypeptide segment
"having cholesterol efflux activity" and "comprising an amino acid sequence having at least 90% or at
least 95% identity with amino acid residue 19-267 or 25-267 of SEQ ID NO:2" is a portion of the
longer polypeptide fusion molecule that, in addition to the specified polypeptide segment having
cholesterol efflux activity, includes other polypeptide segments (e.g., linker(s), dimerizing domain) as
described herein.
The term "expression vector" is used to denote a DNA molecule,
linear or circular,
that comprises a segment encoding a polypeptide of interest operably linked to additional segments
that provide for its transcription. Such additional segments include promoter and terminator
sequences, and may also include one or
more origins of replication, one or more selectable markers,
an enhancer, a polyadenylation signal, etc. Expression vectors are generally derived from plasmid or
viral DNA, or may contain elements of both.
The term "promoter" is used herein for its art-recognized meaning to denote a portion
of a gene containing DNA sequences that provide for the binding of RNA polymerase and initiation
of transcription. Promoter sequences are commonly, but not always, found in the 5' non-coding
regions of genes.
A "secretory signal sequence" is a DNA sequence that encodes a polypeptide (a
"secretory peptide") that, as a component of a larger polypeptide, directs the larger polypeptide
through a secretory pathway of a cell in which it is synthesized. The larger polypeptide is commonly
cleaved to remove the secretory peptide during transit through the secretory pathway.
"Operably linked" means that two or more entities are joined together such that they
function in concert for their intended purposes. When referring to DNA segments, the phrase
example, that coding sequences are joined in the correct reading frame, and transcription
indicates, for
initiates in the promoter and proceeds through the coding segment(s) to the terminator. When
referring to polypeptides, "operably linked" includes both covalently (e.g., by disulfide bonding) and
non-covalently (e.g., by hydrogen bonding, hydrophobic interactions, or salt-bridge interactions)
linked sequences, wherein the desired function(s) of the sequences are retained.
term "recombinant" when used with reference, e.g., to a cell,
nucleic acid,
protein, or vector, indicates that the cell, nucleic acid, protein, or vector, has been modified by the
introduction of a heterologous nucleic acid or protein or the alteration of a native nucleic acid or
protein, or that the cell is derived from a cell so modified. Thus, for example, recombinant cells
express genes that are not found within the native (non-recombinant) form of the cell or express
native genes that are otherwise abnormally expressed, under-expressed or not expressed at all. By the
term "recombinant nucleic acid" herein is meant nucleic acid, originally formed in vitro, in general, by
the manipulation of nucleic acid using, e.g., polymerases and endonucleases, in a form not normally
found in nature. In this manner, operable linkage of different sequences is achieved. Thus an isolated
nucleic acid, in a linear form, or an expression vector formed in vitro by ligating DNA molecules that
are not normally joined, are both considered recombinant for the purposes disclosed herein. It is
understood that once a recombinant nucleic acid is made and reintroduced into a host cell or
organism, it will replicate non-recombinantly, i.e., using the in vivo cellular machinery of the host cell
rather than in vitro manipulations; however, such nucleic
acids, once produced recombinantly,
although subsequently replicated non-recombinantly, are still considered recombinant for the purposes
disclosed
herein. Similarly, a "recombinant protein" is a protein made using
recombinant techniques,
i.e., through the expression of a recombinant nucleic acid as depicted above.
The term "heterologous," when
used with reference to portions of a nucleic acid,
indicates that the nucleic acid comprises two or more subsequences that are not normally found in the
same relationship to each other in nature. For instance, the nucleic acid is typically recombinantly
produced, having two or more sequences, e.g., from unrelated genes arranged to make a new
functional nucleic acid, e.g., a promoter from one source and a coding region from another source.
Similarly, "heterologous," when used in reference to portions of a protein, indicates that the protein
comprises two or more subsequences that are not found in the same relationship to each other in
nature (e.g., two or segments of a fusion polypeptide).
An "immunoglobulin" is a serum protein which functions as an antibody in a
vertebrate organism.
Five classes of "immunoglobulin," or antibody, protein (IgG, IgA,
IgM, IgD,
and IgE) have been identified in higher vertebrates. IgG comprises the major class; it normally exists
as the second most abundant protein
found in plasma. In humans, IgG consists of four subclasses,
designated IgGI, IgG2, IgG3, and IgG4. The heavy chain constant regions of the IgG class are
identified with
the Greek symbol y. For example, immunoglobulins of the IgGI
subclass contain a 71
heavy chain constant region. Each immunoglobulin heavy chain possesses a constant region that
consists of constant region protein domains (CHI, hinge, CH2, and CH3) that are essentially invariant
for a given subclass in a species. DNA sequences encoding human and non-human immunoglobulin
chains are known in the art. See, e.g., Ellison et al., DNA 1:11-18, 1981; Ellison et al., Nuc. Acids
Res. 10:4071-4079, 1982; Kenten et
al., Proc. Natl. Acad. Sci. USA 79:6661-6665, 1982; Seno et al.,
Nuc. Acids Res. 11:719-726, 1983; Riechmann et al., Nature 332:323-327, 1988; Amster et al., Nuc.
Acids Res. 8:2055-2065, 1980; Rusconi and Kohler, Nature 314:330-334, 1985; Boss et al., Nuc.
Acids Res. 12:3791-3806, 1984; Bothwell et al., Nature 298:380-382,
1982; van der Loo et al.,
Immunogenetics 42:333-341, 1995; Karlin et al., J. Mol. Evol. 22:195-208,
1985; Kindsvogel et al.,
DNA 1:335-343, 1982; Breiner et al., Gene 18:165-174, 1982; Kondo et al., Eur. J. Immunol. 23:245
249, 1993; and GenBank Accession No. J00228. For a review of immunoglobulin structure and
function, see Putnam, The Plasma
Proteins, Vol V, Academic Press, Inc., 49-140, 1987; and Padlan,
Mol. Immunol. 31:169-217, 1994.
An "immunoglobulin hinge" is that portion of an immunoglobulin heavy chain
connecting the CHI and CH2 domains. The hinge region of human 71 corresponds approximately to
Eu residues 216-230.
The terms "Fc fragment," "Fc region," or "Fc domain," as used herein, are
synonymous and refer to the portion of an immunoglobulin that is responsible for binding to antibody
receptors on cells and the Clq component of complement (in the
absence of any amino acid changes,
relative to the naturally occurring sequence, to remove such binding activity). Fc stands for "fragment
crystalline," the fragment of an antibody that will readily form a protein crystal. Distinct protein
fragments, which were originally described by proteolytic digestion, can define the overall general
structure of an immunoglobulin protein. As originally defined in the literature, the Fc fragment
consists of the disulfide-linked heavy chain hinge
regions, CH2, and CH3 domains. As used herein,
the term also refers to a single chain consisting of CH3, CH2, and at least a portion of the hinge
sufficient to form a disulfide-linked dimer with a second such chain. As used herein, the term Fc
region further includes variants of naturally occurring sequences, where the variants are capable of
forming dimers and including such variants that have increased or decreased Fc receptor-binding or
complement-binding activity.
"Dimerizing domain," as used herein, refers to a polypeptide having affinity for a
second polypeptide, such that the two polypeptides associate under physiological conditions to form a
dimer. Typically, the second polypeptide is the same polypeptide, although in some variations the
second polypeptide is different. The polypeptides may interact with each other through covalent
and/or non-covalent association(s). Examples of dimerizing domains include an Fc region; a hinge
a leucine zipper domain (e.g., a jun/fos
region; a CH3 domain; a CH4 domain; a CHI or CL domain;
leucine zipper domain, see, e.g., Kostelney et al., J. Immunol., 148:1547-1553, 1992; or a yeast GCN4
leucine zipper domain); an isoleucine zipper domain; a dimerizing region of a dimerizing cell-surface
receptor (e.g., interleukin-8 receptor (IL-8R); or an integrin heterodimer such as LFA-1 or
GPIIb/IIIa); a dimerizing region
of a secreted, dimerizing ligand (e.g., nerve growth factor (NGF),
neurotrophin-3 (NT-3), interleukin-8 (IL-8), vascular endothelial growth factor (VEGF), or brain
derived neurotrophic factor (BDNF); see, e.g., Arakawa et al., J. Biol.
Chem. 269:27833-27839, 1994,
and Radziejewski et al., Biochem. 32:1350, 1993); and a polypeptide comprising at least one cysteine
residue (e.g., from about one, two, or three to about ten cysteine residues) such that disulfide bond(s)
can form between the polypeptide and a second polypeptide comprising at least one cysteine residue
(hereinafter "a synthetic hinge"). A preferred dimerizing domain in accordance with the present
invention is an Fc region.
The term "dimer" or "dimeric protein" as used herein, refers to a multimer of two
("first" and "second") fusion polypeptides as disclosed herein linked together via a dimerizing
domain. Unless the context clearly indicates otherwise, a "dimer" or "dimeric protein" includes
reference to such dimerized first and second fusion polypeptides in the context of higher order
multimers that may form in spherical HDL particles (e.g., trimers), such as through an interaction of
dimerized first and second fusion polypeptides with another ApoA-1 polypeptide that may be present
(e.g., through interaction with a naturally occurring, endogenous ApoA-1 protein). The term also
includes reference to dimerized first and second fusion polypeptides in the context of higher order
multimers that may be created by inclusion of an additional dimerizing domain in a first or second
fusion polypeptide (e.g., a first fusion polypeptide comprising an immunoglobulin light chain and a
second fusion polypeptide comprising an immunoglobulin heavy chain can heterodimerize via the
interaction between the CHI and CL domains, and two such heterodimers may further dimerize via
the Fc region of the immunoglobulin heavy chain, thereby forming a tetramer).
The term "linker" or "polypeptide linker" is used herein to indicate two or more
amino acids joined by peptide bond(s) and linking two discrete, separate polypeptide regions. The
linker is typically designed to allow the separate polypeptide regions to perform their separate
functions (such as, e.g., where a dimerizing domain, linked to other polypeptide regions, associates
with another, corresponding dimerization domain to form a dimer). The linker can be a portion of a
native sequence, a variant thereof, or a synthetic sequence. Linkers are also referred to herein using
the abbreviation "L." The use of a subscript (e.g., "1" or "2") with "L" is used herein to differentiate
among multiple linkers within a polypeptide chain, which linkers may be the same or different with
respect to amino acid sequence.
the context clearly indicates otherwise, reference herein to "ApoA-i" is
Unless
understood to
include naturally occurring ApoA-1 polypeptides as well as functional
variants,
functional fragments, and mimetics thereof. "ApoAl," Apo A-i," "apoA-i," and "apo A-i" are used
herein synonymously with "ApoAl."
Unless the context clearly indicates otherwise, reference
herein "RNase" (e.g.,
"RNase 1"),
"paraoxonase" (e.g., "PONI"), "platelet-activating factor acetylhydrolase"
("PAF-AH"),
"cholesterol ester
transfer protein" ("CETP"),
or "lecithin-cholesterol
acyltransferase"
("LCAT") is
understood to include naturally occurring polypeptides of any of the foregoing, as well as functional
variants and functional fragments thereof.
The term "allelic variant" is used herein to denote any of two or more alternative
forms of a gene occupying the same chromosomal locus. Allelic variation arises naturally through
mutation, and may result in phenotypic polymorphism within populations. Gene mutations can be
silent (no change in the encoded polypeptide) or may encode polypeptides having altered amino acid
sequence. The term allelic variant is also used herein to denote a protein encoded by an allelic variant
of a gene.
ApoA-i fusion polypeptides of the present disclosure may be referred to herein by
formulae such as, for example, "ApoAi-Li-D," "ApoAi-Li-D-L2-P," "ApoAl-Li-[Fc region],"
"ApoAl-Li-D-L2-RNase," "ApoAl-L1-[Fc region] -L2-RNasel," "ApoAl-Li-D-L2-paraoxonase," or
"ApoAl-LI-[Fc region]-L2-PONi." In each such case, unless the context clearly dictates otherwise, a
term referring to a particular segment of a fusion polypeptide (e.g., "ApoAi," "D" (for dimerizing
domain), "Li" (for a first polypeptide linker), "Fc region," "RNase," "paraoxonase," etc.) is
understood to have the meaning ascribed to such term herein and is inclusive of the various
embodiments as described herein.
The term "effective amount," in the context of treatment of a disease by
administration of a soluble fusion polypeptide or dimeric
protein to a subject as described herein,
refers to an amount of such molecule that is sufficient to inhibit the occurrence or ameliorate one or
more symptoms of the disease. For example, in the specific context of treatment of an autoimmune
disease by administration of a dimeric ApoAi fusion protein to a subject as described herein, the term
"effective amount"
refers to an amount of such molecule that is sufficient to modulate an autoimmune
response in the subject so as to inhibit the occurrence or ameliorate one or more symptoms of the
autoimmune disease. An effective amount of an agent is administered according to the methods of the
present invention in an "effective regime." The term "effective regime" refers to a combination of
amount of the agent being administered and dosage frequency adequate to accomplish treatment or
prevention of the disease.
term "patient" or "subject," in the context of treating a disease or disorder as
described herein, includes mammals such as, for example, humans and other primates. The term also
includes domesticated animals such as, e.g., cows, hogs, sheep, horses, dogs, and cats.
The term "combination therapy" refers to a therapeutic regimen that involves the
provision of at least two distinct therapies to achieve an indicated therapeutic effect. For example, a
combination therapy may involve the administration of two or more chemically distinct active
ingredients, or agents, for example, a soluble ApoAl fusion polypeptide or dimeric protein according
to the present invention and another agent such as, e.g., another anti-inflammatory or
immunomodulatory agent. Alternatively, a combination therapy may involve the administration of a
soluble ApoAl fusion polypeptide or dimeric protein according to the present invention, alone or in
conjunction with another agent, as well as the delivery of another therapy (e.g., radiation therapy).
The distinct therapies constituting a combination therapy may
be delivered, e.g., as simultaneous,
overlapping, or sequential dosing regimens. In the context of the administration of two or more
chemically distinct agents, it is understood that the active ingredients may be administered as part of
the same composition or as different compositions. When administered as separate compositions, the
compositions comprising the different active ingredients may be administered at the same or different
times, by the same or different routes, using the same or different dosing regimens, all as the
particular context requires and as determined by the attending physician.
The term "non-ApoAl-mediated immunomodulatory therapy," in the context of
treating cancer, means an immunomodulatory therapy that does not specifically target ApoA-1 or
ApoAmediated signaling pathways.
The term "targeted therapy," in the context of treating cancer, refers to a type of
treatment that uses a therapeutic agent to identify and attack a specific type of cancer cell, typically
with less harm to normal cells. In some embodiments, a targeted therapy blocks the action of an
enzyme or other molecule involved in the growth and spread of cancer cells. In other embodiments, a
targeted therapy either helps the immune system to attack cancer cells or delivers a toxic substance
directly to cancer cells. In certain variations, a targeted therapy uses a small molecule drug or a
monoclonal antibody as a therapeutic agent.
Two amino acid sequences have "100% amino acid sequence identity" if the amino
acid residues of the two amino acid sequences are the same when aligned for maximal
correspondence. Sequence comparisons can be performed using standard software programs such as
those included in the LASERGENE bioinformatics computing suite, which is produced by
DNASTAR (Madison, Wisconsin). Other methods for comparing amino acid sequences by
determining optimal alignment are well-known to those of skill in the art. (See, e.g., Peruski and
Peruski, The Internet and the New Biology: Tools for Genomic and Molecular
Research (ASM Press,
et al. (eds.), "Information Superhighway and Computer Databases of Nucleic Acids
Inc. 1997); Wu
and Proteins,"
in Methods in Gene Biotechnology 123-151 (CRC Press, Inc. 1997);
Bishop (ed.),
Guide to Human Genome Computing (2nd ed., Academic Press, Inc. 1998).) Two amino acid
sequences are considered to have "substantial sequence identity" if the two sequences have at least
80%, at least 90%, or at least 95% sequence identity relative to each other.
Percent sequence identity is determined by conventional methods. See, e.g., Altschul
et al., Bull. Math. Bio. 48:603, 1986, and Henikoff and Henikoff, Proc. Natl. Acad. Sci. USA
89:10915, 1992. For example, two amino acid sequences can be aligned to optimize the alignment
scores using a gap opening penalty of 10, a gap extension penalty of 1, and the "BLOSUM62" scoring
matrix of Henikoff and Henikoff, supra, as shown in Table 1 (amino acids are indicated by the
standard one-letter codes). The percent identity is then calculated as: ([Total number of identical
matches]/ [length of the longer sequence plus the number of gaps introduced into the longer sequence
in order to align the two sequences])(100).
Table 1: BLOSUM62 Scoring Matrix
A R N D C Q E G H I L K M F P S T W Y V
R -1 5
N -2 0 6
D -2 -2 1 6
C 0 -3 -3 -3 9
Q -1 1 0 0 -3 5
E -1 0 0 2 -4 2 5
G 0 -2 0 -1 -3 -2 -2 6
H -2 0 1 -1 -3 0 0 -2 8
-3 -3 -4 -3 4
I -1 -3 -3 -3 -1
L -1 -2 -3 -4 -1 -2 -3 -4 -3 2 4
K -1 2 0 -1 -3 1 1 -2 -1 -3 -2 5
M -1 -1 -2 -3 -1
0 -2 -3 -2 1 2 -1 5
F -2 -3 -3 -3 -2 -3 -3 -3 -1 0 0 -3 0 6
-2 -2 -3 -3 -1 -2 -4 7
P -1 -2 -2 -1 -3 -1 -1
S 1 -1 1 0 -1 0 0 0 -1 -2 -2 0 -1 -2 -1 4
T 0 -1 0 -1 -1 -1 -1 -2 -2 -1 -1 -1 -1 -2 -1 1 5
W -3 -3 -4 -4 -2 -2 -3 -2 -2 -3 -2 -3 -1 1 -4 -3 -2 11
Y -2 -2 -2 -3 -2 -1 -2 -3 2 -1 -1 -2 -1 3 -3 -2 -2 2 7
-1 -2 -2 -3 -3 3 1 -2 1 -1 -2 -2 0 -3 -1 4
V 0 -3 -3 -3
Those skilled in the art appreciate that there are many established algorithms
available to align two amino acid sequences. The "FASTA" similarity search algorithm of Pearson
and Lipman is a suitable protein alignment method for examining the level of identity shared by an
amino acid sequence disclosed herein and a second amino acid sequence. The FASTA algorithm is
described by Pearson and Lipman, Proc. Nat'l Acad. Sci. USA 85:2444, 1988, and by Pearson, Meth.
Enzymol. 183:63, 1990. Briefly, FASTA first characterizes sequence similarity by identifying regions
shared by the query sequence (e.g., residues 19-267 or 25-267 of SEQ ID NO:2) and a test sequence
that have either the highest density of identities (if the ktup variable is 1) or pairs of identities (if
ktup=2), without considering conservative amino acid substitutions, insertions, or deletions. The ten
regions with the highest density of identities are then rescored by comparing the similarity of all
paired amino acids using an amino acid substitution matrix, and the ends of the regions are "trimmed"
to include only those residues that contribute to the highest score. If there are several regions with
scores greater than the "cutoff" value (calculated by a predetermined formula based upon the length of
the sequence and the ktup value), then the trimmed initial regions are examined to determine whether
the regions can be joined to form an approximate alignment with gaps. Finally, the highest scoring
two amino acid sequences are aligned using a modification of the Needleman-Wunsch
regions of the
Sellers algorithm (Needleman and Wunsch, J. Mol. Biol. 48:444, 1970; Sellers, SIAM J. Apple. Math.
26:787, 1974), which allows for amino acid insertions and deletions. Illustrative parameters for
FASTA analysis are: ktup=1, gap opening penalty=10, gap extension penalty=1, and substitution
matrix=BLOSUM62. These parameters can be introduced into a FASTA program by modifying the
scoring matrix file ("SMATRIX"), as explained
in Appendix 2 of Pearson, Meth. Enzymol. 183:63,
1990.
When such a value is expressed as "about" X or "approximately" X, the stated value
of X will be understood to be accurate to ±10%.
BRIEF DESCRIPTION OF THE DRAWINGS
illustrates cholesterol efflux in BHK cell cultures ApoA-1 molecules and
recombinant fusions thereof. ApoAFc fusion protein containing a 26 amino acid linker between
ApoA-1 and the Fc region (ApoA-1(26)Fc) demonstrated increased cholesterol efflux as compared to
either an ApoAFc fusion protein with a two amino acid linker (ApoA-1(2)Fc (Theripion)) or an
ApoAFc fusion protein without a linker (ApoA-1(0)Fc (Sino Biol)) and had activity similar to
wild-type human ApoA-1 (Control ApoA-1). ApoA-1 molecules were incubated for 2 hours with H3
cholesterol labeled BHK cells induced for ABCA1 expression. The Fc proteins were predicted to be
dimers; however, the concentrations shown were calculated and normalized based on the mass of
ApoA-1 per molecule.
FIGS. 2A and 2B show schematic diagrams of certain embodiments of fusion
proteins in accordance with the present disclosure, including component functional domains.
depicts a schematic representation of a human ApoA-1 joined at the carboxyl terminus, via a linker, to
a human IgG Fc region (also referred to herein as a "THER fusion protein" or "THER molecule").
depicts a schematic representation of a THER fusion protein further joined at the carboxyl
terminus, via a linker, to an enzyme region (these fusions are also referred to herein a "Bifunctional
Enzyme Lipid Transport" or "BELT" molecule; a BELT molecule may also be generally referred to
herein as a THER fusion protein or molecule). The linker sequence and the domain present at the
carboxyl terminus of the fusion protein varies depending on the construct.
shows a Western blot of culture supernatants (serum free) from transiently
transfected 293T cells expressing five different THER molecules. Transfections and Western blot
analysis were performed as described in Example 3, infra. From left to right: MOCK - mock
transfection negative control; CD40IgG - CD40IgG DNA transfection positive control; THERO
ApoAl-IgG fusion protein with a linker of two amino acids; THER2 - ApoAl-IgG fusion protein
of 16 amino acids; THER4 - ApoAl-IgG fusion protein with a linker of 26 amino acids;
with a linker
THER6 - ApoAl-IgG fusion protein with a linker of 36 amino acids; THER4RNA - ApoAl-IgG
fusion protein with a linker of 36 amino acids, further linked via a second 18 amino acid linker to
human RNasel.
FIGS. 4A-4E show columnar graphs summarizing the initial screening of stable CHO
clones expressing THERO (), THER2 (), THER4 (), THER6 (), and
THER4RNA2 () ApoA-1 fusion proteins and relative expression levels of the fusion proteins
from 96 well culture supernatants (see Example 4, infra).
FIGS. 5A-5C show results from analysis of a subset of THER clones that expressed
higher levels of fusion protein, assessing their cell growth pattern (), relative cell viability
(), and expression of fusion protein () after six and ten days of culture (see Example
4, infra).
FIGS. 6A and 6B show nonreducing () and reducing () SDS-PAGE
analysis of THER fusion proteins purified from CHO clone spent culture supernatants (see Example
4, infra).
shows Native PAGE gel analysis of the purified THER fusion proteins.
Samples were prepared and BLUE Native PAGE gels were run and stained as described in Example
4, infra.
shows a graph summarizing the relative binding of the different fusion
proteins in a sandwich ELISA, using an anti-IgG capture of fusion proteins and detection step with an
HRP-conjugated anti-ApoA-1 antibody (see Example 5, infra).
shows results from a kinetic enzyme assay measuring the RNase activity
present in samples of serial dilutions of purified ApoAl-IgG-RNase bispecific fusion protein
(THER4RNA2). An RNASEALERTTM assay (IDT, Coralville, IA) was performed as described in
Example 6, infra, using RNase A ("RNase") as a positive control and ApoA1nk26-hIgG
("THER4") as a negative control. Each box displays the relative fluorescence units observed as a
function of time during the course of a 45 minute assay, with a fixed concentration of a non
fluorescent RNA substrate that generates a fluorescent signal upon digestion of the RNA.
shows a subset of the data shown in comparing the RNase enzyme
activity at the 4 pmol/pl protein dilution.
shows the results of a BODIPY-cholesterol efflux assay using purified fusion
proteins and differentiated human monocytic cell line, THP-1. Assays were performed in a 96 well
plate format as described in Example 7, infra, and data are displayed as the mean efflux observed
from 5 replicates, with baseline efflux (media alone) subtracted from all samples.
12 shows the results of a cholesterol efflux assay using the mouse monocyte
FIG.
macrophage cell line J774 A.1 (ATCC, Manassas, VA). Both baseline and cAMP-stimulated efflux
were assessed as described in Example 7, infra.
DESCRIPTION OF THE INVENTION
I. Overview
The present invention provides compositions and methods relating to fusion
polypeptides comprising a first polypeptide segment having cholesterol efflux activity and which is
either an ApoAl polypeptide or functional variant or fragment thereof or, alternatively, an ApoA-1
mimetic. In some aspects, the fusion polypeptide further includes a dimerizing domain with a peptide
linker between the amino-terminal end of the dimerizing domain and the carboxyl-terminal end of the
ApoA-1 polypeptide, variant, fragment, or mimetic, thereby allowing the fusion polypeptide to form
stable dimers. In other, non-mutually exclusive aspects, the fusion polypeptides are bispecific
constructs further comprising a second polypeptide segment carboxyl-terminal to the ApoA-1
polypeptide, variant, fragment, or mimetic and which confers a second biological activity. Exemplary
second polypeptides include RNases, paraoxonases, platelet-activating factor acetylhydrolases (PAF
AHs), cholesterol ester transfer proteins (CETPs), lecithin-cholesterol acyltransferases (LCATs), and
polypeptides that specifically bind to amyloid beta, any of which may be a naturally occurring protein
or a functional variant or fragment thereof.
The fusion molecules of the present invention can be used, for example, to increase
reverse cholesterol transport in a subject and provide therapeutic benefit in the treatment of various
diseases. ApoA-1, the major protein of HDL, has already shown beneficial activity in clinical trials in
patients with acute coronary syndrome. The ApoA- 1 fusion molecules of the present invention can be
used to treat coronary heart disease, acute coronary syndrome, and other cardiovascular diseases
characterized by atherosclerosis such as, e.g., stroke. Fusion molecules of the present invention are
also useful, for example,
for the treatment of autoimmune diseases (e.g., rheumatoid arthritis,
systemic lupus erythematosus), inflammatory diseases, type 2 diabetes, obesity, and
neurodegenerative diseases (e.g., Alzheimer's disease). In some embodiments, fusion proteins of the
present invention are used to replace defective ApoA-1 such as, for example, in the treatment of type
1 diabetes and dementia. In certain variations, fusion proteins as disclosed herein are used to treat
multiple sclerosis (MS). ApoA-1 levels have been shown to be low in patients with MS, and ApoA-1
deficient mice have been shown to exhibit more neurodegeneration and worse disease in experimental
allergic encephalomyelitis (EAE), a model for MS, than wild-type animals. See Meyers et al., J.
Neuroimmunol. 277: 176-185, 2014. Data further suggests a positive neuroprotective effect of ApoA-
1 on the central nervous system. See Gardner et al., Frontiers in Pharmacology: 20 November 2015
doi: 10.3389/fphar.2015.00278.
Several studies support the use of ApoA-1 therapy for autoimmune disease. For
example, patients with systemic lupus erythematosus (SLE) have low HDL-cholesterol levels and the
HDL that is present is often damaged by myeloperoxidase-mediated methionine oxidation and
tyrosine chlorination of ApoA-1, resulting in loss of ABCA1-dependent cholesterol efflux activity.
See Shao et al., J. Biol. Chem. 281:9001-4, 2006; Hewing et al., Arterioscler. Thromb. Vasc. Biol.
34:779-89, 2014. This promotes loss of anti-inflammatory properties and generation of pro
inflammatory HDL seen in patients with SLE. See Skaggs et al., Clin. Immunol. 137:147-156, 2010;
McMahon et al., Athritis Rheum. 60:2428-2437, 2009. Autoantibodies to ApoA-1 are present in
many patients with SLE, and SLE-disease activity assessed by SLEDAI and SLE disease related
organ damage assessed by SLICC/ACR damage index are positively correlated with anti-ApoA-1
antibodies. See Batukla et al., Ann. NYAcad. Sci. 1108:137-146, 2007; Ahmed et al., EXCLI Journal
12:719-732, 2013. Further, increased ApoA-1 concentration attenuated autoimmunity and
glomerulonephritis in lupus
prone SLE 1,2,3 mice. See Black et al., J. Immunol. 195:4685-4698,
2015.
Cholesterol efflux capacity of HDL is also impaired in rheumatoid arthritis patients
with high disease activity and is correlated with systemic inflammation and loss of HDL antioxidant
activity. See Charles-Schoeman et al., Arthritis Rheum. 60:2870-2879, 2009; Charles-Schoeman et
al., Ann. Rheum. Dis. 71:1157-1162, 2012. Treatment of arthritis in the Lewis rat by ApoA-1 and
reconstituted HDL reduced acute and chronic joint inflammation, and decreased macrophage TLR2
expression and activation. See Wu et al., Arterioscler. Thromb. Basc. Biol. 34:543-551, 2014.
Therapy of collagen-induced arthritis in rats with ApoA-1 mimetic peptide D-4F in combination with
pravastatin significantly reduced disease activity. See Charles-Schoeman et al., Clin. Immunol.
127:234-244,
2008.
Fusion molecules of the present invention may also be used in the treatment of
infectious disease. During infection and endotoxemia, significant alterations in lipid metabolism and
lipoprotein composition occur, including a reduction in ApoA-1 and changes in HDL composition and
size. HDL can bind and neutralize Gram-negative
LPS and Gram-positive lipoteichoic acid,
promoting clearance of these inflammatory products. Pharmacological studies support the benefit of
recombinant ApoA-1 during bacterial infection. See, e.g., Pirillo et al., Handb Exp Pharmacol.
224:483-508, 2015.
Bifunctional ApoA-1 fusion molecules of the present invention that contain a
paraoxonase (e.g., PONI) are particularly useful for therapy of patients infected with Pseudomonas
aeruginosa, a gram negative bacterium. This is particularly important for immunocompromised
infections with P. aeruginosa are common. P. aeruginosa secrete virulence factors
patients, where
and form biofilm in response to small signaling molecules called acyl-homoserine lactones in a
concentration-dependent process called quorum sensing (QS). Paroxonase 1 degrades acyl
homoserine lactones and was shown to protect from lethality from P. aeruginosa in a transgenic in
vivo model in Drosophila melanogaster where there are no endogenous PON homologs. See Estin et
al., Adv. Exp. Med. Biol. 660:183-193, 2010.
Fusion molecules of the present invention may also be used in the treatment of
inflammatory disease. For example, ApoA-1 fusion polypeptides and dimeric proteins as described
herein may alter the phenotype of neutrophils, macrophages, and/or antigen-presenting cells to reduce
proinflammatory responses. Molecules of the present invention cause efflux of cholesterol from cell
membranes, mediated by transporter molecules such as, e.g., ABCA1. Efflux of cholesterol from
antigen-presenting cells, including macrophages and dendritic cells, can inhibit proinflammatory
responses mediated by these cells, resulting in reduced production of inflammatory cytokines. Studies
support the benefit of ApoA-1 in mediating anti-inflammatory effects. For example, treatment with
ApoA-1 was shown to inhibit the proinflammatory signaling in macrophages after stimulation of
CD40 by altering the composition of lipid rafts. See Yin et al., J. Atherosclerosis and Thrombosis
19:923-36, 2012. ApoA-1
was also shown to cause a decrease in TRAF-6 recruitment to lipid
rafts,
and a decrease in activation of NF-kB. See id. Another study showed that treatment of human
monocytes and macrophages with ApoA-1 or ApoA-1 mimetic
4F altered their response to LPS,
resulting in decreased production of inflammatory cytokines MCP-1, MIP-1, RANTES, IL-6, and
TNFa, but increased the production of IL-10. See Smythies et al., Am. J. Physiol. Cell Physiol.
298:C1538-48, 2010. doi:1152/ajpcell.00467.2009. Another study showed that treatment with ApoA
1 significantly decreased LPS-induced MCP-1 release from THP-1 cells, and inhibited expression of
CD11b and VCAM-1. See Wang et al., Cytokine 49:194-2000, 2010. Thus ApoA-1 inhibits
activation and adhesion of human monocytes and macrophages, and induces profound functional
changes due to a differentiation to an anti-inflammatory phenotype.
Inflammatory lung diseases are among inflammatory diseases that may be treated
with ApoA-1 fusion molecules as described herein. Serum ApoA-1 was found to be positively
correlated with FEV1 in patients with combined atopy and asthma, but not in atopic and nonatopic
subjects without asthma. See Barochia et al., Am. J. Respir. Crit. Care Med. 191:990-1000, 2015. In
another study, patients with idiopathic pulmonary fibrosis had low levels of ApoA-1 in bronchiolar
lavage fluid compared to controls (P<0.01). See Kim et al., Am. J. Respir. Crit. Care Med. 182:633
642, 2010. Further, intranasal treatment with ApoA-1 in mice treated with bleomycin was very
effective in reducing the number of inflammatory cells and collagen deposition in the lungs. See id.
Obesity is another inflammatory disease amenable to treatment with ApoA-1 fusion
molecules in accordance with the present invention. Evidence supports the use of ApoA-1 and HDL
to combat obesity.
See, e.g., Mineo et al., Circ. Res. 111:1079-1090, 2012.
For example,
overexpression of ApoA-1 or administration of the ApoA-1 mimetic peptide D-4F has been shown to
decrease white adipose mass and insulin resistance and increase energy expenditure in mice fed a
high-fat diet. Further, in ob/ob mice, the ApoA-1 mimetic L-4F was shown to lower adiposity and
inflammation and improve glucose tolerance. Id.
Yet another disorder that may be treated with ApoA-1 fusion molecules in accordance
with the present invention is nephrotic syndrome (NS), which is associated with a higher risk for
cardiovascular disease in patients. Urinary wastage of filterable HDL (i.e., HDL3) and lipid-poor apo
Al is a common feature of patients with nephrotic syndrome. This is typically due to decreased re
uptake of these molecules via cubulin/megalin receptors in the renal
proximal tubule. See Barth et al.,
Trends Cardiovasc. Med. 11:26-31, 2001. ApoA-1 fusion molecules comprising an Fc region as
described herein would bypass the need for reuptake in this usual manner, since the molecules are
being recycled via FcRn due to the presence of the Fc domain.
Fusion molecules as described herein may also be used for therapy of patients with
cancer. It is expected that ApoA-1 fusion polypeptides and
dimeric proteins of the present invention,
while reducing proinflammatory responses, enhance activation and tumor infiltration of CD8' T-cells.
Studies support the efficacy of ApoA-1 therapy in animal models of cancer and have shown that
ApoA-1 therapy can cause a specific increase in CD8' T cells in tumors. See, e.g., Zamanian
Daryoush et al., J. Biol. Chem. 288:21237-21252, 2013. In some aspects, ApoA-1 fusion molecules
of the present invention are useful in combination with one or more other anti-cancer therapies such
as, for example, an anti-cancer immunotherapy.
In certain aspects, the present invention provides a way to stabilize an active ApoA-1
dimer while also controlling the maturation from pre-beta particles to discoid particles and spherical
particles by providing a flexible linker between a dimerizing domain (e.g., an Fc domain) and the C
terminus of the ApoA-1 polypeptide, or functional variant, fragment, or mimetic thereof. A previous
ApoAIg molecule not containing a linker exhibits low activity in cholesterol efflux assays
compared to wild-type ApoAl. In contrast, dimerizing fusion polypeptides of the present invention
retain
ApoA-1 activity in cholesterol efflux assays and also allow for further
improvements such as,
e.g., fusion of an RNase (e.g., RNase 1) or other polypeptide segments C-terminal to the dimerizing
domain. In certain preferred embodiments, the use of an Fc region as the dimerizing domain also
allows for increased half-life of the dimer.
While not intending to be bound by theory, it is believed that the length of the linker
controls the ability of the stable ApoA-1 dimer to expand as it takes up cholesterol. The invention
fusion molecules containing flexible linkers between the C-terminus of an ApoA-1
provides ApoA-1
polypeptide, or variant, fragment, or mimetic thereof, and the N-terminus of a dimerizing domain
such as, e.g., an Fc domain. Linkers are of sufficient length to allow ApoA-1, or the functional
variant, fragment, or mimetic thereof, to mediate cholesterol efflux from cells, an initial and critical
step in Reverse Cholesterol Transport (RCT). Linkers are typically between 2 and 60 amino acids in
length. It is believed that ApoA-1 fusion molecules with alternative linker lengths have distinct
functional properties by controlling the maturation of the HDL particle by constraining the C-terminus
of ApoA-1. HDL discoid particles of intermediate size may have improved atheroprotective
properties, and may have improved CNS transport properties. The molecules of this invention may
change the progress of HDL maturation at these intermediate discoid stages, thereby improving
efficacy of the fusion proteins of the invention relative to wild type ApoA-1 proteins. The molecules
of this invention are likely to affect the structure and composition of spherical HDL particles which
are composed of trimeric ApoA-1 particles (see
Silva et al., Natl. Acad. Sci. USA 105:12176-12181,
2008). It is likely that molecules of this invention will interact with natural ApoA-1 in the formation
of larger spherical HDL particles.
In certain embodiments, the dimerizing domain is a immunoglobulin Fc region.
ApoAFc fusion molecules of the present invention extend ApoA- 1 half-life while retaining ApoA
1 reverse cholesterol efflux and eliminating the requirement for extensive lipid formulation. In
addition, the presence of the Fc region allows purification using immobilized Protein A according to
standard practices in and antibody and Fc fusion protein manufacturing.
Structural studies of ApoA-1 (see, e.g., Gogonea, Frontiers Pharmacol. 6:318, 2016)
show that ApoA-1 assumes multiple conformations as it matures from lipid-free monomer to higher
order forms. Recent data derived from small angle neutron scattering (SANS) show low resolution
structures of ApoA-1 dimers in an open configuration around a lipid core, called the super double
helix (DSH) model. Other structures from SANS studies show ApoA-1 in different open
configurations depending on the composition of the lipid core; in these structures, the C-terminus of
the ApoA-1 monomers are in different positions relative to each other. Similarly, spherical ApoA-1
particles that incorporate a third ApoA-1 monomer show the C-terminus of each monomer in a
different position compared to the positions in dimeric discoid ApoA-1. See, e.g., Gogonea, supra.
The flexible linkers of the present disclosure are of sufficient length to allow ApoA- 1 to assume these
positions without conformational constraint.
In certain embodiments, ApoA- 1-[linker] - [dimerizing domain] molecules of the
present invention include an additional polypeptide segment fused carboxyl-terminal to the dimerizing
domain. Such variations allow for the creation of bispecific molecules with ApoA-1 functional
activity and a second biological activity.
aspects of the present invention, bispecific fusion molecules are provided
In some
comprising a (i) first polypeptide segment with reverse cholesterol transport activity and which is
either an ApoAl polypeptide or functional variant or fragment thereof or, alternatively, an ApoAl
mimetic and (ii) a second polypeptide
segment carboxyl-terminal to the first polypeptide segment,
wherein the second polypeptide segment is selected from an RNase, a paraoxonase, a platelet
activating factor acetylhydrolase (PAF-AH), a cholesterol ester transfer protein (CETP), a lecithin
cholesterol acyltransferase (LCAT), and a polypeptide that specifically binds to amyloid beta. Such
second polypeptides may be a naturally occurring protein or a functional variant or fragment thereof.
In some embodiments, a linker and dimerizing domain is included between the first and second
polypeptides as summarized above. In alternative embodiments, the fusion polypeptide lacks a
dimerizing domain.
In some embodiments of the present ApoA-1 fusion
molecules that lack an Fc region,
the fusion molecule may be conjugated to PEG to provide extended half-life. Such variations may
include bispecific molecules as described herein, such as, e.g., fusion molecules comprising an
RNase, a paraoxonase, a platelet-activating factor acetylhydrolase (PAF-AH), a cholesterol ester
transfer protein (CETP), a lecithin-cholesterol acyltransferase (LCAT), or a polypeptide that
specifically binds to amyloid beta.
In some embodiments of a bispecific molecule as summarized above, the second
polypeptide segment is an RNase. A preferred RNase is human RNase 1 or a functional variant or
fragment thereof. In particular variations, the RNase retains its sensitivity to inhibition by
cytoplasmic inhibitor and has very low toxicity to cells, but is highly active extracellularly. RNase
has anti-inflammatory properties by digestion of inflammatory extracellular RNA and provides
additional therapeutic benefit for treatment of various diseases, including cardiovascular diseases
(e.g., coronary artery disease, stroke), autoimmune diseases,
inflammatory diseases, type 2 diabetes,
infectious disease, and neurodegenerative diseases (e.g., Alzheimer' disease).
For example, a bispecific ApoA-1 fusion molecule comprising an RNase segment as
described herein may be used, e.g., for treatment of an inflammatory disease such as, for example, an
inflammatory lung disease. One study has shown that TLR3, an RNA sensor, has a major role in the
development of ARDS-like pathology in the absence of a viral pathogen. See Murray et al., Am. J.
Respir. Crit. Care Med. 178:1227-1237, 2008. Oxygen therapy is a major therapeutic intervention in
ARDS, but contributes to further lung damage and susceptibility to viral infection. Oxygen therapy
was a major stimulus for increased TLR3 expression and activation in cultured human epithelial
cells, and absence or blockade of TLR3 protected mice from lung injury and inflammation after
exposure to hyperoxic conditions. See Murray et al., supra. Another study has shown that TLR3
activation by extracellular RNA occurs in response to acute hypoxia, and that therapy in mice with
lung inflammation after acute hypoxia. See Biswas et al., Eur. J. Immunol. 45:
RNaseA diminished
3158-3173, 2015. A bispecific ApoA-1 fusion molecule comprising an RNase segment as described
herein may also be used, e.g., for treatment of an autoimmune disease such as, for example, systemic
lupus erythematosus (SLE). Studies show, for example, a role of RNA immune complexes and RNA
receptors, including TLR7, in SLE disease pathogenesis, as well as a protective effect of RNase
overexpression in mouse models of SLE. See, e.g., Sun et al., J. Immunol. 190:2536-2543, 2013.
In other embodiments of a bispecific molecule as summarized above, the second
polypeptide segment is a paraoxonase. A preferred paraoxonase is human paraoxonase 1 (PON1) or a
functional variant or fragment thereof. Paraoxonase bispecific fusion molecules provide additional
therapeutic benefit for the treatment of diseases amenable to ApoAmediated therapy, including, for
example, through its atheroprotective, antioxidant, anti-inflammatory, and/or neuroprotective
properties. In some alternative embodiments, PON1 may attached to an ApoA-1 fusion molecule of
the present invention through its natural, high affinity binding to ApoA-1, which binding is mediated
by Tyr71 of PONi (see Huang et al., J. Clin. Invest. 123:3815-3828, 2013). Incubating an ApoA-1
fusion molecule with recombinant or natural PONi prior to administration will be sufficient to "load"
PON1 onto the ApoA-1 fusion molecule.
A bispecific ApoA-1 fusion molecule comprising a paraoxonase segment as described
herein may be used, e.g., for treatment of an autoimmune disease or an inflammatory disease. For
example, studies support use of a paraoxonase for treatment of autoimmune disease such as systemic
lupus erythematosus (SLE). The autoantibody titer in many patients with systemic lupus
erythematosus (SLE) is correlated with loss of activity of PON1 (see Batukla et al., Ann. NY Acad.
Sci. 1108:137-146, 2007), and SLE-disease activity assessed by SLEDAI and SLE disease related
organ damage assessed by SLICC/ACR damage index are negatively correlated with PONi activity
(see Ahmed et al., EXCLI Journal 12:719-732, 2013). PONi activity is significantly reduced in
patients with SLE, and is a risk factor for atherosclerosis. See Kiss et al., Ann. NYAcad. Sci. 108:83
91, 2007. In addition, other studies support use of a paraoxonase for treatment of inflammatory
disease such as inflammatory lung diseases. One study showed that patients with late lung diseases
long after exposure to sulfur mustard gas (SM), including asthma, chronic obstructive pulmonary
disease (COPD) and bronchiectasis, have significantly reduced levels of PONt in bronchiolar lavage
fluid (p<0.0001). See Golmanesh et al., Immunopharmacol. Immunotoxical. 35:419-425, 2013.
Another study showed that Iranian veterans exposed to SM twenty years ago still have significantly
low serum levels of PONt activity, and low PONt was correlated with lung disease severity. See
Taravati et al., Immunopharmacol. Immunotoxicol. 34:706-713, 2012.
Bispecific ApoA-1 fusion molecules comprising either an RNase segment or a
paraoxonase segment as described herein may also be used, e.g., for treatment of a neurological
bispecific molecules are transported to the brain where they deliver a protective
disease. Such
paraoxonase or RNase enzyme. For example, PON1 is protective in the brain because of its anti
oxidant properties, and RNase is protective by digesting extracellular RNA that promotes
inflammation via stimulation of TLR7 and other RNA receptors. Exemplary neurological diseases
amenable to treatment using an ApoA-1/paraoxonase or ApoAl/RNase bispecific molecule of the
present invention include multiple sclerosis, Parkinson's disease, and Alzheimer's disease.
Attachment of myeloperoxidase (MPO) inhibitors to ApoA-1 fusion molecules of the
present invention may be particularly desirable as a way to protect ApoA-1 from inactivation due to
oxidation mediated by MPO, and can also similarly protect paraoxonase from MPO-mediated
oxidation and inactivation in the context of a bispecific fusion polypeptide comprising a paraoxonase
such as PON1. Myeloperoxidase-mediated oxidation
of ApoA-1 promotes crosslinking of ApoA-1,
and may be implicated in the mechanism that leads to amyloid deposition in atherosclerotic plaques in
vivo. See Chan et al., J. Biol. Chem. 290: 10958-71, 2015. For a review of MPO inhibitors, see Malle
et al., Br J Pharmacol. 152: 838-854, 2007. The attachment of a MPO inhibitor to a molecule of the
present invention can also localize the MPO inhibition to selectively protect ApoA-1 from oxidation
while preserving MPO activity important in anti-microbial activity.
In other embodiments of a bispecific molecule as summarized above, the second
polypeptide segment is selected from a cholesterol ester transfer protein (CETP), and a lecithin
cholesterol acyltransferase (LCAT). CETP is involved in one of the major mechanisms by which
HDL particles can deliver cholesterol to the liver during the process of reverse cholesterol transport
(RCT), specifically, through unloading and transferring of cholesterol to LDL, which then transports
cholesterol back to the liver via LDL receptors. This process of unloading requires CETP. By
improving the initial part of the RCT pathway through the delivery of improved ApoA-1 molecules
such as provided herein, then adding other RCT components can provide an attractive and potentially
synergistic therapeutic approach. Providing more exogenous CETP in the form of a bispecific fusion
molecule containing ApoA- 1 can enhance CETP activity and overall reverse cholesterol transport.
Bispecific fusions containing LCAT can provide an alternative means of enhancing
endogenous CETP. Lecithin-cholesterol acyltransferase (LCAT) is an enzyme that is associated with
HDL and converts free cholesterol to cholesteryl esters, which is then sequestered into the HDL
particle and allows for its spherical shape formation. A human recombinant LCAT given to mice
lacking LCAT significantly improved HDL-C levels, and when given to human ApoA-1 transgenic
mice, the increase in HDL-C was eight-fold, suggesting synergy. See Rousset et al., J Pharmacol Exp
There. 335:140-8, 2010.
A recombinant human LCAT fusion to Fc has been reported (see
Spahr et al.,
Protein Sci. 22:1739-53, 2013), and a bispecific molecule containing both ApoA-1 and LCAT may
also improve RCT more efficiently than a mono-specific protein of either alone.
embodiments of a bispecific molecule as summarized above, the second
In other
polypeptide segment is a polypeptide that specifically binds to amyloid beta (AP). In a specific
variation, the second polypeptide is a An-specific single chain antibody such as, for example, an Ap
specific
scFv. A scFv specific for amyloid beta peptide is described, for example,
by Cattepoel et al.,
PLoS One 6:e18296, 2011. In such embodiments, the An-binding polypeptide is typically fused C
terminal to ApoA-1, or C-terminal to the dimerizing domain, if present. This bispecific fusion
molecule has improved properties for therapy of patients with Alzheimer's disease.
II. Fusion Polypeptides and Dimeric Proteins
Accordingly, in one aspect, the present invention provides a fusion polypeptide
comprising, from an amino-terminal position to a carboxyl-terminal position, ApoAl-L1-D, where
ApoAl is a first polypeptide segment having cholesterol efflux activity and which is selected from (i)
a naturally occurring ApoA-1 polypeptide or a functional variant or fragment thereof and (ii) an
ApoA-1 mimetic; LI is a first polypeptide linker; and D is a dimerizing domain. In some
embodiments, the fusion polypeptide further includes a second polypeptide segment located carboxyl
terminal to the dimerizing domain. In particular variations, the second polypeptide segment is (a) a
naturally occurring RNase, paraoxonase, platelet-activating
factor acetylhydrolase (PAF-AH),
cholesterol ester transfer protein (CETP), or lecithin-cholesterol acyltransferase (LCAT); (b) a
functional variant or fragment of any of the naturally occurring proteins specified in (a); or (c) a
polypeptide that specifically binds to amyloid beta (AP) such as, e.g., an An-specific scFv. Such a
fusion polypeptide comprising a second polypeptide segment may be represented by the formula
ApoAl-L1-D-L2-P
(from an amino-terminal position to a carboxyl-terminal position), where
ApoAl,
LI, and D are each as previously defined, where L2 is a second polypeptide linker and is optionally
present, and where P is the second polypeptide segment.
In another aspect, the present invention provides a fusion polypeptide comprising a
first polypeptide segment having cholesterol efflux activity and which is selected from (i) a naturally
occurring ApoA-1 polypeptide or a functional variant or fragment thereof and (ii) an ApoA-1
mimetic, and a second polypeptide segment located carboxyl-terminal to the first polypeptide
segment, where the second polypeptide
segment is (a) a naturally occurring RNase, paraoxonase,
platelet-activating factor acetylhydrolase (PAF-AH), cholesterol ester transfer protein (CETP), or
lecithin-cholesterol acyltransferase (LCAT); (b) a functional variant or fragment of any of the
naturally occurring proteins specified in (a); or (c) a polypeptide that specifically binds to amyloid
beta (AP) such as, e.g., an An-specific scFv. In some variations, the fusion polypeptide further
includes a linker polypeptide located carboxyl-terminal to the first polypeptide segment and amino-
second polypeptide segment. In some embodiments, the fusion polypeptide further
terminal to the
includes a dimerizing domain, which be located, for example, carboxyl-terminal to the first
polypeptide segment and amino-terminal to the second polypeptide segment.
Functional variants of a particular naturally occurring protein specified above can be
readily identified using routine assays for assessing the variant for a relevant biological or
biochemical
activity corresponding to the natural protein. For example, in the
case of ApoA-1,
variants may be assayed for their ability to induce cholesterol efflux using known cholesterol efflux
assays such as described herein. See, e.g., Tang et al., J Lipid Res. 47:107-14, 2006. In the case of
RNase such as human RNase 1, variants may be assayed for their ability to digest single or double
stranded RNA is known assays to assess ribonuclease
activity. See, e.g., Libonati and Sorrentino,
Methods Enzymol. 341234-248, 2001. Paraoxonase 1 (PON1) variants may be assayed for
phosphotriesterase activity using diethyl p-nitrophenol phosphate (paraoxon) as a substrate, or for
arylesterase activity using phenyl acetate as a substrate. See, e.g., Graves and Scott, Curr Chem
Genomics 2:51-61, 2008. Assays to assess relevant CETP and LCAT activities are also known. For
example, assays for measuring LCAT and CETP enzyme activity are commercially available and
include, e.g., Cell Biolabs Cat. No. STA-615, Sigma-Aldrich Cat. No. MAK107, and Roar
Biomedical Cat. No. RB-LCAT for LCAT, and Abcam Cat. No. ab65383 and Sigma-Aldrich Cat. No.
MAK106 for CETP.
In the case of An-binding activity, polypeptides such as, e.g., single chain antibodies
may be assessed for binding activity using any of various known assays. For example, one assay
system employs a commercially
available biosensor instrument (BIAcore , Pharmacia Biosensor,
Piscataway, NJ), wherein a binding protein (e.g., An-binding candidate, such as an antibody) is
immobilized onto the surface of a sensor chip, and a test sample containing a soluble antigen (e.g., AP
peptide) is passed through the cell. If the immobilized protein has affinity for the antigen, it will bind
to the antigen, causing a change in the refractive index of the medium, which is detected as a change
in surface plasmon resonance of the gold film. This system allows the determination of on- and off
rates, from which binding affinity can be calculated, and assessment of stoichiometry of binding. Use
of this instrument is disclosed, e.g., by Karlsson (J. Immunol. Methods 145:229-240, 1991) and
Cunningham and Wells (J. Mol. Biol. 234:554-563, 1993). An-binding polypeptides can also be used
within other assay systems known in the art. Such systems include Scatchard analysis for
determination of binding affinity (see Scatchard, Ann. NY Acad. Sci. 51: 660-672, 1949) and
calorimetric assays (see Cunningham et al., Science 253:545-548, 1991; Cunningham et al., Science
254:821-825, 1991).
Naturally occurring polypeptide segments for use in accordance with the present
invention (e.g., a naturally occurring ApoA-1 polypeptide, RNase, paraoxonase, or platelet-activating
includes naturally occurring variants such as, for example, allelic variants and
factor acetylhydrolase)
interspecies homologs consistent with the disclosure.
Functional variants of a particular reference polypeptide (e.g., a wild-type human
ApoA-1) are generally characterized as having one or more amino acid substitutions, deletions or
additions relative to the reference polypeptide. These changes are preferably of a minor nature, that is
conservative amino acid substitutions (see, e.g., Table 2, infra, which lists some exemplary
conservative amino acid substitutions) and other substitutions that do not significantly affect the
folding or activity of the protein or polypeptide; small deletions, typically of one to about 30 amino
acids; and small amino- or carboxyl-terminal extensions, such as an amino-terminal methionine
residue, a small linker peptide, or a small extension that facilitates purification (an affinity tag), such
as a poly-histidine tract, protein A (Nilsson et al., EMBO J. 4:1075, 1985; Nilsson et al., Methods
Enzymol. 198:3, 1991), glutathione S transferase (Smith and Johnson, Gene 67:31, 1988), or other
antigenic epitope or binding domain. (See generally Ford et al., Protein Expression and Purification
2:95-107,
1991.) DNAs encoding affinity tags are available from commercial
suppliers (e.g.,
Pharmacia Biotech, Piscataway, NJ). Conservative substitutions may also be selected from the
following: 1) Alanine, Glycine;
2) Aspartate, Glutamate; 3) Asparagine, Glutamine; 4) Arginine,
Lysine; 5) Isoleucine, Leucine, Methionine, Valine; 6) Phenylalanine, Tyrosine, Tryptophan; 7)
Serine, Threonine; and 8) Cysteine, Methionine (see, e.g., Creighton, Proteins (1984)).
Table 2: Conservative amino acid substitutions
Basic Acidic Polar Hydrophobic Aromatic Small
Arginine Glutamate Glutamine Leucine Phenylalanine Glycine
Lysine Aspartate Asparagine Isoleucine Tryptophan Alanine
Histidine Valine Tyrosine Serine
Methionine Threonine
Methionine
Essential amino acids in a naturally occurring polypeptide can be identified according
to procedures known in the art, such as site-directed mutagenesis or alanine-scanning mutagenesis
(Cunningham and Wells, Science 244:1081-1085, 1989; Bass et al., Proc. Natl. Acad. Sci. USA
88:4498-4502, 1991). In the latter technique, single alanine mutations are introduced at every residue
in the molecule, and the resultant mutant molecules are tested for biological activity (e.g., cholesterol
efflux for ApoA-1 variants) to identify amino acid residues that are critical to the activity of the
molecule. In addition, sites of relevant protein interactions can be determined by analysis of crystal
structure as determined by such techniques as nuclear magnetic resonance, crystallography or
labeling. The identities of essential amino acids can also be inferred from analysis of
photoaffinity
homologies with related proteins (e.g., species orthologs retaining the same protein function).
Multiple amino acid substitutions can be made and tested using known methods of
mutagenesis and screening, such as those disclosed
by Reidhaar-Olson and Sauer Science 241:53-57,
1988 or Bowie and Sauer Proc. Natl. Acad. Sci. USA 86:2152-2156, 1989. Briefly, these authors
disclose methods for simultaneously randomizing two or more positions in a polypeptide, selecting
for functional polypeptide, and then sequencing the mutagenized polypeptides to determine the
spectrum of allowable substitutions at each position. Another method that can be used is region
directed mutagenesis (Derbyshire et al., Gene 46:145, 1986; Ner et al., DNA 7:127, 1988).
Variant nucleotide and polypeptide sequences can also be generated through DNA
shuffling. (See, e.g., Stemmer, Nature 370:389, 1994; Stemmer, Proc. Nat'l Acad. Sci. USA
91:10747, 1994; International Publication No. WO 97/20078.) Briefly, variant DNA molecules are
generated by in vitro homologous recombination by random fragmentation of a parent DNA followed
by reassembly using PCR, resulting in randomly introduced point mutations. This technique can be
modified by using a family of parent DNA molecules, such as allelic variants or DNA molecules from
different species, to introduce additional variability into the process. Selection or screening for the
desired activity, followed by additional iterations of mutagenesis and assay provides for rapid
"evolution"
of sequences by selecting
for desirable mutations
while simultaneously
selecting against
detrimental changes.
As previously discussed, a polypeptide fusion in accordance with the present
invention can include a polypeptide segment corresponding to a "functional fragment" of a particular
polypeptide. Routine deletion analyses of nucleic acid molecules can be performed to obtain
functional
fragments of a nucleic acid molecule encoding a given polypeptide.
As an illustration,
ApoAencoding DNA molecules having the nucleotide sequence of residues 70-816 of SEQ ID
NO:1 can be digested with Bal3l nuclease to obtain a series of nested deletions. The fragments are
then inserted into expression vectors in proper reading frame, and the expressed polypeptides are
isolated and tested for the ability to induce cholesterol efflux. One alternative to exonuclease
digestion is to use oligonucleotide-directed mutagenesis to introduce deletions or stop codons to
specify production of a desired fragment. Alternatively, particular fragments of a gene encoding a
polypeptide can be synthesized using the polymerase chain reaction.
Accordingly, using methods such as discussed above, one of ordinary skill in the art
can prepare a variety of polypeptides that (i) are substantially identical to a reference polypeptide
(e.g., residues 19-267 or 25-267 of SEQ ID NO:2 for a human wild-type ApoA-1 polypeptide) and (ii)
retains the desired functional properties of the reference polypeptide.
segments used within the present invention (e.g., polypeptide segments
Polypeptide
corresponding to ApoA-1, RNase, paraoxonase, platelet-activating factor acetyihydrolase, dimerizing
domains such as, e.g., Fc fragments) may be obtained from a variety of species. If the protein is to be
used therapeutically in humans, it is preferred that human polypeptide sequences be employed.
However, non-human sequences can be used, as can variant sequences. For other uses, including in
vitro diagnostic uses and veterinary uses, polypeptide sequences from humans or non-human animals
can be employed, although sequences from the same species as the patient may be preferred for in
vivo veterinary use or for in vitro uses where species specificity of intermolecular reactions is present.
Thus, polypeptide segments for use within the present invention
can be, without limitation, human,
non-human primate, rodent, canine, feline, equine, bovine, ovine, porcine, lagomorph, and avian
polypeptides, as well as variants thereof.
In certain embodiments, the first polypeptide segment is a human wild-type ApoA-1
polypeptide or a functional variant or fragment thereof. For example, in some embodiments, the first
polypeptide segment comprises an amino acid sequence having at least 80% identity with amino acid
residues 19-267 or 25-267 of SEQ ID NO:2. In more particular embodiments, the first polypeptide
segment comprises an amino acid sequence having at least 85%, at least 90%, or at least 95% identity
with amino acid residues 19-267 or 25-267 of SEQ ID NO:2. In yet other embodiments, the first
polypeptide segment comprises an amino acid sequence having at least 96%, at least 97%, at least
98%, at least 99%, or 100% sequence identity with amino acid residues 19-267 or 25-267 of SEQ ID
NO:2. In specific variations, valine at the amino acid position corresponding to position 156 of
mature human wild-type ApoA-1 is replaced by lysine, and/or arginine at the amino acid position
corresponding to position 173 of mature human wild-type ApoA-1 is replaced by cysteine (also
referred to herein, respectively, as V156K and R173C variants or mutations). Position 156 of the
mature human wild-type ApoA-1 corresponds to amino acid position 180 of SEQ ID NO:2, and
position 173 of mature human wild-type ApoA-1 corresponds to amino acid position 197 of SEQ ID
NO:2. V156K and R173C mutations have improved activity and half-life in atherosclerotic mice
compared to wild-type ApoA-1. See Cho et al., Exp Mol Med 41:417, 2009.
In other embodiments,
the first polypeptide segment is an ApoA-1 mimetic
such as,
for example, the 4F peptide (see Song et al., Int. J. Biol. Sci. 5:637-646, 2009). ApoA-1 mimetics are
generally known in the art and are reviewed in Reddy et al., Curr. Opin. Lipidol. 25: 304-308, 2014.
In certain embodiments comprising a second polypeptide segment carboxyl-terminal
to the first polypeptide segment (e.g., carboxyl-terminal to a dimerizing domain), the second
polypeptide segment is an RNase. In some embodiments, the RNase is a human RNAse 1 or a
functional variant or fragment thereof. For example, in some embodiments, the second polypeptide
segment comprises an amino acid sequence having at least 80% identity with amino acid residues
embodiments, the second polypeptide segment
542-675 of SEQ ID NO:4. In more particular
comprises an amino acid sequence having at least 85%, at least 90%, or at least 95% identity with
amino acid residues 542-675 of SEQ ID NO:4. In yet other embodiments, the second polypeptide
segment comprises an amino acid sequence having at least 96%, at least 97%, at least 98%, at least
99%, or 100% sequence identity with amino acid residues 542-675 of SEQ ID NO:4.
In other embodiments comprising a second polypeptide segment carboxyl-terminal to
the first polypeptide segment (e.g., carboxyl-terminal to a dimerizing domain), the second polypeptide
segment is a paraoxonase. In some embodiments, the paraoxonase is a human paraoxonase 1 (PON1)
or a functional variant or fragment thereof. For example, in some embodiments, the second
polypeptide segment comprises an amino acid sequence having at least 80% identity with amino acid
residues 16-355 of SEQ ID NO:12, amino acid residues 16-355 of SEQ ID NO:42, or amino acid
residues 16-355 of SEQ ID NO:44. In more particular embodiments, the second polypeptide segment
comprises an amino acid sequence having at least 85%, at least 90%, or at least 95% identity with
amino acid residues 16-355 of SEQ ID NO:12, amino acid residues 16-355 of SEQ ID NO:42, or
amino acid residues 16-355 of SEQ ID NO:44. In yet other embodiments, the second polypeptide
segment comprises an amino acid sequence having at least 96%, at least 97%, at least 98%, at least
99%, or 100% sequence identity with amino acid residues 16-355 of SEQ ID NO:12, amino acid
residues 16-355 of SEQ ID NO:42, or amino acid residues 16-355 of SEQ ID NO:44.
In yet other embodiments comprising a second polypeptide segment carboxyl
terminal to the first polypeptide segment (e.g., carboxyl-terminal to a dimerizing domain), the second
polypeptide segment is
a platelet-activating factor acetylhydrolase (PAF-AH). In some embodiments,
the platelet-activating factor acetylhydrolase is a human PAF-AH or a functional variant or fragment
thereof. For example, in some embodiments, the second polypeptide segment comprises an amino
acid sequence having at least 80% identity with amino acid residues 22-441 of SEQ ID NO:32. In
more particular embodiments, the second polypeptide segment comprises an amino acid sequence
having at least 85%, at least 90%, or at least 95% identity with amino acid residues 22-441 of SEQ ID
NO:32. In yet other embodiments, the second polypeptide segment comprises an amino acid
sequence having at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity
with amino acid residues 22-441 of SEQ ID NO:32.
In still other embodiments comprising a second polypeptide segment carboxyl
terminal to the first polypeptide segment (e.g., carboxyl-terminal to a dimerizing domain), the second
polypeptide segment is a cholesterol ester transfer protein (CETP). In some embodiments, the
cholesterol ester transfer protein is a human CETP or a functional variant or fragment thereof. For
example, in some embodiments, the second polypeptide segment comprises an amino acid sequence
having at least 80% identity with amino acid residues 18-493 of SEQ ID NO:30. In more particular
the second polypeptide segment comprises an amino acid sequence having at least
embodiments,
85%, at least 90%, or at least 95% identity with amino acid residues 18-493 of SEQ ID NO:30. In yet
other embodiments, the second polypeptide segment comprises an amino acid sequence having at
least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with amino acid
residues 18-493 of SEQ ID NO:30.
Polypeptide linkers for use in accordance with the present invention can be naturally
occurring, synthetic, or a combination of both. The linker joins two separate polypeptide regions
(e.g., a dimerizing domain and an ApoA- 1 polypeptide) and maintains the linked polypeptide regions
as separate and discrete domains of a longer polypeptide. The linker can allow the separate, discrete
domains to cooperate yet maintain separate properties (e.g., in the case of an Fc region dimerizing
domain linked to an ApoA- 1 polypeptide, Fc receptor (e.g., FcRn) binding may be maintained for the
Fc region, while functional properties of the ApoA-1 polypeptide (e.g., lipid binding) will be
maintained). For examples of the use of naturally occurring as well as artificial peptide linkers to
connect heterologous polypeptides, see, e.g., Hallewell et al., J. Biol. Chem. 264, 5260-5268, 1989;
Alfthan et al., Protein Eng. 8, 725-731, 1995; Robinson and Sauer, Biochemistry 35, 109-116, 1996;
Khandekar et al., J. Biol. Chem. 272, 32190-32197, 1997; Fares et al., Endocrinology 139, 2459
2464, 1998; Smallshaw et al., Protein Eng. 12, 623-630, 1999; U.S. Patent No. 5,856,456.
Typically, residues within the linker polypeptide are selected to provide an overall
hydrophilic character and to be non-immunogenic and flexible. As used herein, a "flexible" linker is
one that lacks a substantially stable higher-order conformation in solution, although regions of local
stability are permissible. In general, small, polar, and hydrophilic residues are preferred, and bulky
and hydrophobic residues are undesirable. Areas of local charge are to be avoided; if the linker
polypeptide includes charged residues, they will ordinarily be positioned so as to provide a net neutral
charge within a small region of the polypeptide. It is therefore preferred to place a charged residue
adjacent to a residue of opposite charge. In general, preferred residues for inclusion within the linker
polypeptide
include Gly, Ser, Ala, Thr, Asn, and Gln; more preferred residues
include Gly, Ser, Ala,
Thr; and the most preferred residues are Gly and Ser. In general, Phe,
Tyr, Trp, Pro, Leu, Ile,
Lys, and Arg residues will be avoided (unless present within an immunoglobulin hinge region of the
linker), Pro residues due to their hydrophobicity and lack of flexibility, and Lys and Arg residues due
to potential immunogenicity. The sequence of the linker will also be designed to avoid unwanted
proteolysis.
In certain embodiments, linker LI comprises at least two or at least three amino acid
residues (e.g., at least five, at least 10, at least 16, at least 26, or at least 36 amino acid residues). In
particular variations, LI consists of from two to 60 amino acid residues, from three to 60 amino acid
residues, from five to 40 amino acid residues, or from 15 to 40 amino acid residues. In other
consists of from two to 50, from two to 40, from two to 36, from two to 35, from two to
variations, LI
, from two to 26, from three to 50, from three to 40, from three to 36, from three to 35, from three
to 30, from three to 26, from five to 60, from five to 50, from five to 40, from five to 36, from five to
, from five to 30, from five to 26, from 10 to 60, from 10 to 50, from 10 to 40, from 10 to 36, from
to 35, from 10 to 30, from 10
to 26, from 15 to 60, from 15 to 50, from 15 to 36, from 15 to 35,
from 15 to 30, or from 15 to 26 amino acid residues.
In other variations, LI consists of from 16 to 60,
from 16 to 50, from 16 to 40, or from 16 to 36 amino acid residues. In yet other variations, LI
consists of from 20 to 60, from 20 to 50, from 20 to 40, from 20
to 36, from 25 to 60, from 25 to 50,
from 25 to 40, or from 25 to 36 amino acid residues. In still other variations, LI consists of from 26
to 60, from 26
to 50, from 26 to 40, or from 26 to 36 amino acid residues. In more
specific variations,
LI consists of 16 amino acid residues, 21 amino acid residues, 26 amino acid residues, 31 amino acid
residues, or 36 amino acid residues. In some embodiments, LI comprises or consists of the amino
acid sequence shown in residues 268-293 of
SEQ ID NO:2, residues 268-288 of SEQ ID NO:26,
residues 268-283 of SEQ ID NO:22, SEQ ID NO:54, or residues 268-303 of SEQ ID NO:24.
Exemplary L2 linkers comprise at least three amino acid residues and are typically up
to 60 amino acid residues. In certain variations, L2 linkers have a range of sequence lengths as
described above for Li. In a specific embodiment of a polypeptide comprising the formula ApoAl
Li-D-L2-P and where L2 is present and P is an RNase, L2 comprises or consists of the amino acid
sequence shown in residues 526-541 of SEQ ID NO:4.
In certain embodiments, polypeptide linkers comprise a plurality of glycine resides.
For example, in some embodiments, a polypeptide linker (e.g., LI) comprises a plurality of glycine
residues and optionally at least one serine
residue. In particular variations, a polypeptide linker (e.g.,
LI) comprises the sequence Gly-Gly-Gly-Gly-Ser (SEQ ID NO:15), such as, e.g., two or more
tandem repeats of the amino acid sequence of SEQ ID NO:15. In some embodiments, a linker
comprises the sequence [Gly-Gly-Gly-Gly-Ser], ([SEQ ID NO: 15],), where n is a positive integer
such as, for example, an integer from 1 to 5, from 2 to 5, from 3 to 5, from 1 to 6, from 2 to 6, from 3
to 6, or from 4 to 6. In a specific variation of a polypeptide linker comprising the formula [Gly-Gly
Gly-Gly-Ser],, n is 4. In another specific variation of a polypeptide linker comprising the formula
[Gly-Gly-Gly-Gly-Ser],, n is 3. In yet another specific variation of a polypeptide linker comprising
the formula [Gly-Gly-Gly-Gly-Ser],, n is 5. In still another specific variation of a polypeptide linker
comprising the formula [Gly-Gly-Gly-Gly-Ser], n is 6. In certain embodiments, a polypeptide linker
comprises a series of glycine and serine residues (e.g., [Gly-Gly-Gly-Gly-Ser],, where n is defined as
above) inserted between two other sequences of the polypeptide linker (e.g., any of the polypeptide
linker sequences described herein). In other embodiments, a polypeptide linker includes glycine and
serine residues (e.g., [Gly-Gly-Gly-Gly-Ser], where n is defined as above) attached at one or both
sequence of the polypeptide linker (e.g., any of the polypeptide linker sequences
ends of another
described herein). In one embodiment, a polypeptide linker comprises at least a portion of an upper
hinge region (e.g., derived from an IgGI, IgG2, IgG3, or IgG4 molecule), at least a portion of a
middle hinge region (e.g., derived from an IgGI, IgG2, IgG3, or IgG4 molecule) and a series of
glycine and serine amino acid residues (e.g., [Gly-Gly-Gly-Gly-Ser], wherein n is defined as above).
In another embodiment, a polypeptide linker comprises a non-naturally occurring
immunoglobulin hinge region, e.g., a hinge region that is not naturally found in an immunoglobulin
and/or a hinge region that has been altered so that it differs in amino acid sequence from a naturally
occurring immunoglobulin hinge region. In one embodiment, mutations can be made to a hinge
region to make a polypeptide linker. In one embodiment, a polypeptide linker comprises a hinge
domain that does not comprise a naturally occurring number of cysteines, i.e., the polypeptide linker
comprises either fewer cysteines or a greater number of cysteines than a naturally occurring hinge
molecule.
Various dimerization domains are suitable for use in accordance with the fusion
polypeptides and dimeric fusion proteins as described herein. In certain embodiments, the dimerizing
domain is an immunoglobulin heavy chain constant region, such as an Fc region. The Fc region may
be a native sequence Fc region or a variant Fc region. In some embodiments, the Fc region lacks one
or more effector functions (e.g., one or both of ADCC and CDC effector functions).
In some embodiments, the dimerizing domain is an Fc region of a human antibody
with a mutation in the CH2 region so that the molecule is not glycosylated, including but not limited
to N297 (EU numbering for human IgG heavy chain constant region) (corresponding to amino acid
position 375 of SEQ ID NO:2). In another embodiment, the Fc region is human IgGI (71) with the
three cysteines of the hinge region (C220, C226, C229) each changed to serine, and the proline at
position 238 of the CH2 domain changed to serine. In another preferred embodiment, the Fc region is
human 71 with N297 changed to any other amino acid. In another embodiment, the Fc region is
human 71 with one or more amino acid substitutions between Eu positions 292 and 300. In another
embodiment, the Fc region is human 71 with one or more amino acid additions or deletions at any
position between residues 292 and 300. In another embodiment, the Fc region is human 71 with an
SCC hinge (i.e., with cysteine C220 changed to serine and with a cysteine at each of Eu positions 226
and 229) or an SSS hinge (i.e., each of the three cysteines at Eu positions 220, 226, and 229 changed
to serine). In further embodiments, the Fc region is human 71 with an SCC hinge and a P238
mutation. In another embodiment, the Fc domain is human 71 with mutations that alter binding by Fc
gamma receptors (I, II, III) without affecting FcRn binding important for half-life. In further
embodiments, an Fc region is as disclosed in Ehrhardt and Cooper, Curr. Top. Microbiol. Immunol.
2010 Aug. 3 (Immunoregulatory Roles for Fc Receptor-Like Molecules); Davis et al., Ann. Rev.
Immunol. 25:525-60, 2007 (Fc receptor-like molecules); or Swainson
et al., J. Immunol. 184:3639-47,
2010.
In some embodiments of a fusion polypeptide
comprising an Fc dimerizing domain,
the Fc region comprises an amino acid substitution that alters the antigen-independent effector
functions of the fusion protein. In some such embodiments, the Fc region includes an amino acid
substitution that alters the circulating half-life of the resulting molecule. Such antibody derivatives
exhibit either increased or decreased binding to FcRn when compared to antibodies lacking these
substitutions and, therefore, have an increased or decreased half-life in serum, respectively. Fc
variants with improved affinity for FcRn are anticipated to have longer serum half-lives, and such
antibodies have useful applications in methods of treating mammals where long half-life of the
administered antibody is desired. In contrast, Fc variants with decreased FcRn binding affinity are
expected to have shorter half-lives, and such antibodies are also useful, for example, for
administration to a mammal where a shortened circulation time may be advantageous, e.g., where the
starting antibody has toxic side effects when present in the circulation for prolonged periods. Fc
variants with decreased FcRn binding affinity are also less likely to cross the placenta and, thus, are
also useful in the treatment of diseases or disorders in pregnant women. In addition, other
applications in which reduced FcRn binding affinity may be desired include those applications in
which localization the brain, kidney, and/or liver is desired. In one exemplary embodiment, the
antibodies of the invention exhibit reduced transport across the epithelium of kidney glomeruli from
the vasculature. In another embodiment, the fusion proteins of the invention exhibit reduced transport
across the blood brain barrier (BBB) from the brain, into the vascular space. In one embodiment, a
fusion protein with altered FcRn binding comprises an Fc region having one or more amino acid
substitutions within the "FcRn binding loop" of the Fc domain. Exemplary amino acid substitutions
which altered FcRn binding activity are disclosed in International PCT Publication No. WO
05/047327, which is incorporated by reference herein.
In other embodiments, a fusion polypeptide of the present invention comprises an Fc
variant comprising an amino acid substitution which alters the antigen-dependent effector functions of
the polypeptide, in particular ADCC or complement activation, e.g., as compared to a wild type Fc
region. In an exemplary embodiment, such fusion polypeptides exhibit altered binding to an Fc
gamma receptor (FcyR, e.g., CD16). Such fusion polypeptides exhibit either increased or decreased
binding to FcyR when compared to wild-type polypeptides and, therefore, mediate enhanced or
reduced effector function, respectively. Fc variants with improved affinity for FcyRs are anticipated
to enhance effector function, and such fusion proteins have useful applications in methods of treating
mammals where target molecule destruction is desired. In contrast, Fc variants with decreased FcyR
binding affinity are expected to reduce effector function, and such fusion proteins are also useful, for
treatment of conditions in which target cell destruction is undesirable, e.g., where normal
example, for
cells may express target molecules, or where chronic administration of the antibody might result in
unwanted immune system activation. In one embodiment, the fusion polypeptide comprising an Fc
region exhibits at least one altered antigen-dependent effector function selected from the group
consisting of opsonization, phagocytosis, complement dependent cytotoxicity, antigen-dependent
cellular cytotoxicity (ADCC), or effector cell modulation as compared to a polypeptide comprising a
wild-type Fc region.
In one embodiment, a fusion polypeptide comprising an Fc region exhibits altered
binding to an activating FcyR (e.g., FcyI, FcyIIa, or FcyRIIIa). In another embodiment, the fusion
protein exhibits altered binding affinity to an inhibitory FcyR (e.g., FcyRI~b). Exemplary amino acid
substitutions which altered FcR or complement binding activity are disclosed in International PCT
Publication No. WO 05/063815, which is incorporated by reference herein.
A fusion polypeptide comprising an Fc region may also comprise an amino acid
substitution that alters the glycosylation of the Fc region. For example, the Fc domain of the fusion
protein may have a mutation leading to reduced glycosylation (e.g., N- or O-linked glycosylation) or
may comprise an altered glycoform of the wild-type Fc domain (e.g., a low fucose or fucose-free
glycan). In another embodiment, the molecule has an amino acid substitution near or within a
glycosylation motif, for example, an N-linked glycosylation motif that contains the amino acid
sequence NXT or NXS. Exemplary amino acid substitutions which reduce or alter glycosylation are
disclosed in International PCT Publication No. WO 05/018572 and US Patent Application Publication
No. 2007/0111281, which are incorporated by reference herein.
It will be understood by those of skill in the art that various embodiments of Fc
variants as described herein can be combined
in the fusion polypeptides of the present invention,
unless the context clearly indicates otherwise.
In some embodiments, a dimerizing domain is an Fc region comprising an amino acid
sequence having at least 80%, at least 85%, at least 90%, or at least 95% identity with an amino acid
sequence selected from sequence shown in (i) residues 294-525 or 294-524 of SEQ ID NO:2, or (ii)
residues 294-525 or 294-524 of SEQ ID NO:13. In yet other embodiments, the Fc region comprises
an amino acid sequence having at least 96%, at least 97%, at least 98%, at least 99%, or 100%
sequence identity with the amino acid sequence shown in (i) residues 294-525 or 294-524 of SEQ ID
NO:2, or (ii) residues 294-525 or 294-524 of SEQ ID NO: 13.
In some embodiments of a fusion polypeptide comprising ApoAl-L1-D as described
above, the fusion polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at
least 90%, or at least 95% identity with an amino acid sequence selected from sequence shown in (i)
19-524, 25-525, or 25-524 of SEQ ID NO:2, (ii) residues 19-525, 19-524, 25-525, or
residues 19-525,
-524 of SEQ ID NO:13, (iii) residues 19-501, 19-500, 25-501, or 25-501 of SEQ ID NO:20, (iv)
residues 19-515,
19-514, 25-515, or 25-514 of SEQ ID NO:22, (v) residues 19-520,
19-519, 25-520,
or 25-519 of SEQ ID NO:26, or (vi) residues 19-535, 19-534, 25-535, or 25-534 of SEQ ID NO:24.
In yet other embodiments, the fusion polypeptide comprises an amino acid sequence having at least
96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with the amino acid
sequence shown in (i) residues 19-525, 19-524, 25-525, or 25-524 of SEQ ID NO:2, (ii) residues 19
525, 19-524, 25-525, or 25-524 of SEQ ID NO:13, (iii) residues 19-501, 19-500, 25-501, or 25-501 of
SEQ ID NO:20, (iv) residues 19-515, 19-514, 25-515, or 25-514 of SEQ ID NO:22, (v) residues 19
of SEQ ID NO:26, or (vi) residues 19-535, 19-534, 25-535, or 25-534
520, 19-519, 25-520, or 25-519
of SEQ ID NO:24.
In some embodiments of a fusion polypeptide comprising ApoAl-L1-D-L2-P as
described above and where P is an RNase, the fusion polypeptide comprises an amino acid sequence
having at least 80%, at least 85%, at least 90%, or at least 95% identity with the amino acid sequence
shown in (i) residues 19-675 or 25-675 of SEQ ID NO:4 or (ii) residues 19-675 or 25-675 of SEQ ID
NO:14. In yet other embodiments, the fusion polypeptide comprises an amino acid sequence having
at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with the amino acid
sequence shown in (i) residues 19-675 or 25-675 of SEQ ID NO:4 or (ii) residues 19-675 or 25-675 of
SEQ ID NO:14.
In some embodiments of a fusion polypeptide comprising ApoAl-L1-D-L2-P as
described above and where P is a paraoxonase, the fusion polypeptide comprises an amino acid
sequence having at least 80%, at least 85%, at least 90%, or at least 95% identity with the amino acid
sequence shown in (i) residues 19-883 or 25-883 of SEQ ID NO:28, (ii) residues 19-873 or 25-873 of
SEQ ID NO:38, (iii) residues 19-883 or 25-883 of SEQ ID NO:46, or (iv) residues 19-883 or 25-883
of SEQ ID NO:48. In yet other embodiments, the fusion polypeptide comprises an amino acid
sequence having at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity
with the amino acid sequence shown in (i) residues 19-883 or 25-883 of SEQ ID NO:28, (ii) residues
19-873 or 25-873 of SEQ ID NO:38, (iii) residues 19-883 or 25-883 of SEQ ID NO:46, or (iv)
residues 19-883 or 25-883 of SEQ ID NO:48.
In some embodiments of a fusion polypeptide comprising ApoAl-L1-D-L2-P as
described above and where P is a platelet-activating factor acetylhydrolase (PAF-AH), the fusion
polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, or at
least 95% identity with the amino acid sequence shown in residues 19-963 or 25-963 of SEQ ID
NO:34. In yet other embodiments, the fusion polypeptide comprises an amino acid sequence having
at least 97%, at least 98%, at least 99%, or 100% sequence identity with the amino acid
at least 96%,
sequence shown in residues 19-963 or 25-963 of SEQ ID NO:34.
In some embodiments of a fusion polypeptide comprising ApoAl-L1-D-L2-P as
described above and where P is a cholesterol ester transfer protein (CETP), the fusion polypeptide
comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, or at least 95%
identity with the amino acid sequence shown in residues 19-1019 or 25-1019 of SEQ ID NO:40. In
yet other embodiments, the fusion polypeptide comprises an amino acid sequence having at least
96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity with the amino acid
sequence shown in residues 19-1019 or 25-1019 of SEQ ID NO:40.
The present invention also provides dimeric proteins comprising first and second
polypeptide fusions as described above. Accordingly, in another aspect, the present invention
provides
a dimeric protein comprising a first fusion polypeptide and a second
fusion polypeptide,
where each of the first and second polypeptide fusions comprises, from an amino-terminal position to
a carboxyl-terminal position, ApoAl-L1-D, where ApoAl is a first polypeptide segment having
cholesterol efflux activity and which is selected from (i) a naturally occurring ApoA-1 polypeptide or
a functional variant or fragment thereof and (ii) an ApoA-1 mimetic; LI is a first polypeptide linker;
and D is a dimerizing domain. In some embodiments, each of the first and second fusion polypeptides
further includes a second polypeptide segment located carboxyl-terminal to the dimerizing domain.
In particular
variations, the second polypeptide segment is (a) a naturally
occurring RNase,
paraoxonase, platelet-activating factor acetylhydrolase (PAF-AH), cholesterol ester transfer protein
(CETP), or lecithin-cholesterol acyltransferase (LCAT); (b) a functional variant or fragment of any of
the naturally occurring proteins specified in (a); or (c) a polypeptide that specifically binds to amyloid
beta (AP) such as, e.g., an An-specific scFv. Such a fusion polypeptide comprising a second
polypeptide segment may be represented by the formula ApoAl-L1-D-L2-P (from an amino-terminal
position to a carboxyl-terminal position), where
ApoAl, LI, and D are each as previously defined,
where L2 is a second polypeptide linker and is optionally present, and where P is the second
polypeptide segment.
In another aspect, the present invention provides a dimeric protein comprising a first
fusion polypeptide and a second fusion polypeptide, where each of the first and second fusion
polypeptides comprises a first polypeptide segment, a second polypeptide segment, and a dimerizing
domain, where the first polypeptide segment has cholesterol efflux activity and is selected from (i) a
naturally occurring ApoA- 1 polypeptide or a functional variant or fragment thereof and (ii) an ApoA
1 mimetic, and where the second polypeptide segment is located carboxyl-terminal to the first
polypeptide segment and is (a) a naturally occurring RNase, paraoxonase, platelet-activating factor
acetylhydrolase (PAF-AH), cholesterol ester transfer protein (CETP), or lecithin-cholesterol
of any of the naturally occurring proteins
acyltransferase (LCAT), (b) a functional variant or fragment
specified in (a), or (c) a polypeptide that specifically binds to amyloid beta (AP) such as, e.g., an Ap
specific scFv. In some embodiments, the dimerizing domain is located carboxyl-terminal to the first
polypeptide segment and amino-terminal to the second polypeptide segment.
In another aspect, the present invention provides (a) a first fusion polypeptide
comprising an immunoglobulin heavy chain linked carboxyl-terminal to an ApoA-1 polypeptide or
ApoA-1 mimetic and (b) a second fusion polypeptide comprising an immunoglobulin light chain
linked carboxyl-terminal to the ApoA-1 polypeptide or ApoA-1 mimetic. The first and second fusion
polypeptides can be co-expressed to create a stable tetramer composed of two double belt ApoA-1
dimers, wherein linkers between ApoA-1 and the heavy chain and between ApoA-1 and the light
chain are of sufficient length to allow cholesterol efflux and reverse cholesterol transport.
The fusion polypeptides of the present invention, including
dimeric fusion proteins,
can further be
conjugated to an effector moiety. The effector moiety can be any number
of molecules,
including, e.g., a labeling moiety such as a radioactive label or fluorescent label, a TLR ligand or
binding domain, an enzyme, or a therapeutic moiety. In a particular embodiment, the effector moiety
is a myeloperoxidase (MPO) inhibitor. MPO inhibitors are generally known (see, e.g., Malle et al., Br
J Pharmacol. 152: 838-854, 2007) and may be readily conjugated to fusion polypeptides as described
herein. Exemplary MPO inhibitors include inhibitors based on 3-alkylindole derivatives (see Soubhye
et al., J Med Chem 56:3943-58, 2013; describing studies of 3-alkylindole derivatives as selective and
highly potent myeloperoxidase inhibitors, including a compound with high and selective inhibition of
MPO (IC50 =18nM)); inhibitors based on 3-(aminoalkyl)fluorindoles (see Soubhye et al., J Med
Chem 53: 8747-8759, 2010);
inhibitors based on 2H-indazoles and 1H-indazolones (see Roth et
al.,
Bioorg Med Chem 22: 6422-6429, 2014; describing the evaluation 2H-indazoles and 1H-indazolones
and the identification of compounds with IC50 values <1pM); and benzoic acid hydrazide-containing
compounds (see Huang et al., Arch Biochem Biophys 570: 14-22, 2015; showing inactivation of MPO
by benzoic acid hydrazide-containing compounds, where the light chain subunit of MPO is freed from
the larger heavy chain by cleavage of the ester bond)..
In another embodiment, the fusion polypeptides of the present invention, including
dimeric fusion proteins, are modified to extend half-life, such as, for example, by attaching at least
one molecule to the fusion protein for extending serum half-life. Such molecules for attachment may
include, e.g., a polyethlyene glycol (PEG) group, serum albumin, transferrin, transferrin receptor or
the transferrin-binding portion thereof, or a combination thereof. Methods for such modification are
generally well-known in the art. As used herein, the word "attached" refers to a covalently or
noncovalently conjugated substance. The conjugation may be by genetic engineering or by chemical
means.
III. Materials and Methods for Making Polypeptide Fusions and Dimeric Proteins
The present invention also provides polynucleotide molecules, including DNA and
RNA molecules, that encode the fusion polypeptides disclosed above. The polynucleotides of the
present invention include both single-stranded and double-stranded molecules. Polynucleotides
encoding various segments of a fusion polypeptide (e.g., a dimerizing domain such as an Fc fragment;
ApoAl and P polypeptide segments) can be generated and linked together to form a polynucleotide
encoding a fusion polypeptide as described herein using known methods for recombinant
manipulation of nucleic acids.
sequences encoding ApoA-1, RNases (e.g., RNase 1),
paraoxonases (e.g.,
PON1), platelet-activating factor acetylhydrolase (PAF-AH), cholesterol ester transfer protein
(CETP), and lecithin-cholesterol acyltransferase (LCAT) are known in the art. DNA sequences
encoding various dimerizing domains (e.g., immunoglobulin heavy chain constant regions such as Fc
fragments) are also known. Polynucleotides encoding, e.g., the variable regions of An-binding
antibodies, including scFvs, are also readily identifiable using techniques well-known in the art such
as screening of recombinant antibody expression libraries (e.g., phage display expression libraries).
Additional DNA sequences encoding any of these polypeptides can be readily generated by those of
ordinary skill in the art based on the genetic code. Counterpart RNA sequences can be generated by
substitution of U for T. Those skilled in the art will readily recognize that, in view of the degeneracy
of the genetic code, considerable sequence variation is possible among polynucleotide molecules
encoding a given polypeptide. DNA and RNA encoding functional variants and fragments of such
polypeptides can also be obtained using known recombinant methods to introduce variation into a
polynucleotide sequence, followed by expression of the encoded polypeptide and determination of
functional activity (e.g., cholesterol efflux) using an appropriate screening assay.
Methods for
preparing DNA and RNA are well known in the art. For example,
complementary DNA (cDNA) clones can be prepared from RNA that is isolated from a tissue or cell
that produces large amounts of RNA encoding a polypeptide of interest. Total RNA can be prepared
using guanidine HCl extraction followed by isolation by centrifugation in a CsCl gradient (Chirgwin
et al., Biochemistry 18:52-94, 1979). Poly (A)' RNA is prepared from total RNA using the method of
Aviv and Leder (Proc. Natl. Acad. Sci. USA 69:1408-1412, 1972). Complementary DNA is prepared
from poly(A)* RNA using known methods. In the alternative, genomic DNA can be isolated.
Methods for identifying and isolating cDNA and genomic clones are well known and within the level
of ordinary skill in the art, and include the use of the sequences disclosed herein, or parts thereof, for
probing or priming a library. Polynucleotides encoding polypeptides of interest are identified and
isolated by, for example, hybridization or polymerase chain reaction ("PCR," Mullis, U.S. Patent
receptor
4,683,202). Expression libraries can be probed with antibodies to the polypeptide of interest,
fragments, or other specific binding partners.
The polynucleotides of the present invention can also be prepared by automated
synthesis. The production of short, double-stranded segments (60 to 80 bp) is technically
straightforward and can be accomplished by synthesizing the complementary strands and then
annealing them. Longer segments (typically >300 bp) are assembled in modular form from single
stranded fragments that are from 20 to 100 nucleotides in length. Automated synthesis of
polynucleotides is within the level of ordinary skill in the art, and suitable equipment and reagents are
available from commercial suppliers. See generally Glick and Pasternak, Molecular Biotechnology,
Principles & Applications of Recombinant DNA,
ASM Press, Washington, D.C., 1994; Itakura et al.,
Ann. Rev. Biochem. 53:323-356, 1984; and Climie
et al., Proc. Natl. Acad. Sci. USA 87:633-637,
1990.
In another aspect, materials and methods are provided for producing the polypeptide
fusions of the present invention, including dimeric proteins comprising the fusion polypeptides. The
fusion polypeptides can be produced in genetically engineered host cells according to conventional
techniques. Suitable host cells are those cell types that can be transformed or transfected with
exogenous DNA and grown in culture, and include bacteria, fungal cells, and cultured higher
eukaryotic cells (including cultured cells of multicellular organisms), particularly cultured mammalian
cells. Techniques for manipulating cloned DNA molecules and introducing exogenous DNA into a
variety of host cells are disclosed by Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd
ed., Cold Spring Harbor
Laboratory Press, Cold Spring Harbor, NY, 1989, and Ausubel et al.,
eds.,
Current Protocols in Molecular Biology, Green and Wiley and Sons, NY, 1993.
In general, a DNA sequence encoding a fusion polypeptide is operably linked to other
genetic elements required for its expression, generally including a transcription promoter and
terminator, within an expression vector. The vector will also commonly contain one or more
selectable markers and one or more origins of replication, although those skilled in the art will
recognize that within certain systems selectable markers may be provided on separate vectors, and
replication of the exogenous DNA may be provided by integration into the host cell genome.
Selection of promoters, terminators, selectable markers, vectors and other elements is a matter of
routine design within the level of ordinary skill in the art. Many such elements are described in the
literature and are available through commercial suppliers.
To direct an ApoA-1 fusion polypeptide into the secretory pathway of a host cell, a
secretory signal sequence is provided in the expression vector. The secretory signal sequence may be
that of the native ApoA-1 polypeptide, or may be derived from another secreted protein (e.g., t-PA;
see U.S. Patent No. 5,641,655) or synthesized de novo. An engineered cleavage site may be included
between the secretory peptide and the remainder of the polypeptide fusion to optimize
at the junction
proteolytic processing in the host cell. The secretory signal sequence is operably linked to the DNA
sequence encoding the polypeptide fusion, i.e., the two sequences are joined in the correct reading
frame and positioned to direct the newly synthesized polypeptide fusion into the secretory pathway of
the host cell. Secretory signal sequences are commonly positioned 5' to the DNA sequence encoding
the polypeptide of interest, although certain signal sequences may be positioned elsewhere in the
DNA sequence of interest (see, e.g., Welch et al., U.S. Patent No. 5,037,743; Holland et al., U.S.
Patent No. 5,143,830). Secretory signal sequences suitable for use in accordance with the present
invention include, for example, polynucleotides encoding amino acid residues 1-18 of SEQ ID NO:2.
Expression of fusion polypeptides comprising a dimerizing domain, via a host cell
secretory pathway, is expected to result in the production of dimeric proteins. Accordingly, in another
aspect, the present invention provides dimeric proteins comprising first and second fusion
polypeptides as described above (e.g., a dimeric protein comprising a first fusion polypeptide and a
second fusion polypeptide, where each of the first and second fusion polypeptides comprises, from an
amino-terminal position to a carboxyl-terminal position, ApoAl-L1-D or ApoAl-L1-D-L2-P as
described herein). Dimers may also be assembled in vitro upon incubation of component
polypeptides under suitable conditions. In general, in vitro assembly will include incubating the
protein mixture under denaturing and reducing conditions followed by refolding and reoxidation of
the polypeptides to form dimers. Recovery and assembly of proteins expressed in bacterial cells is
disclosed below.
Cultured mammalian cells are suitable hosts for use within the present invention.
Methods for introducing exogenous DNA into mammalian host cells include calcium phosphate
mediated transfection (Wigler et al., Cell 14:725, 1978; Corsaro and Pearson, Somatic Cell Genetics
7:603, 1981: Graham and Van der Eb, Virology 52:456,
1973), electroporation (Neumann et al.,
EMBO J. 1:841-845, 1982), DEAE-dextran mediated transfection (Ausubel et al., supra), and
liposome-mediated transfection (Hawley-Nelson et al., Focus 15:73, 1993; Ciccarone et al., Focus
:80, 1993). The production of recombinant polypeptides in cultured mammalian cells is disclosed
by, for example, Levinson et al., U.S. Patent No. 4,713,339; Hagen et al., U.S. Patent No. 4,784,950;
Palmiter et al., U.S. Patent No. 4,579,821; and Ringold, U.S. Patent No. 4,656,134. Suitable cultured
mammalian cells include the COS-1 (ATCC No. CRL 1650), COS-7 (ATCC No. CRL 1651), BHK
(ATCC No. CRL 1632), BHK 570 (ATCC No. CRL 10314), 293 (ATCC No. CRL 1573; Graham et
al., J. Gen. Virol. 36:59-72, 1977) and Chinese hamster ovary (e.g., CHO-KI, ATCC No. CCL 61;
CHO-DG44, Urlaub et al., Proc. Natl. Acad. Sci. USA 77:4216-4220, 1980) cell lines. Additional
suitable cell lines are known in the art and available from public depositories such as the American
Type Culture Collection, Manassas, Virginia. Strong transcription promoters can be used, such as
sarcoma virus. See, e.g., U.S. Patent
promoters from SV-40, cytomegalovirus, or myeloproliferative
No. 4,956,288 and U.S. Patent Application Publication No. 20030103986. Other suitable promoters
include those from metallothionein genes (U.S. Patents Nos. 4,579,821 and 4,601,978) and the
adenovirus major late promoter. Expression vectors for use in mammalian cells include pZP-1, pZP
9, and pZMP21, which have been deposited with the American Type Culture Collection, 10801
University Blvd., Manassas, VA USA under
accession numbers 98669, 98668, and PTA-5266,
respectively, and derivatives of these vectors.
Drug selection is generally used to select for cultured mammalian cells into which
foreign DNA has been inserted. Such cells are commonly referred to as "transfectants." Cells that
have been cultured in the presence of the selective agent and are able to pass the gene of interest to
their progeny are referred to as "stable transfectants." An exemplary selectable marker is a gene
encoding resistance to the antibiotic neomycin. Selection is carried out in the presence of a
neomycin-type drug, such as G-418 or the like. Selection systems can also be used to increase the
expression level of the gene of interest, a process referred to as "amplification." Amplification is
carried out by culturing transfectants in the presence of a low level of the selective agent and then
increasing the amount of selective agent to select for cells that produce high levels of the products of
the introduced genes. An exemplary amplifiable selectable marker is dihydrofolate reductase, which
confers resistance to methotrexate. Other drug resistance genes (e.g., hygromycin resistance, multi
drug resistance, puromycin acetyltransferase) can also be used. Cell-surface markers and other
phenotypic selection markers can be used to facilitate identification of transfected cells (e.g., by
fluorescence-activated cell sorting), and include, for example, CD8, CD4, nerve growth factor
receptor, green fluorescent protein, and the like.
Other higher eukaryotic cells can also be used as hosts, including insect cells, plant
cells and avian cells. The use of Agrobacterium rhizogenes as a vector for expressing genes in plant
cells has been reviewed by Sinkar et al., J. Biosci. (Bangalore) 11:47-58, 1987. Transformation of
insect cells and production of foreign polypeptides therein is disclosed by Guarino et al., U.S. Patent
No. 5,162,222 and WIPO publication WO 94/06463.
Insect cells can be infected with recombinant baculovirus, commonly derived from
Autographa californica nuclear polyhedrosis virus (AcNPV). See King and Possee, The Baculovirus
Expression System: A Laboratory Guide, Chapman & Hall, London; O'Reilly et al., Baculovirus
Expression Vectors: A Laboratory Manual, Oxford University Press., New York, 1994; and
Richardson, Ed.,
Baculovirus Expression Protocols. Methods in Molecular Biology, Humana
Press,
Totowa, NJ, 1995. Recombinant baculovirus can also be produced through the use of a transposon
based system described by Luckow et al. (J. Virol. 67:4566-4579, 1993). This system, which utilizes
transfer vectors, is commercially
available in kit form (BAC-TO-BAC kit; Life Technologies,
Life Technologies) contains a Tn7
Gaithersburg, MD). The transfer vector (e.g., PFASTBAC1;
transposon to move the DNA encoding the protein of interest into a baculovirus genome maintained in
E. coli as a large
plasmid called a "bacmid." See Hill-Perkins and Possee, J. Gen. Virol.
71:971-976,
1990; Bonning et al., J. Gen. Virol. 75:1551-1556, 1994; and Chazenbalk and Rapoport, J. Biol.
Chem. 270:1543-1549, 1995. Using techniques known in the art, a transfer vector encoding a
polypeptide fusion is transformed into E. coli host cells, and the cells are screened for bacmids which
contain an interrupted lacZ gene indicative of recombinant baculovirus. The bacmid DNA containing
the recombinant baculovirus genome is isolated, using common techniques, and used to transfect
Spodoptera frugiperda cells, such as Sf9 cells. Recombinant virus that expresses the polypeptide
fusion is subsequently produced. Recombinant viral stocks are made by methods commonly used the
art.
For protein production, the recombinant virus is used to infect host cells, typically a
cell line derived from the fall armyworm, Spodoptera frugiperda (e.g., Sf9 or Sf21 cells) or
Trichoplusia ni (e.g., HIGH FIVE cells; Invitrogen, Carlsbad, CA). See generally Glick and
Pasternak, supra. See also U.S. Patent No. 5,300,435. Serum-free media are used to grow and
maintain the cells. Suitable media formulations are known in the art and can be obtained from
commercial suppliers. The cells are grown up from an inoculation density of approximately 2-5 x 10
cells to a density of 1-2 x 106 cells, at which time a recombinant viral stock is added at a multiplicity
of infection (MOI) of 0.1 to 10, more typically near 3. Procedures used are generally described in
available laboratory manuals (e.g., King and Possee, supra;
O'Reilly et al., supra.; Richardson,
supra).
Fungal cells, including yeast cells, can also be used within the present invention.
Yeast species of particular interest in this
regard include Saccharomyces cerevisiae, Pichia pastoris,
and Pichia methanolica. Methods for transforming S. cerevisiae cells with exogenous DNA and
producing recombinant polypeptides therefrom are disclosed by, for example, Kawasaki, U.S. Patent
No. 4,599,311; Kawasaki et al., U.S. Patent No. 4,931,373; Brake, U.S. Patent No. 4,870,008; Welch
et al., U.S. Patent No. 5,037,743; and Murray et al., U.S. Patent No. 4,845,075. Transformed cells are
selected by phenotype determined by the selectable marker, commonly drug resistance or the ability
to grow in the absence of a particular nutrient (e.g., leucine). An exemplary vector system for use in
Saccharomyces cerevisiae is the POT] vector system disclosed by Kawasaki et al. (U.S. Patent No.
4,931,373), which allows transformed cells to be selected by growth in glucose-containing media.
Suitable promoters and terminators for use in yeast include those from glycolytic enzyme genes (see,
e.g.,
Kawasaki, U.S. Patent No. 4,599,311; Kingsman et al., U.S. Patent
No. 4,615,974; and Bitter,
U.S. Patent No. 4,977,092) and alcohol dehydrogenase genes. See also U.S. Patents Nos. 4,990,446;
,063,154; 5,139,936; and 4,661,454. Transformation systems for other yeasts, including Hansenula
lactis, Kluyveromyces fragilis, Ustilago
polymorpha, Schizosaccharomyces pombe, Kluyveromyces
maydis, Pichia pastoris, Pichia methanolica, Pichia guillermondii, and Candida maltosa are known
in the art. See, e.g., Gleeson et al., J. Gen. Microbiol. 132:3459-3465, 1986; Cregg, U.S. Patent No.
4,882,279; and Raymond et al., Yeast 14:11-23, 1998. Aspergillus cells may be utilized according to
the methods of McKnight et al., U.S. Patent No. 4,935,349. Methods for transforming Acremonium
chrysogenum are disclosed by Sumino et al., U.S. Patent No. 5,162,228. Methods for transforming
Neurospora are disclosed by Lambowitz, U.S. Patent No. 4,486,533. Production of recombinant
proteins in Pichia methanolica is disclosed in U.S. Patents Nos. 5,716,808; 5,736,383; 5,854,039; and
,888,768.
Prokaryotic host cells, including strains of the bacteria Escherichia coli, Bacillus and
other genera are also useful host cells within the present invention. Techniques for transforming these
hosts and expressing foreign DNA sequences cloned therein
are well-known in the art (see, e.g.,
Sambrook et al., supra). When expressing a fusion polypeptide in bacteria such as E. coli, the
polypeptide may be retained in the cytoplasm, typically as insoluble granules, or may be directed to
the periplasmic space by a bacterial secretion sequence. In the former case, the cells are lysed, and
the granules are recovered and denatured using, for example, guanidine HCl or urea. The denatured
polypeptide can then be refolded and dimerized by diluting the denaturant, such as by dialysis against
a solution of urea and a combination of reduced and oxidized glutathione, followed by dialysis against
a buffered saline solution. In the alternative, the protein may be recovered from the cytoplasm in
soluble form and isolated without the use of denaturants. The protein is recovered from the cell as an
aqueous extract in, for example, phosphate buffered saline. To capture the protein of interest, the
extract is applied directly to a chromatographic medium, such as an immobilized antibody or heparin
Sepharose column. Secreted polypeptides can be recovered from the periplasmic space in a soluble
and functional form by disrupting the cells (by, for example, sonication or osmotic shock) and
recovering the protein, thereby obviating
the need for denaturation and refolding. See, e.g., Lu et al.,
J. Immunol. Meth. 267:213-226, 2002.
Transformed or transfected host cells are cultured according to conventional
procedures in a culture medium containing nutrients and other components required for the growth of
the chosen host cells. A variety of suitable media, including defined media and complex media, are
known in the art and generally include
a carbon source, a nitrogen source, essential amino acids,
vitamins and minerals. Media may also contain such components as growth factors or serum, as
required. The growth medium will generally select for cells containing the exogenously added DNA
by, for example, drug selection or deficiency in an essential nutrient which is complemented by the
selectable marker carried on the expression vector or co-transfected into the host cell.
of the present invention are purified by conventional protein purification
Proteins
methods, typically by a combination of chromatographic techniques. See generally Affinity
Chromatography: Principles & Methods, Pharmacia LKB Biotechnology, Uppsala, Sweden, 1988;
and Scopes, Protein Purification: Principles and Practice, Springer-Verlag, New York, 1994.
Proteins comprising an immunoglobulin heavy chain polypeptide can be purified by affinity
chromatography on immobilized protein A. Additional purification steps, such as gel filtration, can
be used to obtain the desired level of purity or to provide for desalting, buffer exchange, and the like.
For example, fractionation and/or conventional purification methods can be used to
obtain fusion polypeptides and dimeric proteins of the present invention purified from recombinant
host cells. In general, ammonium sulfate precipitation and acid or chaotrope extraction may be used
for fractionation of samples. Exemplary purification steps may include hydroxyapatite, size
exclusion, FPLC and reverse-phase high performance liquid chromatography. Suitable
chromatographic media include derivatized dextrans, agarose, cellulose, polyacrylamide, specialty
silicas, and the like. PEI, DEAE, QAE and Q derivatives are suitable. Exemplary chromatographic
media include those media derivatized with phenyl, butyl, or octyl groups, such as Phenyl-Sepharose
FF (Pharmacia), Toyopearl butyl 650 (Toso Haas, Montgomeryville, PA), Octyl-Sepharose
(Pharmacia) and the like; or polyacrylic resins, such as Amberchrom CG 71 (Toso Haas) and the like.
Suitable solid supports include glass beads, silica-based resins, cellulosic resins, agarose beads, cross
linked agarose beads, polystyrene beads, cross-linked polyacrylamide resins and the like that are
insoluble under the conditions in which they are to be used. These supports may be modified with
reactive groups that allow attachment of proteins by amino groups, carboxyl groups, sulfhydryl
groups, hydroxyl groups and/or carbohydrate moieties.
Examples of coupling chemistries include cyanogen bromide activation, N
hydroxysuccinimide activation, epoxide activation, sulfhydryl activation, hydrazide activation, and
carboxyl and amino derivatives for carbodiimide coupling chemistries. These and other solid media
are well-known and widely used in the art, and are available from commercial suppliers. Selection of
a particular method for polypeptide isolation and purification is a matter of routine design and is
determined in part by the properties of the chosen support. See, e.g., Affinity Chromatography:
Principles & Methods (Pharmacia LKB Biotechnology 1988); and Doonan, Protein Purification
Protocols (The Humana Press 1996).
Additional variations in protein isolation and purification can be devised by those of
skill in the art. For example, antibodies that specifically bind a fusion polypeptide or dimeric protein
as described herein (e.g., an antibody that specifically binds a polypeptide segment corresponding to
ApoA-1) can be used to isolate large quantities of protein by immunoaffinity purification.
proteins of the present invention can also be isolated by exploitation of particular
properties. For example, immobilized metal ion adsorption (IMAC) chromatography can be used to
purify histidine-rich proteins, including those comprising polyhistidine tags. Briefly, a gel is first
charged with divalent metal ions to form a chelate (Sulkowski, Trends in Biochem. 3:1, 1985).
Histidine-rich proteins will be adsorbed to this matrix with differing affinities, depending upon the
metal ion used, and will be eluted by competitive elution, lowering the pH, or use of strong chelating
agents. Other methods of purification include purification of glycosylated proteins by lectin affinity
chromatography and ion exchange chromatography (see, e.g., M. Deutscher, (ed.), Meth. Enzymol.
182:529, 1990). Within additional embodiments of the invention, a fusion of the polypeptide of
interest and an affinity tag (e.g., maltose-binding protein, an immunoglobulin domain) may be
constructed to facilitate purification. Moreover, receptor- or ligand-binding properties of a fusion
polypeptide or dimer thereof can be exploited for purification. For example, a fusion polypeptide
comprising an An-binding polypeptide segment may be isolated by using affinity chromatography
wherein amyloid beta (AP) peptide is bound to a column and the fusion polypeptide is bound and
subsequently eluted using standard chromatography methods.
The polypeptides of the present invention are typically purified to at least about 80%
purity, more typically to at least about 90% purity and preferably to at least about 95%, at least about
96%, at least about 97%, at least about 98%, or at least about 99% purity with respect to
contaminating macromolecules, particularly other proteins and nucleic acids, and free of infectious
and pyrogenic agents. The polypeptides of the present invention may also be purified to a
pharmaceutically pure state, which is greater than 99.9% pure. In certain preparations, purified
polypeptide is substantially free of other polypeptides, particularly other polypeptides of animal
origin.
IV. Methods of Use and Pharmaceutical Compositions
The fusion polypeptides and dimeric proteins of the present invention can be used to
provide ApoAmediated therapy for the treatment of various diseases or disorders. In some aspects
relating to bispecific fusions further comprising a second polypeptide segment as described herein
(e.g., an RNase, paraoxonase, platelet-activating factor acetylhydrolase (PAF-AH), cholesterol ester
transfer protein (CETP), or lecithin-cholesterol acyltransferase (LCAT)), the fusion polypeptides and
dimeric proteins may further provide one or more additional biological activities for such treatment.
In particular aspects, the present invention provides methods for treating a disease or
disorder selected from a cardiovascular disease characterized by atherosclerosis, a neurodegenerative
disease, a disease characterized by amyloid deposit, an autoimmune
disease, an inflammatory disease,
disease, obesity, metabolic syndrome, nephrotic syndrome, burns, exposure to sulfur
an infectious
mustard gas, exposure to an organophosphate, and cancer. The methods generally include
administering to a subject having the disease or disorder an effective amount of a fusion polypeptide
or dimeric protein as described herein.
Atherosclerotic cardiovascular diseases amenable to treatment in accordance with the
present invention include, for example, coronary heart disease and stroke. In some variations of
treatment of coronary heart disease, the coronary heart disease is characterized by acute coronary
syndrome. In some embodiments, the atherosclerotic cardiovascular disease is selected from cerebral
artery disease (e.g., extracranial
cerebral artery disease, intracranial cerebral artery disease),
arteriosclerotic aortic disease, renal artery disease, mesenteric artery disease, and peripheral artery
disease (e.g., aortoiliac occlusive disease).
Neurodegenerative diseases amenable to treatment in accordance with the present
invention include, for example, neurodegenerative diseases characterized by amyloid deposit and/or
dementia. An exemplary neurodegenerative disease characterized by amyloid deposit is Alzheimer's
disease. Exemplary neurodegenerative diseases characterized by dementia include Alzheimer's
disease, Parkinson's disease, Huntington's disease, and amylotrophic lateral sclerosis (ALS). In some
embodiments, the neurodegenerative disease is an inflammatory disease such as, for example, a
demyelinating inflammatory disease of the CNS (e.g., multiple sclerosis (MS), including, for
example, spino-optical MS, primary progressive MS (PPMS), and relapsing remitting MS (RRMS)).
In some embodiments of a
method for treating a neurodegenerative disease (e.g.,
Alzheimer's disease or Parkinson's disease), a fusion molecule for the neurodegenerative disease
treatment is a polypeptide having the structure ApoAl-L1-D-L2-RNase (e.g., ApoAl-L1-[Fc region]
L2-RNasel) or ApoAl-L1-D-L2-paraoxonase (e.g., ApoAl-L1-[Fc region]-L2-PON1), or a dimeric
protein formed by dimerization of any of the foregoing fusion polypeptides; in some such
embodiments, the fusion polypeptide comprises or consists of an amino acid sequence having at least
90%, at least 95%, or
100% identity with (i) amino acid residues 19-675 or 25-675 of SEQ
ID NO:4,
(ii) amino acid residues 19-657 or 25-675 of SEQ ID NO:14, (iii) amino acid residues 19-883 or 25
883 of SEQ ID NO:28, (iv) amino acid residues 19-873 or 25-873 of SEQ ID NO:38, (v) amino acid
residues 19-883 or 25-883 of SEQ ID NO:46, or (vi) amino acid acid residues 19-883 or 25-883 of
SEQ ID NO:48.
Autoimmune diseases amenable to treatment in accordance with the present invention
include, for example, rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, and type
1 diabetes. In other embodiments, the autoimmune
disease is selected from coeliac disease, neuritis,
polymyositis, juvenile rheumatoid arthritis, psoriasis, psoriatic arthritis,
vitiligo, Sjogren's syndrome,
autoimmune pancreatitis, an inflammatory bowel disease (e.g., Crohn's
disease, ulcerative colitis),
active chronic hepatitis, glomerulonephritis, lupus nephritis, scleroderma,
antiphospholipid syndrome,
autoimmune vasculitis, sarcoidosis, autoimmune thyroid diseases, Hashimoto's thyroiditis, Graves
disease, Wegener's
granulomatosis, myasthenia gravis, Addison's disease, autoimmune
uveoretinitis,
pemphigus vulgaris, primary biliary
cirrhosis, pernicious anemia, sympathetic opthalmia, uveitis,
autoimmune hemolytic anemia, pulmonary fibrosis, chronic beryllium disease, and idiopathic
pulmonary fibrosis. In some variations, the autoimmune
disease is selected from rheumatoid arthritis,
juvenile rheumatoid arthritis, psoriatic arthritis, systemic lupus
erythematosus, lupus nephritis,
scleroderma, psoriasis, Sjogren's syndrome, type 1 diabetes, antiphospholipid syndrome, and
autoimmune vasculitis.
In some embodiments, a fusion molecule for treatment of an autoimmune disease is a
polypeptide having the structure ApoAl-L1-D (e.g., ApoAl-L1-[Fc region]), ApoAl-L1-D-L2
RNase (e.g., ApoAl-L1-[Fc region] -L2-RNase 1), or ApoAl-Li-D-L2-paraoxonase (e.g., ApoAl-L1
[Fc region]-L2-PON1), or a dimeric protein formed by dimerization of any of the foregoing fusion
polypeptides; in some such embodiments, the fusion polypeptide comprises or consists of an amino
acid sequence having at least 90%, at least 95%, or 100% identity with (i) amino acid residues 19
525, 19-524, 25-525, or 25-524 of SEQ ID NO:2, (ii) amino acid residues 19-525, 19-524, 25-525, or
-524 of SEQ ID NO:13, (iii) amino acid residues 19-501, 19-500, 25-501, or 25-501 of SEQ ID
NO:20, (iv) amino acid residues 19-515, 19-514, 25-515, or 25-514 of SEQ ID NO:22, (v) amino acid
residues 19-535, 19-534, 25-535, or 25-534 of SEQ ID NO:24, (vi) amino acid residues 19-675 or 25
675 of SEQ ID NO:4, (vii) amino acid residues 19-657 or 25-675 of SEQ ID NO:14, (viii) amino acid
residues 19-883 or 25-883 of SEQ ID NO:28, (ix) amino acid residues 19-873 or 25-873 of SEQ ID
NO:38, (x) amino acid residues 19-883 or 25-883 of SEQ ID NO:46, or (xi) amino acid residues 19
883 or 25-883 of SEQ ID NO:48. In some particular variations of a method for treating rheumatoid
arthritis (RA), a fusion molecule for the RA treatment is a polypeptide having the structure ApoAl
L1-D (e.g., ApoAl-L1-[Fc region]), or a dimeric protein formed by dimerization of the foregoing
fusion polypeptide; in some such embodiments, the fusion polypeptide comprises or consists of an
amino acid sequence having at least 90%, at least 95%, or 100% identity with (i) amino acid residues
19-525, 19-524, 25
19-525, 19-524, 25-525, or 25-524 of SEQ ID NO:2, (ii) amino acid residues
525, or 25-524 of SEQ ID NO:13, (iii) amino acid residues 19-501, 19-500, 25-501, or 25-501 of SEQ
ID NO:20, (iv) amino acid residues 19-515, 19-514, 25-515, or 25-514 of SEQ ID NO:22, or (v)
amino acid residues 19-535, 19-534, 25-535, or 25-534 of SEQ ID NO:24. In some particular
variations of a method for treating systemic lupus erythematosus (SLE), a fusion molecule for the
SLE treatment is a polypeptide having the structure ApoAl-L1-D-L2-RNase (e.g., ApoAl-L1-[Fc
region]-L2-RNasel) or ApoAl-Li-D-L2-paraoxonase (e.g., ApoAl-L1-[Fc region]-L2-PON1), or a
dimeric protein formed by dimerization of any of the foregoing fusion polypeptides; in some such
embodiments, the fusion polypeptide comprises or consists of an amino acid sequence having at least
90%, at least 95%, or 100% identity with (i) amino acid residues
19-675 or 25-675 of SEQ ID NO:4,
(ii) amino acid residues 19-657 or 25-675 of SEQ ID NO:14, (iii) amino acid residues 19-883 or 25
883 of SEQ ID NO:28, (iv) amino acid residues 19-873 or 25-873 of SEQ ID NO:38, (v) amino acid
residues 19-883 or 25-883 of SEQ ID NO:46, or (vi) amino acid residues 19-883 or 25-883 of SEQ ID
NO:48. In some particular variations of a method for treating multiple sclerosis (MS), a fusion
molecule for the MS treatment is a polypeptide having the structure ApoAl-L1-D-L2-paraoxonase
(e.g., ApoAl-L1-[Fc region]-L2-PON1), or a dimeric protein formed by dimerization of the foregoing
fusion polypeptide; in some such embodiments, the fusion polypeptide comprises or consists of an
amino acid sequence having at least 90%, at least 95%, or 100% identity with (i) amino acid residues
19-883 or 25-883
of SEQ ID NO:28, (ii) amino acid residues 19-873 or 25-873 of
SEQ ID NO:38,
(iii) amino acid residues 19-883 or 25-883 of SEQ ID NO:46, or (iv) amino acid residues 19-883 or
-883 of SEQ ID NO:48.
Inflammatory diseases amenable to treatment in accordance with the present
invention
include, for example, rheumatoid arthritis, systemic lupus erythematosus,
multiple sclerosis,
type 1 diabetes, type 2 diabetes, and obesity. In some embodiments, the inflammatory disease is an
neurodegenerative inflammatory
disease such as, for example, multiple sclerosis, Alzheimer's disease,
or Parkinson's disease. In other embodiments, the inflammatory disease is an atherosclerotic disease
(e.g., coronary heart disease or stroke). In yet other variations, the inflammatory disease is selected
from hepatitis (e.g., non-alcoholic steatohepatitis), ankylosing
spondylitis, arthritis (e.g.,
osteoarthritis, rheumatoid arthritis (RA), psoriatic arthritis),
Crohn's disease, ulcerative colitis,
dermatitis, diverticulitis, fibromyalgia, irritable bowel syndrome (IBS), and nephritis. In other
embodiments,
the inflammatory disease is an inflammatory lung disease; in some
such embodiments,
the inflammatory lung disease is selected from asthma, chronic obstructive pulmonary disease
(COPD), bronchiectasis, idiopathic pulmonary fibrosis, hyperoxia, hypoxia, and acute respiratory
distress syndrome (ARDS). In some variations, a patient having the inflammatory lung disease is a
patient that has been exposed to sulfur mustard gas (SM). In other variations, a patient having the
inflammatory lung disease is a patient that has been exposed to an organophosphate, such as an
insecticide or other neurotoxin.
In some embodiments, a fusion molecule for treatment of an inflammatory disease
(e.g., an inflammatory lung disease) is a polypeptide having the structure ApoAl-L1-D (e.g., ApoAl
L1-[Fc region]), ApoAl-Li-D-L2-RNase (e.g., ApoAl-L1-[Fc region] -L2-RNase 1), or ApoAl-Li-D
L2-paraoxonase (e.g., ApoAl-L1-[Fc region]-L2-PON1), or a dimeric protein formed by dimerization
of any of the foregoing fusion polypeptides; in some such embodiments, the fusion polypeptide
comprises or consists of an amino acid sequence having at least 90%, at least 95%, or 100% identity
with (i) amino acid residues 19-525, 19-524, 25-525, or 25-524 of SEQ ID NO:2, (ii) amino acid
19-524, 25-525, or 25-524 of SEQ ID NO:13, (iii) amino acid residues 19-501, 19
residues 19-525,
500, 25-501, or 25-501 of SEQ ID NO:20, (iv) amino acid residues 19-515, 19-514, 25-515, or 25
514 of SEQ ID
NO:22, (v) amino acid residues 19-535, 19-534, 25-535, or 25-534 of
SEQ ID NO:24,
(vi) amino acid residues 19-675 or 25-675 of SEQ ID NO:4, (vii) amino acid residues 19-657 or 25
675 of SEQ ID NO:14, (viii) amino acid residues 19-883 or 25-883 of SEQ ID NO:28, (ix) amino
acid residues 19-873 or 25-873 of SEQ ID NO:38, (x) amino acid residues 19-883 or 25-883 of SEQ
ID NO:46, or (xi) amino acid residues 19-883 or 25-883 of SEQ ID NO:48. In some particular
variations of a method for treating idiopathic pulmonary fibrosis, a fusion molecule for the idiopathic
pulmonary fibrosis treatment is a polypeptide having the structure ApoAl-L1-D (e.g., ApoAl-L1-[Fc
region]), or a dimeric protein formed by dimerization of the foregoing fusion polypeptide; in some
such embodiments, the fusion polypeptide comprises or consists of an amino acid sequence having at
least 90%, at least 95%, or 100% identity with (i) amino acid residues 19-525, 19-524, 25-525, or 25
524 of SEQ ID NO:2, (ii) amino acid residues
19-525, 19-524, 25-525, or 25-524 of SEQ ID NO: 13,
(iii) amino acid residues 19-501, 19-500, 25-501, or 25-501 of SEQ ID NO:20, (iv) amino acid
residues 19-515, 19-514, 25-515, or 25-514 of SEQ ID NO:22, or (v) amino acid residues 19-535, 19
534, 25-535, or 25-534 of SEQ ID NO:24. In some particular variations of a method for treating an
inflammatory lung disease in a patient that has been exposed to sulfur mustard gas (SM) or an
organophosphate, a fusion molecule for the treatment is a polypeptide having the structure ApoAl
L1-D-L2-paraoxonase (e.g., ApoAl-L1-[Fc region]-L2-PON1), or a dimeric protein formed by
dimerization of the foregoing fusion polypeptide; in some such embodiments, the fusion polypeptide
comprises or consists of an amino acid sequence having at least 90%, at least 95%, or 100% identity
with (i) amino acid residues 19-883 or 25-883 of SEQ ID NO:28, (ii) amino acid residues 19-873 or
-873 of SEQ ID NO:38, (iii) amino acid residues 19-883 or 25-883 of SEQ ID NO:46, or (iv)
amino acid residues 19-883 or 25-883 of SEQ ID NO:48. In some particular variations of a method
for treating acute respiratory distress syndrome (ARDS), hypoxia, or hyperoxia, a fusion molecule for
the treatment is a polypeptide having the structure ApoAl-L1-D-L2-RNase (e.g., ApoAl-L1-[Fc
region]-L2-RNasel), or a dimeric protein formed by dimerization of any of the foregoing fusion
polypeptides; in some such embodiments, the fusion polypeptide comprises or consists of an amino
acid sequence having at least 90%, at least 95%, or 100% identity with (i) amino acid residues 19-675
or 25-675 of SEQ ID NO:4, or (ii) amino acid residues 19-657 or 25-675 of SEQ ID NO:14. In
certain embodiments, such ApoAl-L1-D-L2-RNase variations are used to treat premature infants that
are treated with oxygen for an extended period of time.
In some embodiments, a fusion molecule for treatment of exposure to sulfur mustard
gas (SM) or exposure to an organophosphate is a polypeptide having the structure ApoAl-L1-D-L2
paraoxonase (e.g., ApoAl-L1-[Fc region]-L2-PON1), or a dimeric protein formed by dimerization of
the foregoing fusion polypeptide; in some such embodiments, the fusion polypeptide comprises or
amino acid sequence having at least 90%, at least 95%, or 100% identity with (i) amino
consists of an
acid residues 19-883 or 25-883 of SEQ ID NO:28, (ii) amino acid residues 19-873 or 25-873 of SEQ
ID NO:38, (iii) amino acid residues 19-883 or 25-883 of SEQ ID NO:46, or (iv) amino acid residues
19-883 or 25-883 of SEQ ID NO:48.
Infectious diseases amenable to treatment in accordance with the present invention
include, for example, bacterial infections and parasitic infections. In some embodiments, the parasitic
infection is a Trypanosoma brucei or Leishmania infection. In other embodiments, the bacterial
infection is a Pseudomonas aeruginosa infection.
In some embodiments of a method
for treating a Pseudomonas aeruginosa infection,
a fusion molecule for the Pseudomonas aeruginosa infection treatment is a polypeptide having the
structure ApoAl-L1-D-L2-paraoxonase (e.g., ApoAl-L1-[Fc region]-L2-PON1), or a dimeric protein
formed by dimerization of the foregoing fusion polypeptide; in some such embodiments, the fusion
polypeptide comprises or consists of an amino acid sequence having at least 90%, at least 95%, or
100% identity with (i) amino acid residues 19-883 or 25-883 of SEQ ID NO:28, (ii) amino acid
residues 19-873 or 25-873 of SEQ ID NO:38, (iii) amino acid residues 19-883 or 25-883 of SEQ ID
NO:46, or (iv) amino acid residues 19-883 or 25-883 of SEQ ID NO:48.
In some embodiments, a fusion molecule for treatment of an infectious
disease (e.g.,
an inflammatory lung disease) is a polypeptide having the structure ApoAl-L1-D (e.g., ApoAl-L1
[Fc region]), or a dimeric protein formed by dimerization of the foregoing fusion polypeptide; in some
such embodiments, the fusion polypeptide comprises or consists of an amino acid sequence having at
least 90%, at least 95%, or 100% identity with (i) amino acid residues 19-525, 19-524, 25-525, or 25
524 of SEQ ID NO:2, (ii) amino acid residues 19-525, 19-524,
-525, or 25-524 of SEQ ID NO:13,
(iii) amino acid residues 19-501, 19-500, 25-501, or 25-501 of SEQ ID NO:20, (iv) amino acid
residues 19-515, 19-514, 25-515, or 25-514 of SEQ ID NO:22, or (v) amino acid residues 19-535, 19
534, 25-535, or 25-534 of SEQ ID NO:24.
Cancers that may be treated in accordance with the present invention include, for
example, the following: a cancer of the head and neck
(e.g., a cancer of the oral cavity, orophyarynx,
nasopharynx, hypopharynx, nasal cavity or paranasal sinuses, larynx, lip, or salivary gland); a lung
cancer (e.g., non-small cell lung cancer, small cell carcinoma, or mesothelimia); a gastrointestinal
tract cancer (e.g., colorectal cancer, gastric cancer, esophageal cancer, or anal cancer); gastrointestinal
stromal tumor (GIST); pancreatic adenocarcinoma; pancreatic acinar cell carcinoma; a cancer of the
small intestine; a cancer of the liver
or biliary tree (e.g., liver cell adenoma, hepatocellular carcinoma,
hemangiosarcoma, extrahepatic or intrahepatic cholangiosarcoma, cancer of the ampulla of vater, or
gallbladder cancer); a breast cancer (e.g., metastatic breast cancer or inflammatory breast cancer); a
gynecologic cancer (e.g., cervical cancer, ovarian cancer, fallopian tube
cancer, peritoneal carcinoma,
vulvar cancer, gestational trophoblastic neoplasia, or uterine cancer, including
vaginal cancer,
endometrial cancer or uterine sarcoma); a cancer of the urinary tract (e.g., prostate cancer; bladder
cancer; penile cancer; urethral cancer, or kidney cancer such as, for example, renal cell carcinoma or
transitional cell carcinoma, including renal pelvis and ureter); testicular cancer; a cancer of the central
nervous system (CNS) such as
an intracranial tumor (e.g., astrocytoma, anaplastic astrocytoma,
glioblastoma, oligodendroglioma, anaplastic oligodendroglioma, ependymoma, primary CNS
lymphoma, medulloblastoma, germ cell tumor, pineal gland neoplasm,
meningioma, pituitary tumor,
tumor of the nerve sheath (e.g., schwannoma), chordoma, craniopharyngioma, a chloroid plexus
tumor (e.g., chloroid plexus carcinoma); or other intracranial tumor of neuronal or glial origin) or a
tumor of the spinal cord (e.g., schwannoma, meningioma); an endocrine neoplasm (e.g., thyroid
cancer such as, for example, thyroid carcinoma, medullary cancer, or thyroid lymphoma; a pancreatic
endocrine tumor such as, for
example, an insulinoma or glucagonoma; an adrenal carcinoma such as,
for example, pheochromocytoma; a carcinoid
tumor; or a parathyroid carcinoma); a skin cancer (e.g.,
squamous cell carcinoma; basal cell carcinoma;
Kaposi's sarcoma; or a malignant melanoma such as,
for example, an intraocular melanoma); a bone cancer (e.g.,
a bone sarcoma such as, for example,
osteosarcoma, osteochondroma, or Ewing's sarcoma); multiple myeloma; a chloroma; a soft tissue
sarcoma (e.g., a fibrous tumor or fibrohistiocytic tumor); a tumor of the smooth muscle or skeletal
muscle; a blood or lymph vessel perivascular tumor (e.g., Kaposi's sarcoma); a synovial tumor; a
mesothelial tumor; a neural tumor; a paraganglionic tumor; an extraskeletal cartilaginous or osseous
tumor; and a pluripotential mesenchymal tumor. In some such embodiments, an ApoA-1 fusion
molecule as described herein is administered to a cancer patient as one of the distinct therapies of a
combination therapy such as, for example, a combination therapy comprising a non-ApoAl-mediated
immunomodulatory therapy (e.g., a therapy comprising an immune checkpoint inhibitor), a radiation
therapy, or a chemotherapy.
In certain embodiments, a combination cancer therapy comprises an ApoA-i fusion
molecule as described herein and a targeted therapy such as, e.g., a therapeutic monoclonal antibody
targeting a specific cell-surface or extracellular antigen, or a small molecule targeting an intracellular
protein (e.g., an intracellular enzyme). Exemplary antibody targeted therapies include anti-VEGF
(e.g.,
bevacizumab), anti-EGFR (e.g., cetuximab), anti-CTLA-4 (e.g., ipilimumab),
anti-PD-1 (e.g.,
nivolumab), and anti-PD-Li (e.g., pembrolizumab). Exemplary small molecule targeted therapies
include proteasome inhibitors (e.g., bortezomib), tyrosine kinase inhibitors (e.g., imatinib), cyclin
dependent kinase inhibitors (e.g., seliciclib); BRAF inhibitors (e.g., vemurafenib or dabrafenib); and
MEK kinase inhibitors (e.g., trametnib).
In some cancer combination therapy variations comprising an immune checkpoint
inhibitor, the combination therapy includes an anti-PD-i/PD-Li therapy, an anti-CTLA-4 therapy, or
aspects, ApoA-I fusion molecules as described herein can increase the response rate
both. In certain
to either anti-CTLA-4 or anti-PD-i/PD-Li therapy, as well as the response rate to the combination of
anti-CTLA-4 plus anti-PD-i/PD-Li therapy. Fusion molecules of the invention may also be useful
for reducing the toxicity associated with anti-CTLA-4, anti-PD-i/PD-Li, or the combination thereof.
In certain variations, a cancer treated in accordance with the present invention is
selected from
malignant melanoma, renal cell carcinoma, non-small cell lung cancer,
bladder cancer,
and head and neck cancer. These cancers have shown responses to immune checkpoint inhibitors
anti-PD-i/PD-Li and anti-CTLA-4. See Grimaldi et al., Expert Opin. Biol. Ther. 16:433-41, 2016;
Gunturi et al., Curr. Treat. Options Oncol. 15:137-46, 2014; Topalian et al., Nat. Rev. Cancer 16:275
87, 2016. Thus, in some more specific variations, any of these cancers is treated with an ApoA-i
fusion molecule as described herein in combination with an anti-PD-i/PD-Li therapy, an anti-CTLA
4 therapy, or both.
In some embodiments, a fusion molecule for treatment of a cancer is a polypeptide
having the structure ApoA1-Li-D
(e.g., ApoAl-Li-[Fc region]), ApoAl-Li-D-L2-RNase
(e.g.,
ApoAl-L1-[Fc region] -L2-RNase i), or ApoAl-Li-D-L2-paraoxonase (e.g., ApoAl-L1-[Fc region]
L2-PONi), or a dimeric protein formed by dimerization of any of the foregoing fusion polypeptides;
in some such embodiments, the fusion polypeptide comprises or consists of an amino acid sequence
having at least 90%, at least 95%, or 100% identity with (i) amino acid residues 19-525, 19-524, 25
525, or 25-524 of SEQ ID NO:2, (ii) amino acid residues 19-525, 19-524, 25-525, or 25-524 of SEQ
ID NO:13, (iii) amino acid residues 19-501, 19-500, 25-501, or 25-501 of SEQ ID NO:20, (iv) amino
acid residues 19-515, 19-514,
-515, or 25-514 of SEQ ID NO:22, (v) amino acid residues
19-535,
19-534, 25-535, or 25-534 of SEQ ID NO:24, (vi) amino acid residues 19-675 or 25-675 of SEQ ID
NO:4, (vii) amino acid residues 19-657 or 25-675 of SEQ ID NO:14, (viii) amino acid residues 19
883 or 25-883 of SEQ ID NO:28, (ix) amino acid residues 19-873 or 25-873 of SEQ ID NO:38, (x)
amino acid residues 19-883 or 25-883 of SEQ ID NO:46, or (xi) amino acid residues 19-883 or 25
883 of SEQ ID NO:48.
For therapeutic use, a fusion polypeptide or dimeric protein as described herein is
delivered in a manner consistent with conventional methodologies associated with management of the
disease or disorder for which treatment is sought. In accordance with the disclosure herein, an
effective amount of the fusion polypeptide or dimeric protein is administered to a subject in need of
such treatment for a time and under conditions sufficient to prevent or treat the disease or disorder.
Subjects for administration of fusion polypeptides or dimeric proteins as described
herein include patients at high risk for developing a particular disease or disorder as well as patients
presenting with an existing disease or disorder. In certain embodiments, the subject has been
diagnosed as having the disease or disorder for which treatment is sought. Further, subjects can be
the course of treatment for any change in the disease or disorder (e.g., for an
monitored during
increase or decrease in clinical symptoms of the disease or disorder). Also, in some variations, the
subject does not suffer from another disease or disorder requiring treatment that involves
administration of an ApoA- 1 protein.
In prophylactic applications, pharmaceutical compositions or medicants are
administered to a patient susceptible to, or otherwise at risk of, a particular disease in an amount
sufficient to
eliminate or reduce the risk or delay the outset of the disease. In therapeutic
applications,
compositions or medicants are administered to a patient suspected of, or already suffering from such a
disease in an amount sufficient to cure, or at least partially arrest, the symptoms of the disease and its
complications. An amount adequate to accomplish this is referred to as a therapeutically or
pharmaceutically effective dose or amount. In both prophylactic and therapeutic regimes, agents are
usually administered in several dosages until a sufficient response (e.g., atherosclerosis regression or
stabilization of existing plaques in coronary heart disease) has been achieved. Typically, the response
is monitored and repeated dosages are given if the desired response starts to fade.
To identify subject patients for
treatment according to the methods of the invention,
accepted screening methods may be employed to determine risk factors associated with a specific
disease or to determine the status of an existing disease identified in a subject. Such methods can
include, for example, determining whether an individual has relatives who have been diagnosed with
a particular disease. Screening methods can also include, for example, conventional work-ups to
determine familial status for a particular disease known to have a heritable component. Toward this
end, nucleotide probes can be routinely employed to identify individuals carrying genetic markers
associated with a particular disease of interest. In addition, a wide variety of immunological methods
are known in the art that are useful to identify markers for specific diseases. Screening may be
implemented as indicated by known patient symptomology, age factors, related risk factors, etc.
These methods allow the clinician to routinely select patients in need of the methods described herein
for treatment. In accordance with these methods, treatment using a fusion polypeptide or dimeric
protein of the present invention may be implemented as an independent treatment program or as a
follow-up, adjunct, or coordinate treatment regimen to other treatments.
For administration, a fusion polypeptide or dimeric protein in accordance with the
present invention is formulated as a pharmaceutical composition. A pharmaceutical composition
comprising a fusion polypeptide or dimeric protein as described herein can be formulated according to
known methods to prepare pharmaceutically useful compositions, whereby the therapeutic molecule is
combined in a mixture with a pharmaceutically acceptable carrier. A composition is said to be a
"pharmaceutically
acceptable
carrier" if its administration
can be
tolerated by a recipient
patient.
Sterile phosphate-buffered saline is one example of a pharmaceutically acceptable carrier. Other
are well-known to those in the art. See, e.g., Gennaro (ed.), Remington's
suitable carriers
Pharmaceutical Sciences (Mack Publishing Company, 19th ed. 1995). Formulations may further
include one or more excipients, preservatives, solubilizers, buffering agents, albumin to prevent
protein loss on vial surfaces, etc.
A pharmaceutical composition comprising a fusion polypeptide or dimeric protein of
the present invention is administered to a subject in an effective amount. The fusion polypeptide or
dimeric protein may be administered to subjects by a variety of administration modes, including, for
example, by
intramuscular, subcutaneous, intravenous, intra-atrial, intra-articular,
parenteral,
intranasal, intrapulmonary, transdermal, intrapleural, intrathecal, and oral routes of administration.
For prevention and treatment purposes, the fusion polypeptide or dimeric protein may be administered
to a subject in a single bolus delivery, via continuous delivery (e.g., continuous transdermal delivery)
over an extended time period, or in a repeated administration protocol (e.g., on an hourly, daily, or
weekly basis).
Determination of effective dosages in this context is typically based on animal model
studies followed up by human clinical trials and is guided by determining effective dosages and
administration protocols that significantly reduce the occurrence or severity of the subject disease or
disorder in model subjects. Effective doses of the compositions of the present invention vary
depending upon many different factors, including means of administration, target site, physiological
state of
the patient, whether the patient is human or an animal, other medications
administered,
whether treatment is prophylactic or therapeutic, as well as the specific activity of the composition
itself and its ability to
elicit the desired response in the individual. Usually, the patient is a
human,
but in some diseases, the patient can be a nonhuman mammal. Typically, dosage regimens are
adjusted to provide an optimum therapeutic response, i.e., to optimize safety and efficacy.
Accordingly, a therapeutically or prophylactically effective amount is also one in which any undesired
collateral effects are outweighed by beneficial effects (e.g., in the case of treatment of atherosclerotic
cardiovascular disease, where any undesired collateral effects are outweighted by any beneficial
effects such as increase in HDL, atherosclerosis regression, and/or plaque stabilization). For
administration of a fusion polypeptide or dimeric protein of the present invention, a dosage typically
ranges from about 0.1 gg to 100 mg/kg or 1 jg/kg to about 50 mg/kg, and more usually 10 jg to 5
mg/kg of the subject's body weight. In more specific embodiments, an effective amount of the agent
is between about 1 jg/kg and about 20 mg/kg, between about 10 jg/kg and about 10 mg/kg, or
between about 0.1 mg/kg and about 5 mg/kg. Dosages within this range can be achieved by single or
multiple administrations, including, e.g., multiple
administrations per day or daily, weekly, bi-weekly,
or monthly administrations. For example, in certain variations, a regimen consists of an initial
administration followed by multiple, subsequent administrations at weekly or bi-weekly intervals.
consists of an initial administration followed by multiple, subsequent
Another regimen
administrations at monthly or bi-monthly intervals. Alternatively, administrations can be on an
irregular basis as indicated by monitoring of clinical symptoms of the disease or disorder and/or
monitoring of disease biomarkers or other disease correlates (e.g., HDL levels in the case of
atherosclerotic cardiovascular disease).
Particularly suitable animal models for evaluating efficacy of an ApoA-1 composition
of the present invention for treatment of atherosclerosis include, for example, known mouse models
that are deficient in the low density lipoprotein receptor (LDLR) or ApoE. LDLR deficient mice
develop atherosclerotic plaques after eating a high fat diet for 12 weeks, and human ApoA-1
(reconstituted with lipids) is effective in reducing plaques in this model. ApoE deficient mice are also
commonly used to study atherosclerosis, and human ApoA-1 (reconstituted with lipids) works rapidly
in this model. Rabbits that are transgenic for hepatic lipase are another known atherosclerosis model
for testing ApoA-1 compositions.
One model of Alzheimer's disease uses overexpression of mutant amyloid-P precursor
protein (APP) and presenilin 1 in mice. In these mice, overexpression of human ApoA-1 prevented
memory and learning deficits. See Lewis et al., J Biol. Chem. 285: 36958-36968, 2010.
Also known is the collagen-induced arthritis (CIA) model for rheumatoid arthritis
(RA). CIA shares similar immunological and pathological features with RA, making it an ideal model
for evaluating efficacy of ApoA-1 compositions. See, e.g., Charles-Schoeman et al., Clin Immunol.
127:234-44, 2008 (describing studies showing efficacy of the ApoA-1 mimetic peptide, D-4F, in the
CIA model). Another known model for RA is PG-polysaccharide (PG-PS)-induced arthritis in female
Lewis rats. In these mice, administration of ApoA- 1 protein or reconstituted HDLs reduced acute and
chronic joint inflammation. Wu et al., Arterioscler Thromb Vasc Biol 34:543-551, 2014.
Animal models for multiple sclerosis (MS) include, for example, experimental
allergic encephalomyelitis (EAE) models that rely on the induction of an autoimmune response in the
CNS by immunization with a CNS antigen (also referred to as an "encephalitogen" in the context of
EAE), which leads to inflammation, demyelination, and weakness. ApoA- 1 deficient mice have been
shown to exhibit more neurodegeneration and worse disease than wild-type animals in this model.
See Meyers et al., J. Neuroimmunol. 277: 176-185, 2014.
Fusion molecules of the present invention can be evaluated for anti-tumor activity in
animal tumor models such as, e.g., B16 melanoma, a poorly immunogenic tumor. Multiple models of
tumor immunotherapy have been studied. See Ngiow et al., Adv. Immunol. 130:1-24, 2016. The B16
melanoma model has been studied extensively with checkpoint inhibitors
anti-CTLA-4, anti-PD-1,
and the combination thereof. Anti-CTLA-4 alone has a potent therapeutic effect in this model only
when combined with GM-CSF transduced tumor vaccine, or combined
with anti-PD-1. See Weber,
Semin. Oncol. 37:430-439, 2010; Ai et al., Cancer Immunol. Immunother. 64:885-92, 2015; Haanen
et al., Prog. Tumor Res. 42:55-66, 2015. Efficacy of an ApoA-1 fusion molecule for treatment of
malignant melanoma is shown, for example, by slowed tumor growth following administration to B16
melanoma mice that have formed palpable subcutaneous tumor nodules. Efficacy of an ApoA-1
fusion molecule can be evaluated in B16 melanoma mice either alone or, alternatively, in combination
with another anti-cancer therapy (e.g., anti-CTLA-4, with or without tumor vaccine or with or without
anti-PD-i/PD-Li). For example, tumor rejection in B16 melanoma mice using a combination of an
ApoA-1 fusion molecule as described herein and anti-CTLA-4,
in the absence of tumor vaccine,
demonstrates an enhanced response to anti-CTLA-4 using the ApoA-1 therapy. In exemplary studies
to evaluate ApoA-1 fusion molecules comprising human protein sequences, which are functionally
active in mice but are expected to be immunogenic in these models (and thereby likely to result in
formation of neutralizing antibodies after 7-10 days), mice may be administered a fusion molecule of
the present invention for a short period (for example, one week, administered in, e.g., two doses of
about 40mg/kg three days apart), and tumor growth then monitored, typically for two to three weeks
after injection with the fusion molecule.
Dosage of the pharmaceutical composition may be varied by the attending clinician to
maintain a desired concentration at a target site. For example, if an intravenous mode of delivery is
selected, local concentration of the agent in the bloodstream at the target tissue may be between about
1-50 nanomoles of the composition per liter, sometimes between
about 1.0 nanomole per liter and 10,
, or 25 nanomoles per liter depending on the subject's status and projected measured response.
Higher or lower concentrations may be selected based on the mode of delivery, e.g., trans-epidermal
delivery versus delivery to a mucosal surface. Dosage should also be adjusted based on the release
rate of the administered formulation, e.g., nasal spray versus powder, sustained release oral or injected
particles, transdermal formulations, etc.
To achieve the same serum concentration level, for example,
slow-release particles with a release rate of 5 nanomolar (under standard conditions) would be
administered at about twice the dosage of particles with a release rate of 10 nanomolar.
A pharmaceutical composition comprising a fusion polypeptide or dimeric protein as
described herein can be furnished in liquid form, in an aerosol, or in solid form. Liquid forms, are
illustrated by injectable solutions, aerosols, droplets, topological solutions and oral suspensions.
Exemplary solid forms include capsules, tablets, and controlled-release forms. The latter form is
illustrated by miniosmotic pumps and implants. See,
e.g., Bremer et al., Pharm. Biotechnol. 10:239,
1997;
Ranade, "Implants in Drug Delivery," in Drug Delivery Systems 95-123
(Ranade and Hollinger,
eds., CRC Press 1995); Bremer et al., "Protein Delivery with Infusion Pumps," in Protein Delivery:
Physical Systems 239-254 (Sanders and Hendren, eds., Plenum Press 1997); Yewey et al., "Delivery
a Controlled Release Injectable Implant," in Protein Delivery: Physical Systems 93
of Proteins from
117 (Sanders and Hendren, eds., Plenum Press 1997). Other solid forms include creams, pastes, other
topological applications, and the like.
Degradable polymer microspheres have been designed to maintain high systemic
levels of therapeutic proteins. Microspheres are prepared from degradable polymers such as
poly(lactide-co-glycolide) (PLG), polyanhydrides, poly (ortho esters), nonbiodegradable ethylvinyl
acetate polymers,
in which proteins are entrapped in the polymer. See, e.g., Gombotz
and Pettit,
Bioconjugate Chem. 6:332, 1995; Ranade, "Role of Polymers in Drug Delivery," in Drug Delivery
Systems 51-93 (Ranade and Hollinger, eds., CRC Press 1995); Roskos and Maskiewicz, "Degradable
Controlled Release Systems Useful for Protein Delivery," in Protein Delivery: Physical Systems 45
92 (Sanders and Hendren, eds., Plenum Press 1997); Bartus et al., Science 281:1161, 1998; Putney
and Burke, Nature Biotechnology 16:153, 1998; Putney, Curr. Opin. Chem. Biol. 2:548, 1998.
Polyethylene glycol (PEG)-coated nanospheres can also provide carriers for intravenous
administration of therapeutic proteins. See, e.g., Gref et al., Pharm. Biotechnol. 10:167, 1997.
Other dosage forms can be devised
by those skilled in the art, as shown by, e.g.,
Ansel and Popovich, Pharmaceutical Dosage Forms and Drug Delivery Systems (Lea & Febiger, 5th
ed. 1990); Gennaro (ed.), Remington's Pharmaceutical Sciences (Mack Publishing Company, 19th ed.
1995), and Ranade and Hollinger, Drug Delivery Systems (CRC Press 1996).
Pharmaceutical compositions as described herein may also be used in the context of
combination therapy. The term "combination therapy" is used herein to denote that a subject is
administered at least one therapeutically effective dose of a fusion polypeptide or dimeric protein as
described herein and another therapeutic agent.
Pharmaceutical compositions may be supplied as a kit comprising a container that
comprises a fusion polypeptide or dimeric protein as described herein. A therapeutic molecule can be
provided, for example, in the form of an injectable solution for single or multiple doses, or as a sterile
powder that will be reconstituted before injection. Alternatively, such a kit can include a dry-powder
disperser, liquid aerosol generator, or nebulizer for administration of a therapeutic protein. Such a kit
may further comprise written information on indications and usage of the pharmaceutical
composition.
The invention is further illustrated by the following non-limiting examples.
Example 1
Molecule Design and Preparation: Two ApoAFc cDNA constructs were
designed, synthesized, expressed by transient transfection of COS7 cells, and the expressed proteins
then purified by Protein A chromatography. One construct had the nucleotide sequence shown in
SEQ ID NO:1 and encoded the fusion polypeptide of SEQ ID NO:2, and is also referred to herein as
ApoA-1(26)Fc or THER4. This construct contained a DNA segment encoding a 26 amino acid linker
(residues 268-293 of SEQ ID NO:2) between the C-terminal end of human ApoA-1 (residues 1-267 of
SEQ ID NO:2) and a human 71 Fc variant (residues 294-525 of SEQ ID NO:2). Upon expression in
mammalian cells and cleavage of the secretory signal peptide (residues 1-18), and any potential
cleavage of the propeptide (residues 19-24), this fusion polypeptide had a predicted amino acid
sequence corresponding to residues 19-525, 19-524, 25-525, or 25-524 of SEQ ID NO:2 (the C
terminal lysine of the Fc region is known to be frequently cleaved in the production of Fc-containing
proteins). The other construct contained ApoA-1 and Fc regions identical to those of the ApoA
1(26)Fc construct, but lacked a (gly4ser) linker between human ApoA-1 and the Fc regions; this
construct is also referred to herein as ApoA-1(2)Fc (Theripion) or as THERO (for no (gly4ser) repeat
units). This construct does contain a two amino acid linker due to insertion of overlapping restriction
sites between the ApoA- 1 region and the hinge region of human IgG 1.
Cholesterol efflux: The cholesterol efflux activity of the ApoA-I fusion proteins were
measured using an in vitro assay. See Tang et al., J Lipid Res. 47:107-14, 2006. In vitro cholesterol
efflux assays were performed using radio-labelled cholesterol and BHK cells expressing a
mifespristone-inducible human ABCA1. H3-cholesterol was added to growth media in order to label
cellular cholesterol 24 hours prior to treatments, and ABCA1 is induced using lOnM mifepristone for
16-20 hours. Cholesterol efflux was measured by incubating cells with or without the fusion proteins
for 2 hours at 37'C, chilled on ice, and medium and cells separated to measure radiolabeled
cholesterol. Wild-type human ApoA-1 protein was used as a positive control. A commercially
available ApoAFc protein, linked directly to Fc without any linker between the ApoA-1 and Fc
regions (APOA1 Recombinant Human Protein, hIgG1-Fc tag; Sino Biological, Inc.), was also tested
and is referred to herein as ApoA-1(0)Fc (Sino Biol). The results of this assay are shown in
Cholesterol efflux was increased in cultures containing ApoAFc with a 26 amino acid linker
(ApoA-1(26)Fc), compared to either ApoAFc with a two amino acid linker (ApoA-1(2)Fc
(Theripion)) or ApoAFc without a linker (ApoA-1(0)Fc (Sino Biol)). ApoA-1(26)Fc also had
activity similar to wild-type human ApoA-1 (Control ApoA-1).
Example 2: Generation of Fusion Constructs and Sequence Verification
Additional ApoAl fusion constructs were designed and the fusion gene sequences
were submitted to Blue Heron (Bothell, WA) for gene synthesis. A basic schematic diagramming the
position of functional domains is shown in FIGS. 2A and 2B for the design of the ApoAl fusion
proteins. Fusion gene constructs inserted into pUC-based vectors isolated by restriction enzyme
digestion, and fragments encoding the fusion genes were subcloned into the mammalian expression
vector pDG. Briefly, HindIll+XbaI flanking restriction sites were used for removal of each
expression gene from the vector, subfragments isolated by gel electrophoresis, DNA extracted using
QlAquick purification columns, and eluted in 30 microliters EB buffer. Fragments were ligated into
Hindll+XbaI digested pDG vector, and ligation reactions transformed into NEB 5-alpha, chemically
competent bacteria. Clones were inoculated into 3 ml LB broth with 100 pg/ml ampicillin, grown at
37'C overnight with shaking at 200 rpm, and plasmid DNA prepared using the QIAGEN spin plasmid
miniprep kits according to manufacturer's instructions. Sequencing primers were obtained from IDT
Integrated DNA Technologies (Coralville, IA) and included the following:
pdgF-2: 5'-ggttttggcagtacatcaatgg-3' (SEQ ID NO:16);
pdgR-2: 5'-ctattgtcttcccaatcctccc-3' (SEQ ID NO:17);
higgras: 5'-accttgcacttgtactcctt-3' (SEQ ID NO:18).
Plasmid DNA (800 ng) and sequencing primers (25 pmol, or 5 pl of a 5 pmol/pl
stock) were mixed and submitted for DNA sequencing by GENEWIZ (South Plainfield, NJ).
Chromatograms were then analyzed, sequences assembled into contigs, and sequence verified using
Vector Nti Advance 11.5 software (Life Technologies, Grand Island, NY).
Example 3: Expression of Fusion Proteins in a Transient HEK 293T Transfection System
This example illustrates transfection of plasmid constructs and expression of fusion
proteins described herein in a mammalian transient transfection system. The Ig fusion gene fragments
with correct sequence were inserted into the mammalian expression vector pDG, and DNA from
positive clones was amplified using QIAGEN plasmid preparation kits (QIAGEN, Valencia, CA).
Five different constructs were generated. These each included the native coding sequence of the
human ApoA-1 gene (nucleotide sequence shown in SEQ ID NO:35, encoding the amino acid
sequence shown in SEQ ID NO:36). Each sequence included the wild-type signal peptide
(nucleotides 1-54 of SEQ ID NO:35, encoding amino acids 1-18 of SEQ ID NO:36) and propeptide
sequences (nucleotides 55-72 of SEQ ID NO:35, encoding amino acids 19-24 of SEQ ID NO:36) for
apolipoprotein A-1. The C-terminal Q (Gln) residues of the ApoA-1 sequence was linked via a
linker segment to the human IgGI hinge, CH2, and CH3 domains to create a single
variable length
chain (ApoA-1)-Ink-human IgG1 Fc fusion gene/protein. The hinge sequence of the human IgG1 is
mutated so that the three cysteines are substituted with serine residues, eliminating disulfide bond
formation in this region or unpaired cysteines that might compromise proper folding of the fusion
protein. The P238 and P331 residues of CH2 are also mutated to serines to eliminate effector
functions such as ADCC and complement fixation. Each construct also included a linker sequence
inserted between the carboxyl terminus of apolipoprotein A-i (ending with the sequence
... TKKLNTQ (SEQ ID NO:35 residues 261-267) ) and the beginning of the hinge sequence of the
human Fc (starting with the motif ...EPKSSDKT... (SEQ ID NO:2 residues 294-301). This linker
sequence ranged from two amino acids (or four if the overlap with the flanking domains is included)
to 36 amino acids in length, depending on the construct.
The shortest linker included only two overlapping restriction sites (BglII and XhoI)
with a linker length of six additional nucleotides or two additional non-native amino acids. The
restriction sites were incorporated into the coding sequence of the molecule so that only two
additional amino acids needed to be added to the amino acid sequence. The BglII site of the linker
overlaps with the codon for the C-terminal glutamine of ApoA-1, and three of the nucleotides
encoding the XhoI site form the codon for the first amino acid of the hinge (E-glutamic acid). The
linker amino acid sequence (including the two overlapping amino acids) is shown in residues 267-270
of SEQ ID NO:20, which is encoded by nucleotides 816-825 of SEQ ID NO:19. The fusion gene and
protein for this construct are identified as THERO (since there are no (gly4ser) repeat units present) or
apoAlnk(2)hIgG. The nucleotide and amino acid sequences for THERO are listed as SEQ ID
NO:19 and SEQ ID NO:20. The figures use the THERO nomenclature to specify this construct.
The second construct included a linker that encodes two (gly4ser) sequences flanked
by restriction sites (16 amino acid linker), and the fusion gene and protein for this construct are
identified as THER2 (or apoAlnk(16)-hIgGl or apoA(g4s)2-hIgGl). The nucleotide and amino
acid sequences for THER2 are listed as SEQ ID NO:21 and SEQ ID NO:22, respectively. The
(gly4ser)2 linker sequence is shown in residues 268-283 of SEQ ID NO:22, and the encoding
nucleotide sequence for the (gly4ser)2 linker is shown in residues 817-864 of SEQ ID NO:21.
The third construct included a linker that encodes four (gly4ser) sequences flanked by
restriction sites (26 amino acid linker), and the fusion gene and protein for this construct is identified
as THER4 (or apoA(g4s)4-mthIgG or apoAlnk(26)-mthlgG), where "4" in "THER4" refers to
the number of (gly4ser) repeat units, and the number 26 refers to the total number of amino acids
encoded in the non-native, introduced linker sequence. The nucleotide and amino acid sequences for
THER4 are listed as SEQ ID NO:1 and SEQ ID NO:2, respectively. The (gly4ser)4 linker sequence is
ID NO:50 (residues 268-293 of SEQ ID NO:2), and the encoding nucleotide sequence
shown in SEQ
for the (gly4ser)4 linker is shown in SEQ ID NO:49 (residues 817-894 of SEQ ID NO: 1).
The fourth construct included a linker that encodes six (gly4ser) sequences flanked by
restriction sites (36 amino acid linker), and the fusion gene and protein for this construct is identified
as THER6 (or apoA(g4s)6-mthIgG or apoAlnk(36)-mthIgG). The nucleotide and amino acid
sequences for THER6 are listed as SEQ ID NO:23 and SEQ ID NO:24, respectively. The (gly4ser)6
linker sequence is shown in SEQ ID NO:52 (residues 268-303 of SEQ ID NO:24), and the encoding
nucleotide sequence for the (gly4ser)6 linker is shown in SEQ ID NO:51 (residues 817-924 of SEQ
ID NO:23).
The fifth construct included a linker that encodes four (gly4ser) sequences flanked by
restriction sites (36 amino acid linker), but in addition, the construct included a second linker and an
enzyme sequence at the carboxyl terminus of the IgG1 domain. The (gly4ser)4 linker sequence is as
described above for THER4 (nucleotide and amino acid sequences shown in SEQ ID NO:49 and SEQ
ID NO:50, respectively). The second linker is an 18 amino acid long sequence
(VDGASSPVNVSSPSVQDI; amino acid residues 1-18 of SEQ ID NO:8, encoded by nucleotides 1
54 of SEQ ID NO:7) that includes an N-linked glycosylation site, followed by a sequence that
encodes human RNasel enzyme activity. The linker sequence is listed as the first 54 nucleotides of
SEQ ID NO 7, or the first 18 amino acids of SEQ ID NO 8, followed by the RNase sequence. The
ApoAlnk-hIgGl segment is fused to the NLG-RNase, and this construct is identified as
THER4RNA2. The nucleotide and amino acid sequences are identified as SEQ ID NO:3 and SEQ ID
NO:4, respectively.
Miniprep DNA for each of the five constructs was prepared and the concentration
checked by Nanodrop analysis.
The day before transfection, approximately 1.2 x 106 293T cells were plated to 60
mm dishes. Mini-plasmid preparations (4.0 pg DNA for 60mm plates) were used for 293T
transfections using the QIAGEN POLYFECT® reagent (Catalog # 301105/301107) and following the
manufacturer's instructions. Culture supernatants were harvested 48-72 hours after transfection. For
most transfections, media was changed to serum-free media 24 hours after transfection, and cultures
incubated for a further 48 hours prior to harvest.
Culture supernatants were used directly for further analysis. 7 pl of each serum-free
culture supernatant from transiently transfected cells was loaded onto gels with a 4X dilution of 4X
LDS sample buffer (Life Technologies, Grand Island, NY) added to each sample to give a final
concentration of 1X LDS loading buffer. For reducing gels, sample reducing agent was added to 1/10
final volume. Samples were heated at 72 C for 10 minutes and loaded on NuPAGE@ 4-12% Bis-Tris
Scientific, Grand Island, NY). Gels were subjected to
gels (Life Technologies/ThermoFisher
electrophoresis in IX NuPAGE® MOPS SDS-PAGE running buffer (NP0001, Life
Technologies/ThermoFisher) at 180 volts for 1.5 hours, and proteins transferred to nitrocellulose
using the XCell IITM Blot Module (Catalog #EI002/EI9051, Life Technologies/ThermoFisher, Grand
Island, NY) at 30 volts for 1 hour. Blots were blocked overnight at 4 C in PBS containing 5% nonfat
milk. Blots were incubated with 1:250,000X dilution of horseradish peroxidase conjugated goat anti
human IgG (Jackson Immunoresearch, Catalog #109098, Lot# 122301). Blots were washed
three times for 30 minutes each in PBS/0.05% Tween 20, and were developed in ThermoScientific
ECL reagent (Catalog #32106) for 1 minute. Blots were exposed to autoradiograph film for 30
seconds to 2 minutes, depending on the blot. shows Western Blot analysis of culture
supernatants from representative 293T transient transfections. Positive and negative controls
(CD40IgG and mock transfection/no DNA, respectively) were included in each transfection series.
Transfected samples are as indicated in lanes from left to right are as follows: Lane #1
mock transfection; Lane #2 - CD40IgG; Lane #3 - MW markers; Lane #4 - THERO; Lane #5
THER2; Lane #6 - THER4; Lane #7 - THER6; Lane #8 - MW marker; Lane #9 - THER4RNA2.
The THERO, THER2, THER4, and THER6 fusion proteins run at a position above the
50 kDa molecular weight marker. The predicted molecular weight for these fusion proteins should be
approximately 55, 56, 56.6, and 57 kDa, respectively. The increasing linker length is evident by
altered mobility for each fusion protein. The THER4RNA2 molecule is predicted to be approximately
73.2 kDa, while ApoA-1 is predicted to run at 28.6 kDa. The CD40IgG control is expected to run at
approximately 55 kDa.
Example 4: Expression of THERmthlgG and Multi-subunitIe Fusion Constructs and Fusion
Proteins in Stable CHO Cell Lines
This example illustrates expression of the different Ig fusion genes described herein
in eukaryotic cell lines and characterization of the expressed fusion proteins by SDS-PAGE and by
IgG sandwich ELISA.
Transfection and selection of stable cell lines expressing fusion proteins
Stable production of the Ig fusion protein was achieved by electroporation of a
selectable, amplifiable plasmid, pDG, containing the THER-mthIgG cDNAs (human apo A-i forms
separated from the hinge and Fc domain of human IgG1 by linkers of varying lengths) under the
control of the CMV promoter, into Chinese Hamster Ovary (CHO) CHO DG44 cells.
The pDG vector is a modified version of pcDNA3 encoding the DHFR selectable
marker with an attenuated promoter to increase selection pressure for the plasmid. Plasmid DNA
maxiprep kits, and purified plasmid was
(200 pg) was prepared using QIAGEN HISPEED@
linearized at a unique AscI site (New England Biolabs, Ipswich, MA Catalog # R0558), purified by
phenol extraction (Sigma-Aldrich, St. Louis, MO), ethanol precipitated, washed, and resuspended in
EX-CELL® 302 tissue culture media, (Catalog #14324, SAFC/Sigma Aldrich, St. Louis, MO).
Salmon sperm DNA (Sigma-Aldrich, St. Louis, MO) was added as carrier DNA just prior to phenol
extraction and ethanol precipitation. Plasmid and carrier DNA were coprecipitated, and the 400 pg
was used to transfect 2x10 CHO DG44 cells by electroporation.
For transfection, CHO DG44 cells were grown to logarithmic phase in EX-CELL®
302 media (Catalog # 13424C,
SAFC Biosciences, St. Louis, MO) containing glutamine (4 mM),
pyruvate, recombinant insulin (1 pg/ml), penicillin-streptomycin, and 2xDMEM nonessential amino
acids (all from Life Technologies, Grand Island, NY), hereafter referred to as "EX-CELL 302
complete" media. Media for untransfected cells and cells to be transfected also contained HT (diluted
from a 100x solution of hypoxanthine and thymidine) (Invitrogen/Life
Technologies, Grand Island,
NY). Electroporations were performed at 280 volts, 950 microFarads, using a BioRad (Hercules, CA)
GENEPULSER@ electroporation unit with capacitance extender. Electroporation was performed in
0.4 cm gap sterile, disposable cuvettes. Electroporated cells were incubated for 5 minutes after
electroporation prior to transfer of culture to non-selective EX-CELL 302 complete media in T75
flasks.
Transfected cells were allowed to recover overnight in non-selective media prior to
selective plating in 96 well flat bottom plates (Costar) at varying serial dilutions ranging from 250
cells/well (2500 cells/ml) to 2000 cells/well (20,000 cells/ml). Culture media for cell cloning was
EX-CELL 302 complete media containing 50 nM methotrexate. Transfection plates were fed at five
day intervals with 80 pl fresh media. After the first couple of feedings, media was removed and
replaced with fresh media. Plates were monitored and individual wells with clones were fed until
clonal outgrowth became close to confluent, after which clones were expanded into 24 well dishes
containing 1 ml media. Aliquots of the culture supernatants from the original 96 well plate were
harvested to a second 96 well plate prior to transfer and expansion of the cells in 24 well plates. This
second plate was frozen until ELISA analysis to estimate IgG concentrations.
Screening culture supernatants for production levels of recombinant fusion proteins
Once clonal outgrowth of initial transfectants was sufficient, serial dilutions of culture
supernatants from master wells were thawed and the dilutions screened for expression of Ig fusion
protein by use of an IgG sandwich ELISA. Briefly, NUNC MAXISORP@ plates were coated
overnight at 4'C with 2 pg/ml F(ab'2) goat anti-human
IgG (Jackson Immunoresearch, West Grove,
PA; Catalog # 109098) in PBS. Plates were blocked in PBS/3% BSA, and serial dilutions of
culture supernatants incubated at room temperature for 2-3 hours or overnight at 4'C. Plates were
times in PBS/0.05 % Tween 20, and incubated with horseradish peroxidase conjugated
washed three
F(ab'2)goat anti-human IgG (Jackson Immunoresearch, West Grove, PA, Catalog # 109098) at
1:7500-1:10,000 in PBS/0.5% BSA, for 1-2 hours at room temperature. Plates were washed five
times in PBS/0.05% Tween 20, and binding detected with SUREBLUE RESERVETM TMB substrate
(KPL Labs, Gaithersburg, MD; catalog #5302). Reactions were stopped by addition of equal
volume of IN HCl, and absorbance per well on each plate was read at 450nM on a SYNERGYTM HT
plate reader (Biotek, Winooski, VT). Concentrations were estimated by comparing the OD450 of the
dilutions of culture supernatants to a standard curve generated using serial dilutions of a known
standard, a protein A purified human IgG fusion protein with an Ig tail identical to that of the THER
clones. Data was collected and analyzed using GEN5TM software (Biotek, Winooski, VT) and
Microsoft Office EXCEL@ spreadsheet software.
The results of initial screening of the CHO transfectants are summarized in Table 3
and FIGS. 4A-4E and 5A-5C. Table 3 shows a summary of the number of clones screened, the range
of expression levels observed from initial 96-well cultures, and the expression observed from initial
T25 and/or 24 well spent cultures. FIGS. 4A-4E show a series of columnar graphs representing the
production levels obtained from each CHO clone of a transfection series. The clones from the
THERO, THER2, THER4, THER6, and THER4RNA2 transfections are displayed as a group in each
of the five panels shown. Each clone was screened at least once by IgG sandwich ELISA to assess
expression level of the fusion protein. FIGS. 5A-5C show three panels showing the results of 6 and
day assays of fusion protein expression from the CHO transfectants with the highest expression
after initial screening. Six and ten day assays were performed by setting up 5 ml cultures at 1x10
viable cells/ml (5x10 initial inoculum) in T25 flasks. The cultures were grown for six days after
which a 1 ml aliquot was removed and live and dead cells counted. Cells were then centrifuged and
the culture supernatants saved for further analysis by IgG sandwich ELISA and other analyses. The
remainder of the cultures were incubated for a further four days until day 10, and the cells counted for
cell number, viability, and a supernatant sample harvested for IgG sandwich ELISA. The results are
tabulated in columnar form for each clone as shown in the
graphs for cell number at day 6 and day 10,
viability, and concentration of fusion protein.
Table 3: Expression of ApoA1-IgG fusion proteins in stably transfected CHO DG44 cells
Construct Clones 96 well sups Spent T25 6/10 day assay
Screened expression top producer on top clones
range (pg/ml) (pg/ml)
(pg/ml)
THERO 45 0-46.5 135 60/230
THER2 135 0-36 145 45/125
THER4 237 0-76 165 70/200
THER6 192 0-57 145 85/250
THER4RNA2 50 0-45 90 50/118
The clones with the highest production of the fusion protein were expanded into T25
and then T75 flasks to provide adequate numbers of cells for freezing and for scaling up production of
the fusion protein. Production levels were further increased in cultures from the four best clones by
progressive amplification in methotrexate-containing culture media. At each successive passage of
cells, the EX-CELL 302 complete media contained an increased concentration of methotrexate, such
that only the cells that amplified the DHFR plasmid could survive. Media for transfections under
selective amplification contained varying
levels of methotrexate (Sigma-Aldrich) as selective agent,
ranging from 50 nM to 1 gM, depending on the degree of amplification achieved.
Purification of fusion proteins from culture supernatants
Supernatants were collected from spent CHO cell cultures expressing the Apo A-1
Ink-mthIgG1 construct, filtered through 0.2 jim PES express filters (Nalgene, Rochester, N.Y.) and
subjected to gravity flow affinity chromatography over a Protein A-agarose (IPA 300 crosslinked
agarose, or IPA 400HC crosslinked agarose) column (Repligen, Waltham, Mass.). The column was
conditioned
with 0.1M citrate buffer, pH2.2, then supernatant adjusted to pH 8.0 with
0.5M NHCO ,
and loaded by gravity flow to allow binding of the fusion proteins. Columns were then washed with
several column volumes column
wash buffer (90mM Tris-Base, 150mM NaCl, 0.05% sodium azide,
pH 8.7) or Dulbecco's modified PBS, pH 7.4 prior to elution. Bound protein was eluted using 0.1 M
citrate buffer, pH 3.2. Fractions (0.8-0.9 ml) were collected into 0.2 ml 0.5M NaCO -NaHCO buffer
to neutralize each fraction. Protein concentration of aliquots (2 pl) from each fraction were
determined at 280nM using a Nanodrop (Wilmington DE) microsample spectrophotometer, with
blank determination using 0.1 M citrate buffer, pH 3.2, 0.5M NaCO at a 10:1 v:v ratio. Fractions
containing fusion protein were pooled, and buffer exchange performed by dialysis using Spectrum
Laboratories G2 (Ranch Dominguez, CA, Catalog #G235057, Fisher Scientific catalog # 08007)
FLOAT-A-LYZER@ units
(MWCO 20kDa) against D-PBS (Hyclone, ThermoFisher Scientific,
Dallas, TX), pH 7.4. Dialysis was performed in sterile, 2.2 liter Corning roller bottles at 4'C
overnight.
dialysis, protein was filtered using 0.2 pM filter units, and aliquots tested for
After
endotoxin contamination using PYROTELL® LAL gel clot system single test vials (STV) (Catalog #
G2006, Associates of Cape Cod, East Falmouth, MA). The predicted OD 280 of a 1 mg/ml solution
of the THER4 fusion protein was determined to be 1.19 (mature protein without either the signal
peptide or the 6 amino acid propeptide) or 1.27 (including the 6 amino acid propeptide) using the
protein analysis tools in the VECTOR NTI@ Version 11.5 Software package (Informax, North
Bethesda, MD) and the predicted cleavage site from the online ExPASy protein analysis tools. It is
unclear whether the fusion protein secreted from the CHO cells is homogeneous with regard to
complete cleavage of the propeptide from the recombinant molecules. The OD280 for each purified
fusion protein was corrected using these tools.
Reducing and Nonreducing SDS-PAGE Analysis of apo A-] Ig Fusion Proteins
Purified fusion proteins were analyzed by electrophoresis on SDS-Polyacrylamide 4
12% Bis-Tris NuPAGE® gels (Life Technologies, Grand Island, NY). Fusion protein samples were
heated at 72 C for 10 minutes in LDS sample buffer with and without reduction of disulfide bonds
and applied to 4-12% BIS-Tris gels (Catalog #NP0301, LIFE Technologies, Grand Island, NY). Five
micrograms of each purified protein was loaded on the gels. The proteins were visualized after
electrophoresis by IMPERIALTM protein staining (Pierce Imperial Protein Stain Reagent, Catalog
#24615, ThermoFisher Scientific/Pierce, Rockford, IL), and destaining in distilled water. Molecular
weight markers were included on the same gel (KALEIDOSCOPETM Prestained Standards, Catalog
#161-0324, Bio-Rad, Hercules, CA). The results from representative nonreducing and reducing gels
are shown in FIGS. 6A and 6B, respectively. Lanes are as follows from left to right: Lane #1
KALEIDOSCOPE prestained MW markers; Lane #2 - THERO; Lane #3 - THER2; Lane #4
THER4; Lane #5 - THER6; Lane #6 - THER4RNA2; Lane #7 - KALEIDOSCOPE Prestained MW
markers. Approximate molecular weights are indicated on the figures.
Again, the linker length difference between the different fusion proteins is evident on
both the reducing and nonreducing gels, with the THERO protein running at just over 50kDa. The
absence of hinge disulfides is evident by the similar mobility for each protein when electrophoresed
under reducing or nonreducing conditions.
Native Gel Electrophoresis of apo A-] IgG Fusion Proteins
The protein A purified fusion proteins were subjected to native PAGE analysis.
BLUE Native PAGE gels were run using 4-16% Bis-Tris NativePAGETM gels (Life
Technologies/ThermoFisher) with cathode and anode buffers prepared according to manufacturer's
instructions. Samples (4.5 pg each fusion protein) were prepared without heating, using 4X sample
buffer, without
detergents. Gels were run for 30 minutes at 150 volts, followed by 1 hour
at 180 volts,
hour at 220 volts. Gels were washed in distilled water and incubated for two hours in
and the final
IMPERIALTM Protein stain. Gels were extensively destained overnight with repeated washes in
distilled water to remove the blue dye present in the cathode buffer used for running the gels.
shows a representative native gel using these conditions. Molecular weight markers were GE
Healthcare high molecular weight calibration markers, a mixture of six large, multicomponent
proteins, resuspended in the loading buffer used for samples, again without added detergents.
Samples were loaded as follows: Lane #1 - ORENCIA® (abatacept; CTLA4hlgG); Lane #2 - anti
mouse CD40 monoclonal antibody iC1O; Lane #3 - THER4RNA2; Lane #4 - GE Healthcare High
MW calibration markers; Lane #5 - THER6; Lane #6 - THER4; Lane #7 - THER2; Lane #8
THERO; Lane #9 - GE Healthcare high MW markers; Lane #10 - Athens Research Apo A-1.
The native ApoAl-IgG fusion proteins run at an approximate molecular weight
somewhere between the 140 and 233 kDa markers and with a similar mobility as ORENCIA®
(abatacept), a CTLA4Jg fusion protein with the same human IgGI Fc domain. The THER4RNase
bispecific fusion protein did not stain well with the IMPERIAL stain possibly due to the highly basic
composition of the RNase domain, but appears to migrate in a more diffuse pattern with the
predominate visible band migrating between the 233 and 440 kDa standards.
Example 5: Use of an IgG/Apo A-1 Sandwich ELISA to Assess Binding of THER Apo A-1
Fusion Proteins
An antigen binding ELISA was performed to assess
the ability of Ig fusion proteins,
captured by immobilized anti-human IgG (Fc-specific) to bind to and be detected by a horseradish
peroxidase conjugated antibody specific for human apolipoprotein A-1. High protein-binding, 96
well ELISA plates (NUNC MAXISORP@ plates, ThermoFisher Scientific) were coated with 1.5
pg/ml goat anti-human IgG (Jackson Immunoresearch). Plates were blocked overnight at 4 C with
PBS/3% BSA. Serial dilutions of each THER fusion protein, starting at 5 pg/ml, were incubated
overnight at 4 C. The plate was washed three times and then incubated with horseradish peroxidase
conjugated anti-human apolipoprotein A-i (ThermoFisher Scientific, catalog # PAI-28965) diluted
1:1500. Plates were incubated at room temperature for 2 hours. Plates were washed four times, then
SUREBLUE RESERVETM TMB substrate (Catalog #: 5302, KPL, Gaithersburg, MD) was added
to the plate at 80g1/well. Development was stopped by addition of 80 pl/well IN HCl. Samples were
read at 450 nm using a SYNERGYTM HT Biotek plate reader (Biotek Instruments, Winooski, VT) and
data analyzed using GEN5TM 2.0 software.
shows the results from a representative Apo A-i binding ELISA. OD450 is
plotted versus concentration of fusion protein. THER 0, 2, 4, 6, and THER4RNA2 fusion proteins all
similar dose-response curves, indicating that the molecules can each be captured by binding
exhibited
to the Ig tail and detected by binding of the Apo A-i domain to the antibody targeted to human Apo
A-1. Human apolipoprotein A-i (Athens Research & Technology, catalog # 16120101) was
included as a control and was not captured by the anti-human Fc specific antibody. At higher
concentrations, the molecule showed weak binding by the antibody targeted to Apo A-1, indicating
that the Apo A-I may have bound weakly to the plastic without capture by the anti-Fc antibody.
Example 6: Expression and Testing of an
RNase Bifunctional Enzyme Lipid Transport Fusion Molecule
For the Apo A-i IgG RNase fusion protein (THER4RNA2), RNase activity was
assayed to determine whether fusion of the enzyme to the carboxyl end of the fusion construct
interfered with ability of the molecule to digest RNA. FIGS. 9 and 10 show the results of an
RNASEALERTTM assay (IDT, Coralville, IA) performed using the fluorescence and kinetic assay
functions of the SYNERGYTM HT plate reader. RNASEALERTTM Substrate is a synthetic RNA
oligonucleotide that has a fluorescein (R) on one end and a dark quencher (Q) on the other end. When
intact, the substrate has little or no fluorescence, but when cleaved by an RNase, the substrate
fluoresces green (490 nm excitation, 520 nm emission) and can be detected with the appropriately
equipped fluorescence plate reader. A positive signal in this assay shows increasing fluorescence
signal over time due to cleavage of the substrate by RNase present in the sample(s). Microplates were
incubated with RNASEALERT substrate (a fixed concentration of 20 pmol/pl), IX RNASEALERT
buffer, and fusion protein or enzyme controls dilutions added to each well of a 96 well plate. Enzyme
activity assays were performed in triplicate for each sample, and the kinetic assay allowed to proceed
for 45 minutes, with successive readings every 60 seconds. The increasing fluorescence at each time
point is displayed for each well as a trace of RFU/well as a function of time in Serial dilutions
of enzyme/fusion protein included 20 pmol/pl, 13.4 pmol/pl, 8.9 pmol/pl, 6 pmol/pl, 4 pmol/pl, 2.7
pmol/pl, 1.8 pmol/pl, and no enzyme. RNase A (Ambion/ThermoFisher, catalog #AM2270) was
included as a positive control, and THER4 (apo A1nk26-hIgG) was included as a negative control
for comparing to the THER4RNA2 fusion protein. Overlays of the traces generated using the 4
pmol/pl enzyme are shown in . Two replicates
of the RNaseA, THER4RNA2, and THER4,
are shown. All enzymes are at 4 pmol/pl and the substrate is present at 20 pmol/pl.
Example 7: Measurement of Cholesterol Efflux to Fusion Protein Acceptors
Using two separate assays, THERO, THER2, THER4, THER6, and THER4RNA2
fusion proteins were assessed for their ability to act as acceptor molecules for reverse cholesterol
pre-loaded monocyte/macrophage mammalian cell lines. The first assay used the
transport from
human monocytic/macrophage
cell line THP-1 and a fluorescently labeled derivative
of cholesterol,
BODIPY-cholesterol or TOPFLUOR-cholesterol (cholesterol compound with fluorescent boron
dipyrromethene difluoride linked to sterol carbon-24) (Avanti Polar Lipids, Alabaster, AL). The
THP-1 cells were grown in RPMI with 4mM glutamine, 10% FBS and maintained in mid-logarithmic
growth prior to plating. The protocol was adapted from the procedures outlined in Sankaranararyanan
et al. (J. Lipid Res. 52:2332-2340, 2011) and Zhang et al. (ASSAY and Drug Development
Technologies: 136-146, 2011). Cells were harvested and plated to 96-well flat bottom tissue culture
plates at 2x10 cells/ml or 2x10 cells/well in 100 pl RPMI media containing 33 ng/ml PMA. Cells
were maintained in culture for 36-48 hours to allow for differentiation to occur prior to the assay.
Culture media was aspirated and plates were washed in 1xPBS. Labeling media consisting of the
following components (Phenol Red free RPMI with media supplements, 0.2% FBS, with ACAT
inhibitor at 2 pg/ml, Sandoz 58-035 (Sigma-Aldrich, St. Louis MO), LXR agonist TO-901317 at 2.5
pM (Sigma-Aldrich, St. Louis, MO), 35 ng/ml PMA (Sigma-Aldrich, St. Louis, MO), and 1.25 mM
methyl beta-cyclodextrin (Sigma-Aldrich, St. Louis, MO), 50 uM cholesterol (Sigma-Aldrich, St.
Louis, MOO, and 25 pM TOPFLUOR cholesterol (Avanti Polar Lipids, Alabaster, AL) was added at a
volume of 100 pl/well and incubated at 37C, 5% C02 for 10-12 hours. Equilibration media, RPMI
complete with 10% FBS, 33 ng/ml PMA (100 ul/well), was added to each well and incubated for 8
hours prior to incubation with acceptors. Labeling/equilibration media was aspirated from plates, and
plates were washed twice with 200 pl/well PBS + 0.15% BSA. Efflux acceptor reagents were added
to individual wells in efflux buffer and incubated for two hours prior to assay. Acceptors were added
to efflux buffer at concentrations ranging from lOOnM to 500nM, depending on the assay. Efflux
buffer was phenol red-free RPMI with growth supplements and 0.15% BSA. Samples were run in
sets of 6-12 per condition/acceptor, and a minimum of five replicates used for statistical analysis.
APO A-i was run as a positive control, and efflux media alone was used as the background negative
control (baseline efflux). The efflux reaction was allowed to proceed for two hours, after which
culture media was harvested to black, flat bottom 96-well plates (media reading). Cell lysates were
prepared by addition of 100 ul 0.1 N NaOH to each well of the efflux plate, and incubation for 15
minutes on a plate shaker at 4'C. Cell lysates were transferred to black, 96-well plates (lysate
reading), and fluorescence for media and lysate samples measured using a SYNERGYTM HT plate
reader with excitation at 485 nm and emission at 528 nm. Efflux was calculated as the ratio of the
fluorescence measurements: (media/(media+lysate) x 100). The specific efflux was calculated by
subtracting the baseline readings of the samples with no acceptor present from the total efflux/sample
for each tested acceptor. Data analysis was performed using GraphPad Prism v 4.0 Software (San
Diego, CA). The assay results are shown in .
The second assay used the mouse macrophage cell line
J774A.1 (ATCC, Manassas,
VA) to assess reverse cholesterol transport (RCT) using a radioactive derivative of cholesterol, [ H]
cholesterol as described by Sankaranararyanan et al. (J. Lipid Res. 52:2332-2340, 2011) and Yancey
et al. (J. Lipid Res. 45:337-346, 2004). Briefly, J774 cells (3.5x105 per well in 24 well plates) were
incubated for 24 hours in 0.25 ml RPMI media supplemented with 5% FBS, ACAT inhibitor Sandoz
58-035 (2 pg/ml), and 4 pCi/ml of [ H]-cholesterol. ACAT inhibitor was present at all times during
the assay. Cells were equilibrated 16-24 hours in media with or without cAMP (0.3mM) prior to
incubation with acceptors. The presence of cAMP upregulates the ABCA1 molecule. Labeled cells
were washed in media containing 1% BSA, then acceptor molecules were added at 50, 100 and
200nM in MEM-HEPES media and incubated for 4 hours prior to measurements. All treatments were
performed in triplicate. The [ H] cholesterol in 100 pl of the media was then measured by liquid
scintillation counting. The percentage efflux is based on the total [ H]cholesterol present in the cells
before the efflux incubation (to sample). To measure the [ H]cholesterol present in the cells, the cell
lipids were extracted by incubating the cell monolayers overnight in isopropanol. After lipid
extraction, the total [ H]cholesterol present in the lipid extract was measured by liquid scintillation
counting. Data analysis was performed using GraphPad Prism software 4.0 (San Diego, CA). The
assay results are shown in .
Example 8: Construction of a PONi Bifunctional Enzyme Lipid Transport Fusion Molecule
In addition to the apoAIgG-RNase
expression constructs described above,
additional molecules which physically link the ApoA-1 phospholipid transport function to the active
sites of other enzyme domains are constructed. One such molecule contains a segment corresponding
to human paraoxanase 1 (PON1), with nucleotide and encoded amino acid sequences as shown in
SEQ ID NO: 11 and SEQ ID NO: 12, respectively. This arylesterase enzyme is present in human
serum exclusively associated with high density lipoprotein (HDL), and inhibits oxidation of low
density lipoprotein molecules. This protection from oxidation also inhibits development of vascular
and coronary diseases. The mature protein form of PON1 is unique in that it retains its amino
terminal signal peptide after secretion (amino acid residues 1 to 15 of SEQ ID NO: 12, encoded by
nucleotide residues 1 to 45 of SEQ ID NO:11). Expression of a mutant form of PON1 with a
cleavable amino terminus demonstrated that PON1 associates with lipoproteins through its amino
terminus by binding to phospholipids directly rather than first binding to ApoA-1. See Sorenson et
al., Arteriosclerosis, Thrombosis, and Vascular Biology 19:2214-2225, 1999. Removal of the signal
sequence was found to eliminate binding of the PONt moiety to phospholipids, proteoliposomes, and
serum lipoproteins. Additionally, in the absence of phospholipid, wild-type PONt does not bind
directly to ApoA-1. See Sorenson et al., supra. These PONt signal sequence mutants showed
reduced enzyme activity, possibly due to inability to bind the optimal phospholipid substrates.
Nevertheless, a recombinant, active form of human PON1 has been expressed in bacteria that is
missing this signal sequence. See Stevens et al., Proc. Natl. Acad. Sci. USA 105:12780-12784, 2008.
The presence of ApoA-1 does appear to stabilize arylesterase activity of the enzyme.
Removal of the amino terminal signal sequence of PON1 (and thereby the
phospholipid binding moiety) and substitution of this region with human apoAlnk-IgG directly
links the enzyme activity with the phospholipid binding domain of ApoA-1, stabilizing the
arylesterase enzyme activity while providing the optimal substrates bound to the ApoA-1 domain.
Such a molecule still traffics and is transported with the phospholipids bound by ApoA-1 and retains
enzyme activity due to replacement of the signal sequence domain with an alternative phospholipid
binding domain. In addition, a bifunctional molecule fusing these two domains exhibits improved
expression and facilitates targeting of the PON1 activity to the choroid plexus through active binding
of apo A-1. PON1 has been expressed at the carboxyl terminus of an insulin receptor targeted
antibody (see Boado et al., Mol. Pharm. 5:1037-1043, 2008; Boado et al., Biotechnology and
Bioengineering 108:186-196, 2011); however, the amino terminal signal peptide was included in this
fusion protein. The fusion gene and protein described here provides a novel method of PONt fusion
protein expression, eliminating the requirement for the signal peptide by a direct physical coupling of
the truncated enzyme to the apo A-i domain, thereby preserving and stabilizing both the binding
function and arylesterase activity of PONt.
Sequences for the fusion gene and protein are shown in SEQ ID NO:27 and SEQ ID
NO:28 for THER4PON1 (nucleotide and amino acid sequences, respectively) and in SEQ ID NO:37
and SEQ ID NO:38 for THER2PON1 (nucleotide and amino acid sequences, respectively). Similar
fusion genes and proteins contain alternative linker forms of ApoA- 1 fused to the hIgG 1 -linker-PONt
segment(s). The PONt sequences within the THER4PON1 and THER2PON1 molecules correspond
to the Q192 allele for human PONt.
Alternative forms of PONt are also used to construct bifunctional fusion molecules
linking PONt to Apo A-1. A sequence polymorphism that affects enzyme activity for different
substrates is present at position 192 of the PONi sequence. See Steven et al., supra. The amino acid
at this position may be glutamine (Q) or arginine (R) in humans, or a lysine (K) in rabbits. The
arginine allele at position 192 has been reported to have a higher catalytic activity in vitro and in vivo.
Similarly, the rabbit form of PONt with a lysine at position 192 has been reported to have a more
stable catalytic activity in vitro and in vivo (see Steven et al., supra; Richter et al., Circulation
Cardiovascular Genetics 1:147-152, 2008). These alternative PONt sequences are shown in SEQ ID
NO:41 (nucleotide) and SEQ ID NO:42 (amino acid) for the PONt Q192K form, and SEQ ID NO:43
(nucleotide) and SEQ ID NO:44 (amino acid) for the PONt Q192R form. The fusion construct
alternate PONI forms and the THER4 sequence (apoA-1(g4s)4hIgGNLG-...) is
between these
designated as THER4PON1 Q192K (nucleotide and amino acid sequences shown in SEQ ID NO:45
and SEQ ID NO:46, respectively) or THER4PON1 Q192R (nucleotide and amino acid sequences
shown in SEQ ID NO:47 and SEQ ID NO:48, respectively), depending on the polymorphism present
in the PONI sequence at amino acid 192 of the PONI sequence (or at amino acid 720 of the
THER4PON1Q variants shown in SEQ ID NO:46 and SEQ ID NO:48). Similarly, a THER2 form of
the fusion gene/protein is indicated as THER2PON1 Q192K or THER2PON1Q192R. For all of these
fusion constructs, the PONI amino terminal signal sequence (amino acids 1-15 of SEQ ID NO:12) is
removed.
Bispecific Enzyme Lipoprotein Transfer Proteins comprising PONI are screened for
arylesterase/PONI activity using the nontoxic substrates 4-(chloromethyl)phenyl acetate (CMPA) and
phenyl acetate (see Richter et al., supra). These substrates are preferable for screening activity since
the substrate and reaction product are relatively nontoxic compared to organophosphate pesticides.
The CMPA substrate (Sigma-Aldrich, Inc. St Louis, MO) is incubated with serial dilutions of fusion
protein and rates of CMPA hydrolysis assayed at 280 nm for 4 minutes at 25 C using ultraviolet
transparent 96-well plates (Costar, Cambridge, Mass). Dilutions are run in triplicate or quadruplicate
and substrate concentration
fixed at 3mmol/L in 20mM Tris-HCl (pH 8.0), 1.0 mM CaCl . Similarly,
arylesterase assays are performed on phenyl acetate as substrate. The rates of PA hydrolysis are
measured at 270nm, for 4 minutes under both high and low salt conditions.
Example 9: Construction of a PAFAH or CETP Bifunctional Enzyme Lipid Transport Fusion
Molecule
In addition to the apoAIgG-RNase and apoAIgG-PON1 expression constructs
described above, additional molecules which physically link the ApoA-1 phospholipid transport
function to the active sites of other enzyme domains are constructed.
One such molecule contains a segment corresponding to human PAFAH (lipoprotein
associated phospholipase A2, human phospholipase A2 group VII, platelet activating factor acetyl
hydrolase), with nucleotide and encoded amino acid sequences as shown in SEQ ID NO:31 and SEQ
ID NO:32, respectively (see also GenBank accession number NM_005084 (transcript variant 1)). The
PAFAH amino acid sequence is encoded by nucleotides 270 to 1592 of SEQ ID NO:31, with the
STOP codon at nucleotides 1593 to 1595. The fusion gene and protein are designed fusing the
PAFAH coding sequence at the carboxyl end of the human IgG with the N-linked glycosylation linker
inserted between the two molecules. The THER4PAFAH nucleotide and encoded amino acid
sequences are shown as SEQ ID NO:33 and SEQ ID NO:34, respectively. The PAFAH sequence
amino acid signal peptide (MVPPKLHVLFCLCGCLAVVYP; residues 1-21 of SEQ
without the 21
ID NO:32) is fused to the NLG linker at amino acid position 544 in SEQ ID NO 34.
Another such molecule contains a segment corresponding to human CETP or
cholesteryl ester transfer protein (CETP), transcript variant 1, with nucleotide and encoded amino acid
sequences as shown in SEQ ID NO:29 and SEQ ID NO:30, respectively (see also GenBank accession
number NM_000078). The CETP protein is encoded by nucleotides 58 to 1537 of SEQ ID NO:29.
The fusion gene and protein are designed fusing the CETP coding sequence at the carboxyl end of the
human IgG with the N-linked glycosylation linker inserted between the two molecules. The
THER4CETP (or human apo A-i-(g4s)4-hIgG-NLG-CETP) nucleotide and encoded amino acid
sequences are shown as SEQ ID NO:39 and SEQ ID NO:40, respectively. The nucleotides (57-107 of
SEQ ID NO 29) encoding the signal peptide (amino acids 1-17 of SEQ ID NO 30) are removed in
order to create the fusion gene between the NLG linker sequence and the CETP mature peptide. The
fusion junction between these two protein domains is located at amino acid 544 of SEQ ID NO 40.
From the foregoing, it will be appreciated that, although specific embodiments of the
invention have been described herein for purposes of illustration, various modifications may be made
without deviating from the spirit and scope of the invention. Accordingly, the invention is not limited
except as by the appended claims. All publications, patents, and patent applications cited herein are
hereby incorporated by reference in their entireties for all purposes.
Claims (25)
1. A fusion polypeptide comprising, from an amino-terminal position to a carboxyl- terminal position, ApoAl-Ll-D, wherein: ApoAl is a first polypeptide segment comprising an amino acid sequence having at least 90% or at least 95% identity with amino acid residues 19-267 or 25-267 of SEQ ID NO:2, wherein said first polypeptide segment has cholesterol efflux activity; L1 is a first polypeptide linker comprising at least five amino acid residues; and D is an immunoglobulin Fc region; wherein the fusion polypeptide has increased ApoA1 cholesterol efflux activity as compared to either an ApoA1-Fc fusion polypeptide with a two amino acid linker or an ApoA1-Fc fusion polypeptide without a linker.
2. The fusion polypeptide of claim 1, wherein L1 comprises at least 10 amino acid residues.
3. The fusion polypeptide of claim 1, wherein L1 comprises at least 16 amino acid residues, consists of from 15 to 40 amino acid residues, or consists of from 16 to 36 amino acid residues.
4. The fusion polypeptide of claim 1, wherein L1 consists of 16 amino acid residues, 21 amino acid residues, 26 amino acid residues, 31 amino acid residues, or 36 amino acid residues.
5. The fusion polypeptide of claim 4, wherein L1 has the amino acid sequence shown in residues 268-283 of SEQ ID NO:22, residues 268-288 of SEQ ID NO:26, residues 268-293 of SEQ ID NO:2, SEQ ID NO:54, or residues 268-303 of SEQ ID NO:24.
6. The fusion polypeptide of any one of claims 1 to 5, wherein the first polypeptide segment has the amino acid sequence shown in residues 19-267 or 25-267 of SEQ ID NO:2.
7. The fusion polypeptide of claim 1, wherein the Fc region is a human Fc region, optionally wherein the human Fc region is an Fc variant comprising one or more amino acid substitutions relative to the wild-type human sequence.
8. The fusion polypeptide of claim 7, wherein the Fc region is a human γ1 Fc region or a human γ3 Fc region.
9. The fusion polypeptide of claim 7, wherein the Fc region is a human γ1 Fc variant in which Eu residue C220 is replaced by serine, optionally wherein Eu residue P331 is replaced by serine.
10. The fusion polypeptide of claim 9, wherein Eu residues C226 and C229 are each replaced by serine, optionally wherein Eu residue P238 is replaced by serine.
11. The fusion polypeptide of claim 1, wherein the Fc region has the amino acid sequence shown in (i) residues 294-525 or 294-524 of SEQ ID NO:2, or (ii) residues 294-525 or 294-524 of SEQ ID NO:13.
12. The fusion polypeptide of claim 1, wherein the fusion polypeptide comprises acid sequence having at least 90% or at least 95% identity with (i) residues 19-525, 19-524, 25-525, or 25-524 of SEQ ID NO:2, (ii) residues 19-525, 19-524, 25-525, or 25-524 of SEQ ID NO:13, (iii) residues 19-515, 19-514, 25-515, or 25-514 of SEQ ID NO:22, (iv) residues 19-520, 19-519, 25-520, or 25-519 of SEQ ID NO:26, or (v) residues 19-535, 19-534, 25-535, or 25-534 of SEQ ID NO:24, optionally wherein the fusion polypeptide comprises the amino acid sequence shown in any one of (i)-(v).
13. The fusion polypeptide of any one of claims 1 to 12, further comprising a second polypeptide segment located carboxyl-terminal to the Fc region, wherein the second polypeptide segment is selected from the group consisting of an RNase, a paraoxonase, a platelet-activating factor acetylhydrolase, and a cholesterol ester transfer protein.
14. The fusion polypeptide of claim 13, wherein the fusion polypeptide comprises, from an amino-terminal position to a carboxyl-terminal position, ApoAl-Ll-D-L2-P, wherein: ApoAl, L1, and D are as defined in claim 1; L2 is a second polypeptide linker, wherein L2 is optionally present; and P is the second polypeptide segment.
15. The fusion polypeptide of claim 13 or 14, wherein the second polypeptide segment is the RNase, optionally wherein the RNase has at least 90% or at least 95% identity with amino acid residues 542-675 of SEQ ID NO:4, optionally wherein the RNase has the amino acid sequence shown in residues 542-675 of SEQ ID NO:4.
16. The fusion polypeptide of claim 15, wherein the fusion polypeptide comprises an amino acid sequence having at least 90% or at least 95% identity with (i) residues 19-675 or 25-675 of SEQ ID NO:4 or (ii) residues 19-675 or 25-675 of SEQ ID NO: 14, optionally wherein the fusion polypeptide comprises the amino acid sequence shown in (i) or (ii).
17. The fusion polypeptide of claim 13 or 14, wherein the second polypeptide segment is the paraoxonase, optionally wherein the paraoxonase has at least 90% or least 95% identity with amino acid residues 16-355 of SEQ ID NO:12, amino acid residues 16-355 of SEQ ID NO:42, or amino acid residues 16-355 of SEQ ID NO:44, optionally wherein the paraoxonase has the amino acid sequence shown in residues 16-355 of SEQ ID NO:12, residues 16-355 of SEQ ID NO:42, or residues 16-355 of SEQ ID NO:44.
18. The fusion polypeptide of claim 17, wherein the fusion polypeptide comprises an amino acid sequence having at least 95% or at least 95% identity with (i) residues 19-883 or 25-883 of SEQ ID NO:28, (ii) residues 19-873 or 25-873 of SEQ ID NO:38, (iii) residues 19-883 or 25-883 of SEQ ID NO:46, or (iv) residues 19-883 or 25-883 of SEQ ID NO:48, optionally wherein the fusion polypeptide comprises the amino acid sequence shown in any one of (i)-(iv).
19. A dimeric protein comprising a first fusion polypeptide and a second fusion polypeptide, wherein each of said first and second fusion polypeptides is a fusion polypeptide as defined in any one of claims 1 to 18.
20. A polynucleotide encoding the fusion polypeptide of any one of claims 1 to 18.
21. An expression vector comprising the following operably linked elements: a transcription promoter; a DNA segment encoding the fusion polypeptide of any one of claims 1 to 18; and a transcription terminator.
22. A cultured cell into which has been introduced the expression vector of claim 21, wherein the cell expresses the DNA segment.
23. A method of making a fusion polypeptide, the method comprising: culturing a cell into which has been introduced the expression vector of claim 21, wherein the cell expresses the DNA segment and the encoded fusion polypeptide is produced; and recovering the fusion polypeptide, optionally wherein the encoded fusion polypeptide is produced and recovered as a dimeric protein.
24. A composition comprising: a fusion polypeptide of any one of claims 1 to 18 or a dimeric protein of claim 19; and a pharmaceutically acceptable carrier.
25. Use of a fusion polypeptide of any one of claims 1 to 18 or a dimeric protein of claim 19 in the manufacture of a medicament for the treatment of a disease, or for the treatment of exposure to sulfur mustard gas or to an organophosphate, wherein the disease is selected from the group consisting of a cardiovascular disease characterized by atherosclerosis, optionally wherein the cardiovascular disease is selected from the group consisting of coronary heart disease and stroke, optionally wherein the coronary heart disease is characterized by acute coronary syndrome; a neurodegenerative disease, optionally wherein the neurodegenerative disease is selected from the group consisting of Alzheimer’s disease, multiple sclerosis, and a neurodegenerative disease is characterized by dementia; a disease characterized by amyloid deposit, optionally wherein the disease characterized by amyloid deposit is Alzheimer’s disease; an autoimmune disease, optionally wherein the autoimmune disease is selected from the group consisting of rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, and type 1 diabetes; an inflammatory disease, optionally wherein the inflammatory disease is selected from the group consisting of rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, type 1 diabetes, type 2 diabetes, obesity, non-alcoholic steatohepatitis, coronary heart disease, stroke, and an inflammatory lung disease, optionally wherein the inflammatory lung disease is selected from the group consisting of asthma, chronic obstructive pulmonary disease (COPD), bronchiectasis, idiopathic pulmonary fibrosis, hyperoxia, hypoxia, and acute respiratory distress syndrome; an infectious disease, optionally wherein the infectious disease is characterized by a bacterial infection; and cancer, optionally wherein the cancer is selected from the group consisting of malignant melanoma, renal cell carcinoma, non-small cell lung cancer, bladder cancer, and head and neck cancer, optionally wherein the cancer treatment is a combination therapy, optionally wherein the combination therapy comprises a therapy selected from the group consisting of a non-ApoA1- mediated immunomodulatory therapy, radiation therapy, chemotherapy, and a targeted therapy, optionally wherein the non-ApoA1-mediated immunomodulatory therapy comprises an anti-PD- 1/PD-L1 therapy, an anti-CTLA-4 therapy, or both.
Applications Claiming Priority (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201562215256P | 2015-09-08 | 2015-09-08 | |
US62/215,256 | 2015-09-08 | ||
PCT/US2016/050405 WO2017044424A1 (en) | 2015-09-08 | 2016-09-06 | Apoa-1 fusion polypeptides and related compositions and methods |
Publications (2)
Publication Number | Publication Date |
---|---|
NZ738963A NZ738963A (en) | 2021-08-27 |
NZ738963B2 true NZ738963B2 (en) | 2021-11-30 |
Family
ID=
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US11472864B2 (en) | Polynucleotides encoding APOA1-PON1 fusion polypeptides | |
EP3875477A1 (en) | Immunomodulatory proteins with tunable affinities | |
US20230250152A1 (en) | Ctla-4 variant immunomodulatory proteins and uses thereof | |
KR20200058322A (en) | Anti-PD-L1 antibody and IL-7 fusion | |
EP3355908A1 (en) | Treatment of bile acid disorders | |
WO2018136163A2 (en) | Tandem apoa-1 fusion polypeptides | |
NZ738963B2 (en) | Apoa-1 fusion polypeptides and related compositions and methods | |
AU2012358248B2 (en) | Serum amyloid P-antibody fusion proteins | |
US20240294889A1 (en) | Dnase fusion polypeptides and related compositions and methods |