WO2018162722A1 - Inhibiteurs de dpp-4 à utiliser dans le traitement de fractures osseuses - Google Patents
Inhibiteurs de dpp-4 à utiliser dans le traitement de fractures osseuses Download PDFInfo
- Publication number
- WO2018162722A1 WO2018162722A1 PCT/EP2018/055931 EP2018055931W WO2018162722A1 WO 2018162722 A1 WO2018162722 A1 WO 2018162722A1 EP 2018055931 W EP2018055931 W EP 2018055931W WO 2018162722 A1 WO2018162722 A1 WO 2018162722A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- dpp
- inhibitor
- cells
- subject
- bone
- Prior art date
Links
- 229940090124 dipeptidyl peptidase 4 (dpp-4) inhibitors for blood glucose lowering Drugs 0.000 title claims abstract description 184
- 208000010392 Bone Fractures Diseases 0.000 title claims abstract description 164
- 210000004027 cell Anatomy 0.000 claims abstract description 211
- 210000000988 bone and bone Anatomy 0.000 claims abstract description 110
- 206010017076 Fracture Diseases 0.000 claims abstract description 84
- 101000884271 Homo sapiens Signal transducer CD24 Proteins 0.000 claims abstract description 73
- 102100038081 Signal transducer CD24 Human genes 0.000 claims abstract description 73
- 238000000034 method Methods 0.000 claims abstract description 70
- 230000002188 osteogenic effect Effects 0.000 claims abstract description 59
- 230000035876 healing Effects 0.000 claims abstract description 57
- 210000002894 multi-fate stem cell Anatomy 0.000 claims abstract description 32
- 230000004069 differentiation Effects 0.000 claims abstract description 25
- 238000002054 transplantation Methods 0.000 claims abstract description 23
- 210000000130 stem cell Anatomy 0.000 claims abstract description 21
- 238000004113 cell culture Methods 0.000 claims abstract description 14
- 229910052500 inorganic mineral Inorganic materials 0.000 claims abstract description 11
- 239000011707 mineral Substances 0.000 claims abstract description 11
- 230000002401 inhibitory effect Effects 0.000 claims abstract description 8
- 210000000577 adipose tissue Anatomy 0.000 claims abstract description 7
- 210000002901 mesenchymal stem cell Anatomy 0.000 claims abstract description 5
- 101000738771 Homo sapiens Receptor-type tyrosine-protein phosphatase C Proteins 0.000 claims description 98
- 102100037422 Receptor-type tyrosine-protein phosphatase C Human genes 0.000 claims description 98
- 102100024616 Platelet endothelial cell adhesion molecule Human genes 0.000 claims description 77
- 239000000203 mixture Substances 0.000 claims description 64
- 239000008194 pharmaceutical composition Substances 0.000 claims description 61
- 230000014509 gene expression Effects 0.000 claims description 48
- 229960004034 sitagliptin Drugs 0.000 claims description 27
- MFFMDFFZMYYVKS-SECBINFHSA-N sitagliptin Chemical compound C([C@H](CC(=O)N1CC=2N(C(=NN=2)C(F)(F)F)CC1)N)C1=CC(F)=C(F)C=C1F MFFMDFFZMYYVKS-SECBINFHSA-N 0.000 claims description 27
- 208000001132 Osteoporosis Diseases 0.000 claims description 25
- 238000009472 formulation Methods 0.000 claims description 20
- 241000282414 Homo sapiens Species 0.000 claims description 17
- 230000002648 chondrogenic effect Effects 0.000 claims description 14
- 230000003111 delayed effect Effects 0.000 claims description 10
- 239000003814 drug Substances 0.000 claims description 10
- 229960003105 metformin Drugs 0.000 claims description 10
- XZWYZXLIPXDOLR-UHFFFAOYSA-N metformin Chemical compound CN(C)C(=N)NC(N)=N XZWYZXLIPXDOLR-UHFFFAOYSA-N 0.000 claims description 10
- -1 vildapliptin Chemical compound 0.000 claims description 10
- 230000005484 gravity Effects 0.000 claims description 9
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 9
- 208000002565 Open Fractures Diseases 0.000 claims description 8
- 230000001737 promoting effect Effects 0.000 claims description 8
- 238000001356 surgical procedure Methods 0.000 claims description 8
- 239000003085 diluting agent Substances 0.000 claims description 6
- 229940124597 therapeutic agent Drugs 0.000 claims description 6
- 230000024245 cell differentiation Effects 0.000 claims description 5
- 238000003745 diagnosis Methods 0.000 claims description 5
- 208000001072 type 2 diabetes mellitus Diseases 0.000 claims description 5
- ZWPRRQZNBDYKLH-VIFPVBQESA-N Gemigliptin Chemical compound C([C@@H](N)CC(=O)N1CC2=C(C(=NC(=N2)C(F)(F)F)C(F)(F)F)CC1)N1CC(F)(F)CCC1=O ZWPRRQZNBDYKLH-VIFPVBQESA-N 0.000 claims description 4
- LTXREWYXXSTFRX-QGZVFWFLSA-N Linagliptin Chemical compound N=1C=2N(C)C(=O)N(CC=3N=C4C=CC=CC4=C(C)N=3)C(=O)C=2N(CC#CC)C=1N1CCC[C@@H](N)C1 LTXREWYXXSTFRX-QGZVFWFLSA-N 0.000 claims description 4
- 229960001667 alogliptin Drugs 0.000 claims description 4
- ZSBOMTDTBDDKMP-OAHLLOKOSA-N alogliptin Chemical compound C=1C=CC=C(C#N)C=1CN1C(=O)N(C)C(=O)C=C1N1CCC[C@@H](N)C1 ZSBOMTDTBDDKMP-OAHLLOKOSA-N 0.000 claims description 4
- 229960002458 gemigliptin Drugs 0.000 claims description 4
- 229960002397 linagliptin Drugs 0.000 claims description 4
- 229960004937 saxagliptin Drugs 0.000 claims description 4
- 108010033693 saxagliptin Proteins 0.000 claims description 4
- QGJUIPDUBHWZPV-SGTAVMJGSA-N saxagliptin Chemical compound C1C(C2)CC(C3)CC2(O)CC13[C@H](N)C(=O)N1[C@H](C#N)C[C@@H]2C[C@@H]21 QGJUIPDUBHWZPV-SGTAVMJGSA-N 0.000 claims description 4
- 229950000034 teneligliptin Drugs 0.000 claims description 4
- WGRQANOPCQRCME-PMACEKPBSA-N teneligliptin Chemical compound O=C([C@H]1NC[C@H](C1)N1CCN(CC1)C1=CC(=NN1C=1C=CC=CC=1)C)N1CCSC1 WGRQANOPCQRCME-PMACEKPBSA-N 0.000 claims description 4
- 101150084229 ATXN1 gene Proteins 0.000 abstract 1
- 201000003624 spinocerebellar ataxia type 1 Diseases 0.000 abstract 1
- 239000003795 chemical substances by application Substances 0.000 description 93
- 102100025012 Dipeptidyl peptidase 4 Human genes 0.000 description 61
- 241000699670 Mus sp. Species 0.000 description 54
- 230000002293 adipogenic effect Effects 0.000 description 48
- 238000011282 treatment Methods 0.000 description 43
- 108010067722 Dipeptidyl Peptidase 4 Proteins 0.000 description 40
- 108090000623 proteins and genes Proteins 0.000 description 32
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 29
- 201000010099 disease Diseases 0.000 description 28
- 230000000694 effects Effects 0.000 description 27
- 238000004458 analytical method Methods 0.000 description 26
- 230000001225 therapeutic effect Effects 0.000 description 26
- 241000699666 Mus <mouse, genus> Species 0.000 description 24
- 241001465754 Metazoa Species 0.000 description 23
- 101000908391 Homo sapiens Dipeptidyl peptidase 4 Proteins 0.000 description 22
- 238000010186 staining Methods 0.000 description 22
- 210000002303 tibia Anatomy 0.000 description 19
- 238000002474 experimental method Methods 0.000 description 18
- 238000011002 quantification Methods 0.000 description 17
- 101150008114 znf423 gene Proteins 0.000 description 17
- 102100034540 Adenomatous polyposis coli protein Human genes 0.000 description 16
- 101000924577 Homo sapiens Adenomatous polyposis coli protein Proteins 0.000 description 16
- 230000009815 adipogenic differentiation Effects 0.000 description 16
- 210000001519 tissue Anatomy 0.000 description 15
- 239000000243 solution Substances 0.000 description 13
- 235000000891 standard diet Nutrition 0.000 description 13
- 206010020649 Hyperkeratosis Diseases 0.000 description 12
- 238000003559 RNA-seq method Methods 0.000 description 12
- 210000001789 adipocyte Anatomy 0.000 description 12
- 238000003556 assay Methods 0.000 description 12
- 238000000338 in vitro Methods 0.000 description 12
- 108010054624 red fluorescent protein Proteins 0.000 description 12
- 238000001727 in vivo Methods 0.000 description 11
- 108090000765 processed proteins & peptides Proteins 0.000 description 11
- 239000005089 Luciferase Substances 0.000 description 10
- 241000124008 Mammalia Species 0.000 description 10
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 10
- 238000012413 Fluorescence activated cell sorting analysis Methods 0.000 description 9
- 108060001084 Luciferase Proteins 0.000 description 9
- 235000009200 high fat diet Nutrition 0.000 description 9
- 238000007912 intraperitoneal administration Methods 0.000 description 9
- 230000004840 osteo-chondrogenic effect Effects 0.000 description 9
- 102000004196 processed proteins & peptides Human genes 0.000 description 9
- 235000018102 proteins Nutrition 0.000 description 9
- 102000004169 proteins and genes Human genes 0.000 description 9
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 8
- 230000003247 decreasing effect Effects 0.000 description 8
- 239000005090 green fluorescent protein Substances 0.000 description 8
- 239000003550 marker Substances 0.000 description 8
- 239000002609 medium Substances 0.000 description 8
- 230000009818 osteogenic differentiation Effects 0.000 description 8
- 229920001184 polypeptide Polymers 0.000 description 8
- WOVKYSAHUYNSMH-RRKCRQDMSA-N 5-bromodeoxyuridine Chemical compound C1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C(Br)=C1 WOVKYSAHUYNSMH-RRKCRQDMSA-N 0.000 description 7
- 101150023417 PPARG gene Proteins 0.000 description 7
- 210000004271 bone marrow stromal cell Anatomy 0.000 description 7
- 150000001875 compounds Chemical class 0.000 description 7
- 230000000875 corresponding effect Effects 0.000 description 7
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 7
- 230000001965 increasing effect Effects 0.000 description 7
- 239000007924 injection Substances 0.000 description 7
- 238000002347 injection Methods 0.000 description 7
- 239000000758 substrate Substances 0.000 description 7
- JNTMAZFVYNDPLB-PEDHHIEDSA-N (2S,3S)-2-[[[(2S)-1-[(2S,3S)-2-amino-3-methyl-1-oxopentyl]-2-pyrrolidinyl]-oxomethyl]amino]-3-methylpentanoic acid Chemical compound CC[C@H](C)[C@H](N)C(=O)N1CCC[C@H]1C(=O)N[C@@H]([C@@H](C)CC)C(O)=O JNTMAZFVYNDPLB-PEDHHIEDSA-N 0.000 description 6
- NPGIHFRTRXVWOY-UHFFFAOYSA-N Oil red O Chemical compound Cc1ccc(C)c(c1)N=Nc1cc(C)c(cc1C)N=Nc1c(O)ccc2ccccc12 NPGIHFRTRXVWOY-UHFFFAOYSA-N 0.000 description 6
- 208000002193 Pain Diseases 0.000 description 6
- HFVAFDPGUJEFBQ-UHFFFAOYSA-M alizarin red S Chemical compound [Na+].O=C1C2=CC=CC=C2C(=O)C2=C1C=C(S([O-])(=O)=O)C(O)=C2O HFVAFDPGUJEFBQ-UHFFFAOYSA-M 0.000 description 6
- 150000001413 amino acids Chemical group 0.000 description 6
- 230000004071 biological effect Effects 0.000 description 6
- 230000000903 blocking effect Effects 0.000 description 6
- 230000007423 decrease Effects 0.000 description 6
- 239000002158 endotoxin Substances 0.000 description 6
- 230000006698 induction Effects 0.000 description 6
- 239000000463 material Substances 0.000 description 6
- 239000002773 nucleotide Substances 0.000 description 6
- 125000003729 nucleotide group Chemical group 0.000 description 6
- 230000011164 ossification Effects 0.000 description 6
- 150000003839 salts Chemical class 0.000 description 6
- 208000024891 symptom Diseases 0.000 description 6
- 108020004414 DNA Proteins 0.000 description 5
- 101150093210 DPP4 gene Proteins 0.000 description 5
- 108090000379 Fibroblast growth factor 2 Proteins 0.000 description 5
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 5
- 210000003486 adipose tissue brown Anatomy 0.000 description 5
- 230000011759 adipose tissue development Effects 0.000 description 5
- 210000000593 adipose tissue white Anatomy 0.000 description 5
- 238000010171 animal model Methods 0.000 description 5
- 230000037396 body weight Effects 0.000 description 5
- 239000000872 buffer Substances 0.000 description 5
- 239000002299 complementary DNA Substances 0.000 description 5
- 229960003957 dexamethasone Drugs 0.000 description 5
- UREBDLICKHMUKA-CXSFZGCWSA-N dexamethasone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1C[C@@H](C)[C@@](C(=O)CO)(O)[C@@]1(C)C[C@@H]2O UREBDLICKHMUKA-CXSFZGCWSA-N 0.000 description 5
- 108010054812 diprotin A Proteins 0.000 description 5
- 108010048367 enhanced green fluorescent protein Proteins 0.000 description 5
- 238000010195 expression analysis Methods 0.000 description 5
- 238000000684 flow cytometry Methods 0.000 description 5
- 238000001990 intravenous administration Methods 0.000 description 5
- 230000001404 mediated effect Effects 0.000 description 5
- 108020004999 messenger RNA Proteins 0.000 description 5
- 230000009467 reduction Effects 0.000 description 5
- 238000012163 sequencing technique Methods 0.000 description 5
- 239000000126 substance Substances 0.000 description 5
- 239000000725 suspension Substances 0.000 description 5
- 210000000689 upper leg Anatomy 0.000 description 5
- 238000005406 washing Methods 0.000 description 5
- FWBHETKCLVMNFS-UHFFFAOYSA-N 4',6-Diamino-2-phenylindol Chemical compound C1=CC(C(=N)N)=CC=C1C1=CC2=CC=C(C(N)=N)C=C2N1 FWBHETKCLVMNFS-UHFFFAOYSA-N 0.000 description 4
- 108700028369 Alleles Proteins 0.000 description 4
- DHCLVCXQIBBOPH-UHFFFAOYSA-N Glycerol 2-phosphate Chemical compound OCC(CO)OP(O)(O)=O DHCLVCXQIBBOPH-UHFFFAOYSA-N 0.000 description 4
- KFZMGEQAYNKOFK-UHFFFAOYSA-N Isopropanol Chemical compound CC(C)O KFZMGEQAYNKOFK-UHFFFAOYSA-N 0.000 description 4
- MIJPAVRNWPDMOR-ZAFYKAAXSA-N L-ascorbic acid 2-phosphate Chemical compound OC[C@H](O)[C@H]1OC(=O)C(OP(O)(O)=O)=C1O MIJPAVRNWPDMOR-ZAFYKAAXSA-N 0.000 description 4
- AUYYCJSJGJYCDS-LBPRGKRZSA-N Thyrolar Chemical compound IC1=CC(C[C@H](N)C(O)=O)=CC(I)=C1OC1=CC=C(O)C(I)=C1 AUYYCJSJGJYCDS-LBPRGKRZSA-N 0.000 description 4
- 230000009471 action Effects 0.000 description 4
- 230000022159 cartilage development Effects 0.000 description 4
- 206010012601 diabetes mellitus Diseases 0.000 description 4
- 235000005911 diet Nutrition 0.000 description 4
- 230000037213 diet Effects 0.000 description 4
- 239000012153 distilled water Substances 0.000 description 4
- 229940079593 drug Drugs 0.000 description 4
- 238000005516 engineering process Methods 0.000 description 4
- 239000012634 fragment Substances 0.000 description 4
- 239000003102 growth factor Substances 0.000 description 4
- 239000001963 growth medium Substances 0.000 description 4
- 238000010166 immunofluorescence Methods 0.000 description 4
- 238000011503 in vivo imaging Methods 0.000 description 4
- 238000010348 incorporation Methods 0.000 description 4
- CGIGDMFJXJATDK-UHFFFAOYSA-N indomethacin Chemical compound CC1=C(CC(O)=O)C2=CC(OC)=CC=C2N1C(=O)C1=CC=C(Cl)C=C1 CGIGDMFJXJATDK-UHFFFAOYSA-N 0.000 description 4
- 230000001939 inductive effect Effects 0.000 description 4
- 238000001802 infusion Methods 0.000 description 4
- 208000014674 injury Diseases 0.000 description 4
- 230000004807 localization Effects 0.000 description 4
- 239000002207 metabolite Substances 0.000 description 4
- 244000005700 microbiome Species 0.000 description 4
- 239000003921 oil Substances 0.000 description 4
- 235000019198 oils Nutrition 0.000 description 4
- 238000002360 preparation method Methods 0.000 description 4
- 239000000651 prodrug Substances 0.000 description 4
- 229940002612 prodrug Drugs 0.000 description 4
- XJMOSONTPMZWPB-UHFFFAOYSA-M propidium iodide Chemical compound [I-].[I-].C12=CC(N)=CC=C2C2=CC=C(N)C=C2[N+](CCC[N+](C)(CC)CC)=C1C1=CC=CC=C1 XJMOSONTPMZWPB-UHFFFAOYSA-M 0.000 description 4
- 230000001105 regulatory effect Effects 0.000 description 4
- 238000012360 testing method Methods 0.000 description 4
- 101150091111 ACAN gene Proteins 0.000 description 3
- 102000011690 Adiponectin Human genes 0.000 description 3
- 108010076365 Adiponectin Proteins 0.000 description 3
- 102100036601 Aggrecan core protein Human genes 0.000 description 3
- 108010067219 Aggrecans Proteins 0.000 description 3
- 108091033409 CRISPR Proteins 0.000 description 3
- 101150061453 Cebpa gene Proteins 0.000 description 3
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 3
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 3
- 102000003974 Fibroblast growth factor 2 Human genes 0.000 description 3
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- 102100031573 Hematopoietic progenitor cell antigen CD34 Human genes 0.000 description 3
- 241000282412 Homo Species 0.000 description 3
- 101000777663 Homo sapiens Hematopoietic progenitor cell antigen CD34 Proteins 0.000 description 3
- 101000741788 Homo sapiens Peroxisome proliferator-activated receptor alpha Proteins 0.000 description 3
- 101000909641 Homo sapiens Transcription factor COE2 Proteins 0.000 description 3
- 101000803403 Homo sapiens Vimentin Proteins 0.000 description 3
- 101150046735 LEPR gene Proteins 0.000 description 3
- 101150063827 LEPROT gene Proteins 0.000 description 3
- 241001529936 Murinae Species 0.000 description 3
- 102000004067 Osteocalcin Human genes 0.000 description 3
- 108090000573 Osteocalcin Proteins 0.000 description 3
- 102000003982 Parathyroid hormone Human genes 0.000 description 3
- 108090000445 Parathyroid hormone Proteins 0.000 description 3
- 102100038831 Peroxisome proliferator-activated receptor alpha Human genes 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- 238000002123 RNA extraction Methods 0.000 description 3
- YASAKCUCGLMORW-UHFFFAOYSA-N Rosiglitazone Chemical compound C=1C=CC=NC=1N(C)CCOC(C=C1)=CC=C1CC1SC(=O)NC1=O YASAKCUCGLMORW-UHFFFAOYSA-N 0.000 description 3
- 101150106167 SOX9 gene Proteins 0.000 description 3
- 102100024204 Transcription factor COE2 Human genes 0.000 description 3
- 102100035071 Vimentin Human genes 0.000 description 3
- 208000027418 Wounds and injury Diseases 0.000 description 3
- 239000000427 antigen Substances 0.000 description 3
- 108091007433 antigens Proteins 0.000 description 3
- 102000036639 antigens Human genes 0.000 description 3
- 210000004204 blood vessel Anatomy 0.000 description 3
- 210000001185 bone marrow Anatomy 0.000 description 3
- 210000000845 cartilage Anatomy 0.000 description 3
- 239000011248 coating agent Substances 0.000 description 3
- 239000013078 crystal Substances 0.000 description 3
- 230000001186 cumulative effect Effects 0.000 description 3
- 210000003275 diaphysis Anatomy 0.000 description 3
- 239000006185 dispersion Substances 0.000 description 3
- 239000002552 dosage form Substances 0.000 description 3
- 238000011156 evaluation Methods 0.000 description 3
- 238000000799 fluorescence microscopy Methods 0.000 description 3
- 230000006870 function Effects 0.000 description 3
- 238000003384 imaging method Methods 0.000 description 3
- 238000003365 immunocytochemistry Methods 0.000 description 3
- 230000006872 improvement Effects 0.000 description 3
- 239000003112 inhibitor Substances 0.000 description 3
- 239000007928 intraperitoneal injection Substances 0.000 description 3
- 238000002955 isolation Methods 0.000 description 3
- 238000013227 male C57BL/6J mice Methods 0.000 description 3
- 108010082117 matrigel Proteins 0.000 description 3
- 239000012528 membrane Substances 0.000 description 3
- 239000000199 parathyroid hormone Substances 0.000 description 3
- 229960001319 parathyroid hormone Drugs 0.000 description 3
- 238000000513 principal component analysis Methods 0.000 description 3
- 230000008569 process Effects 0.000 description 3
- 230000001698 pyrogenic effect Effects 0.000 description 3
- 239000001397 quillaja saponaria molina bark Substances 0.000 description 3
- 108020003175 receptors Proteins 0.000 description 3
- 102000005962 receptors Human genes 0.000 description 3
- 230000006798 recombination Effects 0.000 description 3
- 238000005215 recombination Methods 0.000 description 3
- 238000011084 recovery Methods 0.000 description 3
- 229930182490 saponin Natural products 0.000 description 3
- 150000007949 saponins Chemical class 0.000 description 3
- 239000011780 sodium chloride Substances 0.000 description 3
- 238000007920 subcutaneous administration Methods 0.000 description 3
- 230000003442 weekly effect Effects 0.000 description 3
- APIXJSLKIYYUKG-UHFFFAOYSA-N 3 Isobutyl 1 methylxanthine Chemical compound O=C1N(C)C(=O)N(CC(C)C)C2=C1N=CN2 APIXJSLKIYYUKG-UHFFFAOYSA-N 0.000 description 2
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 description 2
- 108010081589 Becaplermin Proteins 0.000 description 2
- 206010065687 Bone loss Diseases 0.000 description 2
- 102100022545 Bone morphogenetic protein 8B Human genes 0.000 description 2
- 238000010354 CRISPR gene editing Methods 0.000 description 2
- 241000282472 Canis lupus familiaris Species 0.000 description 2
- 241000283707 Capra Species 0.000 description 2
- 102000015775 Core Binding Factor Alpha 1 Subunit Human genes 0.000 description 2
- 108010024682 Core Binding Factor Alpha 1 Subunit Proteins 0.000 description 2
- 108010051219 Cre recombinase Proteins 0.000 description 2
- IGXWBGJHJZYPQS-SSDOTTSWSA-N D-Luciferin Chemical compound OC(=O)[C@H]1CSC(C=2SC3=CC=C(O)C=C3N=2)=N1 IGXWBGJHJZYPQS-SSDOTTSWSA-N 0.000 description 2
- 238000002965 ELISA Methods 0.000 description 2
- 102100040897 Embryonic growth/differentiation factor 1 Human genes 0.000 description 2
- 102400001368 Epidermal growth factor Human genes 0.000 description 2
- 101800003838 Epidermal growth factor Proteins 0.000 description 2
- 108700024394 Exon Proteins 0.000 description 2
- 102000018389 Exopeptidases Human genes 0.000 description 2
- 108010091443 Exopeptidases Proteins 0.000 description 2
- 102000018233 Fibroblast Growth Factor Human genes 0.000 description 2
- 108050007372 Fibroblast Growth Factor Proteins 0.000 description 2
- 102100024785 Fibroblast growth factor 2 Human genes 0.000 description 2
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 2
- 108010070675 Glutathione transferase Proteins 0.000 description 2
- 108010090296 Growth Differentiation Factor 1 Proteins 0.000 description 2
- 108010051696 Growth Hormone Proteins 0.000 description 2
- WZUVPPKBWHMQCE-UHFFFAOYSA-N Haematoxylin Chemical compound C12=CC(O)=C(O)C=C2CC2(O)C1C1=CC=C(O)C(O)=C1OC2 WZUVPPKBWHMQCE-UHFFFAOYSA-N 0.000 description 2
- 102100029100 Hematopoietic prostaglandin D synthase Human genes 0.000 description 2
- 101000976075 Homo sapiens Insulin Proteins 0.000 description 2
- 101000716729 Homo sapiens Kit ligand Proteins 0.000 description 2
- MPBVHIBUJCELCL-UHFFFAOYSA-N Ibandronate Chemical compound CCCCCN(C)CCC(O)(P(O)(O)=O)P(O)(O)=O MPBVHIBUJCELCL-UHFFFAOYSA-N 0.000 description 2
- 108090000723 Insulin-Like Growth Factor I Proteins 0.000 description 2
- 102000014429 Insulin-like growth factor Human genes 0.000 description 2
- 102100020880 Kit ligand Human genes 0.000 description 2
- XUIIKFGFIJCVMT-LBPRGKRZSA-N L-thyroxine Chemical compound IC1=CC(C[C@H]([NH3+])C([O-])=O)=CC(I)=C1OC1=CC(I)=C(O)C(I)=C1 XUIIKFGFIJCVMT-LBPRGKRZSA-N 0.000 description 2
- 102000016267 Leptin Human genes 0.000 description 2
- 108010092277 Leptin Proteins 0.000 description 2
- 102000004058 Leukemia inhibitory factor Human genes 0.000 description 2
- 108090000581 Leukemia inhibitory factor Proteins 0.000 description 2
- 101710175625 Maltose/maltodextrin-binding periplasmic protein Proteins 0.000 description 2
- 108010050258 Mitochondrial Uncoupling Proteins Proteins 0.000 description 2
- 102000015494 Mitochondrial Uncoupling Proteins Human genes 0.000 description 2
- 101100220687 Mus musculus Cidea gene Proteins 0.000 description 2
- 238000009004 PCR Kit Methods 0.000 description 2
- 102000001406 Perilipin Human genes 0.000 description 2
- 108060006002 Perilipin Proteins 0.000 description 2
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N Phenol Chemical compound OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 2
- 108010038512 Platelet-Derived Growth Factor Proteins 0.000 description 2
- 102000010780 Platelet-Derived Growth Factor Human genes 0.000 description 2
- 206010037660 Pyrexia Diseases 0.000 description 2
- IIDJRNMFWXDHID-UHFFFAOYSA-N Risedronic acid Chemical compound OP(=O)(O)C(P(O)(O)=O)(O)CC1=CC=CN=C1 IIDJRNMFWXDHID-UHFFFAOYSA-N 0.000 description 2
- 235000011449 Rosa Nutrition 0.000 description 2
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 2
- 102100038803 Somatotropin Human genes 0.000 description 2
- 108010043267 Sp7 Transcription Factor Proteins 0.000 description 2
- 229910000831 Steel Inorganic materials 0.000 description 2
- 102100032317 Transcription factor Sp7 Human genes 0.000 description 2
- 108010009583 Transforming Growth Factors Proteins 0.000 description 2
- 102000009618 Transforming Growth Factors Human genes 0.000 description 2
- 108700019146 Transgenes Proteins 0.000 description 2
- 108010000134 Vascular Cell Adhesion Molecule-1 Proteins 0.000 description 2
- 108010073929 Vascular Endothelial Growth Factor A Proteins 0.000 description 2
- 102000005789 Vascular Endothelial Growth Factors Human genes 0.000 description 2
- 108010019530 Vascular Endothelial Growth Factors Proteins 0.000 description 2
- 238000002835 absorbance Methods 0.000 description 2
- 238000010521 absorption reaction Methods 0.000 description 2
- 238000009825 accumulation Methods 0.000 description 2
- 239000002253 acid Substances 0.000 description 2
- 239000004480 active ingredient Substances 0.000 description 2
- 230000003321 amplification Effects 0.000 description 2
- 239000005557 antagonist Substances 0.000 description 2
- 239000003963 antioxidant agent Substances 0.000 description 2
- 235000006708 antioxidants Nutrition 0.000 description 2
- 230000008901 benefit Effects 0.000 description 2
- 230000000975 bioactive effect Effects 0.000 description 2
- 230000033558 biomineral tissue development Effects 0.000 description 2
- 230000015572 biosynthetic process Effects 0.000 description 2
- 210000003995 blood forming stem cell Anatomy 0.000 description 2
- 238000010805 cDNA synthesis kit Methods 0.000 description 2
- DEGAKNSWVGKMLS-UHFFFAOYSA-N calcein Chemical compound O1C(=O)C2=CC=CC=C2C21C1=CC(CN(CC(O)=O)CC(O)=O)=C(O)C=C1OC1=C2C=C(CN(CC(O)=O)CC(=O)O)C(O)=C1 DEGAKNSWVGKMLS-UHFFFAOYSA-N 0.000 description 2
- 239000000969 carrier Substances 0.000 description 2
- 239000012876 carrier material Substances 0.000 description 2
- OSASVXMJTNOKOY-UHFFFAOYSA-N chlorobutanol Chemical compound CC(C)(O)C(Cl)(Cl)Cl OSASVXMJTNOKOY-UHFFFAOYSA-N 0.000 description 2
- 230000009816 chondrogenic differentiation Effects 0.000 description 2
- 238000000576 coating method Methods 0.000 description 2
- 230000001332 colony forming effect Effects 0.000 description 2
- 238000002648 combination therapy Methods 0.000 description 2
- 239000003246 corticosteroid Substances 0.000 description 2
- 230000006378 damage Effects 0.000 description 2
- 238000010790 dilution Methods 0.000 description 2
- 239000012895 dilution Substances 0.000 description 2
- 239000003651 drinking water Substances 0.000 description 2
- 235000020188 drinking water Nutrition 0.000 description 2
- 239000003937 drug carrier Substances 0.000 description 2
- 239000003995 emulsifying agent Substances 0.000 description 2
- 229940116977 epidermal growth factor Drugs 0.000 description 2
- 229940126864 fibroblast growth factor Drugs 0.000 description 2
- 239000000834 fixative Substances 0.000 description 2
- 239000000499 gel Substances 0.000 description 2
- 239000008103 glucose Substances 0.000 description 2
- 239000000122 growth hormone Substances 0.000 description 2
- 230000009931 harmful effect Effects 0.000 description 2
- 229940088597 hormone Drugs 0.000 description 2
- 239000005556 hormone Substances 0.000 description 2
- 102000045598 human DPP4 Human genes 0.000 description 2
- 238000009802 hysterectomy Methods 0.000 description 2
- 230000001771 impaired effect Effects 0.000 description 2
- 230000001976 improved effect Effects 0.000 description 2
- 238000011534 incubation Methods 0.000 description 2
- 229960000905 indomethacin Drugs 0.000 description 2
- 239000004615 ingredient Substances 0.000 description 2
- 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 2
- PBGKTOXHQIOBKM-FHFVDXKLSA-N insulin (human) Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)C(=O)N[C@H]1CSSC[C@H]2C(=O)N[C@H](C(=O)N[C@@H](CO)C(=O)N[C@H](C(=O)N[C@H](C(N[C@@H](CO)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC=3C=CC(O)=CC=3)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC=3C=CC(O)=CC=3)C(=O)N[C@@H](CSSC[C@H](NC(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=3C=CC(O)=CC=3)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](C)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=3NC=NC=3)NC(=O)[C@H](CO)NC(=O)CNC1=O)C(=O)NCC(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)NCC(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H]([C@@H](C)O)C(O)=O)C(=O)N[C@@H](CC(N)=O)C(O)=O)=O)CSSC[C@@H](C(N2)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C(C)C)NC(=O)[C@@H](NC(=O)CN)[C@@H](C)CC)[C@@H](C)CC)[C@@H](C)O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC(N)=O)NC(=O)[C@@H](NC(=O)[C@@H](N)CC=1C=CC=CC=1)C(C)C)C1=CN=CN1 PBGKTOXHQIOBKM-FHFVDXKLSA-N 0.000 description 2
- 238000007918 intramuscular administration Methods 0.000 description 2
- 230000001788 irregular Effects 0.000 description 2
- 210000003127 knee Anatomy 0.000 description 2
- 229940039781 leptin Drugs 0.000 description 2
- NRYBAZVQPHGZNS-ZSOCWYAHSA-N leptin Chemical compound O=C([C@H](CO)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CO)NC(=O)CNC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](N)CC(C)C)CCSC)N1CCC[C@H]1C(=O)NCC(=O)N[C@@H](CS)C(O)=O NRYBAZVQPHGZNS-ZSOCWYAHSA-N 0.000 description 2
- 229950008325 levothyroxine Drugs 0.000 description 2
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 description 2
- 238000012423 maintenance Methods 0.000 description 2
- 230000007246 mechanism Effects 0.000 description 2
- 210000003205 muscle Anatomy 0.000 description 2
- 238000003199 nucleic acid amplification method Methods 0.000 description 2
- 229960002378 oftasceine Drugs 0.000 description 2
- 238000003305 oral gavage Methods 0.000 description 2
- 210000000963 osteoblast Anatomy 0.000 description 2
- 210000005009 osteogenic cell Anatomy 0.000 description 2
- 239000012188 paraffin wax Substances 0.000 description 2
- 239000008188 pellet Substances 0.000 description 2
- 102000013415 peroxidase activity proteins Human genes 0.000 description 2
- 108040007629 peroxidase activity proteins Proteins 0.000 description 2
- 230000004962 physiological condition Effects 0.000 description 2
- 102000040430 polynucleotide Human genes 0.000 description 2
- 108091033319 polynucleotide Proteins 0.000 description 2
- 239000002157 polynucleotide Substances 0.000 description 2
- 239000003755 preservative agent Substances 0.000 description 2
- 230000035755 proliferation Effects 0.000 description 2
- 230000000069 prophylactic effect Effects 0.000 description 2
- 239000002510 pyrogen Substances 0.000 description 2
- GZUITABIAKMVPG-UHFFFAOYSA-N raloxifene Chemical compound C1=CC(O)=CC=C1C1=C(C(=O)C=2C=CC(OCCN3CCCCC3)=CC=2)C2=CC=C(O)C=C2S1 GZUITABIAKMVPG-UHFFFAOYSA-N 0.000 description 2
- 230000002829 reductive effect Effects 0.000 description 2
- 238000011160 research Methods 0.000 description 2
- 238000010839 reverse transcription Methods 0.000 description 2
- 230000028327 secretion Effects 0.000 description 2
- 239000011669 selenium Substances 0.000 description 2
- 229910052711 selenium Inorganic materials 0.000 description 2
- 238000000926 separation method Methods 0.000 description 2
- 210000002966 serum Anatomy 0.000 description 2
- 210000000824 sesamoid bone Anatomy 0.000 description 2
- 230000035939 shock Effects 0.000 description 2
- 230000006641 stabilisation Effects 0.000 description 2
- 238000011105 stabilization Methods 0.000 description 2
- 238000007619 statistical method Methods 0.000 description 2
- 239000010959 steel Substances 0.000 description 2
- 210000001562 sternum Anatomy 0.000 description 2
- 239000011550 stock solution Substances 0.000 description 2
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound 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](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 2
- 239000006228 supernatant Substances 0.000 description 2
- 238000007910 systemic administration Methods 0.000 description 2
- 206010043827 tibia fracture Diseases 0.000 description 2
- 238000013518 transcription Methods 0.000 description 2
- 230000035897 transcription Effects 0.000 description 2
- VBEQCZHXXJYVRD-GACYYNSASA-N uroanthelone Chemical compound C([C@@H](C(=O)N[C@H](C(=O)N[C@@H](CS)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CS)C(=O)N[C@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)NCC(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H](CO)C(=O)NCC(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CS)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(O)=O)C(C)C)[C@@H](C)O)NC(=O)[C@H](CO)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@@H](NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H](CCSC)NC(=O)[C@H](CS)NC(=O)[C@@H](NC(=O)CNC(=O)CNC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CS)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)CNC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@H](CO)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CS)NC(=O)CNC(=O)[C@H]1N(CCC1)C(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@@H](N)CC(N)=O)C(C)C)[C@@H](C)CC)C1=CC=C(O)C=C1 VBEQCZHXXJYVRD-GACYYNSASA-N 0.000 description 2
- 239000000080 wetting agent Substances 0.000 description 2
- 239000012224 working solution Substances 0.000 description 2
- XRASPMIURGNCCH-UHFFFAOYSA-N zoledronic acid Chemical compound OP(=O)(O)C(P(O)(O)=O)(O)CN1C=CN=C1 XRASPMIURGNCCH-UHFFFAOYSA-N 0.000 description 2
- DGVVWUTYPXICAM-UHFFFAOYSA-N β‐Mercaptoethanol Chemical compound OCCS DGVVWUTYPXICAM-UHFFFAOYSA-N 0.000 description 2
- KLXQAXYSOJNJRI-KVTDHHQDSA-N (2s,3s,4r,5r)-5-amino-2,3,4,6-tetrahydroxyhexanal Chemical compound OC[C@@H](N)[C@@H](O)[C@H](O)[C@H](O)C=O KLXQAXYSOJNJRI-KVTDHHQDSA-N 0.000 description 1
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 1
- IIZPXYDJLKNOIY-JXPKJXOSSA-N 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCC\C=C/C\C=C/C\C=C/C\C=C/CCCCC IIZPXYDJLKNOIY-JXPKJXOSSA-N 0.000 description 1
- JKMHFZQWWAIEOD-UHFFFAOYSA-N 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 1
- FJKROLUGYXJWQN-UHFFFAOYSA-N 4-hydroxybenzoic acid Chemical compound OC(=O)C1=CC=C(O)C=C1 FJKROLUGYXJWQN-UHFFFAOYSA-N 0.000 description 1
- CKLBXIYTBHXJEH-UHFFFAOYSA-J 75881-23-1 Chemical compound [Cl-].[Cl-].[Cl-].[Cl-].[Cu+2].[N-]1C(N=C2C3=CC=C(CSC(N(C)C)=[N+](C)C)C=C3C(N=C3C4=CC=C(CSC(N(C)C)=[N+](C)C)C=C4C(=N4)[N-]3)=N2)=C(C=C(CSC(N(C)C)=[N+](C)C)C=C2)C2=C1N=C1C2=CC(CSC(N(C)C)=[N+](C)C)=CC=C2C4=N1 CKLBXIYTBHXJEH-UHFFFAOYSA-J 0.000 description 1
- RZVAJINKPMORJF-UHFFFAOYSA-N Acetaminophen Chemical compound CC(=O)NC1=CC=C(O)C=C1 RZVAJINKPMORJF-UHFFFAOYSA-N 0.000 description 1
- 102000007469 Actins Human genes 0.000 description 1
- 108010085238 Actins Proteins 0.000 description 1
- 108010088751 Albumins Proteins 0.000 description 1
- 102000009027 Albumins Human genes 0.000 description 1
- 239000012103 Alexa Fluor 488 Substances 0.000 description 1
- 101100421761 Arabidopsis thaliana GSNAP gene Proteins 0.000 description 1
- 238000011718 B6 albino mouse Methods 0.000 description 1
- 208000008035 Back Pain Diseases 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- 229940122361 Bisphosphonate Drugs 0.000 description 1
- 108010049931 Bone Morphogenetic Protein 2 Proteins 0.000 description 1
- 108010049951 Bone Morphogenetic Protein 3 Proteins 0.000 description 1
- 108010049955 Bone Morphogenetic Protein 4 Proteins 0.000 description 1
- 108010049976 Bone Morphogenetic Protein 5 Proteins 0.000 description 1
- 108010049974 Bone Morphogenetic Protein 6 Proteins 0.000 description 1
- 206010005949 Bone cancer Diseases 0.000 description 1
- 102100028728 Bone morphogenetic protein 1 Human genes 0.000 description 1
- 108090000654 Bone morphogenetic protein 1 Proteins 0.000 description 1
- 102000003928 Bone morphogenetic protein 15 Human genes 0.000 description 1
- 108090000349 Bone morphogenetic protein 15 Proteins 0.000 description 1
- 102100024506 Bone morphogenetic protein 2 Human genes 0.000 description 1
- 102100024504 Bone morphogenetic protein 3 Human genes 0.000 description 1
- 102100024505 Bone morphogenetic protein 4 Human genes 0.000 description 1
- 102100022526 Bone morphogenetic protein 5 Human genes 0.000 description 1
- 102100022525 Bone morphogenetic protein 6 Human genes 0.000 description 1
- 208000018084 Bone neoplasm Diseases 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 206010006895 Cachexia Diseases 0.000 description 1
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 1
- 102000003727 Caveolin 1 Human genes 0.000 description 1
- 108090000026 Caveolin 1 Proteins 0.000 description 1
- 102100032141 Cell death activator CIDE-A Human genes 0.000 description 1
- 101710196996 Cell death activator CIDE-A Proteins 0.000 description 1
- 208000013725 Chronic Kidney Disease-Mineral and Bone disease Diseases 0.000 description 1
- 208000003044 Closed Fractures Diseases 0.000 description 1
- 102000029816 Collagenase Human genes 0.000 description 1
- 108060005980 Collagenase Proteins 0.000 description 1
- 208000024779 Comminuted Fractures Diseases 0.000 description 1
- 208000014311 Cushing syndrome Diseases 0.000 description 1
- 229930105110 Cyclosporin A Natural products 0.000 description 1
- PMATZTZNYRCHOR-CGLBZJNRSA-N Cyclosporin A Chemical compound CC[C@@H]1NC(=O)[C@H]([C@H](O)[C@H](C)C\C=C\C)N(C)C(=O)[C@H](C(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@@H](C)NC(=O)[C@H](C)NC(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)N(C)C(=O)CN(C)C1=O PMATZTZNYRCHOR-CGLBZJNRSA-N 0.000 description 1
- 108010036949 Cyclosporine Proteins 0.000 description 1
- 201000003883 Cystic fibrosis Diseases 0.000 description 1
- 238000000116 DAPI staining Methods 0.000 description 1
- CYCGRDQQIOGCKX-UHFFFAOYSA-N Dehydro-luciferin Natural products OC(=O)C1=CSC(C=2SC3=CC(O)=CC=C3N=2)=N1 CYCGRDQQIOGCKX-UHFFFAOYSA-N 0.000 description 1
- DVJAMEIQRSHVKC-BDAKNGLRSA-N Dutogliptin Chemical compound OB(O)[C@@H]1CCCN1C(=O)CN[C@H]1CNCC1 DVJAMEIQRSHVKC-BDAKNGLRSA-N 0.000 description 1
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 1
- LVGKNOAMLMIIKO-UHFFFAOYSA-N Elaidinsaeure-aethylester Natural products CCCCCCCCC=CCCCCCCCC(=O)OCC LVGKNOAMLMIIKO-UHFFFAOYSA-N 0.000 description 1
- 241000283086 Equidae Species 0.000 description 1
- 241000283074 Equus asinus Species 0.000 description 1
- 206010015548 Euthanasia Diseases 0.000 description 1
- LCDDAGSJHKEABN-MLGOLLRUSA-N Evogliptin Chemical compound C1CNC(=O)[C@@H](COC(C)(C)C)N1C(=O)C[C@H](N)CC1=CC(F)=C(F)C=C1F LCDDAGSJHKEABN-MLGOLLRUSA-N 0.000 description 1
- 101150018889 FABP4 gene Proteins 0.000 description 1
- RZSYLLSAWYUBPE-UHFFFAOYSA-L Fast green FCF Chemical compound [Na+].[Na+].C=1C=C(C(=C2C=CC(C=C2)=[N+](CC)CC=2C=C(C=CC=2)S([O-])(=O)=O)C=2C(=CC(O)=CC=2)S([O-])(=O)=O)C=CC=1N(CC)CC1=CC=CC(S([O-])(=O)=O)=C1 RZSYLLSAWYUBPE-UHFFFAOYSA-L 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 102100037362 Fibronectin Human genes 0.000 description 1
- 108010067306 Fibronectins Proteins 0.000 description 1
- BJGNCJDXODQBOB-UHFFFAOYSA-N Fivefly Luciferin Natural products OC(=O)C1CSC(C=2SC3=CC(O)=CC=C3N=2)=N1 BJGNCJDXODQBOB-UHFFFAOYSA-N 0.000 description 1
- 208000036119 Frailty Diseases 0.000 description 1
- 108010004460 Gastric Inhibitory Polypeptide Proteins 0.000 description 1
- 102100039994 Gastric inhibitory polypeptide Human genes 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- KOSRFJWDECSPRO-WDSKDSINSA-N Glu-Glu Chemical compound OC(=O)CC[C@H](N)C(=O)N[C@@H](CCC(O)=O)C(O)=O KOSRFJWDECSPRO-WDSKDSINSA-N 0.000 description 1
- 102400000322 Glucagon-like peptide 1 Human genes 0.000 description 1
- DTHNMHAUYICORS-KTKZVXAJSA-N Glucagon-like peptide 1 Chemical compound C([C@@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CCCCN)C(=O)NCC(=O)N[C@@H](CCCNC(N)=N)C(N)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CCCCN)NC(=O)[C@H](C)NC(=O)[C@H](C)NC(=O)[C@H](CCC(N)=O)NC(=O)CNC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@H](CC=1C=CC=CC=1)NC(=O)[C@@H](NC(=O)CNC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C)NC(=O)[C@@H](N)CC=1N=CNC=1)[C@@H](C)O)[C@@H](C)O)C(C)C)C1=CC=CC=C1 DTHNMHAUYICORS-KTKZVXAJSA-N 0.000 description 1
- 101800000224 Glucagon-like peptide 1 Proteins 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- 108010043121 Green Fluorescent Proteins Proteins 0.000 description 1
- 102000004144 Green Fluorescent Proteins Human genes 0.000 description 1
- 206010018720 Greenstick fracture Diseases 0.000 description 1
- 102100038367 Gremlin-1 Human genes 0.000 description 1
- 108010041881 Growth Differentiation Factor 10 Proteins 0.000 description 1
- 108010090290 Growth Differentiation Factor 2 Proteins 0.000 description 1
- 108010090293 Growth Differentiation Factor 3 Proteins 0.000 description 1
- 108010090254 Growth Differentiation Factor 5 Proteins 0.000 description 1
- 108010090250 Growth Differentiation Factor 6 Proteins 0.000 description 1
- 102000004858 Growth differentiation factor-9 Human genes 0.000 description 1
- 108090001086 Growth differentiation factor-9 Proteins 0.000 description 1
- 102100040895 Growth/differentiation factor 10 Human genes 0.000 description 1
- 102100040892 Growth/differentiation factor 2 Human genes 0.000 description 1
- 102100035364 Growth/differentiation factor 3 Human genes 0.000 description 1
- 102100035379 Growth/differentiation factor 5 Human genes 0.000 description 1
- 102100035368 Growth/differentiation factor 6 Human genes 0.000 description 1
- 102100035363 Growth/differentiation factor 7 Human genes 0.000 description 1
- 101710204283 Growth/differentiation factor 7 Proteins 0.000 description 1
- 102100039939 Growth/differentiation factor 8 Human genes 0.000 description 1
- 239000007995 HEPES buffer Substances 0.000 description 1
- 102000001554 Hemoglobins Human genes 0.000 description 1
- 108010054147 Hemoglobins Proteins 0.000 description 1
- 101000899368 Homo sapiens Bone morphogenetic protein 8B Proteins 0.000 description 1
- 101001032872 Homo sapiens Gremlin-1 Proteins 0.000 description 1
- 101000939438 Homo sapiens Mitochondrial brown fat uncoupling protein 1 Proteins 0.000 description 1
- 101000684208 Homo sapiens Prolyl endopeptidase FAP Proteins 0.000 description 1
- 101000651373 Homo sapiens Serine palmitoyltransferase small subunit B Proteins 0.000 description 1
- 101000617130 Homo sapiens Stromal cell-derived factor 1 Proteins 0.000 description 1
- 206010020590 Hypercalciuria Diseases 0.000 description 1
- 208000001953 Hypotension Diseases 0.000 description 1
- HEFNNWSXXWATRW-UHFFFAOYSA-N Ibuprofen Chemical compound CC(C)CC1=CC=C(C(C)C(O)=O)C=C1 HEFNNWSXXWATRW-UHFFFAOYSA-N 0.000 description 1
- 102000006496 Immunoglobulin Heavy Chains Human genes 0.000 description 1
- 108010019476 Immunoglobulin Heavy Chains Proteins 0.000 description 1
- 238000012404 In vitro experiment Methods 0.000 description 1
- 206010061218 Inflammation Diseases 0.000 description 1
- 108090001061 Insulin Proteins 0.000 description 1
- 102000004877 Insulin Human genes 0.000 description 1
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 1
- NNJVILVZKWQKPM-UHFFFAOYSA-N Lidocaine Chemical compound CCN(CC)CC(=O)NC1=C(C)C=CC=C1C NNJVILVZKWQKPM-UHFFFAOYSA-N 0.000 description 1
- OYHQOLUKZRVURQ-HZJYTTRNSA-N Linoleic acid Chemical compound CCCCC\C=C/C\C=C/CCCCCCCC(O)=O OYHQOLUKZRVURQ-HZJYTTRNSA-N 0.000 description 1
- DDWFXDSYGUXRAY-UHFFFAOYSA-N Luciferin Natural products CCc1c(C)c(CC2NC(=O)C(=C2C=C)C)[nH]c1Cc3[nH]c4C(=C5/NC(CC(=O)O)C(C)C5CC(=O)O)CC(=O)c4c3C DDWFXDSYGUXRAY-UHFFFAOYSA-N 0.000 description 1
- 238000000585 Mann–Whitney U test Methods 0.000 description 1
- 101710151321 Melanostatin Proteins 0.000 description 1
- 101100539369 Mus musculus Ucp1 gene Proteins 0.000 description 1
- 241000282339 Mustela Species 0.000 description 1
- 108010056852 Myostatin Proteins 0.000 description 1
- 206010028980 Neoplasm Diseases 0.000 description 1
- 102400000064 Neuropeptide Y Human genes 0.000 description 1
- CTQNGGLPUBDAKN-UHFFFAOYSA-N O-Xylene Chemical compound CC1=CC=CC=C1C CTQNGGLPUBDAKN-UHFFFAOYSA-N 0.000 description 1
- 208000008589 Obesity Diseases 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- 208000010191 Osteitis Deformans Diseases 0.000 description 1
- 208000001164 Osteoporotic Fractures Diseases 0.000 description 1
- 108010016731 PPAR gamma Proteins 0.000 description 1
- 101150014691 PPARA gene Proteins 0.000 description 1
- 208000027868 Paget disease Diseases 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 229930182555 Penicillin Natural products 0.000 description 1
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 1
- 102000035195 Peptidases Human genes 0.000 description 1
- 108091005804 Peptidases Proteins 0.000 description 1
- 239000000132 Peptide PHI Substances 0.000 description 1
- 108010084214 Peptide PHI Proteins 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- 239000004365 Protease Substances 0.000 description 1
- 102000014128 RANK Ligand Human genes 0.000 description 1
- 108010025832 RANK Ligand Proteins 0.000 description 1
- 108091030071 RNAI Proteins 0.000 description 1
- 238000011529 RT qPCR Methods 0.000 description 1
- 241000700159 Rattus Species 0.000 description 1
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 1
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 1
- 102100027676 Serine palmitoyltransferase small subunit B Human genes 0.000 description 1
- 206010061363 Skeletal injury Diseases 0.000 description 1
- 108020004459 Small interfering RNA Proteins 0.000 description 1
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 description 1
- 208000005250 Spontaneous Fractures Diseases 0.000 description 1
- 208000013201 Stress fracture Diseases 0.000 description 1
- 102100021669 Stromal cell-derived factor 1 Human genes 0.000 description 1
- 238000000692 Student's t-test Methods 0.000 description 1
- 241000282887 Suidae Species 0.000 description 1
- 241000053227 Themus Species 0.000 description 1
- 102100036407 Thioredoxin Human genes 0.000 description 1
- 101710120037 Toxin CcdB Proteins 0.000 description 1
- 102100040396 Transcobalamin-1 Human genes 0.000 description 1
- 101710124861 Transcobalamin-1 Proteins 0.000 description 1
- 102000040945 Transcription factor Human genes 0.000 description 1
- 108091023040 Transcription factor Proteins 0.000 description 1
- 102000004887 Transforming Growth Factor beta Human genes 0.000 description 1
- 108090001012 Transforming Growth Factor beta Proteins 0.000 description 1
- 239000007983 Tris buffer Substances 0.000 description 1
- 239000013504 Triton X-100 Substances 0.000 description 1
- 229920004890 Triton X-100 Polymers 0.000 description 1
- 206010067584 Type 1 diabetes mellitus Diseases 0.000 description 1
- 241000021375 Xenogenes Species 0.000 description 1
- 101710185494 Zinc finger protein Proteins 0.000 description 1
- 102100023597 Zinc finger protein 816 Human genes 0.000 description 1
- PVNJLUVGTFULAE-UHFFFAOYSA-N [NH4+].[Cl-].[K] Chemical compound [NH4+].[Cl-].[K] PVNJLUVGTFULAE-UHFFFAOYSA-N 0.000 description 1
- 230000001133 acceleration Effects 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 230000004913 activation Effects 0.000 description 1
- 239000008186 active pharmaceutical agent Substances 0.000 description 1
- 239000013543 active substance Substances 0.000 description 1
- 229940037127 actonel Drugs 0.000 description 1
- 230000006978 adaptation Effects 0.000 description 1
- 238000011374 additional therapy Methods 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 238000001042 affinity chromatography Methods 0.000 description 1
- 230000032683 aging Effects 0.000 description 1
- 235000004279 alanine Nutrition 0.000 description 1
- OGSPWJRAVKPPFI-UHFFFAOYSA-M alendronate(1-) Chemical compound NCCCC(O)(P(O)(O)=O)P(O)([O-])=O OGSPWJRAVKPPFI-UHFFFAOYSA-M 0.000 description 1
- KOSRFJWDECSPRO-UHFFFAOYSA-N alpha-L-glutamyl-L-glutamic acid Natural products OC(=O)CCC(N)C(=O)NC(CCC(O)=O)C(O)=O KOSRFJWDECSPRO-UHFFFAOYSA-N 0.000 description 1
- 125000000539 amino acid group Chemical group 0.000 description 1
- 239000003708 ampul Substances 0.000 description 1
- 229950009977 anagliptin Drugs 0.000 description 1
- LDXYBEHACFJIEL-HNNXBMFYSA-N anagliptin Chemical compound C=1N2N=C(C)C=C2N=CC=1C(=O)NCC(C)(C)NCC(=O)N1CCC[C@H]1C#N LDXYBEHACFJIEL-HNNXBMFYSA-N 0.000 description 1
- 230000000202 analgesic effect Effects 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 230000000844 anti-bacterial effect Effects 0.000 description 1
- 230000000692 anti-sense effect Effects 0.000 description 1
- 239000003429 antifungal agent Substances 0.000 description 1
- 229940121375 antifungal agent Drugs 0.000 description 1
- 229960005475 antiinfective agent Drugs 0.000 description 1
- 239000004599 antimicrobial Substances 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- 239000007864 aqueous solution Substances 0.000 description 1
- YBHILYKTIRIUTE-UHFFFAOYSA-N berberine Chemical compound C1=C2CC[N+]3=CC4=C(OC)C(OC)=CC=C4C=C3C2=CC2=C1OCO2 YBHILYKTIRIUTE-UHFFFAOYSA-N 0.000 description 1
- 229940093265 berberine Drugs 0.000 description 1
- QISXPYZVZJBNDM-UHFFFAOYSA-N berberine Natural products COc1ccc2C=C3N(Cc2c1OC)C=Cc4cc5OCOc5cc34 QISXPYZVZJBNDM-UHFFFAOYSA-N 0.000 description 1
- 150000004663 bisphosphonates Chemical class 0.000 description 1
- 210000004369 blood Anatomy 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- 210000001124 body fluid Anatomy 0.000 description 1
- 210000002449 bone cell Anatomy 0.000 description 1
- 230000037182 bone density Effects 0.000 description 1
- 230000010478 bone regeneration Effects 0.000 description 1
- 238000007469 bone scintigraphy Methods 0.000 description 1
- 229940028101 boniva Drugs 0.000 description 1
- 210000001593 brown adipocyte Anatomy 0.000 description 1
- 239000007975 buffered saline Substances 0.000 description 1
- 238000010804 cDNA synthesis Methods 0.000 description 1
- 201000011510 cancer Diseases 0.000 description 1
- 229960004424 carbon dioxide Drugs 0.000 description 1
- 235000011089 carbon dioxide Nutrition 0.000 description 1
- 239000006143 cell culture medium Substances 0.000 description 1
- 230000004709 cell invasion Effects 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 230000004663 cell proliferation Effects 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 229960001927 cetylpyridinium chloride Drugs 0.000 description 1
- YMKDRGPMQRFJGP-UHFFFAOYSA-M cetylpyridinium chloride Chemical compound [Cl-].CCCCCCCCCCCCCCCC[N+]1=CC=CC=C1 YMKDRGPMQRFJGP-UHFFFAOYSA-M 0.000 description 1
- 238000012512 characterization method Methods 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 229960004926 chlorobutanol Drugs 0.000 description 1
- 210000001612 chondrocyte Anatomy 0.000 description 1
- 238000003501 co-culture Methods 0.000 description 1
- 229960002424 collagenase Drugs 0.000 description 1
- 239000003086 colorant Substances 0.000 description 1
- 230000000295 complement effect Effects 0.000 description 1
- 239000002131 composite material Substances 0.000 description 1
- 230000001010 compromised effect Effects 0.000 description 1
- 238000013270 controlled release Methods 0.000 description 1
- 238000001816 cooling Methods 0.000 description 1
- 230000002596 correlated effect Effects 0.000 description 1
- 229960001334 corticosteroids Drugs 0.000 description 1
- 238000004925 denaturation Methods 0.000 description 1
- 229960001251 denosumab Drugs 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 230000001627 detrimental effect Effects 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 230000018109 developmental process Effects 0.000 description 1
- UGMCXQCYOVCMTB-UHFFFAOYSA-K dihydroxy(stearato)aluminium Chemical compound CCCCCCCCCCCCCCCCCC(=O)O[Al](O)O UGMCXQCYOVCMTB-UHFFFAOYSA-K 0.000 description 1
- 239000003603 dipeptidyl peptidase IV inhibitor Substances 0.000 description 1
- 208000035475 disorder Diseases 0.000 description 1
- 239000002270 dispersing agent Substances 0.000 description 1
- 231100000673 dose–response relationship Toxicity 0.000 description 1
- 238000001647 drug administration Methods 0.000 description 1
- 229950003693 dutogliptin Drugs 0.000 description 1
- 230000001674 effect on adipogenesis Effects 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 238000005538 encapsulation Methods 0.000 description 1
- 230000003511 endothelial effect Effects 0.000 description 1
- 210000003038 endothelium Anatomy 0.000 description 1
- 239000003623 enhancer Substances 0.000 description 1
- YQGOJNYOYNNSMM-UHFFFAOYSA-N eosin Chemical compound [Na+].OC(=O)C1=CC=CC=C1C1=C2C=C(Br)C(=O)C(Br)=C2OC2=C(Br)C(O)=C(Br)C=C21 YQGOJNYOYNNSMM-UHFFFAOYSA-N 0.000 description 1
- 210000003743 erythrocyte Anatomy 0.000 description 1
- BEFDCLMNVWHSGT-UHFFFAOYSA-N ethenylcyclopentane Chemical compound C=CC1CCCC1 BEFDCLMNVWHSGT-UHFFFAOYSA-N 0.000 description 1
- LVGKNOAMLMIIKO-QXMHVHEDSA-N ethyl oleate Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OCC LVGKNOAMLMIIKO-QXMHVHEDSA-N 0.000 description 1
- 229940093471 ethyl oleate Drugs 0.000 description 1
- 229940085363 evista Drugs 0.000 description 1
- 229950011259 evogliptin Drugs 0.000 description 1
- 235000019197 fats Nutrition 0.000 description 1
- 210000002950 fibroblast Anatomy 0.000 description 1
- 230000003328 fibroblastic effect Effects 0.000 description 1
- 230000003176 fibrotic effect Effects 0.000 description 1
- 239000000796 flavoring agent Substances 0.000 description 1
- 238000002073 fluorescence micrograph Methods 0.000 description 1
- OGBMKVWORPGQRR-UHFFFAOYSA-N forteo Chemical compound C=1NC=NC=1CC(C(=O)NC(CC(C)C)C(=O)NC(CC(N)=O)C(=O)NC(CO)C(=O)NC(CCSC)C(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NC(C(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC=1C2=CC=CC=C2NC=1)C(=O)NC(CC(C)C)C(=O)NC(CCCNC(N)=N)C(=O)NC(CCCCN)C(=O)NC(CCCCN)C(=O)NC(CC(C)C)C(=O)NC(CCC(N)=O)C(=O)NC(CC(O)=O)C(=O)NC(C(C)C)C(=O)NC(CC=1N=CNC=1)C(=O)NC(CC(N)=O)C(=O)NC(CC=1C=CC=CC=1)C(O)=O)NC(=O)C(CCCCN)NC(=O)CNC(=O)C(CC(C)C)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(CCSC)NC(=O)C(CC(C)C)NC(=O)C(CCC(N)=O)NC(=O)C(NC(=O)C(CCC(O)=O)NC(=O)C(CO)NC(=O)C(NC(=O)C(N)CO)C(C)C)C(C)CC)CC1=CNC=N1 OGBMKVWORPGQRR-UHFFFAOYSA-N 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 230000009368 gene silencing by RNA Effects 0.000 description 1
- 239000011521 glass Substances 0.000 description 1
- 239000003862 glucocorticoid Substances 0.000 description 1
- 239000003292 glue Substances 0.000 description 1
- 108010055341 glutamyl-glutamic acid Proteins 0.000 description 1
- 208000002566 gonadal dysgenesis Diseases 0.000 description 1
- 210000004349 growth plate Anatomy 0.000 description 1
- 230000003394 haemopoietic effect Effects 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 238000007417 hierarchical cluster analysis Methods 0.000 description 1
- 210000001981 hip bone Anatomy 0.000 description 1
- 230000013632 homeostatic process Effects 0.000 description 1
- 239000003688 hormone derivative Substances 0.000 description 1
- 210000002758 humerus Anatomy 0.000 description 1
- 230000036543 hypotension Effects 0.000 description 1
- 229940015872 ibandronate Drugs 0.000 description 1
- 229960001680 ibuprofen Drugs 0.000 description 1
- 238000010191 image analysis Methods 0.000 description 1
- 239000012216 imaging agent Substances 0.000 description 1
- 238000003364 immunohistochemistry Methods 0.000 description 1
- 238000012744 immunostaining Methods 0.000 description 1
- 238000010874 in vitro model Methods 0.000 description 1
- 238000005462 in vivo assay Methods 0.000 description 1
- 230000004054 inflammatory process Effects 0.000 description 1
- 229940125396 insulin Drugs 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 238000007913 intrathecal administration Methods 0.000 description 1
- 239000007951 isotonicity adjuster Substances 0.000 description 1
- 239000000787 lecithin Substances 0.000 description 1
- 235000010445 lecithin Nutrition 0.000 description 1
- 229940067606 lecithin Drugs 0.000 description 1
- 229960004194 lidocaine Drugs 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- OYHQOLUKZRVURQ-IXWMQOLASA-N linoleic acid Natural products CCCCC\C=C/C\C=C\CCCCCCCC(O)=O OYHQOLUKZRVURQ-IXWMQOLASA-N 0.000 description 1
- 235000020778 linoleic acid Nutrition 0.000 description 1
- 239000002502 liposome Substances 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 239000012669 liquid formulation Substances 0.000 description 1
- 238000011068 loading method Methods 0.000 description 1
- 239000003589 local anesthetic agent Substances 0.000 description 1
- 230000033001 locomotion Effects 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 239000000314 lubricant Substances 0.000 description 1
- PKGKOZOYXQMJNG-UHFFFAOYSA-N lupeol Natural products CC(=C)C1CC2C(C)(CCC3C4(C)CCC5C(C)(C)C(O)CCC5(C)C4CCC23C)C1 PKGKOZOYXQMJNG-UHFFFAOYSA-N 0.000 description 1
- MQYXUWHLBZFQQO-QGTGJCAVSA-N lupeol Chemical compound C1C[C@H](O)C(C)(C)[C@@H]2CC[C@@]3(C)[C@]4(C)CC[C@@]5(C)CC[C@@H](C(=C)C)[C@@H]5[C@H]4CC[C@@H]3[C@]21C MQYXUWHLBZFQQO-QGTGJCAVSA-N 0.000 description 1
- 230000002934 lysing effect Effects 0.000 description 1
- 235000019359 magnesium stearate Nutrition 0.000 description 1
- 230000014759 maintenance of location Effects 0.000 description 1
- 208000027202 mammary Paget disease Diseases 0.000 description 1
- 210000004373 mandible Anatomy 0.000 description 1
- 238000002078 massotherapy Methods 0.000 description 1
- 230000035800 maturation Effects 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 208000030159 metabolic disease Diseases 0.000 description 1
- 230000004060 metabolic process Effects 0.000 description 1
- 210000001872 metatarsal bone Anatomy 0.000 description 1
- CWWARWOPSKGELM-SARDKLJWSA-N methyl (2s)-2-[[(2s)-2-[[2-[[(2s)-2-[[(2s)-2-[[(2s)-5-amino-2-[[(2s)-5-amino-2-[[(2s)-1-[(2s)-6-amino-2-[[(2s)-1-[(2s)-2-amino-5-(diaminomethylideneamino)pentanoyl]pyrrolidine-2-carbonyl]amino]hexanoyl]pyrrolidine-2-carbonyl]amino]-5-oxopentanoyl]amino]-5 Chemical compound C([C@@H](C(=O)NCC(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCSC)C(=O)OC)NC(=O)[C@H](CC=1C=CC=CC=1)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CCCCN)NC(=O)[C@H]1N(CCC1)C(=O)[C@@H](N)CCCN=C(N)N)C1=CC=CC=C1 CWWARWOPSKGELM-SARDKLJWSA-N 0.000 description 1
- 239000003094 microcapsule Substances 0.000 description 1
- 239000011859 microparticle Substances 0.000 description 1
- 238000007431 microscopic evaluation Methods 0.000 description 1
- 238000000386 microscopy Methods 0.000 description 1
- 230000000921 morphogenic effect Effects 0.000 description 1
- 239000002105 nanoparticle Substances 0.000 description 1
- 230000001537 neural effect Effects 0.000 description 1
- 239000012457 nonaqueous media Substances 0.000 description 1
- 238000012758 nuclear staining Methods 0.000 description 1
- URPYMXQQVHTUDU-OFGSCBOVSA-N nucleopeptide y Chemical compound C([C@@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(N)=O)NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](C)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)CNC(=O)[C@H]1N(CCC1)C(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CCCCN)NC(=O)[C@H](CO)NC(=O)[C@H]1N(CCC1)C(=O)[C@@H](N)CC=1C=CC(O)=CC=1)C1=CC=C(O)C=C1 URPYMXQQVHTUDU-OFGSCBOVSA-N 0.000 description 1
- 235000020824 obesity Nutrition 0.000 description 1
- 235000008390 olive oil Nutrition 0.000 description 1
- 239000004006 olive oil Substances 0.000 description 1
- 229950000074 omarigliptin Drugs 0.000 description 1
- MKMPWKUAHLTIBJ-ISTRZQFTSA-N omarigliptin Chemical compound C1([C@H]2OC[C@@H](C[C@@H]2N)N2CC3=CN(N=C3C2)S(=O)(=O)C)=CC(F)=CC=C1F MKMPWKUAHLTIBJ-ISTRZQFTSA-N 0.000 description 1
- 150000002895 organic esters Chemical class 0.000 description 1
- 210000004409 osteocyte Anatomy 0.000 description 1
- 208000005368 osteomalacia Diseases 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 210000004417 patella Anatomy 0.000 description 1
- 230000001991 pathophysiological effect Effects 0.000 description 1
- 210000004197 pelvis Anatomy 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- YLBIOQUUAYXLJJ-WZUUGAJWSA-N peptide histidine methionine Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CO)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCSC)C(N)=O)NC(=O)[C@H](CCCCN)NC(=O)[C@H](CCCCN)NC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCC(N)=O)NC(=O)CNC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCCCN)NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C=CC=CC=1)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@H](CC=1C=CC=CC=1)NC(=O)[C@@H](NC(=O)CNC(=O)[C@H](CC(O)=O)NC(=O)[C@H](C)NC(=O)[C@@H](N)CC=1N=CNC=1)C(C)C)[C@@H](C)O)C1=CC=C(O)C=C1 YLBIOQUUAYXLJJ-WZUUGAJWSA-N 0.000 description 1
- 239000002304 perfume Substances 0.000 description 1
- 230000008823 permeabilization Effects 0.000 description 1
- 239000000825 pharmaceutical preparation Substances 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- 238000000554 physical therapy Methods 0.000 description 1
- 239000000419 plant extract Substances 0.000 description 1
- 108010017843 platelet-derived growth factor A Proteins 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 229920002704 polyhistidine Polymers 0.000 description 1
- 229920005862 polyol Polymers 0.000 description 1
- 150000003077 polyols Chemical class 0.000 description 1
- 230000008092 positive effect Effects 0.000 description 1
- 239000000843 powder Substances 0.000 description 1
- 210000000229 preadipocyte Anatomy 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 201000009395 primary hyperaldosteronism Diseases 0.000 description 1
- 238000012545 processing Methods 0.000 description 1
- 239000000047 product Substances 0.000 description 1
- 229940092597 prolia Drugs 0.000 description 1
- 230000002035 prolonged effect Effects 0.000 description 1
- 125000001500 prolyl group Chemical group [H]N1C([H])(C(=O)[*])C([H])([H])C([H])([H])C1([H])[H] 0.000 description 1
- 238000011321 prophylaxis Methods 0.000 description 1
- 235000019419 proteases Nutrition 0.000 description 1
- 230000002685 pulmonary effect Effects 0.000 description 1
- 230000002285 radioactive effect Effects 0.000 description 1
- 229960004622 raloxifene Drugs 0.000 description 1
- 238000003753 real-time PCR Methods 0.000 description 1
- 230000010837 receptor-mediated endocytosis Effects 0.000 description 1
- 229940107023 reclast Drugs 0.000 description 1
- 230000007115 recruitment Effects 0.000 description 1
- 230000008929 regeneration Effects 0.000 description 1
- 238000011069 regeneration method Methods 0.000 description 1
- 201000006409 renal osteodystrophy Diseases 0.000 description 1
- 230000003252 repetitive effect Effects 0.000 description 1
- 208000007442 rickets Diseases 0.000 description 1
- 229940089617 risedronate Drugs 0.000 description 1
- 229960004586 rosiglitazone Drugs 0.000 description 1
- 210000001991 scapula Anatomy 0.000 description 1
- 231100000241 scar Toxicity 0.000 description 1
- 230000035945 sensitivity Effects 0.000 description 1
- 238000007493 shaping process Methods 0.000 description 1
- 238000009097 single-agent therapy Methods 0.000 description 1
- 210000003625 skull Anatomy 0.000 description 1
- 235000019333 sodium laurylsulphate Nutrition 0.000 description 1
- 210000004872 soft tissue Anatomy 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 235000010199 sorbic acid Nutrition 0.000 description 1
- 229940075582 sorbic acid Drugs 0.000 description 1
- 239000004334 sorbic acid Substances 0.000 description 1
- 239000007921 spray Substances 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 239000008227 sterile water for injection Substances 0.000 description 1
- 230000000638 stimulation Effects 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- 210000002536 stromal cell Anatomy 0.000 description 1
- 238000010254 subcutaneous injection Methods 0.000 description 1
- YCUVUDODLRLVIC-VPHDGDOJSA-N sudan black b Chemical compound C1=CC(=C23)NC(C)(C)NC2=CC=CC3=C1\N=N\C(C1=CC=CC=C11)=CC=C1\N=N\C1=CC=CC=C1 YCUVUDODLRLVIC-VPHDGDOJSA-N 0.000 description 1
- 235000000346 sugar Nutrition 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- 230000008093 supporting effect Effects 0.000 description 1
- 239000004094 surface-active agent Substances 0.000 description 1
- 239000000375 suspending agent Substances 0.000 description 1
- 239000003765 sweetening agent Substances 0.000 description 1
- 230000002195 synergetic effect Effects 0.000 description 1
- 239000008399 tap water Substances 0.000 description 1
- 235000020679 tap water Nutrition 0.000 description 1
- ZRKFYGHZFMAOKI-QMGMOQQFSA-N tgfbeta Chemical compound C([C@H](NC(=O)[C@H](C(C)C)NC(=O)CNC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CC(C)C)NC(=O)CNC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](NC(=O)[C@H](C)NC(=O)[C@H](C)NC(=O)[C@@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](N)CCSC)C(C)C)[C@@H](C)CC)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](C)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC(N)=O)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(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](C)C(=O)N[C@@H](CC(C)C)C(=O)N1[C@@H](CCC1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(C)C)C(O)=O)C1=CC=C(O)C=C1 ZRKFYGHZFMAOKI-QMGMOQQFSA-N 0.000 description 1
- 239000002562 thickening agent Substances 0.000 description 1
- 108060008226 thioredoxin Proteins 0.000 description 1
- 229940094937 thioredoxin Drugs 0.000 description 1
- 210000000115 thoracic cavity Anatomy 0.000 description 1
- 238000011200 topical administration Methods 0.000 description 1
- 230000000699 topical effect Effects 0.000 description 1
- 239000012049 topical pharmaceutical composition Substances 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 239000003053 toxin Substances 0.000 description 1
- 231100000765 toxin Toxicity 0.000 description 1
- 108700012359 toxins Proteins 0.000 description 1
- 238000011830 transgenic mouse model Methods 0.000 description 1
- 238000013519 translation Methods 0.000 description 1
- 230000032258 transport Effects 0.000 description 1
- 230000008733 trauma Effects 0.000 description 1
- 229950010728 trelagliptin Drugs 0.000 description 1
- IWYJYHUNXVAVAA-OAHLLOKOSA-N trelagliptin Chemical compound C=1C(F)=CC=C(C#N)C=1CN1C(=O)N(C)C(=O)C=C1N1CCC[C@@H](N)C1 IWYJYHUNXVAVAA-OAHLLOKOSA-N 0.000 description 1
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 1
- 238000002604 ultrasonography Methods 0.000 description 1
- 108010071304 univin Proteins 0.000 description 1
- 235000015112 vegetable and seed oil Nutrition 0.000 description 1
- 239000008158 vegetable oil Substances 0.000 description 1
- 229960001254 vildagliptin Drugs 0.000 description 1
- SYOKIDBDQMKNDQ-XWTIBIIYSA-N vildagliptin Chemical compound C1C(O)(C2)CC(C3)CC1CC32NCC(=O)N1CCC[C@H]1C#N SYOKIDBDQMKNDQ-XWTIBIIYSA-N 0.000 description 1
- 238000001262 western blot Methods 0.000 description 1
- 210000000636 white adipocyte Anatomy 0.000 description 1
- 239000008096 xylene Substances 0.000 description 1
- 229960004276 zoledronic acid Drugs 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/13—Amines
- A61K31/155—Amidines (), e.g. guanidine (H2N—C(=NH)—NH2), isourea (N=C(OH)—NH2), isothiourea (—N=C(SH)—NH2)
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/40—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/40—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
- A61K31/403—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/496—Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/4985—Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
- A61K31/513—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
- A61K31/519—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
- A61K31/519—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
- A61K31/52—Purines, e.g. adenine
- A61K31/522—Purines, e.g. adenine having oxo groups directly attached to the heterocyclic ring, e.g. hypoxanthine, guanine, acyclovir
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- 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
- A61P19/08—Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
-
- 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
- A61P19/08—Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
- A61P19/10—Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
-
- 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
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0652—Cells of skeletal and connective tissues; Mesenchyme
- C12N5/0654—Osteocytes, Osteoblasts, Odontocytes; Bones, Teeth
Definitions
- Bone breaks An average of 8 million bone breaks occurs in the United States each year. Most bone fractures heal without issues. However, about 3-10% of these breaks are slow to heal or do not heal at all with traditional methods. In particular, about 5-10% of all bone fractures also display non-union and/or delayed unions, which have a substantial economic impact and substantial effects on quality of life. Fracture healing is considered to be a complicated metabolic process that requires the interaction of many factors, including the recruitment of reparative cells and expression of corresponding genes. If these factors are inadequate or interrupted, healing is delayed or impaired, resulting in a nonunion of the bone. The causes of nonunions or delayed healings of fractures are usually unknown.
- the present invention relates to a DPP-4 inhibitor for use in treating a bone fracture in a subject in need thereof.
- the present invention relates to a DPP-4 inhibitor for use in preventing non-union of a bone fracture or preventing healing complications following bone fracture in a subject in need thereof.
- the present invention relates to a DPP-4 inhibitor for use in promoting fracture healing in a subject in need thereof.
- the present invention relates to a pharmaceutical composition
- a pharmaceutical composition comprising a DPP-4 inhibitor for use in treating a bone fracture in a subject in need thereof, wherein said pharmaceutical composition further comprises a pharmaceutically-acceptable diluent, excipient, or carrier, and wherein the DPP-4 inhibitor is present in an effective amount to treat or prevent bone fractures.
- the present invention relates to a pharmaceutical composition
- a pharmaceutical composition comprising a DPP-4 inhibitor for use in preventing non-union of a bone fracture or in preventing healing complications following bone fracture in a subject in need thereof, wherein said pharmaceutical composition further comprises a pharmaceutically-acceptable diluent, excipient, or carrier, and wherein the DPP-4 inhibitor is present in an effective amount to treat or prevent bone fractures.
- the bone fracture is a non-union bone fracture, a compound fracture or a fracture with delayed healing.
- the DPP-4 inhibitor for use according to the first, second or third aspect, or of the pharmaceutical composition for use according to the fourth or fifth aspect, is administered systemically.
- the DPP-4 inhibitor for use according to the first, second or third aspect, or of the pharmaceutical composition for use according to the fourth or fifth aspect, the DPP-4 inhibitor is administered locally.
- the DPP-4 inhibitor for use according to the first, second or third aspect, or of the pharmaceutical composition for use according to the fourth or fifth aspect, the DPP-4 inhibitor is administered to the site of fracture.
- the DPP-4 inhibitor for use according to the first, second or third aspect, or of the pharmaceutical composition for use according to the fourth or fifth aspect, is administered in one or more doses over a period of less than 6 months.
- the DPP-4 inhibitor for use according to the first, second or third aspect, or of the pharmaceutical composition for use according to the fourth or fifth aspect, is administered in one or more doses over a period of less than 3 months.
- the DPP-4 inhibitor for use according to the first, second or third aspect, or of the pharmaceutical composition for use according to the fourth or fifth aspect, is administered in one or more doses over a period of less than 1 month.
- the present invention relates to the DPP-4 inhibitor for use according to the first, second or third aspect, or to the pharmaceutical composition for use according to the fourth or fifth aspect, wherein, prior to diagnosis of the bone fracture, the subject was not receiving a DPP-4 inhibitor.
- the present invention relates to the DPP-4 inhibitor for use according to the first, second or third aspect, or to the pharmaceutical composition for use according to the fourth or fifth aspect, wherein the subject in need thereof was not, prior to the bone fracture, diagnosed with or treated for Type II diabetes.
- the subject in need thereof is over age 65.
- the subject in need thereof is under age 40.
- the subject in need thereof has a BMI less than 25.
- the subject in need thereof has a BMI of greater than or equal to 25 and less than 30.
- the subject in need thereof has a BMI of equal to or greater than 30.
- the present invention relates to the DPP-4 inhibitor for use according to the first, second or third aspect, or to the pharmaceutical composition for use according to the fourth or fifth aspect, wherein the DPP-4 inhibitor is administered in one or more doses, and wherein at least one of the doses is administered locally during surgery to set the fracture.
- DPP-4 inhibitor for use according to the first, second or third aspect, or of the pharmaceutical composition for use according to the fourth or fifth aspect, wherein the subject in need thereof is a human subject.
- the healing complications are osteoporosis related healing complications to bone fracture.
- the DPP-4 inhibitor is selected from one or more of alogliptin, linagliptin, saxagliptin, sitagliptin, vildapliptin, gemigliptin, or teneligliptin.
- the use further comprises administering metformin, in the same or a different formulation as the DPP-4 inhibitor.
- the use further comprises administering one or more other therapeutic agent.
- the present invention relates to a DPP-4 inhibitor for use in reducing the inhibitory effects of marrow adipose tissue (MAT) on fracture healing in a subject in need thereof.
- MAT marrow adipose tissue
- the present invention relates to a method of preparing cells for transplantation, comprising
- OPCs osteogenic progenitor cells
- mesenchymal stem cells OPCs
- the present invention relates to a method of preparing cells for transplantation, comprising
- OPCs osteogenic progenitor cells
- the present invention relates to DPP-4 treated cells prepared by the method of the seventh or eights aspect for use in transplantation to a fracture in a subject in need thereof.
- the present invention relates to a DPP-4 inhibitor for use in preventing loss of bone mineral density (BMD) in an astronaut or other individuals exposed to an altered gravity environment.
- BMD bone mineral density
- the astronaut is exposed to an altered gravity environment for greater than one week, and the DPP-4 inhibitor is administered to the astronaut prior to and/or during and/or after the exposure.
- the present invention relates to a DPP-4 inhibitor for use in preventing loss of bone mineral density (BMD) in a subject in need thereof.
- BMD bone mineral density
- Figure 1A shows flow cytometric separation of CD45 CD31 " (upper and lower left quadrants in dot plot) cells by Seal -selection.
- Figure IB shows Oil Red-0 (Adipogenesis), Alizarin Red S (Osteogenesis) and Alician Blue (Chondrogenesis) staining of Scal + and Seal " cells differentiated under corresponding conditions.
- Figure 1C shows FACS analysis plot of CD45 " CD31 " Scal + cells separated by CD24 expression.
- Figure ID shows Oil Red-0 (Adipogenesis), Alizarin Red S (Osteogenesis) and Alician Blue (Chondrogenesis) staining of CD45 " CD31 " Scal + CD24 + and CD45 CD31 " Scal + CD24 " cells differentiated under corresponding conditions.
- Figure 2A shows FACS-analysis of viable cells from 2-month old male -eGFP reporter mice for expression of GFP followed by Seal and CD45/CD31 expression analysis within GFP + cells.
- Figure 2B shows adipogenic (Oil Red O) and osteogenic (Alizarin Red S) differentiation assays of CD45 " CD31 " Scal + Pa + and CD45 " CD31 " Scal " Pa + populations.
- Figure 5 shows flow cytometric dot plot analyses of bone resident CD45 " CD31 " Scal " Pa + cells (left panel) and bone resident CD45 " CD31 " Scal " Pa " cells separated into CD24 " and CD24 + cells by FACS.
- Figure 6 shows FACS-analysis of CD45 CD31 " cells from 2-month old male Zfp423- eGFP reporter mice for expression of Seal and GFP.
- Figure 9 upper panel shows quantification of osteogenic Scal " Pa+, multipotent Scal + Pa + CD24 + and adipogenic Scal + Pa + CD24 " cells in metaphysis or diaphysis of bones derived from -eGFP mice.
- Figure 10 shows quantification of bone marrow- localized Pa-GFP+ cells associated to blood vessels with diameters smaller or larger than 10 ⁇ (upper panel).
- Figure 12A shows transgene alleles of the rep AdlLuc reporter mouse strain:
- the Zfp423- eGFP reporter mouse strain was crossed to a strain expressing Cre-recombinase under control of the Adiponectin promoter (Adipoq-Cre) and a constitutive Luciferase (Luc)-reporter where the Luc-encoding cDNA is suppressed by a loxP-flanked Stop-signal.
- Adipoq-Cre Adipoq-Cre
- Luc constitutive Luciferase
- FIG. 12B shows transgene alleles of the rep tdTom reporter mouse strain: The Zfp423-eGFV reporter was crossed to an mTmG-reporter mouse strain without presence of a Cre-transgene. Thus, the cells maintained constitutive red fluorescence and can be detected by immunofluorescence for tdTomato or by in vivo imaging.
- Figure 13 shows the FACS-gating strategy for the isolation of the four investigated cell populations (PreAd, APC, CD45 CD31 Scal + CD24 + , OPC) from both the rep AdiLuc reporter mouse strain and the rep tdTom reporter mouse strain for subsequent in vivo transplantation assays.
- Figure 14 shows in vivo luciferase imaging (top panels) and macroscopic identification (arrows; middle panels) of transplants 8 weeks after sternal s.c. -injection of the indicated cell populations isolated from repAdiLuc mice. The lower panels show microscopy of corresponding Movat-Pentachrome stains (yellow: mineralized structure; blue: cartilage; purple: nuclei). Scale bars, 30 ⁇ .
- Figure 15 shows FACS-analysis of transplants of initially CD45 CD31 Scal + CD24 + cells identified by tdTomato-expression giving rise to the CD45 ⁇ CD31 ⁇ Scal + CD24 ⁇ population within the transplant.
- Figure 16 shows a summary of sternal transplantation experiments of the four investigated cell populations including numbers of transplanted animals and respective differentiation fates as determined by histological analysis and engraftment efficiency.
- Figure 17 shows a summary of the four investigated cell populations with phenotypic marker expression and differentiation potential performances during in vitro and in vivo experiments.
- markers that are required to define and isolate the respective populations by flow cytometry are labeled in bold.
- Figure 19 shows a western blot analysis of UCP1 protein with ⁇ -Actin as a loading control measured in differentiated CD45 " CD31 " Scal + populations isolated from inguinal white adipose tissue (iWAT), brown adipose tissue (BAT), bone or muscle after adipogenic differentiation in the presence of the browning agent rosiglitazone (Rosi).
- Figure 22 shows hematoxylin and eosin (H&E) stains of femora from 2-month old or
- Figure 23B shows quantification of multipotent CD45 " CD31 " Scal + CD24 + , adipogenic
- CD45 CD31 Scal + CD24- (APC), and osteogenic CD45 CD31 Scal-CD24-Pa + (OPC) subpopulations in 2-month old and 25 -month old male mice fed SD (white bars) or high fat diet for 10 days (lOdHFD) (black bars) (n 9). Results are shown as mean ⁇ SEM (****p ⁇ 0.0001).
- SD standard diet
- LdHFD 1 day high fat diet
- Figure 28 shows red fluorescence in tibiae (top panels) and ⁇ CT images (lower panels) of fracture calluses 14 days after fracture and intratibial injection of the indicated cell populations.
- Figure 29 shows flow cytometric analysis of fracture calluses two weeks after surgery either injected with bone-derived Scal + Pa + cells or Scal " Pa + cells isolated from animals constitutively expressing GFP. Shown are viable cells previously gated for CD45 " CD31 " to show retention of cells after transplantation.
- Figure 3 IB provides immuno florescence showing the contribution of transplanted multipotent CD45 CD31 Scal + CD24 + (upper panels) and osteogenic CD45 CD31 Scal Pa + (OPC, lower panels) cell populations to osteochondrogenic structures in the fractured tibiae that were not observed in adipogenic cell transplants (Red: tdTomato; Blue: DAPI; right panels indicate merge of immuno florescence and light microscopic images). Scale bar, 20 ⁇ .
- Figure 31C provides immuno florescence showing the contribution of transplanted multipotent CD45 " CD31 " Scal + CD24 + (upper panels) and osteogenic CD45 CD3 rScal Pa + (OPC, lower panels) cell populations to endosteal bone linings in the fractured tibiae (Red: tdTomato; Blue: DAPI; dotted lines indicate areas of compact bone as seen in right-side panels of merged immunoflorescence and light microscopic images). Scale bar, 10 ⁇ .
- Figure 3 ID provides immunoflorescence co-staining of tdTomato + cells (red fluorescence) with Osteocalcin to show an osteogenic differentiation fate of transplanted multipotent CD45 " CD31 " Scal + CD24 + (upper panels) osteogenic CD31 " CD45 “ Scal Pa + (lower panels) cell populations. No co-staining detected in adipogenesis-committed populations, e.g. APCs and preAds (not shown). Scale bar, 10 um.
- Figure 3 IE provides immunoflorescence co-staining of tdTomato+ cells (red fluorescence) with Aggrecan to show a chondrogenic differentiation fate of transplanted multipotent CD45 CD31 Scal + CD24 + (upper panels) osteogenic CD45 CD31 Scal Pa + (lower panels) cell populations. No co-staining detected in adipogenesis-committed populations, e.g. APCs and preAds (not shown). Scale bar, 10 um.
- Figure 32 shows characterization results of RNA-Seq samples with read counts (left panel) and the fraction of reads mapped to exons (right panel).
- Figure 33A shows the Principal Component Analysis (PCA) of the RNA-seq samples.
- Figure 33B shows the correlation scores of top 10 genes driving PCI and PC2 in Figure 33 A.
- Figure 33C shows hierarchical clustering analyses of RNA-Seq data from all four cell populations.
- Figure 34 shows a heat map of selected differentially expressed (DE) genes, divided by candidates reported in the literature (known, asterisks indicate no significant DE between individual groups) and novel markers, enriched in CD45 ⁇ CD31 ⁇ Scal + CD24 + cell populations.
- DE differentially expressed
- Figure 35 shows a heat map of selected differentially expressed (DE) genes, divided by candidates reported in the literature (known, asterisks indicate no significant DE between individual groups) and novel markers, enriched in OPC cell populations.
- Figure 36 shows a heat map of selected differentially expressed (DE) genes, divided by candidates reported in the literature (known, asterisks indicate no significant DE between individual groups) and novel markers, enriched in APC cell populations.
- DE differentially expressed
- Figure 37 shows a heat map of selected differentially expressed (DE) genes, divided by candidates reported in the literature (known, asterisks indicate no significant DE between individual groups) and novel markers, enriched in preAd cell populations.
- Figure 38 shows gene expression intensities of Dpp4 from RNA-Seq analysis. Mean ⁇ SEM; *p ⁇ 0.05, **p ⁇ 0.01, ****p ⁇ 0.0001.
- FIG. 39 shows FACS analysis of DPP4/CD26 surface marker expression in
- Figure 42A shows mRNA expression (as a percentage of control) of Runx2 and Osterix
- Figure 43A shows Oil Red-0 staining of CD45 CD31 Scal + CD24 + and APCs either treated with PBS or Sitagliptin (100 ⁇ ) during adipogenic differentiation.
- Figure 47A shows representative Movat Pentachrome stains of fracture calluses from control PBS-treated mice that received osteogenic (PBS/OPC) or adipogenic (PBS/APC) intratibial transplants and animals treated with Sitaglitpin for 1 week after fracture and receiving the same transplants of osteogenic (Sita/OPC) or adipogenic (Sita/APC) cells.
- PBS/OPC osteogenic
- PBS/APC adipogenic
- Figure 47B shows quantification from histomorphometric analysis of fracture calluses from control PBS-treated mice that received osteogenic (PBS/OPC) or adipogenic (PBS/APC) intratibial transplants and animals treated with Sitaglitpin for 1 week after fracture and receiving the same transplants of osteogenic (Sita/OPC) or adipogenic (Sita/APC) cells.
- Figure 48 shows FACS analysis of bone-resident MSCs, APCs, and OPCs from animals treated with Sitagliptin (Sita) or control (Ctrl) for 3 days at a dose of 10 mg/kg body weight, either by intraperitoneal injections (i.p.) or by oral gavage (per oral - p.o.). Results are shown as mean ⁇ SEM.
- the disclosure provides dipeptidyl peptidase-4 (DPP-4) inhibitor agents (which may be referred to herein simply as “agents” or “DPP-4 inhibitors” or “gliptins”) for use in any of the methods or compositions described herein.
- the agents are for use in treating a subject having a bone fracture.
- the agent is a DPP-4 inhibitor.
- the agent is for use in promoting fracture healing in a subject in need thereof (e.g., a subject having a fracture; a subject diagnosed with a fracture).
- the agent is for use in decreasing or preventing complications following fracture, such as non-union.
- the agent is for use in reducing inhibitory effects of marrow adipose tissue (MAT) on fracture healing in a subject in need thereof.
- the agent is for use in preventing loss of bone density.
- the disclosure provides methods of preparing cells for transplantation into a subject, wherein the cells have been treated with the agent in an amount effective to increase osteogenic gene expression and differentiation, e.g. osteogenic (or chondrogenic) bone cell formation. Such cells are suitable for transplantation into a subject having a bone fracture, such as to promote fracture healing or to prevent or decrease complications from bone fracture. Other uses and methods are described in further detail herein.
- compositions comprising a DPP-4 inhibitor, and any such compositions may be used in any of the methods described herein.
- formulations of a DPP-4 inhibitor to be applied systemically or topically/locally in or onto the fracture and any such compositions, such as to promote fracture healing or to prevent or decrease complications from bone fracture.
- the term “has the ability” or “is capable of is meant that the recited agent, proteins or polypeptides will carry out the stated bioactivity under suitable conditions (e.g., physiological conditions or standard laboratory conditions).
- suitable conditions e.g., physiological conditions or standard laboratory conditions.
- the term “can” may be used to describe this ability (e.g., “can bind” or “binds” to a given sequence).
- inhibitor when used to refer to any of the agents disclosed herein, mean that the agent is capable of blocking, reducing, attenuating and/or reversing activation of the protein targeted by the agent (e.g., DPP-4).
- a DPP-4 inhibitor or antagonist is an agent that is capable of blocking, reducing, attenuating and or reversing DPP-4 serine exopeptidase activity.
- the agent for use in any of the methods disclosed herein is a DPP-
- the DPP-4 inhibitor is a small organic molecule. In some embodiments, the DPP-4 inhibitor is a polypeptide or peptide. In some embodiments, the DPP- 4 inhibitor is an antibody (e.g., an antibody that binds to and inhibits the activity of DPP-4). In some embodiments, the DPP-4 inhibitor is a polynucleotide.
- the agent is isolated and/or purified.
- Any of the agents described herein, including those provided in an isolated or purified form, may be provided as a composition, such as a composition comprising an agent formulated with one or more pharmaceutical and/or physiological acceptable carriers and/or excipients.
- Any of the agents described herein, including compositions may be used in any of the methods described herein. Examples of particular pharmaceutical compositions formulated for preferred routes of delivery are provided herein.
- the agents inhibit a biological activity of DPP-4. In some embodiments, the agent binds to DPP-4 and inhibits a biological activity of DPP-4. In some embodiments, the agent binds to another protein or agent and indirectly inhibits a biological activity of DPP-4. In some embodiments, the agent binds to a substrate of DPP-4 and prevents DPP-4 from interacting with that substrate. In some embodiments, the agent is an antibody or antigen-binding fragment that binds to DPP-4 or a DPP-4 substrate in a manner that prevents DPP-4 or the substrate from interacting with each other.
- the agent is capable of binding to a DPP-4 protein, or fragment thereof, having an amino acid sequence that is at least 80%, 85%, 90%, 92%, 95%, 97% or 100% identical to the amino acid sequence of SEQ ID NO: 1 or 3, or a fragment thereof.
- the agent is capable of binding to the caveolin-1 binding domain, the fibronectin binding domain and/or the ADA binding domain of DPP-4.
- the agent is capable of binding to the amino acid residues corresponding to residues 201-211 and/or 603 of SEQ ID NO: 1.
- the agent is capable of binding to residues GWSYG of SEQ ID NO: 1.
- the agent is capable of binding to residues corresponding to Ser630, Asp708 and/or His740 of SEQ ID NO: 1. In some embodiments, the agent inhibits or prevents homodimerization of DPP-4. In some embodiments, the agent inhibits secretion of DPP-4, such as from cells expressing DPP-4 on their surface in or near a fracture site.
- the agent inhibits the expression of DPP-4. In some embodiments, the agent inhibits transcription of the DPP4 gene. In some embodiments, the agent inhibits translation of the DPP4 mRNA transcript. In some embodiments, the agent is capable of binding to a polynucleotide having a nucleotide sequence that is at least 80%>, 85%, 90%, 92%, 95%, 97% or 100% identical to the nucleotide sequence of SEQ ID NO: 2 or 4, or a portion or complement thereof.
- the agent is an antisense molecule, an RNAi molecule, an siRNA or a CRISPR-based therapeutic agent (e.g., a CRISPR/Cas9 complex) that inhibits DPP4 expression.
- the agent inhibits the expression or activity of DPP-4 in a cell by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100% as compared to DPP-4 expression or activity in the same or substantially the same cell in the same or substantially the same conditions in the absence of the agent.
- biological activity By the terms “biological activity”, “bioactivity”, “bioactive” or “functional”, when used in the context of DPP-4, is meant the ability of the DPP-4 polypeptide to carry out one or more functions associated with wildtype DPP-4 polypeptides ⁇ e.g., a polypeptide having the amino acid sequence of SEQ ID NO: 1), for example, serine exopeptidase activity.
- wildtype DPP-4 polypeptides ⁇ e.g., a polypeptide having the amino acid sequence of SEQ ID NO: 1), for example, serine exopeptidase activity.
- biological activity Bioactivity
- bioactivity biological activity
- functional are used interchangeably herein.
- any of the DPP-4 inhibitors disclosed herein is capable of inhibiting activity by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100% of the DPP-4 activity as compared to DPP-4 activity under the same or substantially the same physiological conditions in the absence of the agent.
- the DPP-4 inhibitor preserves the action of DPP-4 substrate molecules, e.g., glucagon- like peptide- 1, gastric inhibitory polypeptide, peptide histidine methionine, substance P, neuropeptide Y, CXCL12, and other molecules typically containing alanine or proline residues in the second aminoterminal position.
- treatment with DPP-4 inhibitors prolongs the duration of action of DPP-4 peptide substrates and increases levels of their intact, undegraded forms.
- any of the agents disclosed herein decreases the half-life (tm) of a DPP-4 polypeptide.
- the half-life of the DPP-4 polypeptide is decreased by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% relative to the half-life of the DPP-4 polypeptide in the absence of the agent.
- the protein half- life is determined in vitro, such as in a buffered saline solution or in serum.
- the protein half-life is an in vivo half life, such as the half-life of the DPP-4 in the serum or other bodily fluid of an animal.
- a DPP-4 inhibitor is also intended to comprise active metabolites and prodrugs thereof, such as active metabolites and prodrugs of DPP-4 inhibitors.
- a “metabolite” is an active derivative of a DPP-4 inhibitor produced when the DPP-4 inhibitor is metabolized.
- a “prodrug” is a compound that is either metabolized to a DPP-4 inhibitor or is metabolized to the same metabolite(s) as a DPP-4 inhibitor.
- the term "a DPP-4 inhibitor” is also intended to comprise pharmaceutical salts thereof.
- DPP-4 inhibitors are known in the art.
- representative DPP- 4 inhibitors are disclosed in WO 98/19998, DE19616 486 Al, WO 00/34241, WO 95/15309, WO 01/72290, WO01/52825, WO03/002553, WO 9310127, WO 99/61431, WO 9925719, WO 9938501, WO 9946272, WO 9967278, WO 9967279, WO 02053548, WO 02067918, WO 02066627, WO 02/068420, W0 02083128, WO 2004/037181, WO 0168603, EP1258480, WO 0181337, WO 02083109, WO 030003250, WO 03035067, WO 03/035057, US2003216450, WO 99/46272, WO 0197808, WO 03002553, WO 01/34594, WO 02051836, EP1245568, EP
- the agent e.g., the DPP-4 inhibitor
- the agent is or comprises alogliptin, sitagliptin, vildagliptin, saxagliptin, gemigliptin, anagliptin, teneligliptin, trelagliptin, omarigliptin, evogliptin, dutogliptin and/or linagliptin or derivatives or pharmaceutically acceptable salts thereof.
- the agent is or comprises diprotin A, berberine and/or lupeol, or a derivative or pharmaceutically acceptable salt thereof.
- the agent is or comprises any natural plant extract or bioactive compounds which inhibit DPP-4 activity, or a derivative or pharmaceutically acceptable salt thereof. Such agents may be provided as pharmaceutical compositions and, as noted above, as prodrugs.
- the agent is combined (e.g., in the same or different formulation) with metformin.
- the methods of the present disclosure include administering a DPP-4 inhibitor and metformin, either as a co-formulation or in separate formulations. When administered as separate formulations, the two may be administered at the same or different times and via the same or different routes of administration.
- metformin is not used (e.g., the methods of the disclosure do not include administering metformin).
- any of the methods disclosed herein may include treatment with a single DPP-4 inhibitor or with more than one DPP-4 inhibitor, such as two DPP-4 inhibitors that act via different mechanisms of action.
- the two agents may be administered at the same or different times and via the same or different routes of administration.
- the methods further include metformin.
- any of the agents disclosed herein is conjugated to a heterologous agent.
- the heterologous agents include, but are not limited to, polyhistidine, Glu-Glu, glutathione S transferase (GST), thioredoxin, protein A, protein G, and an immunoglobulin heavy chain constant region (Fc), maltose binding protein (MBP), which are particularly useful for isolation of the agents by affinity chromatography.
- the agent is conjugated to a detectable moiety.
- the moiety is a fluorescently labeled or radiolabeled detectable moiety.
- Various delivery systems are known and can be used to administer any of the agents of the disclosure, such as any of the DPP-4 inhibitors of the disclosure, e.g., various formulations, encapsulation in liposomes, nanoparticles, microparticles, microcapsules, recombinant cells capable of expressing the compound, receptor-mediated endocytosis (see, e.g., Wu and Wu, 1987, J. Biol. Chem. 262:4429-4432).
- Methods of introduction can be enteral or parenteral, including but not limited to, intraosseous, intradermal, transdermal, intramuscular, intraperitoneal, intravenous, subcutaneous, pulmonary, intranasal, intrathecal, intraocular, epidural, and oral routes.
- parenteral introduction includes intramuscular, subcutaneous, intravenous, intravascular, and intrapericardial administration.
- the agents are administered locally to the subject.
- the agents are administered systemically to the subject.
- the agents are administered directly to a bone fracture site, e.g., during a surgical procedure or when setting a compound fracture.
- the agents are administered as a topical formulation (e.g., as a gel formulation) to the site of the bone fracture.
- routes of administration are combined.
- a DPP4-inhibitor can be administered locally, such as during a surgical procedure to set a bone.
- Subsequent doses of agent over subsequent days, such as over 1-4 weeks, may be by systemic administration (e.g., oral, intravenous, subcutaneous, or intraperitoneal).
- systemic administration e.g., oral, intravenous, subcutaneous, or intraperitoneal.
- the same route of administration may be used throughout a multi-dose regimen, such as systemic administration over 1-4 weeks.
- the agents are administered by means of a device implanted within the subject.
- the implantable device is coated by a composite surface coating comprising any of the DPP-4 inhibitor agents disclosed herein.
- the implantable device delivers drug to the site of bone fracture.
- the implantable device releases DPP-4 inhibitor in a controlled fashion (e.g., a controlled release device).
- the agents of the disclosure are administered in one or more doses over a period of less than 1 year, less than or equal to 9 months, less than or equal to 6 months, less than or equal to 3 months, less than or equal to 1 month, less than or equal to 3 weeks, less than or equal to 2 weeks, or less than or equal to 1 week.
- the total treatment period e.g., 1, 3, 6 months, etc.
- the subject is administered the agent on a dosing schedule.
- the schedule may be daily, every other day, twice weekly, weekly, twice monthly or monthly.
- each infusion is part of an overall treatment plan where a composition of the disclosure is administered according to a regular schedule (e.g., weekly, monthly, etc.).
- the agents of the disclosure are prepared in a formulation/composition appropriate for a specific route of administration.
- the composition and route of administration is chosen depending on the particular use of the technology. For example, a different composition and/or route of administration may be appropriate when using the compositions of the disclosure for research purposes, such as in vitro or in an animal model, versus when using for diagnostic or therapeutic purposes in human patients.
- One of skill in the art can select the appropriate route of administration depending on the particular application of the technology.
- compositions of the disclosure for use in the methods of the present disclosure can be determined by standard clinical techniques and may vary depending on the particular indication or use. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.
- Dosages may be determined by techniques known to those of skill in the art or as taught herein. Toxicity and therapeutic efficacy of any of the agents disclosed herein may be determined by standard pharmaceutical procedures in experimental animals.
- agents of the disclosure are formulated with a pharmaceutically acceptable carrier.
- the disclosure provides a composition comprising an agent of the disclosure formulated with one or more pharmaceutically acceptable carriers and/or excipients.
- Such pharmaceutical compositions include, where applicable, pharmaceutically acceptable salts of a DPP-4 inhibitor.
- the disclosure provides for a pharmaceutical composition for use in treating a bone fracture in a subject in need thereof, comprising a DPP-4 inhibitor in admixture with a pharmaceutically-acceptable diluent, excipient, or carrier, wherein the DPP-4 inhibitor is present in an effective amount to treat or prevent bone fractures.
- the disclosure provides for a pharmaceutical composition for use in preventing non-union of a bone fracture or for use in preventing healing complications following bone fracture in a subject in need thereof, comprising a DPP-4 inhibitor in admixture with a pharmaceutically-acceptable diluent, excipient, or carrier, wherein the DPP-4 inhibitor is present in an effective amount to treat or prevent bone fractures.
- compositions of the disclosure can be administered alone or as a component of a pharmaceutical formulation (composition).
- compositions of the disclosure may be formulated for administration in any convenient way for use in human or veterinary medicine.
- Wetting agents, emulsifiers and lubricants, such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.
- Formulations of the compositions of the disclosure include those suitable for oral, nasal, topical, parenteral, rectal, and/or intravaginal administration.
- the disclosure provides for a composition for administration directly to a bone fracture, e.g., by a liquid formulation applied directly to the site of bone fracture ⁇ e.g., by a spray).
- the disclosure provides for a gel-based formulation for direct or topical administration.
- the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy.
- the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated and the particular mode of administration.
- the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect.
- methods of preparing these formulations or compositions include combining the therapeutic agent and a carrier and, optionally, one or more accessory ingredients.
- the formulations can be prepared with a liquid carrier, or a finely divided solid carrier, or both, and then, if necessary, shaping the product.
- compositions suitable for parenteral administration may comprise one or more compositions of the disclosure in combination with one or more pharmaceutically acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers ⁇ e.g., HEPES buffer), bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
- sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers ⁇ e.g., HEPES buffer), bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
- aqueous and nonaqueous carriers examples include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate.
- polyols such as glycerol, propylene glycol, polyethylene glycol, and the like
- vegetable oils such as olive oil
- injectable organic esters such as ethyl oleate.
- Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
- compositions may also contain adjuvants, such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption, such as aluminum monostearate and gelatin.
- adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents.
- Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium
- the compositions of the disclosure are formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings.
- the composition may also include a solubilizing agent and a local anesthetic such as lidocaine to ease pain at the site of the injection.
- a solubilizing agent such as lidocaine to ease pain at the site of the injection.
- the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
- an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
- compositions of the disclosure are non-pyrogenic.
- the compositions are substantially pyrogen free.
- the formulations of the disclosure are pyrogen- free formulations which are substantially free of endotoxins and/or related pyrogenic substances.
- Endotoxins include toxins that are confined inside a microorganism and are released only when the microorganisms are broken down or die.
- Pyrogenic substances also include fever-inducing, thermostable substances (glycoproteins) from the outer membrane of bacteria and other microorganisms. Both of these substances can cause fever, hypotension and shock if administered to humans. Due to the potential harmful effects, even low amounts of endotoxins must be removed from intravenously administered pharmaceutical drug solutions.
- FDA Food & Drug Administration
- EU endotoxin units
- the endotoxin and pyrogen levels in the composition are less then 10 EU/mg, or less then 5 EU/mg, or less then 1 EU/mg, or less then 0.1 EU/mg, or less then 0.01 EU/mg, or less then 0.001 EU/mg.
- compositions, methods and uses described herein specifically contemplates any combination of the features of compositions of the present disclosure (alone or in combination) with the features described for the various pharmaceutical compositions and route of administration described in this section.
- compositions of the disclosure contemplates the use of any of the compositions of the disclosure (whether alone or in combination with any of the additional therapeutic treatments disclosed herein).
- Compositions of the disclosure may be described based on any combination of structural and/or functional features provided herein.
- the disclosure contemplates the combination of any step or steps of one method or use with any step or steps from another method or use.
- These methods and uses involve administering to an individual in need thereof an effective amount of a compound of the disclosure (including as a pharmaceutical composition) appropriate for the particular disease or condition (e.g., a bone fracture).
- these methods and uses involve delivering any of the agents disclosed herein to the cells of a subject in need thereof.
- the disclosure provides uses in treating a bone fracture in a subject in need thereof. In some embodiments, the disclosure provides a use in preventing nonunion of a bone fracture in a subject in need thereof. In some embodiments, the disclosure provides a use in preventing healing complications following bone fracture in a subject in need thereof. In some embodiments, the disclosure provides a use in promoting fracture healing in a subject in need thereof. In some embodiments, the disclosure provides a use in reducing the inhibitory effects of marrow adipose tissue (MAT) on fracture healing in a subject in need thereof. In some embodiments, the disclosure provides a use in preventing loss of bone mineral density (BMD) in a subject (e.g. , an astronaut) exposed to an altered gravity environment.
- BMD bone mineral density
- any of the methods and uses disclosed herein comprises administering to the subject an effective amount of any of the DPP-4 inhibitors disclosed herein. In some embodiments, any of the methods and uses disclosed herein comprises administering an effective amount of any of the agents disclosed herein (alone or in combination with any of the additional therapeutic treatments disclosed herein), to a subject in need thereof according to a dosing regimen (e.g., a dose and dosing schedule) and/or dosing schedule.
- a dosing regimen e.g., a dose and dosing schedule
- a method or use may comprise administering a DPP-4 inhibitor to a subject in need thereof, such as a subject having a fracture, in one or more doses over a period of time (e.g., a single administration or multiple administrations over a period of less than or equal to 6, 3, 2, 1 month).
- a period of time e.g., a single administration or multiple administrations over a period of less than or equal to 6, 3, 2, 1 month.
- treatment generally mean obtaining a desired pharmacologic and/or physiologic effect, and may also be used to refer to improving, alleviating, and/or decreasing the severity of one or more symptoms of a condition being treated.
- the effect may be prophylactic in terms of completely or partially delaying the onset or recurrence of a disease, condition, or symptoms thereof, and/or may be therapeutic in terms of a partial or complete cure for a disease or condition and/or adverse effect attributable to the disease or condition.
- Treatment covers any treatment of a disease or condition of a mammal, particularly a human, and includes: (a) preventing complications from a fracture or other bone related disease or condition, such as non-union; (b) preventing further complications of an existing disease or condition (e.g. preventing formation of non-union fractures in osteoporosis patients or preventing healing complications in osteoporosis patients having a fracture); (c) inhibiting the disease or condition (e.g., arresting its development or further progression); (d) relieving the disease or condition (e.g., causing regression of the disease or condition, providing improvement in one or more symptoms); promoting healing of a tissue (e.g. , bone) that was damaged or compromised as a result of the disease or condition (e.g. , a bone fracture) ; or (e) acceleration of the healing process or decrease of processes that inhibit the healing process .
- a tissue e.g. , bone
- the disease or condition e.g. , a bone fracture
- the disclosure provides a use in treating a bone-related disease (e.g. , osteoporosis) or bone-related condition (e.g., a bone fracture) or preventing or decreasing complications associated with a bone fracture with any of the agents described herein (alone or in combination with any of the additional therapeutic treatments disclosed herein).
- a bone-related disease e.g. , osteoporosis
- bone-related condition e.g., a bone fracture
- the subject is a human.
- the disclosure provides a use in treating a therapeutically induced bone loss or bone-related disease such as those induced by medication (e.g. corticosteroid-induced bone loss) or a medical procedure (e.g. patients who have undergone a hysterectomy) or preventing or decreasing complications associated with a bone fracture with any of the agents described herein (alone or in combination with any of the additional therapeutic treatments disclosed herein).
- the subject is human. Treating a bone-related disease (e.g., osteoporosis) or bone-related condition (e.g. , a bone fracture) in a subject refers to improving (e.g.
- a bone -related disease e.g., osteoporosis
- bone-related condition e.g., osteoporosis
- a bone fracture) or secondary complications arising after bone injury includes any one or more of: improved or accelerated healing of a bone fracture, reduction in pain associated with a bone fracture (e.g., back pain), preventing height loss, improving posture, reducing frequency of bone fractures, or increasing bone mineral density (BMD) to prevent fractures and to decrease general proneness to bone fractures in subjects (for instance in patients with metabolic diseases such as diabetes and obesity, or in subjects affected by osteoporosis, or in subjects of advanced age alone, or subjects primarily affected by a combination of these disorders and advanced age).
- BMD bone mineral density
- the effects of administration of any of the agents disclosed herein may be determined by assessing the treated subject before and after treatment, and determining whether the treatment has any effect on the bone-related disease (e.g., osteoporosis) or bone-related condition (e.g., a bone fracture).
- the bone-related disease e.g., osteoporosis
- bone-related condition e.g., a bone fracture
- Efficacy can be measured, for example, by: the ability of a bone to heal from a fracture; a reduction in pain experienced by the subject due to the bone fracture; assessing the time for a bone to heal from a fracture or the time for pain associated with a bone fracture to be alleviated; an improvement in posture or height loss. This can be assessed by means of instruments such as X-rays and/or bone scans, or by asking the patient about their degree of pain associated with the site of injury.
- a “therapeutically effective amount” or “effective amount” of a composition is a predetermined amount calculated to achieve the desired result (e.g. , effective in promoting bone fracture healing).
- the activity contemplated by the present methods and uses includes both medical therapeutic and/or prophylactic treatment, as appropriate.
- the specific dose of an agent administered according to this invention to obtain therapeutic and/or prophylactic effects will, of course, be determined by the particular circumstances surrounding the case, including, for example, the agent administered, the route of administration, and the condition being treated.
- a therapeutically effective amount of agent of this invention is typically an amount such that when it is administered in a physiologically tolerable excipient composition, it is sufficient.
- Therapeutically effective amounts may be administered according to a dosing schedule such that the amount of each dose is effective such that, in the aggregate, the combined doses achieve an end result. Each dose is still considered effective even if it adding to an overall therapeutic effect.
- a "patient,” “subject” or “individual” are used interchangeably and refer to either a human or non-human animal.
- the term includes mammals such as humans.
- Bone-related disease e.g. , osteoporosis
- bone-related condition e.g. , a bone fracture
- the mammal is human.
- the mammal is postnatal.
- the mammal is pediatric.
- the subject is at a life stage that is associated with a greater risk of developing a bone-related condition such as osteoporosis.
- the mammal is adult.
- the subject is a human that is at least 50 years in age, at least 60 years in age, at least 65 years in age, at least 70 years in age, at least 80 years of age, at least 90 years of age, at least 100 years of age, at least 1 10 years of age or at least 120 years of age.
- the subject is a human subject under 40 years of age, under 35 years of age, under 30 years of age, or under 20 years of age.
- the subject is a human subject that is still growing (e.g., whose growth plates have not yet fused).
- the subject treated with any of the DPP-4 inhibitors disclosed herein is not a diabetic patient.
- the subject in need thereof has Type I or Type II diabetes.
- the subject was not, prior to the bone fracture, diagosed with or treated for Type II diabetes.
- the subject was not receiving a DPP-4 inhibitor prior to the diagnosis with a bone fracture.
- the subject has not previously received a treatment for diabetes.
- the subject has been administered metformin. In some embodiments, the subject has not been and/or is not being administered metformin.
- the subject has a body mass index (BMI) less than 50, less than 45, less than 40, less than 39, less than 38, less than 37, less than 36, less than 35, less than 34, less than 33, less than 32, less than 31 , less than 30, less than 29, less than 28, less than 27, less than 26 or less than 25.
- BMI body mass index
- the subject has a BMI of greater than or equal to 25 and less than 30.
- the subject has a BMI of equal to or greater than 30.
- the subject has a BMI of 18.5- 24.
- the subject has a BMI of less than 18.5.
- the subject to be treated with any of the DPP-4 inhibitors disclosed herein is a subject having increased marrow adipose tissue (MAT) as compared to a healthy control subject.
- MAT marrow adipose tissue
- the subject in need thereof has, at the time of treatment of or immediately prior to the fracture, an Ale (hemoglobin Ale) level below 5.7%. In some embodiments, the subject in need thereof has, at the time of treatment of or immediately prior to the fracture, an Ale level of 5.7-6.4%. In some embodiments, the subject in need thereof has, at the time of treatment of or immediately prior to the fracture, an Ale level greater than 6.5%.
- the subject treated with any of the DPP-4 inhibitors disclosed herein is a subject to be exposed to, currently exposed to, or previously exposed to altered gravity environment. In some embodiments, the subject is exposed to an altered gravity environment for at least 3 days, one week, two weeks, one month, two months, three months, four months, five months, six months, nine months, and/or one year. In some embodiments, the subject is administered the DPP-4 inhibitor prior to and/or during the exposure. In some embodiments, the subject is an astronaut.
- the agents of the present disclosure may be used to treat a bone- related disease.
- the bone-related disease is selected from the group consisting of osteoporosis, osteogenesis imperfect, primary bone cancer, cancer that has metastasized to bone, rickets, osteomalacia, renal osteodystrophy, and/or Paget's Disease.
- the osteoporosis is primary osteoporosis.
- the primary osteoporosis is idiopathic primary osteoporosis or age-related osteoporosis.
- the osteoporosis is secondary osteoporosis.
- the osteoporosis is caused by idiopathic hyper-calcinuria, cystic fibrosis, glucocorticoid treatment, cyclosporine A treatment, and/or tacromilus treatment.
- the bone related disease occurs in post-meopausal women, patients who have undergone hysterectomy, patients who are undergoing or have undergone long-term administration of corticosteroids, patients suffereing from Cushing's syndrome, patients who display one or more symptoms of the frailty syndrome, patients with cachexia, patients with conditions associated with wasting of body tissues, or patients who have gonadal dysgenesis.
- the disclosure provides any of the foregoing methods or uses comprising administering any of the agents of the disclosure.
- the subject is suffering from more than one of the diseases disclosed herein.
- the subject is suffering from a bone -related condition, such as a fracture of a bone.
- the subject is suffering from a fracture in more than one bone.
- the bone is a flat, long, short, irregular or sesamoid bone.
- the bone is a long bone (e.g., a femur, humerus, tibia, metacarpal, metatarsal and/or phalange).
- the bone is a short bone (e.g., a carpal or tarsal).
- the bone is a flat bone (e.g., a scapula, sternum, cranium, os coxae, pelvis and/or rib).
- the bone is an irregular bone (e.g., vertebrae, sacrum and/or mandible).
- the bone is a sesamoid bone (e.g., knee cap and/or pisiform).
- the fracture is a compound fracture.
- the disclosure provides for a use in treating a bone fracture.
- the fracture is a non-union fracture, a compound fracture, a fracture with delayed healing, a stable fracture, a displaced fracture, a non-displaced fracture, an open fracture, a closed fracture, a Greenstick fracture, a complete fracture, an incomplete fracture, a transverse fracture, an oblique fracture, a comminuted fracture, a buckled fracture, a pathologic fracture, and/or a stress fracture.
- the bone fracture is a non-union bone fracture, a compound fracture or a fracture with delayed healing.
- non-union bone fracture is meant to relate to a permanent failure of healing following a broken bone unless intervention (such as surgery) is performed.
- a "fracture with delayed healing” is meant to relate to a failure to reach bony union by 6 months post-injury. This also includes fractures that are taking longer than expected to heal (ie. distal radial fractures).
- the fracture is the result of a disease or condition such as osteoporosis.
- the fracture is the result of a trauma to the fractured bone.
- the fracture is the result of overuse of the bone (e.g. , as the result of repetitive motion, such as in an athlete).
- the disclosure provides for a use in inducing differentiation of a multipotent stem cell or a progenitor cell into a cell of the bone lineage (e.g., into an osteoblast or an osteocyte) in a subject in need thereof. In some embodiments, the disclosure provides for a use in inducing differentiation of a multipotent stem cell or a progenitor cell into a cell of the cartilage lineage (e.g., into a chondrocyte) in a subject in need thereof.
- the multipotent stem cell or its progeny, the adipogenic progenitor cell is a cell that expresses high levels of genes associated with adipocytic lineage (e.g., CD34, EBF2, VIM, PPARA and/or DPP4).
- the multipotent stem cell or its progeny, the osteochondrogenic progenitor cell is a cell that expresses high levels of genes associated with osteogenic lineage (e.g., AlpI, Dmpl, Collal/2) or is a cell that expresses high levels of genes associated with chondrogenic lineage (e.g., Acan, Col2al, Sox9).
- the multipotent stem cell expresses elevated levels of GREM1 or other markers such as Cxcll2, Kitl/Scf, Vcam-1, Lepr.
- the multipotent stem cell is a CD45 CD31 " Scal + CD24 + multipotent stem cell.
- the adipogenic progenitor cell (APC) is a CD45 ⁇ CD31 ⁇ Scal + CD24 ⁇ cell.
- the osteogenic progenitor cell (OPC) is a CD45 ⁇ CD31 ⁇ Scal ⁇ Pa + cell.
- treatment of the multipotent stem cell with any of the DPP-4 inhibitors disclosed herein enhances osteogenic gene expression.
- treatment of the multipotent stem cell with any of the DPP-4 inhibitors disclosed herein enhances chondrogenic gene expression.
- the subject is a mammal. In some embodiments, the subject is a human.
- the disclosure provides for a method of preparing cells for transplantation into a subject having a bone-related disease ⁇ e.g., osteoporosis) or bone-related condition ⁇ e.g., a bone fracture).
- the method comprises providing a cell culture comprising osteogenic progenitor cells and/or mesenchymal stem cells, contacting the cells with any of the DPP-4 inhibitors disclosed herein in an amount effective to increase osteogenic or chondrogenic gene expression thereby generating a culture comprising DPP-4 treated cells.
- the thus generated DPP-4 treated cells from the culture are transplanted to a fracture in a subject in need thereof.
- the cells for transplantation are multipotent stem cells.
- the multipotent stem cell is a cell that expresses high levels of genes associated with adipocytic lineage ⁇ e.g., CD34, EBF2, VIM, PPARA and/or DPP 4). In some embodiments, the multipotent stem cell is a CD45-CD31 - Scal+CD24+ multipotent stem cell.
- the multipotent stem cell or its progeny, the osteochondrogenic progenitor cell is a cell that expresses high levels of genes associated with osteogenic lineage ⁇ e.g., AlpI, Dmpl, Collal/2) or is a cell that expresses high levels of genes associated with chondrogenic lineage ⁇ e.g., Acan, Col2al, Sox9).
- the OPC is a cell whose surface marker configuration is CD45 " CD31 " Scal " Pa + .
- treatment of the multipotent stem cell with any of the DPP-4 inhibitors disclosed herein enhances osteogenic gene expression and/or osteogenic lineage commitment of the initially multipotent cell.
- any of the agents described herein may be administered in combination with any of the additional therapeutic treatments described herein.
- the additional therapeutic treatment is reduction ⁇ e.g. , closed reduction) and or the use of medical devices (e.g., casts, pins, plates, screws, rods or glue) to hold the fracture in place.
- the additional therapeutic treatments may include treatment with one or more compounds selected from the group consisting of: an anti-infective agent, a pain and/or inflammation reliever (e.g., acetominophen, ibuprofen), a growth factor (e.g., bone morphogenic proteins (BMPs), TGF-beta, insulin-like growth factor (IGF), fibroblast growth factor (FGF), FGF-2, platelet-derived growth factor (PDGF) and vascular endothelial growth factor (VEGF)), a hormone (e.g., parathyroid hormone (PTH) or growth hormone (GH)) and a soluble receptor (e.g., an ActRIIA receptor).
- a pain and/or inflammation reliever e.g., acetominophen, ibuprofen
- a growth factor e.g., bone morphogenic proteins (BMPs), TGF-beta, insulin-like growth factor (IGF), fibroblast growth factor (FGF), FGF-2
- the BMP is selected from the group consisting of: OP-1 , OP-2, OP-3, BMP-2, BMP-3, BMP-4, BMP-5, BMP-6, BMP-8, BMP-9, BMP- 10, BMP-1 1 , BMP- 15, BMP- 16, DPP, Vgl , Vgr-1 , 60A protein, GDF-1 , GDF- 2, GDF-3, GDF-5, GDF-6, GDF-7, GDF-8, GDF-9, GDF-10, GDF-1 1 , GDF-12, NODAL, UNIVIN, SCREW, ADMP, NEURAL, and amino acid sequence variants thereof.
- the additional therapeutic treatment is a compound for treating osteoporosis, such as a bisphosphonate (e.g., Alendronate (Fosamax), Risedronate (Actonel), Ibandronate (Boniva) and Zoledronic acid (Reclast)), a hormone (e.g., raloxifene (Evista)), a RANKL inhibitor (e.g., Denosumab (Prolia)) and/or a synthetic hormone (e.g., a synthetic parathyroid hormone such as Teriparatide (Forteo)).
- a bisphosphonate e.g., Alendronate (Fosamax), Risedronate (Actonel), Ibandronate (Boniva) and Zoledronic acid (Reclast)
- a hormone e.g., raloxifene (Evista)
- a RANKL inhibitor e.g., Denosumab (Prolia)
- Administration "in combination with” one or more further therapeutic agents includes simultaneous (concurrent) and consecutive administration in any order.
- any of the agents described herein are administered to a subject in combination with an additional therapeutic treatment, wherein the subject has a bone-related disease (e.g., osteoporosis) or bone-related condition (e.g., a bone fracture).
- the additional therapeutic treatment is a physical therapy, massage therapy, electrical and electromagnetic stimulation, ultrasound, extracorporeal shock waves and/or rest.
- the combined administration includes coadministration, using separate formulations or a single pharmaceutical formulation, and consecutive administration in either order, wherein preferably there is a time period while both (or all) active agents simultaneously exert their biological activities.
- the additional therapeutic treatment and the agent are administered consecutively.
- the additional therapeutic treatment is administered concurrently with the agent.
- the additional therapeutic treatment is administered prior to the administration of the agent.
- combined therapy results in a cumulative therapeutic effect.
- combined therapy results in a synergistic therapeutic effect.
- the therapeutically effective amount of each of: a) the DPP-4 inhibitor, and/or b) the additional therapeutic treatment is less than that required to achieve a therapeutic effect when one or both agents is administered as a monotherapy.
- compositions of the disclosure have numerous uses.
- the DPP-4 inhibitor agents disclosed herein are useful for studying effects on different cells and tissues in vitro and/or in vivo.
- the agents are useful as imaging agents, such as for ex vivo or in vivo diagnostic applications.
- agents conjugated to a radioactive moiety are useful for ex vivo or in vivo imaging studies.
- the disclosure provides for a method of inducing differentiation of a multipotent stem cell into a cell of the bone lineage (e.g., into an osteoblast).
- the multipotent stem cell is a cell that expresses high levels of genes associated with adipocytic lineage (e.g. , CD34, EBF2, VIM, PPARA and/or DPP4).
- the multipotent stem cell is a CD45-CD31-Scal+CD24+ multipotent stem cell.
- treatment of the multipotent stem cell with any of the DPP-4 inhibitors disclosed herein enhances osteogenic gene expression and/or osteogenic cell differentiation and/or osteogenic lineage commitment of the multipotent cell.
- treatment of the multipotent stem cell with any of the DPP-4 inhibitors disclosed herein enhances chondrogenic gene expression and chondrogenic cell differentiation.
- the multipotent cell is in vitro.
- the multipotent cell is in a subject.
- the subject is a mammal.
- the subject is a human.
- the agents of the disclosure are suitable for identifying binding partners and/or tissue distribution for the agents delivered and for evaluating localization and trafficking. Similarly, effects of the agent on gene expression or DPP-4 half- life may be studied in vivo or in vitro.
- Suitable animal models of bone-related diseases or conditions are known in the art. Suitable animals for studying the effects of any the DPP-4 inhibitors disclosed herein include mice, rats, dogs, and primates. Representative animal models are described in Histing et al, 2011, Bone, 49(4):591-9. An example of a model is also discussed in the Example section below. In this model, anesthetized mice are injected with 1.5 x 10 4 cells in a 50 % matrigel suspension through the proximal articular surface of the tibia. A steel pin (diameter 0.35 mm) is inserted into the medullary cavity for stabilization and a fracture was induced with scissors 0.5 cm distal from the knee. The effects of any of the DPP-4 inhibitors disclosed herein on the healing of the bone-related diseases or conditions may be assessed in these models.
- CD45 CD31 non-hematopoietic, non-endothelial cells
- Sca Stem cell antigen
- Pa Platelet-derived growth factor-a
- a population of unilaterally committed adipogenic progenitors was isolated that was CD45 CD3 l " Scal + CD24 " and a population that displayed tri- lineage differentiation potentials and was CD45 " CD31 " Scal + CD24 + ( Figures 1C and ID).
- Colony forming potentials (CFU-F) and in vitro recovery rates were highest in fibroblastic cells expressing Seal and/or Pa and were highest in the CD45 CD3 l " Scal + Pa + CD24 + subset ( Figures 3 and 4).
- CFU-F potential was enriched in the osteogenic CD45 CD3 rScal " Pa + population but absent in CD45 " CD31 " Scal " Pa " cells.
- the transcription factor zinc finger protein (Zfp)-423 labels adipogenic cells in white adipose tissue (WAT).
- WAT white adipose tissue
- Zfp423-eGFV + (Zfp423 + ) cells occurred as a small subpopulation of less than 1% within the CD45 " CD31 " Sca population while all adipogenic CD45 CD3 l " Scal + cells were GFP " ( Figure 6). This small subset of adipogenic cells was likely not initially detected due to a dilution effect within the strongly osteochondrogenic cell fraction.
- CD45 CD3 Scal ⁇ Zfp423 + cells maintained GFP-expression before and after differentiation into adipocytes.
- all CD45 CD3 l " Scal + cells uniformly acquired GFP-expression only during differentiation, a process that correlated with a concomitant loss of Seal -expression ( Figures 7A, 7B, and 8).
- multipotent cells and osteogenic progenitors were more abundant in the metaphysis compared to the diaphysis whereas adipogenic progenitors were evenly distributed. Further analyses of Pa, Seal and CD24 expression localized osteogenic progenitors to the endosteum. The majority of multipotent and adipogenic progenitors cells resided in a non-endosteal localization within 40 ⁇ of the bone surface and revealed a perivascular association of all non-endosteal Pa + cells to blood vessels of less than 10 ⁇ in diameter ( Figures 9 and 10).
- a tri-potent, perivascular population with stem cell-like characteristics (CD45 ⁇ CD31 ⁇ Scal + CD24 + ), two functionally and anatomically distinct progenitor populations that are fate-committed towards either the osteochondrogenic (CD45 " CD31 Scal Pa + ; also referred to as OPC) or adipogenic (CD45 CD31 Scal + CD24-; also referred to as APC) lineages, and a more mature CD45 CD3 rScal " Zfp423 + adipocyte precursor stage (also referred to as preAd) cell population.
- a triple transgenic mouse strain was generated carrying alleles of the Zfp423-eGFF reporter, Adiponectin (Adipoq)-Cre, and a luciferase reporter within the Rosa26-locus that is only expressed after Cre -mediated recombination, e.g. in mature adipocytes (rep AdlLuc , Figure 12 A) and a second strain with constitutive red fluorescence (mTmG reporter allele) crossed to the Zfp423-eGFF reporter (rep tdTom , Figure 12B).
- RNA-Sequencing was used to further characterize the molecular identity of all four populations ( Figure 32). Principal component and hierarchical clustering analyses clearly supported the distinct nature of each population, providing a second line of evidence for the lineage restriction of adipogenic commitment of the closely related APC and pre Ad populations ( Figure 33 A, Figure 33B, and Figure 33C). Differential expression (DE) analysis produced several sets of known and potential new candidate genes to define each population ( Figures 34-37). For instance, canonical stem cell markers (e.g. Nog, Illrn, Myc) were enriched in the CD45-CD31- Scal+CD24+ multipotent stem cell population ( Figure 34).
- canonical stem cell markers e.g. Nog, Illrn, Myc
- signals known to regulate HSC quiescence and maintenance showed highest expression in this population, along with the highest, but not exclusive, expression level of LepR that was also expressed in the other cells types.
- the OPC population expressed the classical osteogenic (e.g. AlpI, Dmpl, Collal/2) and chondrogenic markers (e.g. Acan, Col2al, Sox9), as well as previously described skeletal stem cell markers (Itga5, CD200) at elevated levels ( Figure 35).
- the adipogenic populations expressed high levels of markers that have been linked to the adipocytic lineage (i.e.
- RNA-Seq analysis confirmed the cellular characteristics of the four populations and establishes the CD45-CD31-Scal+CD24+ multipotent stem cell population as a population expressing elevated levels of Cxcll2 and Lepr that are important regulators of HSCs and osteogenesis.
- Dpp4 Dipeptidyl peptidase-4
- Figure 38 Consistent with the RNA-Seq data, CD26 (the membrane bound form of DPP4), was enriched on the surface of adipogenic cell populations, and only CD45-CD31-Scal+CD24+ and APCs, but not OPCs, released DPP4 into the medium after adipogenic differentiation ( Figures 39 and 40).
- Sitagliptin Sita or control (Ctrl) were administered to the animals daily at a dose of 10 mg/kg body weight, either by intraperitoneal injections (i.p.) or by oral gavage (per oral - p.o.). For each condition, 3 animals were used.
- Flow cytrometric analysis of bone-resident mesenchymal stromal cells (MSCs), APCs, and OPCs was conducted as described below. Per oral administration and intraperitoneal injection of Sitagliptin showed equal effects on the distribution of these cell poulations (Figure 48).
- mice All procedures were approved by the ethics committee for animal welfare of the State Office of Environment, Health, and Consumer Protection (State of Brandenburg, Germany). Animals were housed in a controlled environment (20 ⁇ 2 °C, 12 hour/12 hour light/dark cycle), maintained on a standard diet (SD) (Ssniff, Soest, Germany), or fed a high fat diet (HFD) (45% energy from fat, D12451, Research Diets, New Brunswick, NJ,USA) for 1 or 10 days. Male mice were used for all experiments at the indicated ages, where applicable.
- SD standard diet
- HFD high fat diet
- mice strains were obtained from The Jackson Laboratory: C57BL/6J, B6(Cg)-Tyr c 2J /J (B6-albino), B6.Cg-Tg(Gt(ROSA)26Sor- EGFP)IlAble/J, B6.129S4-Pdgfra tml l(EGFp)So 7J ( ⁇ -eGFP reporter), B6;FVB-Tg(Zfp423- EGFP)7Brsp/J (Zfp423-eGFP reporter), B6.129(Cg)-Gt ⁇ O&4 2 ⁇ or im ⁇ cra - irfromflto - £G ⁇ "°/J (mTmG-reporter), FVBA29S6(B6)-Gt(ROSA)26So ⁇ Jml(Luc)Kael /J (Rosa26-Luciferase reporter), B6;FVB-Tg(Adipoq-
- Zfp423-eGFP reporter mice were either intercrossed with mTmG- reporter mice (rep tdTom ), or to AdipoQ-Cre mice and a lox-Stop-lox reporter strain expressing luciferase after Cre-mediated removal of the fioxed Stop-cassette from the Rosa26-locus (rep AdlLuc ).
- Freshly sorted primary murine cells were used throughout this study and isolated by FACS and cultured as previously described (Schulz et al, 2011, Proc. Natl. Acad. Sci. U. S. A. 108, 143-148 and Steenhuis et al, 2008, Calcif. Tissue Int. 82, 44-56).
- a complex medium of 60% DMEM low glucose (Invitrogen) and 40% MCDB201 (Sigma) was supplemented with 100 U/ml penicillin and 1,000 U/mL streptomycin (Invitrogen).
- 2% FBS, l x insulin-transferrin-selenium (ITS) mix, l x linoleic acid conjugated to BSA, 1 nM dexamethasone, and 0.1 mM L-ascorbic acid 2-phosphate (all from Sigma) were added.
- growth factors were added to the medium: 10 ng/ml epidermal growth factor (PeproTech), 10 ng/ml leukemia inhibitory factor (MerckMillipore), 10 ng/ml platelet-derived growth factor BB (PeproTech), and 5 ng/ml basic fibroblast growth factor (bFGF; Sigma- Aldrich).
- the bFGF was added daily throughout the culture period except where stated otherwise.
- For adipogenic differentiation cells were induced for 48 hours after three days of expansion, followed by a differentiation period of 5 days.
- induction medium growth medium without growth factors
- human insulin Gibcose IGF
- indomethacin 1 ⁇ dexamethasone
- 0.5 ⁇ isobutylmethylxanthine 1 nM 3,3',5- triiodo-L-thyronine (T3) (all from Sigma-Aldrich)
- T3 3,3',5- triiodo-L-thyronine
- Oil Red O staining was performed by fixing cells with 4% Histofix for 15 minutes at room temperature.
- Oil Red O working solution For the preparation of Oil Red O working solution, a 0.5% stock solution in isopropanol was diluted with distilled water at a ratio of 3:2. The working solution was filtered and applied to fixed cells for at least one hour at room temperature. Cells were washed four times with tap water before evaluation. For quantification, Oil Red O was extracted by adding a defined volume of isopropanol and absorbance was read in a micro-plate reader (Synergy HI , BioTek) at 510 nm.
- osteogenic medium DMEM low glucose (Invitrogen)
- FBS fetal bovine serum
- Dexamethasone 0.2 mM L-ascorbic acid 2-phosphate, lOmM ⁇ -glycerophosphate, and 50 ng/ml L-thyroxine
- Cells were then formalin- fixed and stained with 2% Alizarin Red S (Roth) in distilled water.
- Wells were washed twice with PBS and once with distilled water. De-staining was conducted to quantitatively determine mineralization by adding a 10% cetylpyridinium chloride solution. Absorbance was measured in a micro-plate reader (Synergy HI, BioTek) at 570 nm.
- a micromass culture was used for the chondrogenesis assay.
- a 5 ⁇ droplet of cell suspension (appr. 1.5 x 10 7 cells/ml) was pipetted in the center of a well (48-well plate).
- warm chondrogenic media (DMEMhigh (Invitrogen)) with 10% FBS, 100 nM Dexamethasone, 1 ⁇ L-ascorbic acid-2- phosphate, lOx ITS mix, and 10 ng/ml Transforming growth factor ⁇ ) was added.
- Cell media was changed every other day. After 21 days, cells were fixed and stained with 1% Alcian-Blue staining (Sigma) for 30 minutes at room temperature. Cells were rinsed three times with 0.1 M HC1. To neutralize acidity, a washing step with dH20 was conducted before microscopic analysis.
- DPP4 in vitro experiments cell populations were differentiated with adipogenic or osteogenic assays as described above.
- Mouse recombinant DPP4 250 ng/ml; R&D Systems
- the DPP-4 inhibitor Sitagliptin 100 ⁇ ; biomol
- DPP4 secretion into cell culture media was determined by ELISA (ThermoFisher). Either supernatant of freshly isolated tibia explants maintained in culture media for 24 hours or supernatant from cell populations following 10 days of adipogenic differentiation were used.
- CFU-F assay was conducted as follows: Freshly isolated cell populations were seeded in expansion media at 500 cells per 6-well plate. Medium was changed every other day. At day 10, cells were fixed and stained with Crystal Violet (Sigma). Colonies consisting of more than 20 cells were counted as CFU. At least 6 independent assays were performed per cell population. For total recovery rate experiments, cell populations were seeded as described for the CFU-F assay. Analysis of fixed and crystal violet stained cell populations was conducted on day 7, 11, and 15 by quantification of total cell invasion area of well-plate surface using ImageJ software.
- Flow cytometry & cell sorting Flow cytometry and cell sorting were performed on a FACS Aria III cell sorter (BD Biosciences) and analyzed using Flow Jo software (Tree Star). Soft-tissue free bones (tibia/femur) were crushed with bone scissors and incubated for 1 hour in a shaking water bath at 37 °C in 10 ml of 20% FBS/PBS containing 0.5% type-2 collagenase (CellSystems). The suspension was filtered through a 70 ⁇ mesh to remove bone fragments and centrifuged at 1200 rpm for 5 minutes at 4 °C.
- the pellet was re-suspended in ACK (Ammonium-Chloride -Potassium) lysing buffer to eliminate red blood cells and centrifuged again at 1200 rpm for 5 minutes at 4 °C.
- the pellet was re-suspended in 100 ⁇ sorting buffer (2% FBS/PBS) and stained with antibodies for at least 30 minutes at 4 °C.
- the applied FACS antibodies can be found in the Key Resources Table. Living cells were gated for lack of PI (propidium iodide; 1 : 1,000 diluted stock solution: 1 ⁇ g/mL in water) fluorescence and accumulation of Calcein (1 : 1,000 dilution; stock of 1 mg in 215 DMSO).
- Single-cell clonal assays For the co-culture approach, a feeder layer of CD45 CD31 " Pa + cells was isolated from long bones of 8-week old male C57BL/6J mice and seeded in 100 ⁇ of expansion medium at 750 cells per well of a 96-well plate. On the next day, a single CD45 " CD3 Scal + CD24 + tdTomato + cell freshly isolated from 8-week old male Rosa26-mTmG mice was FACS-sorted into each well. Cells were expanded for 10 days to sub-confiuency with media changes every other day. After 10 days, clonal expansion of a single cell was verified by fluorescence microscopy.
- Wells containing a readily detectable single colony of tdTomato + cells were trypsinized, washed, and collected in 100 ⁇ sorting medium.
- Five to ten cells (per condition) of each clone were directly FACS-sorted onto freshly prepared 96-well plate feeder layers of expanded CD45 " CD31 " Pa + cells for adipogenic and osteogenic differentiation protocols, or onto a micromass culture for chondrogenic differentiation.
- clones were analyzed for their differentiation capacity by immunocytochemistry.
- a tdTomato positive clone was considered adipogenic if it co-stained with Perilipin, osteogenic if it co-stained with Osteocalcin, and chondrogenic if it co-stained with Aggrecan.
- a single CD45 CD31 " Scal + CD24 + tdTomato + cell freshly isolated from 8-weeks old male C57BL/6J mice, was FACS-sorted into a well of a 96-well plate without feeder cells. Single cells were expanded for 10 days with media changes every other day. After 10 days, clones giving rise to colonies were re-seeded in a new 96-well plate and expanded until sub-confluency.
- bone marrow regions of 0.05 mm 2 from bone sections were selected on fluorescence images.
- fixated cells in well plates were permeabilized with 0.1% Triton X-100 solution and blocked with 3%) BSA in PBS.
- Antibodies were used as listed in the resources table.
- nuclear staining specimens were treated with DAPI. Sections and cells were analyzed using a Keyence BZ-9000 (Biorevo) fluorescence microscope (for up to two fluorescences) or a Zeiss confocal laser scanning microscope (LSM) 700 (for three fluorescences).
- Sternal transplantation Sorted cell populations from luciferase-expressing rep or tdTomato-expressing r ep tdTom mice were subcutaneously injected at 1.5 x 10 4 cells in a 50% matrigel suspension into the sternal area of B6-albino mice. Eight weeks after transplantation, engrafted tissues were excised, fixed, and histologically analyzed. Mice injected with cells from rep Luc animals were additionally subjected to Luciferase imaging with an IVIS imaging system (Perkin Elmer) before sacrifice. To this end, animals were intraperitoneally injected with luciferin (150 mg/kg) and subsequently anesthetized. After 12 to 18 minutes, the animals were imaged. Image analysis was performed with Living Image 4.4 software (Xenogen).
- mice were i.p. -injected with a single dose of 100 mg BrdU/kg (Sigma Aldrich) diluted in sterile PBS. Mice receiving a SD or HFD for ten days were given BrdU via drinking water at a concentration of 0.5 mg/ml. Drinking water was refreshed every other day.
- BrdU cell proliferation in vivo assay For 24 hour experiments, mice were i.p. -injected with a single dose of 100 mg BrdU/kg (Sigma Aldrich) diluted in sterile PBS. Mice receiving a SD or HFD for ten days were given BrdU via drinking water at a concentration of 0.5 mg/ml. Drinking water was refreshed every other day.
- For single-cell immunostaining was approximately 2 x 10 3 cells/mouse of each population of interest were double-sorted on glass cover slips pre-coated with a 5 ⁇ drop of DMEM(low). Coverslips were incubated for 30 minutes, allowing cells to attach.
- Fracture model Mice were given an analgetic (MediGel, ClearFbO) starting two days prior to surgery. Anesthetized mice were injected with 1.5 x 10 4 cells in a 50% matrigel suspension through the proximal articular surface of the tibia. A steel pin (diameter 0.35 mm) was inserted into the medullary cavity for stabilization and a fracture was induced with scissors 0.5 cm distal from the knee. At the indicated time point after fracture induction, tibiae were harvested for analyses. After removal of the pin from extracted tibiae, ⁇ CT analysis was conducted with LaTheta LCT-200 (Hitachi-Aloka) using manufacturer's pre-defined parameters for isolated bone measurements.
- LaTheta LCT-200 Hitachi-Aloka
- tibiae were fixed and decalcified followed by paraffin embedding and sectioning at 3 ⁇ per slice. Samples were stained using SafraninO/Fast green and Movat Pentachrome. ImageJ software was used for computer-assisted histomorphometric analysis of fracture calluses. Six representative sections of each callus were analyzed for bone, fibrous, and cartilaginous tissue areas in a blinded manner.
- mice received a daily dose of PBS, Diprotin A (5 mg/kg body weight; Sigma) or Sitagliptin (10 mg/kg body weight; biomol) i.p. for 9 consecutive days.
- PBS diprotin A
- Sitagliptin 10 mg/kg body weight; biomol
- RNA extraction, reverse transcription and cDNA pre-amplification, Nextera XT libraries and R A-sequencing of the cell populations was done as previously described.
- Pa + cells were FACS-sorted from bones of 4 mice (the 3 biological replicates were done on 3 different days), collected in a 1.5 ml Eppendorf tube containing 50 ⁇ RLT Plus Buffer (Qiagen) supplemented with 1% 2-Mercaptoethanol, immediately frozen in dry-ice and kept at -80 °C.
- RNA-seq data processing and analysis Sequencing data were aligned to the Mus musculus genome (Ensembl version 38.82) using GSNAP (version 2014-10-07) with default parameters. HTseq-count was used to count the number of reads mapped to each gene (default options). Almost all libraries showed good quality, with sizes ranging between 2-3.5xl0 7 read counts and a fraction of reads mapped to exons greater than 75%. One library yielded less than 300 reads and was excluded from downstream analysis. The data was normalized for sequencing depth using size factors. The union of the top 1,000 genes expressed in each library was selected, which resulted in a list of 2,120 genes.
- RNA-seq data was statistically analyzed using the R-statistical package and Paleontological Statistics (PAST, version 3.10, http://folk.uio.no/ohammer/past/, accessed December 2015).
- PAST paleontological Statistics
- gene expression was compared between all investigated cell populations.
- a p-value of ⁇ 0.05 was used as a cut-off for differentially expressed genes.
- Heat-maps contain representative top-regulated genes, which were further divided by known cell type specific functions as previously described in the literature and unknown novel marker genes.
- RNA isolation and gene expression analysis were conducted using standard methods as described before (Schulz et al, 2011, Proc. Natl. Acad. Sci. U. S. A. 108, 143-148) using column-based RNA-isolation, reserve transcription for cDNA synthesis, and SYBR green-based detection during quantified real-time PCR. Primer sequences were used as noted in the Key Resources Table.
- RNA-seq data generated in this study was deposited at the European Nucleotide Archive (http://www.ebi.ac.uk/ena) under secondary sample accession number (ID code) ERP013883.
- the invention relates to a method of treating a bone fracture in a subject in need thereof, comprising administering an effective amount of a DPP-4 inhibitor to the subject.
- the invention relates to a menthod of preventing non-union of a bone fracture or of preventing healing complications following bone fracture in a subject in need thereof, comprising administering an effective amount of a DPP-4 inhibitor to the subject.
- the invention relates to a method of promoting fracture healing in a subject in need thereof, comprising administering an effective amount of a DPP-4 inhibitor to the subject.
- the bone fracture is a non-union bone fracture, a compound fracture or a delayed fracture healing.
- the DPP-4 inhibitor is administered systemically. In a further embodiment, the DPP-4 inhibitor is administered locally. In a further embodiment, the DPP-4 inhibitor is administered to the site of fracture. In a further embodiment the DPP-4 inhibitor is administered in one or more doses over a period of less than 6 months. In a further embodiment, the DPP-4 inhibitor is administered in one or more doses over a period of less than 3 months. In a further embodiment, the DPP-4 inhibitor is administered in one or more doses over a period of less than 1 month.
- the subject prior to diagnosis of the bone fracture, was not receiving a DPP-4 inhibitor.
- the subject in need thereof was not, prior to the bone fracture, diagnosed with or treated for Type II diabetes.
- the subject in need thereof is over age 65.
- the subject in need thereof is under age 40.
- the subject in need thereof has a BMI less than 25.
- the subject in need thereof has a BMI of greater than or equal to 25 and less than 30.
- the subject in need thereof has a BMI of equal to or greater than 30.
- the DPP-4 inhibitor is administered in one or more doses, and wherein at least one of the doses is administered locally during surgery to set the fracture.
- the subject in need thereof is a human subject.
- the healing complications are osteoporosis related healing complications to bone fracture.
- the DPP-4 inhibitor is selected from one or more of alogliptin, linagliptin, saxagliptin, sitagliptin, vildapliptin, gemigliptin, or teneligliptin.
- the method further comprises administering metformin, in the same or a different formulation as the DPP-4 inhibitor.
- the method further comprises administering one or more other therapeutic agent.
- the invention relates to a method for reducing the inhibitory effects of marrow adipose tissue (MAT) on fracture healing in a subject in need thereof, comprising administering an effective amount of a DPP-4 inhibitor to the subject.
- MAT marrow adipose tissue
- the invention in a further aspect relates to a method of preparing cells for transplantation, comprising providing a cell culture comprising osteogenic progenitor cells (OPCs) and/or mesenchymal stem cells, contacting the cells with a DPP-4 inhibitor in an amount effective to increase osteogenic gene expression, or osteo-/ chondrogenic cell differentiation, or osteogenic lineage commitment, thereby generating a culture comprising DPP-4 treated cells, and transplanting cells from the culture to a fracture in a subject in need thereof.
- OPCs osteogenic progenitor cells
- DPP-4 inhibitor in an amount effective to increase osteogenic gene expression, or osteo-/ chondrogenic cell differentiation, or osteogenic lineage commitment
- the invention in a further aspect relates to a method of preparing cells for transplantation, comprising providing a cell culture comprising CD45 " CD31 " Scal + CD24 + multipotent cells, treating the cell culture to promote lineage commitment and/or differentiation into osteogenic progenitor cells (OPCs) and/or mature cells of the osteogenic lineage/bone tissues, sorting the cells to isolate or enrich for OPCs, and transplanting the OPCs to a fracture in a subject in need thereof.
- OPCs osteogenic progenitor cells
- the invention in a further aspect relates to a method for preventing lose of bone mineral density (BMD) in an astronaut or other individuals exposed to an altered gravity environment, said method comprising administering to the astronaut or individual an effective amount of a DPP-4 inhibitor.
- BMD bone mineral density
- the astronaut is exposed to an altered gravity environment for greater than one week, and the DPP-4 inhibitor is administered prior to and/or during and/or after the exposure.
- the invention relates to a method of preventing loss of bone mineral density (BMD) in a subject in need thereof, comprising administering to the subject an effective amount of a DPP-4 inhibitor.
- BMD bone mineral density
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Veterinary Medicine (AREA)
- Pharmacology & Pharmacy (AREA)
- Medicinal Chemistry (AREA)
- Epidemiology (AREA)
- Engineering & Computer Science (AREA)
- Physical Education & Sports Medicine (AREA)
- Organic Chemistry (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Rheumatology (AREA)
- Orthopedic Medicine & Surgery (AREA)
- Biomedical Technology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Biotechnology (AREA)
- Genetics & Genomics (AREA)
- Wood Science & Technology (AREA)
- Zoology (AREA)
- Cell Biology (AREA)
- Microbiology (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
La présente invention concerne des inhibiteurs de DPP-4 à utiliser pour traiter une fracture osseuse, pour prévenir l'absence de consolidation d'une fracture osseuse ou pour prévenir des complications de cicatrisation à la suite d'une fracture osseuse, pour réduire des effets inhibiteurs de tissus adipeux de moelle (MAT) sur la cicatrisation d'une fracture, ou pour prévenir la perte de densité minérale osseuse (BMD) chez un patient nécessitant un tel traitement. L'invention concerne également des méthodes de préparation de cellules en vue d'une greffe qui consistent à fournir une culture cellulaire comprenant des cellules progénitrices ostéogéniques (OPC) et/ou des cellules souches mésenchymateuses, et à mettre en contact les cellules avec un inhibiteur de DPP-4. L'invention concerne en outre des méthodes de préparation de cellules en vue d'une greffe qui consistent à fournir une culture cellulaire comprenant des cellules multipotentes CD45-CD31- Sca1+ CD24+, et à traiter la culture cellulaire pour favoriser l'implication et/ou la différenciation de lignée en cellules progénitrices ostéogéniques (OPC) et/ou en cellules matures des tissus osseux/de lignée ostéogéniques.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201762469453P | 2017-03-09 | 2017-03-09 | |
US62/469,453 | 2017-03-09 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2018162722A1 true WO2018162722A1 (fr) | 2018-09-13 |
Family
ID=61627101
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/EP2018/055931 WO2018162722A1 (fr) | 2017-03-09 | 2018-03-09 | Inhibiteurs de dpp-4 à utiliser dans le traitement de fractures osseuses |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2018162722A1 (fr) |
Citations (59)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO1993010127A1 (fr) | 1991-11-22 | 1993-05-27 | Boehringer Ingelheim Pharmaceuticals, Inc. | Procede de production d'un ester de prolineboronate |
WO1995015309A1 (fr) | 1993-12-03 | 1995-06-08 | Ferring B.V. | Inhibiteurs de la dp-iv-serine protease |
DE19616486A1 (de) | 1996-04-25 | 1997-10-30 | Knoell Hans Forschung Ev | Verfahren zur Senkung des Blutglukosespiegels in Säugern |
WO1998018763A1 (fr) | 1996-10-25 | 1998-05-07 | Tanabe Seiyaku Co., Ltd. | Derives de tetrahydroisoquinoline |
WO1998019998A2 (fr) | 1996-11-07 | 1998-05-14 | Novartis Ag | 2-cyanopyrrolidines a substitution n |
WO1999025719A1 (fr) | 1997-11-18 | 1999-05-27 | Zaidan Hojin Biseibutsu Kagaku Kenkyu Kai | Substance physiologiquement active, la sulphostine, procede de fabrication et utilisation |
WO1999038501A2 (fr) | 1998-02-02 | 1999-08-05 | Trustees Of Tufts College | Procede de regulation du metabolisme du glucose et reactifs afferents |
WO1999046272A1 (fr) | 1998-03-09 | 1999-09-16 | Fondatech Benelux N.V. | Modulateurs de la serine peptidase |
WO1999061431A1 (fr) | 1998-05-28 | 1999-12-02 | Probiodrug Gesellschaft für Arzneimittelforschung mbH | Nouveaux effecteurs de dipeptidylpeptidase iv |
WO1999067279A1 (fr) | 1998-06-24 | 1999-12-29 | Probiodrug Gesellschaft für Arzneimittelforschung mbH | Composes d'inhibiteurs instables de la dipeptidylpeptidase iv |
WO1999067278A1 (fr) | 1998-06-24 | 1999-12-29 | Probiodrug Gesellschaft für Arzneimittelforschung mbH | Promedicaments d'inhibiteurs de la dipeptidylpeptidase iv |
WO2000034241A1 (fr) | 1998-12-10 | 2000-06-15 | Novartis Ag | 2-cyanopyrrolidines n-substitues |
US6107317A (en) | 1999-06-24 | 2000-08-22 | Novartis Ag | N-(substituted glycyl)-thiazolidines, pharmaceutical compositions containing them and their use in inhibiting dipeptidyl peptidase-IV |
US6124305A (en) | 1996-11-07 | 2000-09-26 | Novartis Ag | Use of N-(substituted glycyl)-2-cyanopyrrolidines in inhibiting dipeptidyl peptidase-IV |
WO2001034594A1 (fr) | 1999-11-12 | 2001-05-17 | Guilford Pharmaceuticals, Inc. | Inhibiteurs de la dipeptidyl peptidase iv; methodes de fabrication et d'utilisation desdits inhibiteurs |
WO2001052825A2 (fr) | 2000-01-21 | 2001-07-26 | Novartis Ag | Combinaisons comprenant un inhibiteur de la dipeptidylpeptidase - iv |
WO2001055105A1 (fr) | 2000-01-24 | 2001-08-02 | Novo Nordisk A/S | 2-cyanopyroles et -pyrrolines a substitution n inhibant l'enzyme dpp-iv |
WO2001068603A2 (fr) | 2000-03-10 | 2001-09-20 | Bristol-Myers Squibb Co. | Inhibiteurs de la dipeptidyl peptidase iv, a base de pyrrolidone fusionnee a du cyclopropyle, et procede |
WO2001072290A2 (fr) | 2000-03-31 | 2001-10-04 | Probiodrug Ag | Procede destine a ameliorer le signalement d'ilots dans le diabete sucre et a prevenir ce dernier |
WO2001081337A1 (fr) | 2000-04-26 | 2001-11-01 | Ferring B.V. | Inhibiteurs de dipeptidyl peptidase iv |
WO2001097808A1 (fr) | 2000-06-19 | 2001-12-27 | Smithkline Beecham Plc | Combinaisons d'inhibiteurs de peptidase iv de dipeptidyl et d'autres agents antidiabetiques pour traiter le diabete sucre |
WO2002002560A2 (fr) | 2000-07-04 | 2002-01-10 | Novo Nordisk A/S | Composes heterocycliques inhibiteurs de l'enzyme dpp-iv |
WO2002014271A1 (fr) | 2000-08-10 | 2002-02-21 | Mitsubishi Pharma Corporation | Dérivés de proline et leur utilisation comme médicaments |
WO2002038541A1 (fr) | 2000-11-10 | 2002-05-16 | Taisho Pharmaceutical Co., Ltd. | Derives de cyanopyrrolidine |
WO2002051836A1 (fr) | 2000-12-27 | 2002-07-04 | Kyowa Hakko Kogyo Co., Ltd. | Inhibiteur de dipeptidyl peptidase iv |
WO2002053548A1 (fr) | 2000-12-27 | 2002-07-11 | Banyu Pharmaceutical Co.,Ltd. | Derives de la benzothiazepine |
WO2002062764A1 (fr) | 2001-02-02 | 2002-08-15 | Takeda Chemical Industries, Ltd. | Composes heterocycliques condenses |
WO2002066627A1 (fr) | 2001-02-16 | 2002-08-29 | Bayer Aktiengesellschaft | Regulation de la dipeptidyle peptidase 8 humaine |
WO2002067918A1 (fr) | 2001-02-27 | 2002-09-06 | Banyu Pharmaceutical Co., Ltd. | Nouveau derive de diallylmethylamine |
WO2002068420A1 (fr) | 2001-02-24 | 2002-09-06 | Boehringer Ingelheim Pharma Gmbh & Co. Kg | Derives xanthine, fabrication et utilisations en tant qu'agents pharmaceutiques |
EP1245568A1 (fr) | 2001-03-28 | 2002-10-02 | Les Laboratoires Servier | Dérives sulfonyles d'-aminoacides et leur utilisation en tant qu'inhibiteurs de dipeptidyl-peptidase IV ( DPP IV) |
WO2002076450A1 (fr) | 2001-03-27 | 2002-10-03 | Merck & Co., Inc. | Inhibiteurs de peptidase dipeptidyl destines au traitement ou a la prevention du diabete |
WO2002083109A1 (fr) | 2001-04-11 | 2002-10-24 | Ferring Bv | Traitement du diabete de type 2 a l'aide d'inhibiteurs de dipeptidylpeptidase iv |
WO2002083128A1 (fr) | 2001-04-12 | 2002-10-24 | Bristol-Myers Squibb Company | Inhibiteurs a base de 2,1-oxazoline et 1,2-pyrazoline de la dipeptidyl peptidase iv et methode associee |
WO2002088090A2 (fr) | 2001-04-27 | 2002-11-07 | Vertex Pharmaceuticals Incorporated | Inhibiteurs de kinases derives du pyrazole |
US6482844B1 (en) | 2000-04-07 | 2002-11-19 | Neurogen Corporation | 1-benzylimidazole derivatives |
EP1258480A1 (fr) | 2001-05-18 | 2002-11-20 | Eisai Co., Ltd. | Dérivés de N-Carbamoylazole et leur utilisation pharmaceutique |
EP1258476A1 (fr) | 2001-05-15 | 2002-11-20 | Les Laboratoires Servier | Dérivés d'alpha-amino-acides, leur procédé de préparation ainsi que leur utilisation en tant qu'inhibiteurs de dipeptidyl-peptidase IV (DPP IV) |
WO2003000180A2 (fr) | 2001-06-20 | 2003-01-03 | Merck & Co., Inc. | Inhibiteurs de dipeptidyle peptidase pour le traitement du diabete |
WO2003000181A2 (fr) | 2001-06-20 | 2003-01-03 | Merck & Co., Inc. | Inhibiteurs de dipeptidyl peptidase utilises dans le traitement du diabete |
WO2003003250A1 (fr) | 2001-06-28 | 2003-01-09 | Hywire Ltd. | Memoire adressable par son contenu constituee de plages |
WO2003003727A1 (fr) | 2001-06-27 | 2003-01-09 | Nokia Corporation | Guide de programmes electronique presentant une fonctionnalite de visualisation d'echantillons et d'historique par fenetres a incrustation d'images |
WO2003002553A2 (fr) | 2001-06-27 | 2003-01-09 | Smithkline Beecham Corporation | Fluoropyrrolidines inhibitrices de la dipeptidyl peptidase |
WO2003002942A1 (fr) | 2001-05-18 | 2003-01-09 | Faeger Jan G | Procede pour determiner la position et/ou l'orientation d'un etre vivant par rapport a un environnement |
WO2003004498A1 (fr) | 2001-07-06 | 2003-01-16 | Merck & Co., Inc. | Pyrazines beta-amino tetrahydroimidazo (1, 2-a) et pyrazines tetrahydrotrioazolo (4, 3-a) utilisees en tant qu'inhibiteurs de la dipeptidyl peptidase dans le traitement ou la prevention du diabete |
WO2003004496A1 (fr) | 2001-07-03 | 2003-01-16 | Novo Nordisk A/S | Derives de purine inhibiteurs de dpp-iv pour le traitement du diabete |
WO2003024965A2 (fr) | 2001-09-19 | 2003-03-27 | Novo Nordisk A/S | Composes heterocycliques constituant des inhibiteurs de l'enzyme dpp-iv |
WO2003035057A1 (fr) | 2001-10-23 | 2003-05-01 | Ferring B.V. | Inhibiteurs de la dipeptidyl peptidase iv |
WO2003035067A1 (fr) | 2001-10-23 | 2003-05-01 | Ferring B.V. | Inhibiteurs de dipeptidyl peptidase iv (dp-iv) tenant lieu d'agents anti-diabetiques |
US20030096857A1 (en) | 1999-11-30 | 2003-05-22 | Evans David Michael | Novel antidiabetic agents |
WO2003068757A1 (fr) | 2002-02-13 | 2003-08-21 | F. Hoffmann-La Roche Ag | Nouveaux derives de pyridine et de pyrimidine |
WO2003074500A2 (fr) | 2002-03-06 | 2003-09-12 | Sanofi-Aventis | Nouveaux composes |
US20030216450A1 (en) | 2000-04-26 | 2003-11-20 | Evans David Michael | Inhibitors of dipeptidyl peptidase IV |
US20030225102A1 (en) | 2002-04-08 | 2003-12-04 | Torrent Pharmaceuticals Ltd. | Novel compounds and therapeutic uses thereof |
WO2004037181A2 (fr) | 2002-10-23 | 2004-05-06 | Bristol-Myers Squibb Company | Inhibiteurs de dipeptidyl peptidase iv a base de glycinenitrile et procedes correspondants |
WO2005095339A1 (fr) * | 2004-03-31 | 2005-10-13 | Pfizer Products Inc. | Dicyanopyrrolidines inhibiteurs de la dipeptidyl peptidase iv |
WO2007120702A2 (fr) * | 2006-04-11 | 2007-10-25 | Arena Pharmaceuticals, Inc. | Agonistes du récepteur de gpr119 dans des procédés d'augmentation de la masse osseuse et de traitement de l'ostéoporose et autres états se caractérisant par une masse osseuse faible, et thérapie de combinaison associée |
KR20080093203A (ko) * | 2007-04-16 | 2008-10-21 | 경희대학교 산학협력단 | 알기닌, 신남산, 다이드진, 푸에라린, 팔마틴, 베르베린또는 베타-디-글루코피라노사이드로부터 선택된 화합물을유효성분으로 함유하는 골절 질환의 예방 및 치료용 조성물 |
WO2009069838A1 (fr) * | 2007-11-29 | 2009-06-04 | Seoul National University Industry Foundation | Composition comprenant la berbérine pour activer la différenciation des ostéoblastes |
-
2018
- 2018-03-09 WO PCT/EP2018/055931 patent/WO2018162722A1/fr active Application Filing
Patent Citations (60)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO1993010127A1 (fr) | 1991-11-22 | 1993-05-27 | Boehringer Ingelheim Pharmaceuticals, Inc. | Procede de production d'un ester de prolineboronate |
WO1995015309A1 (fr) | 1993-12-03 | 1995-06-08 | Ferring B.V. | Inhibiteurs de la dp-iv-serine protease |
DE19616486A1 (de) | 1996-04-25 | 1997-10-30 | Knoell Hans Forschung Ev | Verfahren zur Senkung des Blutglukosespiegels in Säugern |
WO1998018763A1 (fr) | 1996-10-25 | 1998-05-07 | Tanabe Seiyaku Co., Ltd. | Derives de tetrahydroisoquinoline |
US6124305A (en) | 1996-11-07 | 2000-09-26 | Novartis Ag | Use of N-(substituted glycyl)-2-cyanopyrrolidines in inhibiting dipeptidyl peptidase-IV |
WO1998019998A2 (fr) | 1996-11-07 | 1998-05-14 | Novartis Ag | 2-cyanopyrrolidines a substitution n |
WO1999025719A1 (fr) | 1997-11-18 | 1999-05-27 | Zaidan Hojin Biseibutsu Kagaku Kenkyu Kai | Substance physiologiquement active, la sulphostine, procede de fabrication et utilisation |
WO1999038501A2 (fr) | 1998-02-02 | 1999-08-05 | Trustees Of Tufts College | Procede de regulation du metabolisme du glucose et reactifs afferents |
WO1999046272A1 (fr) | 1998-03-09 | 1999-09-16 | Fondatech Benelux N.V. | Modulateurs de la serine peptidase |
WO1999061431A1 (fr) | 1998-05-28 | 1999-12-02 | Probiodrug Gesellschaft für Arzneimittelforschung mbH | Nouveaux effecteurs de dipeptidylpeptidase iv |
WO1999067278A1 (fr) | 1998-06-24 | 1999-12-29 | Probiodrug Gesellschaft für Arzneimittelforschung mbH | Promedicaments d'inhibiteurs de la dipeptidylpeptidase iv |
WO1999067279A1 (fr) | 1998-06-24 | 1999-12-29 | Probiodrug Gesellschaft für Arzneimittelforschung mbH | Composes d'inhibiteurs instables de la dipeptidylpeptidase iv |
WO2000034241A1 (fr) | 1998-12-10 | 2000-06-15 | Novartis Ag | 2-cyanopyrrolidines n-substitues |
US6107317A (en) | 1999-06-24 | 2000-08-22 | Novartis Ag | N-(substituted glycyl)-thiazolidines, pharmaceutical compositions containing them and their use in inhibiting dipeptidyl peptidase-IV |
WO2001034594A1 (fr) | 1999-11-12 | 2001-05-17 | Guilford Pharmaceuticals, Inc. | Inhibiteurs de la dipeptidyl peptidase iv; methodes de fabrication et d'utilisation desdits inhibiteurs |
US20030096857A1 (en) | 1999-11-30 | 2003-05-22 | Evans David Michael | Novel antidiabetic agents |
WO2001052825A2 (fr) | 2000-01-21 | 2001-07-26 | Novartis Ag | Combinaisons comprenant un inhibiteur de la dipeptidylpeptidase - iv |
WO2001055105A1 (fr) | 2000-01-24 | 2001-08-02 | Novo Nordisk A/S | 2-cyanopyroles et -pyrrolines a substitution n inhibant l'enzyme dpp-iv |
WO2001068603A2 (fr) | 2000-03-10 | 2001-09-20 | Bristol-Myers Squibb Co. | Inhibiteurs de la dipeptidyl peptidase iv, a base de pyrrolidone fusionnee a du cyclopropyle, et procede |
WO2001072290A2 (fr) | 2000-03-31 | 2001-10-04 | Probiodrug Ag | Procede destine a ameliorer le signalement d'ilots dans le diabete sucre et a prevenir ce dernier |
US6482844B1 (en) | 2000-04-07 | 2002-11-19 | Neurogen Corporation | 1-benzylimidazole derivatives |
US20030216450A1 (en) | 2000-04-26 | 2003-11-20 | Evans David Michael | Inhibitors of dipeptidyl peptidase IV |
WO2001081337A1 (fr) | 2000-04-26 | 2001-11-01 | Ferring B.V. | Inhibiteurs de dipeptidyl peptidase iv |
WO2001097808A1 (fr) | 2000-06-19 | 2001-12-27 | Smithkline Beecham Plc | Combinaisons d'inhibiteurs de peptidase iv de dipeptidyl et d'autres agents antidiabetiques pour traiter le diabete sucre |
WO2002002560A2 (fr) | 2000-07-04 | 2002-01-10 | Novo Nordisk A/S | Composes heterocycliques inhibiteurs de l'enzyme dpp-iv |
WO2002014271A1 (fr) | 2000-08-10 | 2002-02-21 | Mitsubishi Pharma Corporation | Dérivés de proline et leur utilisation comme médicaments |
WO2002038541A1 (fr) | 2000-11-10 | 2002-05-16 | Taisho Pharmaceutical Co., Ltd. | Derives de cyanopyrrolidine |
WO2002051836A1 (fr) | 2000-12-27 | 2002-07-04 | Kyowa Hakko Kogyo Co., Ltd. | Inhibiteur de dipeptidyl peptidase iv |
WO2002053548A1 (fr) | 2000-12-27 | 2002-07-11 | Banyu Pharmaceutical Co.,Ltd. | Derives de la benzothiazepine |
WO2002062764A1 (fr) | 2001-02-02 | 2002-08-15 | Takeda Chemical Industries, Ltd. | Composes heterocycliques condenses |
WO2002066627A1 (fr) | 2001-02-16 | 2002-08-29 | Bayer Aktiengesellschaft | Regulation de la dipeptidyle peptidase 8 humaine |
WO2002068420A1 (fr) | 2001-02-24 | 2002-09-06 | Boehringer Ingelheim Pharma Gmbh & Co. Kg | Derives xanthine, fabrication et utilisations en tant qu'agents pharmaceutiques |
WO2002067918A1 (fr) | 2001-02-27 | 2002-09-06 | Banyu Pharmaceutical Co., Ltd. | Nouveau derive de diallylmethylamine |
WO2002076450A1 (fr) | 2001-03-27 | 2002-10-03 | Merck & Co., Inc. | Inhibiteurs de peptidase dipeptidyl destines au traitement ou a la prevention du diabete |
US20030087950A1 (en) | 2001-03-28 | 2003-05-08 | Denanteuil Guillaume | New alpha-amino acid sulphonyl compounds |
EP1245568A1 (fr) | 2001-03-28 | 2002-10-02 | Les Laboratoires Servier | Dérives sulfonyles d'-aminoacides et leur utilisation en tant qu'inhibiteurs de dipeptidyl-peptidase IV ( DPP IV) |
WO2002083109A1 (fr) | 2001-04-11 | 2002-10-24 | Ferring Bv | Traitement du diabete de type 2 a l'aide d'inhibiteurs de dipeptidylpeptidase iv |
WO2002083128A1 (fr) | 2001-04-12 | 2002-10-24 | Bristol-Myers Squibb Company | Inhibiteurs a base de 2,1-oxazoline et 1,2-pyrazoline de la dipeptidyl peptidase iv et methode associee |
WO2002088090A2 (fr) | 2001-04-27 | 2002-11-07 | Vertex Pharmaceuticals Incorporated | Inhibiteurs de kinases derives du pyrazole |
EP1258476A1 (fr) | 2001-05-15 | 2002-11-20 | Les Laboratoires Servier | Dérivés d'alpha-amino-acides, leur procédé de préparation ainsi que leur utilisation en tant qu'inhibiteurs de dipeptidyl-peptidase IV (DPP IV) |
WO2003002942A1 (fr) | 2001-05-18 | 2003-01-09 | Faeger Jan G | Procede pour determiner la position et/ou l'orientation d'un etre vivant par rapport a un environnement |
EP1258480A1 (fr) | 2001-05-18 | 2002-11-20 | Eisai Co., Ltd. | Dérivés de N-Carbamoylazole et leur utilisation pharmaceutique |
WO2003000181A2 (fr) | 2001-06-20 | 2003-01-03 | Merck & Co., Inc. | Inhibiteurs de dipeptidyl peptidase utilises dans le traitement du diabete |
WO2003000180A2 (fr) | 2001-06-20 | 2003-01-03 | Merck & Co., Inc. | Inhibiteurs de dipeptidyle peptidase pour le traitement du diabete |
WO2003003727A1 (fr) | 2001-06-27 | 2003-01-09 | Nokia Corporation | Guide de programmes electronique presentant une fonctionnalite de visualisation d'echantillons et d'historique par fenetres a incrustation d'images |
WO2003002553A2 (fr) | 2001-06-27 | 2003-01-09 | Smithkline Beecham Corporation | Fluoropyrrolidines inhibitrices de la dipeptidyl peptidase |
WO2003003250A1 (fr) | 2001-06-28 | 2003-01-09 | Hywire Ltd. | Memoire adressable par son contenu constituee de plages |
WO2003004496A1 (fr) | 2001-07-03 | 2003-01-16 | Novo Nordisk A/S | Derives de purine inhibiteurs de dpp-iv pour le traitement du diabete |
WO2003004498A1 (fr) | 2001-07-06 | 2003-01-16 | Merck & Co., Inc. | Pyrazines beta-amino tetrahydroimidazo (1, 2-a) et pyrazines tetrahydrotrioazolo (4, 3-a) utilisees en tant qu'inhibiteurs de la dipeptidyl peptidase dans le traitement ou la prevention du diabete |
WO2003024965A2 (fr) | 2001-09-19 | 2003-03-27 | Novo Nordisk A/S | Composes heterocycliques constituant des inhibiteurs de l'enzyme dpp-iv |
WO2003035067A1 (fr) | 2001-10-23 | 2003-05-01 | Ferring B.V. | Inhibiteurs de dipeptidyl peptidase iv (dp-iv) tenant lieu d'agents anti-diabetiques |
WO2003035057A1 (fr) | 2001-10-23 | 2003-05-01 | Ferring B.V. | Inhibiteurs de la dipeptidyl peptidase iv |
WO2003068757A1 (fr) | 2002-02-13 | 2003-08-21 | F. Hoffmann-La Roche Ag | Nouveaux derives de pyridine et de pyrimidine |
WO2003074500A2 (fr) | 2002-03-06 | 2003-09-12 | Sanofi-Aventis | Nouveaux composes |
US20030225102A1 (en) | 2002-04-08 | 2003-12-04 | Torrent Pharmaceuticals Ltd. | Novel compounds and therapeutic uses thereof |
WO2004037181A2 (fr) | 2002-10-23 | 2004-05-06 | Bristol-Myers Squibb Company | Inhibiteurs de dipeptidyl peptidase iv a base de glycinenitrile et procedes correspondants |
WO2005095339A1 (fr) * | 2004-03-31 | 2005-10-13 | Pfizer Products Inc. | Dicyanopyrrolidines inhibiteurs de la dipeptidyl peptidase iv |
WO2007120702A2 (fr) * | 2006-04-11 | 2007-10-25 | Arena Pharmaceuticals, Inc. | Agonistes du récepteur de gpr119 dans des procédés d'augmentation de la masse osseuse et de traitement de l'ostéoporose et autres états se caractérisant par une masse osseuse faible, et thérapie de combinaison associée |
KR20080093203A (ko) * | 2007-04-16 | 2008-10-21 | 경희대학교 산학협력단 | 알기닌, 신남산, 다이드진, 푸에라린, 팔마틴, 베르베린또는 베타-디-글루코피라노사이드로부터 선택된 화합물을유효성분으로 함유하는 골절 질환의 예방 및 치료용 조성물 |
WO2009069838A1 (fr) * | 2007-11-29 | 2009-06-04 | Seoul National University Industry Foundation | Composition comprenant la berbérine pour activer la différenciation des ostéoblastes |
Non-Patent Citations (9)
Title |
---|
AMBROSI THOMAS H ET AL: "Adipocyte Accumulation in the Bone Marrow during Obesity and Aging Impairs Stem Cell-Based Hematopoietic and Bone Regeneration", CELL STEM CELL, vol. 20, no. 6, 1 June 2017 (2017-06-01), pages 771, XP085053510, ISSN: 1934-5909, DOI: 10.1016/J.STEM.2017.02.009 * |
DATABASE WPI Week 200920, Derwent World Patents Index; AN 2009-E85101, XP002781195 * |
HISTING ET AL., BONE, vol. 49, no. 4, 2011, pages 591 - 599 |
SCHULZ ET AL., PROC. NATL. ACAD. SCI. U. S. A., vol. 108, 2011, pages 143 - 148 |
STEENHUIS ET AL., CALCIF. TISSUE INT., vol. 82, 2008, pages 44 - 56 |
THE UNITED STATES PHARMACOPEIAL CONVENTION, PHARMACOPEIAL FORUM, vol. 26, no. 1, 2000, pages 223 |
WU, J. BIOL. CHEM., vol. 262, 1987, pages 4429 - 4432 |
YINQIU YANG ET AL: "Effect of Dipeptidyl Peptidase-4 Inhibitors on Bone Metabolism and the Possible Underlying Mechanisms", FRONTIERS IN PHARMACOLOGY, vol. 8, 25 July 2017 (2017-07-25), XP055476027, DOI: 10.3389/fphar.2017.00487 * |
YOUNG SIL EOM ET AL: "Protective Effects of Vildagliptin against Pioglitazone-Induced Bone Loss in Type 2 Diabetic Rats", PLOS ONE, vol. 11, no. 12, 20 December 2016 (2016-12-20), pages e0168569, XP055476253, DOI: 10.1371/journal.pone.0168569 * |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Kumar et al. | Mobilization of bone marrow mesenchymal stem cells in vivo augments bone healing in a mouse model of segmental bone defect | |
US11612639B2 (en) | Methods and compositions for rejuvenating skeletal muscle stem cells | |
RU2525913C1 (ru) | Новый пептид и его применение | |
US20160250224A1 (en) | Orexin-control of bone formation and loss | |
Rivera et al. | Local injections of β-NGF accelerates endochondral fracture repair by promoting cartilage to bone conversion | |
US20200016233A1 (en) | Molecular Composition for Enhancing and Rejuvenating Maintenance and Repair of Mammalian Tissues | |
CN101553565B (zh) | 从血液特别是外周血扩增成体干细胞的方法及在医药领域的相关应用 | |
Xu et al. | Rosuvastatin treatment activates JAK-STAT pathway and increases efficacy of allogeneic mesenchymal stem cell transplantation in infarcted hearts | |
EP2937088B1 (fr) | Composition présentant une activité de réparation tissulaire et son utilisation | |
KR20140013887A (ko) | 눈 손상 및 질환 치료용 성체 줄기 세포/전구 세포 및 줄기 세포 단백질 | |
JP2008530004A (ja) | サルコペニア(加齢性筋肉減退性疾患)を治療するためのミオスタチン(gdf−8)拮抗物質の使用 | |
US20090038022A1 (en) | IGF-1 Novel peptides | |
US9220746B2 (en) | Compounds and methods for treating bone disorders and controlling weight | |
EP3921029B1 (fr) | Composition comprenant ccl5 pour utilisation dans le traitement de la rupture osseuse, de la fracture osseuse, de la dégénérescence osseuse et de l'ostéoporose | |
US20210379104A1 (en) | Pharmaceutical composition comprising isolated mitochondria for preventing or treating tendinopathy | |
Deng et al. | Bone marrow-derived mesenchymal stem cells overexpressed with miR-182-5p protects against brain injury in a mouse model of cerebral ischemia | |
KR101736280B1 (ko) | 관절염 예방 또는 치료용 약학적 조성물 | |
JP7562066B2 (ja) | 変形性関節症の予防又は治療剤、及び変形性関節症の予防又は治療用医薬組成物 | |
WO2018162722A1 (fr) | Inhibiteurs de dpp-4 à utiliser dans le traitement de fractures osseuses | |
Shi et al. | Nicotinamide mononucleotide enhances fracture healing by promoting skeletal stem cell proliferation | |
JP7366249B2 (ja) | タウタンパク質の蓄積、凝集及びタングル形成抑制用組成物及びその抑制方法 | |
KR20130023797A (ko) | 자연살해세포 억제제 및 간엽줄기세포를 유효성분으로 포함하는 이식편대숙주질환의 예방 또는 치료를 위한 세포치료제 조성물 | |
WO2014055986A1 (fr) | Méthodes et compositions pour la multiplication de cellules souches et progénitrices hématopoïétiques | |
US20210177908A1 (en) | Anabolic targeting stem cell gene therapy for osteoporosis | |
WO2022232116A1 (fr) | Activation biochimique de cellules souches squelettiques dysfonctionnelles pour la régénération squelettique |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 18710839 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 18710839 Country of ref document: EP Kind code of ref document: A1 |