TW201130977A - Mesenchymal stem cells (MSCs) isolated from mobilized peripheral blood - Google Patents
Mesenchymal stem cells (MSCs) isolated from mobilized peripheral blood Download PDFInfo
- Publication number
- TW201130977A TW201130977A TW099142522A TW99142522A TW201130977A TW 201130977 A TW201130977 A TW 201130977A TW 099142522 A TW099142522 A TW 099142522A TW 99142522 A TW99142522 A TW 99142522A TW 201130977 A TW201130977 A TW 201130977A
- Authority
- TW
- Taiwan
- Prior art keywords
- peripheral blood
- individual
- csf
- cells
- msc
- Prior art date
Links
- 239000011886 peripheral blood Substances 0.000 title claims abstract description 351
- 210000005259 peripheral blood Anatomy 0.000 title claims abstract description 350
- 210000002901 mesenchymal stem cell Anatomy 0.000 title claims description 590
- 238000000034 method Methods 0.000 claims abstract description 244
- 210000000130 stem cell Anatomy 0.000 claims abstract description 175
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 claims abstract description 153
- 102000004457 Granulocyte-Macrophage Colony-Stimulating Factor Human genes 0.000 claims abstract description 153
- 230000001483 mobilizing effect Effects 0.000 claims abstract description 49
- 108010017080 Granulocyte Colony-Stimulating Factor Proteins 0.000 claims abstract 10
- 102000004269 Granulocyte Colony-Stimulating Factor Human genes 0.000 claims abstract 10
- 210000004027 cell Anatomy 0.000 claims description 429
- 239000003795 chemical substances by application Substances 0.000 claims description 78
- 210000004369 blood Anatomy 0.000 claims description 51
- 239000008280 blood Substances 0.000 claims description 50
- 238000011282 treatment Methods 0.000 claims description 45
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 40
- 238000000926 separation method Methods 0.000 claims description 40
- 201000010099 disease Diseases 0.000 claims description 38
- 230000001965 increasing effect Effects 0.000 claims description 20
- 102100032912 CD44 antigen Human genes 0.000 claims description 19
- 101000868273 Homo sapiens CD44 antigen Proteins 0.000 claims description 19
- 101000800116 Homo sapiens Thy-1 membrane glycoprotein Proteins 0.000 claims description 19
- 102100033523 Thy-1 membrane glycoprotein Human genes 0.000 claims description 19
- 238000000338 in vitro Methods 0.000 claims description 19
- 238000004091 panning Methods 0.000 claims description 16
- 238000002560 therapeutic procedure Methods 0.000 claims description 16
- 230000001172 regenerating effect Effects 0.000 claims description 15
- 101000935043 Homo sapiens Integrin beta-1 Proteins 0.000 claims description 14
- 102100025304 Integrin beta-1 Human genes 0.000 claims description 14
- 102100022464 5'-nucleotidase Human genes 0.000 claims description 13
- 101000678236 Homo sapiens 5'-nucleotidase Proteins 0.000 claims description 13
- 210000004504 adult stem cell Anatomy 0.000 claims description 12
- 206010036790 Productive cough Diseases 0.000 claims description 11
- 210000003802 sputum Anatomy 0.000 claims description 11
- 208000024794 sputum Diseases 0.000 claims description 11
- 238000002955 isolation Methods 0.000 claims description 10
- 108050003558 Interleukin-17 Proteins 0.000 claims description 8
- 102000013691 Interleukin-17 Human genes 0.000 claims description 8
- 230000002093 peripheral effect Effects 0.000 claims description 8
- 238000010187 selection method Methods 0.000 claims description 8
- 238000005534 hematocrit Methods 0.000 claims description 7
- RCINICONZNJXQF-MZXODVADSA-N taxol Chemical compound O([C@@H]1[C@@]2(C[C@@H](C(C)=C(C2(C)C)[C@H](C([C@]2(C)[C@@H](O)C[C@H]3OC[C@]3([C@H]21)OC(C)=O)=O)OC(=O)C)OC(=O)[C@H](O)[C@@H](NC(=O)C=1C=CC=CC=1)C=1C=CC=CC=1)O)C(=O)C1=CC=CC=C1 RCINICONZNJXQF-MZXODVADSA-N 0.000 claims description 7
- 239000002458 cell surface marker Substances 0.000 claims description 6
- 210000001671 embryonic stem cell Anatomy 0.000 claims description 6
- PCHJSUWPFVWCPO-UHFFFAOYSA-N gold Chemical compound [Au] PCHJSUWPFVWCPO-UHFFFAOYSA-N 0.000 claims description 6
- 229910052737 gold Inorganic materials 0.000 claims description 6
- 239000010931 gold Substances 0.000 claims description 6
- 229930012538 Paclitaxel Natural products 0.000 claims description 5
- 229960001592 paclitaxel Drugs 0.000 claims description 5
- ZDZOTLJHXYCWBA-VCVYQWHSSA-N N-debenzoyl-N-(tert-butoxycarbonyl)-10-deacetyltaxol Chemical compound O([C@H]1[C@H]2[C@@](C([C@H](O)C3=C(C)[C@@H](OC(=O)[C@H](O)[C@@H](NC(=O)OC(C)(C)C)C=4C=CC=CC=4)C[C@]1(O)C3(C)C)=O)(C)[C@@H](O)C[C@H]1OC[C@]12OC(=O)C)C(=O)C1=CC=CC=C1 ZDZOTLJHXYCWBA-VCVYQWHSSA-N 0.000 claims description 2
- 238000002617 apheresis Methods 0.000 claims description 2
- 229960003668 docetaxel Drugs 0.000 claims description 2
- 238000002372 labelling Methods 0.000 claims description 2
- 241000218691 Cupressaceae Species 0.000 claims 1
- 230000000903 blocking effect Effects 0.000 claims 1
- 229910052594 sapphire Inorganic materials 0.000 claims 1
- 239000010980 sapphire Substances 0.000 claims 1
- 239000002689 soil Substances 0.000 claims 1
- 210000001185 bone marrow Anatomy 0.000 abstract description 91
- 238000001727 in vivo Methods 0.000 abstract description 16
- 210000004271 bone marrow stromal cell Anatomy 0.000 abstract description 10
- 210000003995 blood forming stem cell Anatomy 0.000 abstract description 5
- 230000001105 regulatory effect Effects 0.000 abstract description 2
- 239000000523 sample Substances 0.000 description 97
- 239000000203 mixture Substances 0.000 description 62
- 210000001519 tissue Anatomy 0.000 description 52
- 108090000623 proteins and genes Proteins 0.000 description 50
- 230000004069 differentiation Effects 0.000 description 36
- 239000002609 medium Substances 0.000 description 35
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 31
- 150000001875 compounds Chemical class 0.000 description 28
- 102000004169 proteins and genes Human genes 0.000 description 28
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 27
- 239000000047 product Substances 0.000 description 27
- 239000003814 drug Substances 0.000 description 26
- 206010017076 Fracture Diseases 0.000 description 24
- 238000003860 storage Methods 0.000 description 23
- 239000000243 solution Substances 0.000 description 20
- 230000000694 effects Effects 0.000 description 19
- 102100031573 Hematopoietic progenitor cell antigen CD34 Human genes 0.000 description 18
- 101000777663 Homo sapiens Hematopoietic progenitor cell antigen CD34 Proteins 0.000 description 18
- 101000958041 Homo sapiens Musculin Proteins 0.000 description 18
- 210000000988 bone and bone Anatomy 0.000 description 18
- 230000014509 gene expression Effects 0.000 description 18
- 102000046949 human MSC Human genes 0.000 description 18
- 230000008569 process Effects 0.000 description 18
- 208000010392 Bone Fractures Diseases 0.000 description 17
- 208000027418 Wounds and injury Diseases 0.000 description 17
- 239000007788 liquid Substances 0.000 description 17
- 210000000056 organ Anatomy 0.000 description 17
- 238000002659 cell therapy Methods 0.000 description 16
- 230000006870 function Effects 0.000 description 16
- 238000012360 testing method Methods 0.000 description 16
- 108010002386 Interleukin-3 Proteins 0.000 description 15
- 102000000646 Interleukin-3 Human genes 0.000 description 15
- 241001465754 Metazoa Species 0.000 description 15
- 238000003556 assay Methods 0.000 description 15
- 229940076264 interleukin-3 Drugs 0.000 description 15
- 230000035755 proliferation Effects 0.000 description 15
- 230000035876 healing Effects 0.000 description 14
- 239000007924 injection Substances 0.000 description 14
- 238000002347 injection Methods 0.000 description 14
- 238000005138 cryopreservation Methods 0.000 description 13
- 229940079593 drug Drugs 0.000 description 13
- 210000000265 leukocyte Anatomy 0.000 description 13
- 239000000126 substance Substances 0.000 description 13
- 238000002054 transplantation Methods 0.000 description 13
- 102100027286 Fanconi anemia group C protein Human genes 0.000 description 12
- 101000738771 Homo sapiens Receptor-type tyrosine-protein phosphatase C Proteins 0.000 description 12
- 102100037422 Receptor-type tyrosine-protein phosphatase C Human genes 0.000 description 12
- 210000003714 granulocyte Anatomy 0.000 description 12
- 238000001990 intravenous administration Methods 0.000 description 12
- 150000007523 nucleic acids Chemical class 0.000 description 12
- 241000282412 Homo Species 0.000 description 11
- 206010052428 Wound Diseases 0.000 description 11
- 238000004458 analytical method Methods 0.000 description 11
- 230000008859 change Effects 0.000 description 11
- 210000002540 macrophage Anatomy 0.000 description 11
- 230000001225 therapeutic effect Effects 0.000 description 11
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 10
- 150000001413 amino acids Chemical class 0.000 description 10
- 239000012595 freezing medium Substances 0.000 description 10
- 239000010410 layer Substances 0.000 description 10
- YIQPUIGJQJDJOS-UHFFFAOYSA-N plerixafor Chemical compound C=1C=C(CN2CCNCCCNCCNCCC2)C=CC=1CN1CCCNCCNCCCNCC1 YIQPUIGJQJDJOS-UHFFFAOYSA-N 0.000 description 10
- 239000000725 suspension Substances 0.000 description 10
- 230000029663 wound healing Effects 0.000 description 10
- OZFAFGSSMRRTDW-UHFFFAOYSA-N (2,4-dichlorophenyl) benzenesulfonate Chemical compound ClC1=CC(Cl)=CC=C1OS(=O)(=O)C1=CC=CC=C1 OZFAFGSSMRRTDW-UHFFFAOYSA-N 0.000 description 9
- 102100031650 C-X-C chemokine receptor type 4 Human genes 0.000 description 9
- 239000012591 Dulbecco’s Phosphate Buffered Saline Substances 0.000 description 9
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 9
- 101000922348 Homo sapiens C-X-C chemokine receptor type 4 Proteins 0.000 description 9
- 230000001464 adherent effect Effects 0.000 description 9
- 238000004113 cell culture Methods 0.000 description 9
- 239000000306 component Substances 0.000 description 9
- 230000006378 damage Effects 0.000 description 9
- 230000007774 longterm Effects 0.000 description 9
- 239000003550 marker Substances 0.000 description 9
- 102000039446 nucleic acids Human genes 0.000 description 9
- 108020004707 nucleic acids Proteins 0.000 description 9
- 230000007170 pathology Effects 0.000 description 9
- 229960002169 plerixafor Drugs 0.000 description 9
- 239000002243 precursor Substances 0.000 description 9
- 230000008439 repair process Effects 0.000 description 9
- 210000002966 serum Anatomy 0.000 description 9
- 238000007920 subcutaneous administration Methods 0.000 description 9
- 108091006905 Human Serum Albumin Proteins 0.000 description 8
- 102000008100 Human Serum Albumin Human genes 0.000 description 8
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 8
- 238000009472 formulation Methods 0.000 description 8
- 239000003102 growth factor Substances 0.000 description 8
- 238000001802 infusion Methods 0.000 description 8
- 108020004999 messenger RNA Proteins 0.000 description 8
- 230000037361 pathway Effects 0.000 description 8
- 239000002953 phosphate buffered saline Substances 0.000 description 8
- 241000894007 species Species 0.000 description 8
- 108010073385 Fibrin Proteins 0.000 description 7
- 102000009123 Fibrin Human genes 0.000 description 7
- BWGVNKXGVNDBDI-UHFFFAOYSA-N Fibrin monomer Chemical compound CNC(=O)CNC(=O)CN BWGVNKXGVNDBDI-UHFFFAOYSA-N 0.000 description 7
- 102000000589 Interleukin-1 Human genes 0.000 description 7
- 108010002352 Interleukin-1 Proteins 0.000 description 7
- -1 Stro-1 Proteins 0.000 description 7
- 210000002798 bone marrow cell Anatomy 0.000 description 7
- 230000001413 cellular effect Effects 0.000 description 7
- 210000001175 cerebrospinal fluid Anatomy 0.000 description 7
- 210000002808 connective tissue Anatomy 0.000 description 7
- 210000003743 erythrocyte Anatomy 0.000 description 7
- 229950003499 fibrin Drugs 0.000 description 7
- 239000012530 fluid Substances 0.000 description 7
- 239000000499 gel Substances 0.000 description 7
- 238000009396 hybridization Methods 0.000 description 7
- 238000004519 manufacturing process Methods 0.000 description 7
- 239000000463 material Substances 0.000 description 7
- 210000003716 mesoderm Anatomy 0.000 description 7
- 210000003205 muscle Anatomy 0.000 description 7
- 210000000440 neutrophil Anatomy 0.000 description 7
- 230000001575 pathological effect Effects 0.000 description 7
- 230000004044 response Effects 0.000 description 7
- 238000012216 screening Methods 0.000 description 7
- 230000004936 stimulating effect Effects 0.000 description 7
- 238000010257 thawing Methods 0.000 description 7
- 238000012546 transfer Methods 0.000 description 7
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 7
- 102000004127 Cytokines Human genes 0.000 description 6
- 108090000695 Cytokines Proteins 0.000 description 6
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 6
- 102000004890 Interleukin-8 Human genes 0.000 description 6
- 108090001007 Interleukin-8 Proteins 0.000 description 6
- 206010028980 Neoplasm Diseases 0.000 description 6
- 108091034117 Oligonucleotide Proteins 0.000 description 6
- 239000004698 Polyethylene Substances 0.000 description 6
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 6
- 201000011510 cancer Diseases 0.000 description 6
- 230000003833 cell viability Effects 0.000 description 6
- 238000001816 cooling Methods 0.000 description 6
- 238000007667 floating Methods 0.000 description 6
- 239000012634 fragment Substances 0.000 description 6
- 210000001654 germ layer Anatomy 0.000 description 6
- 208000014674 injury Diseases 0.000 description 6
- 229940096397 interleukin-8 Drugs 0.000 description 6
- XKTZWUACRZHVAN-VADRZIEHSA-N interleukin-8 Chemical compound C([C@H](NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@@H](NC(C)=O)CCSC)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H]([C@@H](C)O)C(=O)NCC(=O)N[C@@H](CCSC)C(=O)N1[C@H](CCC1)C(=O)N1[C@H](CCC1)C(=O)N[C@@H](C)C(=O)N[C@H](CC(O)=O)C(=O)N[C@H](CCC(O)=O)C(=O)N[C@H](CC(O)=O)C(=O)N[C@H](CC=1C=CC(O)=CC=1)C(=O)N[C@H](CO)C(=O)N1[C@H](CCC1)C(N)=O)C1=CC=CC=C1 XKTZWUACRZHVAN-VADRZIEHSA-N 0.000 description 6
- 210000001616 monocyte Anatomy 0.000 description 6
- 102000040430 polynucleotide Human genes 0.000 description 6
- 108091033319 polynucleotide Proteins 0.000 description 6
- 239000002157 polynucleotide Substances 0.000 description 6
- 238000002360 preparation method Methods 0.000 description 6
- 230000000750 progressive effect Effects 0.000 description 6
- 239000000758 substrate Substances 0.000 description 6
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 5
- 239000000427 antigen Substances 0.000 description 5
- 108091007433 antigens Proteins 0.000 description 5
- 102000036639 antigens Human genes 0.000 description 5
- 230000015572 biosynthetic process Effects 0.000 description 5
- 210000000601 blood cell Anatomy 0.000 description 5
- 210000001772 blood platelet Anatomy 0.000 description 5
- 230000004663 cell proliferation Effects 0.000 description 5
- 210000001612 chondrocyte Anatomy 0.000 description 5
- 238000012258 culturing Methods 0.000 description 5
- 229960004397 cyclophosphamide Drugs 0.000 description 5
- 230000007613 environmental effect Effects 0.000 description 5
- 210000003979 eosinophil Anatomy 0.000 description 5
- 108020001507 fusion proteins Proteins 0.000 description 5
- 102000037865 fusion proteins Human genes 0.000 description 5
- 230000002068 genetic effect Effects 0.000 description 5
- 230000012010 growth Effects 0.000 description 5
- 239000001963 growth medium Substances 0.000 description 5
- 230000036541 health Effects 0.000 description 5
- 208000015181 infectious disease Diseases 0.000 description 5
- 229910052757 nitrogen Inorganic materials 0.000 description 5
- 210000004940 nucleus Anatomy 0.000 description 5
- 210000000963 osteoblast Anatomy 0.000 description 5
- 239000000546 pharmaceutical excipient Substances 0.000 description 5
- 239000002504 physiological saline solution Substances 0.000 description 5
- 230000009467 reduction Effects 0.000 description 5
- 150000003839 salts Chemical class 0.000 description 5
- 239000011780 sodium chloride Substances 0.000 description 5
- 210000000952 spleen Anatomy 0.000 description 5
- 239000006228 supernatant Substances 0.000 description 5
- 108091023037 Aptamer Proteins 0.000 description 4
- 108010039209 Blood Coagulation Factors Proteins 0.000 description 4
- 102000015081 Blood Coagulation Factors Human genes 0.000 description 4
- 206010017088 Fracture nonunion Diseases 0.000 description 4
- 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 4
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 4
- 241001529936 Murinae Species 0.000 description 4
- 230000004913 activation Effects 0.000 description 4
- 210000001789 adipocyte Anatomy 0.000 description 4
- 230000003321 amplification Effects 0.000 description 4
- 238000010171 animal model Methods 0.000 description 4
- 230000008901 benefit Effects 0.000 description 4
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 4
- 238000001574 biopsy Methods 0.000 description 4
- 239000003114 blood coagulation factor Substances 0.000 description 4
- 210000001124 body fluid Anatomy 0.000 description 4
- 239000010839 body fluid Substances 0.000 description 4
- 210000004413 cardiac myocyte Anatomy 0.000 description 4
- 210000000845 cartilage Anatomy 0.000 description 4
- 230000024245 cell differentiation Effects 0.000 description 4
- 238000006243 chemical reaction Methods 0.000 description 4
- 230000001684 chronic effect Effects 0.000 description 4
- 230000002338 cryopreservative effect Effects 0.000 description 4
- 238000001514 detection method Methods 0.000 description 4
- 210000003981 ectoderm Anatomy 0.000 description 4
- 210000001900 endoderm Anatomy 0.000 description 4
- 210000004700 fetal blood Anatomy 0.000 description 4
- 238000000684 flow cytometry Methods 0.000 description 4
- 230000003394 haemopoietic effect Effects 0.000 description 4
- 230000002757 inflammatory effect Effects 0.000 description 4
- 150000002484 inorganic compounds Chemical class 0.000 description 4
- 229910010272 inorganic material Inorganic materials 0.000 description 4
- 230000002175 menstrual effect Effects 0.000 description 4
- 238000000386 microscopy Methods 0.000 description 4
- 230000035772 mutation Effects 0.000 description 4
- 210000002569 neuron Anatomy 0.000 description 4
- 238000003199 nucleic acid amplification method Methods 0.000 description 4
- 238000005457 optimization Methods 0.000 description 4
- 150000002894 organic compounds Chemical class 0.000 description 4
- 108090000765 processed proteins & peptides Proteins 0.000 description 4
- 238000011084 recovery Methods 0.000 description 4
- 239000012679 serum free medium Substances 0.000 description 4
- 150000003384 small molecules Chemical class 0.000 description 4
- 206010006002 Bone pain Diseases 0.000 description 3
- 108010012236 Chemokines Proteins 0.000 description 3
- 102000019034 Chemokines Human genes 0.000 description 3
- 208000035473 Communicable disease Diseases 0.000 description 3
- 108020004414 DNA Proteins 0.000 description 3
- 108091060211 Expressed sequence tag Proteins 0.000 description 3
- 231100000111 LD50 Toxicity 0.000 description 3
- 241000124008 Mammalia Species 0.000 description 3
- 241000699670 Mus sp. Species 0.000 description 3
- 108091028043 Nucleic acid sequence Proteins 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- 241000239226 Scorpiones Species 0.000 description 3
- 208000013201 Stress fracture Diseases 0.000 description 3
- 102000036693 Thrombopoietin Human genes 0.000 description 3
- 108010041111 Thrombopoietin Proteins 0.000 description 3
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 description 3
- 239000000853 adhesive Substances 0.000 description 3
- 230000001070 adhesive effect Effects 0.000 description 3
- 210000000577 adipose tissue Anatomy 0.000 description 3
- 239000000556 agonist Substances 0.000 description 3
- 230000000735 allogeneic effect Effects 0.000 description 3
- 230000001580 bacterial effect Effects 0.000 description 3
- 239000012503 blood component Substances 0.000 description 3
- 230000010478 bone regeneration Effects 0.000 description 3
- 239000000872 buffer Substances 0.000 description 3
- 150000001720 carbohydrates Chemical class 0.000 description 3
- 235000014633 carbohydrates Nutrition 0.000 description 3
- 230000000747 cardiac effect Effects 0.000 description 3
- 239000006285 cell suspension Substances 0.000 description 3
- 150000005829 chemical entities Chemical class 0.000 description 3
- 238000002512 chemotherapy Methods 0.000 description 3
- 238000011109 contamination Methods 0.000 description 3
- 235000005911 diet Nutrition 0.000 description 3
- 230000037213 diet Effects 0.000 description 3
- 239000003085 diluting agent Substances 0.000 description 3
- 238000010790 dilution Methods 0.000 description 3
- 239000012895 dilution Substances 0.000 description 3
- 239000002552 dosage form Substances 0.000 description 3
- 239000000975 dye Substances 0.000 description 3
- 230000003511 endothelial effect Effects 0.000 description 3
- 239000002158 endotoxin Substances 0.000 description 3
- 238000005516 engineering process Methods 0.000 description 3
- 230000001747 exhibiting effect Effects 0.000 description 3
- 239000007789 gas Substances 0.000 description 3
- 238000010353 genetic engineering Methods 0.000 description 3
- 210000004602 germ cell Anatomy 0.000 description 3
- 239000008103 glucose Substances 0.000 description 3
- 238000002513 implantation Methods 0.000 description 3
- 230000001976 improved effect Effects 0.000 description 3
- 210000002570 interstitial cell Anatomy 0.000 description 3
- 238000007918 intramuscular administration Methods 0.000 description 3
- 238000007912 intraperitoneal administration Methods 0.000 description 3
- 229920006008 lipopolysaccharide Polymers 0.000 description 3
- 239000011159 matrix material Substances 0.000 description 3
- 238000005259 measurement Methods 0.000 description 3
- 238000002493 microarray Methods 0.000 description 3
- 230000004048 modification Effects 0.000 description 3
- 238000012986 modification Methods 0.000 description 3
- 230000000877 morphologic effect Effects 0.000 description 3
- 229930014626 natural product Natural products 0.000 description 3
- 210000005036 nerve Anatomy 0.000 description 3
- 230000002974 pharmacogenomic effect Effects 0.000 description 3
- 210000002826 placenta Anatomy 0.000 description 3
- 102000004196 processed proteins & peptides Human genes 0.000 description 3
- 102000005962 receptors Human genes 0.000 description 3
- 108020003175 receptors Proteins 0.000 description 3
- 210000002460 smooth muscle Anatomy 0.000 description 3
- 239000007787 solid Substances 0.000 description 3
- 238000001228 spectrum Methods 0.000 description 3
- 239000003381 stabilizer Substances 0.000 description 3
- 210000002536 stromal cell Anatomy 0.000 description 3
- 239000007929 subcutaneous injection Substances 0.000 description 3
- 230000004083 survival effect Effects 0.000 description 3
- 208000024891 symptom Diseases 0.000 description 3
- 230000009885 systemic effect Effects 0.000 description 3
- 238000012549 training Methods 0.000 description 3
- 230000032258 transport Effects 0.000 description 3
- 239000003981 vehicle Substances 0.000 description 3
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 2
- 206010001935 American trypanosomiasis Diseases 0.000 description 2
- 102000008076 Angiogenic Proteins Human genes 0.000 description 2
- 108010074415 Angiogenic Proteins Proteins 0.000 description 2
- 241000894006 Bacteria Species 0.000 description 2
- 241000283690 Bos taurus Species 0.000 description 2
- 241000282472 Canis lupus familiaris Species 0.000 description 2
- 235000002566 Capsicum Nutrition 0.000 description 2
- 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 2
- 238000002965 ELISA Methods 0.000 description 2
- 108010029961 Filgrastim Proteins 0.000 description 2
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 2
- 102000003886 Glycoproteins Human genes 0.000 description 2
- 108090000288 Glycoproteins Proteins 0.000 description 2
- ZRALSGWEFCBTJO-UHFFFAOYSA-N Guanidine Chemical compound NC(N)=N ZRALSGWEFCBTJO-UHFFFAOYSA-N 0.000 description 2
- 206010019233 Headaches Diseases 0.000 description 2
- HTTJABKRGRZYRN-UHFFFAOYSA-N Heparin Chemical compound OC1C(NC(=O)C)C(O)OC(COS(O)(=O)=O)C1OC1C(OS(O)(=O)=O)C(O)C(OC2C(C(OS(O)(=O)=O)C(OC3C(C(O)C(O)C(O3)C(O)=O)OS(O)(=O)=O)C(CO)O2)NS(O)(=O)=O)C(C(O)=O)O1 HTTJABKRGRZYRN-UHFFFAOYSA-N 0.000 description 2
- 101000946889 Homo sapiens Monocyte differentiation antigen CD14 Proteins 0.000 description 2
- 108090001030 Lipoproteins Proteins 0.000 description 2
- 102000004895 Lipoproteins Human genes 0.000 description 2
- 102100035877 Monocyte differentiation antigen CD14 Human genes 0.000 description 2
- 206010028813 Nausea Diseases 0.000 description 2
- 241001494479 Pecora Species 0.000 description 2
- 241000758706 Piperaceae Species 0.000 description 2
- 108010076504 Protein Sorting Signals Proteins 0.000 description 2
- 241000700159 Rattus Species 0.000 description 2
- 101100247004 Rattus norvegicus Qsox1 gene Proteins 0.000 description 2
- 108020004511 Recombinant DNA Proteins 0.000 description 2
- 108010083644 Ribonucleases Proteins 0.000 description 2
- 102000006382 Ribonucleases Human genes 0.000 description 2
- 206010072170 Skin wound Diseases 0.000 description 2
- 108020004459 Small interfering RNA Proteins 0.000 description 2
- 241000282887 Suidae Species 0.000 description 2
- 102000040945 Transcription factor Human genes 0.000 description 2
- 108091023040 Transcription factor Proteins 0.000 description 2
- 241000223109 Trypanosoma cruzi Species 0.000 description 2
- 108090000631 Trypsin Proteins 0.000 description 2
- 102000004142 Trypsin Human genes 0.000 description 2
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 2
- 241000700605 Viruses Species 0.000 description 2
- 230000009471 action Effects 0.000 description 2
- 239000004480 active ingredient Substances 0.000 description 2
- 239000002671 adjuvant Substances 0.000 description 2
- 230000032683 aging Effects 0.000 description 2
- 229940035676 analgesics Drugs 0.000 description 2
- 239000000730 antalgic agent Substances 0.000 description 2
- 239000003242 anti bacterial agent Substances 0.000 description 2
- 230000003367 anti-collagen effect Effects 0.000 description 2
- 239000002260 anti-inflammatory agent Substances 0.000 description 2
- 229940121363 anti-inflammatory agent Drugs 0.000 description 2
- 230000001754 anti-pyretic effect Effects 0.000 description 2
- 239000003146 anticoagulant agent Substances 0.000 description 2
- 229940127219 anticoagulant drug Drugs 0.000 description 2
- 239000002221 antipyretic Substances 0.000 description 2
- 229940125716 antipyretic agent Drugs 0.000 description 2
- 230000006907 apoptotic process Effects 0.000 description 2
- 238000003491 array Methods 0.000 description 2
- 238000011130 autologous cell therapy Methods 0.000 description 2
- 239000011324 bead Substances 0.000 description 2
- 230000003115 biocidal effect Effects 0.000 description 2
- 230000004071 biological effect Effects 0.000 description 2
- 238000010322 bone marrow transplantation Methods 0.000 description 2
- 239000011575 calcium Substances 0.000 description 2
- 239000006143 cell culture medium Substances 0.000 description 2
- 239000002771 cell marker Substances 0.000 description 2
- 210000000170 cell membrane Anatomy 0.000 description 2
- 230000012292 cell migration Effects 0.000 description 2
- 239000003153 chemical reaction reagent Substances 0.000 description 2
- 230000004087 circulation Effects 0.000 description 2
- 239000002299 complementary DNA Substances 0.000 description 2
- 239000012141 concentrate Substances 0.000 description 2
- 235000008504 concentrate Nutrition 0.000 description 2
- 238000009833 condensation Methods 0.000 description 2
- 230000005494 condensation Effects 0.000 description 2
- 230000001276 controlling effect Effects 0.000 description 2
- 230000034994 death Effects 0.000 description 2
- 230000003247 decreasing effect Effects 0.000 description 2
- 230000007547 defect Effects 0.000 description 2
- 230000002950 deficient Effects 0.000 description 2
- 238000013461 design Methods 0.000 description 2
- 230000018109 developmental process Effects 0.000 description 2
- 208000035475 disorder Diseases 0.000 description 2
- 238000004090 dissolution Methods 0.000 description 2
- 238000009826 distribution Methods 0.000 description 2
- 230000007717 exclusion Effects 0.000 description 2
- 206010016256 fatigue Diseases 0.000 description 2
- 230000008175 fetal development Effects 0.000 description 2
- 238000001415 gene therapy Methods 0.000 description 2
- 229960001031 glucose Drugs 0.000 description 2
- 235000011187 glycerol Nutrition 0.000 description 2
- 210000000224 granular leucocyte Anatomy 0.000 description 2
- 231100000869 headache Toxicity 0.000 description 2
- 238000010438 heat treatment Methods 0.000 description 2
- 210000000777 hematopoietic system Anatomy 0.000 description 2
- 229920000669 heparin Polymers 0.000 description 2
- 229960002897 heparin Drugs 0.000 description 2
- 210000003494 hepatocyte Anatomy 0.000 description 2
- 239000012676 herbal extract Substances 0.000 description 2
- 238000007901 in situ hybridization Methods 0.000 description 2
- 230000001939 inductive effect Effects 0.000 description 2
- 239000003112 inhibitor Substances 0.000 description 2
- 230000005764 inhibitory process Effects 0.000 description 2
- 238000007913 intrathecal administration Methods 0.000 description 2
- 238000010253 intravenous injection Methods 0.000 description 2
- 235000015110 jellies Nutrition 0.000 description 2
- 239000008274 jelly Substances 0.000 description 2
- 230000003902 lesion Effects 0.000 description 2
- 150000002632 lipids Chemical class 0.000 description 2
- 210000004185 liver Anatomy 0.000 description 2
- 210000004698 lymphocyte Anatomy 0.000 description 2
- 239000006166 lysate Substances 0.000 description 2
- 230000007246 mechanism Effects 0.000 description 2
- 210000003593 megakaryocyte Anatomy 0.000 description 2
- 230000005012 migration Effects 0.000 description 2
- 238000013508 migration Methods 0.000 description 2
- 235000013336 milk Nutrition 0.000 description 2
- 239000008267 milk Substances 0.000 description 2
- 210000004080 milk Anatomy 0.000 description 2
- 210000005087 mononuclear cell Anatomy 0.000 description 2
- 208000025113 myeloid leukemia Diseases 0.000 description 2
- 210000000107 myocyte Anatomy 0.000 description 2
- 230000008693 nausea Effects 0.000 description 2
- 229940029345 neupogen Drugs 0.000 description 2
- 230000001537 neural effect Effects 0.000 description 2
- 210000001178 neural stem cell Anatomy 0.000 description 2
- 230000001722 neurochemical effect Effects 0.000 description 2
- 210000004967 non-hematopoietic stem cell Anatomy 0.000 description 2
- 125000003729 nucleotide group Chemical group 0.000 description 2
- 230000000399 orthopedic effect Effects 0.000 description 2
- 238000007911 parenteral administration Methods 0.000 description 2
- 230000036961 partial effect Effects 0.000 description 2
- 210000004976 peripheral blood cell Anatomy 0.000 description 2
- 230000003836 peripheral circulation Effects 0.000 description 2
- 239000002831 pharmacologic agent Substances 0.000 description 2
- 210000001778 pluripotent stem cell Anatomy 0.000 description 2
- 102000054765 polymorphisms of proteins Human genes 0.000 description 2
- 239000000843 powder Substances 0.000 description 2
- 210000001236 prokaryotic cell Anatomy 0.000 description 2
- 230000002062 proliferating effect Effects 0.000 description 2
- 230000001737 promoting effect Effects 0.000 description 2
- 238000004445 quantitative analysis Methods 0.000 description 2
- 238000010188 recombinant method Methods 0.000 description 2
- 238000005057 refrigeration Methods 0.000 description 2
- 230000010076 replication Effects 0.000 description 2
- 230000002441 reversible effect Effects 0.000 description 2
- 230000036573 scar formation Effects 0.000 description 2
- 230000028327 secretion Effects 0.000 description 2
- 238000004062 sedimentation Methods 0.000 description 2
- 230000019491 signal transduction Effects 0.000 description 2
- 210000003491 skin Anatomy 0.000 description 2
- 210000004872 soft tissue Anatomy 0.000 description 2
- 239000002904 solvent Substances 0.000 description 2
- 230000002269 spontaneous effect Effects 0.000 description 2
- 238000010186 staining Methods 0.000 description 2
- 238000007619 statistical method Methods 0.000 description 2
- 239000008174 sterile solution Substances 0.000 description 2
- 230000035882 stress Effects 0.000 description 2
- 238000010254 subcutaneous injection Methods 0.000 description 2
- 235000000346 sugar Nutrition 0.000 description 2
- 150000008163 sugars Chemical class 0.000 description 2
- 230000008093 supporting effect Effects 0.000 description 2
- 238000001356 surgical procedure Methods 0.000 description 2
- 238000003786 synthesis reaction Methods 0.000 description 2
- 239000006188 syrup Substances 0.000 description 2
- 235000020357 syrup Nutrition 0.000 description 2
- 231100001274 therapeutic index Toxicity 0.000 description 2
- 230000000451 tissue damage Effects 0.000 description 2
- 231100000827 tissue damage Toxicity 0.000 description 2
- 230000017423 tissue regeneration Effects 0.000 description 2
- 231100000331 toxic Toxicity 0.000 description 2
- 230000002588 toxic effect Effects 0.000 description 2
- 230000001988 toxicity Effects 0.000 description 2
- 231100000419 toxicity Toxicity 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- 239000012588 trypsin Substances 0.000 description 2
- 210000004881 tumor cell Anatomy 0.000 description 2
- 210000003954 umbilical cord Anatomy 0.000 description 2
- 239000013598 vector Substances 0.000 description 2
- 239000013603 viral vector Substances 0.000 description 2
- 239000011782 vitamin Substances 0.000 description 2
- 229940088594 vitamin Drugs 0.000 description 2
- 229930003231 vitamin Natural products 0.000 description 2
- 235000013343 vitamin Nutrition 0.000 description 2
- LGLVVVCSQBZONM-HCCLCSBVSA-N (2r)-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-acetamido-5-(diaminomethylideneamino)pentanoyl]amino]-5-(diaminomethylideneamino)pentanoyl]amino]-5-(diaminomethylideneamino)pentanoyl]amino]-5-(diaminomethylideneamino)pentano Chemical compound NC(N)=NCCC[C@@H](NC(=O)C)C(=O)N[C@H](CCCN=C(N)N)C(=O)N[C@H](CCCN=C(N)N)C(=O)N[C@H](CCCN=C(N)N)C(=O)N[C@H](CCCN=C(N)N)C(=O)N[C@H](CCCN=C(N)N)C(=O)N[C@H](CCCN=C(N)N)C(=O)N[C@H](CCCN=C(N)N)C(=O)N[C@H](CCCN=C(N)N)C(N)=O LGLVVVCSQBZONM-HCCLCSBVSA-N 0.000 description 1
- CPKVUHPKYQGHMW-UHFFFAOYSA-N 1-ethenylpyrrolidin-2-one;molecular iodine Chemical compound II.C=CN1CCCC1=O CPKVUHPKYQGHMW-UHFFFAOYSA-N 0.000 description 1
- GZCWLCBFPRFLKL-UHFFFAOYSA-N 1-prop-2-ynoxypropan-2-ol Chemical compound CC(O)COCC#C GZCWLCBFPRFLKL-UHFFFAOYSA-N 0.000 description 1
- PUAQLLVFLMYYJJ-UHFFFAOYSA-N 2-aminopropiophenone Chemical compound CC(N)C(=O)C1=CC=CC=C1 PUAQLLVFLMYYJJ-UHFFFAOYSA-N 0.000 description 1
- 208000030507 AIDS Diseases 0.000 description 1
- 108010051457 Acid Phosphatase Proteins 0.000 description 1
- 102000013563 Acid Phosphatase Human genes 0.000 description 1
- 241000251468 Actinopterygii Species 0.000 description 1
- 108020004774 Alkaline Phosphatase Proteins 0.000 description 1
- 102000002260 Alkaline Phosphatase Human genes 0.000 description 1
- 244000144730 Amygdalus persica Species 0.000 description 1
- 101100100054 Arabidopsis thaliana TPLATE gene Proteins 0.000 description 1
- 241000531806 Aramus guarauna Species 0.000 description 1
- BHELIUBJHYAEDK-OAIUPTLZSA-N Aspoxicillin Chemical compound C1([C@H](C(=O)N[C@@H]2C(N3[C@H](C(C)(C)S[C@@H]32)C(O)=O)=O)NC(=O)[C@H](N)CC(=O)NC)=CC=C(O)C=C1 BHELIUBJHYAEDK-OAIUPTLZSA-N 0.000 description 1
- 208000023275 Autoimmune disease Diseases 0.000 description 1
- 208000013883 Blast injury Diseases 0.000 description 1
- 108010007726 Bone Morphogenetic Proteins Proteins 0.000 description 1
- 102000007350 Bone Morphogenetic Proteins Human genes 0.000 description 1
- 102100022525 Bone morphogenetic protein 6 Human genes 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- 206010006187 Breast cancer Diseases 0.000 description 1
- 208000026310 Breast neoplasm Diseases 0.000 description 1
- 210000003771 C cell Anatomy 0.000 description 1
- 108010061299 CXCR4 Receptors Proteins 0.000 description 1
- 102000012000 CXCR4 Receptors Human genes 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 208000024172 Cardiovascular disease Diseases 0.000 description 1
- 208000024699 Chagas disease Diseases 0.000 description 1
- 108010077544 Chromatin Proteins 0.000 description 1
- 108090000317 Chymotrypsin Proteins 0.000 description 1
- 208000032544 Cicatrix Diseases 0.000 description 1
- 206010053567 Coagulopathies Diseases 0.000 description 1
- 108091026890 Coding region Proteins 0.000 description 1
- 102000008186 Collagen Human genes 0.000 description 1
- 108010035532 Collagen Proteins 0.000 description 1
- 102000007644 Colony-Stimulating Factors Human genes 0.000 description 1
- 108010071942 Colony-Stimulating Factors Proteins 0.000 description 1
- 241000938605 Crocodylia Species 0.000 description 1
- 108010053770 Deoxyribonucleases Proteins 0.000 description 1
- 102000016911 Deoxyribonucleases Human genes 0.000 description 1
- 238000009007 Diagnostic Kit Methods 0.000 description 1
- 241000196324 Embryophyta Species 0.000 description 1
- 108010022894 Euchromatin Proteins 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 108010080379 Fibrin Tissue Adhesive Proteins 0.000 description 1
- 102100037362 Fibronectin Human genes 0.000 description 1
- 108010067306 Fibronectins Proteins 0.000 description 1
- 238000012413 Fluorescence activated cell sorting analysis Methods 0.000 description 1
- 241000233866 Fungi Species 0.000 description 1
- 108060005986 Granzyme Proteins 0.000 description 1
- 102000001398 Granzyme Human genes 0.000 description 1
- 206010018910 Haemolysis Diseases 0.000 description 1
- 239000012981 Hank's balanced salt solution Substances 0.000 description 1
- 206010019237 Healing abnormal Diseases 0.000 description 1
- 206010019280 Heart failures Diseases 0.000 description 1
- 208000002250 Hematologic Neoplasms Diseases 0.000 description 1
- 101000899390 Homo sapiens Bone morphogenetic protein 6 Proteins 0.000 description 1
- 101000746373 Homo sapiens Granulocyte-macrophage colony-stimulating factor Proteins 0.000 description 1
- 101000599852 Homo sapiens Intercellular adhesion molecule 1 Proteins 0.000 description 1
- 101000835093 Homo sapiens Transferrin receptor protein 1 Proteins 0.000 description 1
- 241000713772 Human immunodeficiency virus 1 Species 0.000 description 1
- 206010020751 Hypersensitivity Diseases 0.000 description 1
- 206010061218 Inflammation Diseases 0.000 description 1
- 102100037877 Intercellular adhesion molecule 1 Human genes 0.000 description 1
- 102100021592 Interleukin-7 Human genes 0.000 description 1
- 108010002586 Interleukin-7 Proteins 0.000 description 1
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 1
- FEWJPZIEWOKRBE-JCYAYHJZSA-L L-tartrate(2-) Chemical compound [O-]C(=O)[C@H](O)[C@@H](O)C([O-])=O FEWJPZIEWOKRBE-JCYAYHJZSA-L 0.000 description 1
- 102000001846 Low Density Lipoprotein Receptor-Related Protein-2 Human genes 0.000 description 1
- 108010015372 Low Density Lipoprotein Receptor-Related Protein-2 Proteins 0.000 description 1
- 102000009571 Macrophage Inflammatory Proteins Human genes 0.000 description 1
- 108010009474 Macrophage Inflammatory Proteins Proteins 0.000 description 1
- 102000018697 Membrane Proteins Human genes 0.000 description 1
- 108010052285 Membrane Proteins Proteins 0.000 description 1
- 208000034578 Multiple myelomas Diseases 0.000 description 1
- 241000699666 Mus <mouse, genus> Species 0.000 description 1
- 241000699660 Mus musculus Species 0.000 description 1
- 101100335081 Mus musculus Flt3 gene Proteins 0.000 description 1
- 108010021466 Mutant Proteins Proteins 0.000 description 1
- 102000008300 Mutant Proteins Human genes 0.000 description 1
- 241000204031 Mycoplasma Species 0.000 description 1
- 201000003793 Myelodysplastic syndrome Diseases 0.000 description 1
- 108010056642 N-alpha-acetyl-nona-D-arginine amide acetate Proteins 0.000 description 1
- CHJJGSNFBQVOTG-UHFFFAOYSA-N N-methyl-guanidine Natural products CNC(N)=N CHJJGSNFBQVOTG-UHFFFAOYSA-N 0.000 description 1
- 229920002274 Nalgene Polymers 0.000 description 1
- 206010029260 Neuroblastoma Diseases 0.000 description 1
- 238000000636 Northern blotting Methods 0.000 description 1
- 108010038807 Oligopeptides Proteins 0.000 description 1
- 102000015636 Oligopeptides Human genes 0.000 description 1
- 108700020796 Oncogene Proteins 0.000 description 1
- 102000043276 Oncogene Human genes 0.000 description 1
- 241000366596 Osiris Species 0.000 description 1
- 208000002193 Pain Diseases 0.000 description 1
- 206010057249 Phagocytosis Diseases 0.000 description 1
- 206010035226 Plasma cell myeloma Diseases 0.000 description 1
- 229920003171 Poly (ethylene oxide) Polymers 0.000 description 1
- 241000219000 Populus Species 0.000 description 1
- 101710150336 Protein Rex Proteins 0.000 description 1
- 102000016971 Proto-Oncogene Proteins c-kit Human genes 0.000 description 1
- 108010014608 Proto-Oncogene Proteins c-kit Proteins 0.000 description 1
- 235000006040 Prunus persica var persica Nutrition 0.000 description 1
- 239000012980 RPMI-1640 medium Substances 0.000 description 1
- 208000006265 Renal cell carcinoma Diseases 0.000 description 1
- 208000017442 Retinal disease Diseases 0.000 description 1
- 206010038923 Retinopathy Diseases 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 1
- 206010061363 Skeletal injury Diseases 0.000 description 1
- 208000002152 Splenic rupture Diseases 0.000 description 1
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 1
- 229930006000 Sucrose Natural products 0.000 description 1
- 108010045306 T134 peptide Proteins 0.000 description 1
- 108010025037 T140 peptide Proteins 0.000 description 1
- 108700012920 TNF Proteins 0.000 description 1
- 102100038126 Tenascin Human genes 0.000 description 1
- 206010043276 Teratoma Diseases 0.000 description 1
- 108090000190 Thrombin Proteins 0.000 description 1
- 102100026144 Transferrin receptor protein 1 Human genes 0.000 description 1
- 108700019146 Transgenes Proteins 0.000 description 1
- GLNADSQYFUSGOU-GPTZEZBUSA-J Trypan blue Chemical compound [Na+].[Na+].[Na+].[Na+].C1=C(S([O-])(=O)=O)C=C2C=C(S([O-])(=O)=O)C(/N=N/C3=CC=C(C=C3C)C=3C=C(C(=CC=3)\N=N\C=3C(=CC4=CC(=CC(N)=C4C=3O)S([O-])(=O)=O)S([O-])(=O)=O)C)=C(O)C2=C1N GLNADSQYFUSGOU-GPTZEZBUSA-J 0.000 description 1
- 102000005789 Vascular Endothelial Growth Factors Human genes 0.000 description 1
- 108010019530 Vascular Endothelial Growth Factors Proteins 0.000 description 1
- 230000005856 abnormality Effects 0.000 description 1
- 230000002745 absorbent Effects 0.000 description 1
- 239000002250 absorbent Substances 0.000 description 1
- 239000002253 acid Substances 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 230000003679 aging effect Effects 0.000 description 1
- 230000029936 alkylation Effects 0.000 description 1
- 238000005804 alkylation reaction Methods 0.000 description 1
- 208000030961 allergic reaction Diseases 0.000 description 1
- 125000003277 amino group Chemical group 0.000 description 1
- 239000003708 ampul Substances 0.000 description 1
- 239000002870 angiogenesis inducing agent Substances 0.000 description 1
- 210000004102 animal cell Anatomy 0.000 description 1
- 229940088710 antibiotic agent Drugs 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- 239000007864 aqueous solution Substances 0.000 description 1
- 238000005899 aromatization reaction Methods 0.000 description 1
- 125000003118 aryl group Chemical group 0.000 description 1
- 230000003416 augmentation Effects 0.000 description 1
- 230000004888 barrier function Effects 0.000 description 1
- 238000010923 batch production Methods 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 229940064804 betadine Drugs 0.000 description 1
- 210000001109 blastomere Anatomy 0.000 description 1
- 238000004820 blood count Methods 0.000 description 1
- 229940125691 blood product Drugs 0.000 description 1
- 210000004204 blood vessel Anatomy 0.000 description 1
- 230000037396 body weight Effects 0.000 description 1
- 210000002449 bone cell Anatomy 0.000 description 1
- 238000009583 bone marrow aspiration Methods 0.000 description 1
- 229940112869 bone morphogenetic protein Drugs 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- 239000007853 buffer solution Substances 0.000 description 1
- 239000007975 buffered saline Substances 0.000 description 1
- 108010054191 butyrylesterase Proteins 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 210000003321 cartilage cell Anatomy 0.000 description 1
- 238000013354 cell banking Methods 0.000 description 1
- 230000022131 cell cycle Effects 0.000 description 1
- 230000011712 cell development Effects 0.000 description 1
- 230000032823 cell division Effects 0.000 description 1
- 230000010261 cell growth Effects 0.000 description 1
- 230000006037 cell lysis Effects 0.000 description 1
- 230000008668 cellular reprogramming Effects 0.000 description 1
- 239000001913 cellulose Substances 0.000 description 1
- 229920002678 cellulose Polymers 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 238000012512 characterization method Methods 0.000 description 1
- 239000013043 chemical agent Substances 0.000 description 1
- 238000007385 chemical modification Methods 0.000 description 1
- 239000002975 chemoattractant Substances 0.000 description 1
- 210000003483 chromatin Anatomy 0.000 description 1
- 208000022371 chronic pain syndrome Diseases 0.000 description 1
- 229960002376 chymotrypsin Drugs 0.000 description 1
- 210000003040 circulating cell Anatomy 0.000 description 1
- 239000007979 citrate buffer Substances 0.000 description 1
- 238000005352 clarification Methods 0.000 description 1
- 210000003109 clavicle Anatomy 0.000 description 1
- 230000035602 clotting Effects 0.000 description 1
- 230000004186 co-expression Effects 0.000 description 1
- 239000000701 coagulant Substances 0.000 description 1
- 239000011248 coating agent Substances 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 229920001436 collagen Polymers 0.000 description 1
- 239000000515 collagen sponge Substances 0.000 description 1
- 230000001332 colony forming effect Effects 0.000 description 1
- 238000010835 comparative analysis Methods 0.000 description 1
- 230000000052 comparative effect Effects 0.000 description 1
- 230000000295 complement effect Effects 0.000 description 1
- 239000008139 complexing agent Substances 0.000 description 1
- 238000002591 computed tomography Methods 0.000 description 1
- 238000004590 computer program Methods 0.000 description 1
- 230000009073 conformational modification Effects 0.000 description 1
- 239000002872 contrast media Substances 0.000 description 1
- 230000008878 coupling Effects 0.000 description 1
- 238000010168 coupling process Methods 0.000 description 1
- 238000005859 coupling reaction Methods 0.000 description 1
- 210000000695 crystalline len Anatomy 0.000 description 1
- 210000004748 cultured cell Anatomy 0.000 description 1
- 208000035250 cutaneous malignant susceptibility to 1 melanoma Diseases 0.000 description 1
- 230000009089 cytolysis Effects 0.000 description 1
- 238000004163 cytometry Methods 0.000 description 1
- 210000000805 cytoplasm Anatomy 0.000 description 1
- 230000001472 cytotoxic effect Effects 0.000 description 1
- 230000002354 daily effect Effects 0.000 description 1
- 238000007405 data analysis Methods 0.000 description 1
- 230000032459 dedifferentiation Effects 0.000 description 1
- 230000008260 defense mechanism Effects 0.000 description 1
- 238000000151 deposition Methods 0.000 description 1
- 210000004207 dermis Anatomy 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 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 1
- 239000008121 dextrose Substances 0.000 description 1
- 206010012601 diabetes mellitus Diseases 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- 238000002405 diagnostic procedure Methods 0.000 description 1
- 238000010586 diagram Methods 0.000 description 1
- 238000009792 diffusion process Methods 0.000 description 1
- AYXBAIULRDEVAS-UHFFFAOYSA-N dimethyl-[[4-[[3-(4-methylphenyl)-8,9-dihydro-7h-benzo[7]annulene-6-carbonyl]amino]phenyl]methyl]-(oxan-4-yl)azanium;iodide Chemical compound [I-].C1=CC(C)=CC=C1C1=CC=C(CCCC(=C2)C(=O)NC=3C=CC(C[N+](C)(C)C4CCOCC4)=CC=3)C2=C1 AYXBAIULRDEVAS-UHFFFAOYSA-N 0.000 description 1
- SWSQBOPZIKWTGO-UHFFFAOYSA-N dimethylaminoamidine Natural products CN(C)C(N)=N SWSQBOPZIKWTGO-UHFFFAOYSA-N 0.000 description 1
- 238000005553 drilling Methods 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 230000009977 dual effect Effects 0.000 description 1
- 238000004043 dyeing Methods 0.000 description 1
- 230000004064 dysfunction Effects 0.000 description 1
- 230000002526 effect on cardiovascular system Effects 0.000 description 1
- 230000002500 effect on skin Effects 0.000 description 1
- 235000013601 eggs Nutrition 0.000 description 1
- 210000002308 embryonic cell Anatomy 0.000 description 1
- 238000002674 endoscopic surgery Methods 0.000 description 1
- 210000003038 endothelium Anatomy 0.000 description 1
- 230000002255 enzymatic effect Effects 0.000 description 1
- 210000000981 epithelium Anatomy 0.000 description 1
- 150000002148 esters Chemical class 0.000 description 1
- 210000000632 euchromatin Anatomy 0.000 description 1
- 230000003203 everyday effect Effects 0.000 description 1
- 238000011124 ex vivo culture Methods 0.000 description 1
- 230000005284 excitation Effects 0.000 description 1
- 238000002474 experimental method Methods 0.000 description 1
- 239000000284 extract Substances 0.000 description 1
- 210000003414 extremity Anatomy 0.000 description 1
- 208000030533 eye disease Diseases 0.000 description 1
- 210000004709 eyebrow Anatomy 0.000 description 1
- 210000002436 femur neck Anatomy 0.000 description 1
- 239000012894 fetal calf serum Substances 0.000 description 1
- 230000001605 fetal effect Effects 0.000 description 1
- 210000003754 fetus Anatomy 0.000 description 1
- 210000002950 fibroblast Anatomy 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 238000011049 filling Methods 0.000 description 1
- 238000001914 filtration Methods 0.000 description 1
- 108700014844 flt3 ligand Proteins 0.000 description 1
- MHMNJMPURVTYEJ-UHFFFAOYSA-N fluorescein-5-isothiocyanate Chemical compound O1C(=O)C2=CC(N=C=S)=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 MHMNJMPURVTYEJ-UHFFFAOYSA-N 0.000 description 1
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 1
- 239000007850 fluorescent dye Substances 0.000 description 1
- 238000005194 fractionation Methods 0.000 description 1
- 230000005714 functional activity Effects 0.000 description 1
- 230000002538 fungal effect Effects 0.000 description 1
- 238000002309 gasification Methods 0.000 description 1
- 230000002496 gastric effect Effects 0.000 description 1
- 210000001035 gastrointestinal tract Anatomy 0.000 description 1
- 238000010363 gene targeting Methods 0.000 description 1
- 238000007429 general method Methods 0.000 description 1
- 230000002518 glial effect Effects 0.000 description 1
- 150000004676 glycans Chemical class 0.000 description 1
- 239000008187 granular material Substances 0.000 description 1
- 210000003128 head Anatomy 0.000 description 1
- 230000005802 health problem Effects 0.000 description 1
- 230000009033 hematopoietic malignancy Effects 0.000 description 1
- 230000008588 hemolysis Effects 0.000 description 1
- 230000011132 hemopoiesis Effects 0.000 description 1
- 210000003897 hepatic stem cell Anatomy 0.000 description 1
- 125000000623 heterocyclic group Chemical group 0.000 description 1
- 229920006158 high molecular weight polymer Polymers 0.000 description 1
- 230000001744 histochemical effect Effects 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- 229940088597 hormone Drugs 0.000 description 1
- 238000001794 hormone therapy Methods 0.000 description 1
- 102000046157 human CSF2 Human genes 0.000 description 1
- 210000005260 human cell Anatomy 0.000 description 1
- 238000005286 illumination Methods 0.000 description 1
- 238000010166 immunofluorescence Methods 0.000 description 1
- 230000002163 immunogen Effects 0.000 description 1
- 238000011532 immunohistochemical staining Methods 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 229910052738 indium Inorganic materials 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 239000011261 inert gas Substances 0.000 description 1
- 230000008595 infiltration Effects 0.000 description 1
- 238000001764 infiltration Methods 0.000 description 1
- 230000004054 inflammatory process Effects 0.000 description 1
- 239000003978 infusion fluid Substances 0.000 description 1
- 239000004615 ingredient Substances 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 229940102223 injectable solution Drugs 0.000 description 1
- 238000011081 inoculation Methods 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 229940100994 interleukin-7 Drugs 0.000 description 1
- 230000000968 intestinal effect Effects 0.000 description 1
- 238000001361 intraarterial administration Methods 0.000 description 1
- 230000003834 intracellular effect Effects 0.000 description 1
- 239000007928 intraperitoneal injection Substances 0.000 description 1
- 238000007914 intraventricular administration Methods 0.000 description 1
- NBQNWMBBSKPBAY-UHFFFAOYSA-N iodixanol Chemical compound IC=1C(C(=O)NCC(O)CO)=C(I)C(C(=O)NCC(O)CO)=C(I)C=1N(C(=O)C)CC(O)CN(C(C)=O)C1=C(I)C(C(=O)NCC(O)CO)=C(I)C(C(=O)NCC(O)CO)=C1I NBQNWMBBSKPBAY-UHFFFAOYSA-N 0.000 description 1
- 229960004359 iodixanol Drugs 0.000 description 1
- 230000007794 irritation Effects 0.000 description 1
- 208000028867 ischemia Diseases 0.000 description 1
- 230000000302 ischemic effect Effects 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 108010013469 leridistim Proteins 0.000 description 1
- 229950003059 leridistim Drugs 0.000 description 1
- 239000002502 liposome Substances 0.000 description 1
- 238000004811 liquid chromatography Methods 0.000 description 1
- 239000007791 liquid phase Substances 0.000 description 1
- 230000033001 locomotion Effects 0.000 description 1
- 210000003141 lower extremity Anatomy 0.000 description 1
- 239000007937 lozenge Substances 0.000 description 1
- 210000004072 lung Anatomy 0.000 description 1
- 210000003738 lymphoid progenitor cell Anatomy 0.000 description 1
- 239000003120 macrolide antibiotic agent Substances 0.000 description 1
- 208000002780 macular degeneration Diseases 0.000 description 1
- 210000001161 mammalian embryo Anatomy 0.000 description 1
- 210000004373 mandible Anatomy 0.000 description 1
- 230000000873 masking effect Effects 0.000 description 1
- 238000004949 mass spectrometry Methods 0.000 description 1
- 230000005541 medical transmission Effects 0.000 description 1
- 238000002483 medication Methods 0.000 description 1
- 201000001441 melanoma Diseases 0.000 description 1
- QSHDDOUJBYECFT-UHFFFAOYSA-N mercury Chemical compound [Hg] QSHDDOUJBYECFT-UHFFFAOYSA-N 0.000 description 1
- 229910052753 mercury Inorganic materials 0.000 description 1
- 230000004060 metabolic process Effects 0.000 description 1
- 229930182817 methionine Natural products 0.000 description 1
- 239000011325 microbead Substances 0.000 description 1
- 150000007522 mineralic acids Chemical class 0.000 description 1
- 108010032806 molgramostim Proteins 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 239000000178 monomer Substances 0.000 description 1
- PJUIMOJAAPLTRJ-UHFFFAOYSA-N monothioglycerol Chemical compound OCC(O)CS PJUIMOJAAPLTRJ-UHFFFAOYSA-N 0.000 description 1
- 229940074923 mozobil Drugs 0.000 description 1
- 210000004877 mucosa Anatomy 0.000 description 1
- 210000002464 muscle smooth vascular Anatomy 0.000 description 1
- 210000001665 muscle stem cell Anatomy 0.000 description 1
- 206010028537 myelofibrosis Diseases 0.000 description 1
- 230000002107 myocardial effect Effects 0.000 description 1
- 208000010125 myocardial infarction Diseases 0.000 description 1
- 210000004165 myocardium Anatomy 0.000 description 1
- 210000000822 natural killer cell Anatomy 0.000 description 1
- 230000001338 necrotic effect Effects 0.000 description 1
- 239000013642 negative control Substances 0.000 description 1
- 210000000276 neural tube Anatomy 0.000 description 1
- 210000004498 neuroglial cell Anatomy 0.000 description 1
- 230000007935 neutral effect Effects 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 231100000956 nontoxicity Toxicity 0.000 description 1
- 239000002773 nucleotide Substances 0.000 description 1
- 238000011580 nude mouse model Methods 0.000 description 1
- 235000015097 nutrients Nutrition 0.000 description 1
- 210000004248 oligodendroglia Anatomy 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 230000008520 organization Effects 0.000 description 1
- 150000002902 organometallic compounds Chemical class 0.000 description 1
- 230000002188 osteogenic effect Effects 0.000 description 1
- 230000003647 oxidation Effects 0.000 description 1
- 238000007254 oxidation reaction Methods 0.000 description 1
- 239000012188 paraffin wax Substances 0.000 description 1
- 244000045947 parasite Species 0.000 description 1
- 244000052769 pathogen Species 0.000 description 1
- 230000001717 pathogenic effect Effects 0.000 description 1
- 230000008807 pathological lesion Effects 0.000 description 1
- HQQSBEDKMRHYME-UHFFFAOYSA-N pefloxacin mesylate Chemical compound [H+].CS([O-])(=O)=O.C1=C2N(CC)C=C(C(O)=O)C(=O)C2=CC(F)=C1N1CCN(C)CC1 HQQSBEDKMRHYME-UHFFFAOYSA-N 0.000 description 1
- 108010044644 pegfilgrastim Proteins 0.000 description 1
- 229960001373 pegfilgrastim Drugs 0.000 description 1
- 239000008188 pellet Substances 0.000 description 1
- 239000000816 peptidomimetic Substances 0.000 description 1
- 230000010412 perfusion Effects 0.000 description 1
- 210000001428 peripheral nervous system Anatomy 0.000 description 1
- 230000008782 phagocytosis Effects 0.000 description 1
- 239000008194 pharmaceutical composition Substances 0.000 description 1
- 239000012071 phase Substances 0.000 description 1
- 230000035790 physiological processes and functions Effects 0.000 description 1
- 239000000902 placebo Substances 0.000 description 1
- 229940068196 placebo Drugs 0.000 description 1
- 239000004033 plastic Substances 0.000 description 1
- 229920003023 plastic Polymers 0.000 description 1
- 230000004983 pleiotropic effect Effects 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 229920000642 polymer Polymers 0.000 description 1
- 229920001184 polypeptide Polymers 0.000 description 1
- 229920001282 polysaccharide Polymers 0.000 description 1
- 239000005017 polysaccharide Substances 0.000 description 1
- 239000013641 positive control Substances 0.000 description 1
- 230000004481 post-translational protein modification Effects 0.000 description 1
- 230000002980 postoperative effect Effects 0.000 description 1
- 238000004321 preservation Methods 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 230000003449 preventive effect Effects 0.000 description 1
- 230000037452 priming Effects 0.000 description 1
- 238000004393 prognosis Methods 0.000 description 1
- 238000011321 prophylaxis Methods 0.000 description 1
- 238000001814 protein method Methods 0.000 description 1
- 238000000575 proteomic method Methods 0.000 description 1
- 238000003908 quality control method Methods 0.000 description 1
- 230000005855 radiation Effects 0.000 description 1
- 238000001959 radiotherapy Methods 0.000 description 1
- 239000013643 reference control Substances 0.000 description 1
- 239000013074 reference sample Substances 0.000 description 1
- 230000008929 regeneration Effects 0.000 description 1
- 238000011069 regeneration method Methods 0.000 description 1
- 238000007634 remodeling Methods 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 230000000241 respiratory effect Effects 0.000 description 1
- 108091008146 restriction endonucleases Proteins 0.000 description 1
- 230000000717 retained effect Effects 0.000 description 1
- 210000001525 retina Anatomy 0.000 description 1
- 238000012340 reverse transcriptase PCR Methods 0.000 description 1
- 238000012552 review Methods 0.000 description 1
- 231100000241 scar Toxicity 0.000 description 1
- 230000037387 scars Effects 0.000 description 1
- 238000005204 segregation Methods 0.000 description 1
- 239000002453 shampoo Substances 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- 239000002356 single layer Substances 0.000 description 1
- 210000002027 skeletal muscle Anatomy 0.000 description 1
- 159000000000 sodium salts Chemical class 0.000 description 1
- 210000001988 somatic stem cell Anatomy 0.000 description 1
- 238000010183 spectrum analysis Methods 0.000 description 1
- 239000007921 spray Substances 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 210000004500 stellate cell Anatomy 0.000 description 1
- 210000000603 stem cell niche Anatomy 0.000 description 1
- 238000009168 stem cell therapy Methods 0.000 description 1
- 238000009580 stem-cell therapy Methods 0.000 description 1
- 150000003431 steroids Chemical class 0.000 description 1
- 230000000638 stimulation Effects 0.000 description 1
- 210000002784 stomach Anatomy 0.000 description 1
- 210000003699 striated muscle Anatomy 0.000 description 1
- 125000000547 substituted alkyl group Chemical group 0.000 description 1
- 239000005720 sucrose Substances 0.000 description 1
- 239000013589 supplement Substances 0.000 description 1
- 230000001502 supplementing effect Effects 0.000 description 1
- 239000000829 suppository Substances 0.000 description 1
- 210000005222 synovial tissue Anatomy 0.000 description 1
- 238000007910 systemic administration Methods 0.000 description 1
- 230000008685 targeting Effects 0.000 description 1
- 229940095064 tartrate Drugs 0.000 description 1
- 210000002435 tendon Anatomy 0.000 description 1
- 229940124597 therapeutic agent Drugs 0.000 description 1
- 229940035024 thioglycerol Drugs 0.000 description 1
- 229960004072 thrombin Drugs 0.000 description 1
- 229940033618 tisseel Drugs 0.000 description 1
- 238000011200 topical administration Methods 0.000 description 1
- 238000011830 transgenic mouse model Methods 0.000 description 1
- 238000013519 translation Methods 0.000 description 1
- 238000011269 treatment regimen Methods 0.000 description 1
- 230000002476 tumorcidal effect Effects 0.000 description 1
- 230000000381 tumorigenic effect Effects 0.000 description 1
- 210000000623 ulna Anatomy 0.000 description 1
- 210000001364 upper extremity Anatomy 0.000 description 1
- 210000000626 ureter Anatomy 0.000 description 1
- 210000003708 urethra Anatomy 0.000 description 1
- 210000003932 urinary bladder Anatomy 0.000 description 1
- 230000002485 urinary effect Effects 0.000 description 1
- 201000002282 venous insufficiency Diseases 0.000 description 1
- 210000002845 virion Anatomy 0.000 description 1
- 230000000007 visual effect Effects 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 230000003442 weekly effect Effects 0.000 description 1
- 230000037314 wound repair Effects 0.000 description 1
- 210000000707 wrist Anatomy 0.000 description 1
- DGVVWUTYPXICAM-UHFFFAOYSA-N β‐Mercaptoethanol Chemical compound OCCS DGVVWUTYPXICAM-UHFFFAOYSA-N 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/12—Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
- A61K35/28—Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/19—Cytokines; Lymphokines; Interferons
- A61K38/193—Colony stimulating factors [CSF]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/19—Cytokines; Lymphokines; Interferons
- A61K38/20—Interleukins [IL]
-
- 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
- C12N5/0662—Stem cells
- C12N5/0665—Blood-borne mesenchymal stem cells, e.g. from umbilical cord blood
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Zoology (AREA)
- Chemical & Material Sciences (AREA)
- Developmental Biology & Embryology (AREA)
- Immunology (AREA)
- General Health & Medical Sciences (AREA)
- Biomedical Technology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Biotechnology (AREA)
- Medicinal Chemistry (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Cell Biology (AREA)
- Animal Behavior & Ethology (AREA)
- Epidemiology (AREA)
- Pharmacology & Pharmacy (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Gastroenterology & Hepatology (AREA)
- Hematology (AREA)
- Organic Chemistry (AREA)
- Genetics & Genomics (AREA)
- Wood Science & Technology (AREA)
- Rheumatology (AREA)
- Microbiology (AREA)
- General Engineering & Computer Science (AREA)
- Biochemistry (AREA)
- Virology (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
Abstract
Description
201130977 六、發明說明: 【發明所屬之技術領域】 本發明係關於幹細胞,特定言之,用於自經調動之周邊 血中分離人類間質幹細胞(MSC)之最佳時序的方法及組合 物’及分離之MSC隨後諸如超低溫保存及/或用於治療有 需要之個體的用途,諸如在自體再生醫學中》 【先前技術】 間質幹細胞(MSC)為在細胞療法領域具有重大效用之多 能細胞。MSC可分化成至少3個下游間質細胞譜系(亦即骨 母細胞、軟骨母細胞及脂肪細胞)。迄今為止,尚未鑑別 出獨特MSC標記。因而,利用形態及功能標準鑑別此等細 胞。參見Horwitz E等人,「Clarification of the nomenclature for MSC: the International Society for Cellular Therapy position statement,」Cytotherapy 7:393 (2005);及201130977 VI. Description of the Invention: [Technical Field of the Invention] The present invention relates to stem cells, in particular, methods and compositions for separating the optimal timing of human mesenchymal stem cells (MSCs) from mobilized peripheral blood. And isolated MSCs are then used, such as cryopreservation and/or for the treatment of individuals in need thereof, such as in autologous regenerative medicine. [Prior Art] Mesenchymal stem cells (MSC) are versatile in the field of cell therapy. cell. MSCs can differentiate into at least three downstream mesenchymal cell lineages (i.e., osteoblasts, chondroblasts, and adipocytes). To date, unique MSC markers have not been identified. Thus, these cells are identified using morphological and functional criteria. See Horwitz E et al., "Clarification of the nomenclature for MSC: the International Society for Cellular Therapy position statement," Cytotherapy 7:393 (2005);
Dominici M 等人,「Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement,」 Cytotherapy 8:315 (2006)。因為MSC可分化成許多細胞類 型,所以此項技術涵蓋供基於細胞之療法、再生醫學及重 建醫學用的分化MSC之方法。 製備MSC組合物的典型方法係由根據MSC之塑膠黏著性 質分離MSC及隨後對其培養擴增所組成。當在胎兒發育期 間自人類體液分離MSC時,此方法相對容易。然而,自成 人體液獲得MSC要複雜得多且可再現性要低得多。 152716.doc 201130977 通常,自成人骨髓、脂肪、軟骨及肌肉分離MSC。 Pittenger F 等人,「Multilineage potential of adult human mesenchymal stem cells,」Science 284:143-147 (1999); Zuk P等人,「Multilineage cells from human adipose tissue: implications for cell-based therapies,」Tissue Eng. 7:211-228 (2001);及 Young H等人,「Human reserve pluripotent mesenchymal stem cells are present in the connective tissues of skeletal muscle and dermis derived from fetal, adult, and geriatric donors,」 Anat. Rec. 264:51-62 (2001)。 亦可自人類新生組織,諸如華頓氏膠質(Wharton’s jelly)(Wang Η 等人,「Mesenchymal stem cells in the Wharton's jelly of the human umbilical cord,」Stem Cells 22:1330-1337 (2004))、人類胎盤(Fukuchi Y等人,「Human placenta-derived cells have mesenchymal stem/progenitor cell potential,」Stem Cells 22:649-658 (2004));及臍帶血 (Erices A 等人,「Mesenchymal progenitor cells in human umbilical cord blood,」Br. J. Haematol. 109:235-242 (2000))及人類胎兒組織(Campagnoli C等人, 「Identification of mesenchymal stem/progenitor cells in human first-trimester fetal blood,liver,and bone marrow,」 Blood 98:2396-2402 (2001))分離MSC。 此項技術受限於不能分離出足夠的MSC供隨後分化及使 用。在可利用適合供者的情況下,為分離甚至有限數目之 152716.doc 201130977 MSC所需的侵入性程序,諸如骨髓生檢,會給供者帶來顯 著風險。亦難以維持所分離之MSC處於長期培養狀態及維 持此等培養物不被細菌或病毒污染。 先前已報導可自人類非經調動之周邊血中分離Msc。 Kassis I等人報導「使用血纖維蛋白微珠自經g_csf調動之 人類周邊金中分離間質幹細胞(Is〇lati〇n 〇f mesenchymal stem cells from G-CSF-mobilized human peripheral blood using fibrin microbeads,) d Bone Marrow Transplant. 37:967-976 (2GG6)〇 Kassis I等人報導自經調動之周邊血及 在較小程度上自非經調動之周邊血獲得MSC。已報導藉由 投與粒細胞群落刺激因子(Gr_l〇cyte_c〇l〇ny如則1峋Dominici M et al., "Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement," Cytotherapy 8:315 (2006). Because MSCs can differentiate into many cell types, this technology encompasses methods for differentiated MSCs for cell-based therapies, regenerative medicine, and reconstituted medicine. A typical method for preparing MSC compositions consists of isolating MSCs based on the plastic adhesion properties of MSCs and subsequently culturing them. This method is relatively easy when MSCs are isolated from human body fluids during fetal development. However, obtaining MSCs from human body fluids is much more complicated and reproducible much lower. 152716.doc 201130977 Typically, MSCs are isolated from adult bone marrow, fat, cartilage, and muscle. Pittenger F et al., "Multilineage potential of adult human mesenchymal stem cells," Science 284: 143-147 (1999); Zuk P et al., "Multilineage cells from human adipose tissue: implications for cell-based therapies," Tissue Eng. 7:211-228 (2001); and Young H et al., "Human reserve pluripotent mesenchymal stem cells are present in the connective tissues of skeletal muscle and dermis derived from fetal, adult, and geriatric donors," Anat. Rec. 264: 51-62 (2001). It can also be derived from human newborn tissue, such as Wharton's jelly (Wang Η et al., "Mesenchymal stem cells in the Wharton's jelly of the human umbilical cord," Stem Cells 22: 1330-1337 (2004)), humans. Placenta (Fukuchi Y et al., "Human placenta-derived cells have mesenchymal stem/progenitor cell potential," Stem Cells 22: 649-658 (2004)); and cord blood (Erices A et al., "Mesenchymal progenitor cells in human umbilical Cord blood,"Br. J. Haematol. 109:235-242 (2000)) and human fetal tissue (Campagnoli C et al., "Identification of mesenchymal stem/progenitor cells in human first-trimester fetal blood, liver, and bone marrow , Blood 98: 2396-2402 (2001)) Separation of MSCs. This technique is limited in that sufficient MSC cannot be isolated for subsequent differentiation and use. Invasive procedures required to isolate even a limited number of 152716.doc 201130977 MSCs, such as bone marrow biopsy, present a significant risk to the donor, where appropriate donors are available. It is also difficult to maintain the isolated MSCs in a long-term culture state and to maintain such cultures free from contamination by bacteria or viruses. It has previously been reported that Msc can be isolated from peripheral blood that is not mobilized by humans. Kassis I et al. reported that "Is〇lati〇n 〇f mesenchymal stem cells from G-CSF-mobilized human peripheral blood using fibrin microbeads, using fibrin beads from G_csf d Bone Marrow Transplant. 37:967-976 (2GG6)〇Kassis I et al. reported that MSCs were obtained from mobilized peripheral blood and, to a lesser extent, from non-mobilized peripheral blood. It has been reported that by administering granulocyte communities Stimulus factor (Gr_l〇cyte_c〇l〇ny is 1)
Factor,G-CSF)為期至少5天來將Msc調動至周邊血中。 然而’此方法似乎有缺陷且可再現性不良,因為僅較小百 分比之魔經分離’且此等Msc的生長及擴增潛力有限。 因此,有必要找到自周邊血中獲得MSC之允許製備富集 MSC群體的改良方法。 引用以上文獻並不意欲承纫 欲承w任何前述事項為相關先前技 術。關於此等文獻之日期之拚 之所有陳述或内容之所有表述皆 基於本申請人可用之資 且並非承認關於此等文獻之曰期 或内容之正確性。 【發明内容】 本發明大體上係關於投盥 价隹拉〜认 一肩動劑及自經調動之周邊血中 收集特疋幹細胞群之最佳時 崎序的發現。特定言之,本發明 人已發現在投與調動劑(諸Factor, G-CSF) mobilizes Msc to peripheral blood for at least 5 days. However, this method appears to be defective and poorly reproducible because only a small percentage of the magic is separated' and the growth and expansion potential of these Mscs is limited. Therefore, it is necessary to find an improved method for obtaining MSCs from peripheral blood that allows for the preparation of enriched MSC populations. The citation of the above documents is not intended to be a commitment to any of the foregoing matters as related prior art. All representations of all statements or content on the date of these documents are based on the information available to the applicant and are not an admission as to the correctness of the period or content of such documents. SUMMARY OF THE INVENTION The present invention is generally directed to the discovery of the optimal time sequence for collecting scorpion stem cell populations in the peripheral blood of the sputum. In particular, the inventors have discovered that in the administration of mobilizing agents (the
如(但不限於)GM-CSF及G-CSF 152716.doc 201130977 或AM3100(Plerixafor(普樂沙福)))之後自經調動之周邊血 中收集並分離間質幹細胞(MSC)的最佳時序。換言之,本 發明人已驚人地發現一種達成優良MSC產量的方法,其係 藉由在投與調動劑之後在經調動之周邊血中之MSC含量最 大時的最佳時間點分離經調動之MSC。 本發明人已驚人地發現’可在時間上調控MSC向周邊血 中之調動’且在向人類個體投與調動劑(諸如G-CSF或GM-CSF)之後,人類周邊血中之循環MSC之數目達到峰值的時 間與其他經調動之幹細胞群(諸如HSC)不同。因此,本發 明人已證明’使用G-CSF或GM-CSF最佳化MSC向周邊血 中調動之方案不同於通常用於將其他幹細胞(諸如CD3 4+ HSC)調動至周邊血中的方案。 因此’本發明係關於以時間調控方式自骨髓調動不同幹 細胞群的發現。特定言之’本發明人已發現,在投與特定 幹細胞調動劑之後的不同時間,骨髓釋放或產生不同幹細 胞群’且因此需要協調自經調動之周邊血中收集或分離某 一幹細胞群之時序以使特定相關幹細胞群之分離最佳化。 舉例而言’本發明人已發現向個體投與幹細胞調動劑將在 不同時間以不同速率 '不同持續時間及不同量調動幹細 胞’且因此本發明人已發現欲自經調動之周邊血中收集之 相關幹細胞將決定協調給藥方案及自經調動之周邊血中收 集幹細胞。舉例而言,本發明人已發現周邊血調動因子 (諸如GM-CSF及/或G-CSF)以獨特時間樣式調動不同幹細 胞群(諸如造血幹細胞(HSC)、間質幹細胞(MSC)及其他幹 152716.doc 201130977 細胞群,諸如極小胚胎樣(VSEL)幹細胞(「VSEL·」))進入 周邊血中。 驚人的是,不同於GM-CSF及/或G-CSF之習知用途(投與 其為期至少5天且在第6天收集HSC),本發明人已意外發現 可在投與GM-CSF及/或G-CSF約2天之後,在經調動之血液 中達成MSC高濃度。因此,本發明人已發現獲得最佳MSc 產量之方法,其中在投與該種調動劑至少1天,但至多4天 之後自經調動之周邊血中收集或分離MSC。在一實施例 中’投與該種調動劑約2天’且在投與調動劑之最後一天 或在次曰收集MSC »存在具有不同時間反應之其他調動 劑’且其可用於在投與調動劑之後調動MSC至少1天且小 於4天。 以習知劑量(例如通常為期5天,每天約5-1 〇 pg/kg)之生 長因子(諸如G-CSF或GM-CSF)調動幹細胞價格昂貴,且具 有不良副作用,包括引起廣泛骨疼痛、頭痛、疲勞、潛在 自發性脾破裂’且對正常供者具有未知副作用,包括造血 惡性疾病及骨髓白血病(AML)之風險增加(參見Casheen等 人,Bone Marrow Transplant, 2007,39; 577-588)。另外, 習知調動方案不可預測,導致產量較低,且通常會花費長 達約4週之G-CSF或GM-CSF處理以收集足夠用於移植之材 料。在生長因子處理之後’ HSC CD34+細胞在血液中達到 2-4%之最大含量。 通常自骨髓獲得MSC »先前已報導]vise以極低含量存在 於周邊血及經調動之周邊血中(參見Wexler等人,British J. 1527I6.doc 201130977Optimal timing for collecting and isolating mesenchymal stem cells (MSCs) from mobilized peripheral blood such as, but not limited to, GM-CSF and G-CSF 152716.doc 201130977 or AM3100 (Plerixafor) . In other words, the inventors have surprisingly discovered a method of achieving superior MSC production by isolating the mobilized MSC at the optimum point in time when the MSC content in the circulated peripheral blood is maximized after administration of the mobilizer. The present inventors have surprisingly found that 'the mobilization of MSCs into peripheral blood can be modulated in time' and after circulating a mobilizing agent (such as G-CSF or GM-CSF) to human subjects, circulating MSCs in human peripheral blood The number of peaks is different from other mobilized stem cell populations (such as HSC). Thus, the inventors have demonstrated that the regime of using G-CSF or GM-CSF to optimize mobilization of MSCs into peripheral blood differs from the regimen typically used to mobilize other stem cells, such as CD3 4 + HSC, into peripheral blood. Thus, the present invention relates to the discovery that different stem cell populations are mobilized from bone marrow in a time-controlled manner. Specifically, the inventors have discovered that at different times after administration of a particular stem cell mobilizer, the bone marrow releases or produces a different stem cell population' and therefore needs to coordinate the timing of collecting or isolating a stem cell population from the peripheral blood mobilized. To optimize the separation of specific related stem cell populations. For example, the inventors have discovered that administration of a stem cell mobilizer to an individual will mobilize stem cells at different rates 'different durations and different amounts at different times' and thus the inventors have discovered that they are to be collected from the peripheral blood of the mobilization. The relevant stem cells will determine the coordinated dosing regimen and collect stem cells from the peripheral blood of the mobilization. For example, the inventors have discovered that peripheral blood mobilization factors (such as GM-CSF and/or G-CSF) mobilize different stem cell populations (such as hematopoietic stem cells (HSC), mesenchymal stem cells (MSC), and other stems in a unique time pattern. 152716.doc 201130977 Cell populations, such as very small embryo-like (VSEL) stem cells ("VSEL·")) enter the peripheral blood. Surprisingly, unlike the conventional use of GM-CSF and/or G-CSF (with a period of at least 5 days and collecting HSC on day 6), the inventors have unexpectedly discovered that GM-CSF and/or can be administered. Or about 2 days after G-CSF, a high concentration of MSC is achieved in the mobilized blood. Accordingly, the inventors have discovered a method of obtaining optimal MSc yield in which MSCs are collected or isolated from the mobilized peripheral blood for at least 1 day, but up to 4 days after administration of the mobilizer. In one embodiment, 'administering the mobilizer for about 2 days' and on the last day of administration of the mobilizer or collecting MSCs in the second sputum, there are other mobilizers with different time responses' and they can be used for administration in the administration The MSC is then mobilized for at least 1 day and less than 4 days. The mobilization of stem cells at a conventional dosage (eg, typically 5 days, about 5-1 〇pg/kg per day), such as G-CSF or GM-CSF, is expensive and has undesirable side effects, including extensive bone pain, Headache, fatigue, and potentially spontaneous splenic rupture' have an unknown side effect on normal donors, including increased risk of hematopoietic malignancies and myeloid leukemia (AML) (see Casheen et al., Bone Marrow Transplant, 2007, 39; 577-588) . In addition, conventional transfer protocols are unpredictable, resulting in lower yields and typically take up to about 4 weeks of G-CSF or GM-CSF treatment to collect enough material for transplantation. After treatment with growth factors, HSC CD34+ cells reached a maximum of 2-4% in the blood. Usually obtained from bone marrow MSC » previously reported] Vise is present in peripheral blood and mobilized peripheral blood at very low levels (see Wexler et al., British J. 1527I6.doc 201130977)
Haematology, 2003; 121; 368-374;「Adult Bone Marrow is a rich source of human mesenchymal「stem」cells but umbilical cord and mobilized adult blood are not」)。此 外,報導已論述並非所有經調動之血液皆可產生MSC(參 見 Kassis 等人,Bone Marrow Transplantation,2006; 37; 967-976)。此外,即使MSC係自非經調動之周邊血中收 集,其亦不能足量收集用於未來的自體移植,即使培養擴 增殖亦如此,原因為MSC具有衰老之性質且在喪失其多能 潛力之前傳代次數有限。因此,本發明係關於以下驚人發 現:與先前報導相反,在MSC於循環中處於最高產量時的 最佳時間自經調動之周邊血中獲得或收集MSC時,可自經 調動之周邊血中獲得供治療性使用的足量MSC。 本發明之另一態樣係關於以下發現:在稱為「調動」之 過程中自骨髓遷移至周邊血中的MSC亦在周邊血中增殖。 因此,如本文揭示之調動劑(諸如GM-CSF及G-CSF或其促 效劑)可⑴投與個體以增強MSC自骨髓向周邊血中之調動 以增加周邊血中#趱源磔MSC(在本文中稱為「BM源性 MSC」)之數目,及/或(ii)可使個體之周邊血與調動劑(諸 如GM-CSF及G-CSF)離體接觸以調動及增強MSC在周邊血 中之增殖以增加周邊血中眉邊泌旗從在本文中稱為 「PB源性MSC」)之數目。欲用於實施本發明之其他調動 劑包括選自由介白素-17、AMD3100、環磷醯胺 (cyclophosphamide ; Cy)歐洲紫杉醇(Docetaxel)及(DXT)組 成之群之彼等調動劑。 152716.doc 201130977 因此,本發明之一態樣係關於向個體投與GM-CSF及/或 G-CSF以增強MSC之活體内調動;及使個體周邊血中經調 動之BM源性MSC及PB源性MSC的分離最佳化。 因此,本發明係關於自個鱧獲得周邊血源性人類間質幹 細胞之方法,其包含: a. 向該個體投與有效量之G-CSF或GM-CSF為期4天或 小於4天; b. 自得自該個體之周邊血樣品獲得人類間質幹細胞 群,其中該等人類間質幹細胞對於CD34-、CD45-細 胞表面標記呈陰性,且對CD44+、CD90+、 CD105+、CD29+及CD73 +細胞表面標記呈陽性;及 c. 相對於周邊血中之其他成體幹細胞(somatic stem cell)分離人類間質幹細胞。 本發明另外係關於增加個體之周邊血中人類間質幹細胞 群的方法,其包含: a_向個體之周邊投與有效量之G-CSF或GM-CSF為期4天 或小於4天; b·自得自該個體之周邊血樣品獲得人類間質幹細胞群, 其中該等人類間質幹細胞對於CD34-、CD45-細胞表面 標記呈陰性,且對於CD44+、CD90+、CD105+、CD29+ 及CD73 +細胞表面標記呈陽性;及 c.相對於周邊血中其他成體幹細胞分離人類間質幹細 胞。 在本發明之一特定實施例中,向個體投與有效量之G- 152716.doc •10· 201130977 CSF或GM-CSF為期3天或小於3天,為期2天或小於2天, 或為期1天或小於1天。在本發明之另一實施例中,個體為 健康個體。 本發明之另一態樣係關於使個體之周邊血與GM-CSF及/ 或G-CSF離體接觸;及使得自該個體之周邊血中經調動之 PB源性MSC的分離最佳化《因此,本發明係關於自個體獲 得人類間質幹細胞之方法,其包含: a. 使得自該個體之周邊血樣品與有效量之g-CSF或 GM-CSF接觸以增加周邊血樣品中人類間質幹細胞 之數目; b. 自周邊血樣品獲得人類間質幹細胞群,其中該等人 類間質幹細胞對於CD34-、CD45-細胞表面標記呈陰 性,且對於CD44+、CD90+、CD105+、CD29+及 CD73 +細胞表面標記呈陽性;及 c. 相對於周邊血中其他成體幹細胞分離人類間質幹細 胞。 本發明亦關於自個體獲得人類間質幹細胞之方法,其包 含: a.使周邊血樣品與有效量之g-CSF或GM-CSF活體外接 觸以增加周邊血樣品中人類間質幹細胞之數目; b_自周邊血樣品中分離人類間質幹細胞群,其中該等 人類間質幹細胞對於CD34-、CD45-細胞表面標記呈 陰性,且對於CD44+、CD90+、CD105+、CD29+及 CD73 +細胞表面標記呈陽性。 152716.doc •11· 201130977 在本發明之一實施例中’周邊血樣品可經由血液析離術 (apheresis)自個體獲得。周邊血樣品可為經培養之周邊血 樣品。在本發明之另一實施例中,周邊血樣品可為新鮮周 邊血樣品》得自周邊血之幹細胞群可經培養擴增。另外, 得自周邊血之人類間質幹細胞可經超低溫保存。 在本發明之一實施例中,人類間質幹細胞群係使用基於 尺寸之分離方法自周邊血樣品獲得》人類間質幹細胞群亦 可使用排除非間質幹細胞之負向選擇法(negative selection)自周邊血樣品獲得’其中自包含間質幹細胞群及 非間質幹細胞群的樣品中分離並收集人類間質幹細胞群。 人類間質幹細胞群亦可使用任何以下方法之組合自周邊血 中獲得:基於細胞表面標記之正向選擇、基於細胞表面標 記之負向選擇、基於尺寸之正向選擇、基於尺寸之負向選 擇。 本發明亦關於包含使用本發明方法所分離之實質上純人 類間質幹細胞群的純系細胞株。純系細胞株可置放於包含 適合培養基之容器中。另外’含有適合培養基及人類間質 幹細胞群之純系細胞株的容器可經超低溫保存。 因此,相較於G-CSF或GM-CSF用於活體内調動之習知 用途,本發明人提供的關鍵優點為顯著降低個體之風險及 與調動程序相關之成本’甚至提供一種使收集細胞之時序 最佳化為發現血液中之細胞(例如MSC)濃度最大時的時 間’藉此增加經調動之細胞之產量及減少非MSC之污染的 方法。 1527l6.doc •12· 201130977 本發明之另一態樣係關於自周邊血離體調動MSC,其中 使個體之周邊血與調動劑(諸如GM-CSF及G-CSF)接觸,且 自經調動之周邊金離體收集並分離周邊企源性MSC。在一 些實施例中,周邊血自個體(諸如人類個體)新鮮分離。在 一些實施例中,周邊血來自企庫或已儲存於血庫中。在一 些實施例中,周邊血已儲存或保藏。在一些實施例中,周 邊血為月經血,如美國專利申請案2008/0241113中所揭 示,該案以全文引用的方式併入本文中。 本發明之另一態樣係關於在周邊血中來自自骨髓(例如 BM源性MSC)或自周邊血(例如PB源性MSC)調動之MSC數 目到達峰值之時間點分離間質幹細胞(MSC)。在一些實施 例中,BM源性MSC及PB源性MSC可用於產生純系人類間 質祖細胞株,且更特定言之,在一實施例中,提供一種利 用BM源性MSC及PB源性MSC產生個別(例如個體特異性或 個人化)純系MSC株之方法,該等MSC自個體之活體内或 離體調動之周邊血獲得。 如本文所論述,本發明人已證明活體内投與低劑量之 GM-CSF及G-CSF(例如相較於GM-CSF及G-CSF作為調動劑 之習知用途,劑量更低)對於獲得周邊血中MSC數目之峰 值數目及對於獲得存在於人類個體之周邊血中之經調動的 PB源性及BM源性MSC之高產量最佳。因此,本發明人已 證明低劑量之GM-CSF及G-CSF或其促效劑可活體内用於 使自經調動之周邊血收集MSC最佳化,以便使用得自個體 (例如人類周邊血)之MSC製備用於細胞療法之組合物。 152716.doc -13- 201130977 在一些實施例中,本文所述之方法包括自個體獲得骨髓 源性MSC或周邊血源性MSC。接著可將所有BM源性MSC 或PB源性MSC或其亞群投與個體,例如MSC所來源之同一 個體(例如同種異體移植(all〇genic transplantation))或在某 些情形中,投與第二個體(例如非同種異體移植)(HLA型匹 配之第二個體)。在一些實施例中,方法亦包括例如使用 血液析離術或白血球血液析離術(leuk〇pheresis)自血液分 離BM源性MSC及/或PB源性MSC。在一些實施例中,分離 之BM源性MSC或PB源性MSC可投與有需要之個體,其中 該個體與MSC所來源個體為同一個體(例如同種異體移 植)’或在某些情形中投與第二個體(例如非同種異體移 植)’諸如HLA型匹配之第二個體。在一些實施例中,有 需要之個體患有選自由癌症及自體免疫疾病組成之群之病 狀或需要再生療法。 在另一態樣中,本文所述之方法包括自個體獲得周邊 血’使周邊血與有效量(例如足以離體增加周邊血中PB源 性MSC之數目之量)之如本文所述之調動劑(諸如g_cj§f或 GM-CSF)接觸。在一些實施例中,周邊血中之pB源性msc 可在投與個體之前經分離、富集或純化。視情況而定,在 一些實施例中’經處理之周邊血接著投與個體,例如同一 個體或第二個體,例如HLA型匹配之第二個體。 本發明之一態樣係關於藉由自經調動之周邊血中分離間 質幹細胞(MSC)且培養所獲得的組合物β此等組合物係使 用GM-CSF及G-CSF作為調動劑獲得。 152716.doc -14- 201130977 本發明係關於人類間質幹細胞群或其分化子代群户療有 需要之個體之疾病或病症的用途,其中該人類間質幹細胞 群係使用任何以上技術方案之方法分離,且其中向有需要 之個體投與該人_質幹細料或其分化子代群以達成自 體再生療法。 在一實施例中,本發明係有關治療人類個體之方法,其 中MSC之數目得以增加’且Msc經收集且用於細胞移植。 本發明方法採用如專利及公開案中所述之Msc調動劑,包 括GM-CSF及G-CSF,該等專利及公開案以引用的方式併 入本文中。 因此,本發明提供用自體間質幹細胞(MSC)治療個體之 疾病或病症之方法,其包含: a. 利用周邊血樣品之MSC群,其中該周邊血樣品得自 調動劑已投與4天或小於4天之個體,且其中該Msc 群為相對於周邊企中之其他幹細胞經富集的Msc 群;及 b. 向該個體投與該MSC群以治療該疾病或病症。 在本發明之一實施例中,可藉由正向選擇法或藉由淘洗 法(elutriation)富集MSC。富集之MSC群可包含至少1〇% MSC。富集之MSC群可為實質上純MSC群。富集之Msc群 可包含非M S C細胞。 另外,MSC群體可為已經超低溫保存之細胞群。另外, MSC細胞群可在投與個體之前已經活體外擴增。 本發明之一態樣係有關增加周邊血樣品中之MSC群的方 152716.doc •15· 201130977 法,其中該周邊血樣品存在於個體,諸如人類個體内。 本發明之另一態樣係關於活體内或離體富集來自個體之 周邊血樣品中之BM源性MSC及/或PB源性MSC群的方法。 在一些實施例中,活體内方法包含藉由以下步驟自個體之 虽集間質幹細胞之周邊血中分離人類間質幹細胞群:⑴向 該個體投與至少一種調動劑(諸如G_CSF或gm-CSF),為期 4天或小於4天;(Η)自該個體獲得周邊血樣品;(iii)使MSC 群與該個體之周邊血中之其他細胞分離,及視情況(iv)培 養MSC。培養及分離步驟亦可顛倒順序❶亦即,如本文揭 示之PB源性MSC或BM源性MSC可自周邊血分離且接著加 以培養擴增。此分離之方法包括白血球血液析離術、密度 梯度部分分離(density gradient fractionation)、免疫選擇及 差異黏著分離。培養基可為化學上限定之無血清培養基或 可為「完全培養基」,諸如DMEM或含有1 g血清之 DMEM。適合的化學上限定之無血清培養基描述於1995年 6月5曰申請之美國專利第08/464 599號中,且「完全培養 基」描述於1996年1月23日頒予之美國專利第5,486,359號 中。在一些實施例中,得自個體之BM源性MSC及/或PB源 性MSC在保藏(例如超低溫保存)之前經活體外擴增或培 養。 在一些實施例中,實質上純人類MSC群係得自至少一種 調動劑(例如G-CSF及/或GM-CSF)已投與4天或小於4天之 人類個體’該群體為人類MSC之實質上純異源群體,包含 BM源性人類MSC與PB源性人類MSC。 152716.doc -16· 201130977 在一些實施例中’得自至少一種調動劑(例如g_csf及/ 或GM-CSF)已投與4天或小於4天之人類個體的實質上純人 類MSC群包含BM源性人類MSC之實質上純群體。在替代 性貫施例中’得自至少一種調動劑(例如g_csf& /或gm-CSF)已投與4天或小於4天之人類個體的實質上純人類MSC 群包含PB源性人類MSC之實質上純群體。 本發明之另一態樣係關於離體富集個體之周邊血樣品中 PB源性MSC群的方法。在一些實施例中,離體方法包含藉 由以下步驟自個體之富集間質幹細胞之周邊血中分離人類 間質幹細胞群:使先前得自該個體之周邊血樣品與至少一 種調動劑(諸如G-CSF或GM-CSF)接觸為期4天或小於4天; 使MSC群與周邊血樣品中之其他細胞分離,及視情況培養 MSC。在一些實施例中’得自先前得自個體之周邊血樣品 的PB源性MSC可經保藏。在一些實施例中,保藏為超低溫 保存。在一些實施例中,PB源性MSC在保藏(例如超低溫 保存)之前經活體外擴增或培養。 本發明之另一態樣另外係關於離體保藏藉由如,本文揭示 之方法獲得之經分離PB源性MSC或BM源性MSC的方法, 其中該方法包含⑴向個體投與至少一種調動劑(諸如G-CSF 或GM-CSF)為期4天或小於4天;(ii)自個體獲得周邊血樣 品;(iii)使MSC群與個體之周邊血中之其他細胞分離,及 視情況(iv)培養MSC ;及(v)保藏經分離之PB源性MSC或 BM源性MSC。保藏較佳為超低溫保存。 本發明之另一態樣係關於用藉由如本文揭示之方法獲得 152716.doc -17- 201130977 之PB源性MSC及/或BM源性MSC的實質上純群體治療需要 治療之個體的方法,該方法包含向個體投與PB源性MSC或 BM源性MSC之實質上純群體。在一些實施例中,PB源性 MSC或BM源性MSC已藉由如本文揭示之方法新鮮分離, 或在一些實施例中,PB源性MSC或BM源性MSC已藉由熟 習此項技術者通常使用之已知方法加以超低溫保存。在一 些實施例中,PB源性MSC及/或BM源性MSC之實質上純群 體可藉由例如全身性輸注或局部植入(諸如藉由切開或關 節鏡(arthroscopic)程序)需要產生新生組織之部位内加以投 與。細胞可在再投與之前經保藏。 本發明之一態樣係關於自個體獲得周邊血源性人類間質 幹細胞之方法,其包含:(a)向該個體投與有效量之G-CSF 或GM-CSF為期4天或小於4天;(b)自得自該個體之周邊血 樣品獲得人類間質幹細胞群,其中該等人類間質幹細胞對 於CD34-、CD45-細胞表面標記呈陰性,且對於CD44+、 CD90+ ' CD105+、CD29+及CD73 +細胞表面標記呈陽性。 在一些實施例中,本發明係關於增加個體之周邊血中人類 間質幹細胞群之方法,其包含:(a)向該個體投與有效量之 G-CSF或GM-CSF為期4天或小於4天;(b)自得自該個體之 周邊血樣品獲得人類間質幹細胞群,其中該等人類間質幹 細胞對於CD34-、CD45-細胞表面標記呈陰性,且對於 CD44+、CD90+、CD105+、CD29+及 CD73 +細胞表面標記 呈陽性,及(c)相對於周邊血中之成體幹細胞分離人類間質 幹細胞。 152716.doc -18· 201130977 本發明之另一態樣係關於自個體獲得人類間質幹細胞之 方法,其包含:使得自該個體之周邊血樣品與有效量之G-CSF或GM-CSF接觸以增加周邊血樣品中人類間質幹細胞 之數目;(b)自周邊血樣品獲得人類間質幹細胞群,其中該 等人類間質幹細胞對於CD34-、CD45-細胞表面標記呈陰 性,且對於CD44+、CD90+、CD105+、CD29+及CD73 +細 胞表面標記呈陽性;及(c)相對於周邊血中之成體幹細胞分 離人類間質幹細胞。 在一些實施例中’方法包含向個體投與調動劑(諸如有 效量之G-CSF或GM-CSF、或AM3100)為期3天或小於3天。 在一些實施例中,向個體投與有效量之調動劑(諸如G-CSF 或GM-CSF或AM3100)為期2天或小於2天,或為期1天或小 於1天。在一些實施例中,向健康個體投與有效量之調動 劑。 在一些實施例中’人類間質幹細胞群係使用基於尺寸之 分離方法(諸如淘洗)自周邊血樣品獲得。在一些實施例 中’人類間質幹細胞群係使用排除非間質幹細胞之負向選 擇法自周邊企樣品獲得,其中自包含間質幹細胞群及非間 質幹細胞群的樣品分離並收集人類間質幹細胞群。 在一些實施例中,人類間質幹細胞群係使用任何以下方 法之組合自周邊血獲得:基於細胞表面標記之正向選擇、 基於細胞表面標記之負向選擇、基於尺寸之正向選擇、基 於尺寸之負向選擇。 本發明之另一態樣係關於自個體獲得人類間質幹細胞之 152716.doc •19· 201130977 方法,其包含:使周邊血樣品與有效量之G-CSF或GM-CSF活體外接觸以增加周邊血樣品中人類間質幹細胞之數 目;(b)自周邊血樣品分離人類間質幹細胞群,其中該等人 類間質幹細胞對於CD34-、CD45-細胞表面標記呈陰性, 且對於CD44+、CD90+、CD105+、CD29+及CD73 +細胞表 面標記呈陽性,及(c)相對於周邊血中之成體幹細胞分離人 類間質幹細胞。 在一些實施例中,MSC係自所培養之經調動的周邊血樣 品獲得。在一些實施例中,MSC係自作為新鮮周邊血樣品 之周邊血樣品獲得。在一些實施例中,MSC係自得自周邊 血樣品之經培養擴增的人類間質幹細胞群獲得。在一些實 施例中,得自周邊血樣品之人類間質幹細胞群經超低溫保 存。 在一些實施例中,個體為人類個體。本發明之另一態樣 係關於純系細胞株,其包含使用任何以上技術方案之方法 所分離之實質上純人類間質幹細胞群。 本發明之另一態樣係關於包含適合培養基及藉由如本文 揭示之方法獲得之人類間質幹細胞群的純系細胞株之容 器。在一些實施例中,適合培養基及人類間質幹細胞群之 純系細胞株經超低溫保存》 本發明之另一態樣係關於超低溫保存之藉由任何如本文 揭示之方法獲得的人類間質幹細胞群。本發明之另一態樣 係關於人類間質幹細胞群或其分化子代群治療有需要之個 體之疾病或病症的用途,其中人類間質幹細胞群係使用任 152716.doc • 20· 201130977 技術方案之方法加以分離,且其中向有需要之個體 w類間質幹細胞群或其分化子代群以達成自體再生 療法。 本發明之另-態樣係關於用自體間質幹細胞(M S C)治療 個體之疾病或病症之方法,其包含:⑷利用周邊血樣品之 MSC群’其中該周邊血樣品係自調動劑已投與4天或小於4 天之個體獲得,且其中該Msc群相對於血液中之其他幹細 胞群為經富集的MSC群;及(b)向該個體投與該Msc群以治 療該疾病或病症。在一些實施例中,藉由正向選擇法富集 MSC ’在替代性實施例中,藉由淘洗富集Msc。 在一些實施例中’ MSC群已經超低溫保存。在一些實施 例中,富集之MSC群包含非MSC細胞,或在一些實施例 中’富集之MSC群包含至少10% MSC或實質上純]^8(:群。 在一些實施例中,MSC群在投與個體之前已經活體外擴 增。在一些實施例中,個體為人類個體。 本發明另外提供自個體獲得周邊血源性人類幹細胞之方 法,其包含: a. 向該個體投與有效量之調動劑為期4天或小於4天; b. 自得自該個體之周邊血樣品獲得人類幹細胞群;及 c. 相對於周邊血中其他成體幹細胞分離人類幹細胞。 在本發明之一特定實施例中’幹細胞為間質幹細胞或 「極小胚胎樣幹細胞(VSEL)」。另外’調動劑可為G-CSF 或GM-CSF。或者,調動劑係選自由介白素]7、 AMD3 100、環磷醢胺(Cy)、歐洲紫杉醇及(DXT)組成之 152716.doc -21- 201130977 群ο 【實施方式】 間質幹細胞(MSC)為通常自骨趙獲得之非造血幹細胞。 MSC存在於周邊血中的含量極低且自周邊血中收集導致用 於治療性用途的量不足,即使其經活體外擴增亦如此。本 發明人已發現可自經調動之周邊血中獲得MSC。特定言 之’可自調動劑(例如(但不限於)G-CSF、GM-CSF、 AMD3 100等)已投與4天或小於4天(諸如約2天)之個體獲得 並收集MSC(使用血液析離術及淘洗法收集)^在一些實施 例中’自經調動之周邊血中所分離之人類MSC可經培養擴 增及/或經超低溫保存以供未來使用,例如其中Ms〇可在 再生療法中單獨或與其他細胞組合用於未來自體治療性應 用,諸如創口癒合及骨修復及其他矯形適應症。 發明者已驚人地發現,可在時間上調控MSC向周邊血中 之調動且在向人類個體投與調動劑(諸如G-CSF或GM-CSF) 後至少1天之後,人類周邊血中之循環MSC之數目達到峰 值。因此’本發明人已證明使用G_CSF或GM-CSF最佳化 MSC向周邊血中調動之方案不同於通常用於將其他幹細胞 (諸如CD34+HSC)調動至周邊血中的方案。 本發明人已意外地發現,向人類個體投與G-CSF或GM-CSF之後短期(例如為期至少2天,但小於4天)便會在經調 動之周邊血中得到MSC的最高產量,而非習知使用向個體 投與5天G-CSF或GM-CSF之方案。因此,本發明人已確 定’對於特定所要應用而言’投與幹細胞調動劑(諸如G_ 152716.doc •22· 201130977 CSF或GM-CSF)之方案需要藉由以下步驟加以最佳化:在 不同條件下投與G-CSF或GM-CSF ’且接著根據熟習此項 技術者通常所知及如本文揭示之方法監測周邊血中之所要 幹細胞亞群(諸如MSC、佩、造血細胞)的輸出。應監測 調動劑(諸如G-CSF或GM-CSF)投與後可供最佳回收或誘導 血液中之特定細胞群的時限,因為在投與調動劑之後的期 間會依序出現不同細胞類型》 本發明之另一態樣係關於得自個體經調動之周邊血之 MSC的用途,其用於個人化醫學應用,諸如自體治療性用 途’以及用於個人化檢定中以評估個人飲食、藥物遺傳 學、神經化學品、生活方式對MSC功能及活力之影響。 定4 _ 為方便起見,此處彙集在本文、說明書、實例及隨附申 請專利範圍中所採用之某些術語。除非另作陳述或由上下 文暗示,否則以下術語及片語包括下文提供之含義。除非 另作明確陳述或由上下文顯而易知,否則以下術語及片語 不排除術語或片語在其所屬技術中已獲得之含義。提供定 義以幫助描述特定實施例’且不意欲限制所主張之本發 明,因為本發明之範疇僅由申請專利範圍限制。除非另外 疋義,否則本文使用之所有技術及科學術語皆具有與本發 明所屬技術中之一般技術者通常理解相同的含義。 術語「間質幹細胞」在本文中亦稱為r MSC」,其係指 能夠分化成一種以上特定類型之間質或結締組織(亦即支 持特異化元件之身體組織;例如脂肪結締組織、骨結締組 152716.doc -23· 201130977 織、基質結締組織、軟骨結締組織、彈性結締組織及纖維 結締組織)的萬能幹細胞《人類間質幹細胞(hMSC)與稱為 SH2、SH3及SH4之某些單株抗體具有反應性。(參見美國 專利第5,486,359號,其以全文引用的方式併入本文中)。 MSC可基於其免疫特異性概況而與HSC區分,且其中Msc 為SH2+/CD14·且人類HSC為SH27CD14+。出於鑑別之目 的,可基於以下鑑別人類MSC : (i)CD34·、CD45·、 CD90+、CD105 +及CD44+之表型標記表現,(ii)功能表型, 包括在如本文實例中揭示之CFA檢定中形成群落形成單位 (colony forming unit)之能力,及分化成支持特異化元件 (包括(但不限於)軟骨細胞、軟骨及脂肪細胞)之組織之能 力。由MSC表現之其他標記在此項技術中為已知的且包括 (但不限於)CD71、CD73、Stro-1、CD166及 CD271。在某 些實施例中,MSC為lin·。 術語「極小胚胎樣幹細胞」在本文中亦稱為「VSEL幹 細胞」且係指萬能幹細胞。在一些實施例中,VSEL幹細 胞(「VSEL」)為人類VSEL且可表徵為lin·、CD45·及 CD34+。在一些實施例中,VSEL為人類VSEL且可表徵為 lin·、CD45·及CD133+。在一些實施例中,VSEL為人類 VSEL且可表徵為lin-、CD45·及CXCR4+。在一些實施例 中,VSEL為人類VSEL且可表徵為lin·、CD45·、CXCR4+、 CD133 +及CD34+ » 人類 VSEL 表現 SSEA-4、Oct-4、Rex-1及 Nanog之至少一者且具有由窄邊緣之細胞質包圍之大核, 且含有胚胎型無定形染色質。VSEL亦具有高端粒酶活 152716.doc -24- 201130977 性。在一些實施例中,VSEL為人類VSEL且可表徵為lin·、 CD45-、CXCR4+、CD133+、Oct 4+、SSEA4 +及 CD34+。在 一些實施例中,人類VSEL可具有較小原生性且可表徵為 lin·、CD45-、CXCR4+、CD133·及 CD34+。在一些實施例 中,人類VSEL可富含萬能胚胎轉錄因子,例如Oct-4、 Sox2及Nanog。在一些實施例中,人類VSEL可具有4-5 μιη ' 4-6 μηι、4-7 μηι、5-6 μηι、5,8 μηι、6-9 μπι或 7-10 μπι 之直徑。 如本文所用之術語「周邊血源性MSC」或「PB-MSC」 係指僅自周邊血調動之MSC,且可包括周邊血中之MSC的 擴增/增殖。在一些實施例中,可藉由根據如本文揭示之 方法使周邊血與調動劑活體内或離體接觸來增加周邊血中 循環PB-MSC之數目。 如本文所用之術語「骨髓源性MSC」或「BM-MSC」係 指自骨髓調動至周邊血中之MSC,且可包括已自BM遷移 之MSC。在一些實施例中,周邊血中之BM-MSC包括在遷 移至周邊血中之前已在骨髓中增殖之MSC,或者在自骨髓 遷移之後已在周邊血中增殖之MSC。在一些實施例中,可 根據如本文揭示之方法、藉由使周邊血與調動劑活體内接 觸來增加周邊血中循環BM-MSC之數目。 亦稱為「HSC」之術語「造血幹細胞」係指尤其在骨髓 中及周邊血中發現之能夠分化成任何特定類型之造血細胞 或血球(諸如紅血球、淋巴細胞、巨嗤細胞及巨核細胞)的 所有幹細胞或祖細胞。HSC與目前視為對造血細胞具有特 152716.doc -25« 201130977 異性之某些單株抗體(例如CD34)具有反應性。 亦稱為「HSC」之術語「造血細胞」係指在自我更新造 金幹細胞經由各種血系之不成熟前驅細胞分化成(且包括) 成熟功能血球之整個過程中之所有類型的造血細胞,如熟 習此項技術者所瞭解。 如本文所用之術語「調動」係指細胞藉此離開骨髓且進 入血液之過程。調動可藉由化學引誘劑(例如細胞激素)與 存在於周邊組織及骨趙之幹細胞小生境中之幹細胞集合池 或群體喪失黏附性的組合來實現。 「有效量」為足以實現周邊血中之MSC之數目及/或頻 率顯著增加的量。有效量可以一或多次投藥、用藥或給藥 投與。調動劑之治療有效量視所選調動劑(例如G_csf或 GM-CSF)而定。諸如G_CSF或Gm-CSF之調動劑可每天投 與一或多次至每週投與一或多次;包括每隔一天投與一 次。熟習此項技術者將瞭解某些因素可影響為有效治療個 體所需之劑量及時序,該等因素包括(但不限於)先前治 療、個體之综合健康及/或年齡、及存在之其他疾病。此 外’用如本文所述之治療有效量之G-CSi^^GM-CSF治療 個體可包括早一治療或一系列治療。 術語「間質細胞」或「間質」在本文中可互換使用且在 一些情況下係指在幼小胚胎之外胚層與内胚層之間所發現 的紡錘狀或星狀細胞;大多數間質細胞來源於所建立之中 胚層,但在頭部區域中,其亦由神經脊或神經管外胚層發 育而成。間質細胞具有萬能能力,特定言之,胚胎主體中 152716.doc •26- 201130977 之胚胎間質細胞能夠在不同位置發育成任何類型之結締組 織或支持組織、平滑肌、血管内皮及血球。 如本文所用,術語「幹細胞」在廣義上使用且包括傳統 幹細胞、袓細胞、先祖細胞、儲備細胞(reserve cell)及其 類似細胞。術語「幹細胞」或「祖細胞」在本文中可互換 使用,且係指能夠增殖及產生更多祖細胞之未分化細胞, 該等祖細胞能夠產生許多母細胞,該等母細胞又可產生經 分化或可分化之子細胞。子細胞自身可經誘導而增殖且產 生子代,隨後分化成一或多種成熟細胞類型,同時亦使— 或多種細胞保留親本發育潛力。術語「幹細胞」則指具有 在特定情況下可分化成更特異化或經分化表型之能力或潛 力的細胞,及保留在某些情況下增殖而不實質上分化之能 力的細胞。在一實施例中,術語祖細胞或幹細胞係指廣義 母細胞’其後代(子代)通常在不同方向上藉由分化而特異 化,例如獲得完全個別特徵,如胚細胞及組織之漸進多樣 化中所發生一樣。細胞分化為一種通常經由多次細胞分裂 而發生之複雜過程。分化細胞可來源於多能細胞,多能細 胞自身來源於多能細胞’諸如此類。儘管此等多能細胞之 各者可視為幹細胞,但各者可產生之細胞類型之範圍可顯 著變化。一些分化細胞亦具有產生發育潛力較大之細胞的 能力。此能力可為天然的或可在各種因子處理時經人工誘 導。在許多生物學情況下,幹細胞亦具有「多能」,因為 其可產生一種以上獨特細胞類型之子代,但此不為「主幹 性質(stem-ness)」所必需。自我更新為幹細胞定義之另一 152716.doc •27· 201130977 經典部分,且其如本文件中所用為重要的。在理論上,自 我更新可藉由兩種主要機制之任一者而發生。幹細胞可不 對稱分裂,其中一個子代保留主幹狀態且另一子代表現— 些獨特之其他特定功能及表型。或者,群體中之一些幹細 胞可對稱分裂成兩個幹細胞,從而總體上維持群體中之— 些幹細胞,而群體中之其他細胞僅產生分化子代。在形式 上’以幹細胞形式開始之細胞有可能向分化表型進展,但 接著「逆轉」且再表現幹細胞表型,此過程用通常稱為 「反分化(dedifferentiation)」之術語描述。 例示性幹細胞包括胚胎幹細胞、成人幹細胞、萬能幹細 胞、神經幹細胞、肝幹細胞、肌肉幹細胞、肌肉前驅幹細 胞、内皮祖細胞、骨髓幹細胞、軟骨性幹細胞、淋巴樣幹 細胞、間質幹細胞、造血幹細胞、中樞神經系統幹細胞、 周邊神經系統幹細胞及其類似幹細胞。對幹細胞之描述 (包括分離及培養幹細胞之方法)尤其可見於EmbryonicHaematology, 2003; 121; 368-374; "Adult Bone Marrow is a rich source of human mesenchymal "stem" cells but umbilical cord and mobilized adult blood are not"). In addition, the report has stated that not all mobilized blood can produce MSCs (see Kassis et al., Bone Marrow Transplantation, 2006; 37; 967-976). In addition, even if the MSC is collected from non-mobilized peripheral blood, it cannot be collected for future autologous transplantation, even if culture is expanded, because MSC has aging properties and loses its pluripotency potential. The number of previous passages was limited. Thus, the present invention relates to the surprising discovery that, contrary to previous reports, the optimal time for MSC to be at the highest yield in the circulation can be obtained from the mobilized peripheral blood when the MSC is obtained or collected from the circulated peripheral blood. A sufficient amount of MSC for therapeutic use. Another aspect of the present invention relates to the discovery that MSCs that migrate from bone marrow to peripheral blood during the process known as "mobilization" also proliferate in peripheral blood. Thus, a mobilizing agent (such as GM-CSF and G-CSF or an agonist thereof) as disclosed herein can (1) be administered to an individual to enhance mobilization of MSCs from the bone marrow into the peripheral blood to increase peripheral blood 趱 (趱 source 磔 MSC ( The number of "BM-derived MSCs" referred to herein, and/or (ii) ex vivo contact of the peripheral blood of the individual with a mobilizing agent (such as GM-CSF and G-CSF) to mobilize and enhance MSCs in the periphery. The proliferation in the blood increases the number of eyebrows in the peripheral blood from the "PB-derived MSC" herein. Other mobilizers for use in the practice of the invention include those selected from the group consisting of interleukin-17, AMD3100, cyclophosphamide (C), docetaxel, and (DXT). 152716. Doc 201130977 Accordingly, one aspect of the present invention relates to the administration of GM-CSF and/or G-CSF to an individual to enhance in vivo mobilization of MSC; and BM-derived MSC and PB-derived mobilization in peripheral blood of an individual The separation of MSCs is optimized. Accordingly, the present invention relates to a method for obtaining peripheral blood-derived human mesenchymal stem cells from a sputum, comprising: a. Administering to the individual an effective amount of G-CSF or GM-CSF for 4 days or less; b. A human mesenchymal stem cell population is obtained from a peripheral blood sample of the individual, wherein the human mesenchymal stem cells are negative for CD34-, CD45-cell surface markers, and surface markers of CD44+, CD90+, CD105+, CD29+, and CD73+ cells are present. Positive; and c. Human mesenchymal stem cells are isolated relative to other somatic stem cells in the peripheral blood. The invention further relates to a method for increasing a population of human mesenchymal stem cells in peripheral blood of an individual, comprising: a_ administering an effective amount of G-CSF or GM-CSF to the periphery of the individual for 4 days or less; b· A human mesenchymal stem cell population is obtained from a peripheral blood sample of the individual, wherein the human mesenchymal stem cells are negative for CD34-, CD45-cell surface markers, and are surface markers for CD44+, CD90+, CD105+, CD29+, and CD73+ cells. Positive; and c. Human mesenchymal stem cells are isolated relative to other adult stem cells in the peripheral blood. In a particular embodiment of the invention, an effective amount of G-152716 is administered to the individual. Doc •10· 201130977 CSF or GM-CSF for 3 days or less, for 2 days or less, or for 1 day or less. In another embodiment of the invention, the individual is a healthy individual. Another aspect of the present invention relates to contacting peripheral blood of an individual with GM-CSF and/or G-CSF in vitro; and optimizing separation of mobilized PB-derived MSCs from peripheral blood of the individual. Accordingly, the present invention is directed to a method of obtaining human mesenchymal stem cells from an individual comprising: a. Causing a peripheral blood sample from the individual to contact an effective amount of g-CSF or GM-CSF to increase the number of human mesenchymal stem cells in the peripheral blood sample; b. Human mesenchymal stem cell population obtained from peripheral blood samples, wherein the human mesenchymal stem cells are negative for CD34-, CD45-cell surface markers, and positive for CD44+, CD90+, CD105+, CD29+, and CD73+ cell surface markers; . Human mesenchymal stem cells are isolated relative to other adult stem cells in the peripheral blood. The invention also relates to a method of obtaining human mesenchymal stem cells from an individual, comprising: a. The peripheral blood sample is contacted with an effective amount of g-CSF or GM-CSF in vitro to increase the number of human mesenchymal stem cells in the peripheral blood sample; b_ separating the human mesenchymal stem cell population from the peripheral blood sample, wherein the human human The stem cells were negative for CD34-, CD45-cell surface markers and positive for CD44+, CD90+, CD105+, CD29+ and CD73+ cell surface markers. 152716. Doc • 11· 201130977 In one embodiment of the invention, a peripheral blood sample can be obtained from an individual via apheresis. The peripheral blood sample can be a cultured peripheral blood sample. In another embodiment of the invention, the peripheral blood sample can be a fresh peripheral blood sample. The stem cell population derived from peripheral blood can be expanded by culture. In addition, human mesenchymal stem cells derived from peripheral blood can be cryopreserved. In one embodiment of the invention, the human mesenchymal stem cell population is obtained from a peripheral blood sample using a size-based separation method. The human mesenchymal stem cell population may also use a negative selection method that excludes non-mesenchymal stem cells. Peripheral blood samples were obtained from which a human mesenchymal stem cell population was isolated and collected from samples containing mesenchymal stem cell populations and non-mesenchymal stem cell populations. Human mesenchymal stem cell populations can also be obtained from peripheral blood using any combination of the following methods: positive selection based on cell surface markers, negative selection based on cell surface markers, positive selection based on size, negative selection based on size . The invention also relates to a pure lineage cell line comprising a substantially pure human mesenchymal stem cell population isolated using the methods of the invention. The pure cell line can be placed in a container containing a suitable medium. Further, a container containing a pure cell line suitable for the medium and the human mesenchymal stem cell population can be cryopreserved. Thus, in contrast to the conventional use of G-CSF or GM-CSF for in vivo mobilization, the inventors provide a key advantage of significantly reducing the risk to the individual and the cost associated with the procedure, even providing a means of collecting cells. Timing optimization is the method of finding the time at which the concentration of cells (e.g., MSCs in the blood) is maximized, thereby increasing the yield of mobilized cells and reducing non-MSC contamination. 1527l6. Doc •12· 201130977 Another aspect of the invention relates to the mobilization of MSCs from peripheral blood, wherein the peripheral blood of the individual is contacted with a mobilizing agent such as GM-CSF and G-CSF, and the mobilized peripheral gold Peripheral MSCs were collected and isolated in vitro. In some embodiments, peripheral blood is freshly separated from an individual, such as a human individual. In some embodiments, the peripheral blood is from an enterprise library or has been stored in a blood bank. In some embodiments, peripheral blood has been stored or preserved. In some embodiments, the peripheral blood is menstrual blood, as disclosed in U.S. Patent Application Serial No. 2008/0241, the entire disclosure of which is incorporated herein by reference. Another aspect of the invention relates to the separation of mesenchymal stem cells (MSCs) at a time point in the peripheral blood from the time when the number of MSCs mobilized from bone marrow (e.g., BM-derived MSC) or from peripheral blood (e.g., PB-derived MSC) peaks. . In some embodiments, BM-derived MSCs and PB-derived MSCs can be used to generate pure human mesenchymal progenitor cell lines, and more particularly, in one embodiment, provide a utilization of BM-derived MSCs and PB-derived MSCs. Methods of producing individual (e.g., individual-specific or personalized) pure lineage MSC strains obtained from the peripheral blood of an individual in vivo or ex vivo. As discussed herein, the inventors have demonstrated that in vivo administration of low doses of GM-CSF and G-CSF (e.g., compared to conventional uses of GM-CSF and G-CSF as mobilizers, lower doses) is obtained. The peak number of MSCs in peripheral blood and the high yield of mobilized PB-derived and BM-derived MSCs present in peripheral blood of human subjects are optimal. Accordingly, the inventors have demonstrated that low doses of GM-CSF and G-CSF or agonists thereof can be used in vivo to optimize peripheral blood collection MSCs from mobilization for use in individuals (eg, human peripheral blood) MSCs prepare compositions for cell therapy. 152716. Doc-13-201130977 In some embodiments, the methods described herein comprise obtaining bone marrow-derived MSCs or peripheral blood-derived MSCs from an individual. All BM-derived MSCs or PB-derived MSCs or subpopulations thereof can then be administered to an individual, such as the same individual from which the MSC originated (eg, all〇genic transplantation) or, in some cases, administered Two individuals (eg, non-allogeneic transplants) (the second individual with an HLA type match). In some embodiments, the method also includes separating BM-derived MSCs and/or PB-derived MSCs from the blood, for example, using bloodpheresis or leuk〇pheresis. In some embodiments, the isolated BM-derived MSC or PB-derived MSC can be administered to an individual in need thereof, wherein the individual is the same individual as the MSC-derived individual (eg, allograft)' or in some cases A second individual, such as a non-allograft, is matched to a second individual (eg, an HLA type). In some embodiments, the individual in need has a condition selected from the group consisting of cancer and autoimmune diseases or requires regenerative therapy. In another aspect, the methods described herein comprise obtaining peripheral blood from an individual to mobilize peripheral blood with an effective amount (eg, an amount sufficient to increase the number of PB-derived MSCs in the peripheral blood ex vivo) as described herein. The agent (such as g_cj§f or GM-CSF) is contacted. In some embodiments, the pB-derived msc in the peripheral blood can be isolated, enriched, or purified prior to administration to the individual. As the case may be, in some embodiments the treated peripheral blood is then administered to an individual, such as the same individual or a second individual, such as a second individual that is HLA type matched. One aspect of the present invention relates to a composition β obtained by isolating mesenchymal stem cells (MSC) from peripheral blood mobilized and culturing such compositions using GM-CSF and G-CSF as a mobilizing agent. 152716. Doc -14- 201130977 The present invention relates to the use of a human mesenchymal stem cell population or a differentiated progeny group thereof for the treatment of a disease or condition in an individual in need thereof, wherein the human mesenchymal stem cell population is isolated using any of the above technical means, And wherein the individual is administered to the individual in need thereof, or the differentiated progeny group thereof, to achieve autologous regenerative therapy. In one embodiment, the invention relates to a method of treating a human subject wherein the number of MSCs is increased' and Msc is collected and used for cell transplantation. The method of the present invention employs Msc mobilizers as described in the patents and publications, including GM-CSF and G-CSF, which are incorporated herein by reference. Accordingly, the invention provides a method of treating a disease or condition in an individual with autologous mesenchymal stem cells (MSC), comprising: a. Utilizing a MSC population of peripheral blood samples, wherein the peripheral blood sample is obtained from an individual in which the mobilizer has been administered for 4 days or less, and wherein the Msc population is an Msc population enriched relative to other stem cells in the surrounding enterprise; And b. The MSC population is administered to the individual to treat the disease or condition. In one embodiment of the invention, the MSC can be enriched by a forward selection method or by elutriation. The enriched MSC population may comprise at least 1% MSC. The enriched MSC population can be a substantially pure MSC population. The enriched Msc population can comprise non-M S C cells. Alternatively, the MSC population can be a population of cells that have been cryopreserved. In addition, the MSC cell population can be expanded in vitro prior to administration to an individual. One aspect of the invention relates to the addition of a MSC population in a peripheral blood sample 152716. Doc • 15. 201130977, wherein the peripheral blood sample is present in an individual, such as a human individual. Another aspect of the invention is a method of enriching a BM-derived MSC and/or a PB-derived MSC population in a peripheral blood sample from an individual in vivo or ex vivo. In some embodiments, the in vivo method comprises isolating a population of human mesenchymal stem cells from peripheral blood of an individual collecting mesenchymal stem cells by the following steps: (1) administering to the individual at least one mobilizing agent (such as G_CSF or gm-CSF) ), for 4 days or less; (Η) obtaining peripheral blood samples from the individual; (iii) isolating the MSC population from other cells in the peripheral blood of the individual, and (iv) culturing the MSC as appropriate. The culture and isolation steps can also reverse the sequence, i.e., PB-derived MSCs or BM-derived MSCs as disclosed herein can be isolated from peripheral blood and subsequently cultured for amplification. Methods of isolation include leukocyte hemolysis, density gradient fractionation, immune selection, and differential adhesion separation. The medium may be a chemically defined serum-free medium or may be a "complete medium" such as DMEM or DMEM containing 1 g of serum. A suitable chemically defined serum-free medium is described in U.S. Patent No. 5,486,359 issued to Jun. 5, 1995, which is incorporated herein by reference. in. In some embodiments, BM-derived MSCs and/or PB-derived MSCs from an individual are expanded or cultured ex vivo prior to storage (e.g., cryopreservation). In some embodiments, the substantially pure human MSC population is derived from a human subject in which at least one mobilizing agent (eg, G-CSF and/or GM-CSF) has been administered for 4 days or less. A substantially pure heterologous population comprising BM-derived human MSCs and PB-derived human MSCs. 152716. Doc -16· 201130977 In some embodiments, a substantially pure human MSC population of a human individual who has been administered with at least one mobilizer (eg, g_csf and/or GM-CSF) for 4 days or less comprises BM source. A substantially pure population of human MSCs. In an alternative embodiment, a substantially pure human MSC population of a human individual from which at least one mobilizer (eg, g_csf & / or gm-CSF) has been administered for 4 days or less comprises PB-derived human MSCs. Essentially pure group. Another aspect of the invention is a method for ex vivo enrichment of a PB-derived MSC population in a peripheral blood sample of an individual. In some embodiments, the ex vivo method comprises isolating a human mesenchymal stem cell population from peripheral blood of an individual enriched mesenchymal stem cell by the step of: circumventing a peripheral blood sample previously obtained from the individual with at least one mobilizing agent (such as G-CSF or GM-CSF) exposure is for 4 days or less; the MSC population is separated from other cells in the peripheral blood sample, and MSCs are cultured as appropriate. In some embodiments, a PB-derived MSC obtained from a peripheral blood sample previously obtained from an individual can be deposited. In some embodiments, the deposit is cryopreserved. In some embodiments, the PB-derived MSC is expanded or cultured in vitro prior to storage (e.g., cryopreservation). Another aspect of the invention is additionally directed to a method of ex vivo isolation of PB-derived MSCs or BM-derived MSCs obtained by a method as disclosed herein, wherein the method comprises (1) administering to the individual at least one mobilizing agent (such as G-CSF or GM-CSF) for 4 days or less; (ii) obtaining peripheral blood samples from the individual; (iii) separating the MSC population from other cells in the peripheral blood of the individual, and optionally (iv) Cultivating MSC; and (v) preserving isolated PB-derived MSCs or BM-derived MSCs. The preservation is preferably cryopreservation. Another aspect of the invention pertains to obtaining 152716 by the method as disclosed herein. Doc -17-201130977 A substantially pure population treatment of a subject in need of treatment with a substantially pure population of PB-derived MSCs and/or BM-derived MSCs, the method comprising administering to the individual substantially pure PB-derived MSCs or BM-derived MSCs group. In some embodiments, the PB-derived MSC or BM-derived MSC has been freshly isolated by methods as disclosed herein, or in some embodiments, PB-derived MSCs or BM-derived MSCs have been known to those skilled in the art. It is usually cryopreserved using known methods. In some embodiments, a substantially pure population of PB-derived MSCs and/or BM-derived MSCs may be required to produce new tissue by, for example, systemic infusion or local implantation, such as by incision or arthroscopic procedures. The part is administered. Cells can be preserved prior to re-injection. One aspect of the invention relates to a method of obtaining peripheral blood-derived human mesenchymal stem cells from an individual comprising: (a) administering to the individual an effective amount of G-CSF or GM-CSF for 4 days or less. (b) obtaining human mesenchymal stem cell populations from peripheral blood samples obtained from the individual, wherein the human mesenchymal stem cells are negative for CD34-, CD45-cell surface markers, and for CD44+, CD90+ 'CD105+, CD29+, and CD73+ The cell surface marker is positive. In some embodiments, the invention relates to a method of increasing a human mesenchymal stem cell population in a peripheral blood of an individual comprising: (a) administering to the individual an effective amount of G-CSF or GM-CSF for a period of 4 days or less 4 days; (b) obtaining human mesenchymal stem cell populations from peripheral blood samples obtained from the individual, wherein the human mesenchymal stem cells are negative for CD34-, CD45-cell surface markers, and for CD44+, CD90+, CD105+, CD29+ and The CD73+ cell surface marker is positive, and (c) the human mesenchymal stem cells are isolated relative to the adult stem cells in the peripheral blood. 152716. Doc -18· 201130977 Another aspect of the invention relates to a method of obtaining human mesenchymal stem cells from an individual comprising: contacting a peripheral blood sample from the individual with an effective amount of G-CSF or GM-CSF to increase the periphery The number of human mesenchymal stem cells in the blood sample; (b) the human mesenchymal stem cell population obtained from the peripheral blood sample, wherein the human mesenchymal stem cells are negative for CD34-, CD45-cell surface markers, and for CD44+, CD90+, CD105+ , CD29+ and CD73 + cell surface markers are positive; and (c) isolation of human mesenchymal stem cells relative to adult stem cells in peripheral blood. In some embodiments the method comprises administering to the individual a mobilizing agent (such as an effective amount of G-CSF or GM-CSF, or AM3100) for a period of 3 days or less. In some embodiments, an effective amount of a mobilizing agent (such as G-CSF or GM-CSF or AM3100) is administered to an individual for a period of 2 days or less, or for 1 day or less. In some embodiments, an effective amount of a mobilizing agent is administered to a healthy individual. In some embodiments, the human mesenchymal stem cell population is obtained from a peripheral blood sample using a size-based separation method such as panning. In some embodiments, the 'human mesenchymal stem cell population is obtained from a peripheral sample using a negative selection method that excludes non-mesenchymal stem cells, wherein the human mesenchyme is isolated and collected from a sample comprising a mesenchymal stem cell population and a non-stromal stem cell population. Stem cell population. In some embodiments, the human mesenchymal stem cell population is obtained from peripheral blood using any combination of the following methods: positive selection based on cell surface markers, negative selection based on cell surface markers, positive selection based on size, based on size Negative choice. Another aspect of the invention relates to obtaining human mesenchymal stem cells from an individual 152716. Doc • 19·201130977 Method comprising: contacting a peripheral blood sample with an effective amount of G-CSF or GM-CSF in vitro to increase the number of human mesenchymal stem cells in a peripheral blood sample; (b) separating humans from peripheral blood samples a mesenchymal stem cell population, wherein the human mesenchymal stem cells are negative for CD34-, CD45-cell surface markers, and positive for CD44+, CD90+, CD105+, CD29+, and CD73+ cell surface markers, and (c) relative to peripheral blood Adult mesenchymal stem cells are isolated from adult stem cells. In some embodiments, the MSC is obtained from the mobilized peripheral blood sample being cultured. In some embodiments, the MSC is obtained from a peripheral blood sample that is a fresh peripheral blood sample. In some embodiments, the MSC is obtained from a population of human mesenchymal stem cells that have been cultured and expanded from peripheral blood samples. In some embodiments, the human mesenchymal stem cell population derived from the peripheral blood sample is preserved at an ultra-low temperature. In some embodiments, the individual is a human individual. Another aspect of the invention pertains to a pure lineage cell line comprising a population of substantially pure human mesenchymal stem cells isolated using the method of any of the above aspects. Another aspect of the invention pertains to a container comprising a pure lineage of a human mesenchymal stem cell population suitable for the culture medium and obtained by the methods disclosed herein. In some embodiments, a pure cell line suitable for culture and human mesenchymal stem cell populations is cryopreserved. Another aspect of the invention pertains to cryopreservation of a human mesenchymal stem cell population obtained by any of the methods disclosed herein. Another aspect of the invention relates to the use of a human mesenchymal stem cell population or a differentiated progeny population thereof for treating a disease or condition in a subject in need thereof, wherein the human mesenchymal stem cell population is used in any of 152,716. Doc • 20· 201130977 The method of the technical protocol is isolated, and the individual w-type mesenchymal stem cell population or its differentiated progeny group is required to achieve autologous regenerative therapy. A further aspect of the invention relates to a method for treating a disease or condition in an individual with autologous mesenchymal stem cells (MSC), comprising: (4) using a MSC population of peripheral blood samples, wherein the peripheral blood sample is self-mobilizing Obtained with an individual for 4 days or less, and wherein the Msc population is an enriched MSC population relative to other stem cell populations in the blood; and (b) administering the Msc population to the individual to treat the disease or condition . In some embodiments, the MSC is enriched by a forward selection method. In an alternative embodiment, the Msc is enriched by panning. In some embodiments the 'MSC population has been cryopreserved. In some embodiments, the enriched MSC population comprises non-MSC cells, or in some embodiments the 'enriched MSC population comprises at least 10% MSC or substantially pure ^8 (: population). In some embodiments, The MSC population has been expanded in vitro prior to administration to the individual. In some embodiments, the individual is a human subject. The invention further provides methods of obtaining peripheral blood-derived human stem cells from an individual comprising: a. Administering to the individual an effective amount of the mobilizer for a period of 4 days or less; b. Obtaining a human stem cell population from a peripheral blood sample of the individual; and c. Human stem cells are isolated relative to other adult stem cells in the peripheral blood. In a particular embodiment of the invention, the stem cells are mesenchymal stem cells or "very small embryonic stem cells (VSELs)". Further, the mobilizer may be G-CSF or GM-CSF. Alternatively, the mobilizing agent is selected from the group consisting of interleukin-7, AMD3 100, cyclophosphamide (Cy), European paclitaxel, and (DXT) 152716. Doc -21- 201130977 Group ο [Embodiment] Mesenchymal stem cells (MSC) are non-hematopoietic stem cells usually obtained from Bone. The presence of MSCs in peripheral blood is extremely low and is collected from peripheral blood resulting in insufficient amounts for therapeutic use, even if it is expanded in vitro. The inventors have discovered that MSCs can be obtained from the peripheral blood of the mobilization. Specifically, 'self-adjustable agents (such as, but not limited to, G-CSF, GM-CSF, AMD3 100, etc.) have been administered and collected by individuals for 4 days or less (such as about 2 days). Blood Separation and Panning Collection] In some embodiments, 'human MSCs isolated from the peripheral blood of the mobilization can be cultured and/or cryopreserved for future use, for example, where Ms can be used. Used alone or in combination with other cells in regenerative therapy for non-therapeutic applications such as wound healing and bone repair and other orthopedic indications. The inventors have surprisingly discovered that it is possible to modulate the mobilization of MSCs into peripheral blood over time and to circulate in human peripheral blood at least one day after administration of a mobilizer (such as G-CSF or GM-CSF) to a human subject. The number of MSCs peaks. Thus, the inventors have demonstrated that the use of G_CSF or GM-CSF to optimize mobilization of MSCs into peripheral blood differs from the regimen typically used to mobilize other stem cells, such as CD34+HSC, into peripheral blood. The inventors have surprisingly discovered that short-term (eg, at least 2 days, but less than 4 days) after administration of G-CSF or GM-CSF to a human subject will result in the highest yield of MSC in the mobilized peripheral blood, It is not customary to use a regimen that gives individuals 5 days of G-CSF or GM-CSF. Thus, the inventors have determined that 'for a particular application to be applied' a stem cell mobilizer (such as G_152716. Doc •22· 201130977 CSF or GM-CSF) The program needs to be optimized by: G-CSF or GM-CSF under different conditions and then generally known and familiar to those skilled in the art The methods disclosed herein monitor the output of a desired subset of stem cells (such as MSCs, pebbles, hematopoietic cells) in peripheral blood. The time limit for transfer of a mobilizer (such as G-CSF or GM-CSF) to optimally recover or induce a specific cell population in the blood should be monitored, as different cell types will occur sequentially after administration of the mobilizer. Another aspect of the invention pertains to the use of MSCs derived from individual mobilized peripheral blood for personalized medical applications, such as autologous therapeutic uses' and for personalization assays to assess personal diet, drugs The effects of genetics, neurochemicals, and lifestyle on MSC function and vitality. 4 _ For convenience, some of the terms used in this document, the description, the examples, and the accompanying claims are hereby incorporated. Unless otherwise stated or implied by the context, the following terms and phrases include the meanings provided below. Unless otherwise stated or apparent from the context, the following terms and phrases do not exclude the meaning of the term or phrase in the art to which it belongs. The definitions are provided to help describe a particular embodiment and are not intended to limit the claimed invention, as the scope of the invention is limited only by the scope of the claims. All technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs, unless otherwise claimed. The term "mesenchymal stem cells" is also referred to herein as r MSC", which refers to a body tissue that is capable of differentiating into more than one specific type of interstitial or connective tissue (ie, supporting specialized elements; eg, fat connective tissue, bone connective tissue) Group 152716. Doc -23· 201130977 omnipotent stem cells of woven, matrix connective tissue, cartilage connective tissue, elastic connective tissue and fibrous connective tissue. Human mesenchymal stem cells (hMSC) react with certain monoclonal antibodies called SH2, SH3 and SH4. Sex. (See U.S. Patent No. 5,486,359, incorporated herein by reference in its entirety). MSCs can be distinguished from HSCs based on their immunospecific profiles, and wherein Msc is SH2+/CD14 and human HSC is SH27CD14+. For identification purposes, human MSCs can be identified based on: (i) phenotypic expression of CD34., CD45., CD90+, CD105+, and CD44+, (ii) functional phenotype, including CFA as disclosed in the Examples herein. The ability to form a colony forming unit in assays and to differentiate into tissues that support specialized elements including, but not limited to, chondrocytes, cartilage, and fat cells. Other markers represented by MSC are known in the art and include, but are not limited to, CD71, CD73, Stro-1, CD166, and CD271. In some embodiments, the MSC is lin. The term "very small embryonic stem cells" is also referred to herein as "VSEL stem cells" and refers to omnipotent stem cells. In some embodiments, the VSEL stem cells ("VSEL") are human VSELs and can be characterized as lin·, CD45·, and CD34+. In some embodiments, the VSELs are human VSELs and can be characterized as lin·, CD45·, and CD133+. In some embodiments, the VSELs are human VSELs and can be characterized as lin-, CD45, and CXCR4+. In some embodiments, the VSEL is a human VSEL and can be characterized as lin·, CD45·, CXCR4+, CD133+, and CD34+ » human VSELs exhibit at least one of SSEA-4, Oct-4, Rex-1, and Nanog and have The nucleus of the narrow margin is surrounded by a large nucleus and contains embryonic amorphous chromatin. VSEL also has high-end granzyme activity 152,716. Doc -24- 201130977 Sex. In some embodiments, the VSEL is a human VSEL and can be characterized as lin·, CD45-, CXCR4+, CD133+, Oct 4+, SSEA4+, and CD34+. In some embodiments, human VSELs can be less native and can be characterized as lin·, CD45-, CXCR4+, CD133·, and CD34+. In some embodiments, human VSELs can be enriched with universal embryonic transcription factors, such as Oct-4, Sox2, and Nanog. In some embodiments, the human VSEL can have a diameter of 4-5 μιη ' 4-6 μηι, 4-7 μηι, 5-6 μηι, 5,8 μηι, 6-9 μπι, or 7-10 μπι. The term "peripheral blood-derived MSC" or "PB-MSC" as used herein refers to an MSC that is only mobilized from peripheral blood and may include amplification/proliferation of MSCs in peripheral blood. In some embodiments, the number of circulating PB-MSCs in the peripheral blood can be increased by contacting the peripheral blood with the mobilizing agent in vivo or ex vivo according to methods as disclosed herein. The term "bone marrow-derived MSC" or "BM-MSC" as used herein refers to MSCs that are mobilized from bone marrow to peripheral blood and may include MSCs that have migrated from BM. In some embodiments, the BM-MSCs in the peripheral blood include MSCs that have proliferated in the bone marrow prior to migrating to the peripheral blood, or MSCs that have proliferated in peripheral blood after migration from the bone marrow. In some embodiments, the number of circulating BM-MSCs in the peripheral blood can be increased by contacting the peripheral blood with the mobilizing agent in vivo according to the methods as disclosed herein. Also known as "HSC", the term "hematopoietic stem cells" refers to the ability to differentiate into any particular type of hematopoietic cells or blood cells (such as red blood cells, lymphocytes, giant cells, and megakaryocytes), especially found in the bone marrow and in peripheral blood. All stem or progenitor cells. HSC is currently considered to have a special 152716 for hematopoietic cells. Doc -25« 201130977 Some monoclonal antibodies (such as CD34) of the opposite sex are reactive. Also known as "HSC", the term "hematopoietic cells" refers to all types of hematopoietic cells in the process of self-renewal of gold-derived stem cells into (and including) mature functional blood cells through immature precursor cells of various blood lines, such as Those familiar with this technology know. The term "mobilization" as used herein refers to the process by which cells leave the bone marrow and enter the bloodstream. The mobilization can be achieved by a combination of a chemoattractant (e.g., a cytokine) and a pool of stem cell pools or groups that are present in the surrounding tissue and in the stem cell niche of the bone. An "effective amount" is an amount sufficient to achieve a significant increase in the number and/or frequency of MSCs in the peripheral blood. An effective amount can be administered by one or more administrations, administrations or administrations. The therapeutically effective amount of the mobilizing agent will depend on the selected mobilizing agent (e.g., G_csf or GM-CSF). A mobilizing agent such as G_CSF or Gm-CSF may be administered one or more times per day to one or more times per week; including once every other day. Those skilled in the art will appreciate that certain factors may affect the dosage and timing required to effectively treat an individual, including, but not limited to, prior treatment, overall health and/or age of the individual, and other conditions present. Further, treating a subject with a therapeutically effective amount of G-CSi^^GM-CSF as described herein can include an early treatment or a series of treatments. The term "interstitial cells" or "interstitial" is used interchangeably herein and in some cases refers to spindle-shaped or stellate cells found between young embryonic ectodermal and endoderm; most interstitial cells Originated from the established mesoderm, but in the head region, it is also developed by the neural ridge or neural tube ectoderm. Interstitial cells have versatile ability, specifically, in the embryonic body 152716. Doc •26- 201130977 Embryonic mesenchymal cells can develop into any type of connective tissue or supporting tissue, smooth muscle, vascular endothelium and blood cells at different locations. As used herein, the term "stem cell" is used broadly and includes conventional stem cells, tendon cells, progenitor cells, reserve cells, and the like. The term "stem cell" or "progenitor cell" is used interchangeably herein and refers to an undifferentiated cell that is capable of proliferating and producing more progenitor cells that are capable of producing a plurality of mother cells, which in turn can produce Differentiated or differentiated daughter cells. The daughter cells themselves can be induced to proliferate and produce offspring, which then differentiate into one or more mature cell types, while also allowing - or multiple cells to retain parental developmental potential. The term "stem cell" refers to a cell that has the ability or potential to differentiate into a more specific or differentiated phenotype under specific conditions, and that retains the ability to proliferate without substantial differentiation in some cases. In one embodiment, the term progenitor or stem cell line refers to a generalized mother cell 'the progeny (progeny) are usually differentiated by differentiation in different directions, for example to obtain completely individual features, such as progressive differentiation of embryonic cells and tissues. The same happens in the middle. Cells differentiate into a complex process that usually occurs through multiple cell divisions. The differentiated cells may be derived from pluripotent cells, and the pluripotent cells themselves are derived from pluripotent cells' and the like. Although each of these pluripotent cells can be considered a stem cell, the range of cell types that each can produce can vary significantly. Some differentiated cells also have the ability to produce cells with greater developmental potential. This ability can be natural or can be artificially induced when processed with various factors. In many biological situations, stem cells also have "pluripotent" because they produce offspring of more than one unique cell type, but this is not required for "stem-ness." Self-renewal is another definition of stem cells. Doc •27· 201130977 Classic part, and it is important as used in this document. In theory, self-renewal can occur by either of two main mechanisms. Stem cells can be asymmetrically split, with one progeny retaining the trunk state and the other progeny exhibiting some unique other specific functions and phenotypes. Alternatively, some of the stem cells in the population can be symmetrically split into two stem cells, thereby generally maintaining some of the stem cells in the population, while other cells in the population only produce differentiated progeny. Cells that formally start in the form of stem cells are likely to progress toward a differentiated phenotype, but then "reverse" and re-express the stem cell phenotype, a process that is commonly referred to as "dedifferentiation." Exemplary stem cells include embryonic stem cells, adult stem cells, omnipotent stem cells, neural stem cells, hepatic stem cells, muscle stem cells, muscle precursor stem cells, endothelial progenitor cells, bone marrow stem cells, cartilage stem cells, lymphoid stem cells, mesenchymal stem cells, hematopoietic stem cells, central nervous cells. System stem cells, peripheral nervous system stem cells, and similar stem cells. Description of stem cells (including methods for isolating and culturing stem cells) can be found especially in Embryonic
Stem Cells,Methods and Protocols,Turksen編,Humana Press,2002 ; Weisman等人,Annu. Rev. Cell. Dev. Biol. 17:387 403 ; Pittinger等人,Science,284:143 47, 1999 ; Animal Cell Culture,Masters編 ’ Oxford University Press, 2000 ; Jackson等人,PNAS 96(25):14482 86,1999 ; Zuk等 人 ’ Tissue Engineering, 7:211 228,2001(「Zuk等人」); Atala等人’尤其第33 41章;及美國專利第5,559,022號、 第5,672,346號及第5,827,735號。對基質細胞之描述(包括 分離基質細胞之方法)尤其可見於Prockop,Science,276:71 -28 · 152716.doc 201130977 74,1997 ; Theise 等人,Hepatology,31:235 40,2000 ; Current Protocols in Cell Biology, Bonifacino等人編,John Wiley & Sons,2000(包括直至2002年3月之更新);及美國 專利第4,963,489號。 術語「祖細胞」在本文中用於指相對於可藉由分化產生 之細胞,更具原生性細胞表型之細胞(例如處於比完全分 化細胞早之沿發育路徑之階段或進程)》通常,祖細胞亦 具有顯著或極高增殖潛力。視發育路徑及細胞發育及分化 之環境而定,祖細胞可產生多種獨特分化細胞類型或單一 分化細胞類型。 如上所指示,可依據「幹細胞」之一般定義將細胞分為 不同等級或種類。存在「全能(totipotent)」、「萬能 (pluripotent)」及「多能(multipotent)」幹細胞。術語「全 能」係指幹細胞可產生體内任何組織或細胞類型。「萬 能」幹細胞可產生體内除生殖系細胞之外的任何類型細 胞。可產生較小或有限數目之不同細胞類型的幹細胞一般 稱為「多能」。因此,全能細胞分化成可產生大多數(但非 所有)為胎兒發育所必需之組織的萬能細胞。萬能細胞進 步分化成專門產生具有特定功能之細胞的多能細胞。舉 例而言,多能造血幹細胞產生血液中之紅血球、白血球及 血小板。 如本文所用之術語「萬能」係指細胞能夠在不同條件下 分化成為所有3個生殖細胞層(内胚層、中胚層及外胚層)所 特有之細胞類型《萬能細胞的主要特徵為其能夠分化成所 152716.doc -29· 201130977 有3個胚層(使用例如裸小鼠畸胎瘤(teratoma)形成檢定)。 萬能性亦藉由表現胚胎幹(ES)細胞標記證實,但萬能性之 較佳測試為證明可分化成3個胚層之每一者之細胞的能 力。在一些實施例中,萬能細胞為未分化細胞。 如本文所用之術語「萬能性」或「多能狀態」係指細胞 能夠分化成所有3個胚胎胚層:内胚層(消化道組織)、中胚 層(包括血液、肌肉及血管)、及外胚層(諸如皮膚及神 經)’且通常具有可長期(例如1年以上或傳代30次以上)活 體外分裂的潛力》 術語「多能」在關於「多能細胞」使用時係指細胞能夠 分化成一些(但非所有)來源於所有3個胚層之細胞。因此, 多能細胞為部分分化細胞。多能細胞在此項技術中已熟 知,且多能細胞之實例包括成人幹細胞,諸如造血幹細胞 及神經幹細胞。多能意謂幹細胞可形成指定譜系中許多類 型之細胞,而非其他譜系之細胞。舉例而言,多能血液幹 細胞可形成許多不同類型之血球(紅血球、白血球、血小 板等),但其不能形成神經元。 術s吾「多能性」係指細胞發育多功能性(versatiHty)程度 小於全能及萬能。 術語「全能性」係指細胞具有描述形成成人體内所有細 胞以及胚胎外組織(包括胎盤)之能力的分化程度。與早期 分裂細胞(分裂球(blastomere))—樣,受精卵(合子)具有全 能性。 如本文所用之術語「經分離細胞」係指已自個體移除之 152716.doc •30- 201130977 細胞或該種細胞之後代,其中該細胞最初在該個體中被發 現。細胞視情況已例如在其他細胞存在下經活體外培養。 視情況隨後將藉由如本文揭示之方法產生之細胞(例如經 分離PB源性MSC或經分離BM源性MSC)引入第二個體中或 再引入已分離(例如同種異體移植)之該細胞(或其所來源之 細胞)所來源之個體中。 如本文所用之關於經分離細胞群之術語「經分離群體」 係指已移除且與混合或異質細胞群分離之細胞群。在一些 實施例中,相較於細胞已分離或富集之異質群體,經分離 群體為實質上純細胞群。在一些實施例中,經分離群體為 經分離重新編程之細胞群,其相較於包含重新編程之細胞 及該等重新編程之細胞所來源之細胞之異質細胞群為重新 編程之細胞的實質上純群體 關於特定細胞群之術語「實質上純」係指就組成總細胞 群之細胞而言,細胞群之純度為至少約75%、較佳至少約 85°/。、更佳至少約90%且最佳至少約95%。換而言之,關 於使用如本文揭示之方法所分離之PB源性MSC或BM源性 MSC群體的術δ吾「貫質上純」或「基本上經純化」係指pB 源性MSC或BM源性MSC群含有小於約2〇%、更佳小於約 15〇/〇、10%、8%、7%、最佳小於約 5〇/〇、4〇/〇、3%、2%、 1 %或小於1 〇/〇的細胞不為如本文術語所定義之p B源性M s C 或BM源性MSC。在一些實施例中,本發明涵蓋擴增抑源 性MSC或BM源性MSC群之方法,其中經擴增之叩源性 MSC或BM源性MSC群為抑源性MSqBM源性之實質 152716.doc -31 _ 201130977 上純群體。 二:文所用,「增殖」係指群體中之細胞數目藉助於細 二刀4而增加(生長)。細胞增殖通常理解為由多個信號 導路徑回應於環境(包括生長因子及其他有絲分㈣)之協 調活化所致。亦可藉由脫離阻斷或負性影響細胞增殖之细 胞内或細胞外信號及機制之作用而促進細胞增殖。 術語「再生」意謂在疾病或損傷之後,細胞群、器官或 組織之再生長。 術語「富集」或「經富集」在本文中可互換使用且意謂 某一類型之細胞之產量(分數)比彼類型之細胞在起始培養 物或製備物中之分數增加至少10〇/〇。 術語「更新」或「自我更新」或「增殖」在本文中可互 換使用,且係指細胞產生該細胞自身之更多複本之過程 (例如複製)。在一些實施例中,重新編程之細胞能夠藉由 長期及/或多月至多年分裂成相同未分化細胞(例如萬能或 非特異化細胞類型)而自我更新。在一些情況下,增殖係 指重新編程之細胞藉由單個細胞重複分裂成兩個相同子細 胞而擴增。 如本文所用之術語「譜系」係指描述具有共同袓先之細 胞或具有共同發育命運之細胞(例如來源於相同PB源性 MSC或BM源性MSC之細胞或其子代)的術語。 如本文所用,術語「純系細胞株」係指可在培養中維持 且有無限繁殖之潛力的細胞譜系。純系細胞株可為幹細胞 株(例如PB源性MSC或BM源性MSC細胞株)或來源於pb源 152716.doc •32· 201130977 性MSC或BM源性MSC,且當該純系細胞株用於包含PB源 性MSC或BM源性MSC之純系細胞株之情形中時,該術語 係指已在允許數月至數年增殖而不分化之活體外條件下培 養的PB源性MSC或BM源性MSC。此等純系幹細胞株(例如 PB源性MSC或BM源性MSC)可具有沿來自原始幹細胞之若 干細胞譜系分化的潛力。 在細胞個體發育之情形下,形容詞「分化」或「正分 化」為相關術語。「分化細胞」為已沿著發育路徑進一步 發展(與其所比較之細胞相比)的細胞。因此,幹細胞可分 化成受譜系限制的前驅細胞(諸如中胚層幹細胞),其又可 分化成沿路徑進一步發展之其他類型之前驅細胞(諸如心 肌細胞前驅體)’且接著分化成末期分化細胞,其在某一 組織類型中起特徵性作用,且可或可不保留進一步增殖之 能力。 本文中之術語「分化」意謂形成表現標記之細胞,該等 標記已知與更特異化及更接近變為不能進一步分化之末期 分化細胞的細胞相關。細胞自分化方向尚未確定之細胞進 展成分化方向日益確定成特定細胞類型之細胞且最终發展 成末期分化細胞所沿的路徑稱為漸進分化或漸進定型。更 特異化(例如已開始沿漸進分化之路徑進展)但尚未經末期 刀化之細胞稱為部分分化。分化為細胞藉此呈現特異化表 型,例如獲得一或多種不同於其他細胞類型之特徵或功能 的發育過程。在一些情況下,分化表型係指某一發育路徑 中之成熟終點時的細胞表型(所謂末期分化細胞)。在許多 152716.doc -33- 201130977 * _斤有)組織t,分化過程與退出細胞週期有聯繫。在 *月況下末期分化細胞喪失或極大限制其增殖能力。 :而,應注意在本說明書之情形下,術語「分化」或「經 刀化」係指細胞在其命運或功能方面比其發育中之先前點 時更特異化’ 包括末期分化之細胞與儘管未末期分化但 比其發月中之先前點時更特異化之細胞。細胞自未定型細 胞(例如幹細胞)發育成分化程度日益確定成特定分化細胞 類5L之細胞且最終發育成末期分化細胞稱為漸進分化或漸 進疋型。相對於袓細胞「經分化」之細胞相對於彼祖細胞 具有一或多種表型差異。表型差異包括(但不限於)形態學 差異及基因表現及生物活性差異,不僅包括存在或不存在 經表現之標記,而且包括標記之量之差異及一組標記之共 表現樣式之差異。 如本文所用之術語「分化」係指細胞自原生階段向更成 熟(亦即原生性較小)細胞之細胞發育。 如本文所用之術語「定向分化」係指經由遺傳及/或環 境操作迫使細胞自未分化細胞類型(例如更具原生性細胞) 分化成更成熟細胞類型(亦即原生性較小之細胞)^在一些 實施例中,如本文揭示重新編程之細胞定向分化成特定細 胞類型,諸如神經元細胞類型、肌細胞類型及其類似細胞 類型。 如本文提及之術語「培養基」為維持組織或細胞群或培 養細胞群的介質(例如「培養基」),其含有維持細胞活力 及支持增殖之養分。細胞培養基可含有以下任一者之適當 152716.doc -34- 201130977 組合:鹽類、緩衝劑類、胺基酸、葡萄糖或其他糖類、抗 生素血或血π替代物、及其他组分,諸如狀生長因子 等。-般用於特定細胞類型之細胞培養基為熟習此項技術 者所知。 術5吾「表型」係指在一組特定環境條件及因素下定義細 . 胞或有機體之一種或許多總生物特徵,而不論實際基因 型。 如本文所用之「標記」描述細胞之特徵及/或表型。標 記可用於選擇包含相關特徵之細胞。標記將隨特定細胞而 變化。標記為特定針對一種細胞類型或該細胞類型所表現 之为子的特徵(不論形態、功能或生物化學(酶促)特徵)。 此等標記較佳為蛋白質,且更佳具有針對此項技術中可用 之抗體或其他結合分子的抗原決定基。然而,標記可由細 胞中所發現之任何分子(包括(但不限於)蛋白質(肽及多 狀)、脂質、多醣、核酸及類固醇)組成,形態特徵或性狀 之實例包括(但不限於)形狀、尺寸、及核與細胞質之比 率。功能特徵或性狀之實例包括(但不限於)黏附於特定受 質之能力、合併或排除特定染料之能力、在特定條件下遷 • 移之能力、及沿特定譜系分化之能力。可藉由熟習此項技 術者可用之任何方法偵測標記。 如本文關於活體内接觸周邊血樣品所用之術語「接觸」 可包含經由適當投藥途徑向個體投與調動劑(視情況含於 組合物中)以使化合物活體内接觸周邊血樣品。如本文關 於離體接觸周邊血樣品所用之術語「接觸」可包含向周邊 152716.doc -35- 201130977 血樣品中投與調動劑(視情況含於組合物中)以使該調動劑 離體接觸該周邊血樣品。 如本文所用’術語「投與」、「引入」及「移植」可互換 使用且係指藉由使人類PB源性MSC或BM源性MSC至少部 分定位在所要部位處之方法或途徑將如本文所述之周邊血 源性MSC或骨髓源性MSC置入個體中。人類PB源性MSC或 BM源性MSC可藉由可傳遞至個體中之所要位置之任何適 當途徑投與,其中至少一部分人類PB源性MSC或BM源性 MSC仍存活^細胞投與個體之後的存活期可短至數小時 (例如24小時)至數天,至長達數年。 如本文所用,術語「供者」係指所收集之待移植器官、 組織或細胞所來源之個體。 如本文所用,術語「接受者」係指將接受所移植器官、 組織或細胞之個體。 如本文所用之術語「移植」係指藉以將游離(未附著)細 胞、組織或器官在移植入個體中之後整合入組織中的過 程。 術語「同種異體移植物」係指來源於相同物種之不同動 物之所移植細胞、組織或器官。 如本文所用之術語「異種移植物(Xen〇graft/Xen〇transplant)」 係指來源於不同物種之動物的所移植細胞、組織或器官。 在一些實施例中,異種移植物為來自某一物種之組織藉由 手術植入不同物種、屬或科中的移植物。舉例而言,自狒 狒植入人體中之移植物為異種移植物。 152716.doc •36· 201130977 術語「異種移植」係指活細胞、組織或器官自某一物種 移植至另一物種(諸如自豬移植至人體中)之過程。 術語「個體(subject)」與「個體(individual)」在本文中 可互換使用’且係指可根據本文所述之方法及組合物分離 且收集之如本文揭示之PB源性MSC所來源的動 物’例如人類,且個體視情況可接受移植(例如pB源性 MSC或BM源性MSC可植入個體中)用於例如治療,包括預 防比/。療疾,病。對於治療為諸如人類個體之特定動物所特 有之疾病狀態’術語「個體」係指彼特定動物。術語「非 類動物」及非人類哺乳動物」在本文中可互換使用, 且包括哺乳動物’諸如大鼠、小鼠、4、綿羊、貓 '犬、 奶牛、豬及非人類靈長類動物。術語「個體」亦涵蓋任何 脊椎動物,包括(但不限於 丨氏於)商乳動物、爬行動物、兩棲動 物及魚。然而》個體ig» SL· . 一 體且為诸如人類之哺乳動物,或其他哺Stem Cells, Methods and Protocols, Turksen, ed., Humana Press, 2002; Weisman et al, Annu. Rev. Cell. Dev. Biol. 17:387 403; Pittinger et al, Science, 284: 143 47, 1999; Animal Cell Culture , Masters, ed., Oxford University Press, 2000; Jackson et al, PNAS 96(25): 14482 86, 1999; Zuk et al., Tissue Engineering, 7:211 228, 2001 ("Zuk et al."); Atala et al. In particular, Chapters 33, 41; and U.S. Patent Nos. 5,559,022, 5,672,346 and 5,827,735. Descriptions of stromal cells (including methods for isolating stromal cells) are found in particular in Prockop, Science, 276: 71 -28 152 716. doc 2011 30 977 74, 1997; Theise et al, Hepatology, 31: 235 40, 2000; Current Protocols in Cell Biology, edited by Bonifacino et al., John Wiley & Sons, 2000 (including updates up to March 2002); and U.S. Patent No. 4,963,489. The term "progenitor cell" is used herein to refer to a cell that is more prototypically phenotypically relative to a cell that can be produced by differentiation (eg, at a stage or process that is earlier than the fully differentiated cell). Progenitor cells also have significant or extremely high proliferative potential. Depending on the developmental pathway and the environment in which the cells are developed and differentiated, progenitor cells can produce a variety of uniquely differentiated cell types or single differentiated cell types. As indicated above, cells can be classified into different grades or species according to the general definition of "stem cells." There are "totipotent", "pluripotent" and "multipotent" stem cells. The term "all-round" means that stem cells can produce any tissue or cell type in the body. "Universal" stem cells produce any type of cell in the body other than germline cells. Stem cells that produce a smaller or limited number of different cell types are commonly referred to as "pluripotent." Thus, totipotent cells differentiate into pluripotent cells that produce most, but not all, tissues necessary for fetal development. The unipotent cells are further differentiated into pluripotent cells that specifically produce cells with specific functions. For example, pluripotent hematopoietic stem cells produce red blood cells, white blood cells, and platelets in the blood. The term "universal" as used herein refers to a cell type that cells can differentiate into all three germ cell layers (endoderm, mesoderm, and ectoderm) under different conditions. The main feature of omnipotent cells is their ability to differentiate into 152716.doc -29· 201130977 There are 3 germ layers (using, for example, a nude mouse teratoma formation assay). The versatility is also confirmed by the expression of embryonic stem (ES) cell markers, but a better test of omnipotence is the ability to demonstrate differentiation into cells of each of the three germ layers. In some embodiments, the omnipotent cells are undifferentiated cells. The term "universal" or "pluripotent state" as used herein refers to a cell that is capable of differentiating into all three embryonic germ layers: endoderm (digestive tract tissue), mesoderm (including blood, muscle and blood vessels), and ectoderm ( Such as skin and nerves' and usually have the potential for long-term (for example, more than 1 year or more than 30 passages) in vitro. The term "multi-energy" refers to the ability of cells to differentiate into some when used with "pluripotent cells". (but not all) cells derived from all three germ layers. Therefore, pluripotent cells are partially differentiated cells. Pluripotent cells are well known in the art, and examples of pluripotent cells include adult stem cells, such as hematopoietic stem cells and neural stem cells. Multi-meaning means that stem cells can form many types of cells in a given lineage, rather than cells of other lineages. For example, pluripotent blood stem cells can form many different types of blood cells (red blood cells, white blood cells, blood platelets, etc.), but they cannot form neurons. The term "versatility" refers to the degree of versatiability (versatiHty) of cells that is less than omnipotent and omnipotent. The term "totipotency" refers to the degree of differentiation of a cell that describes the ability to form all cells in an adult as well as extraembryonic tissues, including the placenta. As with early dividing cells (blastomere), fertilized eggs (zygotes) are pluripotent. The term "isolated cells" as used herein refers to 152716.doc • 30-201130977 cells or progeny of such cells that have been removed from an individual, wherein the cells were originally discovered in the individual. The cells have been cultured in vitro, for example, in the presence of other cells, as appropriate. The cells produced by the methods disclosed herein (eg, isolated PB-derived MSCs or isolated BM-derived MSCs) are then introduced into the second individual or re-introduced into the isolated (eg, allograft) cells, as appropriate ( Or the individual from which the cell from which it is derived. The term "isolated population" as used herein with respect to an isolated population of cells refers to a population of cells that have been removed and separated from a mixed or heterogeneous population of cells. In some embodiments, the isolated population is a substantially pure population of cells compared to a heterogeneous population in which the cells have been isolated or enriched. In some embodiments, the isolated population is a reprogrammed cell population that is substantially reprogrammed by a heterogeneous population of cells comprising reprogrammed cells and cells from which the reprogrammed cells are derived. Pure population The term "substantially pure" with respect to a particular cell population means that the cell population has a purity of at least about 75%, preferably at least about 85°, for the cells that make up the total cell population. More preferably, it is at least about 90% and optimally at least about 95%. In other words, the use of a PB-derived MSC or a BM-derived MSC population isolated by the methods disclosed herein refers to a pB-derived MSC or BM. The source MSC population contains less than about 2%, more preferably less than about 15%/〇, 10%, 8%, 7%, optimally less than about 5〇/〇, 4〇/〇, 3%, 2%, 1 Cells of % or less than 1 〇/〇 are not p B-derived M s C or BM-derived MSCs as defined herein. In some embodiments, the invention encompasses a method of amplifying a BM-derived MSC or a BM-derived MSC population, wherein the amplified sputum-derived MSC or BM-derived MSC population is the source of the priming MSqBM source 152716. Doc -31 _ 201130977 on the pure group. 2: As used herein, "proliferation" means that the number of cells in a population is increased (growth) by means of a fine knife 4. Cell proliferation is generally understood to be caused by coordinated activation of multiple signaling pathways in response to the environment, including growth factors and other fractions (4). Cell proliferation can also be promoted by the action of intracellular or extracellular signals and mechanisms that block or negatively affect cell proliferation. The term "regeneration" means the regrowth of a cell population, organ or tissue after a disease or injury. The terms "enriched" or "enriched" are used interchangeably herein and mean that the yield (fraction) of a cell of a certain type is increased by at least 10% compared to the fraction of cells of that type in the starting culture or preparation. /〇. The terms "update" or "self-renewal" or "proliferation" are used interchangeably herein and refer to the process by which a cell produces more copies of the cell itself (e. g., replication). In some embodiments, the reprogrammed cells can self-renew by dividing into the same undifferentiated cells (e.g., omnipotent or non-specific cell types) over a long period of time and/or months to years. In some cases, proliferation refers to the reprogramming of cells that are amplified by repeated splitting of individual cells into two identical daughter cells. The term "lineage" as used herein refers to a term describing a cell having a common cell or a cell having a common developmental fat (e.g., a cell derived from the same PB-derived MSC or BM-derived MSC or a progeny thereof). As used herein, the term "pure cell line" refers to a cell lineage that is maintained in culture and has the potential for unlimited reproduction. The pure cell line may be a stem cell strain (for example, a PB-derived MSC or a BM-derived MSC cell strain) or a source derived from the pb source 152716.doc •32·201130977 MSC or BM-derived MSC, and when the pure cell line is used for inclusion In the case of a pure cell line of PB-derived MSC or BM-derived MSC, the term refers to PB-derived MSC or BM-derived MSC that has been cultured under in vitro conditions that allow proliferation for several months to several years without differentiation. . Such pure stem cell lines (e. g., PB-derived MSCs or BM-derived MSCs) may have the potential to differentiate along the stem cell lineage from the original stem cells. In the case of cell development, the adjectives "differentiation" or "positive differentiation" are related terms. A "differentiated cell" is a cell that has been further developed along the developmental pathway (compared to the cell to which it is compared). Thus, stem cells can differentiate into lineage-restricted precursor cells (such as mesoderm stem cells), which in turn can differentiate into other types of pro-driver cells (such as cardiomyocyte precursors) that further develop along the pathway and then differentiate into terminally differentiated cells, It plays a characteristic role in a certain tissue type and may or may not retain the ability to further proliferate. The term "differentiation" as used herein means the formation of cells expressing markers which are known to be associated with more specialized and closer cells that become terminally differentiated cells that are unable to differentiate further. The path along which the cells have not yet been determined from the direction of differentiation is increasingly determined to be a cell of a particular cell type and eventually develops into a terminally differentiated cell called progressive differentiation or progressive typing. More specific (e.g., cells that have begun to progress along the path of progressive differentiation) but have not yet undergone end-stage disease are referred to as partial differentiation. Differentiating into cells thereby presenting a specific phenotype, such as obtaining one or more developmental processes that differ from the characteristics or functions of other cell types. In some cases, a differentiated phenotype refers to the cellular phenotype at the maturity endpoint in a developmental pathway (so-called terminally differentiated cells). In many 152716.doc -33- 201130977 * _ jin has) tissue t, the differentiation process is linked to the withdrawal of the cell cycle. At the end of the month, the differentiated cells lose or greatly limit their ability to proliferate. In addition, it should be noted that in the context of this specification, the term "differentiation" or "knife-in" refers to a cell that is more specific in its fate or function than its pre-developmental point, including cells that are terminally differentiated, although A cell that has not been differentiated but is more specialized than its previous point in the month of the month. The degree of developmental differentiation of cells from unshaped cells (e.g., stem cells) is increasingly determined to be a cell of a specific differentiated cell type 5L and eventually developed into a terminally differentiated cell called a progressive differentiation or progressive sputum type. Cells that are "differentiated" relative to sputum cells have one or more phenotypic differences relative to progenitor cells. Phenotypic differences include, but are not limited to, morphological differences and differences in gene expression and biological activity, including not only the presence or absence of a manifested marker, but also differences in the amount of labeling and differences in the co-expression patterns of a set of markers. The term "differentiation" as used herein refers to the development of cells from the native stage to cells of more mature (i.e., less native) cells. The term "directed differentiation" as used herein refers to the forced differentiation of cells from undifferentiated cell types (eg, more native cells) into more mature cell types (ie, less primitive cells) via genetic and/or environmental manipulation^ In some embodiments, reprogrammed cells are disclosed as directed herein to differentiate into specific cell types, such as neuronal cell types, myocyte types, and the like. The term "medium" as referred to herein is a medium (e.g., "medium") that maintains a tissue or cell population or a population of culture cells that contains nutrients that maintain cell viability and support proliferation. The cell culture medium may contain any of the following: 152716.doc -34 - 201130977 combination: salts, buffers, amino acids, glucose or other sugars, antibiotic blood or blood π substitutes, and other components, such as Growth factors, etc. Cell culture media for use in a particular cell type are well known to those skilled in the art. A "phenotype" refers to the definition of one or many of the total biological characteristics of a cell or organism, regardless of the actual genotype, under a specific set of environmental conditions and factors. As used herein, "label" describes the characteristics and/or phenotype of a cell. Markers can be used to select cells that contain related features. The label will vary with the particular cell. Marked as a characteristic (whether morphological, functional or biochemical (enzymatic) characteristic) that is specific for a cell type or for that cell type. Such labels are preferably proteins, and more preferably have epitopes for antibodies or other binding molecules useful in the art. However, the label may be composed of any molecule found in the cell including, but not limited to, proteins (peptides and polymorphs), lipids, polysaccharides, nucleic acids, and steroids, examples of which may include, but are not limited to, shapes, Size, and ratio of nuclear to cytoplasm. Examples of functional features or traits include, but are not limited to, the ability to adhere to a particular receptor, the ability to incorporate or exclude a particular dye, the ability to migrate under specific conditions, and the ability to differentiate along a particular lineage. The marker can be detected by any method available to those skilled in the art. The term "contacting" as used herein with respect to contacting a peripheral blood sample in vivo may comprise administering to the individual via a suitable route of administration a mobilizing agent, optionally contained in the composition, such that the compound contacts the peripheral blood sample in vivo. The term "contacting" as used herein with respect to ex vivo contact with a peripheral blood sample can include administering a mobilizing agent (as appropriate in the composition) to a peripheral blood sample 152716.doc-35-201130977 to allow the mobilizing agent to be in contact with the body. The peripheral blood sample. As used herein, the terms 'administering,' "introducing," and "transplanting" are used interchangeably and refer to a method or pathway by which a human PB-derived MSC or BM-derived MSC is at least partially localized at a desired site. The peripheral blood-derived MSC or bone marrow-derived MSC is placed in the individual. Human PB-derived MSCs or BM-derived MSCs can be administered by any suitable route that can be delivered to a desired location in an individual, wherein at least a portion of the human PB-derived MSCs or BM-derived MSCs are still viable after the cells are administered to the individual. The survival period can be as short as several hours (for example, 24 hours) to several days, up to several years. As used herein, the term "donor" refers to the individual from which the organ, tissue or cell to be transplanted is collected. As used herein, the term "recipient" refers to an individual who will receive a transplanted organ, tissue or cell. The term "transplantation" as used herein refers to the process by which free (unattached) cells, tissues or organs are integrated into a tissue after implantation into an individual. The term "allograft" refers to a transplanted cell, tissue or organ derived from a different animal of the same species. The term "Xen〇graft/Xen〇transplant" as used herein refers to a transplanted cell, tissue or organ derived from an animal of a different species. In some embodiments, the xenograft is a graft from a species of a species that is surgically implanted into a different species, genus, or family. For example, a graft implanted into a human body is a xenograft. 152716.doc •36· 201130977 The term “xenograft” refers to the process by which a living cell, tissue or organ is transplanted from one species to another, such as from a pig to a human. The terms "subject" and "individual" are used interchangeable herein and refer to an animal from which PB-derived MSCs as disclosed herein can be isolated and collected according to the methods and compositions described herein. 'For example, humans, and individuals may be transplanted as appropriate (eg, pB-derived MSC or BM-derived MSC-implantable individuals) for, for example, treatment, including prophylaxis. Treating diseases, illness. For a disease state peculiar to a particular animal, such as a human individual, the term "individual" refers to a particular animal. The terms "non-animal" and non-human mammals are used interchangeably herein and include mammals such as rats, mice, 4, sheep, cats, dogs, cows, pigs, and non-human primates. The term "individual" also covers any vertebrate, including (but not limited to) 商 于, commercial animals, reptiles, amphibians and fish. However, "individual ig» SL· . is a mammal such as a human, or other
乳動物’诸如馬丨丨養哺受丨包I 你、…^ €例如犬、猶、馬及其類似動 # + 奶牛綿羊、豬及其類似動物)亦Milk animals, such as the horses, feeding the quilt, I, ... ^ € such as dogs, squad, horses and similar movements # + cows, sheep, pigs and similar animals)
涵蓋在術語個體中。 W 術語「組織」係指類似 貝似特異化細胞之群組或層,1丘 執行某些特殊功能。椒4「 ’、八同 織之細胞之來源特徵或限定性特徵。 ^自特疋組 如本文所用之術語「藥劑」意謂任何 如(但不限於)小分子、枋 物次物質,诸 「藥劑I可Λ杯打儿内 樂物離子專。 樂劑」T為任何化學品、f 合成及天然存在之蛋白及非蛋白實體:在=不限於) «祖在一些實施例中, 152716.doc •37- 201130977 藥劑為核酸;核酸類似物;蛋白質;抗體;肽;適體;核 酸、胺基酸或碳水化合物之寡聚物,包括(但不限於)蛋白 質、寡核苷酸、核糖核酸酶、DNA酶、醣蛋白、siRNA、 脂蛋白、適體及其修飾形式及組合等。在某些實施例中, 藥劑為具有化學部分之小分子。舉例而言,化學部分包括 未經取代或經取代烷基、芳族或雜環基部分’包括巨環内 醋(maCrolide)、來普黴素(丨ept〇mycin)及其相關天然產物或 類似物。化合物可已知具有所要活性及/或性質,或可選 自不同化合物之集合庫。 如本文所用,術語「小分子」係指化學藥劑,其可包括 (但不限於)肽、肽模擬物、胺基酸、胺基酸類似物、聚核 苷酸、聚核苷酸類似物、適體、核苷酸、核苷酸類似物、 分子量小於約10,000公克/莫耳之有機或無機化合物(例如 包括雜有機化合物及有機金屬化合物)、分子量小於約 5,000 a克/莫耳之有機或無機化合物、分子量小於約1,⑽〇 公克/莫耳之有機或無機化合物、分子量小於約5〇〇公克/莫 耳之有機或無機化合物、及此等化合物之鹽、酯及醫藥學 上可接受之其他形式。 術§吾「疾病」或「病症」在本文中可互換使用,且係指 身體或一些器官狀態之任何改變,從而中斷或干擾功能之 執灯及/或引起罹患者或與個體接觸者之症狀,諸如不 適、功能障礙、痛苦、或甚至死亡。疾病或病症亦可指瘟 熱、不舒服、失調、混亂、異常、。惡心、病患、不適、微 恙或病變。 1527l6.doc -38 - 201130977 如本文所用之術語「病理」係指造成疾病或病症之症 狀,例如細胞、組織或器官之結構及功能變化。舉例而 言’病理可與特定核酸序列或「病理性核酸」相關,該病 理性核酸係指完全或部分造成病理之核酸序列,舉例而 5 ’病理性核酸可為編碼具有引起特定病理或與病理相關 之突變或多形現象(polymorphism)之基因的核酸序列。病 理可與完全或部分造成與特定疾病或病症相關之病理之病 理性蛋白質或病理性多肽的表現相關。在另一實施例中, 病理例如與其他因素(例如缺血及其類似因素)相關。 如本文所用,術語「治療」包括減輕或緩和病狀、疾病 或病症之至少一種不良反應或症狀。 如本文所用之片語「非經腸投藥」意謂除經腸及局部投 藥以外之投藥方式,通常為注射,且包括(但不限於)靜脈 内、肌肉内、動脈内、鞘内、心室内、囊内、眶内、心 内、真皮内、腹膜内、經氣管、皮下、表皮下、關節内、 囊下、蛛網膜下、脊椎内、腦脊髓内、及胸骨内注射及輸 注。如本文所用之片語「全身性投藥」、「周邊投藥」意謂 PB源性MSC或BM源性MSC或其分化子代及/或其子代及/ 或化合物及/或其他物質以使其進入動物全身中且從而經 受代謝及其他類似過程的方式投與(例如皮下或靜脈内投 與),而非直接進入個體中。 片語「醫藥學上可接受」在本文中用於指在合理醫學判 斷之㈣内適用於與人類及動物組織接觸而無過度毒性、 刺激、過敏反應或其他問題或併發症且與合理效益/風險 152716.doc •39- 201130977 比率相稱的彼等化合物、物質、組合物及/或劑型。 如本文所用之片語「醫藥學上可接受之載劑」意謂在自 身體之某一器官或部分傳送或輸送標的藥劑至身體之另一 器官或部分中所涉及的醫藥學上可接受之物質、組合物或 媒劑,諸如液體或固體填充劑、稀釋劑、賦形劑、溶劑或 囊封物質。各載劑在可與調配物之其他成分相容之意義上 必須為「可接受」。 如本文所用之術語「藥物」或「化合物」係指投與個體 以治療或預防或控制疾病或病狀之化學實體或生物製品, 或化學實體或生物製品之組合。化學實體或生物製品較佳 (但非必定)為低分子量化合物,但亦可為較大化合物,例 如核酸、胺基酸、或碳水化合物之寡聚物,包括(但不限 於)蛋白質、寡核苷酸、核糖核酸酶、DNA酶、醣蛋白、 siRNA、脂蛋白、適體及其修飾形式及組合。 如本文所用之術語「移植」係指將新細胞(例如PB源性 MSC或BM源性MSC或其分化子代)或組織(諸如由pb源性 MSC或BM源性MSC產生之分化細胞)或器官引入宿主(亦即 移植物接受者或移植物個體)中。 術語「調節j的用法與其在此項技術中之用法一致,例 如引起或促進相關過程、路徑或現象之定性或定量變化、 改變或修改。此變化可為(不限於)過程、路徑或現象之不 同組分或分支之相對強度或活性的增加、減小或變化。 「調節劑」為引起或促進相關過程、路徑或現象之性質或 數量變化、改變或修改之藥劑。 I52716.doc -40· 201130977 術語減少」、「降低」或「抑制」在本文中 意謂以統計學顯著量減少。然而,為避免疑問,「降; 或。「減少」或「抑制」意謂相較於參考含量減少至二 腕,例如減少至少約鳩、或至少約鳩、或至/ 娜、或至少約鄉、或至少約6Q%、或至少物%/至 於春者揭。 或多達且包括1〇〇%降幅(例如相較 樣。°,不存在含量),或相較於參考含量為介於1〇 100%之間的任一降幅。 1於10- 術語「增加」或「增強」或「活化」在本文中一般皆用 於意“胃以統計學顯著量增加;為避免任何疑問,術語「辦 加」或㉟強」或「活化」意謂相較於參考含量增加至少 0例如增加至少約20%、或至少約30%、或至少約 桃、或至少㈣%、或至少約峨、或至少約聰、或至 少=〇%、或至少約90%或多達且包括刚%增幅或相較 於> 考含量為介於1(M00%之間的任一增幅,或為參考含 量之至少約2倍 '或至少約3倍、或至少約4倍、或至少約$ 倍或至少約H)倍’或為介於2倍與1G倍或iq倍以上之間的 任一增幅。 j語「統計學顯著」《「顯著」係指統計學顯著性且通 $意4比標記之標準或較低濃度低兩倍標準差(2SD)。該 ΓΓ 系指證實存在差異的統計學證據。其定義為當虛無假 -貫際上真實時’決定排除虛無假設 作出決定。如本文所用之術語「實質上」意= 6〇/。、或較佳至少約观或至少約嶋 '或至少約娜、至 152716.doc •41- 201130977 少約95%、至少約97%或至少約99%或99%以上,或介於 7 0 %與1 〇 〇 %之間之任何整數的比例。 如本說明書及隨附申請專利範圍中所使用,除非上下文 另外明確規定,否則單數形式「一」及「該」包括複數個 參考物。目此’舉例而言,提及「該方法」包括本文所述 及/或熟習此項技術者在閱讀本發明時將顯而易知的一或 多個方法及/或步驟類型,諸如此類。 應暸解以上詳細說明及以下實例僅具有說明性且不應視 為限制本發明L可在殘離本發明之㈣及範相 情況下對所揭示之實施例進行各種修改及潤飾,此等修改 及潤飾將為熟習此項技術者顯而易知。另外,所鑑別之所 有專利、專射請案及公開案皆以引用的方式明確併入本 文中,以用於描述及揭示例如此等公開案中所述之可配合 本發明使用之方法的㈣^在本中請案之申請日期之前, 此等公開案僅為揭示其所提供。就此而言,不應理解為承 認本發明人因先前發明或任何其他原因而無權將本發明之 曰期提前。關於此等文獻之曰期之所有陳述或内容之所有 表述皆係基於本申請人可用夕咨 用之資訊且不構成關於此等文獻 之日期或内容之正確性的任何承認。 MSC調動劑 在本發明之-態樣中,提供治療方法,其包含向哺乳鸯 物(例如人類)投與有效量之調動劑,例如某-形式之G CSF或GM-CSF及其醫藥學上可接受之鹽以增強㈠, 激)MSC產生/釋放,如依據表型、物理或化學性質或熟習 152716.doc •42· 201130977 此項技術者用於鑑別MSC型細胞之任何其他特徵所量度。 在一實施例中,在約2次每日一次注射投與諸如GM-CSF 或G-CSF之調動劑之後,得到循環MSC的峰值數目。周邊 血中循環MSC之峰值在至少4次每日一次注射之後下降。 任何間質調動劑或間質調動劑之組合可在規定時間投與 個體以最佳化MSC向周邊血中之調動。在一些實施例中, MSC調動劑為GM-CSF或G-CSF(粒細胞群落刺激因子)。在 一些實施例中,可以單一調動劑或此等調動劑之組合形式 向個體投與其他調動劑,包括(但不限於)G-CSF、GM-CSF、Flt-3配位體、幹細胞因子(SCF)、地塞米松 (dexamethazone)、CXCR4受體抑制劑、介白素-l(IL-l)、 介白素-3(IL-3)、戴尼普司亭(Diniplestim)(IL-3促效劑)、 來立司亭(Leridistim)(IL-3促效劑-G-CSF喪合分子)、祖細 胞生成素-l(Progenipoietin-l)(Flt-3 配位體-G-CSF 嵌合分 子)、聚乙二醇化非格司亭(peg-filgrastim) (NEULASTATM)、介白素-8(IL-8)、PIXY-321(GM-CSF/IL-3 融合蛋白)、巨噬細胞發炎性蛋白、幹細胞因子(SCF)、血 小板生成素(thrombopoietin)及生長相關之致癌基因。其他 調動劑包括例如(但不限於)揭示於美國專利第7,169,750號 中之 CXCR4抑制劑,諸如 AMD3100、ALX40-4C、T22、 T134、T140及TAK-779,該專利以全文引用的方式併入本 文中。 在一些實施例中,諸如G-CSF或GM-CSF、或介白素-17 (IL-17)、AMD3100、環磷酿胺(Cy)及歐洲紫杉醇(DXT)及 152716.doc • 43- 201130977 其組合之調動劑可混合投與個體或離體投與周邊血樣品以 刺激PB源性MSC。 在—些實施例中’諸如G-CSF或GM-CSF之調動劑可與 療上或營養上適用之其他活性成分混合投與個體或離體 投與周邊血樣品,該等其他活性成分諸如抗生素、維生 素、草本萃取物、消炎劑、葡萄糖、解熱劑、止痛劑、粒 細胞-巨噬細胞群落刺激因子(GM_CSF)、介白素_1(IL1)、 介白素-3(IL-3)、介白素 _8(IL-8)、PIXY-321(GM-CSF/IL-3 融合蛋白)、巨噬細胞發炎性蛋白、幹細胞因子、血小板 生成素及其類似物。 使用例如G-CSF或GM-CSF之調動劑所產生之經調動的 周邊血幹細胞集群(PBSC)可移植入例如癌症患者中。此等 集群可為經調動之白血球或自經調動之周邊血獲得之純化 HSC的總群體。Hsc能夠使有機體之造血系統重組,此需 要HSC之自我更新以及分化成各種造血譜系之細胞。cD34 標記為HSC所特有。使用此等調動劑調動的其他細胞包括 多形核白血球(介導發炎及病原體清除之細胞)、單核白血 球(淋巴細胞及單核細胞)及紅血球祖細胞(紅血球母細 胞)。 CD34+ HSC之調動為一種獲得用於自體或同種異體造血 移植之材料的快速擴增臨床技術。此外,多形核白血球之 調動為沉重化學療法之重要輔助手段以維持先天防禦機 制。當前,兩種方法均依賴昂貴、會引起骨疼痛且對正常 供者具有未知副作用的生長因子(❻/”或GM_CSF)進行調 1527J6.doc • 44 - 201130977 動。花費長達約4週之處理來收集足夠用於移植之材料。 在生長因子處理之後,CD34+細胞在血液中達到2-4%之最 大含量。 G-CSF : 在一些實施例中,用於如本文揭示之方法中之G-CSF可 使用重組方法製備且可使用G-CSF之變異體。根據本發明 之術語G-CSF或G-CSF變異體涵蓋所有天然存在之G-CSF 變異體、以及來源於其之藉由重組DNA技術修飾之G-CSF 蛋白,尤其除G-CSF部分外亦含有其他蛋白質序列之融合 蛋白。就此含義而言,尤其較佳為在位置1具有N端Met殘 基之G-CSF突變蛋白(mutein),其適於在原核細胞中表 現。可根據WOA-91/11520製備之不含曱硫胺酸之重組G-CSF變異體同樣適合,該專利以全文引用的方式併入本文 中。術語「G-CSF變異體」應理解為包含一或多個胺基酸 可缺失或經其他胺基酸置換之彼等G-CSF分子,其具有G-CSF之基本性質,特定言之能夠調動大量保留之骨髓細 胞。適合G-CSF突變蛋白描述於例如EP0,456,200中,該專 利以全文引用的方式併入本文中。 在一些實施例中,如本文揭示之方法及組合物中可使用 熟習此項技術者所知之任何市售形式之G-CSF,例如(但不 限於)Neupogen®(F. Hoffmann-La Roche Ltd., Basel, Switzerland)及其變異體。 GM-CSF : 根據本發明,可使用利用重組方法所製備之GM-CSF(粒 152716.doc -45- 201130977 細胞-巨噬細胞群落刺激因子)及其變異體。根據本發明之 術語GM-CSF或GM-CSF變異體涵蓋所有天然存在之GM-CSF變異體、以及來源於其之藉由重組DNA技術修飾之 GM-CSF蛋白,尤其除GM-CSF部分外亦含有其他蛋白質序 列之融合蛋白。就此含義而言,尤其較佳為在位置1具有N 端Met殘基之GM-CSF突變蛋白,其適於在原核細胞中表 現。可根據WOA-91/11520製備之不含甲硫胺酸之重組GM-CSF變異體同樣適合,該專利以全文引用的方式併入本文 中。術語「GM-CSF變異體」應理解為包含一或多個胺基 酸可缺失或經其他胺基酸置換之彼等G-CSF分子,其具有 GM-CSF之基本性質,特定言之能夠調動大量保留之骨髓 細胞。適合GM-CSF突變蛋白描述於例如ΕΡ0,456,200中, 該專利以全文引用的方式併入本文中。 在4種『粒細胞-巨噬細胞』CSF中,GM-CSF最先經分離 及表徵。GM-CSF顯示可誘導含有粒細胞及巨噬細胞祖細 胞之鼠類骨髓源性或脾源性造血細胞增殖,從而產生主要 含有粒細胞及巨噬細胞前驅體之群落。就此而言,GM-CSF似乎與隨後表徵之IL-3具有共同生物性質。然而,更 近的研究表明相較於IL-3,GM-CSF對『更晚期』多能細 胞起作用。此外,GM-CSF刺激紅血球系及巨核細胞前驅 體增殖之活性似乎小於IL-3。然而,與IL-3類似,GM-CSF 顯示可對粒細胞及巨噬細胞譜系之成熟細胞具有活性。 不希望受理論束缚,GM-CSF(粒細胞-巨噬細胞群落刺 激因子)直接且選擇性地對粒細胞/巨噬細胞祖細胞起作用 152716.doc -46- 201130977 以刺激屬於此等譜系之細胞(例如嗜中性白血球、嗜伊紅 血球、巨噬細胞)的活體外生長及分化^亦已證明重組GM-CSF之此等多效性活性。除調控骨髓譜系之祖細胞/前驅細 胞之增殖及分化之外,GM-CSF亦已顯示可活化成熟骨髓 細胞類型之功能。舉例而言,已發現GM-CSF可誘導巨噬 細胞對抗惡性黑素瘤細胞株A375之殺腫瘤活性。IFNy亦可 充當巨噬細胞活化因子,但與GM-CSF相反,其需要另一 種二級刺激物(例如細菌LPS)以誘發殺腫瘤活性》此外, GM-CSF活化巨嗔細胞以抑制克氏錐蟲(Trypanosoma cruzi)(—種單細胞寄生蟲,其為卻格司氏病(Chagas disease)或美洲錐蟲病(American trypanosomiasis)之發病原 因)複製且增強呼吸氧化過程。此外,HIV-1在人類單核細 胞細胞株U937中之複製已顯示可被GM-CSF中度抑制,且 可被GM-CSF與IFNY之組合更有效抑制。 在嗜中性白血球及嗜伊紅血球中,GM-CSF刺激許多功 能。特定言之,GM-CSF增強嗜中性白血球對細菌及酵母 之吞噬作用。經純化之重組人類GM-CSF亦已顯示可增強 嗜中性白血球及嗜伊紅血球對抗經抗體包覆之目標細胞的 細胞毒性活性。 已報導當小鼠用重組鼠類GM-CSF重複腹膜内注射時, 腹膜巨嗔細胞、粒細胞(嗜中性白血球及嗜伊紅血球)之數 目及功能活性快速且持續增加且循環單核細胞之數目亦增 加。(GM-CSF通常花費約兩週以起作用)。將重組人類gm-CSF注射於AIDS患者及非人類靈長類動物中之後,亦已觀 152716.doc •47- 201130977 測到嗜中性白血球、嗜伊紅企球及單核細胞數目之明顯增 加。然而,可能存在與GM_CSF療法相關之併發症。 Metcalf及同事已證明,含有組成性表現之鼠類gm csf* 因的轉殖基因小鼠在出生後不久即於各種組織(包括晶狀 體、視網膜及橫紋肌)中出現由活化之巨嗤細胞浸潤所引 起的病理性病變。(活化之巨噬細胞已知會產生許多可誘 發組織損傷之發炎性介體,包括細胞激素,諸如TNFa及 IL-1) 〇 與GM-CSF之生長刺激作用相反,GM-CSF可充當分化因 子。其對例如成熟巨噬細胞及嗜中性白血球之作用可視為 其分化誘導能力的結果《—種限制腫瘤細胞增殖之方式為 使生長因子驅動之自我更新與生長因子誘導之分化脫離聯 繫。換§之,『分化』腫瘤細胞越多,其增殖能力越小。 就此而言,GM-CSF已顯示可誘導骨髓白血病細胞株HL6〇 分化且抑制其自我更新。然而,在若干其他研究中,GM_ CSF刺激HL60細胞增殖。可藉由量測各種質膜相關抗原 (例如CD14(單核細胞/巨噬細胞標記)及cd57(NK細胞標 記))之表現來監測分化。 在些貫施例中’如本文揭示之方法及組合物中可使用 熟習此項技術者所知之任何市售形式之GM-CSF,例如(但 不限於)Leucomax®(Schering Pl〇Ugh-Sandoz Pharma Ltd”Covered in the term individual. W The term "tissue" refers to a group or layer resembling a shell-like cell, and a hill performs some special functions. The source or characteristic characteristics of the peppers of the peppers and the scorpion. The term "agent" as used herein means any such as, but not limited to, small molecules, sputum sub-substances, Pharmacy I can be used to sip a cup of syrup. The agent "T is any chemical, f synthetic and naturally occurring protein and non-protein entities: in = not limited to" «祖 in some embodiments, 152716.doc •37- 201130977 Pharmacy is a nucleic acid; nucleic acid analog; protein; antibody; peptide; aptamer; oligomer of nucleic acid, amino acid or carbohydrate, including but not limited to protein, oligonucleotide, ribonuclease , DNase, glycoprotein, siRNA, lipoprotein, aptamer and modified forms and combinations thereof. In certain embodiments, the agent is a small molecule having a chemical moiety. For example, the chemical moiety includes unsubstituted or substituted alkyl, aromatic or heterocyclic moieties' including maCrolide, 丨ept〇mycin and related natural products or the like. Things. The compounds are known to have a desired activity and/or property, or a pool of different compounds. As used herein, the term "small molecule" refers to a chemical agent, which may include, but is not limited to, peptides, peptidomimetics, amino acids, amino acid analogs, polynucleotides, polynucleotide analogs, Aptamers, nucleotides, nucleotide analogs, organic or inorganic compounds having a molecular weight of less than about 10,000 grams per mole (for example including heteroorganic compounds and organometallic compounds), organics having a molecular weight of less than about 5,000 agram per mole Inorganic compound, organic or inorganic compound having a molecular weight of less than about 1, (10) gramm/mole, an organic or inorganic compound having a molecular weight of less than about 5 gram/mole, and salts, esters and pharmaceutically acceptable salts of such compounds Other forms. § "Disease" or "condition" is used interchangeably herein and refers to any change in the state of the body or organs that interrupts or interferes with the function of the lamp and/or causes symptoms of the patient or the person who is in contact with the individual. , such as discomfort, dysfunction, pain, or even death. A disease or condition can also mean heat, discomfort, disorders, confusion, abnormalities. Nausea, illness, discomfort, microspasm or disease. 1527l6.doc -38 - 201130977 The term "pathology" as used herein refers to a condition causing a disease or condition, such as a change in the structure and function of a cell, tissue or organ. For example, 'pathology can be related to a specific nucleic acid sequence or a "pathological nucleic acid", which refers to a nucleic acid sequence that completely or partially causes pathology, for example, 5' pathological nucleic acid can be encoded to have a specific pathology or pathology. A nucleic acid sequence of a gene associated with a mutation or polymorphism. The pathology may be associated with the performance of pathological proteins or pathological polypeptides that completely or partially cause pathology associated with a particular disease or condition. In another embodiment, the pathology is associated, for example, with other factors, such as ischemia and the like. As used herein, the term "treating" includes alleviating or alleviating at least one of the adverse effects or symptoms of a condition, disease or condition. As used herein, the phrase "parenteral administration" means a mode of administration other than enteral and topical administration, usually injection, and includes (but is not limited to) intravenous, intramuscular, intraarterial, intrathecal, intraventricular. , intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subepidermal, intra-articular, subcapsular, subarachnoid, intraspinal, cerebrospinal, and intrasternal injections and infusions. As used herein, the phrase "systemic administration" or "peripheral administration" means PB-derived MSC or BM-derived MSC or its differentiated progeny and/or its progeny and/or compounds and/or other substances to Administration into the body of the animal and thereby undergoing metabolism and other similar processes (eg, subcutaneous or intravenous administration) rather than directly into the individual. The phrase "pharmaceutically acceptable" is used herein to mean that it is suitable for contact with humans and animal tissues in a reasonable medical judgment (4) without excessive toxicity, irritation, allergic reaction or other problems or complications and reasonable benefits/ Risk 152716.doc •39- 201130977 The proportion of their compounds, substances, compositions and/or dosage forms. The phrase "pharmaceutically acceptable carrier" as used herein means pharmaceutically acceptable in connection with the delivery or delivery of a target agent to another organ or part of the body in an organ or part of the body. A substance, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation. The term "drug" or "compound" as used herein refers to a chemical entity or biological product, or a combination of chemical entities or biological products, administered to an individual to treat or prevent or control a disease or condition. Chemical entities or biological products are preferably, but not necessarily, low molecular weight compounds, but may also be larger compounds such as nucleic acids, amino acids, or oligomers of carbohydrates including, but not limited to, proteins, oligos Glycosylates, ribonucleases, DNases, glycoproteins, siRNAs, lipoproteins, aptamers, and modified forms and combinations thereof. The term "transplantation" as used herein refers to the administration of new cells (eg, PB-derived MSCs or BM-derived MSCs or their differentiated progeny) or tissues (such as differentiated cells produced by pb-derived MSCs or BM-derived MSCs) or The organ is introduced into the host (i.e., the recipient of the graft or the individual of the graft). The term "modulation j" is used in accordance with its usage in the art, for example to cause or promote qualitative or quantitative changes, changes or modifications of related processes, paths or phenomena. This change may be, without limitation, a process, path or phenomenon. An increase, decrease, or change in the relative strength or activity of a different component or branch. A "regulator" is an agent that causes or promotes a change, change, or modification in the nature or quantity of a related process, pathway, or phenomenon. I52716.doc -40· 201130977 The term "reduction", "reduction" or "inhibition" is used herein to mean a statistically significant reduction. However, for the avoidance of doubt, "decreasing; or "reducing" or "suppressing" means reducing the reference content to two wrists, for example, reducing at least about 鸠, or at least about 鸠, or to / 娜, or at least about 乡, or at least about 6Q%, or at least %% / as for the spring. Or up to and including 1% reduction (e.g., comparable, °, no content), or any decrease from 1 to 100% compared to the reference content. 1 in 10 - the terms "increase" or "enhancement" or "activation" are generally used herein to mean "the stomach is increased by a statistically significant amount; to avoid any doubt, the term "doing" or "35" or "activation" By means of an increase of at least 0, for example at least about 20%, or at least about 30%, or at least about peach, or at least (four)%, or at least about 峨, or at least about Cong, or at least = 〇%, compared to the reference content. Or at least about 90% or up to and including just % increase or as compared to > the test content is between 1 (M00% of any increase, or at least about 2 times the reference content' or at least about 3 times Or at least about 4 times, or at least about $ times or at least about H) times or any increase between 2 times and 1G times or more. J is "statistically significant" and "significant" means statistically significant and is less than two standard deviations (2SD) lower than the standard or lower concentration of the marker. This ΓΓ refers to statistical evidence that confirms the difference. It is defined as the decision to exclude the null hypothesis when the illusion is false - the truth is true. The term "substantially" as used herein means 6 〇 /. Or preferably at least about or at least about 嶋' or at least 约娜, to 152716.doc •41- 201130977 less than about 95%, at least about 97% or at least about 99% or more, or between 70% The ratio of any integer between 1 and 〇〇%. As used in the specification and the appended claims, the singular forms " By way of example, reference to "the method" includes one or more methods and/or types of steps which are described herein and/or which are apparent to those skilled in the art in reading the invention, and so forth. The above detailed description and the following examples are to be construed as illustrative and not restrictive of the invention. Retouching will be apparent to those skilled in the art. In addition, all patents, specific disclosures, and publications identified herein are hereby expressly incorporated by reference for the purposes of the disclosure of the disclosure of the disclosure of ^ These publications are for disclosure only, prior to the filing date of the request. In this regard, it is not to be understood that the inventors have the right to advance the present invention for the prior invention or for any other reason. All representations of all statements or content in the sequel to these documents are based on the information available to the applicant and do not constitute any recognition as to the correctness of the date or content of such documents. MSC mobilizers, in aspects of the invention, provide a method of treatment comprising administering to a mammal (eg, a human) an effective amount of a mobilizing agent, such as a certain form of G CSF or GM-CSF, and its medicinal Acceptable salts are used to enhance (i), stimulate MSC production/release, as measured by phenotypic, physical or chemical properties or by any other feature used by the skilled artisan to identify MSC-type cells. In one embodiment, the peak number of circulating MSCs is obtained after about 2 injections per day of administration of a mobilizer such as GM-CSF or G-CSF. The peak of circulating MSCs in the peripheral blood decreased after at least 4 daily injections. Any combination of interstitial or interstitial agents can be administered to an individual at a defined time to optimize mobilization of the MSC into the peripheral blood. In some embodiments, the MSC mobilizing agent is GM-CSF or G-CSF (granulocyte community stimulating factor). In some embodiments, the individual may be administered a combination of other agents, including, but not limited to, G-CSF, GM-CSF, Flt-3 ligand, stem cell factor (eg, but not limited to) a single mobilizer or a combination of such mobilizers ( SCF), dexamethazone, CXCR4 receptor inhibitor, interleukin-1 (IL-1), interleukin-3 (IL-3), and Diniplestim (IL-3) Agonist), Leridistim (IL-3 agonist-G-CSF fungus), progenipoietin-l (Flt-3 ligand-G-CSF) Chimeric molecule), peg-filgrastim (NEULASTATM), interleukin-8 (IL-8), PIXY-321 (GM-CSF/IL-3 fusion protein), macrophage Cellular inflammatory proteins, stem cell factor (SCF), thrombopoietin, and growth-related oncogenes. Other mobilizing agents include, for example, but are not limited to, CXCR4 inhibitors such as AMD3100, ALX40-4C, T22, T134, T140, and TAK-779, which are disclosed in U.S. Patent No. 7,169,750, hereby incorporated hereinby entirety in. In some embodiments, such as G-CSF or GM-CSF, or interleukin-17 (IL-17), AMD3100, cyclophosphamide (Cy), and European paclitaxel (DXT) and 152716.doc • 43- 201130977 The combined mobilizer can be administered to the individual or ex vivo to the peripheral blood sample to stimulate the PB-derived MSC. In some embodiments, a mobilizing agent such as G-CSF or GM-CSF may be administered to an individual or ex vivo to a peripheral blood sample in combination with other therapeutically or nutritionally active ingredients, such as antibiotics. , vitamins, herbal extracts, anti-inflammatory agents, glucose, antipyretics, analgesics, granulocyte-macrophage community stimulating factor (GM_CSF), interleukin-1 (IL1), interleukin-3 (IL-3) , interleukin-8 (IL-8), PIXY-321 (GM-CSF/IL-3 fusion protein), macrophage inflammatory protein, stem cell factor, thrombopoietin and analogs thereof. A mobilized peripheral blood stem cell cluster (PBSC) produced using a mobilizer such as G-CSF or GM-CSF can be transplanted into, for example, a cancer patient. Such clusters may be the total population of purified HSCs obtained from mobilized white blood cells or from mobilized peripheral blood. Hsc is able to reorganize the hematopoietic system of organisms, which requires self-renewal of HSCs and differentiation into cells of various hematopoietic lineages. The cD34 is labeled as unique to the HSC. Other cells mobilized using such mobilizers include polymorphonuclear leukocytes (cells that mediate inflammation and pathogen clearance), mononuclear white blood cells (lymphocytes and monocytes), and red blood cell progenitor cells (red blood cells). The mobilization of CD34+ HSC is a rapid amplification clinical technique for obtaining materials for autologous or allogeneic hematopoietic transplantation. In addition, the mobilization of polymorphonuclear leukocytes is an important aid to heavy chemotherapy to maintain an innate defense mechanism. Currently, both methods rely on expensive growth factors (❻/" or GM_CSF) that cause bone pain and have unknown side effects to normal donors. It takes about 4 weeks to process. To collect enough material for transplantation. After growth factor treatment, CD34+ cells reach a maximum of 2-4% in blood. G-CSF: In some embodiments, for G- in a method as disclosed herein CSF can be prepared using recombinant methods and variants of G-CSF can be used. The term G-CSF or G-CSF variant according to the invention encompasses all naturally occurring G-CSF variants, as well as recombinant DNA derived therefrom The technically modified G-CSF protein, in particular, a fusion protein of other protein sequences in addition to the G-CSF moiety. In this sense, a G-CSF mutant protein having an N-terminal Met residue at position 1 is particularly preferred (mutein). It is suitable for expression in prokaryotic cells. Recombinant G-CSF variants which are prepared according to WOA-91/11520 and which are free of guanidine thioglycol are also suitable, which is incorporated herein by reference in its entirety. G-CSF variant Comprising one or more amino acids may be deleted or replaced by other amino acids of their G-CSF molecule having the essential nature of G-CSF, a large number of specific words can mobilize bone marrow cells reservations. Suitable G-CSF muteins are described, for example, in EP 0,456,200, the disclosure of which is incorporated herein in its entirety. In some embodiments, any commercially available form of G-CSF known to those skilled in the art can be used, such as, but not limited to, Neupogen® (F. Hoffmann-La Roche Ltd), as disclosed herein. ., Basel, Switzerland) and its variants. GM-CSF: According to the present invention, GM-CSF (granule 152716.doc -45-201130977 cell-macrophage colony stimulating factor) prepared by a recombinant method and a variant thereof can be used. The term GM-CSF or GM-CSF variant according to the invention encompasses all naturally occurring GM-CSF variants, as well as GM-CSF proteins derived therefrom modified by recombinant DNA techniques, especially in addition to the GM-CSF moiety. A fusion protein containing other protein sequences. In this sense, a GM-CSF mutein having an N-terminal Met residue at position 1 is particularly preferred, which is suitable for expression in prokaryotic cells. Also suitable are recombinant GM-CSF variants which are free of methionine prepared according to WOA-91/11520, which is incorporated herein by reference in its entirety. The term "GM-CSF variant" is understood to include those G-CSF molecules which may be deficient in one or more amino acids or substituted by other amino acids, which have the basic properties of GM-CSF, specifically capable of mobilizing A large number of preserved bone marrow cells. Suitable GM-CSF muteins are described, for example, in U.S. Patent No. 0,456,200, the disclosure of which is incorporated herein in its entirety. Among the four "granulocyte-macrophage" CSFs, GM-CSF was first isolated and characterized. GM-CSF has been shown to induce proliferation of murine bone marrow-derived or spleen-derived hematopoietic cells containing granulocytes and macrophage progenitor cells, resulting in a community of granulocyte- and macrophage precursors. In this regard, GM-CSF appears to have a common biological property with the subsequently characterized IL-3. However, recent studies have shown that GM-CSF acts on "more advanced" pluripotent cells compared to IL-3. In addition, GM-CSF appears to stimulate the proliferation of red blood cell lines and megakaryocyte precursors to be less active than IL-3. However, similar to IL-3, GM-CSF has been shown to be active against mature cells of the granulocyte and macrophage lineages. Without wishing to be bound by theory, GM-CSF (granulocyte-macrophage colony stimulating factor) acts directly and selectively on granulocyte/macrophage progenitor cells 152716.doc -46- 201130977 to stimulate belonging to these lineages In vitro growth and differentiation of cells (e.g., neutrophils, eosinophils, macrophages) have also demonstrated such pleiotropic activities of recombinant GM-CSF. In addition to regulating the proliferation and differentiation of progenitor/precursor cells of the myeloid lineage, GM-CSF has also been shown to activate the function of mature bone marrow cell types. For example, GM-CSF has been found to induce tumorigenic activity of macrophages against malignant melanoma cell line A375. IFNy can also act as a macrophage activating factor, but in contrast to GM-CSF, it requires another secondary stimulator (such as bacterial LPS) to induce tumoricidal activity. In addition, GM-CSF activates giant scorpion cells to inhibit Krebs cones. Trypanosoma cruzi (a single-celled parasite that is responsible for the onset of Chagas disease or American trypanosomiasis) replicates and enhances the respiratory oxidation process. Furthermore, replication of HIV-1 in human monocyte cell line U937 has been shown to be moderately inhibited by GM-CSF and is more effectively inhibited by the combination of GM-CSF and IFNY. In neutrophils and eosinophils, GM-CSF stimulates many functions. In particular, GM-CSF enhances the phagocytosis of neutrophils to bacteria and yeast. Purified recombinant human GM-CSF has also been shown to enhance the cytotoxic activity of neutrophils and eosinophils against antibody-coated target cells. It has been reported that when mice are repeatedly intraperitoneally injected with recombinant murine GM-CSF, the number and functional activity of peritoneal giant sputum cells, granulocytes (neutrophils and eosinophils) are rapidly and continuously increased and circulating monocytes are The number has also increased. (GM-CSF usually takes about two weeks to work). After injection of recombinant human gm-CSF into AIDS patients and non-human primates, it has also been observed 152716.doc •47- 201130977 A significant increase in the number of neutrophils, eosinophils and monocytes was detected. . However, there may be complications associated with GM_CSF therapy. Metcalf and colleagues have shown that transgenic mice containing constitutively expressed murine gm csf* are caused by activated giant cell infiltration in various tissues (including lens, retina, and striated muscle) shortly after birth. Pathological lesions. (Activated macrophages are known to produce many inflammatory mediators that can induce tissue damage, including cytokines such as TNFa and IL-1.) 相反 In contrast to the growth stimulating effects of GM-CSF, GM-CSF acts as a differentiation factor. Its effects on, for example, mature macrophages and neutrophils can be seen as a result of their differentiation-inducing ability. A way to limit tumor cell proliferation is to link growth factor-driven self-renewal with growth factor-induced differentiation. In other words, the more "differentiated" tumor cells, the smaller their proliferation ability. In this regard, GM-CSF has been shown to induce differentiation of myeloid leukemia cell line HL6 and inhibit self-renewal. However, in several other studies, GM_CSF stimulated HL60 cell proliferation. Differentiation can be monitored by measuring the performance of various plasma membrane associated antigens such as CD14 (monocyte/macrophage marker) and cd57 (NK cell marker). In some embodiments, any commercially available form of GM-CSF known to those skilled in the art, such as, but not limited to, Leucomax® (Schering Pl〇 Ugh-Sandoz) may be used in the methods and compositions disclosed herein. Pharma Ltd"
Basel, Switzerland)及其變異體。 投藥劑董、持續期、頻率、途徑(腹膜内(i.p>、皮下(sc)) G-CSF或GM-CSF之劑量可視各種因素(諸如用藥方式、 152716.doc -48· 201130977 物種、年齡或個體狀況)而定。在一些實施例中,皮下 (S.C.)施用G-CSF或GM-CSF的劑量為每天約100至700 pg之 G-CSF或GM-CSF。歷時至少2天、或至少3天且至多4天每 天至少一次投與G-CSF或GM-CSF。 G-CSF或GM-CSF之不同劑量包括(但不限於)每天約至少 100 pg、或至少約200 pg、或至少約300 pg、或至少約400 pg、或至少約500 pg、或至少約600 pg、或至少約700 pg、或700 pg以上G-CSF或GM-CSF。在一些實施例中,劑 量不超過每天1000 pg,歷時2天。在一些實施例中,G-CSF或GM-CSF之劑量為每天約450 pg,為期至少1天但小 於4天《在一些實施例中,G-CSF或GM-CSF之劑量為每天 約400 pg或每天約500 pg,為期至少1天但小於4天。在一 些實施例中,G-CSF之給藥持續期為小於5天,且因為此 等劑量已與嚴重副作用相關聯,所以較佳為每天小於1 〇 pg/kg、或每天小於7.5 pg/kg,該等副作用包括脾長度增 加、頭痛、噁心 '疲勞及骨疼痛,且在一些情況下包括自 發性脾破裂(參見以下評述:Cashen等人,Bone marrow transplantation, 2007; 39; 577-588 及 Pusic 等人,Current Pharmaceutical Design,2008; 14; 1950-1961,其以全文引 用的方式併入本文中)。 如本文所揭示,本發明人已證明,每天450 pg之劑量在 皮下投藥至少1天之後得到PB源性MSC的大部分調動。另 外,熟習此項技術者顯而易知,可嘗試增加或減少G-CSF 或GM-CSF之劑量以找到針對各用途之最佳劑量》此可簡 152716.doc -49- 201130977 單地如下進行:向個體投與遞增劑量之g_CSF4Gmcsf 且分析血液樣品,例如如本文所述獲取之血液樣品。此 外,視欲調動之細胞而定,可以可能高於或低於每天45〇 gg之最佳濃度每小時(或以其他間隔時間)輸注g_csf或 GM-CSF,而非依序增加基於每曰之劑量。亦可檢查投與 G-CSF或GM-CSF之不同頻率及持續期。 G-CSF或GM-CSF之適合劑量範圍視此等考量而變化, 但一般而言,化合物的投與劑量範圍為每天約1〇〇盹、或 至少約200 pg、或至少約300 、或至少約4〇〇叫、或至 少約500 、或至少約600 pg、或至少約7〇〇吨、或7〇〇 pg以上之G_CSF或GM-CSF。相較於例如靜脈内投藥,當 化合物經口或經皮投與時,劑量可較高。 對於如本文所揭示向個體投與g_CSF4GM_csf,可使 用此項技術中熟知之普遍瞭解調配技術調配G_CSF或GM_ CSF以投與動物個體,諸如人類.適用於特定投藥方式之 調配物可見於Remingt〇n,s Pharmaceuticai Sciences,最新 版,Mack Publishing Company,Easton,Pa 中。 在一些實施例中,使用標準投藥形式(其中注射溶液較 佳)向諸如人類之個體投與G_CSF4GM-CSF。水較佳用作 注射介質,其包括注射溶液中常見之佐劑,諸如穩定劑、 增溶劑及緩衝劑。舉例而言,此等佐劑為酒石酸鹽及檸檬 酸鹽緩衝劑;乙醇;錯合劑,諸如乙二胺四乙酸及其無毒 鹽;用於控制黏度之高分子量聚合物,諸如液體聚氧化乙 烯。注射溶液之液體媒劑必須無菌且較佳裝填入安瓿中。 152716.doc .50· 201130977 G-CSF或GM-CSF較佳藉由注射,最佳藉由靜脈内注 射’但亦藉由皮下或腹膜内注射及其類似方式投與諸如人 類之個體。其他非經腸投藥途徑包括肌肉内及關節内注 射。對於靜脈内或非經腸投藥,G_CSF或GM_CSF必要時 可用賦形劑調配成適合液體形式。G_CSF4GM_CSF可於 脂質體或其他適合載劑中投與個體,冑如人類個體。對於 靜脈内注射’使用諸如亨克氏溶液(Hank,s s〇luti〇n)之標準 製劑使溶液具有等張性。 除注射之外,亦可使用其他投藥途徑。在一些實施例 中,待投與個體(諸如人類個體)之G_CSF*GM_CSF可調配 成錠劑、膠囊劑、糖漿、散劑或適於經口投與之其他形 式。藉由使用適合賦形劑,g_CSF4GM_csf可使用栓劑 或鼻内喷霧劑經由黏膜投與。經皮投藥亦可藉由使用適合 滲透劑且控制釋放速率實現。 除靜脈内輸注外,G-CSF或GM-CSF之不同投藥途徑亦 有效。舉例而言,皮下或腹膜内所置放之G_CSF或gm· CSF之「儲槽(depot)」可提供長期連續輸注且為提供g_ CSF或GM-CSF之便利且有效方法。若皮下或腹膜内投與 G-CSF或GM-CSF,則可達成例如連續輪注且隨時間監測 個體。或者,可經口投與G-CSF或GM-CSF。本發明包括 藉由此等手段及如由熟習此項技術者所瞭解之所有其他手 段投與G-CSF或GM-CSF。 不同的G-CSF或GM-CSF投藥方案(包括不同的g_csf或 GM-CSF劑量、G-CSF或GM-CSF投藥途徑及持續期)將調 152716.doc •51· 201130977 動不同幹細胞群,諸如MSC。如上所論述,本發明人已驚 人地發現,用於最佳化調動]^8(:的(}]^_(:汀及(3_(:§17投藥 方案不同於習知用於調動HSC之方案。因此,可針對特定 所要應用最佳化方案:在不同條件下投與g_csf或 CSF ’接著根據如本文揭示之方法監測血液中所要幹細胞 亞群(諸如MSC或造血細胞)之輸出。因為如本文實例中所 指示,在投與調動劑之後的時段,不同細胞類型會依序出 現,所以應監測輸注之後達成最佳化回收或誘導血液中之 特定細胞群的時間。 所選調配物及投藥途徑應適合個別個體、待治療之個體 病狀之性質、及一般而言主治從醫者之判斷。 在一些實施例中’ G-CSF或GM-CSF可以單次快速給 藥、隨時間給藥(如靜脈内或經皮投藥)或多次給藥投與個 體,諸如人類個體。 除直接投與個體外,G-CSF或GM-CSF可用於離體處理 方案中以自個體之周邊血樣品中分離MSC,其可用於製備 MSC培養物,該等培養物可視情況經超低溫保存及儲存且 隨後在需要MSC時用於個體之基於細胞的再生治療。在— 些實施例中’ MSC之此離體產生可用於分離並收集自體 MSC ’該等自體MSC自周邊血或在一些情況下自匹配供者 之同種異體移植物收集。單獨或與其他藥劑(諸如巨嗔細 胞發炎性蛋白)組合離體接觸周邊血的調動劑之濃度為常 規最佳化的問題。 藉由投與G-CSF或GM-CSF ’可觀測到脾中粒細胞大量 152716.doc -52· 201130977 生成及脾重量顯著增加,根據Bungart等人,Brit j 76, 174-179, 1990,此可歸因於幹細胞調動或MSC調動。 在一些實施例中,用於如本文揭示之方法之周邊血樣品 係自哺乳動物個體’諸如人類個體獲得。在一些實施例 中,人類個體先前已投與調動劑以增加周邊血中循環幹細 胞之產量或數目。任何調動劑皆可諸如根據此項技術中通 常所知之方法,且包括例如(但不限於)如美國專利申請案 6,261,549及美國專利申請案2009/0155225中揭示之方法加 以使用’該等專利申請案以全文引用之方式併入本文中。 在一些實施例中,諸如G-CSF或GM-CSF之調動劑可與 任何一或多種其他調動劑混合投與個體或離體投與周邊血 樣品’該等其他調動劑例如為選自由以下組成之群之藥 劑:介白素-17(IL-17)、AMD3100、環磷醯胺(Cy)及歐洲 紫杉醇(DXT)及其組合。 在一些實施例中,諸如G-CSF或GM-CSF之調動劑可與 治療上或營養上適用之其他活性成分混合投與個體或離體 投與周邊血樣品,該等其他活性成分諸如抗生素、維生 素、草本萃取物、消炎劑、葡萄糖、解熱劑、止痛劑、粒 細胞-巨嗤細胞群落刺激因子(GM-CSF)、介白素-1 (IL-1)、 介白素-3(IL-3)、介白素-8(IL-8)、PIXY-321(GM-CSF/IL-3 融合蛋白)、巨嗟細胞發炎性蛋白、幹細胞因子、血小板 生成素及其類似物。 任何個體皆可使用如本文揭示之方法進行MSC調動,且 可自得自任何個體(諸如人類個體,包括成人或非新生兒 152716.doc •53· 201130977 兒童)之周邊血調動並收集MSC »此外,MSC可以-或多 個收集步驟或收集週期收集。舉例而言,彳自個體之經調 動周,血收集(例如使用血液析離術)Μ%至少2次、至少3 或至/ 5-人《在各收集步驟期間,自每公斤體重個體 所收集之總有核細胞的數目可為—百萬(ΐχΐ()6)或一百萬以 上(例如卜1〇7、1><1〇8、1><1〇9、1><1〇1〇、1><1〇11、1><1〇12、 1X10:: 1X101:、I,15、、1X10-、1X101«、 1〇 1X10 )。在較佳實施例中,視供者個體之體重及 年齡而在單批收集中所收集之析離產物(其用於Μ% 與非MSC之分離)中的細胞數目可等於或大於ΐχΐ〇丨5個總有 核細胞(TNC),或至少約 1χ1〇Μ、ΐχΐ〇13、ΐχΐ〇丨2、 lxio6 、 lxio5個 lxlO1 丨、1x10 丨。、1χ10、1χ1〇8、1χ1〇7 總有核細胞。 視隋形及可使用如本文揭示之方法自任何人類個體供者 獲得之MSCM,較佳可在人類個體供者處於「成人」或 成熟j年齡(除非另外在特定情境中使用以獲得不同含 義’否則如本文所用之術語「成人」係指且包括成人及非 新生兒)及/或某-最小體重時自其收集幹細&。舉例而 吕,當個體在10至200 kg之範圍内(根據本發明之一實施 例)或此範圍内之任何範圍内時,諸如2〇至4〇 kg内時可 收集MSC。或者或此外,根據本發明之一實施例,可要求 個體具有2-80歲範圍(例如2_1〇、1〇_15、1218、16 2()、 20-26 、 26-30 、 30-35 、 30-40 、 40-45 、 40-50 、 55-60 、 60- 65、60_70及70-80歲)内之某一年齡。 152716.doc •54· 201130977 MSC與周邊金中其他幹細胞群之分離 自周邊血獲得並分離PB源性MSC及BM源性MSC之方法 在此項技術+已熟知,且包括白血球企液析離術、密度梯 度部分分離、免疫選擇及差異黏著分離。實質上純MSC群 可於培養基(諸如化學上限定之無血清培養基或完全培養 基(諸如DMEM或含有1 g血清之DMEM))中收集。適合的化 學上限定之無血清培養基描述於1995年6月$日申請之美國 專利第08M64,599號中,且「完全培養基」描述於1996年1 月23日頒予之美國專利第5 486,359號中,該等專利以引用 的方式併入本文中。 本發明之一態樣為使用離心淘洗自經調動之周邊血中分 離MSC。不希望受理論束縛,在一種淘洗方法中,將周邊 血樣品引入位於旋轉離心機上之漏斗狀分離室中。接著將 液體淘洗緩衝液流或低密度液流引入含有周邊血樣品之腔 至中。當通過腔室之液體淘洗緩衝溶液之流速增加(通常 以逐步方式)時’液體將尺寸較小、沈降較慢之細胞掠向 腔室内之淘洗邊界’而沈降較快的較大細胞遷移至離心力 與沈降(拖曳(drag))力平衡之室内區域。因此,當周邊血 (例如經調動之周邊血)包含許多不同幹細胞群時,本發明 之一態樣係關於使用淘洗法,將存在於周邊血中之此等不 同幹細胞群分離成不同群體,其中分離出較小幹細胞與尺 寸較大之幹細胞(fractionate) 〇 在本發明之一些態樣中’任何淘洗裝置皆可用於獲得PB 源性MSC及BM源性MSC且使其與周邊血中其他幹細胞群 152716.doc -55- 201130977 分離。在一些實施例中,淘洗裝置係使用析離產物(其中 血小板及血球返回至個體供者中),且剩餘周邊血產物為 析離產物,其以某一流速經淘選,以將周邊血樣品部分分 離成包含MSC群之部分《在一些實施例中,市售淘洗裝置 (例如由Gambro BCT,Inc製造之ELUTRA®離心機)可用於 自周邊血中分離 MSC » ELUTRA CELL SEPARATION SYSTEM®能夠利用尺寸與密度,將經調動之周邊血分離 成包含MSC之部分,從而使細胞之富集、排除 (depletion)、濃縮及洗滌皆能夠在封閉式功能系統内進 行。ELUTRA CELL SEPARATION SYSTEM®能夠直接自 白血球析離產物富集幹細胞群,而無需抗體或在小於1小 時内進行預處理。Elutra Cell Separation System®係使用 逆流離心淘洗法,其中流體在離心場中流過細胞層,以分 離細胞群。 在一些實施例中,其他淘洗裝置(包括(但不限於)亦由 Gambro BTC Inc.製造之COBE® Spectra析離系統、Trima® 系統及Trima Accel®系統,以及用於分離血液組分之其他 市售淘洗裝置)可用於自周邊血中,分離MSC。 在一些實施例中,MSC群可使用細胞分離器(諸如 COBE® Spectra析離系統),自經調動之周邊血樣品獲得。 COBE® SPECTRA™離心機描述於美國專利4,425,172、 4,708,712及6,022,306中,該等專利以引用的方式併入本文 中。在該實施例中,將經調動之周邊血樣品(或自已根據 如本文揭示之方法投與調動劑之個體獲得的析離產物)引 152716.doc •56· 201130977 入細胞分離器(諸如C0BE 離系統)中,且視情況 添加抗凝血劑溶液至血液中,以阻止其在過程期間凝結。 血液/抗凝血劑混合物經過離心機循環,以將周邊血樣品 分離成MSC群,且自其他血液組分及血漿中分離單核細 胞。系統將分離之MSC抽入收集袋中儲存,而其他血液組 刀及血毁返回至患者中。所用全部管組(tubing set)及針皆 無菌’因此不存在疾病傳染風險。 可使用其他血液分離裝置,諸如描述於以下中之裝置: 1998年3月3日頒予之美國專利第5 722 926號;1999年9月 14曰頒予之美國專利第5 951 877號;2〇〇〇年4月25日頒予 之美國專利第6,053,856號;2002年1月1日頒予之美國專利 第6,3 34,842號;2004年7月1日申請之美國專利申請案第 1〇/884,877號;美國專利 7,201,848、美國專利 6 〇22 3〇6、 美國專利6,589,526、美國專利申請案2〇〇8/〇〇35585 ' US2008/03 18756及2009/0104626,此等美國專利及專利申 請案之每一者之全部揭示内容以引用的方式併入本文中。 本發明人已發現,宜調整血液析離術所用之血球比容範 圍以增強所收集之細胞群的性質。舉例而言,在一實施例 中,血球比容範圍經選擇可增加VSEL之比例。在另一實 施例中,血球比容範圍經選擇可增強所收集之VSEL的超 低溫保存’例如藉由使粒細胞之收集減至最少。在另一實 施例中,血球比容範圍經選擇可確保同時收集多個幹細胞 群,諸如VSEL及MSC。在一實施例中,血球比容範圍選 擇為2-3%。在另一實施例中’血球比容範圍選擇為3_ 152716.doc •57- 201130977 4%。Basel, Switzerland) and its variants. Dosing, duration, frequency, route (intraperitoneal (i.p>, subcutaneous (sc)) G-CSF or GM-CSF dose can be seen by various factors (such as medication, 152716.doc -48· 201130977 species, Depending on age or individual condition. In some embodiments, the dose of G-CSF or GM-CSF administered subcutaneously (SC) is about 100 to 700 pg of G-CSF or GM-CSF per day for at least 2 days, or G-CSF or GM-CSF is administered at least once a day for at least 3 days and up to 4 days. Different doses of G-CSF or GM-CSF include, but are not limited to, at least about 100 pg per day, or at least about 200 pg, or at least About 300 pg, or at least about 400 pg, or at least about 500 pg, or at least about 600 pg, or at least about 700 pg, or more than 700 pg of G-CSF or GM-CSF. In some embodiments, the dose does not exceed each day. 1000 pg for 2 days. In some embodiments, the dose of G-CSF or GM-CSF is about 450 pg per day for at least 1 day but less than 4 days. In some embodiments, G-CSF or GM-CSF The dose is about 400 pg per day or about 500 pg per day for at least 1 day but less than 4 days. In some embodiments, the duration of administration of G-CSF is less than 5 And because these doses have been associated with severe side effects, preferably less than 1 〇pg/kg per day, or less than 7.5 pg/kg per day, such side effects include increased spleen length, headache, nausea, fatigue, and bone pain. And in some cases including spontaneous spleen rupture (see review below: Cashen et al, Bone marrow transplantation, 2007; 39; 577-588 and Pusic et al, Current Pharmaceutical Design, 2008; 14; 1950-1961, full text The manner of reference is incorporated herein. As disclosed herein, the inventors have demonstrated that most of the mobilization of PB-derived MSCs is obtained after a subcutaneous administration of a dose of 450 pg per day for at least one day. However, it is easy to know that it is possible to increase or decrease the dose of G-CSF or GM-CSF to find the optimal dose for each use. This can be done as follows: 152716.doc -49- 201130977 g_CSF4Gmcsf and analyzes a blood sample, such as a blood sample obtained as described herein. Further, depending on the cell to be mobilized, it may be higher or lower than the optimal concentration of 45 gg per day per (Or at other intervals) g_csf infusion or GM-CSF, rather than sequentially increasing the dose on each of said. It is also possible to check the different frequencies and durations of G-CSF or GM-CSF. Suitable dosage ranges for G-CSF or GM-CSF will vary depending on such considerations, but in general, the compound will be administered in a dosage range of about 1 每天, or at least about 200 pg, or at least about 300, or at least About 4 〇〇, or at least about 500, or at least about 600 pg, or at least about 7 ton, or more than 7 〇〇 pg of G_CSF or GM-CSF. The dose may be higher when the compound is administered orally or transdermally, as compared to, for example, intravenous administration. For administration of g_CSF4GM_csf to an individual as disclosed herein, G_CSF or GM_CSF can be formulated for administration to an individual animal, such as a human, using a universal demodulation technique well known in the art. Formulations suitable for a particular mode of administration can be found in Remingt〇n , s Pharmaceuticai Sciences, latest edition, Mack Publishing Company, Easton, Pa. In some embodiments, G_CSF4GM-CSF is administered to an individual such as a human using a standard mode of administration wherein the injectable solution is preferred. Water is preferably used as an injection medium, which includes adjuvants commonly found in injectable solutions, such as stabilizers, solubilizers, and buffers. For example, such adjuvants are tartrate and citrate buffers; ethanol; complexing agents such as ethylenediaminetetraacetic acid and its non-toxic salts; high molecular weight polymers for controlling viscosity, such as liquid polyethylene oxide. The liquid vehicle for injecting the solution must be sterile and preferably filled into the ampoule. 152716.doc .50· 201130977 G-CSF or GM-CSF is preferably administered by injection, preferably by intravenous injection, but also by subcutaneous or intraperitoneal injection and the like, such as humans. Other parenteral routes of administration include intramuscular and intra-articular injections. For intravenous or parenteral administration, G_CSF or GM_CSF may be formulated into a suitable liquid form if necessary. G_CSF4GM_CSF can be administered to an individual, such as a human subject, in a liposome or other suitable carrier. The solution is made isotonic with respect to intravenous injection using standard formulations such as Henk's solution (Hank, s s〇luti〇n). In addition to injection, other routes of administration may be used. In some embodiments, the G_CSF*GM_CSF to be administered to an individual, such as a human individual, may be formulated as a lozenge, capsule, syrup, powder, or other form suitable for oral administration. g_CSF4GM_csf can be administered via the mucosa using a suppository or an intranasal spray by using a suitable excipient. Transdermal administration can also be achieved by using a suitable penetrant and controlling the release rate. In addition to intravenous infusion, different routes of administration of G-CSF or GM-CSF are also effective. For example, a "depot" of G_CSF or gm·CSF placed subcutaneously or intraperitoneally can provide a long-term continuous infusion and is a convenient and efficient method of providing g_CSF or GM-CSF. If G-CSF or GM-CSF is administered subcutaneously or intraperitoneally, for example, continuous rounds can be achieved and the individual monitored over time. Alternatively, G-CSF or GM-CSF can be administered orally. The present invention encompasses the administration of G-CSF or GM-CSF by such means and by all other means as would be appreciated by those skilled in the art. Different G-CSF or GM-CSF dosing regimens (including different g_csf or GM-CSF doses, G-CSF or GM-CSF route of administration and duration) will be adjusted to 152716.doc • 51· 201130977 to move different stem cell populations, such as MSC. As discussed above, the inventors have surprisingly found that the optimization of the transfer of ^8 (: (}) ^ _ (: Ting and (3_ (: § 17 dosing scheme is different from the conventional use for mobilizing HSC Thus, an optimization scheme can be applied for a particular application: g_csf or CSF is administered under different conditions. Then the output of a subset of the desired stem cells (such as MSCs or hematopoietic cells) in the blood is monitored according to the methods as disclosed herein. As indicated in the examples herein, different cell types will occur sequentially during the period after administration of the mobilizer, so the time to optimize recovery or induce a specific cell population in the blood after the infusion should be monitored. Selected formulations and administration The route should be suitable for the individual individual, the nature of the individual's condition to be treated, and generally the judgment of the attending physician. In some embodiments, 'G-CSF or GM-CSF can be administered in a single dose, administered over time. (eg, intravenous or transdermal administration) or multiple administrations to an individual, such as a human individual. In addition to direct administration to the body, G-CSF or GM-CSF can be used in an ex vivo treatment regimen to extract blood from the periphery of the individual. Separation of MSC It can be used to prepare MSC cultures which can optionally be cryopreserved and stored and subsequently used for individual cell-based regenerative therapies when MSCs are required. In some embodiments, this ex vivo production of MSC is available. Separating and collecting autologous MSCs. These autologous MSCs are collected from peripheral blood or, in some cases, from allografts of matching donors. In vitro or in combination with other agents, such as megalin cell inflammatory proteins. The concentration of the mobilizer in the peripheral blood is a problem of conventional optimization. A large number of granulocytes in the spleen can be observed by administration of G-CSF or GM-CSF '152716.doc -52·201130977 and the spleen weight is significantly increased, according to Bungart et al, Brit j 76, 174-179, 1990, this can be attributed to stem cell mobilization or MSC mobilization. In some embodiments, peripheral blood samples for use in methods as disclosed herein are from a mammalian individual such as a human Individually obtained. In some embodiments, a human subject has previously administered a mobilizing agent to increase the yield or number of circulating stem cells in the peripheral blood. Any mobilizing agent can be used, for example, according to the art. A commonly known method, and includes, for example, but not limited to, the methods disclosed in U.S. Patent Application No. 6,261,549, and U.S. Patent Application Serial No. 2009/0155225, the disclosure of each of In some embodiments, a mobilizing agent such as G-CSF or GM-CSF can be administered to an individual or ex vivo to a peripheral blood sample in combination with any one or more other mobilizing agents, such as selected from The following composition of the agents: interleukin-17 (IL-17), AMD3100, cyclophosphamide (Cy) and European paclitaxel (DXT) and combinations thereof. In some embodiments, a mobilizing agent such as G-CSF or GM-CSF can be administered to an individual or ex vivo to a peripheral blood sample, such as an antibiotic, in combination with other therapeutically or nutritionally active ingredients. Vitamins, herbal extracts, anti-inflammatory agents, glucose, antipyretics, analgesics, granulocyte-maize cell community stimulating factor (GM-CSF), interleukin-1 (IL-1), interleukin-3 (IL) -3), interleukin-8 (IL-8), PIXY-321 (GM-CSF/IL-3 fusion protein), megakaryocytic inflammatory protein, stem cell factor, thrombopoietin, and the like. Any individual can perform MSC mobilization using the methods disclosed herein, and can mobilize and collect MSCs from peripheral blood of any individual (such as a human individual, including adult or non-neonatal 152716.doc • 53·201130977 children). The MSC can be collected - or multiple collection steps or collection cycles. For example, from the individual's mobilization week, blood collection (eg, using blood separation) Μ% at least 2 times, at least 3 or to / 5- people "collected from each kilogram of body weight during each collection step The total number of nucleated cells may be - million (ΐχΐ()6) or more than one million (e.g., 1〇7, 1><1〇8,1><1〇9,1><<> 1〇1〇, 1><1〇11,1><1〇12, 1X10:: 1X101:, I, 15, 1, 1X10-, 1X101«, 1〇1X10). In a preferred embodiment, the number of cells in the single batch collection of segregation products (which are used for the separation of Μ% from non-MSCs) may be equal to or greater than ΐχΐ〇丨 depending on the weight and age of the donor individual. 5 total nucleated cells (TNC), or at least about 1χ1〇Μ, ΐχΐ〇13, ΐχΐ〇丨2, lxio6, lxio5 lxlO1 丨, 1x10 丨. , 1χ10, 1χ1〇8, 1χ1〇7 Total nucleated cells. Visual form and MSCM that can be obtained from any human individual donor using methods as disclosed herein, preferably at a human adult donor at an "adult" or mature j age (unless otherwise used in a particular context to achieve a different meaning) Otherwise, the term "adult" as used herein refers to and includes both adult and non-neonatal) and/or a certain minimum weight from which dry fines & By way of example, MSCs may be collected when the individual is in the range of 10 to 200 kg (according to an embodiment of the invention) or any range within this range, such as within 2 Torr to 4 〇 kg. Alternatively or in addition, according to an embodiment of the invention, the individual may be required to have a range of 2-80 years old (eg 2_1〇, 1〇_15, 1218, 16 2(), 20-26, 26-30, 30-35, An age within 30-40, 40-45, 40-50, 55-60, 60-65, 60_70, and 70-80 years old. 152716.doc •54· 201130977 Isolation of MSCs from other stem cell populations in peripheral gold The method of obtaining and isolating PB-derived MSCs and BM-derived MSCs from peripheral blood is well known in the art and includes leukocyte separation. , partial separation of density gradients, immune selection, and differential adhesion separation. Substantially pure MSC populations can be collected in a culture medium such as a chemically defined serum-free medium or a complete medium (such as DMEM or DMEM containing 1 g serum). A suitable chemically defined serum-free medium is described in U.S. Patent No. 08, 464, 359, filed on Jun. These patents are incorporated herein by reference. One aspect of the invention is the separation of MSCs from the peripheral blood mobilized using centrifugal panning. Without wishing to be bound by theory, in a panning process, a peripheral blood sample is introduced into a funnel-shaped separation chamber located on a rotary centrifuge. A liquid panning buffer stream or a low density stream is then introduced into the chamber containing the peripheral blood sample. When the flow rate of the liquid panning buffer solution through the chamber is increased (usually in a stepwise manner), the liquid will migrate to the eluting boundary of the chamber with smaller size and slower sedimentation cells, and the larger cell migration will settle faster. The indoor area to the balance of centrifugal force and sedimentation (drag) force. Thus, when peripheral blood (e.g., mobilized peripheral blood) comprises a plurality of different stem cell populations, one aspect of the present invention relates to the use of panning to separate such different populations of stem cells present in peripheral blood into different populations. Wherein smaller stem cells and larger fractions are isolated. In some aspects of the invention, 'any panning device can be used to obtain PB-derived MSCs and BM-derived MSCs and other peripheral blood. Stem cell population 152716.doc -55- 201130977 Isolation. In some embodiments, the panning device uses a separation product (in which platelets and blood cells are returned to the individual donor), and the remaining peripheral blood product is a separation product that is panned at a certain flow rate to peripheral blood. The sample is partially separated into a fraction comprising a MSC population. In some embodiments, commercially available panning devices (such as the ELUTRA® centrifuge manufactured by Gambro BCT, Inc.) can be used to separate MSCs from peripheral blood » ELUTRA CELL SEPARATION SYSTEM® Using the size and density, the circulated peripheral blood is separated into a portion containing MSC, so that cell enrichment, depletion, concentration, and washing can be performed in a closed functional system. ELUTRA CELL SEPARATION SYSTEM® is capable of enriching stem cell populations directly from leukocyte detachment products without the need for antibodies or pretreatment in less than 1 hour. The Elutra Cell Separation System® uses a countercurrent centrifugal panning method in which fluid flows through the cell layer in a centrifugal field to separate the cell population. In some embodiments, other panning devices (including but not limited to) COBE® Spectra separation systems, Trima® systems, and Trima Accel® systems, also manufactured by Gambro BTC Inc., and others for separating blood components Commercially available panning devices can be used to separate MSCs from peripheral blood. In some embodiments, the MSC population can be obtained from a mobilized peripheral blood sample using a cell separator, such as a COBE® Spectra separation system. The COBE® SPECTRATM centrifuges are described in U.S. Patent Nos. 4,425,172, 4,708, 712, and 6, 022, 306, incorporated herein by reference. In this embodiment, the mobilized peripheral blood sample (or the isolated product obtained from an individual who has administered the mobilizer according to the methods disclosed herein) is introduced into a cell separator (such as C0BE). In the system), and optionally add anticoagulant solution to the blood to prevent it from clotting during the process. The blood/anticoagulant mixture is circulated through a centrifuge to separate peripheral blood samples into MSC populations and to separate mononuclear cells from other blood components and plasma. The system draws the separated MSC into a collection bag for storage, while other blood sets and blood is returned to the patient. All tubing sets and needles used are sterile' so there is no risk of disease transmission. Other blood separation devices may be used, such as those described in the following: U.S. Patent No. 5,722,926, issued March 3, 1998, and U.S. Patent No. 5,951,877, issued on September 14, 1999; U.S. Patent No. 6,053,856, issued on Apr. 25, the entire disclosure of U.S. Patent No. 6,34,842, issued Jan. 1, 2002; U.S. Patent No. 7, 198, 848, U.S. Patent No. 6, 222, 362, U.S. Patent No. 6, 589, 526, U.S. Patent Application Serial No. PCT/A No. The entire disclosure of each of the patent applications is incorporated herein by reference. The inventors have discovered that it is desirable to adjust the hematocrit range used for bloodpheresis to enhance the properties of the collected cell population. For example, in one embodiment, the hematocrit range is selected to increase the ratio of VSELs. In another embodiment, the hematocrit range is selected to enhance cryopreservation of the collected VSELs', e.g., by minimizing granulocyte collection. In another embodiment, the hematocrit range is selected to ensure simultaneous collection of multiple stem cell populations, such as VSELs and MSCs. In one embodiment, the hematocrit range is selected to be 2-3%. In another embodiment, the hematocrit range is selected to be 3_152716.doc • 57- 201130977 4%.
鑑別MSC 如本文所揭示,可藉由FAC分選、以及使用光顯微術及 CFU-F檢定之免疫選擇及表型分析鑑別MSC。僅舉例而 言,可對培養板中或經EDTA分離之細胞之細胞離心 (cytospin)製備物中的經萊特-吉姆沙(Wright-Giemsa)染色 之細胞進行光顯微鏡檢查。組織化學染色可使用針對蘇丹 黑(Sudan Black)、過埃酸-希夫染料(Periodic Acid-Schiff ; PAS)、丁酸a-萘基酯酯酶、酸性磷酸酯酶及鹼性磷酸酯酶 之診斷套組(Sigma)藉由標準方案進行。 對於免疫螢光研究,MSC可用固定/滲透試劑 (Cytoperm®, Serotech Ltd·,Oxford, England)處理且用適當 的游離單株抗體或經FITC結合之單株抗體(如本文所論述) 標記且在Nikon顯微鏡上在使用汞氣燈的50 nm紫外線照明 下加以偵‘。 對於流動式細胞測量分析,經EDTA釋放之MSC或析離 產物中之MSC可用純單株抗體或經FITC或PE結合之單株 抗體染色。非特異性同型匹配抗體可用於測定背景螢光。 可在FACScan流式細胞儀(Becton Dickinson, BD)上分析 MSC且可根據此項技術中通常所知之程序用FAC Scan Lysis II(BD)研究軟體進行資料獲取。 以下單株抗體可用於偵測MSC :抗CD-45-FITC、抗 CD14-PE、抗 CD34-PE(Becton Dickinson);抗膠原蛋白 I、 抗膠原蛋白III及抗纖維結合蛋白(fibronectin)(Sigma);抗 152716.doc * 58 - 201130977 膠原蛋白 VI(Gibco BRL,Grand Island,N.Y.,USA);抗 ICAM-1(CD54)及抗 VCAM-1 (CD 106)(R&D Systems, Minneapolis, Minn.,USA)。對照小鼠 IgG 1-PE、IgG 1-FITC、IgG 2a-PE 及 F(ab')2-PE(Becton Dickinson)。單株抗 體 SH-2(IgG 1)及 SH-3.(IgG 2b)(Case Western Reserve University, Cleveland, Ohio·,USA 或 Osiris Therapeutics, Inc.(Baltimore, Md.,USA))。此等抗體識別MSC細胞表面 上之抗原,但不能與BM源性HSC以及骨母細胞或骨細胞 之細胞表面反應。 可藉由錐蟲藍(Trypan blue)經由染料排除(dye exclusion) 來測試實質上純MSC群之任何所收集樣品的總細胞計數及 細胞活力。亦可分析實質上純MSC群之樣品的表現細胞標 記之細胞之存在性。 處理前、處理後及任何其他細胞樣品之總細胞計數及細 胞活力可藉由血球計手工計數、流式細胞儀、或適於獲得 細胞計數之其他手段,諸如ViCell(Beckman Coulter)或適 於對顯微影像上所顯示之細胞計數之軟體加以定量。 超低溫保存 在一些實施例中,藉由如本文揭示之方法獲得之實質上 純人類MSC群可在液氮溫度下超低溫保存(例如冷凍)且長 期儲存,從而在解凍時能夠使用。若經冷凍,則MSC將通 常儲存於 10% DMSO、50% FCS、40% RPMI 1640培養基 中。人類MSC —經解凍,則可在生長因子及/或滋養層 (feeder layers)存在下離體培養及擴增適當時段,隨後植入 152716.doc -59- 201130977 有需要之個體中。 超低溫保存諸如MSC之幹細胞之方法通常為一般技術者 所知,且揭示於美國專利申請案2008/0220520、 2009/0022693、2008/0241113 及 2005/0106554 及美國專利 5,759,764及 7,112,576及 7,604,930 中。 對於超低溫保存,可將藉由如本文揭示之方法獲得之實 質上純MSC群懸浮於DPBS中,且可在冰上置放至少約15 分鐘,為超低溫保存作準備。該準備過程可包括添加超低 溫保存介質至實質上純MSC群中,接著利用控速冷凍器或 其他合適冷凍系統(監測凍結(dump-freeze)之冷凍系統或冷 凍容器(Nalgene)),對混合物執行若干降溫步驟,以使實 質上純MSC群之溫度降低至約-90°C的最終溫度。合適控 速冷床器包括(但不限於)Cryomed Thermo Forma控速冷象 器 7454(Thermo Electron, Corp.)、平面控速冷床器 Kryo 10/16(TS Scientific) ' Gordinier ' Bio-Cool--FTS Systems ' 及 Asymptote EF600、BIOSTOR CBS 2100 系列。 可製備包含培養基及DMSO之超低溫保存介質。可將約3 ml DPBS添加至容器(諸如50 ml圓錐形管)中。可向約3 ml DPBS中添加約1 ml人類血清白蛋白(HSA)至,接著在冰上 冷卻約10分鐘。向HSA及DPBS中添加約1 ml經冷卻99% DMSO,以製備最終超低溫保存介質。超低溫保存介質及 實質上純MSC群樣品可接著在冰上置放約15分鐘,隨後添 加超低溫保存介質至細胞樣品中。採用分批處理法,將超 低溫保存介質等分加至細胞樣品中。舉例而言,可取一等 152716.doc •60. 201130977 份約100 μΐ實質上純MSC群之懸浮液與約3 ml DPBS、1 ml HSA及約1 ml 99% DMSO合併。可取2等份約200 μΐ MSC 細胞懸浮液與約6 ml DPBS、2 ml HSA及約2 ml 99〇/0 DMSO合併。可取約5等分細胞樣品與約15以DPBS、約5 ml HSA及約5 ml 99% DMSO合併。可取約10等分細胞樣品 與約 30 ml DPBS、約 10 ml HSA 及約 10 ml 99% DMSO 合 併。 在一替代性實施例中,可使用其他超低溫保存介質。舉 例而言,含有超低溫保存劑之超低溫保存介質(諸如 CryoStor CS10或CS5(Bi〇life)、補充有丙二醇及蔗糖之胚 胎超低溫保存介質(Vitr〇life)、或SAGE介質(c〇〇per Surgical))可用於維持解凍後之高細胞活力結果。甘油可與 其他超低溫保存劑(諸如DMSO)—起使用或可依約1〇%之濃 度,單獨用於含有合適蛋白質之介質中。 超低溫保存介質可添加至實質上純Msc細胞群樣品中。 超低溫保存介質可逐滴添加至懸浮有實質上純Msc群之 DPBS中,直至所懸浮之經血幹細胞與超低溫保存介質之 總混合物體積達到超低溫保存細胞混合物之最終所要體 積。最終超低溫保存細胞介質可轉移至2><5 mi有條碼之冷 凍瓶(Cry〇quat)中,其中qC樣品儲存在5 mi小瓶之瓶蓋 I。使用移液器取出約 μ1#分樣品,且等分加至Q. 蓋中。在填滿瓶蓋之後,冑剩餘4.8 ml試樣添加入5⑹小 瓶中。樣品應送去進行傳染病分析及其他分析。冷珠小瓶 應置放於CRYOMED冷涞器中且經控速降溫至約.机或低 152716.doc -61 - 201130977 於-85°C之溫度。超低溫保存之試樣應轉移至儲槽(〜以打) 中,以在-150t或低於-uot的液氮蒸汽中儲存。對傳染 病測試呈陽性之任何樣品皆應檢疫。對傳染病測試呈陰性 之任何樣品可轉移至不同永久儲槽中。 以下降溫步驟可於控速冷凍器中加以程式化:首先使超 低溫保存劑與經血幹細胞之混合物降溫。與超低溫保存劑 合併之經血幹細胞可經受控速降溫以為最終儲存於冷凍器 中作準備》控速降溫經設計可維持細胞活力。Cry〇_Med冷 凍器(Thermo Electron Corp.)、液氮鋼瓶及攜帶型Cry〇_Identification of MSCs As disclosed herein, MSCs can be identified by FAC sorting, and immunoselection and phenotypic analysis using light microscopy and CFU-F assays. By way of example only, Wright-Giemsa stained cells in cytospin preparations in culture plates or cells isolated from EDTA may be subjected to light microscopy. Histochemical staining can be used for Sudan Black, Peric Acid-Schiff (PAS), a-naphthyl butyrate esterase, acid phosphatase and alkaline phosphatase. The diagnostic kit (Sigma) is performed by standard protocols. For immunofluorescence studies, MSCs can be treated with a fixation/permeation reagent (Cytoperm®, Serotech Ltd., Oxford, England) and labeled with a suitable free monoclonal antibody or FITC-conjugated monoclonal antibody (as discussed herein) and The Nikon microscope was Detected under 50 nm UV illumination using a mercury gas lamp. For flow cytometric analysis, MSCs in EDTA-released MSCs or isolates can be stained with either monoclonal antibodies or FITC- or PE-conjugated monoclonal antibodies. Non-specific isotype matched antibodies can be used to determine background fluorescence. MSCs can be analyzed on a FACScan flow cytometer (Becton Dickinson, BD) and can be accessed using the FAC Scan Lysis II (BD) study software according to procedures generally known in the art. The following monoclonal antibodies can be used to detect MSC: anti-CD-45-FITC, anti-CD14-PE, anti-CD34-PE (Becton Dickinson); anti-collagen I, anti-collagen III and fibronectin (Sigma) ); anti-152716.doc * 58 - 201130977 Collagen VI (Gibco BRL, Grand Island, NY, USA); anti-ICAM-1 (CD54) and anti-VCAM-1 (CD 106) (R&D Systems, Minneapolis, Minn .,USA). Control mice IgG 1-PE, IgG 1-FITC, IgG 2a-PE and F(ab')2-PE (Becton Dickinson). Monoclonal antibodies SH-2 (IgG 1) and SH-3. (IgG 2b) (Case Western Reserve University, Cleveland, Ohio, USA or Osiris Therapeutics, Inc. (Baltimore, Md., USA)). These antibodies recognize antigens on the surface of MSC cells but do not react with BM-derived HSCs and the cell surface of osteoblasts or bone cells. The total cell count and cell viability of any of the collected samples of the substantially pure MSC population can be tested by dye exclusion by Trypan blue. The presence of cells expressing the cell marker of a sample of substantially pure MSC population can also be analyzed. Total cell count and cell viability before, after, and on any other cell sample can be manually counted by a hemocytometer, flow cytometry, or other means suitable for obtaining cell counts, such as ViCell (Beckman Coulter) or suitable for The soft body of the cell count shown on the microscopic image was quantified. Cryopreservation In some embodiments, a substantially pure human MSC population obtained by a method as disclosed herein can be cryopreserved (e.g., frozen) at liquid nitrogen temperature and stored for extended periods of time for use during thawing. If frozen, the MSC will typically be stored in 10% DMSO, 50% FCS, 40% RPMI 1640 medium. Human MSCs - after thawing, can be cultured and expanded in vitro in the presence of growth factors and/or feeder layers for appropriate periods of time, and subsequently implanted in individuals in need of 152716.doc -59 - 201130977. Methods for cryopreservation of stem cells, such as MSCs, are generally known to those of ordinary skill in the art and are disclosed in U.S. Patent Application Nos. 2008/0220520, 2009/0022693, 2008/0241113, and 2005/0106554, and U.S. Patent Nos. 5,759,764 and 7,112,576 and 7,604,930. For cryopreservation, the substantially pure MSC population obtained by the methods disclosed herein can be suspended in DPBS and placed on ice for at least about 15 minutes in preparation for cryopreservation. The preparation process can include adding a cryopreservation medium to the substantially pure MSC population, followed by performing a mixture on a mixture using a controlled speed freezer or other suitable refrigeration system (dump-freeze refrigeration system or cryogenic vessel (Nalgene)) Several cooling steps are performed to reduce the temperature of the substantially pure MSC population to a final temperature of about -90 °C. Suitable speed-controlled cold bed devices include, but are not limited to, Cryomed Thermo Forma Cooling Camera 7544 (Thermo Electron, Corp.), Plane Controlled Cooling Machine Kryo 10/16 (TS Scientific) ' Gordinier ' Bio-Cool- -FTS Systems ' and Asymptote EF600, BIOSTOR CBS 2100 series. An ultra-low temperature storage medium comprising a medium and DMSO can be prepared. Approximately 3 ml of DPBS can be added to a container (such as a 50 ml conical tube). About 1 ml of human serum albumin (HSA) can be added to about 3 ml of DPBS, followed by cooling on ice for about 10 minutes. About 1 ml of cooled 99% DMSO was added to HSA and DPBS to prepare a final cryopreservation medium. The cryopreservation medium and the substantially pure MSC population sample can then be placed on ice for about 15 minutes, followed by the addition of cryopreservation medium to the cell sample. Aliquots of cryopreservation medium were added to the cell samples using a batch process. For example, a first-class 152716.doc • 60. 201130977 portion of a suspension of approximately 100 μΐ of substantially pure MSC population is combined with approximately 3 ml of DPBS, 1 ml of HSA, and approximately 1 ml of 99% DMSO. Approximately 2 aliquots of approximately 200 μM MSC cell suspension are combined with approximately 6 ml DPBS, 2 ml HSA, and approximately 2 ml 99 〇/0 DMSO. Approximately 5 aliquots of the cell sample can be combined with about 15 in DPBS, about 5 ml HSA, and about 5 ml 99% DMSO. Approximately 10 aliquots of the cell sample may be combined with about 30 ml of DPBS, about 10 ml of HSA, and about 10 ml of 99% DMSO. In an alternative embodiment, other cryogenic storage media can be used. For example, cryopreservation media containing cryopreservatives (such as CryoStor CS10 or CS5 (Bi〇life), embryo cryopreservation media supplemented with propylene glycol and sucrose (Vitr〇life), or SAGE media (c〇〇per Surgical) ) can be used to maintain high cell viability results after thawing. Glycerin may be used with other cryopreservatives (such as DMSO) or may be used alone in a medium containing a suitable protein at a concentration of about 1%. The cryopreservation medium can be added to a substantially pure Msc cell population sample. The cryopreservation medium can be added dropwise to the DPBS in which the substantially pure Msc population is suspended until the total volume of the suspended hematopoietic stem cells and cryopreservation medium reaches the final desired volume of the cryopreserved cell mixture. The final cryopreserved cell medium can be transferred to a 2<5 mi bar coded cryo-roller (Cry® quat) where the qC sample is stored in a 5 mi vial cap I. Use a pipette to remove approximately μ1 of the sample and add the aliquot to the Q. lid. After filling the cap, add 4.8 ml of the remaining sample to the 5 (6) vial. Samples should be sent for infectious disease analysis and other analysis. The cold bead vials should be placed in a CRYOMED cold-squeezing device and cooled to a temperature of about 152716.doc -61 - 201130977 at -85 °C. Samples cryopreserved should be transferred to a reservoir (~ to beat) for storage in liquid nitrogen vapor at -150t or below -uot. Any sample that is positive for an infectious disease test should be quarantined. Any sample that is negative for an infectious disease test can be transferred to a different permanent reservoir. The step of lowering the temperature can be programmed in a controlled speed freezer: first, the mixture of cryopreservative and menstrual blood stem cells is cooled. The menstrual blood stem cells combined with the cryopreservation can be cooled at a controlled rate to be finally stored in the freezer. The controlled rate cooling is designed to maintain cell viability. Cry〇_Med Freezer (Thermo Electron Corp.), Liquid Nitrogen Cylinder and Portable Cry〇_
Med冷凍器可用於控速降溫以為最終儲存於冷凍器中作準 備。細胞可於冷凍小瓶或冷凍袋中經受控速降溫以達到 約-90°C之溫度。 對於收集於冷凍袋中之實質上純MSC群樣品,細胞可經 受以下控速降溫概況:在約4t)下等待、以每分鐘丨〇(>c 至-6.0°C (樣品)' 以每分鐘25 〇〇c降至_5〇 〇t (腔室)、以每 分鐘10.0C降至_M.0eC (腔室)、以每分鐘1.〇。(:降 至-45.0 C (腔室)、以每分鐘1〇 〇〇c降至_9〇 〇<>c (腔室)、及 結束(樣品在-85.0°C下或低於_85.(Tc下)。 對於收集於冷凍小瓶中之實質上純MSC群樣品,細胞可 經受以下控速降溫概況:在4 (rc下等待、以每分鐘1〇<t 降至-3.0C(腔室)、以每分鐘⑺』^降至_2〇〇t>c(腔室)、以 每分鐘1.0C降至-40.0°C (腔室)、以每分鐘1〇〇〇c降 至-90.0°C (腔室)、及結束。 一旦超低溫保存劑與經血幹細胞之混合物處於約_85工 152716.doc -62· 201130977 或低於-85°C,即將超低溫保存小瓶轉移至極冷儲存裝置 (storage unit)中且在約^^或低於_13Γ(:溫度的液氮蒸 汽中儲存,或者,小瓶可儲存於液氮之液相中。舉例而 s ’適合的極冷儲存裝置包括(但不限於)ln2冷束器MVE 1830(Chart Industries) « 亦可藉由流動式細胞測量術分析實質上純MSC群之新鮮 樣品及先前超低溫保存之實質上純MSC群的解凍樣品以分 析細胞表面標記、細胞活力、及其他細胞特徵。亦可根據 以下方案在細胞溶解之後分析實質上純MSC群之新鮮樣 品。 超低溫保存之MSC必須根據本文所述之解康方法解康。 在MSC細胞樣品必須解凍之情況下,實質上純Msc群可能 需要在解凍之後或在培養細胞之後即刻進行流動式細胞測 量術分析以評估某一細胞傳代。超低溫保存之樣品可於 3 7 C水浴中攪拌,而不讓細胞完全解凍。細胞可轉移入約 1 ml冷卻洗滌介質中且藉由倒置加以混合。樣品可在約 2000 rpm下離心約7分鐘。可移除上清液且將細胞再懸浮 於約100 μΐ洗滌介質(25% HSA、DNAse、肝素及HBSS w/ Ca+及Mg+)中。再懸浮之細胞可接著於血庫血清離心機 (Blood Bank Serofuge)中離心約1分鐘。可傾析上清液且將 細胞再懸浮於約1.2 ml鞘液(Sheath fluid)中並加以滿旋。 中期至長期儲存所有或一部分藉由如本文揭示之方法獲 知之人類MSC(諸如中期或長期儲存於細胞庫中)亦在本發 明的範疇内。細胞於細胞庫中之此中期至長期儲存揭示於 152716.doc -63 - 201130977 美國專利申請案第2003/0054331號及專利申請案第 W003/024215號中,且該等專利以全文引用的方式併入本 文中。在處理結束時,如本文揭示之人類Msc可裝載入諸 如注射器之傳遞裝置中以藉由一般技術者所知之任何手段 置放入接受者個體中。 在一些實施例中,藉由如本文揭示之方法獲得之人類 MSC可視情況在適合介質存在下包裝於適合容器中。在一 些實施例中,包裝進一步包含用於所要目的之書面說明 書,諸如將人類MSC植入個體中以改善或治療疾病之方法 (及視情況存在之儲存方法及/或解凍方法(若經超低溫保 存)),或用於再生醫學或療法之治療方案之書面說明書。 如本文揭示之一態樣係關於治療需要增加幹細胞(諸如 MSC)數目之個體的方法。在一些實施例中,個體預定或 意欲捐獻諸如MSC之幹細胞用於例如異源或自體移植。一 般而言,方法包括向需要此治療或已確定需要此治療之個 體投與治療有效量之如本文所述的調動劑,諸如gm_csf 或G-CSF。為治療此等個體而投與治療有效量之如本文所 述之調動劑(諸如G-CSF或GM-CSF)為期至少2天或至少3天 但至多4天將使周邊血中之MSC數目及/或頻率增加。在一 些實施例中,此投藥將使周邊血中之MSC數目增加約1 〇_ 500倍。量測此等增幅之方法在此項技術中已知,其揭示 於本文中,且參見例如Neben等人,Blood. 1993; 81(7):1960-7 ; Ashihara 等人 Exp. Hematol. 2000; 152716.doc •64- 201130977The Med chiller can be used to control the temperature drop for final storage in the freezer. The cells can be cooled at a controlled rate in a frozen vial or freezer bag to a temperature of about -90 °C. For a substantially pure MSC population sample collected in a freezer bag, the cells can be subjected to the following rate-controlled cooling profile: wait at about 4 t), 丨〇 (>c to -6.0 ° C (sample) per minute per Minute 25 〇〇c drops to _5〇〇t (chamber), drops to _M.0eC (chamber) at 10.0C per minute, at 1.〇 per minute. (: down to -45.0 C (chamber) ), to 1 〇〇〇c per minute to _9 〇〇 <> c (chamber), and end (sample at -85.0 ° C or below _85. (Tc under). For collection A substantially pure MSC population sample in a frozen vial, the cells can withstand the following rate-controlled cooling profile: waiting at 4 (rc, 1 〇 per minute) to -3.0 C (chamber), per minute (7) ^ falls to _2〇〇t>c (chamber), drops to -40.0 ° C (chamber) at 1.0 C per minute, drops to -90.0 ° C (chamber) at 1 ° C per minute, And at the end. Once the mixture of cryopreservative and menstrual blood stem cells is at about _75 workers 152716.doc -62·201130977 or below -85 °C, the cryopreservation vials are transferred to a very cold storage unit and at about ^ ^ or below _13Γ (: temperature of liquid nitrogen Stored in steam, or vials can be stored in liquid nitrogen. For example, s 'suitable extremely cold storage devices include (but are not limited to) ln2 cold bundler MVE 1830 (Chart Industries) « can also be flowed Cytometry analyzes fresh samples of substantially pure MSC populations and thawing samples of previously purely cryopreserved substantially pure MSC populations for analysis of cell surface markers, cell viability, and other cellular characteristics. The substance can also be analyzed after cell lysis according to the following protocol Fresh samples from the pure MSC population. The cryopreserved MSC must be resolved according to the method described in this article. In the case that MSC cell samples must be thawed, the substantially pure Msc population may need to be after thawing or immediately after culturing the cells. Flow cytometry analysis is performed to assess passage of a certain cell. The cryopreserved sample can be stirred in a 37 C water bath without allowing the cells to completely thaw. The cells can be transferred to about 1 ml of the cooled wash medium and inverted by inversion. Mix and mix. The sample can be centrifuged at about 2000 rpm for about 7 minutes. The supernatant can be removed and the cells resuspended in about 100 μL. (25% HSA, DNAse, heparin and HBSS w/ Ca+ and Mg+). The resuspended cells can then be centrifuged in a Blood Bank Serofuge for about 1 minute. The supernatant can be decanted and the cells re-exposed. Suspension in about 1.2 ml of Sheath fluid and full rotation. Medium to long term storage of all or a portion of human MSCs (such as medium or long term storage in cell banks) known by the methods disclosed herein is also in the present invention. Within the scope of the. The medium to long term storage of the cells in the cell bank is disclosed in U.S. Patent Application Serial No. 2003/005433, filed on Jan. Into this article. At the end of the treatment, the human Msc as disclosed herein can be loaded into a delivery device such as a syringe for placement into a recipient individual by any means known to those of ordinary skill. In some embodiments, a human MSC obtained by a method as disclosed herein can be packaged in a suitable container in the presence of a suitable medium, as appropriate. In some embodiments, the package further comprises written instructions for the desired purpose, such as methods of implanting human MSCs into an individual to ameliorate or treat the disease (and, where appropriate, storage methods and/or thawing methods (if cryopreserved) )), or a written instruction for a treatment plan for regenerative medicine or therapy. One aspect as disclosed herein relates to a method of treating an individual in need of an increase in the number of stem cells, such as MSCs. In some embodiments, the individual is scheduled or intended to donate stem cells such as MSCs for, for example, heterologous or autologous transplantation. In general, the methods comprise administering to a subject in need of such treatment or having determined that such treatment is administered a therapeutically effective amount of a mobilizing agent, such as gm_csf or G-CSF, as described herein. Administration of a therapeutically effective amount of a mobilizing agent (such as G-CSF or GM-CSF) as described herein for the treatment of such individuals for a period of at least 2 days or at least 3 days but up to 4 days will result in the number of MSCs in the peripheral blood and / or frequency increases. In some embodiments, the administration will increase the number of MSCs in the peripheral blood by about 1 〇 _ 500 times. Methods for measuring such increases are known in the art and are disclosed herein, and see, for example, Neben et al, Blood. 1993; 81(7): 1960-7; Ashihara et al. Exp. Hematol. 2000; 152716.doc •64- 201130977
28(3):311-7 ; Pruijt等人,proc. Natl. Acad. Sci. U.S.A 1999; 96(19):10863-8。 諸如G-CSF或GM-CSF之調動劑之劑量、毒性及治療功 效可藉由標準醫藥程序’例如用於測定LD50(群體50%致 死劑量)及ED50(群體50%治療有效劑量)之標準醫藥程序, 於例如細胞培養物或實驗動物中加以確定。毒性效應與治 療效應之間的劑量比率為治療指數且其可表示為比率 LD50/ED50。展現高治療指數之調動劑(諸如G-CSF或GM- CSF)較佳》儘管可使用展現毒副作用之化合物,但應慎重 設計可使此等化合物靶向受影響組織之位點的傳遞系統以 使對未感染細胞之潛在損傷最小化且藉此降低副作用。 自細胞培養檢定及動物研究獲得之資料可用於確定供人 類使用的劑量範圍。此等化合物之劑量較佳在包括具有少 許或不具有毒性之ED50之循環濃度的範圍内。視所用劑 型及所用投藥途徑而定,劑量可在此範圍内變彳卜本發明 方法所用之任何化合物的治療有效劑量初始皆可利用細胞 培養檢定估計。㈣可在動物模财確定以獲得循環金毁 濃度範圍’其包括如細胞培養中所測定之IC5〇(亦即達成 症狀之半數最大抑制之測試化合物丨農度卜此資訊可用於 剧確定適用於人類之劑量。血漿中之含量可藉由例如 南效液相層析量測。 根據本發明之另一態樣’提供在其他臨床程序之前/期 間將MSC作為單—藥劑自某—組織調動至另—組織中之方 法。此等細胞包括非造血正f細胞且在其生命週期中之至 152716.doc •65· 201130977 少一個分化階段期間具有經歷活體内調動/遷移的潛能。 根據本發明之另一態樣,提供藉由輸注有效量之調動劑 (諸如GM-CSF或G-CSF)而在收集待用於器官移植之組織之 刖及期間調動MSC的方法。在一些實施例中,本發明亦提 供使用諸如GM-CSF或G-CSF之離體調動劑將MSC從已自 個體收集之離體器官或周邊血樣品調出的方法。此包括例 如在自個趙收集PB源性或BM源性MSC之前,活體内灌注 來自合法死亡器官供者之活體内周邊血。 丨 在一實施例中’向個體投與有效量之調動劑,諸如G_ CSF或GM-CSF ’以調動該個體之周邊血中之bm源性 MSC »在另一實施例中’向個體投與有效量之調動劑,諸 如G-CSF或GM-CSF,以調動該個體之周邊血中之pB源性 MSC »在另一實施例中’使有效量之之調動劑(諸如g_csf 或GM-CSF)與個體之周邊血樣品接觸以離體調動周邊血中 之PB源性MSC。「有效量」為足以實現周邊血中之msc數 目及/或頻率顯著增加的量。有效量可以一或多次投藥、 用藥或給藥投與。調動劑之治療有效量視所選調動劑(例 如G-CSF或GM-CSF)而定。諸如G-CSF或GM-CSF之調動劑 可每天投與一或多次至每週投與一或多次;包括每隔一天 一次。熟習此項技術者將瞭解某些因素可能影響為有效治 療個體所必需之劑量及時序,該等因素包括(但不限於)先 前治療、個體之综合健康及/或年齡、及存在之其他疾 病。此外,用如本文所述之治療有效量之G-CSF或GM-CSF治療個體可包括單一治療或一系列治療。 152716.doc -66- 201130977 在一些實施例中,本文所述之治療方法包括投與另一種 調動劑,例如選自由以下組成之群但不限於以下之藥劑: 介白素· 17(IL-17 ; Journal of Immunol. 2001; 167: 2081-2086)、AMD3100(Flomenberg等人,Acta Haematol. 2005; 114(4):198-205)、環磷醯胺(Cy)、歐洲紫杉醇及 (DXT)(Ojeifo 等人,Experimental Hematology 2000; 28:451-459)。 在一些實施例中,方法包括向個體投與PB源性MSC或 BM源性MSC或其分化子代,例如將細胞再引入同一個體 中或將細胞移植入第二個體(例如HLA型匹配之第二個體) 中。 在一些實施例中,可使用PB源性MSC或BM源性MSC有 效治療之個體包括可通常用骨髓或幹細胞移植物治療之任 何個體,例如患有癌症,例如神經母細胞瘤 (neuroblastoma)(產生於不成熟神經細胞中且主要影響嬰兒 及兒童之癌症)、骨趙發育不良(myelodysplasia)、骨髓纖 維化(myelofibrosis)、乳癌、腎細胞癌或多發性骨髓瘤之 個體。舉例而言,PB源性MSC或BM源性MSC可移植入患 有對放射療法或化學療法治療具有抗性之癌症的個體中, 以例如恢復用於治療癌症之高劑量化學療法及/或放射療 法所破壞的幹細胞。 在一些實施例中,MSC可接著於血庫血清離心機中離心 約1分鐘。可傾析上清液且將細胞再懸浮於約1.2 ml鞠液中 並加以滿旋。 152716.doc -67- 201130977 中期至長期儲存所有或一部分藉由如本文揭示之方法獲 得之MSC群或實質上純MSC群(諸如中期或長期儲存於細 胞庫中)亦在本發明的範_内。細胞於細胞庫中之此中期 至長期儲存揭示於美國專利申請案第2003/0054331號及專 和申凊案第W003/024215號中,且該等專利以全文引用的 方式併入本文中。在處理結束時,如本文揭示之MSC群或 實質上純MSC群可裝載入諸如注射器之傳遞裝置中以藉由 一般技術者所知之任何手段置放入接受者個體中。在一些 實施例中,使用如本文揭示之方法獲得之MSC群可懸浮於 適於輸注入個體中之短期儲存介質中,諸如美國專利 5,955,257中揭示之介質,該專利以全文引用的方式併入本 文中。在-些實施例中,使用如本文揭示之方法收集並富 集之MSC可經儲存(例如供供者(例如同種異體)或另一個體 未來治療使用)或直接移植,例如用於促進手術後癒合。 在本發明之一態樣中,藉由如本文揭示之方法收集之複 數個MSC可進一步分選成至少2個亞群,其可分別或一起 (例如含於同一小瓶中)超低溫保存。在一些實施例中至 少2個MSC亞群可為2個不同MSC亞群。舉例而言,14|5(:可 分選成至少2個細胞亞群,例如(但不限於)(1)幹細胞群或 富含MSC群及(2)非幹細胞群或MSC排除群。此外,亦設想 2個亞群(亦即以上(1)及(2))可一起超低溫保存。在一 ^實 施例中,MSC分為亞群之此分選可為陽性或陰性分選或其 組合。 在一些實施例中,細胞表面蛋白標記可用於正向選擇或 152716.doc -68- 201130977 負向選擇或移除幹細胞。此等標記之實例在此項技術中已 熟知且展示於表1中: 表1 細胞類型 標記 造血幹細胞(HSC) CD34+、CD45+、CXCR4+ 内皮祖細胞 CD34+、CD45+、CD133+、KDR+ 極小胚胎樣細胞(VSEL) CD34+、CD133+、CXCR4+、SSEA4+ 間質幹細胞(MSC) CD34·、CD45_、CD90+、CD105+、 CD106+、CD44+ 在一些實施例中,藉由如本文揭示之方法獲得之MSC群 或實質上純MSC群可視情況在適合介質存在下包裝在適合 容器中。在一些實施例中,包裝進一步包含用於所要目的 之書面說明書,諸如將MSC或實質上純MSC群植入個體中 以改善或治療疾病之方法(及視情況存在之儲存方法及/或 解凍方法(若經超低溫保存)),或用於再生醫學或療法之治 療方案之書面說明書。 用於自短細胞療法之MSC : 在本發明之一實施例中,藉由如本文揭示之方法自個體 之經調動之周邊血收集的MSC在該個體需要此細胞療法 (例如自體細胞療法)時可引入或移植回至個體中。在一些 實施例中,MSC可單獨或與其他細胞組合用於再生療法中 之未來自體治療應用,諸如創口癒合、心臟修復及骨修復 及其他橋形適應症,如本文在實例中所揭示。在一些實施 例中,藉由如本文揭示之方法自個體之經調動之周邊血收 152716.doc -69- 201130977 集的MSC可用於修復或治療眼病’諸如視網膜病或黃斑變 性,如 Vossmerbaeumer等人 ’ Cytotherapy,2009; 11; 177-188中所論述,該文獻以全文引用的方式併入本文中。在 一些實施例中,藉由如本文揭示之方法自個體之經調動之 周邊血收集的MSC可分化成任何數目之起源於中胚層的不 同細胞類型,包括脂肪細胞、骨母細胞及軟骨細胞,如 Musina等人,Cell Technologies in Biology and Medicine, 2006; 2; 147-151中所論述,該文獻以全文引用的方式併入 本文中。植入MSC以治療心血管病症及其他病症之方法為 一般技術者所熟知,且揭示於國際專利W0 2008/0548 19及 美國專利 6,387,369、7514074、6,174,333及 6,225,119及美 國專利申請案2009/0214493中,該等專利以全文引用的方 式併入本文中。 在本發明之一實施例中,自個體經調動之周邊血獲得之 MSC可用於個人化醫學應用,諸如自體治療性用途,以及 用於個人化檢定中以評估個人飲食、藥物遺傳學、神經化 學品、生活方式對MSC功能及活力之影響。 包含一部分或所有藉由如本文揭示之方法自個體之周邊 血收集的MSC之MSC組合物可用於經由增補 (augmentation)受損組織來修復、治療或改善各種美觀或 功能性病狀(例如缺陷)^藉由如本文揭示之方法自個體之 周邊血收集的MSC為再建或增補受損組織提供重要資源。 在一實施例中’藉由如本文揭示之方法自個體之周邊血收 集的MSC可用於組織工程改造及再生醫學中以置換已由發 152716.doc •70· 201130977 月缺陷、損傷、疾病或老化磨損損壞的身體部分。藉由如 本文揭示之方法自個體之周邊血收集的MSC提供其中msc 可分化成不同細胞且產生相同個體之特定譜系或基因型的 獨特系統。因此,藉由如本文揭示之方法自個體之周邊血 收集的MSC提供個別化幹細胞療法之顯著優點。 此外,在不存在疾病或損傷之情況下,藉由如本文揭示 之方法自個體之周邊血收集的此等Msc及其組合物可藉由 向軟組織區域 '裂口、凹陷或空隙中添加主體(諸如用於 修平」來用於增補與損傷無關之軟組織。本發明亦包括 多次及連續投與藉由如本文揭示之方法自個體之周邊血收 集的MSC。 對於基於幹細胞之治療,藉由如本文揭示之方法自個體 之周邊血收集的MSC群體較佳自自體來源收集。接著如本 文所述製備並分離MSC組合物,且特定言之,與存在於經 調動之周邊血中之其他幹細胞群分離。為了根據本發明將 藉由如本文揭示之方法自個體之周邊金收集的及/或 其組合物引入或移植入人類或動物接受者中,製備單核細 胞之懸浮液。單核細胞懸浮液含有MSC之濃縮物,且可與 存在於周邊血中之其他幹細胞分離,且收集Msc並再懸浮 於生理學上可接受之載劑、賦形劑或稀釋劑中。或者, MSC懸浮物可含於無血清無菌溶液(諸如超低溫保存溶液) 中。在一些實施例中,亦可使用富含MSC之製劑。在一些 貫施例中,MSC懸浮液可接著經由例如注射引入供者組織 之一或多個部位中。 152716.doc •71 · 201130977 經濃縮或經富集之幹細胞可以含有生理學上可接受之載 劑、賦形劑或稀釋劑之醫藥學上或生理學上可接受的製劑 或組合物形式投與,且投與相關接受者有機體(包括人類 及非人類動物)之組織。可藉由將細胞再懸浮於適合液體 或溶液(諸如無菌生理鹽水或其他生理學上可接受之可注 射水性液體)中I製備含有MSC(组合物。此等組合物中 各組分之使用量可由熟習此項技術者以常規方式確定。 在-些實施例中’藉由如本文揭示之方法自個趙之周邊 血收集的MSC群體或其組合物可如下投與:將幹細胞懸浮 液置放於吸收性或黏著性材料(亦即膠原蛋白海綿基質)上 且將含有MSC之材料插於相關部位中或相關部位上。或 者’藉由如本文揭不之方法自個體之周邊血收集的Μ%群 體可藉由非經腸注射途徑(包括皮下、靜脈内、肌肉内及 胸骨内注射)投與。其他投藥模式包括(但不限於)鼻内、鞘 内、皮内、經皮、經腸及舌下投藥。在一實施例中,藉由 如揭示之方法自個體之周邊血收集的MSC群體可藉由内窺 鏡手術投與。 對於可’主射投藥’包含藉由如本文揭示之方法自個體之 周邊血收集的MSC之組合物可懸浮於無菌溶液或懸浮液中 或可再懸浮於醫藥學上及生理學上可接受之水性或油性媒 劑中,其可含有防劑、穩定劑及使溶液或懸浮液與接受 者之體液(亦即血液)等張之物質。適用之賦形劑之非限制 性實例包括水、破酸鹽緩衝鹽水(pH 7 4)、〇 15 M氣化納 水溶液、右旋糖、甘油、稀乙醇及其類似物、及其混合 152716.doc -72· 201130977 物。說明性穩定劑為可單獨或混合使用之聚乙二醇、蛋白 質、醣、胺基酸、無機酸及有機酸。用量或數量以及所用 投藥途徑視個體而定,且相當於熟習此項技術者所知之類 似服用類型或適應症中所使用的量β 依照本發明,藉由如本文揭示之方法自個體之周邊血收 集的MSC群體可投與身體組織,包括上皮組織(亦即皮 膚、内腔等)、肌肉組織(亦即平滑肌)、血液、腦及各種器 官組織,諸如與泌尿系統相關之彼等器官(亦即膀胱、尿 道、輸尿管、腎等)。 根據本文所述之一般治療方法,藉由如本文揭示之方法 自個體之周邊血收集的MSC群體可包含其他細胞(例如包 含目標幹細胞群之細胞部分)且可投與個體,例如藉由靜 脈内(ι·ν.)導管輸注入個體之血流中(類似於任何其他靜脈 内流體)。或者,然而,可產生MSC群之個別化混合物, 以諸如提供特定用於個體之治療需要的細胞療法混合物。 諸如經由析離方法獲得之全面細胞混合物可經表徵分選 且分離成不同細胞群。細胞標記(諸如VSEL、MSC及HSC 標記)或組織特異性標記可用於在表型上表徵自周邊血收 集之細胞群。使用此等標記,有可能基於細胞類型進行分 離及分選。從而將細胞混合物轉變成細胞群,其可大致分 成兩部分:幹細胞部分及非幹細胞部分。非幹細胞部分可 進一步分成祖細胞或纖維母細胞部分及功能細胞或完全分 化細胞部分。周邊血細胞混合物一經分選,即可將MSC群 及非幹細胞部分分別超低溫保存及儲存。以此方式,可產 152716.doc •73- 201130977 生來自個趙之不同細胞群之集合庫或儲存庫。或者,Msc 群及非幹細胞部分可一起超低溫保存,接著在使用之前分 選及分離。 在一些實施例中,藉由如本文揭示之方法自個體之周邊 血收集的MSC群體可用於產生或分化成某一細胞類型之任 何群體’其由胚層(亦即内胚層、中胚層及外胚層)發育而 成。此等細胞包括(但不限於)分化細胞、神經祖細胞或分 化細胞、神經膠質祖細胞或分化細胞、寡樹突神經膠質細 胞祖細胞或分化細胞、皮膚祖細胞或分化細胞、肝祖細胞 或分化細胞、肌肉祖細胞或分化細胞、骨祖細胞或分化細 胞、間質幹細胞或祖細胞、胰臟祖細胞或分化細胞、祖細 胞或分化軟骨細胞、基質祖細胞或分化細胞、培養擴增之 幹細胞或祖細胞、經培養之分化幹細胞或祖細胞、或其組 合。亦關注祖細胞,諸如造血、神經、基質、肌肉(包括 平滑肌)、肝、肺、胃腸及間質祖細胞。亦關注已分化之 MSC群’諸如分化成細胞,諸如(但不限於)骨母細胞、肝 細胞、粒細胞、軟骨細胞、肌細胞、脂肪細胞、神經元細 胞、胰臟細胞、或其組合及混合物。 在一些實施例中’藉由如本文揭示之方法自個體之周邊 血收集的MSC群可組合、重組或混配成適於治療個體疾病 及/或特定組織再生的細胞療法細胞混合物。藉由如本文 揭示之方法自個趙之周邊血收集的MS C群體與組織特異性 祖細胞及視情況存在之功能細胞的組合可用於例如增強所 移植之MSC的嫁接性。 152716.doc • 74· 201130977 因此,在一實施例中,本發明提供使用藉由如本文揭示 之方法自個體之周邊血收集的MSC群之自體混合物的方法 及產品。此MCS群可單獨或與其他適用細胞組合使用。在 一些實施例中,MSC可與得自周邊血之其他幹細胞、或其 他功能適用非幹細胞組合。 因此,個體MSC可單獨或經選擇性重組(定製混合)用於 再生療法中之個別化自體治療應用。換言之,自體MSC群 可與其他細胞(包括幹細胞,諸如得自周邊血之其他非 MSC幹細胞)選擇性組合以用於自體細胞療法。在一些實 施例中,混合MSC群可包含約10%至約90%之藉由如本文 揭示之方法自個體之周邊血收集的MSC、約10%至約 80%、或約10%至約60%、或約10%至約40%、或約10%至 約90%之藉由如本文揭示之方法自個體之周邊血收集的 MSC。在一些實施例中,MSC群亦可包含非幹細胞(例如 功能細胞)及/或亦得自周邊血之不為MSC之幹細胞的群 體,諸如約5%至約50%功能細胞及/或非MSC幹細胞、約 5%至約40%功能細胞及/或非MSC幹細胞、約5%至約30% 功能細胞及/或非MSC幹細胞、約5%至約20%功能細胞及/ 或非MSC幹細胞、或約5%至約10%功能細胞及/或非MSC 幹細胞。 上述細胞療法產品之一適合實例為HSC與MSC或造血系 統之其他功能細胞的自體混合物。另一實例為包含自體 MSC群與其他細胞混合的細胞療法產品,該等其他細胞諸 如以下細胞類型中之至少一者或任何組合:HSC、PBSC、 152716.doc -75- 201130977 心肌祖細胞及視情況存在之心肌細胞。 因此,在另一實施例中,提供治療有需要之患者之方 法,其包含向個體投與單獨或組合(分別或混合)之藉由如 本文揭示之方法自個體之周邊血收集的MSC群之自體混人 物0 幹細胞错藏(Banking) 在本發明之另一態樣中,藉由如本文揭示之方法自個體 之周邊血收集的MSC群可儲存於細胞庫中以支持任選健康 照護保險模型,從而有效保護群體成員防止未來疾病。當 個別個體處於健康狀態時,其可選擇處理及保藏自周邊: 收集之其自身幹細胞群以用於未來其健康照護需要之配給 (distribution)。 因此,在-實施例中,藉由如本文揭示之方法自個體之 周邊血收集的MSC群「儲藏」在幹細胞庫或寄存處或儲存 設施、或為保管而保存目標幹細胞群之任何地方以供未來 使用。儲存絲可以倘若發生諸如核攻擊之災難性事件, 藉由如本文揭示之方法自個體之周邊▲收集的目標幹細胞 群得以保持安全的方式加以設計m施例中,儲存 設施可在地τ ’洞中或地窖中。在其他實施例中,其可在 山之側面或在外太空中。儲在执 料4可嵌㈣諸如錯之遮蔽 材料中。 根據另-實施例,提供具有4步驟幹細胞儲藏方 ⑽包括向個體投與一或多種調動劑以增加該個體之周邊 血中之魔群的量。步驟以括藉由如本文 152716.doc • 76 · 201130977 周邊血收集MSC群,其中該個體無即刻感知之健康病狀, 例如無其中個體需要使用其自身目標幹細胞群進行其自體 移植來治療的病狀。步驟C包括以保藏之細胞群形式保藏 藉由如本文揭示之方法自周邊血收集的MSC群。步驟D包 括取得藉由如本文揭示之方法收集之保藏MSC群以將MSC 群自體移植入個體中。 將詳細參考本發明之實施例,其實例在一些隨附圖式中 加以說明。在本發明之一些實施例中,包括使用 Lakewood, Colorado之 Gambro BCT,Inc_ 所製造之 Elutra® 離心機自經調動之血液中分離MSC。儘管本發明之實施例 係聯合Elutra®離心機描述,但此提及僅出於例示性目的 且不意欲在任何意義上限制本發明。應瞭解本發明之實施 例可使用其他離心機,包括(但不限於)亦由Gambro BTC Inc.製造之COBE® Spectra析離系統、Trima®系統及Trima Accel®系統、以及用於分離血液組分之其他淘洗裝置。 MSC用於個人化醫學診斷及預後性檢定之用途。 本發明之另一態樣係關於得自個體經調動之周邊血之 MSC的用途,其用於個人化醫學應用,諸如自體治療性用 途,以及用於個人化檢定中以評估個人飲食、藥物遺傳 學、神經化學品、生活方式對MSC功能及活力之影響。此 等檢定當前在此項技術中已熟知,且包括其中使化合物、 藥劑或環境刺激物與MSC接觸之高產量篩檢,且可量測化 合物、藥劑或環境刺激物對MSC功能(例如分化潛力、增 殖、存活)及活力之影響,且結果與參考對照樣品(例如不 152716.doc -77· 201130977 存在化合物 物)比較》 或陽性對照樣品 ’諸如存在BMP6及其類似 在本發明之-態樣中,自個體獲得之奶C群體可用於高 產量筛檢中以評估個體對藥劑(諸如對特定藥物、激素治 療或醫藥或療法)之個別反應。簡言之,使MSC細胞群與 相關藥劑接觸,且藉由監測輸出參數(諸如標記表現、細 广舌力77化特徵、多能能力及其類似參數)對藥劑作用 2仃-平估。MSC可使用如本文揭示之方法新鮮分離,或在 些實施例中,在用於檢定t之前經培養、超低溫保存或 遺傳工程改造。在-些實施例中,個體職可為純系培養 物之環境料變㈣:例如分裂成獨立培杨且在獨特條 件(例如有或無病毒;存在或不存在其他細胞激素或其组 合)下生長。 重要的是,因為MSC對於個體遺傳組成而言具有特異 性,所以MSC之使用提供對MSC進行藥物遺傳分析之工 具二舉例而言,個體MSC可具有會導致獨特病理性特徵之 特定變異體。舉例而言,因為Msc來自於個體(例如自 體)’所以其具有所要病理性㈣,例如造以病病變或 導致對治療藥物之反應不良或增強之突變及/或多形現 象。因此,藉由如本文揭示之方法獲得之Msc可用於評估 個體對特定化合物、藥劑之MSC反應且在一些情況下此可 適用於疾病之預後。在該實施例中,本發明方法可用於篩 檢緩和病變之藥劑、或正面影響MSC增殖或功能之藥劑。 在替代性實施例中,本發明方法可用於篩檢相較於來自 152716.doc .78 · 201130977 其他個體之MSC(例如無突變及/或多形現象),引起個體之 不同MSC反應(歸因於固有遺傳組成或特定突變及/或多形 現象)之藥劑,因此該等方法可用於例如評估相較於其他 人及/或MSC細胞,特定藥物及/或藥劑對個體Msc群體之 影響,因此充當個人化醫學及/或藥物遺傳學之高產量篩 檢。MSC對藥劑(特定言之,藥理學藥劑)反應之方式(包括 反應時序)為細胞生理狀態之重要反映。 參數為MSC之可定量組分,特定言之,宜於高產量系統 中準確量測之組分。參數可為任何細胞組分或細胞產物, 包括細胞表面決定子(determinant)、受體、蛋白質或其構 形或轉譯後修飾形式、脂質、碳水化合物、有機或無機分 子、核酸(例如mRNA、DNA等)、或來源於該種細胞組分 之一部分、或其組合》儘管大多數參數將提供定量讀數, 但在一些情況下,半定量或定性結果可接受。讀數可包括 單一測定值,或可包括平均值、中位值或偏差等。利用許 多相同檢定將獲得各參數之參數讀數值之特徵性範圍。預 期存在可變性(Variability)且將使用標準統計方法及用於提 供單一值之常見統計方法獲得各測試參數組之值範圍。 包括候選藥劑之化合物係自廣泛多種來源(包括合成或 天然化合物之集合庫)獲得。舉例而言,眾多手段可用於 隨機及定向合成廣泛多種有機化合物,包括生物分子,包 括表現隨機化寡聚核苷酸及寡肽。或者,呈細菌、真菌、 植物及動物萃取物形式之天然化合物集合庫可利用或容易 產生。另夕卜,天然或合成產生之集合庫及化合物容易經由 152716.doc -79· 201130977 習知之化學、物理及生物化學手段修飾且可用於產生組合 集合庫。可對已知藥理學藥劑進行定向或隨機化學修飾, 諸如醯化、烧基〖、醋化、酿胺化等,以產生結構類似 物。 針對藥劑對幹細胞群的影響來篩檢藥劑,此係藉由將藥 劑(通常連同缺乏藥劑之細胞一起)添加至至少一個且通常 複數個幹細胞樣品中。量測對藥劑有反應之參數變化,且 藉由與參考培養物(例如在藥劑存在及不存在下,用其他 藥劑獲得等)比較來評估結果。 在一些實施例中,藥劑宜以溶液形式或易溶形式添加至 培養中之幹細胞的培養基中。藥劑可以呈間歇或連續流形 式之流過式系統(flow-through system)添加,或者,逐一 或遞增添加化合物之大丸劑至另外靜止之溶液中。在流過 式系統中’使用兩種流體,其中一種流體為生理學上中性 溶液,且另一種流體為與所添加之測試化合物相同之溶 液。第一流體及第二流體依次越過幹細胞\在單一溶液方 法中’添加測試化合物之大丸劑至包圍細胞之—定體積的 培養基中。培養基組分之總濃度不應隨大丸劑添加而顯著 變化,或在流過式方法中,不應在兩種溶液之間顯著變 化。在一些實施例中,藥劑調配物不包括可對總調配物具 有顯著影響之其他組分,諸如防腐劑。因此,較佳調g己物 基本上由生物活性化合物及生理學上可接受之載齊丨^列如 水、乙醇、DMSO等)組成。然而,若化合物為無溶劑液 體,則調配物可基本上由化合物自身組成。 152716.doc -80- 201130977 複數個檢定可以不同藥劑濃度平行操作以獲得對各種濃 度之差異反應。如此項技術中所知,通常使用依1:10或其 他對數尺度、稀釋度產生的一系列濃度確定藥劑的有效濃 度。必要時,濃度可用第二系列之稀釋度進一步改進。通 常,此等濃度之一充當陰性對照,亦即處於零濃度或低於 藥劑偵測含量或處於或低於不產生可偵測之表型變化之藥 劑濃度。 視情況’用於篩檢之個體MSC可經操作以表現所要基因 產物’以例如賦予MSC所要生理特徵及/或修復或置換 MSC之遺傳組成中之不合需要的突變及/或多形現象。因 此’在個體MSC自體移植回個體中之前,對個體msc離體 進行此基因療法》在一些實施例中,MSC可經遺傳修飾以 置換基因產物或添加或抑制基因產物。在一些實施例中, 遺傳工程改造可促進組織再生、治療疾病或改良MSC在植 入個體中之後的存活(亦即防止排斥)。用於轉染細胞之技 術在此項技術中已知。 若干細胞用於自體移植之目的,則熟習此項技術者可設 想眾多基因可向經轉染之個體間質細胞傳送有益性質。在 一些實施例中,視基因及實施例而定,所添加之基因可最 終保留在接受者MSC及所有其子代中,或可僅短暫保留》 舉例而言,編碼血管生成因子之基因可轉染入自周邊血分 離之MSC中。當MSC用於分化成心肌細胞及修復心肌時, 此等基因將適用於誘導側血管形成。在一些情形下,可能 需要用一種以上基因轉染細胞。 152716.doc -81 - 201130977 在一些情況下’需要使基因產物分泌。在此等情況下, 基因產物較佳含有促進蛋白質分泌之分泌信號序列。舉例 而言’若所要基因產物為血管生成蛋白,則熟習此項技術 者可選擇具有天然信號序列之血管生成蛋白,例如 VEGF ’或可使用常規遺傳操作(參見Nabel等人,ι993)修 飾基因產物以含有該種序列。 在本發明之一實施例中,自個體經調動之周邊血獲得之 MSC可以個別化檢定形式用於研究及瞭解個體MSC生長及 分化之信號傳導路徑。使用本發明之個體MSC適用於幫助 開發用於先天性及成人心臟衰竭之治療應用。使用本發明 之此等個體MSC能夠對特定分化成不同譜系(例如成骨譜 系、心臟譜系)進行研究,而不需要複雜、耗時的動物模 型。在另一實施例中,個趙MSC可經遺傳修飾以攜帶特定 疾病及/或特定疾病或病症之病理性性狀及表型。 在一實施例中’檢定包含複數個本發明之個體或其 分化子代。在一實施例中,檢定可用於研究個體Msc之分 化路徑,例如(但不限於)沿心肌細胞譜系、平滑肌譜系、 内皮譜系及此等譜系之亞群之分化。 在另一實施例中,檢定可用於研究個體Msc之病理性特 徵,例如疾病及/或與疾病或病症相關之遺傳特徵。在一 些實施例中,疾病或病症為創口癒合病症、骨病症或心血 管病症或疾病。在一些實施例中,個體Msc已經遺傳工程 改造以包含與疾病或病症相關之特徵。遺傳工程改造個體 MSC之此等方法為熟習此項技術者所熟知,且包括藉助於 152716.doc •82- 201130977 轉染(例如(但不限於)使用病毒載體轉染)或藉由此項技術 中已知之其他手段將核酸引入細胞中。 在一些實施例中,個體MSC可易於於提供靶向譜系分化 以及純系均質性之優點且能夠操作外部環境之實驗系統中 加以操作。此外,因為以實驗方式改變人類生殖系在倫理 上不可接受,所以ES細胞轉殖基因途徑不可為包括操作人 類基因之實驗所用。本發明之人類個體Msc中之基因靶向 允§午其中系統因倫理問題而不能活體内測試之領域中的重 要應用,且可適當重演彼特定個體之人類生物學或疾病過 程。 在另一實施例中’本發明之個體MSC可用於製備相對而 言未經優先表現於其他譜系之細胞中之cDna污染的cdna 集合庫。 本發明之個體MSC亦可用於製備對心肌細胞及其前驅體 之標記具有特異性的抗體。多株抗體可藉由以免疫原性形 式之本發明細胞注射脊椎動物加以製備。單株抗體之產生 描述於諸如美國專利第4,491,632號、第4,472,500號及第 4,444,887號、及 Methods in Enzymology 73B:3 (1981)之標 準參考文獻中。特異性抗體分子亦可藉由使免疫勝任細胞 或病毒粒子之集合庫與目標抗原接觸且長出經正向選擇之 純系來產生。參見Marks等人,New Eng. J. Med. 335:730, 1996 及 McGuiness 等人,Nature Biotechnol. 14:1449, 1996 °另一替代方案為將隨機DNA片段再裝配入抗體編碼 區域中,如EP專利申請案1〇94,1〇8 a中所述。 1527l6.doc •83· 201130977 抗體又可用於自混合細胞群中鑑別或補救(例如恢復表 型)具有所要表型的細胞,以達成諸如在使用組織樣品之 免疫診斷期間共染色、及使前驅細胞與末期分化個體Msc 及其他譜系之細胞分離的目的。 在另一實施例中’個體MSC可用於檢查在本發明之個體 MSC之分化期間及之後的基因表現概況。所表現之基因組 可與如此項技術中已知之來自不同個體或對照MSC樣品之 其他MSC亞群進行比較。可使用此項技術中已知之用於债 測特定mRNA之任何適合定性或定量方法》mRNA可藉由 例如與微陣列雜交、於組織切片中原位雜交、藉由逆轉錄 酶-PCR、或於含有p〇iy a+ mRNA之北方墨點中加以偵 測。熟習此項技術者可容易使用此等方法確定兩個樣品之 間的mRNA轉錄物之分子尺寸或分子量差異。 任何適用於偵測及比較樣品中之mRNA表現量之方法皆 可聯合本發明方法使用。舉例而言,樣品中之mRNA表現 量可藉由自樣品產生經表現之序列標籤(EST)之集合庫加 以測定。集合庫内EST之相對表現度(relative representati〇n) 之列舉可用於估計起始樣品内基因轉錄物之相對表現度。 測試樣品之EST分析之結果可接著與參考樣品的EST分析 相比較以確定所選聚核苷酸(特定言之,對應於本文所述 之一或多種經差異表現之基因的聚核苷酸)的相對表現 量。 或者,測試樣品中之基因表現可使用基因表現連續分析 (SAGE)方法(Velculescu 等人,Science (1995) 270:484)進 I52716.doc -84 - 201130977 打。簡而言之,SAGE包括自各轉錄物内之特定位置分離 短獨特序列標籤。序列標籤可經串聯、選殖及定序。起始 樣品内之特定轉錄物之頻率藉由相關序列標蕺與序列群相 遇之次數反映。 測試樣品中之基因表現亦可使用差異呈現(DD)方法分 析在DD中,由特定序列定界符(例如限制酶位點)限定之 片段用作基因之唯一識別符,偶合有關於片段長度或片段 在經表現基因内之位置的資訊。接著可依據所有可能片段 之集合池内與彼基因相關之片段的相對表現度估計樣品内 之經表現基因之相對表現度。進RDD之方法及組合物在 此項技術中已熟知,參見例如美國專利第5,776,683號;及 美國專利第5,807,6嶋。或者,制依據核Μ相互作用. 之特異性的雜交分析偵測樣品中之基因表現。募核苦酸或 cDNA可用於選擇性鑑別或捕捉具有特定序列組成之dna 或RNA,且定性或定量測定與已知捕捉序列雜交之rna或 cDNA之量以提供樣品中之細胞訊息集合池内特定訊息之 相對表現度的資訊。可設計雜交分析以允許藉由使用例如 具有高密度格式(包括過濾器、顯微鏡载片或微晶片)之陣 列基技術或使用光譜分析(例如質譜)之溶液基技術並行篩 檢數百至數千基因之相對表現。以下更詳細描述陣列在本 發明診斷方法中之一種例示性用途。 可進行與陣列之雜交’其中該料列可根據此項技術中 已知之任何適合方法產生。舉例而言,使用光引導合成技 術產生寡核苦酸之大陣列之方法描述於美國專利第 152716.doc •85· 201130977 5,134,854號及美國專利第5,445,934號中《使用電腦控制系 統’經由在許多反應位點同時偶合可將單體之異質陣列轉 化成聚合物之異質陣列。或者,藉由在固體基板上沈積預 合成之寡核苷酸來產生微陣列’例如如pct公開申請案第 WO 95/35505號中所述。用於收集樣品與陣列雜交之資料 之方法亦為此項技術中熟知的。舉例而言,細胞樣品之聚 核苷酸可使用可偵測螢光標記產生,且藉由針對可偵測標 記之存在掃描微陣列來偵測樣品中聚核苷酸之雜交。在裝 置上偵測經螢光標記之目標之方法及裝置在此項技術中已 知。一般而言,此等偵測裝置包括顯微鏡及將光導向基板 的光源。光子計數器偵測來自基板之螢光,而xy平移台 (translation stage)改變基板位置。可用於標的方法中之共 焦偵測裝置描述於美國專利第5,631,734號中。掃描雷射顯 微鏡描述於 Shalon等人 ’ Genome Res. (1996) 6:639 中。使 用適當激發光線(excitation line)對所用各螢光團進行掃 描。接著將掃描所產生之數位影像組合用於隨後分析。對 於任何特定陣列元件,來自一樣品之螢光信號之比率與來 自另一樣品之螢光信號相比較,且確定相對信號強度。用 於分析自雜交陣列所收集之資料之方法在此項技術中已熟 知。舉例而言’當雜交偵測包括螢光標記時,資料分析可 包括以下步驟:根據所收集之資料確定隨基板位置而變之 勞光強度’移除離群值’亦即偏離預定統計分佈之資料; 及根據剩餘資料計算目標之相對結合親和力。所得資料可 以影像呈現,其中各區域中之強度隨目標與探針之間的結 152716.doc -86- 201130977 合親和力而變化。樣式匹配可手動進行,或可使用電腦程 式進行。製備基板基質(例如陣列)、設計可供此等基質使 用之寡核苷酸、探針標記、雜交條件、掃描雜交基質、及 分析所產生樣式(包括比較分析)的方法描述於例如美國專 利第5,800,992號中。分子及細胞生物化學中之一般方法亦 可見於諸如以下之標準教科書中:A Laboratory Manual, 第 3 版(Sambrook 等人,Harbor Laboratory Press 2001); Short Protocols in Molecular Biology,第 4版(Ausubel等人 編,John Wiley & Sons 1999) ; Protein Methods(Bollag等 人,John Wiley & Sons 1996) ; Nonviral Vectors for Gene Therapy(Wagner 等人編,Academic Press 1999) ; Viral Vectors(Kaplift 及 Loewy 編,Academic Press 1995); Immunology Methods Manual(I. Lefkovits 編,Academic Press 1997);及 Cell and Tissue Culture: Laboratory Procedures in Biotechnology(Doyle及 Griffiths, John Wiley & Sons 1998)。本發明中提及之用於遺傳操作之試劑、選 殖載體及套組可購自商業供應商,諸如BioRad、 Stratagene、Invitrogen、Sigma-Aldrich及 ClonTech 〇 實例 方法 於灕衮序。所有個體皆在投與細胞以調動幹細胞之前接 受皮下注射G-CSF(Neupogen)480 pg為期至少2天或小於4 天,或240 pg/kg單次劑量之AMD3100(普樂沙福, Mozobil®,Genzyme Corporation, Cambridge, MA)且將在 152716.doc -87 - 201130977 投藥後當天經歷析離術。在經歷析離術之前,將抽取周邊 血樣品(第1天、第2天、第3天、第4天及第5天)以監測白血 球計數(WBC)且在第4天及第5天進行FACS分析以評估循環 中CD34 +細胞之調動功效以及測定析離參數。若個體WBC 高於60,000/mm3,則中止G-CSF之傳遞且儘早執行析離 術。析離術將在位於 Beth Israel Medical Center,Petrie Division之New York Blood Center執行(參見醫學主任28(3): 311-7; Pruijt et al., proc. Natl. Acad. Sci. U. S. A 1999; 96(19): 10863-8. The dosage, toxicity and therapeutic efficacy of a mobilizer such as G-CSF or GM-CSF can be determined by standard medical procedures such as standard medicine for the determination of LD50 (50% lethal dose in the population) and ED50 (50% therapeutically effective dose in the population). The program is determined, for example, in a cell culture or an experimental animal. The dose ratio between the toxic effect and the therapeutic effect is the therapeutic index and can be expressed as the ratio LD50/ED50. A mobilizer (such as G-CSF or GM-CSF) exhibiting a high therapeutic index is preferred. Although compounds exhibiting toxic side effects can be used, care should be taken to design a delivery system that targets these compounds to the site of the affected tissue. Minimize potential damage to uninfected cells and thereby reduce side effects. Information obtained from cell culture assays and animal studies can be used to determine the range of doses for human use. The dosage of such compounds is preferably in the range of circulating concentrations including ED50 with little or no toxicity. Depending on the dosage form employed and the route of administration employed, the dosage may vary within this range. The therapeutically effective dose of any of the compounds used in the methods of the invention can be initially estimated using cell culture assays. (4) It can be determined in the animal model to obtain the circulating gold concentration range 'which includes IC5 as determined in cell culture (that is, the test compound 丨 农度 which achieves the half maximum inhibition of symptoms). This information can be used for the determination of the application. The dose in humans. The amount in plasma can be measured by, for example, Southern Liquid Chromatography. According to another aspect of the invention, the MSC is mobilized as a single-agent from a certain tissue to/before other clinical procedures. Another method in tissue. These cells include non-hematopoietic positive f cells and in their life cycle to 152,716. Doc •65· 201130977 The potential to experience in vivo mobilization/migration during one stage of differentiation. According to another aspect of the present invention, there is provided a method of mobilizing an MSC during collection and during collection of an organ to be used for organ transplantation by infusion of an effective amount of a mobilizing agent such as GM-CSF or G-CSF. In some embodiments, the invention also provides methods of transferring MSCs from ex vivo or peripheral blood samples that have been collected from an individual using an ex vivo mobilizer such as GM-CSF or G-CSF. This includes, for example, in vivo perfusion of in vivo peripheral blood from a donor of a legitimate death organ prior to collection of PB-derived or BM-derived MSCs from a Zhao. In one embodiment, 'administering an effective amount of a mobilizing agent, such as G_CSF or GM-CSF', to the individual to mobilize the bm-derived MSC in the peripheral blood of the individual » In another embodiment, 'administering to the individual' An effective amount of a mobilizing agent, such as G-CSF or GM-CSF, to mobilize the pB-derived MSC in the peripheral blood of the individual » in another embodiment 'to make an effective amount of a mobilizing agent (such as g_csf or GM-CSF) Contacting a peripheral blood sample of the individual to mobilize the PB-derived MSC in the peripheral blood ex vivo. The "effective amount" is an amount sufficient to achieve a significant increase in the number and/or frequency of msc in the peripheral blood. An effective amount can be administered by one or more administrations, administrations, or administrations. The therapeutically effective amount of the mobilizing agent will depend on the chosen mobilizing agent (e.g., G-CSF or GM-CSF). A mobilizing agent such as G-CSF or GM-CSF may be administered one or more times per day to one or more times per week; including every other day. Those skilled in the art will appreciate that certain factors may affect the dosage and timing necessary to effectively treat an individual, including, but not limited to, prior treatment, overall health and/or age of the individual, and other conditions present. Furthermore, treating a subject with a therapeutically effective amount of G-CSF or GM-CSF as described herein can include a single treatment or a series of treatments. 152716. Doc-66-201130977 In some embodiments, the methods of treatment described herein comprise administering another mobilizing agent, for example, an agent selected from the group consisting of, but not limited to, the following: Interleukin-17 (IL-17; Journal Of Immunol. 2001; 167: 2081-2086), AMD3100 (Flomenberg et al., Acta Haematol. 2005; 114(4): 198-205), cyclophosphamide (Cy), European paclitaxel and (DXT) (Ojeifo et al., Experimental Hematology 2000; 28: 451-459). In some embodiments, the method comprises administering to the individual a PB-derived MSC or a BM-derived MSC or a differentiated progeny thereof, eg, reintroducing the cell into the same individual or transplanting the cell into a second individual (eg, an HLA-type match) Two individuals). In some embodiments, an individual that can be effectively treated with PB-derived MSCs or BM-derived MSCs includes any individual that can be typically treated with a bone marrow or stem cell transplant, such as having a cancer, such as a neuroblastoma (produced) Individuals in immature neurons that primarily affect cancer in infants and children, myelodysplasia, myelofibrosis, breast cancer, renal cell carcinoma, or multiple myeloma. For example, PB-derived MSCs or BM-derived MSCs can be transplanted into an individual with a cancer that is resistant to radiation therapy or chemotherapy, for example to restore high dose chemotherapy and/or radiation for the treatment of cancer. Stem cells destroyed by therapy. In some embodiments, the MSC can then be centrifuged in a blood bank serum centrifuge for about 1 minute. The supernatant can be decanted and the cells resuspended at about 1. 2 ml of sputum and fully swirled. 152716. Doc-67-201130977 Medium to long term storage All or a portion of the MSC population obtained by the methods disclosed herein or a substantially pure MSC population (such as intermediate or long term storage in a cell bank) is also within the scope of the invention. The medium to long term storage of the cells in the cell bank is disclosed in U.S. Patent Application Serial No. 2003/005433, the entire disclosure of which is incorporated herein by reference. At the end of the treatment, the MSC population or substantially pure MSC population as disclosed herein can be loaded into a delivery device such as a syringe for placement into the recipient individual by any means known to those of ordinary skill. In some embodiments, a population of MSCs obtained using the methods disclosed herein can be suspended in a short-term storage medium suitable for infusion into an individual, such as the medium disclosed in U.S. Patent No. 5,955,257, the disclosure of in. In some embodiments, MSCs collected and enriched using methods as disclosed herein can be stored (eg, for a donor (eg, allogeneic) or another body for future treatment) or directly transplanted, eg, to facilitate post-operative use Heal. In one aspect of the invention, the plurality of MSCs collected by the methods disclosed herein can be further sorted into at least two subpopulations which can be cryopreserved separately or together (e.g., contained in the same vial). In some embodiments at least two MSC subpopulations can be two different MSC subpopulations. For example, 14|5(: can be sorted into at least 2 cell subpopulations such as, but not limited to, (1) a stem cell population or a MSC rich group and (2) a non-stem cell population or an MSC exclusion group. It is also contemplated that two subpopulations (i.e., above (1) and (2)) may be cryopreserved together. In one embodiment, the sorting of MSCs into subpopulations may be positive or negative sorting or a combination thereof. In some embodiments, cell surface protein markers can be used for positive selection or 152716. Doc -68- 201130977 Select or remove stem cells negatively. Examples of such markers are well known in the art and are shown in Table 1: Table 1 Cell type markers Hematopoietic stem cells (HSC) CD34+, CD45+, CXCR4+ Endothelial progenitor cells CD34+, CD45+, CD133+, KDR+ Very small embryo-like cells (VSEL CD34+, CD133+, CXCR4+, SSEA4+ mesenchymal stem cells (MSC) CD34·, CD45_, CD90+, CD105+, CD106+, CD44+ In some embodiments, a MSC population or a substantially pure MSC population obtained by the methods disclosed herein can be visualized The condition is packaged in a suitable container in the presence of a suitable medium. In some embodiments, the package further comprises written instructions for the desired purpose, such as methods of implanting a MSC or a substantially pure MSC population into an individual to ameliorate or treat the disease (and, where appropriate, storage methods and/or thawing methods) (If cryopreserved), or a written instruction for a treatment plan for regenerative medicine or therapy. MSC for use in short cell therapy: In one embodiment of the invention, MSCs collected from peripheral blood mobilized by an individual as described herein require such cell therapy (e.g., autologous cell therapy) in the individual. It can be introduced or transplanted back into the individual. In some embodiments, MSCs can be used alone or in combination with other cells for untreated therapeutic applications in regenerative therapies, such as wound healing, cardiac repair and bone repair, and other bridge-shaped indications, as disclosed herein in the Examples. In some embodiments, the peripheral blood is mobilized from the individual by a method as disclosed herein 152716. Doc-69-201130977 The collection of MSCs can be used to repair or treat eye diseases such as retinopathy or macular degeneration, as discussed in Vossmerbaeumer et al., Cytotherapy, 2009; 11; 177-188, which is incorporated herein by reference in its entirety. in. In some embodiments, MSCs collected from peripheral blood mobilized by an individual as disclosed herein can differentiate into any number of different cell types originating from mesoderm, including adipocytes, osteoblasts, and chondrocytes, As discussed in Musina et al., Cell Technologies in Biology and Medicine, 2006; 2; 147-151, which is incorporated herein by reference in its entirety. Methods of implanting MSCs for the treatment of cardiovascular disorders and other disorders are well known to those of ordinary skill in the art and are disclosed in International Patent No. WO 2008/0548 19 and U.S. Patent Nos. 6,387,369, 7,514,074, 6,174,333 and 6,225,119, and U.S. Patent Application Serial No. These patents are incorporated herein by reference in their entirety in the entirety of the entire disclosure. In one embodiment of the invention, MSCs obtained from individual mobilized peripheral blood can be used in personalized medical applications, such as autologous therapeutic uses, and in personalized assays to assess personal diet, pharmacogenetics, nerves The impact of chemicals and lifestyles on MSC function and vitality. A MSC composition comprising some or all of the MSCs collected from peripheral blood of an individual by methods as disclosed herein can be used to repair, treat or ameliorate various aesthetic or functional conditions (eg, defects) via augmentation of damaged tissue^ MSCs collected from peripheral blood of an individual by methods as disclosed herein provide an important resource for rebuilding or supplementing damaged tissue. In one embodiment, MSCs collected from peripheral blood of an individual by methods as disclosed herein can be used in tissue engineering and regenerative medicine to replace 152716. Doc •70· 201130977 The body part of the defect, injury, disease or aging wear and tear. MSCs collected from peripheral blood of an individual by methods as disclosed herein provide a unique system in which msc can differentiate into distinct cells and produce a particular lineage or genotype of the same individual. Thus, MSCs collected from peripheral blood of an individual by the methods disclosed herein provide significant advantages of individualized stem cell therapy. Moreover, in the absence of disease or injury, such Mscs and compositions thereof collected from peripheral blood of an individual by methods as disclosed herein may be added to the soft tissue region 'splits, depressions or voids (such as For flattening to supplement soft tissue unrelated to injury. The invention also encompasses multiple and continuous administration of MSCs collected from peripheral blood of an individual by methods as disclosed herein. For stem cell-based treatment, by this document Methods of Revealing MSC populations collected from peripheral blood of an individual are preferably collected from an autologous source. The MSC composition is then prepared and isolated as described herein, and in particular, with other stem cell populations present in the peripheral blood of the mobilized Separation. In order to introduce or transplant a mononuclear cell suspension from a peripheral gold collection of an individual and/or a composition thereof into a human or animal recipient by a method as disclosed herein. The solution contains a concentrate of MSC and can be separated from other stem cells present in the peripheral blood, and the Msc is collected and resuspended in a physiologically acceptable load. Or in an excipient or diluent. Alternatively, the MSC suspension may be contained in a serum-free sterile solution (such as a cryopreservation solution). In some embodiments, an MSC-rich formulation may also be used. In some embodiments The MSC suspension can then be introduced into one or more sites of the donor tissue via, for example, injection. Doc • 71 · 201130977 The concentrated or enriched stem cells can be administered in the form of a pharmaceutically or physiologically acceptable formulation or composition containing a physiologically acceptable carrier, excipient or diluent, and An organization that deals with the recipient's organisms, including humans and non-human animals. MSCs can be prepared by resuspending the cells in a suitable liquid or solution, such as sterile saline or other physiologically acceptable injectable aqueous liquid. (Composition. The amount of each component in such compositions. It can be determined in a conventional manner by those skilled in the art. In some embodiments, 'a population of MSCs or a combination thereof collected from a peripheral blood of Zhao as disclosed herein can be administered as follows: a stem cell suspension is placed Adhering to an absorbent or adhesive material (ie, a collagen sponge matrix) and inserting the material containing the MSC into the relevant site or related site, or 'by collecting blood from the peripheral blood of the individual by methods not disclosed herein. % of the population can be administered by parenteral injection (including subcutaneous, intravenous, intramuscular, and intrasternal injection). Other modes of administration include, but are not limited to, intranasal, intrathecal, intradermal, transdermal, and intestinal. And sublingual administration. In one embodiment, the MSC population collected from the peripheral blood of the individual by the method disclosed can be administered by endoscopic surgery. Method of Revealing MSCs The composition of MSCs collected from the peripheral blood of an individual can be suspended in a sterile solution or suspension or can be resuspended in a pharmaceutically and physiologically acceptable aqueous or oily vehicle, which may contain a preventive agent. Stabilizers and substances which cause the solution or suspension to be isotonic with the body fluid (ie, blood) of the recipient. Non-limiting examples of suitable excipients include water, sodium salt buffered saline (pH 7 4), 〇15 M gasification aqueous solution, dextrose, glycerin, dilute ethanol and their analogues, and their mixtures 152,716. Doc -72· 201130977 Things. Illustrative stabilizers are polyethylene glycols, proteins, sugars, amino acids, inorganic acids, and organic acids that can be used alone or in combination. The amount or amount and the route of administration employed will depend on the individual and will be equivalent to the amount employed in a similar dosage form or indication known to those skilled in the art, in accordance with the present invention, by the method disclosed herein from the periphery of the individual. Blood-collected MSC populations can be administered to body tissues, including epithelial tissue (ie, skin, lumen, etc.), muscle tissue (ie, smooth muscle), blood, brain, and various organ tissues, such as those associated with the urinary system ( That is, bladder, urethra, ureter, kidney, etc.). According to the general treatment methods described herein, a population of MSCs collected from peripheral blood of an individual by methods as disclosed herein may comprise other cells (eg, a portion of a cell comprising a population of target stem cells) and may be administered to the individual, eg, by intravenous (ι·ν. The catheter is infused into the bloodstream of the individual (similar to any other intravenous fluid). Alternatively, however, an individualized mixture of MSC populations can be generated, such as to provide a cell therapy mixture specifically for the therapeutic needs of the individual. A comprehensive cell mixture such as obtained via a separation method can be characterized for sorting and separation into different cell populations. Cell markers (such as VSEL, MSC, and HSC markers) or tissue-specific markers can be used to phenotypically characterize a population of cells harvested from peripheral blood. Using these markers, it is possible to separate and sort based on cell type. The cell mixture is thereby converted into a population of cells which can be broadly divided into two parts: a stem cell fraction and a non-stem cell fraction. The non-stem cell fraction can be further divided into progenitor cells or fibroblast fractions as well as functional cells or fully differentiated cell fractions. Once the peripheral blood cell mixture is sorted, the MSC group and the non-stem cell parts can be cryopreserved and stored separately. In this way, it can produce 152716. Doc •73- 201130977 Born from a collection or repository of different cell groups of Zhao. Alternatively, the Msc population and non-stem cell fractions can be cryopreserved together and then sorted and separated prior to use. In some embodiments, a population of MSCs collected from peripheral blood of an individual by methods as disclosed herein can be used to produce or differentiate into any population of a certain cell type, which is derived from germ layers (ie, endoderm, mesoderm, and ectoderm) ) developed. Such cells include, but are not limited to, differentiated cells, neural progenitor or differentiated cells, glial progenitor or differentiated cells, oligodendrocyte glial progenitor or differentiated cells, dermal progenitor or differentiated cells, hepatic progenitor cells or Differentiated cells, muscle progenitor cells or differentiated cells, osteoprogenitor or differentiated cells, mesenchymal stem cells or progenitor cells, pancreatic progenitor cells or differentiated cells, progenitor cells or differentiated chondrocytes, stromal progenitor cells or differentiated cells, cultured and expanded Stem cells or progenitor cells, cultured differentiated stem or progenitor cells, or a combination thereof. Progenitor cells are also of interest, such as hematopoiesis, nerves, stroma, muscle (including smooth muscle), liver, lung, gastrointestinal and mesenchymal progenitor cells. Also contemplated are differentiated MSC populations such as differentiated into cells such as, but not limited to, osteoblasts, hepatocytes, granulocytes, chondrocytes, myocytes, adipocytes, neuronal cells, pancreatic cells, or combinations thereof and mixture. In some embodiments, a population of MSCs collected from peripheral blood of an individual by methods as disclosed herein can be combined, recombinantly or compounded into a mixture of cell therapy cells suitable for treating disease and/or tissue regeneration in a subject. The combination of a MS C population collected from a peripheral blood of Zhao and a tissue-specific progenitor cell and optionally a functional cell by a method as disclosed herein can be used, for example, to enhance the graftability of the transplanted MSC. 152716. Doc • 74· 201130977 Accordingly, in one embodiment, the present invention provides methods and products for using an autologous mixture of MSC populations collected from peripheral blood of an individual by methods as disclosed herein. This MCS population can be used alone or in combination with other suitable cells. In some embodiments, the MSC can be combined with other stem cells derived from peripheral blood, or other functionally suitable non-stem cells. Thus, individual MSCs can be used alone or selectively recombined (customized mix) for individualized autologous therapeutic applications in regenerative therapies. In other words, the autologous MSC population can be selectively combined with other cells, including stem cells, such as other non-MSC stem cells derived from peripheral blood, for autologous cell therapy. In some embodiments, the mixed MSC population can comprise from about 10% to about 90% of MSCs collected from peripheral blood of the individual by methods as disclosed herein, from about 10% to about 80%, or from about 10% to about 60%. %, or from about 10% to about 40%, or from about 10% to about 90% of MSCs collected from peripheral blood of an individual by methods as disclosed herein. In some embodiments, the MSC population may also comprise non-stem cells (eg, functional cells) and/or populations of stem cells that are also derived from peripheral blood, such as from about 5% to about 50% functional cells and/or non-MSCs. Stem cells, from about 5% to about 40% functional cells and/or non-MSC stem cells, from about 5% to about 30% functional cells and/or non-MSC stem cells, from about 5% to about 20% functional cells and/or non-MSC stem cells, Or from about 5% to about 10% functional cells and/or non-MSC stem cells. One of the above cell therapy products is suitable as an example of an autologous mixture of HSC and MSC or other functional cells of the hematopoietic system. Another example is a cell therapy product comprising an autologous MSC population mixed with other cells, such as at least one or any combination of the following cell types: HSC, PBSC, 152716. Doc -75- 201130977 Cardiac progenitor cells and cardiomyocytes as appropriate. Accordingly, in another embodiment, a method of treating a patient in need thereof, comprising administering to the individual, alone or in combination (separately or in combination), the MSC population collected from peripheral blood of the individual by a method as disclosed herein Automated Character 0 Stem Cell Banking In another aspect of the invention, a population of MSCs collected from peripheral blood of an individual by methods as disclosed herein can be stored in a cell bank to support optional health care insurance. Models that effectively protect group members from future diseases. Individual individuals can be treated and preserved from the periphery when they are in a healthy state: collect their own stem cell population for future distribution of their health care needs. Thus, in an embodiment, the MSC population collected from the peripheral blood of the individual is "stored" in a stem cell bank or a storage or storage facility, or in a place where the target stem cell population is preserved for storage by a method as disclosed herein. Future use. The storage wire can be designed in such a way that a catastrophic event such as a nuclear attack can be designed in a manner that is safe from the periphery of the individual by the method disclosed herein. In or in the cellar. In other embodiments, it may be on the side of the mountain or in outer space. Stored in the mask 4 can be embedded in (4) such as the wrong masking material. According to a further embodiment, providing a 4-step stem cell reservoir (10) comprises administering to the individual one or more mobilizing agents to increase the amount of moths in the peripheral blood of the individual. The steps are included by 152716. Doc • 76 · 201130977 Peripheral blood collects MSC populations, where the individual does not have immediate health conditions, such as no conditions in which individuals need to use their own target stem cell population for their own autologous transplantation. Step C includes preserving the population of MSCs collected from peripheral blood by a method as disclosed herein in the form of a deposited cell population. Step D includes obtaining a deposited MSC population collected by a method as disclosed herein to autologously transplant the MSC population into the individual. Reference will be made in detail to the embodiments of the invention, examples of which are illustrated in the accompanying drawings. In some embodiments of the invention, the MSC is separated from the mobilized blood using an Elutra® centrifuge manufactured by Gambro BCT, Inc. of Lakewood, Colorado. Although the embodiments of the present invention are described in connection with an Elutra® centrifuge, this reference is for illustrative purposes only and is not intended to limit the invention in any sense. It will be appreciated that embodiments of the invention may use other centrifuges including, but not limited to, also by Gambro BTC Inc. COBE® Spectra separation system, Trima® system and Trima Accel® system, as well as other panning devices for separating blood components. MSC is used for personalized medical diagnosis and prognostic testing. Another aspect of the invention relates to the use of MSCs derived from individual mobilized peripheral blood for personalized medical applications, such as autotherapeutic uses, and for personalization assays to assess personal diet, medications The effects of genetics, neurochemicals, and lifestyle on MSC function and vitality. Such assays are currently well known in the art and include high yield screening in which a compound, agent or environmental stimulator is contacted with MSC, and can measure the function of a compound, agent or environmental stimulator against MSC (eg, differentiation potential) , proliferation, survival) and vitality effects, and the results are compared with reference control samples (eg not 152716. Doc -77· 201130977 The presence of a compound) or a positive control sample 'such as the presence of BMP6 and the like in the aspect of the invention, the milk C population obtained from the individual can be used in high yield screening to assess the individual versus the agent Individual reactions (such as for specific drugs, hormone therapy, or medicine or therapy). Briefly, the MSC cell population is contacted with the relevant agent and the agent is effected by monitoring the output parameters (such as marker expression, fine tongue feature, pluripotency, and the like). The MSC can be freshly isolated using methods as disclosed herein, or in some embodiments, cultured, cryopreserved, or genetically engineered prior to use in assaying t. In some embodiments, the individual's position may be an environmental change of the pure line culture (4): for example, splitting into independent poplar and growing under unique conditions (eg, with or without virus; presence or absence of other cytokines or combinations thereof) . Importantly, because MSCs are specific to individual genetic makeup, the use of MSCs provides a tool for pharmacogenetic analysis of MSCs. For example, individual MSCs can have specific variants that result in unique pathological features. For example, because Msc is derived from an individual (e.g., autologous)', it has the desired pathology (4), e.g., a diseased lesion or a mutation and/or polymorphism that results in a poor or enhanced response to a therapeutic agent. Thus, Msc obtained by the methods as disclosed herein can be used to assess an individual's response to a particular compound, an agent's MSC, and in some cases this may be applicable to the prognosis of the disease. In this embodiment, the method of the invention can be used to screen for agents that alleviate lesions, or agents that positively affect MSC proliferation or function. In an alternative embodiment, the method of the invention can be used for screening as compared to 152716. Doc . 78 · 201130977 Other individual MSCs (eg, no mutations and/or polymorphisms) that cause different MSC responses in an individual (due to intrinsic genetic composition or specific mutations and/or polymorphisms), so these methods are available For example, assessing the effects of specific drugs and/or agents on individual Msc populations compared to other human and/or MSC cells, thus acting as a high yield screening for personalized medicine and/or pharmacogenetics. The manner in which MSCs react to a drug (specifically, a pharmacological agent) (including the timing of the reaction) is an important reflection of the physiological state of the cell. The parameters are quantifiable components of MSC, in particular, components that are accurately measured in high yield systems. The parameter can be any cellular component or cellular product, including cell surface determinants, receptors, proteins or their conformational or post-translational modifications, lipids, carbohydrates, organic or inorganic molecules, nucleic acids (eg, mRNA, DNA) Etc.), or derived from a portion of the cellular component, or a combination thereof. Although most parameters will provide a quantitative reading, in some cases semi-quantitative or qualitative results are acceptable. The readings may include a single measurement, or may include an average, median or deviation, and the like. A characteristic range of parameter readings for each parameter will be obtained using many of the same assays. Variability is expected and a range of values for each test parameter set will be obtained using standard statistical methods and common statistical methods for providing a single value. Compounds including candidate agents are obtained from a wide variety of sources, including pools of synthetic or natural compounds. For example, a variety of means are available for the random and directed synthesis of a wide variety of organic compounds, including biomolecules, including the expression of randomized oligonucleotides and oligopeptides. Alternatively, a collection of natural compound pools in the form of bacterial, fungal, plant and animal extracts may be utilized or readily produced. In addition, collections and compounds produced naturally or synthetically are readily accessible via 152716. Doc -79· 201130977 Conventional chemical, physical and biochemical modifications and can be used to generate combinatorial collection libraries. Oriented or random chemical modifications can be made to known pharmacological agents, such as deuteration, alkylation, acetification, aromatization, etc., to produce structural analogs. The agent is screened for the effect of the agent on the stem cell population by adding the drug (usually along with the cells lacking the agent) to at least one and typically a plurality of stem cell samples. The change in parameters that are responsive to the agent is measured and the results are evaluated by comparison to a reference culture (e.g., obtained with other agents in the presence and absence of the agent, etc.). In some embodiments, the agent is preferably added to the culture medium of the stem cells in culture in solution or in a soluble form. The agent may be added in a flow or through system in the form of a batch or continuous flow, or the compound bolus may be added one by one or incrementally to another still solution. In a flow through system, 'two fluids' are used, one of which is a physiologically neutral solution and the other is the same solution as the added test compound. The first fluid and the second fluid are sequentially passed over the stem cells' in a single solution method' the test compound bolus is added to a volumetric medium surrounding the cells. The total concentration of the components of the medium should not vary significantly with the addition of the bolus, or in the flow-through method, it should not vary significantly between the two solutions. In some embodiments, the pharmaceutical formulation does not include other components, such as preservatives, that can have a significant effect on the overall formulation. Therefore, it is preferred that the substance is substantially composed of a biologically active compound and a physiologically acceptable carrier such as water, ethanol, DMSO or the like. However, if the compound is a solventless liquid, the formulation may consist essentially of the compound itself. 152716. Doc -80- 201130977 Multiple tests can be run in parallel with different drug concentrations to obtain differential responses to various concentrations. As is known in the art, a range of concentrations produced on a 1:10 or other logarithmic scale, dilution, is typically used to determine the effective concentration of the agent. If necessary, the concentration can be further improved by the dilution of the second series. Typically, one of these concentrations acts as a negative control, i.e., at or below the level of detection of the agent or at or below the concentration of the drug that does not produce a detectable phenotypic change. The individual MSCs used for screening can be manipulated to express the desired gene product, e.g., to confer undesirable physiological and/or polymorphic phenomena in the genetic makeup of the MSC. Thus, the gene therapy is performed on the individual msc ex vivo before the individual MSC is autologously transplanted back into the individual. In some embodiments, the MSC can be genetically modified to replace the gene product or to add or inhibit the gene product. In some embodiments, genetic engineering can promote tissue regeneration, treat disease, or improve survival (i.e., prevent rejection) of MSCs after implantation in an individual. Techniques for transfecting cells are known in the art. A number of cells are used for autologous transplantation, and those skilled in the art will be able to contemplate that a wide variety of genes can transfer beneficial properties to transfected individual mesenchymal cells. In some embodiments, depending on the optic gene and the examples, the added gene may ultimately remain in the recipient MSC and all of its progeny, or may be only transiently retained. For example, a gene encoding an angiogenic factor may be transduced. Dyeing into MSCs isolated from peripheral blood. When MSCs are used to differentiate into cardiomyocytes and repair myocardium, these genes will be useful for inducing lateral vascularization. In some cases, it may be desirable to transfect cells with more than one gene. 152716. Doc -81 - 201130977 In some cases, it is necessary to secrete the gene product. In such cases, the gene product preferably contains a secretion signal sequence that promotes secretion of the protein. For example, if the desired gene product is an angiogenic protein, those skilled in the art can select an angiogenic protein with a native signal sequence, such as VEGF' or can modify the gene product using conventional genetic manipulation (see Nabel et al., ι 993). To contain such a sequence. In one embodiment of the invention, MSCs obtained from individual mobilized peripheral blood can be individually assayed for use in studying and understanding the signaling pathways of individual MSC growth and differentiation. The individual MSCs of the invention are useful for helping to develop therapeutic applications for innate and adult heart failure. The use of such individual MSCs of the present invention enables the study of specific differentiation into different lineages (e.g., osteogenic lineage, cardiac lineage) without the need for complex, time consuming animal models. In another embodiment, a MSC can be genetically modified to carry a particular disease and/or a pathological trait and phenotype of a particular disease or condition. In one embodiment, the assay comprises a plurality of individuals of the invention or their differentiated progeny. In one embodiment, the assay can be used to study the differentiation pathways of individual Msc, such as, but not limited to, differentiation along the myocardial cell lineage, the smooth muscle lineage, the endothelial lineage, and subpopulations of such lineages. In another embodiment, the assay can be used to study the pathological features of an individual Msc, such as a disease and/or genetic characteristics associated with the disease or condition. In some embodiments, the disease or condition is a wound healing condition, a bone condition, or a cardiovascular condition or disease. In some embodiments, the individual Msc has been genetically engineered to include features associated with the disease or condition. These methods of genetically engineering individual MSCs are well known to those skilled in the art and include by means of 152716. Doc • 82- 201130977 Transfection (such as, but not limited to, transfection with a viral vector) or introduction of nucleic acids into cells by other means known in the art. In some embodiments, an individual MSC can be readily manipulated in an experimental system that provides advantages in targeting lineage differentiation as well as homologous homogeneity and is capable of operating an external environment. Furthermore, because experimentally altering the human germline is ethically unacceptable, the ES cell transgene pathway cannot be used for experiments involving the manipulation of human genes. The gene targeting in the human individual Msc of the present invention allows for the important application of the system in which the system cannot be tested in vivo due to ethical issues, and can appropriately reproduce the human biology or disease process of a particular individual. In another embodiment, the individual MSCs of the invention can be used to prepare cDna-contaminated cdna pools of libraries that are relatively unexpressed in cells of other lineages. The individual MSCs of the invention can also be used to prepare antibodies specific for the signature of cardiomyocytes and their precursors. Multiple antibodies can be prepared by injecting vertebrate cells in the immunogenic form of the cells of the invention. The production of monoclonal antibodies is described in standard references such as U.S. Patent Nos. 4,491,632, 4,472,500 and 4,444,887, and Methods in Enzymology 73B:3 (1981). Specific antibody molecules can also be produced by contacting a pool of immunocompetent cells or virions with a target antigen and growing a positively selected line. See Marks et al., New Eng. J. Med. 335:730, 1996 and McGuiness et al., Nature Biotechnol. Another alternative to 14:1449, 1996 ° is to reassemble random DNA fragments into the antibody coding region as described in EP Patent Application Serial No. 94,1,8 a. 1527l6. Doc •83· 201130977 Antibodies can in turn be used to identify or remediate (eg, restore phenotype) cells from a mixed cell population with the desired phenotype to achieve co-staining, such as during immunodiagnosis using tissue samples, and to make precursor cells and terminal cells The purpose of cell differentiation of differentiated individuals Msc and other lineages. In another embodiment, an individual MSC can be used to examine gene expression profiles during and after differentiation of individual MSCs of the invention. The expressed genome can be compared to other MSC subpopulations from different individual or control MSC samples known in the art. Any suitable qualitative or quantitative method known in the art for the determination of a particular mRNA can be used. mRNA can be hybridized, for example, to a microarray, in situ hybridization in a tissue section, by reverse transcriptase-PCR, or The northern blot of p〇iy a+ mRNA is detected. Those skilled in the art can readily use these methods to determine the molecular size or molecular weight difference of mRNA transcripts between two samples. Any method suitable for detecting and comparing the amount of mRNA expression in a sample can be used in conjunction with the methods of the invention. For example, the amount of mRNA expression in a sample can be determined by generating a pool of expressed sequence tags (ESTs) from the sample. An enumeration of relative representations of ESTs in the pool can be used to estimate the relative degree of expression of the gene transcripts within the starting sample. The results of the EST analysis of the test sample can then be compared to the EST analysis of the reference sample to determine the selected polynucleotide (specifically, a polynucleotide corresponding to one or more of the differentially expressed genes described herein) The relative amount of performance. Alternatively, gene expression in test samples can be performed using the Gene Expression Continuous Analysis (SAGE) method (Velculescu et al., Science (1995) 270: 484) into I52716. Doc -84 - 201130977 Hit. Briefly, SAGE involves the isolation of short unique sequence tags from specific locations within each transcript. Sequence tags can be linked, colonized, and sequenced. The frequency of a particular transcript within the starting sample is reflected by the number of times the associated sequence label meets the sequence group. The gene expression in the test sample can also be analyzed using the differential representation (DD) method in DD, and the fragment defined by the specific sequence delimiter (eg, restriction enzyme site) is used as the unique identifier of the gene, coupled to the length of the fragment or Information about the location of the fragment within the expressed gene. The relative degree of expression of the expressed genes in the sample can then be estimated based on the relative performance of the fragments associated with the gene within the pool of all possible fragments. Methods and compositions for entering RDD are well known in the art, see, for example, U.S. Patent No. 5,776,683; and U.S. Patent No. 5,807,6. Alternatively, the system is based on nuclear interactions. A specific hybridization assay detects gene expression in the sample. The nucleus acid or cDNA can be used to selectively identify or capture a DNA or RNA having a specific sequence composition, and qualitatively or quantitatively determine the amount of rna or cDNA that hybridizes to a known capture sequence to provide a specific message within the pool of cell information in the sample. Information on the relative performance. Hybridization assays can be designed to allow for hundreds to thousands of screenings by using, for example, array-based techniques with high density formats (including filters, microscope slides or microchips) or solution-based techniques using spectral analysis (eg, mass spectrometry) The relative performance of genes. An exemplary use of the array in the diagnostic method of the present invention is described in more detail below. Hybridization with the array can be performed' wherein the column can be produced according to any suitable method known in the art. For example, a method for producing a large array of oligonucleotides using photo-guided synthesis techniques is described in U.S. Patent No. 152,716. The heterogeneous array of monomers can be converted to a heterogeneous array of polymers by simultaneous coupling at a number of reaction sites in U.S. Patent No. 5,445,934 and U.S. Patent No. 5,445,934. Alternatively, the microarray can be produced by depositing a pre-synthesized oligonucleotide on a solid substrate, e.g., as described in PCT Publication No. WO 95/35505. Methods for collecting data for hybridization of a sample to an array are also well known in the art. For example, a polynucleotide of a cell sample can be generated using a detectable fluorescent label and the hybridization of the polynucleotide in the sample can be detected by scanning the microarray for the presence of the detectable label. Methods and apparatus for detecting fluorescently-marked objects on a device are known in the art. Generally, such detection devices include a microscope and a light source that directs light to the substrate. The photon counter detects the fluorescence from the substrate and the xy translation stage changes the substrate position. Confocal detection devices that can be used in the subject method are described in U.S. Patent No. 5,631,734. Scanning laser microscopy is described in Shalon et al.' Genome Res. (1996) 6:639. Each of the fluorophores used was scanned using an appropriate excitation line. The digital images produced by the scan are then combined for subsequent analysis. For any particular array element, the ratio of the fluorescent signal from one sample is compared to the fluorescent signal from another sample and the relative signal strength is determined. Methods for analyzing data collected from hybrid arrays are well known in the art. For example, when the hybridization detection includes a fluorescent marker, the data analysis may include the steps of: determining, according to the collected data, the intensity of the light that changes with the position of the substrate, 'removing the outliers', that is, deviating from the predetermined statistical distribution. Data; and calculate the relative binding affinity of the target based on the remaining data. The resulting data can be presented as images, with the intensity in each region following the knot between the target and the probe 152716. Doc -86- 201130977 Change with affinity. Style matching can be done manually or using a computer program. Methods of preparing substrate matrices (eg, arrays), designing oligonucleotides for use with such matrices, probe labels, hybridization conditions, scanning hybrid matrices, and analyzing the resulting patterns (including comparative analysis) are described, for example, in US Patent No. No. 5,800,992. General methods in molecular and cellular biochemistry can also be found in standard textbooks such as: A Laboratory Manual, 3rd edition (Sambrook et al., Harbor Laboratory Press 2001); Short Protocols in Molecular Biology, 4th edition (Ausubel et al.) Edited by John Wiley & Sons 1999); Protein Methods (Bollag et al., John Wiley & Sons 1996); Nonviral Vectors for Gene Therapy (edited by Wagner et al., Academic Press 1999); Viral Vectors (edited by Kaplift and Loewy, Academic Press 1995); Immunology Methods Manual (I. Lefkovits, ed., Academic Press 1997); and Cell and Tissue Culture: Laboratory Procedures in Biotechnology (Doyle and Griffiths, John Wiley & Sons 1998). The reagents, selection vectors and kits for genetic manipulation referred to in the present invention are commercially available from commercial suppliers such as BioRad, Stratagene, Invitrogen, Sigma-Aldrich and ClonTech®. All individuals received a subcutaneous injection of G-CSF (Neupogen) 480 pg for at least 2 days or less, or 240 pg/kg of a single dose of AMD3100 (Pulesaf, Mozobil®, before administration of cells to mobilize stem cells). Genzyme Corporation, Cambridge, MA) and will be at 152716. Doc -87 - 201130977 After the administration of the drug, it was subjected to separation. Peripheral blood samples (Day 1, Day 2, Day 3, Day 4, and Day 5) were taken to monitor white blood cell count (WBC) and on Days 4 and 5 before undergoing the separation procedure. FACS analysis was performed to assess the modulating efficacy of CD34+ cells in the circulation and to determine the separation parameters. If the individual WBC is higher than 60,000/mm3, the delivery of G-CSF is discontinued and the separation is performed as early as possible. Separation will be performed at the New York Blood Center at the Petri Division in Beth Israel Medical Center (see Medical Director)
Vijay Shah之隨附證明信)。 MSC分雜及播智。所有個體之析離產物皆如下所述進行 處理以根據標準技術分離用於細胞療法之MSC » 經調動之PB中之各種細胞群將使用菲科-帕克(Fic〇I1_ Paque)、隨後藉由於50 ml錐形管中梯度離心加以分離。在 已4分入50 ml官中之各10 ml菲科-帕克液體上形成2〇如 PB層。此藉由緩慢及溫和地使pb沿管側向下流至流趙上 達成。分層管在室溫下在400x g下離心30分鐘^ pB一經離 心,即將中間白血球層置入T25培養燒瓶中。每個τ25燒瓶 將使用7 ml MSC完全培養基。每週兩次藉由光學顯微術檢 查細胞之污染情況及細胞形態’且更換培養基。在更換培 養基時洗滌未黏附細胞。在細胞匯合之後,其將傳代至 T75燒瓶中》對於患者之細胞療法,將使用早期傳代(至多 5次)。在向患者傳遞細胞之前,對最終細胞產品進行品質 控制測試’包括表型表徵、核型分析、支原體 (mycoplasma)及細菌測試。細胞劑量將為“〇〇,〇〇〇個細 胞。 152716.doc -88- 201130977 f瀠身邀培#之M5T。自T-75燒瓶移除培養基。用1〇如 lx無菌PBS洗滌各燒瓶2分鐘。移除PBS且添加丨.2 mi姨蛋 白酶(TryPsin)-EDTA至各燒瓶中。旋轉燒瓶以使胰蛋白酶 完全包覆各燒瓶之底部。使燒瓶返回至培育箱中維持i 〇分 鐘。藉由檢查細胞圓整性及細胞移動性來檢查適當姨蛋白 酶處理(trypsinization)。藉由添加5 ml MSC培養基至各燒 瓶中使腺蛋白酶- EDTA不活化。使用吸液管來回緩和洗蘇 佈滿瓶底的培養基以使細胞捕捉最大化。使用同一吸液管 將細胞/培養基混合物轉移至離心管中。將來自所有燒瓶 之細胞及培養基合併入1個50 ml離心管中。在室溫下在 400 g(1400 rpm)下離心細胞7分鐘。小心移除上清液且將 細胞小球再懸浮於10 ml無菌生理鹽水(自藥房獲得之標準 0.9〇/。生理鹽水)中。在室溫下在400 g(14〇〇 rpm)下離心細 胞7分鐘。重複此程序2次。小心移除上清液且藉由將其懸 浮於1.5 ml鹽水中並獲取此懸浮液稀釋10倍(1〇 4於1〇〇 ^ 培養基中)之少量等分試樣來對細胞計數。血球計孔需要 10 μΐ流體樣品。 向窗邀授#以浴療;σ瘡合:MSC接著與如下所述之血 纖維蛋白凝膠混合且以4週間隔施用於創口 3次。 製備及施用稀釋之血纖維蛋白凝膠/細胞混合物。认飞 說明利用 1 ml TISSEEL VH套組(Baxter Healthcare,inc.) 製備稀釋之血纖維蛋白凝膠。此套組係利用兩個液相可 經由雙室施用器擠壓或經由具有惰性氣體載劑之施用器 喷灑。在此稀釋血纖維蛋白凝膠中,塗封蛋白(sealer 152716.doc -89 - 201130977 protein)及凝血因子(thrombin)之最終濃度分別為25單位/毫 升。 藉由添加0.221 g CaClz粉末至50 ml無菌生理鹽水中且 在組織培養通風櫥下經0.22 μπι管頂過濾器過濾混合物來 製備30 mM鈣(CaCh)於生理鹽水(0·9% NaCl)中之無菌混 合物。為了配製凝血因子溶液,1 ml瓶裝凝血因子於加熱 混合器中溶解20分鐘。一旦此混合物溶解,即用注射器自 瓶抽出0.5 ml且與4.5 ml無菌CaCb混合物混合。此混合物 於加熱混合器中保持溫熱。在初始溶解步驟之4小時内使 用此稀釋凝血因子溶液。為了配製塗封蛋白,使用i W注 射器添加1 ml無菌生理鹽水至塗封蛋白瓶中且於加熱混合 器中溶解20分鐘。一旦此生理鹽水溶解,即用注射器自瓶 抽取0.5 ml且與4.5 ml無菌生理鹽水混合。此混合物於加 熱混合器中保持溫熱。在初始溶解步驟之4小時内使用此 稀釋塗封蛋白。如下形成凝膠:首先視創口尺寸而定,決 定凝膠之施用總量。兩種液體之每一者將為此總量之一 半。在1 ml吸液管中’向上抽吸如塗封蛋白/細胞混合物之 最終體積所必需之相同量的塗封蛋白。藉由於含有細胞之 離心管中藉由緩和地向上及向下吸移(4次)來混合細胞與塗 封蛋白。將塗封蛋白/細胞混合物向上抽入喊保氣泡已移 除之1 ml注射器中。將與塗封蛋白/細胞混合物之體積相同 體積之凝血因子溶液向上抽人另—1 ml注射nt ;再次敲 出任何氣泡。移除針且將兩個注射器扣人紅色施用器中。 接著將白色./透明施用器接頭置於注射器開口上。嗔霧 152716.doc 201130977 時,將無菌氣體軟管與施用器接頭連接且在施用期間保持 氣體供應流動以防止在施用器接頭中凝結。若將擠出而非 喷出凝膠,則持續穩定緩慢擠壓直至所有流體用盡以避免 在施用器接頭中凝結。 CFU-C測試(群落形成單位培養) 調整MEM(流動)中之股骨髓細胞數目至2 5 x i 〇6個細胞/ 毫升。0.2 ml此懸浮液與〇·5 mi馬血清、〇.丨mi硫代甘油 (20 mM ’用MEM稀釋4倍)、1.〇 mi曱基纖維素於mem 中)、〇·6 ml MEM(流動)及O.i mi額外培養基或標準化經刺 激小鼠血清(在腹膜内投與2.5 mg/kg脂多醣(LPS)之後3小 時抽取之血清之200倍稀釋液)或5 ng/ml rhG-CSF混合。充 分混合之半固體懸浮液吸入直徑為4 CIn之皮氏培養皿 (Petri dish)中且在37°C ' 5% C02及95%相對濕度下培育6 天。在添加0.5 ml對蛾确基四β坐鐵(p-i〇d〇nitrotetrazolium) 紫色溶液(0.5 mg/ml PBS)之後,將培養皿再培育24小時。 群落使用群落計數器計數且校準至1〇 6個骨髓細胞。 實例1 自周邊血中分離MSC且擴增 本發明人係利用來源於經調動之周邊血之MSC,其中將 粒細胞群落刺激因子(G-CSF)投與個體以自骨髓調動MSC 至周邊循環中。藉由菲科分離及收集含有單核細胞之白血 球層來使經調動之周邊血富含幹細胞。來自白血球層之細 胞接著接種於含有MSC生長培養基之細胞培養燒瓶中。在 前兩週之定期(每3天)培養基更換期間洗去未黏附細胞。黏 152716.doc •91· 201130977 附細胞接著於MSC生長培養基中擴增至多5次傳代。 證明MSC由G-CSF調動至周邊血中 本本發明人已證明,作為對2次連續皮下注射G-CSF(每 天480 pg)的反應’ MSC被調動至健康個體之周邊血中·β在 此實驗中’本發明人證明’在藉由多參數流動式細胞測量 分析法所分析之調動前及經調動之周邊血樣品中,富含表 現 MSC 表型(亦即 CD45-、CD34-、CD105+、CD90+、 CD29+)之大量細胞。圖6顯示在G-CSF調動之後,健康人 類供者(η=5)之周邊血中之MSC數目顯著增加。單尾配對τ 檢驗已顯示’呈 CD45-、CD34-、CD90+、CD105+、 CD29+之MSC對G-CSF調動充分反應且相較於調動前樣 品、,調動後樣品中之MSC數目顯著增加(ρ=〇.03)。 自經調動之周邊血獲得之MSC樣品的用途 慢性缺血性、糖尿病性及靜脈鬱滯創口影響數百萬美國 人且導致大量病態(morbidity),包括功能喪失、慢性疼痛 症候群、及全身性感染、甚至致命性感染之風險增加。甚 至當潛伏病狀已經改善時,皮膚創口痊癒之阻力亦可與以 下相關:創口内之細胞(特定言之,「組織中」之幹細胞)衰 老’基於疾病的對存在於成功創口療合中之正常細胞-細 胞及細胞-基質信號傳導之異常反應;或壞死性碎片及/或 瘢痕,其對粗及顯微組織產生破壞,阻止體液或細胞對癒 合過程之作用。特定言之,骨髓源性幹細胞之全身性細胞 移行(trafficking)可在創口癒合中起重要作用且針對嫁接之 此等内源性障壁可為重要的。使用已自具有慢性創口之患 152716.doc •92· 201130977 者分離,經離體培養且施用於創口的人類自體骨髓源性 (非周邊血源性)間質幹細胞(MSC)可對受影響肢體之所有 組織層進行顯著功能性組織恢復且瘢痕形成最小。因此, 本發明之一態樣係關於根據如本文揭示之方法自個體獲得 之PB源性MSC的用途,MSC可自具有已自骨髓調動之更容 易得到之循環MSC的個體(其中該等個體已經投與粒細胞 群落刺激因子)獲得且自周邊血獲得。因此,該方法顯著 優於直接自患者骨髓分離細胞之先前研究(一種可為費 力、疼痛且昂貴之程序且在一些情況下不實用)。本發明 人已證明自血液分離並捕捉Msc之程序使得獲得用於創口 癒合之MSC之程序更簡單且實際上允許可在損傷之前收集 並儲存幹細胞以供隨後使用。因此,本發明人證明直接對 慢性未癒合皮膚創口施用經培養之自體循環Msc可達成顯 著改善或完全創口癒合及功能復原及最小瘢痕形成。G_ CSF誘導MSC自骨髓調動至周邊循環中,隨後執行行析離 術以分離MSC。MSC在培養中生長,傳代至多5次,接著 (以血纖維蛋白混合物形式)在3個時間點局部施用於創口, 其中各時間點之間隔為4週。 鍺廣从SC廣忑兴未4砰龙居若]^^(:用於創口修復及癒 合’則並非所有創口的癒合程度皆可能相同。判定各患者 中可能傾向於或抑制創口癒合之潛伏病狀將為有用的。因 此,可儲藏MSC用於未來分析。可能之預期輔助研究包括 (但不限於)比較定量分析(藉由ELISA個別地或經由全面蛋 白質組研究篩檢)調動前及調動後相關循環MSC群、血清 152716.doc •93· 201130977 細胞激素及趨化因子;來自創口之鑽孔活檢體中細胞及擴· 散性因子之分化、活化、增殖及細胞凋亡標記隨時間的免 疫組織化學及原位雜交分析。本發明人可在治療前及治療 後使用MSC來預先對患者循環細胞群、血清及創口部位取 樣且儲藏此等樣品以用於包括(但不限於)以下之進一步研 究。 析離前及治療之每一天(但在治療之前)、及最終治療之 後1個月之血清。此等樣品可用於相關個別循環趨化因子/ 細胞激素之ELISA評估,或有希望用於各患者及患者之間 在不同時間點之完全蛋白質組研究分析及比較。 治療之每一天(但在治療之前)、最終治療之後1個月之 創口之周邊及中心的鑽孔活檢體。各活檢體將對半分割, 一片用於冷凍組織,一片用於福馬林固定及石蠟包埋。此 等組織可用於根據標準方案對細胞群及局部擴散細胞激素 及趨化因子及丼在細胞膜上之受體、增殖及細胞凋亡標記 進行免疫組織化學染色。 在用G-CSF調動之前及在析離時(亦即GSF後)對周邊血 中循環幹細胞之不同亞群進行定量,該等亞群包括(但不 限於):CD34+、c-kit+、thy-11。、Sca-1+、lin.(「KTLS 細 胞」)、極小胚胎樣幹細胞(VSELS)及MSC。 實例2 使用如本文所述之方法獲得之MSC可用於治療或修復骨 損壞及用於其他橋形適應症。尤其關注的是PB源性MSC及 BM源性MSC治療以降低劇烈體格訓練期間應力骨折(stress 152716.doc -94· 201130977 fracture)之發病率、或修復骨損傷(使用習知治療方法(例 如月刺激、激素及固定)而未癒合)的用途。在美國人口中 每年有約800萬例骨折且近1〇%癒合受損。一般而言,在 軍隊中’在基本訓練期Μ ’男性中之應力骨折率多達 5%,在女性中比率多達21%。在陸軍中,男性骨折數字為 2·6%且女㈣折數字8.1%。處於骨折風險中之特^骨路部 位為尺骨(步搶訓練);股頸(應力骨折);上肢及下肢及下 頜骨、顱面骨及鎖骨之暴發性損傷(blast injury)。此等骨 折導致軍隊之所有分支每年_萬美元醫藥成本及值勤時 間損失。因此’使用如本文揭示之方法自個體獲得之自體 MSC可促進不癒合骨折之癒合。 使用經皮置放、培養擴增之自體間質幹細胞進行骨折不 癒合治療 長骨骨折不癒合為大幅增加平均骨折治療成本之嚴重健 康問題。治療可採用若干形式,通常需要手術,包括骨移 植及/或初始切開復位、内固定(〇RIF)程序之檢核 (revision)。更保守選項包括配合手術方法使用骨形態發生 蛋白及骨刺激物。此等程序具有相對較高失敗率,表明明 顯需要更新穎更有效方法來促進不癒合骨折之治療。 在此研究中,本發明人證明藉由如本文揭示之方法獲得 之PB源性MSC及BM源性MSC可安全且有效用於修復骨損 壞’且證明經由放射照相引導之注射投與PB源性Mg匸及 BM源性MSC具有促進骨折不癒合中之骨再生的初步功 效。此等PB源性MSC及BM源性MSC藉由增加骨再生來增 152716.doc -95· 201130977 強癒合之組織環境以及恢復功能。 不希望受理論束缚,骨為經歷涉及新形成與再吸收之間 平衡之連續重塑的特異化組織。當骨被破壞時,間質幹細 胞(MSC)募集至損傷部位。MSC為可見於人類成人組織, 包括骨髓(BM)、滑液組織、脂肪組織及周邊血(PB)中之多 能細胞。因為其來源於中胚層,所以MSC能夠分化成骨、 軟骨、肌肉及脂肪組織。正常骨折癒合不能在不存在内源 可用MSC下進行,因為此等細胞會分化成為骨修復所需之 多種細胞。儘管藥物尚未特別有效促進骨折癒合,但許多 研究已表明對不癒合骨折部位直接施用MSC可使受損骨有 效癒合。MSC譜系已在許多動物模型及人類中成功再生軟 骨及骨。因此,藉由如本文揭示之方法獲得之自體PB源性 MSC及BM源性MSC可以允許細胞分化成骨母細胞及為癒 合所需之其他成熟細胞之方式向骨折部位投與。因此,如 本文揭示之PB源性MSC及BM源性MSC適用作治療不癒合 骨折之骨再生產品。 因此,根據如本文揭示之方法自經調動之血液獲得的PB 源性MSC及BM源性MSC可用於治療各種病狀,包括骨折 不癒合。 本發明人已證明自經調動之周邊血分離有活力之MSC的 方法,且此等MSC可用於針對慢性創口及矯形適應症之自 體MSC細胞療法的方法中。因為MSC具有自體性,所以排 斥反應之風險有限或無風險。本發明人已證明投與G-CSF (480 gg,2天,皮下)顯示可將MSC自人類BM調動至PB。 152716.doc •96· 201130977 或者,本發明人亦已證明MSC可藉由單次劑量之AMD3100 (240 pg/kg,皮下)調動至周邊血中。本發明人證明PB源性 MSC及BM源性MSC可藉由如本文揭示之方法收集且視情 況隨後擴增及/或超低溫保存。 自體BM-MSC及PB-MSC治療骨折不癒合之功效 為了評估MSC治療不癒合骨折之功效,本發明人執行12 個月隨機安慰劑對照雙盲多中心1期臨床試驗,該試驗採 用總計100名患者,分成4組,每組25名患者。一組用BM-MSC治療,第二組用PB-MSC治療且2個對照組用自體血小 板溶解產物(PL,用於擴增MSC)或磷酸鹽緩衝鹽水(PBS) 治療。患者經招募且在4所矯形專業醫院加以治療。細胞 在1個中心細胞培養機構經處理並擴增。藉由骨折部位之 薄層CT量測可判定不癒合骨折癒合的主要終點。若骨折不 癒合顯著小於(P<〇.05)對照治療組,則將評估MSC之功 效。 方法: #禮衮序汔括.· 1.全骨髓抽吸:與骨髓收集程序一致, 抽取200立方公分(CC)肝素化靜脈内靜脈血以用於產生自體 PL。將骨髓吸出物抽入2個含有30,000 IU肝素之30 ml注射 器中。分離有核細胞且於PBS中洗滌1次,計數,且接著再 懸浮於DMEM+10% PL中且連同已收集之200 ml全靜脈内 血液一起置放於4°C運輸包裝中且在24小時内運輸至細胞 培養場所。 2. MSO之####謂磨及從# .·藉由在收集之前4小時 152716.doc -97- 201130977 使用AMD3100(240 pg/ kg,皮下)來調動患者。執行析離 術以收集約300 cc經調動之周邊血。抽取約2〇〇 cc肝素化 靜脈内靜脈血以用於產生自體血小板溶解產物(pL)。析離 產物連同已收集之200 ml全靜脈内血液一起置放於4。〇運 輸包裝中且經由24小時運輸傳遞至細胞培養場所。 3 ·冷邀MSC#增.·細胞以1 X〖〇6個細胞/cm2接種於單層燒 瓶中且在37 C /5% C〇2下於含濕氣環境中培育。3天後更換 培養基(含有自體PL),移除未黏附細胞,MSC群將在接種 之後6-12天形成。各培養物在達到4〇_5〇%匯合之後以1:3 傳代。MSC生長至第1次-第3次傳代,接著懸浮於磷酸鹽 緩衝鹽水(PB S)及符合其最大活體外培養擴增率之自體pL 濃縮物中。 矣治#產忑屢理。移植程序:最終MSC劑量為可以高達 1.0xlO7個細胞之預期產量利用的最大劑量。MSC依次置放 入無菌注射器、無菌傳遞袋及4»c冷卻器中以便運輸至操 作室中。工作人員所需之說明書將連同產品一起包括在 内。患者將返回至無菌操作室中且不癒合部位之區域將使 用必達淨(betadine)及無菌手套進行預備處理。無菌套管針 接著在c形臂(c-arm)上在若干位置處插入骨折部位中。用 PBS稀釋2倍之威視派克(visipaque)將用於確定骨折部位中 放射照相對比劑之流動。將注射樣品注射入可確定染料流 動之區域中。移除套管針。指示患者保持靜止3〇分鐘以允 許細胞附著。 結果終點量測:使用操作前、1、3、6及12個月時骨折 152716.doc -98- 201130977 部位之薄層CT掃描確定MSC改善骨折部位癒合的功效。檢 視膠片(scout film)用於確定骨折部位面積。為了限制輻射 曝露,視野確定為骨折部位近端2 cm及遠端2 cm。 實例3 本發明人已事先證明,人類臍帶血含有小(尺寸小於紅 血球)CXCR4+CD133+CD34+SSEA-4+Oct-4+liiTCD45·細胞群 2007:21;297-303)且此等細胞在組織器官損壞期 間調動至周邊血中,如在例如心梗塞〇/. Co//.Vijay Shah's accompanying letter). MSC is mixed and broadcasted. All individual isolates were processed as described below to isolate MSCs for cell therapy according to standard techniques. » The various cell populations in the mobilized PB will use Fic〇I1_ Paque, followed by 50 The ml conical tube was separated by gradient centrifugation. Two layers of PB, such as PB, were formed on each of the 10 ml Fico-Parker liquids that had been divided into 4 ml of 50 ml. This is achieved by slowly and gently flowing pb down the tube side to the flow. The layered tube was centrifuged at 400 x g for 30 minutes at room temperature. Once pB was centrifuged, the intermediate white blood cell layer was placed in a T25 culture flask. Each τ25 flask will use 7 ml MSC complete medium. The contamination and cell morphology of the cells were examined by light microscopy twice a week and the medium was changed. Wash unadhered cells when changing the medium. After the cells are confluent, they will be passaged into T75 flasks. For cell therapy for patients, early passage (up to 5 times) will be used. Quality control tests were performed on the final cell product prior to delivery of the cells to the patient' including phenotypic characterization, karyotyping, mycoplasma and bacterial testing. The cell dose will be "〇〇, 〇〇〇 cells. 152716.doc -88- 201130977 f潆身培培# of M5T. Remove the medium from the T-75 flask. Wash each flask with 1〇 as lx sterile PBS 2 Minutes. Remove PBS and add 2.2 mi chymotrypsin (TryPsin)-EDTA to each flask. Rotate the flask to allow trypsin to completely coat the bottom of each flask. Return the flask to the incubator for i 〇 minutes. Check for proper trypsinization by examining cell roundness and cell mobility. Adenosine-EDTA is not activated by adding 5 ml of MSC medium to each flask. Use a pipette to gently relax the bottle of shampoo Bottom medium to maximize cell capture. Transfer the cell/culture mixture to the centrifuge tube using the same pipette. Combine cells and media from all flasks into one 50 ml centrifuge tube. At room temperature at 400 The cells were centrifuged for 7 minutes at g (1400 rpm). The supernatant was carefully removed and the pellet was resuspended in 10 ml sterile saline (standard 0.9 〇 / saline obtained from the pharmacy). 400 g (14〇 The cells were centrifuged for 7 minutes at rpm. This procedure was repeated twice. Carefully remove the supernatant and dilute it 10 times by suspending it in 1.5 ml of saline and obtaining this suspension (1〇4 in 1〇〇^ medium) A small aliquot of the sample is used to count the cells. The hemacytometer well requires 10 μΐ of the fluid sample. The window is invited to # bath therapy; sputum combination: MSC is then mixed with the fibrin gel as described below and 4 Weekly intervals were applied to the wound 3 times. Preparation and application of diluted fibrin gel/cell mixture. Description of the fly The diluted fibrin gel was prepared using 1 ml TISSEEL VH kit (Baxter Healthcare, inc.). The system utilizes two liquid phases that can be extruded via a dual chamber applicator or via an applicator with an inert gas carrier. In this diluted fibrin gel, the coated protein (sealer 152716.doc -89 - 201130977) The final concentration of protein and thrombin was 25 units/ml, respectively. Prepared by adding 0.221 g of CaClz powder to 50 ml of sterile physiological saline and filtering the mixture through a 0.22 μπι top filter in a tissue culture hood. 30 A sterile mixture of mM calcium (CaCh) in physiological saline (0.99% NaCl). To prepare the clotting factor solution, dissolve 1 ml of the bottled clotting factor in a heated mixer for 20 minutes. Once the mixture is dissolved, use a syringe from the bottle. 0.5 ml was withdrawn and mixed with 4.5 ml of sterile CaCb mixture. This mixture was kept warm in the heating mixer. This diluted clotting factor solution was used within 4 hours of the initial dissolution step. To prepare the encapsulated protein, 1 ml of sterile physiological saline was added to the coated protein vial using an i W syringe and dissolved in a heated mixer for 20 minutes. Once the physiological saline was dissolved, 0.5 ml was taken from the bottle with a syringe and mixed with 4.5 ml of sterile physiological saline. This mixture was kept warm in the heating mixer. This diluted coating protein was used within 4 hours of the initial dissolution step. The gel is formed as follows: first depending on the size of the wound, the total amount of application of the gel is determined. Each of the two liquids will be one-half of this total. The same amount of encapsulated protein as necessary to coat the final volume of the protein/cell mixture was aspirated in a 1 ml pipette. The cells and the coated protein were mixed by gently pipetting up and down (4 times) in the centrifuge tube containing the cells. The coated protein/cell mixture is pumped up into a 1 ml syringe that has been removed. The volume of the coagulation factor solution of the same volume as the coated protein/cell mixture is pumped up another 1 ml to inject nt; any bubbles are knocked out again. The needle was removed and the two syringes were snapped into the red applicator. The white./transparent applicator fitting is then placed over the syringe opening.嗔 fog 152716.doc 201130977, the sterile gas hose is connected to the applicator joint and the gas supply flow is maintained during application to prevent condensation in the applicator joint. If the gel is to be extruded rather than ejected, continue to steadily and slowly squeeze until all fluid is used up to avoid condensation in the applicator fitting. CFU-C test (community forming unit culture) Adjust the number of femoral bone marrow cells in MEM (flow) to 2 5 x i 〇 6 cells/ml. 0.2 ml of this suspension with 〇·5 mi horse serum, 〇.丨mi thioglycerol (20 mM 'diluted 4 times with MEM), 1. 〇mi曱 based cellulose in mem), 〇·6 ml MEM ( Flow) and Oi mi additional medium or standardized stimulated mouse serum (200-fold dilution of serum taken 3 hours after intraperitoneal administration of 2.5 mg/kg lipopolysaccharide (LPS)) or 5 ng/ml rhG-CSF . The fully mixed semi-solid suspension was drawn into a 4 CIn Petri dish and incubated for 6 days at 37 ° C '5% C02 and 95% relative humidity. After adding 0.5 ml of a purple solution of p-i〇d〇nitrotetrazolium (0.5 mg/ml PBS), the culture dish was incubated for another 24 hours. The community was counted using a community counter and calibrated to 1 to 6 bone marrow cells. Example 1 Isolation of MSCs from Peripheral Blood and Amplification The present inventors utilized MSCs derived from mobilized peripheral blood, wherein granulocyte community stimulating factor (G-CSF) was administered to individuals to mobilize MSCs from the bone marrow to the peripheral circulation. . The mobilized peripheral blood is enriched in stem cells by Fico separation and collection of leukocytes containing monocytes. The cells from the white blood cell layer were then seeded in a cell culture flask containing MSC growth medium. Unadhered cells were washed off during the first two weeks of regular (every 3 days) medium change. Adhesive 152716.doc •91· 201130977 The attached cells were then expanded for up to 5 passages in MSC growth medium. Proof that MSC is mobilized from G-CSF to peripheral blood. The inventors have demonstrated that as a response to two consecutive subcutaneous injections of G-CSF (480 pg per day), MSCs are mobilized to the peripheral blood of healthy individuals. The 'inventors' demonstrated that the MSC phenotype (ie, CD45-, CD34-, CD105+, CD90+) is enriched in the pre-mobilization and mobilized peripheral blood samples analyzed by multiparameter flow cytometry analysis. , CD29+) a large number of cells. Figure 6 shows a significant increase in the number of MSCs in the peripheral blood of healthy human donors (η = 5) after G-CSF mobilization. The one-tailed paired τ test has shown that 'MSCs with CD45-, CD34-, CD90+, CD105+, CD29+ respond adequately to G-CSF mobilization and the number of MSCs in the sample after mobilization is significantly increased compared to the sample before mobilization (ρ= 〇.03). Use of MSC samples obtained from mobilized peripheral blood Chronic ischemic, diabetic, and venous stasis wounds affect millions of Americans and cause a large number of morbidities, including loss of function, chronic pain syndrome, and systemic infections Even the risk of fatal infections increases. Even when the latent condition has improved, the resistance to skin wound healing can be related to the following: cells in the wound (specifically, stem cells in the "tissue") aging' disease-based pair exists in successful wound healing Abnormal reactions of normal cell-cell and cell-matrix signaling; or necrotic debris and/or scars that disrupt coarse and microscopic tissue, preventing the action of body fluids or cells on the healing process. In particular, systemic cell migration of bone marrow-derived stem cells can play an important role in wound healing and can be important for grafting such endogenous barriers. Human autologous bone marrow-derived (non-peripheral blood-derived) mesenchymal stem cells (MSC) isolated from ex vivo culture and applied to wounds can be affected by isolation from patients with chronic wounds 152716.doc •92· 201130977 Significant functional tissue recovery and minimal scar formation were observed in all tissue layers of the limb. Thus, one aspect of the invention pertains to the use of PB-derived MSCs obtained from an individual according to methods disclosed herein, which may be from individuals having circulating MSCs that have been more readily available from bone marrow mobilization (where such individuals have Induction of granulocyte community stimulating factor) was obtained and obtained from peripheral blood. Thus, this approach is significantly superior to previous studies directly isolated from patient bone marrow cells (a procedure that can be laborious, painful, and expensive, and in some cases not practical). The inventors have demonstrated that the procedure for isolating and capturing Msc from blood makes the procedure for obtaining MSC for wound healing simpler and actually allows for the collection and storage of stem cells prior to injury for subsequent use. Thus, the inventors have demonstrated that direct administration of cultured autologous circulating Msc to chronic non-healing skin wounds can achieve significant improvement or complete wound healing and functional recovery with minimal scar formation. G_CSF induces MSC transfer from the bone marrow to the peripheral circulation, followed by performing a ligation procedure to isolate the MSC. MSCs were grown in culture, passaged up to 5 times, and then topically applied (in the form of a fibrin mixture) to the wound at 3 time points with an interval of 4 weeks at each time point.锗广从SC广忑兴4砰龙居若]^^(:for wound repair and healing', not all wounds may have the same degree of healing. Determine the latent disease that may tend to or inhibit wound healing in each patient It will be useful. Therefore, MSCs can be stored for future analysis. It is possible to expect complementary studies including, but not limited to, comparative quantitative analysis (by ELISA alone or via comprehensive proteomic research screening) before and after mobilization. Related circulating MSC group, serum 152716.doc •93· 201130977 cytokines and chemokines; differentiation, activation, proliferation and apoptosis markers of cells and expansion and diffusion factors in the biopsy of the mouth Histochemistry and in situ hybridization analysis. The inventors can use MSC to pre-sample and recycle the patient's circulating cell population, serum and wound sites before and after treatment for use including, but not limited to, the following further Study. Pre-separation and each day of treatment (but before treatment), and serum 1 month after the final treatment. These samples can be used for related individual cycles. ELISA for chemokine/cytokine assessment, or promising for complete proteomic analysis and comparison between patients and patients at different time points. Every day of treatment (but before treatment), 1 month after final treatment Drilling biopsy around the wound and at the center. Each biopsy will be split in half, one for frozen tissue and one for formalin fixation and paraffin embedding. These tissues can be used to target cell populations and parts according to standard protocols. Immunohistochemical staining of diffuse cytokines and chemokines and receptors, proliferation and apoptosis markers on the cell membrane. Before mobilization with G-CSF and at the time of separation (ie after GSF) in peripheral blood Different subpopulations of circulating stem cells are included, including but not limited to: CD34+, c-kit+, thy-11, Sca-1+, lin. ("KTLS cells"), minimal embryonic stem cells ( VSELS) and MSC. Example 2 MSCs obtained using the methods described herein can be used to treat or repair bone damage and for other bridge-shaped indications. Of particular concern are PB-derived MSCs and BM-derived MSCs. Use of stress fractures during stress physical training (stress 152716.doc -94·201130977 fracture), or repair of bone injuries (using conventional treatments (eg, monthly stimulation, hormones and fixation) without healing). There are about 8 million fractures and nearly 1% of the fractures are damaged every year. In general, the stress fracture rate in men in the basic training period is up to 5%, and the ratio in women is as high as 21%. In the Army, the male fracture number is 2.6% and the female (four) discount number is 8.1%. The special bone site at the risk of fracture is the ulna (step grab training); the femoral neck (stress fracture); the upper and lower limbs and A blast injury of the mandible, craniofacial bone, and clavicle. These fractures result in a 10,000-dollar medical cost and loss of duty time per year for all branches of the military. Thus, autologous MSCs obtained from an individual using methods as disclosed herein can promote healing of non-healing fractures. Fracture nonunion treatment using percutaneous placement and culture of expanded autologous mesenchymal stem cells Non-healing of long bone fractures is a serious health problem that significantly increases the cost of average fracture treatment. Treatment can take several forms and usually requires surgery, including bone grafting and/or initial open reduction, internal fixation (〇RIF) procedures for revision. More conservative options include the use of bone morphogenetic proteins and bone stimulators in conjunction with surgical procedures. These procedures have a relatively high failure rate, indicating a clear need for newer and more effective methods to promote the treatment of non-healing fractures. In this study, the inventors demonstrated that PB-derived MSCs and BM-derived MSCs obtained by the methods disclosed herein are safe and effective for repairing bone damage 'and demonstrate that radiographically-directed injections are administered to PB-derived Mg匸 and BM-derived MSCs have the initial effect of promoting bone regeneration in fracture nonunion. These PB-derived MSCs and BM-derived MSCs increased the tissue environment and recovery function of 152716.doc-95·201130977 by increasing bone regeneration. Without wishing to be bound by theory, bone is a specialized tissue that undergoes continuous remodeling involving a balance between new formation and resorption. When the bone is destroyed, the mesenchymal stem cells (MSC) are recruited to the site of injury. MSCs are pluripotent cells found in human adult tissues, including bone marrow (BM), synovial tissue, adipose tissue, and peripheral blood (PB). Because it is derived from the mesoderm, MSCs are able to differentiate into bone, cartilage, muscle and adipose tissue. Normal fracture healing cannot be performed in the absence of endogenous MSCs, as these cells differentiate into the various cells required for bone repair. Although drugs have not been particularly effective in promoting fracture healing, many studies have shown that direct administration of MSC to non-healing fracture sites can effectively heal damaged bone. The MSC lineage has successfully regenerated soft bone and bone in many animal models and humans. Thus, autologous PB-derived MSCs and BM-derived MSCs obtained by the methods disclosed herein can allow cells to differentiate into osteoblasts and to deliver to the fracture site in a manner that is desirable for other mature cells. Thus, PB-derived MSCs and BM-derived MSCs as disclosed herein are useful as bone regeneration products for the treatment of non-unsaturated fractures. Thus, PB-derived MSCs and BM-derived MSCs obtained from mobilized blood according to the methods disclosed herein can be used to treat a variety of conditions, including fracture non-healing. The inventors have demonstrated a method of isolating viable MSCs from mobilized peripheral blood, and such MSCs are useful in methods of autologous MSC cell therapy for chronic wounds and orthopedic indications. Because MSCs are self-contained, the risk of rejection is limited or risk free. The inventors have demonstrated that administration of G-CSF (480 gg, 2 days, subcutaneous) shows that MSC can be mobilized from human BM to PB. 152716.doc • 96· 201130977 Alternatively, the inventors have demonstrated that MSC can be mobilized into peripheral blood by a single dose of AMD 3100 (240 pg/kg, subcutaneously). The inventors demonstrate that PB-derived MSCs and BM-derived MSCs can be collected by methods as disclosed herein and subsequently expanded and/or cryopreserved as appropriate. Efficacy of autologous BM-MSCs and PB-MSCs in the treatment of fracture nonunion In order to evaluate the efficacy of MSC in the treatment of nonunion fractures, the inventors performed a 12-month randomized, placebo-controlled, double-blind, multicenter phase 1 clinical trial using a total of 100 trials. The patients were divided into 4 groups of 25 patients each. One group was treated with BM-MSC, the second group was treated with PB-MSC and the two controls were treated with autologous platelet lysate (PL, for amplifying MSC) or phosphate buffered saline (PBS). Patients were recruited and treated in 4 orthopaedic professional hospitals. The cells were processed and expanded in a central cell culture facility. The primary endpoint of nonunion fracture healing can be determined by thin-layer CT measurements of the fracture site. If the fracture nonunion is significantly less than the (P<〇.05) control treatment group, the efficacy of the MSC will be assessed. METHODS: #礼衮序汔·· 1. Whole bone marrow aspiration: Consistent with the bone marrow collection procedure, 200 cubic centimeters (CC) of heparinized intravenous venous blood was drawn for the production of autologous PL. The bone marrow aspirate was drawn into two 30 ml syringes containing 30,000 IU of heparin. The nucleated cells were isolated and washed once in PBS, counted, and then resuspended in DMEM + 10% PL and placed in a 4 ° C shipping package along with the collected 200 ml of total intravenous blood for 24 hours. Transport to the cell culture site. 2. MSO's ####说磨和从# .· Use AMD3100 (240 pg/kg, subcutaneous) to mobilize the patient 4 hours before collection 152716.doc -97- 201130977. Separation was performed to collect approximately 300 cc of mobilized peripheral blood. About 2 cc of heparinized intravenous venous blood was drawn for the production of autologous platelet lysate (pL). The separated product was placed in 4 along with the collected 200 ml of total intravenous blood. Transfer to the cell culture site via a 24-hour transport. 3 · Coldly invite MSC# to increase cells. The cells were inoculated in a single-layer flask at 1 X [〇6 cells/cm2 and cultured in a moisture-containing environment at 37 C /5% C〇2. After 3 days, the medium (containing autologous PL) was replaced, and unadhered cells were removed, and the MSC group was formed 6-12 days after inoculation. Each culture was passaged 1:3 after reaching 4〇_5〇% confluence. MSCs were grown to passage 1st through passage 3, followed by suspension in phosphate buffered saline (PB S) and autologous pL concentrates that meet their maximum in vitro culture expansion rates.矣治# 忑 忑 忑 忑. Transplant procedure: The final MSC dose is the maximum dose that can be utilized for up to 1.0 x 107 cells. The MSCs are placed in a sterile syringe, sterile delivery bag, and 4»c cooler for transport to the operating room. The instructions required by the staff will be included with the product. The patient will return to the sterile operating room and the area of the non-healing site will be prepared using betadine and sterile gloves. The sterile trocar is then inserted into the fracture site at several locations on the c-arm. Visipaque, diluted 2 times with PBS, will be used to determine the flow of radiographic contrast agents in the fracture site. The injected sample is injected into the area where the dye flow can be determined. Remove the trocar. The patient is instructed to remain stationary for 3 minutes to allow for cell attachment. RESULTS Endpoint measurement: Fractures were used before, at 1, 3, 6 and 12 months of operation. 152716.doc -98- 201130977 The thin-layer CT scan of the site determined the efficacy of MSC in improving the healing of the fracture site. A scout film is used to determine the area of the fracture site. To limit radiation exposure, the field of view was determined to be 2 cm proximal to the fracture site and 2 cm distal. Example 3 The inventors have previously demonstrated that human umbilical cord blood contains small (size smaller than red blood cells) CXCR4+CD133+CD34+SSEA-4+Oct-4+liiTCD45·cell population 2007:21; 297-303) and these cells are During tissue damage, mobilize to the surrounding blood, such as in the case of myocardial infarction / Co / /.
CarAo/.,2009:53;1-9.)及中風(《Siroh. 2009:40;1237_)中所 見。亦已報導鼠類器官中的類似細胞,且更重要的是,已 描述此等細胞可活體外分化成所有3個胚層之細胞 (Zewkwz’a 2006:20;857-869)。為了研究人類 VSEL可成為 再生醫學中之多能幹細胞來源之可能性,本發明人開發出 自成人患者分離此等細胞之高效策略》 本發明人已證明,VSEL與其鼠類對應物類似,可在注 射粒細胞群落刺激因子(G-CSF)之後調動至周邊血中(汾em Ce//i 2008:26;2083-2092)。本發明人藉由招募一組使用G-CSF(每天480 pg,皮下)連續2天經調動之年輕健康供者來 證明此現象。在第3天,藉由血液析離術收集有核細胞 (TNC)。 本發明人評估G-CSF調動之前及之後周邊血(PB)樣品中 之VSEL數目以及析離產物中之最終數目。獲得至少1〇〇萬 個TNC且藉由FACS Diva軟體加以分析。分析富含VSEL (Lin7CD457CD133+ 、 Lin7CD457CD34+ 、 Lin*/CD457 152716.doc ·99· 201130977 CXCR4+)之非造血幹細胞、以及其CD45陽性造血對應物之 3個不同部分。根據各個別樣品之各群體之百分比含量及 TNC計數計算1 pL樣品中所含之各群細胞的絕對數。 本發明人確定在G-CSF調動之後,人類周邊血含有表現 0又0114、€034及€0133抗原之如-€045-單核細胞之群體。 此等lin_CD45XXCR4+CD133+CD34+細胞高度富含核内萬 能胚胎轉錄因子(諸如Oct-4、Sox2及Nanog)之mRNA。更 重要的是,本發明人發現,〇ct-4表現於經調動之VSEL之 核中且此等細胞亦表現細胞表面標記SSEA-4,其為通常用 作未分化萬能人類胚胎幹細胞之標記的早期胚胎醣酯抗 原"本發明人觀測到此等成人周邊血源性VSEL略微大於 其在成年鼠類骨髓中鑑別之對應物,但仍然極小。此外, 此等人類周邊血源性VSEL具有含有胚胎型無定形真染色 質(euchromatin)之大核。 本發明人明確證明,在G-CSF調動之前,在周邊血中僅 極少VSEL可偵測,而在G-CSF誘導之調動之後,析離產物 中佔TNC小於0.01%之VSEL極顯著地增加,超過106個。本 發明人證明儘管VSEL為相對稀少之細胞,但其可調動至 周邊血中且G-CSF誘導之調動適用於獲得人類萬能幹細胞 以用於再生醫學。 參考文獻 . 本文及說明書整篇引用之所有參考文獻、專利及專利申 請案皆以全文引用的方式併入本文中。 【圖式簡單說明】 152716.doc -100- 201130977 圖1 A-1D顯示經調動之周邊血源性人類MSC活體外黏附 及擴增。圖1A及1B展示培養於含有1〇°/。FBS(胎牛血清)之 DMEM中且以1,〇〇〇,〇〇〇個細胞/板(圖1A)及2,000,000個細 胞/板(圖1B)接種的經調動之周邊血源性人類MSC。圖1C 及1D展示培養於MesenCult-MSC(**商標?)中且以1,000,000 個細胞/板(圖1C)及2,000,000個細胞/板(圖1D)接種之經調 動之周邊血源性人類MSC。 圖2A-2D顯示周邊血源性人類MSC為CD45-及CD105+。 圖2A顯示基於CD45之標記表現之析離產物的FAC。圖2B 顯示基於CD45之標記表現之黏附細胞的FAC。黏附細胞富 含不表現CD45之細胞。圖2C顯示基於CD 105之標記表現 之析離產物的FAC。圖2D顯示基於CD105之標記表現之黏 附細胞的FAC »黏附細胞富含對CD105表現呈陽性之細 胞。 圖3八-30顯示周邊血源性人類1\^0:為€090+及00105+。 圖3A顯示基於CD90之標記表現之漂浮細胞的FAC »大多 數漂浮細胞對CD90表現呈陰性。圖3B顯示基於CD45之標 記表現之1次傳代(P1)後黏附細胞的FAC。黏附細胞(1次傳 代)富含表現CD90之細胞。圖3C顯示基於CD105之標記表 現之漂浮細胞的FAC。大多數漂浮細胞對CD105表現呈陰 性。圖3D顯示基於CD105之標記表現之1次傳代(P1)後黏 附細胞的FAC。黏附細胞(1次傳代)富含表現CD 105之細 胞。 圖4A-4B顯示周邊血源性人類MSC為CD44+。圖4A顯示 152716.doc -101 - 201130977 基於CD44之標記表現之漂浮細胞的FAC。大多數漂浮細胞 對CD44表現呈陰性。圖4B顯示基於CD44之標記表現之1 次傳代(P1)後黏附細胞的FAC。黏附細胞(1次傳代)富含表 現CD44之細胞。 圖5為說明用於自經調動之周邊血中分離MSC群之步驟 的示意圖。 圖6顯示G-CSF調動前及調動後周邊血中MSC之數目。 152716.doc -102-CarAo/., 2009:53;1-9.) and stroke (see Siroh. 2009:40;1237_). Similar cells in rodent organs have also been reported, and more importantly, cells that have been described to differentiate into all three germ layers in vitro have been described (Zewkwz'a 2006: 20; 857-869). In order to investigate the possibility that human VSELs can be a source of pluripotent stem cells in regenerative medicine, the inventors have developed an efficient strategy for isolating such cells from adult patients. The inventors have demonstrated that VSELs are similar to their murine counterparts and can be injected. The granulocyte community stimulating factor (G-CSF) is then mobilized into the peripheral blood (汾em Ce//i 2008:26; 2083-2092). The present inventors demonstrated this phenomenon by recruiting a group of young healthy donors who were mobilized using G-CSF (480 pg per day, subcutaneous) for 2 consecutive days. On day 3, nucleated cells (TNC) were collected by bloodpheresis. The inventors evaluated the number of VSELs in the peripheral blood (PB) samples before and after G-CSF mobilization and the final number in the fractionated product. At least 1 million TNCs were obtained and analyzed by FACS Diva software. Three different fractions of non-hematopoietic stem cells rich in VSEL (Lin7CD457CD133+, Lin7CD457CD34+, Lin*/CD457 152716.doc ·99·201130977 CXCR4+) and their CD45-positive hematopoietic counterparts were analyzed. The absolute number of cells contained in the 1 pL sample was calculated based on the percentage content of each population of each sample and the TNC count. The inventors determined that after G-CSF mobilization, human peripheral blood contains a population of 0-114, €034, and €0133 antigens such as - 045-monocytes. These lin_CD45XXCR4+CD133+CD34+ cells are highly enriched in mRNA of nuclear intraprimary embryonic transcription factors such as Oct-4, Sox2 and Nanog. More importantly, the inventors have found that 〇ct-4 is expressed in the nucleus of the mobilized VSEL and that these cells also exhibit the cell surface marker SSEA-4, which is commonly used as a marker for undifferentiated universal human embryonic stem cells. Early embryonic glycoester antigens" The inventors observed that the blood-borne VSELs of such adults were slightly larger than their counterparts identified in the bone marrow of adult rats, but were still extremely small. In addition, such human peripheral blood-derived VSELs have large nuclei containing embryonic amorphous euchromatin. The inventors have clearly demonstrated that only very few VSELs are detectable in peripheral blood prior to G-CSF mobilization, whereas after G-CSF-induced mobilization, VSELs in the separation products that account for less than 0.01% of TNC increase significantly. More than 106. The inventors have demonstrated that although VSELs are relatively rare cells, they are mobilized into peripheral blood and G-CSF-induced mobilization is suitable for obtaining human pluripotent stem cells for use in regenerative medicine. REFERENCES. All references, patents, and patent applications, which are hereby incorporated by reference in its entirety herein in its entirety herein in its entirety [Simple illustration] 152716.doc -100- 201130977 Figure 1 A-1D shows in vitro adhesion and expansion of mobilized peripheral blood-derived human MSCs. Figures 1A and 1B show cultures containing 1 〇 ° /. The mobilized peripheral blood-derived human MSCs in FMEM (fetal calf serum) in DMEM and inoculated with 1, 〇〇〇, 细胞 cells/plates (Fig. 1A) and 2,000,000 cells/plate (Fig. 1B). Figures 1C and 1D show mobilized peripheral blood-derived humans incubated in MesenCult-MSC (**Trademark?) and inoculated with 1,000,000 cells/plate (Figure 1C) and 2,000,000 cells/plate (Figure 1D). MSC. Figures 2A-2D show peripheral blood-derived human MSCs as CD45- and CD105+. Figure 2A shows the FAC of the isolated product expressed based on the label of CD45. Figure 2B shows the FAC of adherent cells expressed on CD45-based markers. Adhesive cells are rich in cells that do not express CD45. Figure 2C shows the FAC of the isolated product expressed based on the label of CD 105. Figure 2D shows that FACs based on adherent cells expressed by CD105 markers are adherent to cells that are positive for CD105. Figure 3-8 shows peripheral blood-borne humans 1\^0: €090+ and 00105+. Figure 3A shows FAC of floating cells expressed on CD90-based markers. Most floating cells are negative for CD90 expression. Fig. 3B shows the FAC of adherent cells after one passage (P1) based on the expression of CD45. Adherent cells (1 passage) are rich in cells expressing CD90. Figure 3C shows the FAC of floating cells based on the label of CD105. Most floating cells show negative for CD105. Figure 3D shows the FAC of adherent cells after one passage (P1) based on the label of CD105. Adherent cells (1 passage) are rich in cells expressing CD 105. Figures 4A-4B show peripheral blood-derived human MSCs as CD44+. Figure 4A shows the FAC of floating cells expressed by the CD44-based marker 152716.doc -101 - 201130977. Most floating cells were negative for CD44. Figure 4B shows FAC of adherent cells after 1 passage (P1) based on the CD44 marker. Adherent cells (1 passage) are rich in cells expressing CD44. Figure 5 is a schematic diagram showing the steps for separating MSC populations from the peripheral blood of the mobilization. Figure 6 shows the number of MSCs in peripheral blood before and after G-CSF mobilization. 152716.doc -102-
Claims (1)
Priority Applications (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| TW099142522A TW201130977A (en) | 2009-12-04 | 2010-12-06 | Mesenchymal stem cells (MSCs) isolated from mobilized peripheral blood |
Applications Claiming Priority (4)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US26682509P | 2009-12-04 | 2009-12-04 | |
| TW99104520 | 2010-02-10 | ||
| PCT/US2010/058980 WO2011069121A1 (en) | 2009-12-04 | 2010-12-03 | Mesenchymal stem cells (mscs) isolated from mobilized peripheral blood |
| TW099142522A TW201130977A (en) | 2009-12-04 | 2010-12-06 | Mesenchymal stem cells (MSCs) isolated from mobilized peripheral blood |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| TW201130977A true TW201130977A (en) | 2011-09-16 |
Family
ID=44115330
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| TW099142522A TW201130977A (en) | 2009-12-04 | 2010-12-06 | Mesenchymal stem cells (MSCs) isolated from mobilized peripheral blood |
Country Status (2)
| Country | Link |
|---|---|
| TW (1) | TW201130977A (en) |
| WO (1) | WO2011069121A1 (en) |
Cited By (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN110402286A (en) * | 2017-11-28 | 2019-11-01 | 熊仲川 | Method for preparing mesenchymal stem cell population from peripheral blood and use thereof |
Families Citing this family (15)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN102864123B (en) * | 2011-07-06 | 2015-04-22 | 北京大学第三医院 | Acquisition method of peripheral blood mesenchymal stem cells and application thereof |
| WO2013067038A1 (en) * | 2011-11-01 | 2013-05-10 | Neostem, Inc. | Adult mesenchymal stem cell (msc) compositions and methods for preparing the same |
| US20130244948A1 (en) * | 2012-03-15 | 2013-09-19 | Scharp Technologies, Inc. | Compositions of cells, media, and methods thereof |
| WO2013181642A1 (en) * | 2012-05-31 | 2013-12-05 | Neostem, Inc. | Human very small embryonic-like (vsel) stem cells for treatment of ocular disease |
| BE1020480A5 (en) * | 2012-10-01 | 2013-11-05 | Global Stem Cell Technology | METHOD FOR INSULATION OF MESENCHYMAL STEM CELLS FROM BIRD BLOOD AND USE THEREOF |
| EP3243519B1 (en) | 2013-05-09 | 2023-08-09 | Advanced Neuroregenerative Therapies LLC | Composition to improve the levels of anti-ageing biomarkers in a recipient |
| US20180161366A1 (en) * | 2015-05-20 | 2018-06-14 | Biokine Therapeutics Ltd. | Methods of obtaining mononuclear blood cells and uses thereof |
| CA2986705A1 (en) | 2015-07-16 | 2017-01-19 | Biokine Therapeutics Ltd. | A cxcr4 inhibitor and a pdi antagonist for use in treating cancer |
| EP3419645B1 (en) | 2016-02-23 | 2020-09-02 | BioLineRx Ltd. | Method of selecting a treatment regimen in acute myeloid leukemia (aml) |
| CA3019659A1 (en) * | 2016-04-06 | 2017-10-12 | University Of Florida Research Foundation, Inc. | Therapeutic use of electroacupuncture-induced mesenchymal stem cells |
| WO2017176267A1 (en) * | 2016-04-07 | 2017-10-12 | Dragonfly Foundation For Research & Development Corp | Method and system for repairing damaged tissue using nucleated plasma particles (nuc-p2s) and mesodermal stem cells (mesoscs) |
| WO2018187469A1 (en) * | 2017-04-05 | 2018-10-11 | Senlin Li | Methods and compositions for non-cytotoxic stem cell transplantation |
| TR201911506A2 (en) * | 2019-07-30 | 2021-02-22 | T C Erciyes Ueniversitesi | FIBRIN-WELDED MESENCHIMAL STEM CELLS RICH THROMBOSITE |
| CN111671896B (en) * | 2020-07-31 | 2020-12-15 | 深圳迈吉赛尔生物科技有限公司 | Application of bone marrow mesenchymal stem cells and monoclonal antibody in combined treatment of cancer |
| IT202000030692A1 (en) * | 2020-12-14 | 2022-06-14 | Centro Di Riferimento Oncologico | METHOD FOR PRODUCING MODIFIED MESENCHIMAL STEM STEM CELLS WITH IMPROVED PROPERTIES, MODIFIED CELLS OBTAINED BY THIS METHOD, COMPOSITION INCLUDING SUCH CELLS AND THEIR USE. |
Family Cites Families (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US7015037B1 (en) * | 1999-08-05 | 2006-03-21 | Regents Of The University Of Minnesota | Multiponent adult stem cells and methods for isolation |
| AUPR703601A0 (en) * | 2001-08-15 | 2001-09-06 | Peter Maccallum Cancer Institute, The | Identification and isolation of somatic stem cells and uses thereof |
| US20090004661A1 (en) * | 2003-05-26 | 2009-01-01 | Reliance Life Sciences Pvt Ltd. | Method of growing mesenchymal stem cells from bone marrow |
| EP2489728A1 (en) * | 2006-06-15 | 2012-08-22 | Neostem, Inc | Processing procedure for peripheral blood stem cells |
-
2010
- 2010-12-03 WO PCT/US2010/058980 patent/WO2011069121A1/en not_active Ceased
- 2010-12-06 TW TW099142522A patent/TW201130977A/en unknown
Cited By (2)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN110402286A (en) * | 2017-11-28 | 2019-11-01 | 熊仲川 | Method for preparing mesenchymal stem cell population from peripheral blood and use thereof |
| CN110402286B (en) * | 2017-11-28 | 2023-05-26 | 熊仲川 | Method for preparing mesenchymal stem cell population from peripheral blood and use thereof |
Also Published As
| Publication number | Publication date |
|---|---|
| WO2011069121A1 (en) | 2011-06-09 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| TW201130977A (en) | Mesenchymal stem cells (MSCs) isolated from mobilized peripheral blood | |
| US8986744B2 (en) | Stem cell populations and methods of use | |
| JP6285901B2 (en) | Use of mesenchymal stem cells | |
| Kim et al. | Cultured human bone marrow–derived CD31+ cells are effective for cardiac and vascular repair through enhanced angiogenic, adhesion, and anti-inflammatory effects | |
| WO2005063967A1 (en) | Induction of myocardial cell with the use of mammalian bone marrow cell or cord blood-origin cell and fat tissue | |
| JP2008504290A (en) | Cell-based treatment of ischemia | |
| TW200804599A (en) | Isolation and purification of hematopoietic stem cells from post-liposuction lipoaspirates | |
| EP2214497A2 (en) | Uses and isolation of very small embryonic-like (vsel) stem cells | |
| CA2705450A1 (en) | Stem cells for treating lung diseases | |
| KR102213527B1 (en) | Mesenchymal stromal cells for treating sepsis | |
| TW201130978A (en) | Method of isolation of stem cell populations from peripheral blood using sized-based separation (elutriation) | |
| WO2007024441A2 (en) | Compositions of cells enriched for combinations of various stem and progenitor cell populations, methods of use thereof and methods of private banking thereof | |
| KR20080042761A (en) | Stem Cell Proliferation Using Leptin | |
| EP1771551B1 (en) | Novel cell populations and uses thereof | |
| US20160158292A1 (en) | Method and apparatus for recovery of umbilical cord tissue derived regenerative cells and uses thereof | |
| CN113853206A (en) | Functional recovery after cerebral infarction | |
| CN114901806B (en) | Cell population and method for obtaining the same | |
| EP2205251B1 (en) | A method to amplify cardiac stem cells in vitro and in vivo | |
| CN104995295A (en) | A method of generating multilineage potential cells | |
| CN110882276B (en) | Cell therapy compositions and methods for treating vascular disorders | |
| CN110402286B (en) | Method for preparing mesenchymal stem cell population from peripheral blood and use thereof | |
| CN101506353A (en) | Processing procedure for peripheral blood stem cells | |
| US11674131B2 (en) | Indirect ultrasonic cavitation-derived perivascular cells and methods of use thereof | |
| RU2454247C1 (en) | Method for prevention of acute graft-versus-host disease following marrow allografting | |
| RU2800644C2 (en) | Method of producing fibroblasts and g-csf-positive fibroblast mass |