WO2010008561A2 - Methods and compositions for protecting against cytotoxic therapy - Google Patents
Methods and compositions for protecting against cytotoxic therapy Download PDFInfo
- Publication number
- WO2010008561A2 WO2010008561A2 PCT/US2009/004110 US2009004110W WO2010008561A2 WO 2010008561 A2 WO2010008561 A2 WO 2010008561A2 US 2009004110 W US2009004110 W US 2009004110W WO 2010008561 A2 WO2010008561 A2 WO 2010008561A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- pthrp
- cancer
- derivative
- fragment
- analog
- Prior art date
Links
- 238000000034 method Methods 0.000 title claims abstract description 140
- 239000000203 mixture Substances 0.000 title claims abstract description 106
- 238000002560 therapeutic procedure Methods 0.000 title claims abstract description 101
- 230000002633 protecting effect Effects 0.000 title claims description 6
- 230000001472 cytotoxic effect Effects 0.000 title description 29
- 231100000433 cytotoxic Toxicity 0.000 title description 27
- 108700020797 Parathyroid Hormone-Related Proteins 0.000 claims abstract description 456
- 102000043299 Parathyroid hormone-related Human genes 0.000 claims abstract description 456
- 238000002512 chemotherapy Methods 0.000 claims abstract description 92
- 238000001959 radiotherapy Methods 0.000 claims abstract description 71
- 239000003814 drug Substances 0.000 claims abstract description 57
- 210000003958 hematopoietic stem cell Anatomy 0.000 claims abstract description 31
- 206010065553 Bone marrow failure Diseases 0.000 claims abstract description 22
- 230000002441 reversible effect Effects 0.000 claims abstract description 22
- 230000005855 radiation Effects 0.000 claims abstract description 18
- 210000004027 cell Anatomy 0.000 claims description 89
- 239000012634 fragment Substances 0.000 claims description 87
- 229940127089 cytotoxic agent Drugs 0.000 claims description 56
- 230000003394 haemopoietic effect Effects 0.000 claims description 56
- 239000002254 cytotoxic agent Substances 0.000 claims description 51
- 231100000599 cytotoxic agent Toxicity 0.000 claims description 51
- 206010028980 Neoplasm Diseases 0.000 claims description 49
- -1 PDGF Proteins 0.000 claims description 48
- 102000004127 Cytokines Human genes 0.000 claims description 47
- 108090000695 Cytokines Proteins 0.000 claims description 47
- 201000011510 cancer Diseases 0.000 claims description 41
- 230000001988 toxicity Effects 0.000 claims description 38
- 231100000419 toxicity Toxicity 0.000 claims description 38
- 210000000130 stem cell Anatomy 0.000 claims description 32
- 108010017080 Granulocyte Colony-Stimulating Factor Proteins 0.000 claims description 31
- 108010002350 Interleukin-2 Proteins 0.000 claims description 29
- 102000000588 Interleukin-2 Human genes 0.000 claims description 29
- 230000001225 therapeutic effect Effects 0.000 claims description 27
- 108010046938 Macrophage Colony-Stimulating Factor Proteins 0.000 claims description 24
- 102000007651 Macrophage Colony-Stimulating Factor Human genes 0.000 claims description 24
- 108010002386 Interleukin-3 Proteins 0.000 claims description 22
- 102100039064 Interleukin-3 Human genes 0.000 claims description 22
- 102100039620 Granulocyte-macrophage colony-stimulating factor Human genes 0.000 claims description 18
- 230000007423 decrease Effects 0.000 claims description 18
- 230000009467 reduction Effects 0.000 claims description 18
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 claims description 17
- 206010033661 Pancytopenia Diseases 0.000 claims description 16
- 210000001744 T-lymphocyte Anatomy 0.000 claims description 16
- 208000024389 cytopenia Diseases 0.000 claims description 16
- 210000000056 organ Anatomy 0.000 claims description 16
- 108090000394 Erythropoietin Proteins 0.000 claims description 15
- 102000003951 Erythropoietin Human genes 0.000 claims description 15
- 229940105423 erythropoietin Drugs 0.000 claims description 15
- OXCMYAYHXIHQOA-UHFFFAOYSA-N potassium;[2-butyl-5-chloro-3-[[4-[2-(1,2,4-triaza-3-azanidacyclopenta-1,4-dien-5-yl)phenyl]phenyl]methyl]imidazol-4-yl]methanol Chemical compound [K+].CCCCC1=NC(Cl)=C(CO)N1CC1=CC=C(C=2C(=CC=CC=2)C2=N[N-]N=N2)C=C1 OXCMYAYHXIHQOA-UHFFFAOYSA-N 0.000 claims description 15
- 229940124597 therapeutic agent Drugs 0.000 claims description 15
- 230000004044 response Effects 0.000 claims description 14
- 102000003972 Fibroblast growth factor 7 Human genes 0.000 claims description 13
- 108090000385 Fibroblast growth factor 7 Proteins 0.000 claims description 13
- GHASVSINZRGABV-UHFFFAOYSA-N Fluorouracil Chemical compound FC1=CNC(=O)NC1=O GHASVSINZRGABV-UHFFFAOYSA-N 0.000 claims description 13
- 108090000978 Interleukin-4 Proteins 0.000 claims description 13
- 102000004388 Interleukin-4 Human genes 0.000 claims description 13
- 108090001005 Interleukin-6 Proteins 0.000 claims description 13
- 102100020880 Kit ligand Human genes 0.000 claims description 13
- 101100335081 Mus musculus Flt3 gene Proteins 0.000 claims description 13
- 108010039445 Stem Cell Factor Proteins 0.000 claims description 13
- 102100031294 Thymic stromal lymphopoietin Human genes 0.000 claims description 13
- 108010029307 thymic stromal lymphopoietin Proteins 0.000 claims description 13
- 108090000176 Interleukin-13 Proteins 0.000 claims description 12
- 108010002616 Interleukin-5 Proteins 0.000 claims description 12
- 102000004889 Interleukin-6 Human genes 0.000 claims description 12
- 108010002586 Interleukin-7 Proteins 0.000 claims description 12
- 108090001007 Interleukin-8 Proteins 0.000 claims description 12
- 108010002335 Interleukin-9 Proteins 0.000 claims description 12
- 108010041111 Thrombopoietin Proteins 0.000 claims description 12
- 102000036693 Thrombopoietin Human genes 0.000 claims description 12
- 229960002949 fluorouracil Drugs 0.000 claims description 12
- BLQSIKSVAXFNRH-ZMWZJJJDSA-N semparatide Chemical compound C([C@@H](C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCCCN)C(=O)NCC(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CO)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC=1N=CNC=1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](C)C(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC=1N=CNC=1)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@H](C)N)C(C)C)C1=CNC=N1 BLQSIKSVAXFNRH-ZMWZJJJDSA-N 0.000 claims description 12
- 229950009144 semparatide Drugs 0.000 claims description 12
- 206010009944 Colon cancer Diseases 0.000 claims description 11
- 230000037396 body weight Effects 0.000 claims description 11
- 230000000977 initiatory effect Effects 0.000 claims description 11
- 102000003812 Interleukin-15 Human genes 0.000 claims description 10
- 210000004369 blood Anatomy 0.000 claims description 10
- 239000008280 blood Substances 0.000 claims description 10
- 210000000601 blood cell Anatomy 0.000 claims description 10
- 208000004235 neutropenia Diseases 0.000 claims description 10
- 239000002246 antineoplastic agent Substances 0.000 claims description 9
- 230000003247 decreasing effect Effects 0.000 claims description 9
- 210000000777 hematopoietic system Anatomy 0.000 claims description 9
- 210000000963 osteoblast Anatomy 0.000 claims description 9
- 102000049772 Interleukin-16 Human genes 0.000 claims description 8
- 102000013691 Interleukin-17 Human genes 0.000 claims description 8
- 108050003558 Interleukin-17 Proteins 0.000 claims description 8
- 230000000973 chemotherapeutic effect Effects 0.000 claims description 8
- 238000001802 infusion Methods 0.000 claims description 8
- 230000003039 myelosuppressive effect Effects 0.000 claims description 8
- 206010006187 Breast cancer Diseases 0.000 claims description 7
- 208000026310 Breast neoplasm Diseases 0.000 claims description 7
- 108090000177 Interleukin-11 Proteins 0.000 claims description 7
- 102000003815 Interleukin-11 Human genes 0.000 claims description 7
- 108010065805 Interleukin-12 Proteins 0.000 claims description 7
- 230000000116 mitigating effect Effects 0.000 claims description 7
- 239000000126 substance Substances 0.000 claims description 7
- 210000001519 tissue Anatomy 0.000 claims description 7
- 206010006002 Bone pain Diseases 0.000 claims description 6
- 206010033128 Ovarian cancer Diseases 0.000 claims description 6
- 206010061535 Ovarian neoplasm Diseases 0.000 claims description 6
- 208000007502 anemia Diseases 0.000 claims description 6
- 210000000988 bone and bone Anatomy 0.000 claims description 6
- 208000029742 colonic neoplasm Diseases 0.000 claims description 6
- 210000003714 granulocyte Anatomy 0.000 claims description 6
- 208000032839 leukemia Diseases 0.000 claims description 6
- 238000011275 oncology therapy Methods 0.000 claims description 6
- 238000002054 transplantation Methods 0.000 claims description 6
- 206010005003 Bladder cancer Diseases 0.000 claims description 5
- 208000001333 Colorectal Neoplasms Diseases 0.000 claims description 5
- 206010014733 Endometrial cancer Diseases 0.000 claims description 5
- 206010014759 Endometrial neoplasm Diseases 0.000 claims description 5
- 208000000461 Esophageal Neoplasms Diseases 0.000 claims description 5
- 208000017604 Hodgkin disease Diseases 0.000 claims description 5
- 208000021519 Hodgkin lymphoma Diseases 0.000 claims description 5
- 208000010747 Hodgkins lymphoma Diseases 0.000 claims description 5
- 208000007766 Kaposi sarcoma Diseases 0.000 claims description 5
- 208000008839 Kidney Neoplasms Diseases 0.000 claims description 5
- 206010058467 Lung neoplasm malignant Diseases 0.000 claims description 5
- 206010025323 Lymphomas Diseases 0.000 claims description 5
- 208000015914 Non-Hodgkin lymphomas Diseases 0.000 claims description 5
- 206010030155 Oesophageal carcinoma Diseases 0.000 claims description 5
- 206010061902 Pancreatic neoplasm Diseases 0.000 claims description 5
- 206010060862 Prostate cancer Diseases 0.000 claims description 5
- 208000000236 Prostatic Neoplasms Diseases 0.000 claims description 5
- 206010038389 Renal cancer Diseases 0.000 claims description 5
- 208000005718 Stomach Neoplasms Diseases 0.000 claims description 5
- 208000007097 Urinary Bladder Neoplasms Diseases 0.000 claims description 5
- 230000030833 cell death Effects 0.000 claims description 5
- 238000011393 cytotoxic chemotherapy Methods 0.000 claims description 5
- 201000004101 esophageal cancer Diseases 0.000 claims description 5
- 206010017758 gastric cancer Diseases 0.000 claims description 5
- 201000010536 head and neck cancer Diseases 0.000 claims description 5
- 208000014829 head and neck neoplasm Diseases 0.000 claims description 5
- 201000010982 kidney cancer Diseases 0.000 claims description 5
- 201000005202 lung cancer Diseases 0.000 claims description 5
- 208000020816 lung neoplasm Diseases 0.000 claims description 5
- 208000015486 malignant pancreatic neoplasm Diseases 0.000 claims description 5
- 201000001441 melanoma Diseases 0.000 claims description 5
- 201000002528 pancreatic cancer Diseases 0.000 claims description 5
- 208000008443 pancreatic carcinoma Diseases 0.000 claims description 5
- 201000011549 stomach cancer Diseases 0.000 claims description 5
- 208000024891 symptom Diseases 0.000 claims description 5
- 206010043554 thrombocytopenia Diseases 0.000 claims description 5
- 201000005112 urinary bladder cancer Diseases 0.000 claims description 5
- 108090000174 Interleukin-10 Proteins 0.000 claims description 4
- 206010027476 Metastases Diseases 0.000 claims description 4
- XQRLCLUYWUNEEH-UHFFFAOYSA-L diphosphonate(2-) Chemical compound [O-]P(=O)OP([O-])=O XQRLCLUYWUNEEH-UHFFFAOYSA-L 0.000 claims description 4
- 210000002540 macrophage Anatomy 0.000 claims description 4
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 claims description 4
- 206010005949 Bone cancer Diseases 0.000 claims description 3
- 208000018084 Bone neoplasm Diseases 0.000 claims description 3
- GUTLYIVDDKVIGB-OUBTZVSYSA-N Cobalt-60 Chemical compound [60Co] GUTLYIVDDKVIGB-OUBTZVSYSA-N 0.000 claims description 3
- HCWPIIXVSYCSAN-IGMARMGPSA-N Radium-226 Chemical compound [226Ra] HCWPIIXVSYCSAN-IGMARMGPSA-N 0.000 claims description 3
- TVFDJXOCXUVLDH-RNFDNDRNSA-N cesium-137 Chemical compound [137Cs] TVFDJXOCXUVLDH-RNFDNDRNSA-N 0.000 claims description 3
- GKOZUEZYRPOHIO-IGMARMGPSA-N iridium-192 Chemical compound [192Ir] GKOZUEZYRPOHIO-IGMARMGPSA-N 0.000 claims description 3
- 230000009401 metastasis Effects 0.000 claims description 3
- 210000003719 b-lymphocyte Anatomy 0.000 claims description 2
- 210000002997 osteoclast Anatomy 0.000 claims description 2
- 102000004269 Granulocyte Colony-Stimulating Factor Human genes 0.000 claims 4
- 102000004457 Granulocyte-Macrophage Colony-Stimulating Factor Human genes 0.000 claims 1
- 230000000699 topical effect Effects 0.000 claims 1
- 229940079593 drug Drugs 0.000 abstract description 39
- 230000003013 cytotoxicity Effects 0.000 abstract description 6
- 231100000135 cytotoxicity Toxicity 0.000 abstract description 6
- 230000004224 protection Effects 0.000 abstract description 6
- 238000011282 treatment Methods 0.000 description 55
- 108090000623 proteins and genes Proteins 0.000 description 53
- 108090000765 processed proteins & peptides Proteins 0.000 description 51
- 102000004169 proteins and genes Human genes 0.000 description 46
- 235000018102 proteins Nutrition 0.000 description 42
- 239000003795 chemical substances by application Substances 0.000 description 37
- 230000000694 effects Effects 0.000 description 37
- 210000000265 leukocyte Anatomy 0.000 description 37
- 102000004196 processed proteins & peptides Human genes 0.000 description 34
- 102100039619 Granulocyte colony-stimulating factor Human genes 0.000 description 28
- 238000004820 blood count Methods 0.000 description 28
- 230000014509 gene expression Effects 0.000 description 26
- 229920001184 polypeptide Polymers 0.000 description 26
- 210000001772 blood platelet Anatomy 0.000 description 25
- 210000003743 erythrocyte Anatomy 0.000 description 21
- 210000001185 bone marrow Anatomy 0.000 description 18
- 239000003102 growth factor Substances 0.000 description 15
- 229920001223 polyethylene glycol Polymers 0.000 description 15
- 239000002202 Polyethylene glycol Substances 0.000 description 14
- 239000000306 component Substances 0.000 description 14
- 210000004976 peripheral blood cell Anatomy 0.000 description 13
- 108020004414 DNA Proteins 0.000 description 12
- 150000001413 amino acids Chemical class 0.000 description 12
- 239000013604 expression vector Substances 0.000 description 12
- 208000015181 infectious disease Diseases 0.000 description 12
- 238000001990 intravenous administration Methods 0.000 description 12
- 239000003053 toxin Substances 0.000 description 12
- 231100000765 toxin Toxicity 0.000 description 12
- 108700012359 toxins Proteins 0.000 description 12
- 239000013598 vector Substances 0.000 description 12
- 241001465754 Metazoa Species 0.000 description 11
- 150000007523 nucleic acids Chemical class 0.000 description 11
- 238000002360 preparation method Methods 0.000 description 11
- 239000000243 solution Substances 0.000 description 11
- 108020004707 nucleic acids Proteins 0.000 description 10
- 102000039446 nucleic acids Human genes 0.000 description 10
- 239000008194 pharmaceutical composition Substances 0.000 description 10
- 229920000642 polymer Polymers 0.000 description 10
- 229940024606 amino acid Drugs 0.000 description 9
- 235000001014 amino acid Nutrition 0.000 description 9
- 150000001875 compounds Chemical class 0.000 description 9
- 239000003937 drug carrier Substances 0.000 description 9
- 229940127121 immunoconjugate Drugs 0.000 description 9
- 210000004698 lymphocyte Anatomy 0.000 description 9
- 210000000440 neutrophil Anatomy 0.000 description 9
- 108090000445 Parathyroid hormone Proteins 0.000 description 8
- VWQVUPCCIRVNHF-OUBTZVSYSA-N Yttrium-90 Chemical compound [90Y] VWQVUPCCIRVNHF-OUBTZVSYSA-N 0.000 description 8
- 238000009472 formulation Methods 0.000 description 8
- 238000001361 intraarterial administration Methods 0.000 description 8
- 238000007912 intraperitoneal administration Methods 0.000 description 8
- 230000002829 reductive effect Effects 0.000 description 8
- 108091028043 Nucleic acid sequence Proteins 0.000 description 7
- 102100036893 Parathyroid hormone Human genes 0.000 description 7
- 239000000562 conjugate Substances 0.000 description 7
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 7
- 238000007913 intrathecal administration Methods 0.000 description 7
- 238000011084 recovery Methods 0.000 description 7
- 150000003839 salts Chemical group 0.000 description 7
- 238000011285 therapeutic regimen Methods 0.000 description 7
- AOJJSUZBOXZQNB-TZSSRYMLSA-N Doxorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 AOJJSUZBOXZQNB-TZSSRYMLSA-N 0.000 description 6
- 102000004190 Enzymes Human genes 0.000 description 6
- 108090000790 Enzymes Proteins 0.000 description 6
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 6
- 208000001388 Opportunistic Infections Diseases 0.000 description 6
- 239000004480 active ingredient Substances 0.000 description 6
- 201000010099 disease Diseases 0.000 description 6
- 230000001965 increasing effect Effects 0.000 description 6
- 230000004048 modification Effects 0.000 description 6
- 238000012986 modification Methods 0.000 description 6
- 239000002773 nucleotide Substances 0.000 description 6
- 125000003729 nucleotide group Chemical group 0.000 description 6
- 238000000746 purification Methods 0.000 description 6
- 238000002741 site-directed mutagenesis Methods 0.000 description 6
- 238000007920 subcutaneous administration Methods 0.000 description 6
- 239000007929 subcutaneous injection Substances 0.000 description 6
- 238000010254 subcutaneous injection Methods 0.000 description 6
- 241000699670 Mus sp. Species 0.000 description 5
- 230000004913 activation Effects 0.000 description 5
- 239000013543 active substance Substances 0.000 description 5
- 239000003242 anti bacterial agent Substances 0.000 description 5
- 230000001093 anti-cancer Effects 0.000 description 5
- 230000001413 cellular effect Effects 0.000 description 5
- 239000006185 dispersion Substances 0.000 description 5
- 230000006870 function Effects 0.000 description 5
- 230000000670 limiting effect Effects 0.000 description 5
- 244000005700 microbiome Species 0.000 description 5
- 238000012544 monitoring process Methods 0.000 description 5
- 210000000066 myeloid cell Anatomy 0.000 description 5
- 230000035755 proliferation Effects 0.000 description 5
- 230000008685 targeting Effects 0.000 description 5
- 238000012360 testing method Methods 0.000 description 5
- 210000004881 tumor cell Anatomy 0.000 description 5
- ZOXJGFHDIHLPTG-BJUDXGSMSA-N Boron-10 Chemical group [10B] ZOXJGFHDIHLPTG-BJUDXGSMSA-N 0.000 description 4
- 108091026890 Coding region Proteins 0.000 description 4
- NKANXQFJJICGDU-QPLCGJKRSA-N Tamoxifen Chemical compound C=1C=CC=CC=1C(/CC)=C(C=1C=CC(OCCN(C)C)=CC=1)/C1=CC=CC=C1 NKANXQFJJICGDU-QPLCGJKRSA-N 0.000 description 4
- 230000002411 adverse Effects 0.000 description 4
- 239000000443 aerosol Substances 0.000 description 4
- 238000003556 assay Methods 0.000 description 4
- 239000012503 blood component Substances 0.000 description 4
- 238000002648 combination therapy Methods 0.000 description 4
- 230000006378 damage Effects 0.000 description 4
- 239000002612 dispersion medium Substances 0.000 description 4
- 238000009826 distribution Methods 0.000 description 4
- 230000012010 growth Effects 0.000 description 4
- 238000002513 implantation Methods 0.000 description 4
- 238000002347 injection Methods 0.000 description 4
- 239000007924 injection Substances 0.000 description 4
- 238000007918 intramuscular administration Methods 0.000 description 4
- 230000002147 killing effect Effects 0.000 description 4
- 125000005647 linker group Chemical group 0.000 description 4
- 230000007774 longterm Effects 0.000 description 4
- 239000003550 marker Substances 0.000 description 4
- 239000013612 plasmid Substances 0.000 description 4
- 230000008488 polyadenylation Effects 0.000 description 4
- 238000009258 post-therapy Methods 0.000 description 4
- 230000000069 prophylactic effect Effects 0.000 description 4
- 238000011363 radioimmunotherapy Methods 0.000 description 4
- 230000003439 radiotherapeutic effect Effects 0.000 description 4
- 238000003259 recombinant expression Methods 0.000 description 4
- 230000001105 regulatory effect Effects 0.000 description 4
- 239000002904 solvent Substances 0.000 description 4
- 230000000638 stimulation Effects 0.000 description 4
- 230000004083 survival effect Effects 0.000 description 4
- 238000013518 transcription Methods 0.000 description 4
- 230000035897 transcription Effects 0.000 description 4
- 230000037317 transdermal delivery Effects 0.000 description 4
- 230000003612 virological effect Effects 0.000 description 4
- ZOWOHMFPXMYFKJ-WBTWNKCNSA-N (4S)-4-[[(2S)-2-[[(2S,3S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-5-amino-2-[[(2S,3S)-2-[[(2S)-2-[[(2S)-6-amino-2-[[2-[[(2S)-6-amino-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-5-amino-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-aminopropanoyl]amino]-3-methylbutanoyl]amino]-3-hydroxypropanoyl]amino]-4-carboxybutanoyl]amino]-3-(1H-imidazol-5-yl)propanoyl]amino]-5-oxopentanoyl]amino]-4-methylpentanoyl]amino]-4-methylpentanoyl]amino]-3-(1H-imidazol-5-yl)propanoyl]amino]-3-carboxypropanoyl]amino]hexanoyl]amino]acetyl]amino]hexanoyl]amino]-3-hydroxypropanoyl]amino]-3-methylpentanoyl]amino]-5-oxopentanoyl]amino]-3-carboxypropanoyl]amino]-4-methylpentanoyl]amino]-5-carbamimidamidopentanoyl]amino]-5-carbamimidamidopentanoyl]amino]-5-carbamimidamidopentanoyl]amino]-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]-3-(1H-imidazol-5-yl)propanoyl]amino]-3-(1H-imidazol-5-yl)propanoyl]amino]-4-methylpentanoyl]amino]-3-methylpentanoyl]amino]propanoyl]amino]-5-[[(2S,3S)-1-[[(2S)-1-[[(2S,3R)-1-[[(1S)-1-carboxyethyl]amino]-3-hydroxy-1-oxobutan-2-yl]amino]-3-(1H-imidazol-5-yl)-1-oxopropan-2-yl]amino]-3-methyl-1-oxopentan-2-yl]amino]-5-oxopentanoic acid Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CC=1NC=NC=1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](C)C(O)=O)NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=1C=CC=CC=1)NC(=O)[C@H](CC=1C=CC=CC=1)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](NC(=O)[C@H](CO)NC(=O)[C@H](CCCCN)NC(=O)CNC(=O)[C@H](CCCCN)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@H](C)N)C(C)C)[C@@H](C)CC)C1=CN=CN1 ZOWOHMFPXMYFKJ-WBTWNKCNSA-N 0.000 description 3
- QTBSBXVTEAMEQO-UHFFFAOYSA-M Acetate Chemical compound CC([O-])=O QTBSBXVTEAMEQO-UHFFFAOYSA-M 0.000 description 3
- 208000023275 Autoimmune disease Diseases 0.000 description 3
- 241000894006 Bacteria Species 0.000 description 3
- 206010051779 Bone marrow toxicity Diseases 0.000 description 3
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 3
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 3
- 102000001554 Hemoglobins Human genes 0.000 description 3
- 108010054147 Hemoglobins Proteins 0.000 description 3
- 102000008072 Lymphokines Human genes 0.000 description 3
- 108010074338 Lymphokines Proteins 0.000 description 3
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 3
- 239000004472 Lysine Substances 0.000 description 3
- 241000124008 Mammalia Species 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 3
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 3
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 3
- 206010040047 Sepsis Diseases 0.000 description 3
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 3
- 206010052779 Transplant rejections Diseases 0.000 description 3
- 238000010521 absorption reaction Methods 0.000 description 3
- 230000009471 action Effects 0.000 description 3
- 238000004458 analytical method Methods 0.000 description 3
- 230000000844 anti-bacterial effect Effects 0.000 description 3
- 229940121375 antifungal agent Drugs 0.000 description 3
- 239000003429 antifungal agent Substances 0.000 description 3
- 239000007864 aqueous solution Substances 0.000 description 3
- 230000009286 beneficial effect Effects 0.000 description 3
- 230000008901 benefit Effects 0.000 description 3
- 230000015572 biosynthetic process Effects 0.000 description 3
- 231100000366 bone marrow toxicity Toxicity 0.000 description 3
- 239000000872 buffer Substances 0.000 description 3
- 230000024245 cell differentiation Effects 0.000 description 3
- 230000004087 circulation Effects 0.000 description 3
- 238000000576 coating method Methods 0.000 description 3
- 125000004122 cyclic group Chemical group 0.000 description 3
- 229960004397 cyclophosphamide Drugs 0.000 description 3
- 230000002435 cytoreductive effect Effects 0.000 description 3
- 230000002939 deleterious effect Effects 0.000 description 3
- 238000012217 deletion Methods 0.000 description 3
- 230000037430 deletion Effects 0.000 description 3
- 238000012377 drug delivery Methods 0.000 description 3
- 239000003623 enhancer Substances 0.000 description 3
- 230000002489 hematologic effect Effects 0.000 description 3
- 230000001981 hematoprotective effect Effects 0.000 description 3
- 230000028993 immune response Effects 0.000 description 3
- 238000001727 in vivo Methods 0.000 description 3
- 230000002458 infectious effect Effects 0.000 description 3
- 239000004615 ingredient Substances 0.000 description 3
- 238000003780 insertion Methods 0.000 description 3
- 230000037431 insertion Effects 0.000 description 3
- 239000007951 isotonicity adjuster Substances 0.000 description 3
- 239000007788 liquid Substances 0.000 description 3
- 238000004519 manufacturing process Methods 0.000 description 3
- 206010061289 metastatic neoplasm Diseases 0.000 description 3
- 108010054971 parathyroid hormone-related protein (1-34) Proteins 0.000 description 3
- 238000010647 peptide synthesis reaction Methods 0.000 description 3
- 230000002093 peripheral effect Effects 0.000 description 3
- 239000012071 phase Substances 0.000 description 3
- 230000001766 physiological effect Effects 0.000 description 3
- 239000000843 powder Substances 0.000 description 3
- 238000011321 prophylaxis Methods 0.000 description 3
- 238000011362 radionuclide therapy Methods 0.000 description 3
- 230000004223 radioprotective effect Effects 0.000 description 3
- 230000001172 regenerating effect Effects 0.000 description 3
- 238000011160 research Methods 0.000 description 3
- 238000006467 substitution reaction Methods 0.000 description 3
- 238000003786 synthesis reaction Methods 0.000 description 3
- 230000002103 transcriptional effect Effects 0.000 description 3
- 230000014616 translation Effects 0.000 description 3
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 3
- 229920003169 water-soluble polymer Polymers 0.000 description 3
- PNDPGZBMCMUPRI-HVTJNCQCSA-N 10043-66-0 Chemical compound [131I][131I] PNDPGZBMCMUPRI-HVTJNCQCSA-N 0.000 description 2
- 241000272517 Anseriformes Species 0.000 description 2
- PVSNBTCXCQIXSE-JYJNAYRXSA-N Arg-Arg-Phe Chemical compound NC(=N)NCCC[C@H](N)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@H](C(O)=O)CC1=CC=CC=C1 PVSNBTCXCQIXSE-JYJNAYRXSA-N 0.000 description 2
- 206010003445 Ascites Diseases 0.000 description 2
- ZOXJGFHDIHLPTG-UHFFFAOYSA-N Boron Chemical group [B] ZOXJGFHDIHLPTG-UHFFFAOYSA-N 0.000 description 2
- 241000283690 Bos taurus Species 0.000 description 2
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 2
- 108010022366 Carcinoembryonic Antigen Proteins 0.000 description 2
- 102100025475 Carcinoembryonic antigen-related cell adhesion molecule 5 Human genes 0.000 description 2
- 108010035563 Chloramphenicol O-acetyltransferase Proteins 0.000 description 2
- 241000196324 Embryophyta Species 0.000 description 2
- 241000588724 Escherichia coli Species 0.000 description 2
- 108010070675 Glutathione transferase Proteins 0.000 description 2
- 102000005720 Glutathione transferase Human genes 0.000 description 2
- 108091027305 Heteroduplex Proteins 0.000 description 2
- BRQKGRLDDDQWQJ-MBLNEYKQSA-N His-Thr-Ala Chemical compound [H]N[C@@H](CC1=CNC=N1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](C)C(O)=O BRQKGRLDDDQWQJ-MBLNEYKQSA-N 0.000 description 2
- 241000282412 Homo Species 0.000 description 2
- KYIIALJHAOIAHF-KKUMJFAQSA-N Leu-Leu-His Chemical compound CC(C)C[C@H](N)C(=O)N[C@@H](CC(C)C)C(=O)N[C@H](C(O)=O)CC1=CN=CN1 KYIIALJHAOIAHF-KKUMJFAQSA-N 0.000 description 2
- YSZNURNVYFUEHC-BQBZGAKWSA-N Lys-Ser Chemical compound NCCCC[C@H](N)C(=O)N[C@@H](CO)C(O)=O YSZNURNVYFUEHC-BQBZGAKWSA-N 0.000 description 2
- 102000005650 Notch Receptors Human genes 0.000 description 2
- 108010070047 Notch Receptors Proteins 0.000 description 2
- 108091034117 Oligonucleotide Proteins 0.000 description 2
- 241000283973 Oryctolagus cuniculus Species 0.000 description 2
- 229930012538 Paclitaxel Natural products 0.000 description 2
- 108010058828 Parathyroid Hormone Receptors Proteins 0.000 description 2
- 102000006461 Parathyroid Hormone Receptors Human genes 0.000 description 2
- METZZBCMDXHFMK-BZSNNMDCSA-N Phe-Leu-His Chemical compound CC(C)C[C@@H](C(=O)N[C@@H](CC1=CN=CN1)C(=O)O)NC(=O)[C@H](CC2=CC=CC=C2)N METZZBCMDXHFMK-BZSNNMDCSA-N 0.000 description 2
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N Phenol Chemical compound OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 2
- 108010076504 Protein Sorting Signals Proteins 0.000 description 2
- 241000700159 Rattus Species 0.000 description 2
- 241000906446 Theraps Species 0.000 description 2
- 241000723873 Tobacco mosaic virus Species 0.000 description 2
- JXLYSJRDGCGARV-WWYNWVTFSA-N Vinblastine Natural products O=C(O[C@H]1[C@](O)(C(=O)OC)[C@@H]2N(C)c3c(cc(c(OC)c3)[C@]3(C(=O)OC)c4[nH]c5c(c4CCN4C[C@](O)(CC)C[C@H](C3)C4)cccc5)[C@@]32[C@H]2[C@@]1(CC)C=CCN2CC3)C JXLYSJRDGCGARV-WWYNWVTFSA-N 0.000 description 2
- 241000700605 Viruses Species 0.000 description 2
- 239000003070 absorption delaying agent Substances 0.000 description 2
- 239000002253 acid Substances 0.000 description 2
- RJURFGZVJUQBHK-UHFFFAOYSA-N actinomycin D Natural products CC1OC(=O)C(C(C)C)N(C)C(=O)CN(C)C(=O)C2CCCN2C(=O)C(C(C)C)NC(=O)C1NC(=O)C1=C(N)C(=O)C(C)=C2OC(C(C)=CC=C3C(=O)NC4C(=O)NC(C(N5CCCC5C(=O)N(C)CC(=O)N(C)C(C(C)C)C(=O)OC4C)=O)C(C)C)=C3N=C21 RJURFGZVJUQBHK-UHFFFAOYSA-N 0.000 description 2
- 229940009456 adriamycin Drugs 0.000 description 2
- 230000000172 allergic effect Effects 0.000 description 2
- 125000000539 amino acid group Chemical group 0.000 description 2
- 238000010171 animal model Methods 0.000 description 2
- 229940124650 anti-cancer therapies Drugs 0.000 description 2
- 230000000845 anti-microbial effect Effects 0.000 description 2
- 229940088710 antibiotic agent Drugs 0.000 description 2
- 238000011319 anticancer therapy Methods 0.000 description 2
- 230000006907 apoptotic process Effects 0.000 description 2
- 238000013459 approach Methods 0.000 description 2
- 108010092854 aspartyllysine Proteins 0.000 description 2
- 208000010668 atopic eczema Diseases 0.000 description 2
- 230000001580 bacterial effect Effects 0.000 description 2
- 210000003651 basophil Anatomy 0.000 description 2
- 230000004071 biological effect Effects 0.000 description 2
- 230000008512 biological response Effects 0.000 description 2
- 238000010241 blood sampling Methods 0.000 description 2
- 210000002798 bone marrow cell Anatomy 0.000 description 2
- 239000011575 calcium Substances 0.000 description 2
- 229910052791 calcium Inorganic materials 0.000 description 2
- 230000010261 cell growth Effects 0.000 description 2
- 230000011748 cell maturation Effects 0.000 description 2
- 230000004663 cell proliferation Effects 0.000 description 2
- 230000007541 cellular toxicity Effects 0.000 description 2
- 239000012829 chemotherapy agent Substances 0.000 description 2
- OSASVXMJTNOKOY-UHFFFAOYSA-N chlorobutanol Chemical compound CC(C)(O)C(Cl)(Cl)Cl OSASVXMJTNOKOY-UHFFFAOYSA-N 0.000 description 2
- 230000000295 complement effect Effects 0.000 description 2
- 230000021615 conjugation Effects 0.000 description 2
- 238000013270 controlled release Methods 0.000 description 2
- 229920001577 copolymer Polymers 0.000 description 2
- 230000002596 correlated effect Effects 0.000 description 2
- 230000004069 differentiation Effects 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- 210000003979 eosinophil Anatomy 0.000 description 2
- VJJPUSNTGOMMGY-MRVIYFEKSA-N etoposide Chemical compound COC1=C(O)C(OC)=CC([C@@H]2C3=CC=4OCOC=4C=C3[C@@H](O[C@H]3[C@@H]([C@@H](O)[C@@H]4O[C@H](C)OC[C@H]4O3)O)[C@@H]3[C@@H]2C(OC3)=O)=C1 VJJPUSNTGOMMGY-MRVIYFEKSA-N 0.000 description 2
- 230000007717 exclusion Effects 0.000 description 2
- 238000005194 fractionation Methods 0.000 description 2
- 108020001507 fusion proteins Proteins 0.000 description 2
- 102000037865 fusion proteins Human genes 0.000 description 2
- 230000002068 genetic effect Effects 0.000 description 2
- 231100000226 haematotoxicity Toxicity 0.000 description 2
- 230000036541 health Effects 0.000 description 2
- 230000011132 hemopoiesis Effects 0.000 description 2
- 108010028295 histidylhistidine Proteins 0.000 description 2
- 230000003054 hormonal effect Effects 0.000 description 2
- 229940088597 hormone Drugs 0.000 description 2
- 239000005556 hormone Substances 0.000 description 2
- 210000005260 human cell Anatomy 0.000 description 2
- 239000002955 immunomodulating agent Substances 0.000 description 2
- 229940121354 immunomodulator Drugs 0.000 description 2
- 238000002650 immunosuppressive therapy Methods 0.000 description 2
- 230000006698 induction Effects 0.000 description 2
- 230000001939 inductive effect Effects 0.000 description 2
- 230000002401 inhibitory effect Effects 0.000 description 2
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 description 2
- 238000004255 ion exchange chromatography Methods 0.000 description 2
- 238000001155 isoelectric focusing Methods 0.000 description 2
- 210000003734 kidney Anatomy 0.000 description 2
- 239000003446 ligand Substances 0.000 description 2
- 210000004185 liver Anatomy 0.000 description 2
- 238000011068 loading method Methods 0.000 description 2
- 238000012423 maintenance Methods 0.000 description 2
- 210000004962 mammalian cell Anatomy 0.000 description 2
- 230000035800 maturation Effects 0.000 description 2
- 230000007246 mechanism Effects 0.000 description 2
- 230000001394 metastastic effect Effects 0.000 description 2
- 239000003607 modifier Substances 0.000 description 2
- 210000001616 monocyte Anatomy 0.000 description 2
- 238000002703 mutagenesis Methods 0.000 description 2
- 231100000350 mutagenesis Toxicity 0.000 description 2
- 230000035772 mutation Effects 0.000 description 2
- 229960001592 paclitaxel Drugs 0.000 description 2
- 238000007911 parenteral administration Methods 0.000 description 2
- 230000008807 pathological lesion Effects 0.000 description 2
- 108010044644 pegfilgrastim Proteins 0.000 description 2
- 108010073025 phenylalanylphenylalanine Proteins 0.000 description 2
- 230000006461 physiological response Effects 0.000 description 2
- 230000001323 posttranslational effect Effects 0.000 description 2
- 239000002243 precursor Substances 0.000 description 2
- 238000002203 pretreatment Methods 0.000 description 2
- 230000008569 process Effects 0.000 description 2
- 238000012545 processing Methods 0.000 description 2
- 229940002612 prodrug Drugs 0.000 description 2
- 239000000651 prodrug Substances 0.000 description 2
- 230000002035 prolonged effect Effects 0.000 description 2
- 238000001742 protein purification Methods 0.000 description 2
- RXWNCPJZOCPEPQ-NVWDDTSBSA-N puromycin Chemical compound C1=CC(OC)=CC=C1C[C@H](N)C(=O)N[C@H]1[C@@H](O)[C@H](N2C3=NC=NC(=C3N=C2)N(C)C)O[C@@H]1CO RXWNCPJZOCPEPQ-NVWDDTSBSA-N 0.000 description 2
- 102000005962 receptors Human genes 0.000 description 2
- 108020003175 receptors Proteins 0.000 description 2
- 230000008929 regeneration Effects 0.000 description 2
- 238000011069 regeneration method Methods 0.000 description 2
- 230000022532 regulation of transcription, DNA-dependent Effects 0.000 description 2
- 230000010076 replication Effects 0.000 description 2
- WUAPFZMCVAUBPE-IGMARMGPSA-N rhenium-186 Chemical compound [186Re] WUAPFZMCVAUBPE-IGMARMGPSA-N 0.000 description 2
- 239000011780 sodium chloride Substances 0.000 description 2
- 239000007787 solid Substances 0.000 description 2
- 239000007790 solid phase Substances 0.000 description 2
- 241000894007 species Species 0.000 description 2
- 210000000603 stem cell niche Anatomy 0.000 description 2
- 238000003860 storage Methods 0.000 description 2
- CIOAGBVUUVVLOB-OUBTZVSYSA-N strontium-89 Chemical compound [89Sr] CIOAGBVUUVVLOB-OUBTZVSYSA-N 0.000 description 2
- 239000004094 surface-active agent Substances 0.000 description 2
- 238000001356 surgical procedure Methods 0.000 description 2
- 238000007910 systemic administration Methods 0.000 description 2
- 230000001839 systemic circulation Effects 0.000 description 2
- 229960001603 tamoxifen Drugs 0.000 description 2
- 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 description 2
- 231100000331 toxic Toxicity 0.000 description 2
- 230000002588 toxic effect Effects 0.000 description 2
- 238000011269 treatment regimen Methods 0.000 description 2
- GETQZCLCWQTVFV-UHFFFAOYSA-N trimethylamine Chemical compound CN(C)C GETQZCLCWQTVFV-UHFFFAOYSA-N 0.000 description 2
- 230000004614 tumor growth Effects 0.000 description 2
- 230000002792 vascular Effects 0.000 description 2
- 229960003048 vinblastine Drugs 0.000 description 2
- JXLYSJRDGCGARV-XQKSVPLYSA-N vincaleukoblastine Chemical compound C([C@@H](C[C@]1(C(=O)OC)C=2C(=CC3=C([C@]45[C@H]([C@@]([C@H](OC(C)=O)[C@]6(CC)C=CCN([C@H]56)CC4)(O)C(=O)OC)N3C)C=2)OC)C[C@@](C2)(O)CC)N2CCC2=C1NC1=CC=CC=C21 JXLYSJRDGCGARV-XQKSVPLYSA-N 0.000 description 2
- FDKXTQMXEQVLRF-ZHACJKMWSA-N (E)-dacarbazine Chemical compound CN(C)\N=N\c1[nH]cnc1C(N)=O FDKXTQMXEQVLRF-ZHACJKMWSA-N 0.000 description 1
- LKJPYSCBVHEWIU-KRWDZBQOSA-N (R)-bicalutamide Chemical compound C([C@@](O)(C)C(=O)NC=1C=C(C(C#N)=CC=1)C(F)(F)F)S(=O)(=O)C1=CC=C(F)C=C1 LKJPYSCBVHEWIU-KRWDZBQOSA-N 0.000 description 1
- IIZPXYDJLKNOIY-JXPKJXOSSA-N 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCC\C=C/C\C=C/C\C=C/C\C=C/CCCCC IIZPXYDJLKNOIY-JXPKJXOSSA-N 0.000 description 1
- WUAPFZMCVAUBPE-NJFSPNSNSA-N 188Re Chemical compound [188Re] WUAPFZMCVAUBPE-NJFSPNSNSA-N 0.000 description 1
- STQGQHZAVUOBTE-UHFFFAOYSA-N 7-Cyan-hept-2t-en-4,6-diinsaeure Natural products C1=2C(O)=C3C(=O)C=4C(OC)=CC=CC=4C(=O)C3=C(O)C=2CC(O)(C(C)=O)CC1OC1CC(N)C(O)C(C)O1 STQGQHZAVUOBTE-UHFFFAOYSA-N 0.000 description 1
- 206010000060 Abdominal distension Diseases 0.000 description 1
- 102000007469 Actins Human genes 0.000 description 1
- 108010085238 Actins Proteins 0.000 description 1
- QGZKDVFQNNGYKY-UHFFFAOYSA-O Ammonium Chemical compound [NH4+] QGZKDVFQNNGYKY-UHFFFAOYSA-O 0.000 description 1
- 102100026189 Beta-galactosidase Human genes 0.000 description 1
- 102100023995 Beta-nerve growth factor Human genes 0.000 description 1
- 108010006654 Bleomycin Proteins 0.000 description 1
- 208000018240 Bone Marrow Failure disease Diseases 0.000 description 1
- COVZYZSDYWQREU-UHFFFAOYSA-N Busulfan Chemical compound CS(=O)(=O)OCCCCOS(C)(=O)=O COVZYZSDYWQREU-UHFFFAOYSA-N 0.000 description 1
- KLWPJMFMVPTNCC-UHFFFAOYSA-N Camptothecin Natural products CCC1(O)C(=O)OCC2=C1C=C3C4Nc5ccccc5C=C4CN3C2=O KLWPJMFMVPTNCC-UHFFFAOYSA-N 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- 206010007559 Cardiac failure congestive Diseases 0.000 description 1
- 241000701489 Cauliflower mosaic virus Species 0.000 description 1
- 241000700198 Cavia Species 0.000 description 1
- 206010055114 Colon cancer metastatic Diseases 0.000 description 1
- 102000007644 Colony-Stimulating Factors Human genes 0.000 description 1
- 108010071942 Colony-Stimulating Factors Proteins 0.000 description 1
- 208000035473 Communicable disease Diseases 0.000 description 1
- 241000699800 Cricetinae Species 0.000 description 1
- 241000699802 Cricetulus griseus Species 0.000 description 1
- 241000701022 Cytomegalovirus Species 0.000 description 1
- 102000053602 DNA Human genes 0.000 description 1
- 102000004594 DNA Polymerase I Human genes 0.000 description 1
- 108010017826 DNA Polymerase I Proteins 0.000 description 1
- 230000005778 DNA damage Effects 0.000 description 1
- 231100000277 DNA damage Toxicity 0.000 description 1
- 102000004163 DNA-directed RNA polymerases Human genes 0.000 description 1
- 108090000626 DNA-directed RNA polymerases Proteins 0.000 description 1
- 108010092160 Dactinomycin Proteins 0.000 description 1
- 102100024746 Dihydrofolate reductase Human genes 0.000 description 1
- 102400001368 Epidermal growth factor Human genes 0.000 description 1
- 101800003838 Epidermal growth factor Proteins 0.000 description 1
- YQYJSBFKSSDGFO-UHFFFAOYSA-N Epihygromycin Natural products OC1C(O)C(C(=O)C)OC1OC(C(=C1)O)=CC=C1C=C(C)C(=O)NC1C(O)C(O)C2OCOC2C1O YQYJSBFKSSDGFO-UHFFFAOYSA-N 0.000 description 1
- 241000283086 Equidae Species 0.000 description 1
- JOYRKODLDBILNP-UHFFFAOYSA-N Ethyl urethane Chemical compound CCOC(N)=O JOYRKODLDBILNP-UHFFFAOYSA-N 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 102000018233 Fibroblast Growth Factor Human genes 0.000 description 1
- 108050007372 Fibroblast Growth Factor Proteins 0.000 description 1
- 108010029961 Filgrastim Proteins 0.000 description 1
- 241000233866 Fungi Species 0.000 description 1
- 241000287828 Gallus gallus Species 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 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 1
- 206010061188 Haematotoxicity Diseases 0.000 description 1
- 206010019280 Heart failures Diseases 0.000 description 1
- 102000012428 Hematopoietic Cell Growth Factors Human genes 0.000 description 1
- 108010022580 Hematopoietic Cell Growth Factors Proteins 0.000 description 1
- 241000238631 Hexapoda Species 0.000 description 1
- 101001002657 Homo sapiens Interleukin-2 Proteins 0.000 description 1
- 101001135738 Homo sapiens Parathyroid hormone-related protein Proteins 0.000 description 1
- 108010000521 Human Growth Hormone Proteins 0.000 description 1
- 241000701024 Human betaherpesvirus 5 Species 0.000 description 1
- AVXURJPOCDRRFD-UHFFFAOYSA-N Hydroxylamine Chemical compound ON AVXURJPOCDRRFD-UHFFFAOYSA-N 0.000 description 1
- 229920002153 Hydroxypropyl cellulose Polymers 0.000 description 1
- 102000026633 IL6 Human genes 0.000 description 1
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 1
- 108700002232 Immediate-Early Genes Proteins 0.000 description 1
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 1
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 1
- 108700001097 Insect Genes Proteins 0.000 description 1
- 102000004877 Insulin Human genes 0.000 description 1
- 108090001061 Insulin Proteins 0.000 description 1
- 108010002352 Interleukin-1 Proteins 0.000 description 1
- 102000010789 Interleukin-2 Receptors Human genes 0.000 description 1
- 102000000743 Interleukin-5 Human genes 0.000 description 1
- 102000015696 Interleukins Human genes 0.000 description 1
- 108010063738 Interleukins Proteins 0.000 description 1
- ZQISRDCJNBUVMM-UHFFFAOYSA-N L-Histidinol Natural products OCC(N)CC1=CN=CN1 ZQISRDCJNBUVMM-UHFFFAOYSA-N 0.000 description 1
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 1
- ZQISRDCJNBUVMM-YFKPBYRVSA-N L-histidinol Chemical compound OC[C@@H](N)CC1=CNC=N1 ZQISRDCJNBUVMM-YFKPBYRVSA-N 0.000 description 1
- FBOZXECLQNJBKD-ZDUSSCGKSA-N L-methotrexate Chemical compound C=1N=C2N=C(N)N=C(N)C2=NC=1CN(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FBOZXECLQNJBKD-ZDUSSCGKSA-N 0.000 description 1
- 108060001084 Luciferase Proteins 0.000 description 1
- 239000005089 Luciferase Substances 0.000 description 1
- 101710175625 Maltose/maltodextrin-binding periplasmic protein Proteins 0.000 description 1
- 102000018697 Membrane Proteins Human genes 0.000 description 1
- 108010052285 Membrane Proteins Proteins 0.000 description 1
- 206010027452 Metastases to bone Diseases 0.000 description 1
- 108700005443 Microbial Genes Proteins 0.000 description 1
- 229930192392 Mitomycin Natural products 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- 101001013152 Mycobacterium avium Major membrane protein 1 Proteins 0.000 description 1
- 101001013151 Mycobacterium leprae (strain TN) Major membrane protein I Proteins 0.000 description 1
- 241000522131 Myroconger compressus Species 0.000 description 1
- NWIBSHFKIJFRCO-WUDYKRTCSA-N Mytomycin Chemical compound C1N2C(C(C(C)=C(N)C3=O)=O)=C3[C@@H](COC(N)=O)[C@@]2(OC)[C@@H]2[C@H]1N2 NWIBSHFKIJFRCO-WUDYKRTCSA-N 0.000 description 1
- 125000001429 N-terminal alpha-amino-acid group Chemical group 0.000 description 1
- 229930193140 Neomycin Natural products 0.000 description 1
- 108010025020 Nerve Growth Factor Proteins 0.000 description 1
- 102400000058 Neuregulin-1 Human genes 0.000 description 1
- 108090000556 Neuregulin-1 Proteins 0.000 description 1
- 208000001132 Osteoporosis Diseases 0.000 description 1
- 108091006006 PEGylated Proteins Proteins 0.000 description 1
- 102000003982 Parathyroid hormone Human genes 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 102000035195 Peptidases Human genes 0.000 description 1
- 108091005804 Peptidases Proteins 0.000 description 1
- 241000286209 Phasianidae Species 0.000 description 1
- 108010038512 Platelet-Derived Growth Factor Proteins 0.000 description 1
- 102000010780 Platelet-Derived Growth Factor Human genes 0.000 description 1
- 108010039918 Polylysine Proteins 0.000 description 1
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- 239000004365 Protease Substances 0.000 description 1
- 108020004511 Recombinant DNA Proteins 0.000 description 1
- 241000714474 Rous sarcoma virus Species 0.000 description 1
- 208000003837 Second Primary Neoplasms Diseases 0.000 description 1
- 241000700584 Simplexvirus Species 0.000 description 1
- 108020004682 Single-Stranded DNA Proteins 0.000 description 1
- 241000282887 Suidae Species 0.000 description 1
- 201000001322 T cell deficiency Diseases 0.000 description 1
- 108091008874 T cell receptors Proteins 0.000 description 1
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 1
- 108010049264 Teriparatide Proteins 0.000 description 1
- 102000006601 Thymidine Kinase Human genes 0.000 description 1
- 108020004440 Thymidine kinase Proteins 0.000 description 1
- 108700005077 Viral Genes Proteins 0.000 description 1
- 108010084455 Zeocin Proteins 0.000 description 1
- 210000001015 abdomen Anatomy 0.000 description 1
- 238000010317 ablation therapy Methods 0.000 description 1
- 230000021736 acetylation Effects 0.000 description 1
- 238000006640 acetylation reaction Methods 0.000 description 1
- RJURFGZVJUQBHK-IIXSONLDSA-N actinomycin D Chemical compound C[C@H]1OC(=O)[C@H](C(C)C)N(C)C(=O)CN(C)C(=O)[C@@H]2CCCN2C(=O)[C@@H](C(C)C)NC(=O)[C@H]1NC(=O)C1=C(N)C(=O)C(C)=C2OC(C(C)=CC=C3C(=O)N[C@@H]4C(=O)N[C@@H](C(N5CCC[C@H]5C(=O)N(C)CC(=O)N(C)[C@@H](C(C)C)C(=O)O[C@@H]4C)=O)C(C)C)=C3N=C21 RJURFGZVJUQBHK-IIXSONLDSA-N 0.000 description 1
- 125000002015 acyclic group Chemical group 0.000 description 1
- 230000010933 acylation Effects 0.000 description 1
- 238000005917 acylation reaction Methods 0.000 description 1
- 108060000200 adenylate cyclase Proteins 0.000 description 1
- 102000030621 adenylate cyclase Human genes 0.000 description 1
- 238000009098 adjuvant therapy Methods 0.000 description 1
- 238000001042 affinity chromatography Methods 0.000 description 1
- 238000011256 aggressive treatment Methods 0.000 description 1
- 229930013930 alkaloid Natural products 0.000 description 1
- 229940100198 alkylating agent Drugs 0.000 description 1
- 239000002168 alkylating agent Substances 0.000 description 1
- 150000003862 amino acid derivatives Chemical class 0.000 description 1
- 125000003277 amino group Chemical group 0.000 description 1
- BFNBIHQBYMNNAN-UHFFFAOYSA-N ammonium sulfate Chemical compound N.N.OS(O)(=O)=O BFNBIHQBYMNNAN-UHFFFAOYSA-N 0.000 description 1
- 229910052921 ammonium sulfate Inorganic materials 0.000 description 1
- 235000011130 ammonium sulphate Nutrition 0.000 description 1
- 239000001166 ammonium sulphate Substances 0.000 description 1
- 230000001195 anabolic effect Effects 0.000 description 1
- 210000004102 animal cell Anatomy 0.000 description 1
- 238000000137 annealing Methods 0.000 description 1
- 208000022531 anorexia Diseases 0.000 description 1
- 230000002280 anti-androgenic effect Effects 0.000 description 1
- 230000003466 anti-cipated effect Effects 0.000 description 1
- 229940046836 anti-estrogen Drugs 0.000 description 1
- 230000001833 anti-estrogenic effect Effects 0.000 description 1
- 230000000719 anti-leukaemic effect Effects 0.000 description 1
- 230000000690 anti-lymphoma Effects 0.000 description 1
- 230000000340 anti-metabolite Effects 0.000 description 1
- 230000000840 anti-viral effect Effects 0.000 description 1
- 239000000051 antiandrogen Substances 0.000 description 1
- 229940030495 antiandrogen sex hormone and modulator of the genital system Drugs 0.000 description 1
- 238000011230 antibody-based therapy Methods 0.000 description 1
- 230000010056 antibody-dependent cellular cytotoxicity Effects 0.000 description 1
- 238000011394 anticancer treatment Methods 0.000 description 1
- 239000000427 antigen Substances 0.000 description 1
- 108091007433 antigens Proteins 0.000 description 1
- 102000036639 antigens Human genes 0.000 description 1
- 229940100197 antimetabolite Drugs 0.000 description 1
- 239000002256 antimetabolite Substances 0.000 description 1
- 239000004599 antimicrobial Substances 0.000 description 1
- 238000003782 apoptosis assay Methods 0.000 description 1
- 239000012736 aqueous medium Substances 0.000 description 1
- VSRXQHXAPYXROS-UHFFFAOYSA-N azanide;cyclobutane-1,1-dicarboxylic acid;platinum(2+) Chemical compound [NH2-].[NH2-].[Pt+2].OC(=O)C1(C(O)=O)CCC1 VSRXQHXAPYXROS-UHFFFAOYSA-N 0.000 description 1
- 229960002170 azathioprine Drugs 0.000 description 1
- LMEKQMALGUDUQG-UHFFFAOYSA-N azathioprine Chemical compound CN1C=NC([N+]([O-])=O)=C1SC1=NC=NC2=C1NC=N2 LMEKQMALGUDUQG-UHFFFAOYSA-N 0.000 description 1
- 239000013602 bacteriophage vector Substances 0.000 description 1
- 230000004888 barrier function Effects 0.000 description 1
- 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 1
- 108010005774 beta-Galactosidase Proteins 0.000 description 1
- 229960000997 bicalutamide Drugs 0.000 description 1
- 125000002619 bicyclic group Chemical group 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 230000003115 biocidal effect Effects 0.000 description 1
- 239000003124 biologic agent Substances 0.000 description 1
- 230000008827 biological function Effects 0.000 description 1
- 239000012620 biological material Substances 0.000 description 1
- 230000033228 biological regulation Effects 0.000 description 1
- 238000001815 biotherapy Methods 0.000 description 1
- 229960001561 bleomycin Drugs 0.000 description 1
- OYVAGSVQBOHSSS-UAPAGMARSA-O bleomycin A2 Chemical compound N([C@H](C(=O)N[C@H](C)[C@@H](O)[C@H](C)C(=O)N[C@@H]([C@H](O)C)C(=O)NCCC=1SC=C(N=1)C=1SC=C(N=1)C(=O)NCCC[S+](C)C)[C@@H](O[C@H]1[C@H]([C@@H](O)[C@H](O)[C@H](CO)O1)O[C@@H]1[C@H]([C@@H](OC(N)=O)[C@H](O)[C@@H](CO)O1)O)C=1N=CNC=1)C(=O)C1=NC([C@H](CC(N)=O)NC[C@H](N)C(N)=O)=NC(N)=C1C OYVAGSVQBOHSSS-UAPAGMARSA-O 0.000 description 1
- 229920001400 block copolymer Polymers 0.000 description 1
- 230000036765 blood level Effects 0.000 description 1
- 230000036770 blood supply Effects 0.000 description 1
- 150000001639 boron compounds Chemical class 0.000 description 1
- 150000001642 boronic acid derivatives Chemical class 0.000 description 1
- 108010006025 bovine growth hormone Proteins 0.000 description 1
- 229960002092 busulfan Drugs 0.000 description 1
- 229940127093 camptothecin Drugs 0.000 description 1
- VSJKWCGYPAHWDS-FQEVSTJZSA-N camptothecin Chemical compound C1=CC=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)[C@]5(O)CC)C4=NC2=C1 VSJKWCGYPAHWDS-FQEVSTJZSA-N 0.000 description 1
- 238000005251 capillar electrophoresis Methods 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 229960004562 carboplatin Drugs 0.000 description 1
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 1
- 230000021523 carboxylation Effects 0.000 description 1
- 238000006473 carboxylation reaction Methods 0.000 description 1
- 125000002843 carboxylic acid group Chemical group 0.000 description 1
- 230000021164 cell adhesion Effects 0.000 description 1
- 230000036755 cellular response Effects 0.000 description 1
- 230000004700 cellular uptake Effects 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- 238000006243 chemical reaction Methods 0.000 description 1
- 230000001767 chemoprotection Effects 0.000 description 1
- 235000013330 chicken meat Nutrition 0.000 description 1
- 229960004630 chlorambucil Drugs 0.000 description 1
- JCKYGMPEJWAADB-UHFFFAOYSA-N chlorambucil Chemical compound OC(=O)CCCC1=CC=C(N(CCCl)CCCl)C=C1 JCKYGMPEJWAADB-UHFFFAOYSA-N 0.000 description 1
- 229960004926 chlorobutanol Drugs 0.000 description 1
- 238000011210 chromatographic step Methods 0.000 description 1
- 210000000349 chromosome Anatomy 0.000 description 1
- DQLATGHUWYMOKM-UHFFFAOYSA-L cisplatin Chemical compound N[Pt](N)(Cl)Cl DQLATGHUWYMOKM-UHFFFAOYSA-L 0.000 description 1
- 229960004316 cisplatin Drugs 0.000 description 1
- 238000003776 cleavage reaction Methods 0.000 description 1
- 238000010367 cloning Methods 0.000 description 1
- 239000011248 coating agent Substances 0.000 description 1
- AGVAZMGAQJOSFJ-WZHZPDAFSA-M cobalt(2+);[(2r,3s,4r,5s)-5-(5,6-dimethylbenzimidazol-1-yl)-4-hydroxy-2-(hydroxymethyl)oxolan-3-yl] [(2r)-1-[3-[(1r,2r,3r,4z,7s,9z,12s,13s,14z,17s,18s,19r)-2,13,18-tris(2-amino-2-oxoethyl)-7,12,17-tris(3-amino-3-oxopropyl)-3,5,8,8,13,15,18,19-octamethyl-2 Chemical compound [Co+2].N#[C-].[N-]([C@@H]1[C@H](CC(N)=O)[C@@]2(C)CCC(=O)NC[C@@H](C)OP(O)(=O)O[C@H]3[C@H]([C@H](O[C@@H]3CO)N3C4=CC(C)=C(C)C=C4N=C3)O)\C2=C(C)/C([C@H](C\2(C)C)CCC(N)=O)=N/C/2=C\C([C@H]([C@@]/2(CC(N)=O)C)CCC(N)=O)=N\C\2=C(C)/C2=N[C@]1(C)[C@@](C)(CC(N)=O)[C@@H]2CCC(N)=O AGVAZMGAQJOSFJ-WZHZPDAFSA-M 0.000 description 1
- 229940047120 colony stimulating factors Drugs 0.000 description 1
- 238000009096 combination chemotherapy Methods 0.000 description 1
- 230000002301 combined effect Effects 0.000 description 1
- 239000002299 complementary DNA Substances 0.000 description 1
- 230000001010 compromised effect Effects 0.000 description 1
- 238000002591 computed tomography Methods 0.000 description 1
- 230000003750 conditioning effect Effects 0.000 description 1
- 230000001268 conjugating effect Effects 0.000 description 1
- 238000010276 construction Methods 0.000 description 1
- 238000011254 conventional chemotherapy Methods 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 238000011443 conventional therapy Methods 0.000 description 1
- 230000000875 corresponding effect Effects 0.000 description 1
- 239000003246 corticosteroid Substances 0.000 description 1
- 238000010168 coupling process Methods 0.000 description 1
- 238000004132 cross linking Methods 0.000 description 1
- WZHCOOQXZCIUNC-UHFFFAOYSA-N cyclandelate Chemical compound C1C(C)(C)CC(C)CC1OC(=O)C(O)C1=CC=CC=C1 WZHCOOQXZCIUNC-UHFFFAOYSA-N 0.000 description 1
- 230000001086 cytosolic effect Effects 0.000 description 1
- 229960000640 dactinomycin Drugs 0.000 description 1
- STQGQHZAVUOBTE-VGBVRHCVSA-N daunorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(C)=O)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 STQGQHZAVUOBTE-VGBVRHCVSA-N 0.000 description 1
- 229960000975 daunorubicin Drugs 0.000 description 1
- 206010061428 decreased appetite Diseases 0.000 description 1
- 230000001934 delay Effects 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 230000000994 depressogenic effect Effects 0.000 description 1
- 238000010511 deprotection reaction Methods 0.000 description 1
- 238000001212 derivatisation Methods 0.000 description 1
- 238000013461 design Methods 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 235000005911 diet Nutrition 0.000 description 1
- 230000037213 diet Effects 0.000 description 1
- 208000015799 differentiated thyroid carcinoma Diseases 0.000 description 1
- 238000009792 diffusion process Methods 0.000 description 1
- 108020001096 dihydrofolate reductase Proteins 0.000 description 1
- UGMCXQCYOVCMTB-UHFFFAOYSA-K dihydroxy(stearato)aluminium Chemical compound CCCCCCCCCCCCCCCCCC(=O)O[Al](O)O UGMCXQCYOVCMTB-UHFFFAOYSA-K 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- 208000035475 disorder Diseases 0.000 description 1
- VSJKWCGYPAHWDS-UHFFFAOYSA-N dl-camptothecin Natural products C1=CC=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)C5(O)CC)C4=NC2=C1 VSJKWCGYPAHWDS-UHFFFAOYSA-N 0.000 description 1
- 239000003534 dna topoisomerase inhibitor Substances 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- 231100000673 dose–response relationship Toxicity 0.000 description 1
- 229960004679 doxorubicin Drugs 0.000 description 1
- 238000001647 drug administration Methods 0.000 description 1
- 229940126534 drug product Drugs 0.000 description 1
- 238000002651 drug therapy Methods 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 230000007613 environmental effect Effects 0.000 description 1
- 229940116977 epidermal growth factor Drugs 0.000 description 1
- 210000000267 erythroid cell Anatomy 0.000 description 1
- 239000000328 estrogen antagonist Substances 0.000 description 1
- 102000015694 estrogen receptors Human genes 0.000 description 1
- 108010038795 estrogen receptors Proteins 0.000 description 1
- BEFDCLMNVWHSGT-UHFFFAOYSA-N ethenylcyclopentane Chemical compound C=CC1CCCC1 BEFDCLMNVWHSGT-UHFFFAOYSA-N 0.000 description 1
- 229960005420 etoposide Drugs 0.000 description 1
- 229940126864 fibroblast growth factor Drugs 0.000 description 1
- 229960004177 filgrastim Drugs 0.000 description 1
- 238000001914 filtration Methods 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 239000012458 free base Substances 0.000 description 1
- 238000004108 freeze drying Methods 0.000 description 1
- 230000004927 fusion Effects 0.000 description 1
- BTCSSZJGUNDROE-UHFFFAOYSA-N gamma-aminobutyric acid Chemical compound NCCCC(O)=O BTCSSZJGUNDROE-UHFFFAOYSA-N 0.000 description 1
- 238000001502 gel electrophoresis Methods 0.000 description 1
- 238000001641 gel filtration chromatography Methods 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 238000001476 gene delivery Methods 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- 102000006602 glyceraldehyde-3-phosphate dehydrogenase Human genes 0.000 description 1
- 108020004445 glyceraldehyde-3-phosphate dehydrogenase Proteins 0.000 description 1
- 230000013595 glycosylation Effects 0.000 description 1
- 238000006206 glycosylation reaction Methods 0.000 description 1
- 210000002216 heart Anatomy 0.000 description 1
- 238000003505 heat denaturation Methods 0.000 description 1
- 238000005534 hematocrit Methods 0.000 description 1
- 238000004128 high performance liquid chromatography Methods 0.000 description 1
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 1
- 238000001794 hormone therapy Methods 0.000 description 1
- 230000007236 host immunity Effects 0.000 description 1
- 208000008750 humoral hypercalcemia of malignancy Diseases 0.000 description 1
- 238000009396 hybridization Methods 0.000 description 1
- 235000011167 hydrochloric acid Nutrition 0.000 description 1
- 238000004191 hydrophobic interaction chromatography Methods 0.000 description 1
- 150000004679 hydroxides Chemical class 0.000 description 1
- 125000002887 hydroxy group Chemical group [H]O* 0.000 description 1
- 229910052588 hydroxylapatite Inorganic materials 0.000 description 1
- 238000012872 hydroxylapatite chromatography Methods 0.000 description 1
- 239000001863 hydroxypropyl cellulose Substances 0.000 description 1
- 235000010977 hydroxypropyl cellulose Nutrition 0.000 description 1
- 229960001101 ifosfamide Drugs 0.000 description 1
- HOMGKSMUEGBAAB-UHFFFAOYSA-N ifosfamide Chemical compound ClCCNP1(=O)OCCCN1CCCl HOMGKSMUEGBAAB-UHFFFAOYSA-N 0.000 description 1
- 210000002865 immune cell Anatomy 0.000 description 1
- 230000001900 immune effect Effects 0.000 description 1
- 210000000987 immune system Anatomy 0.000 description 1
- 230000002584 immunomodulator Effects 0.000 description 1
- 230000001024 immunotherapeutic effect Effects 0.000 description 1
- 230000001976 improved effect Effects 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 239000012535 impurity Substances 0.000 description 1
- 238000011065 in-situ storage Methods 0.000 description 1
- 208000000509 infertility Diseases 0.000 description 1
- 230000036512 infertility Effects 0.000 description 1
- 231100000535 infertility Toxicity 0.000 description 1
- 238000011221 initial treatment Methods 0.000 description 1
- 239000007972 injectable composition Substances 0.000 description 1
- 150000007529 inorganic bases Chemical class 0.000 description 1
- 229910052500 inorganic mineral Inorganic materials 0.000 description 1
- 229940125396 insulin Drugs 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 230000035992 intercellular communication Effects 0.000 description 1
- 229940047122 interleukins Drugs 0.000 description 1
- 230000006525 intracellular process Effects 0.000 description 1
- 239000007928 intraperitoneal injection Substances 0.000 description 1
- 238000011835 investigation Methods 0.000 description 1
- JJWLVOIRVHMVIS-UHFFFAOYSA-N isopropylamine Chemical compound CC(C)N JJWLVOIRVHMVIS-UHFFFAOYSA-N 0.000 description 1
- 229940067606 lecithin Drugs 0.000 description 1
- 235000010445 lecithin Nutrition 0.000 description 1
- 239000000787 lecithin Substances 0.000 description 1
- 230000003902 lesion Effects 0.000 description 1
- 231100000636 lethal dose Toxicity 0.000 description 1
- 206010024378 leukocytosis Diseases 0.000 description 1
- 230000029226 lipidation Effects 0.000 description 1
- 239000002502 liposome Substances 0.000 description 1
- 238000004811 liquid chromatography Methods 0.000 description 1
- 229920002521 macromolecule Polymers 0.000 description 1
- 230000036210 malignancy Effects 0.000 description 1
- 238000007726 management method Methods 0.000 description 1
- 229960004961 mechlorethamine Drugs 0.000 description 1
- HAWPXGHAZFHHAD-UHFFFAOYSA-N mechlorethamine Chemical compound ClCCN(C)CCCl HAWPXGHAZFHHAD-UHFFFAOYSA-N 0.000 description 1
- 230000001404 mediated effect Effects 0.000 description 1
- 238000002483 medication Methods 0.000 description 1
- 229960001924 melphalan Drugs 0.000 description 1
- SGDBTWWWUNNDEQ-LBPRGKRZSA-N melphalan Chemical compound OC(=O)[C@@H](N)CC1=CC=C(N(CCCl)CCCl)C=C1 SGDBTWWWUNNDEQ-LBPRGKRZSA-N 0.000 description 1
- 238000002844 melting Methods 0.000 description 1
- 230000008018 melting Effects 0.000 description 1
- 230000003340 mental effect Effects 0.000 description 1
- 108020004999 messenger RNA Proteins 0.000 description 1
- 208000011645 metastatic carcinoma Diseases 0.000 description 1
- 229960000485 methotrexate Drugs 0.000 description 1
- 230000000813 microbial effect Effects 0.000 description 1
- 230000005012 migration Effects 0.000 description 1
- 238000013508 migration Methods 0.000 description 1
- 239000011707 mineral Substances 0.000 description 1
- 150000007522 mineralic acids Chemical class 0.000 description 1
- 229960004857 mitomycin Drugs 0.000 description 1
- 108010032806 molgramostim Proteins 0.000 description 1
- 229960003063 molgramostim Drugs 0.000 description 1
- 125000002950 monocyclic group Chemical group 0.000 description 1
- 239000000178 monomer Substances 0.000 description 1
- 238000010172 mouse model Methods 0.000 description 1
- 239000003471 mutagenic agent Substances 0.000 description 1
- 230000001400 myeloablative effect Effects 0.000 description 1
- 210000003887 myelocyte Anatomy 0.000 description 1
- 230000002410 myeloprotective effect Effects 0.000 description 1
- 208000010125 myocardial infarction Diseases 0.000 description 1
- 229940086322 navelbine Drugs 0.000 description 1
- 229960004927 neomycin Drugs 0.000 description 1
- 210000005170 neoplastic cell Anatomy 0.000 description 1
- 230000001613 neoplastic effect Effects 0.000 description 1
- 229940053128 nerve growth factor Drugs 0.000 description 1
- 210000000653 nervous system Anatomy 0.000 description 1
- 229940071846 neulasta Drugs 0.000 description 1
- 230000007935 neutral effect Effects 0.000 description 1
- 238000009206 nuclear medicine Methods 0.000 description 1
- 230000000474 nursing effect Effects 0.000 description 1
- 239000003921 oil Substances 0.000 description 1
- 235000019198 oils Nutrition 0.000 description 1
- 238000005457 optimization Methods 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 150000007530 organic bases Chemical class 0.000 description 1
- 210000001672 ovary Anatomy 0.000 description 1
- 239000000199 parathyroid hormone Substances 0.000 description 1
- 229960001319 parathyroid hormone Drugs 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 230000001575 pathological effect Effects 0.000 description 1
- 230000007170 pathology Effects 0.000 description 1
- 230000037361 pathway 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
- 229960001373 pegfilgrastim Drugs 0.000 description 1
- 230000006320 pegylation Effects 0.000 description 1
- XYJRXVWERLGGKC-UHFFFAOYSA-D pentacalcium;hydroxide;triphosphate Chemical compound [OH-].[Ca+2].[Ca+2].[Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O XYJRXVWERLGGKC-UHFFFAOYSA-D 0.000 description 1
- 230000000737 periodic effect Effects 0.000 description 1
- 230000003836 peripheral circulation Effects 0.000 description 1
- 230000035699 permeability Effects 0.000 description 1
- 239000000825 pharmaceutical preparation Substances 0.000 description 1
- 229960003742 phenol Drugs 0.000 description 1
- CWCMIVBLVUHDHK-ZSNHEYEWSA-N phleomycin D1 Chemical compound N([C@H](C(=O)N[C@H](C)[C@@H](O)[C@H](C)C(=O)N[C@@H]([C@H](O)C)C(=O)NCCC=1SC[C@@H](N=1)C=1SC=C(N=1)C(=O)NCCCCNC(N)=N)[C@@H](O[C@H]1[C@H]([C@@H](O)[C@H](O)[C@H](CO)O1)O[C@@H]1[C@H]([C@@H](OC(N)=O)[C@H](O)[C@@H](CO)O1)O)C=1N=CNC=1)C(=O)C1=NC([C@H](CC(N)=O)NC[C@H](N)C(N)=O)=NC(N)=C1C CWCMIVBLVUHDHK-ZSNHEYEWSA-N 0.000 description 1
- 108010079892 phosphoglycerol kinase Proteins 0.000 description 1
- 235000011007 phosphoric acid Nutrition 0.000 description 1
- 150000003016 phosphoric acids Chemical class 0.000 description 1
- 230000026731 phosphorylation Effects 0.000 description 1
- 238000006366 phosphorylation reaction Methods 0.000 description 1
- 229920000656 polylysine Polymers 0.000 description 1
- 230000000379 polymerizing effect Effects 0.000 description 1
- 108091033319 polynucleotide Proteins 0.000 description 1
- 102000040430 polynucleotide Human genes 0.000 description 1
- 239000002157 polynucleotide Substances 0.000 description 1
- 229920005862 polyol Polymers 0.000 description 1
- 150000003077 polyols Chemical class 0.000 description 1
- 230000004481 post-translational protein modification Effects 0.000 description 1
- 239000011591 potassium Substances 0.000 description 1
- 229910052700 potassium Inorganic materials 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- 244000144977 poultry Species 0.000 description 1
- 235000013594 poultry meat Nutrition 0.000 description 1
- 238000001556 precipitation Methods 0.000 description 1
- 230000002028 premature Effects 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 230000002335 preservative effect Effects 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 230000003449 preventive effect Effects 0.000 description 1
- 238000009117 preventive therapy Methods 0.000 description 1
- MFDFERRIHVXMIY-UHFFFAOYSA-N procaine Chemical compound CCN(CC)CCOC(=O)C1=CC=C(N)C=C1 MFDFERRIHVXMIY-UHFFFAOYSA-N 0.000 description 1
- 229960004919 procaine Drugs 0.000 description 1
- 229960000624 procarbazine Drugs 0.000 description 1
- CPTBDICYNRMXFX-UHFFFAOYSA-N procarbazine Chemical compound CNNCC1=CC=C(C(=O)NC(C)C)C=C1 CPTBDICYNRMXFX-UHFFFAOYSA-N 0.000 description 1
- 230000005522 programmed cell death Effects 0.000 description 1
- 230000002062 proliferating effect Effects 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- 239000003223 protective agent Substances 0.000 description 1
- 230000004952 protein activity Effects 0.000 description 1
- 239000003528 protein farnesyltransferase inhibitor Substances 0.000 description 1
- 208000020016 psychiatric disease Diseases 0.000 description 1
- 238000010926 purge Methods 0.000 description 1
- 229950010131 puromycin Drugs 0.000 description 1
- 239000002510 pyrogen Substances 0.000 description 1
- 239000012857 radioactive material Substances 0.000 description 1
- GZUITABIAKMVPG-UHFFFAOYSA-N raloxifene Chemical compound C1=CC(O)=CC=C1C1=C(C(=O)C=2C=CC(OCCN3CCCCC3)=CC=2)C2=CC=C(O)C=C2S1 GZUITABIAKMVPG-UHFFFAOYSA-N 0.000 description 1
- 229960004622 raloxifene Drugs 0.000 description 1
- 238000010188 recombinant method Methods 0.000 description 1
- 230000007115 recruitment Effects 0.000 description 1
- 230000008439 repair process Effects 0.000 description 1
- 230000002629 repopulating effect Effects 0.000 description 1
- 230000004043 responsiveness Effects 0.000 description 1
- 230000000284 resting effect Effects 0.000 description 1
- 238000004366 reverse phase liquid chromatography Methods 0.000 description 1
- 238000004007 reversed phase HPLC Methods 0.000 description 1
- 210000003705 ribosome Anatomy 0.000 description 1
- 108010038379 sargramostim Proteins 0.000 description 1
- 229960002530 sargramostim Drugs 0.000 description 1
- 230000007017 scission Effects 0.000 description 1
- 238000004062 sedimentation Methods 0.000 description 1
- 238000001542 size-exclusion chromatography Methods 0.000 description 1
- 201000008261 skin carcinoma Diseases 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 229940075582 sorbic acid Drugs 0.000 description 1
- 235000010199 sorbic acid Nutrition 0.000 description 1
- 239000004334 sorbic acid Substances 0.000 description 1
- 238000001228 spectrum Methods 0.000 description 1
- 230000001954 sterilising effect Effects 0.000 description 1
- 238000004659 sterilization and disinfection Methods 0.000 description 1
- 210000002536 stromal cell Anatomy 0.000 description 1
- 239000000758 substrate Substances 0.000 description 1
- 235000000346 sugar Nutrition 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- 230000003319 supportive effect Effects 0.000 description 1
- 230000001629 suppression Effects 0.000 description 1
- 238000013268 sustained release Methods 0.000 description 1
- 239000012730 sustained-release form Substances 0.000 description 1
- 230000002194 synthesizing effect Effects 0.000 description 1
- 201000000596 systemic lupus erythematosus Diseases 0.000 description 1
- 230000035488 systolic blood pressure Effects 0.000 description 1
- 238000002626 targeted therapy Methods 0.000 description 1
- OGBMKVWORPGQRR-UMXFMPSGSA-N teriparatide Chemical compound C([C@H](NC(=O)[C@H](CCSC)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@@H](N)CO)C(C)C)[C@@H](C)CC)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC(C)C)C(=O)NCC(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CC=1N=CNC=1)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCSC)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC=1N=CNC=1)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC=1C=CC=CC=1)C(O)=O)C1=CNC=N1 OGBMKVWORPGQRR-UMXFMPSGSA-N 0.000 description 1
- RTKIYNMVFMVABJ-UHFFFAOYSA-L thimerosal Chemical compound [Na+].CC[Hg]SC1=CC=CC=C1C([O-])=O RTKIYNMVFMVABJ-UHFFFAOYSA-L 0.000 description 1
- 229940033663 thimerosal Drugs 0.000 description 1
- 230000002446 thrombocytic effect Effects 0.000 description 1
- 230000002992 thymic effect Effects 0.000 description 1
- 229940044693 topoisomerase inhibitor Drugs 0.000 description 1
- 229960000303 topotecan Drugs 0.000 description 1
- UCFGDBYHRUNTLO-QHCPKHFHSA-N topotecan Chemical compound C1=C(O)C(CN(C)C)=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)[C@]5(O)CC)C4=NC2=C1 UCFGDBYHRUNTLO-QHCPKHFHSA-N 0.000 description 1
- 238000001890 transfection Methods 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 230000009261 transgenic effect Effects 0.000 description 1
- 230000010474 transient expression Effects 0.000 description 1
- 238000013519 translation Methods 0.000 description 1
- 230000005909 tumor killing Effects 0.000 description 1
- 230000002476 tumorcidal effect Effects 0.000 description 1
- 238000009281 ultraviolet germicidal irradiation Methods 0.000 description 1
- 241000701447 unidentified baculovirus Species 0.000 description 1
- 241001515965 unidentified phage Species 0.000 description 1
- VBEQCZHXXJYVRD-GACYYNSASA-N uroanthelone Chemical compound C([C@@H](C(=O)N[C@H](C(=O)N[C@@H](CS)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CS)C(=O)N[C@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)NCC(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H](CO)C(=O)NCC(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CS)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(O)=O)C(C)C)[C@@H](C)O)NC(=O)[C@H](CO)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@@H](NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H](CCSC)NC(=O)[C@H](CS)NC(=O)[C@@H](NC(=O)CNC(=O)CNC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CS)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)CNC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@H](CO)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CS)NC(=O)CNC(=O)[C@H]1N(CCC1)C(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@@H](N)CC(N)=O)C(C)C)[C@@H](C)CC)C1=CC=C(O)C=C1 VBEQCZHXXJYVRD-GACYYNSASA-N 0.000 description 1
- 238000001291 vacuum drying Methods 0.000 description 1
- 235000015112 vegetable and seed oil Nutrition 0.000 description 1
- 239000008158 vegetable oil Substances 0.000 description 1
- 239000003981 vehicle Substances 0.000 description 1
- 210000003501 vero cell Anatomy 0.000 description 1
- 229960004528 vincristine Drugs 0.000 description 1
- OGWKCGZFUXNPDA-XQKSVPLYSA-N vincristine Chemical compound C([N@]1C[C@@H](C[C@]2(C(=O)OC)C=3C(=CC4=C([C@]56[C@H]([C@@]([C@H](OC(C)=O)[C@]7(CC)C=CCN([C@H]67)CC5)(O)C(=O)OC)N4C=O)C=3)OC)C[C@@](C1)(O)CC)CC1=C2NC2=CC=CC=C12 OGWKCGZFUXNPDA-XQKSVPLYSA-N 0.000 description 1
- OGWKCGZFUXNPDA-UHFFFAOYSA-N vincristine Natural products C1C(CC)(O)CC(CC2(C(=O)OC)C=3C(=CC4=C(C56C(C(C(OC(C)=O)C7(CC)C=CCN(C67)CC5)(O)C(=O)OC)N4C=O)C=3)OC)CN1CCC1=C2NC2=CC=CC=C12 OGWKCGZFUXNPDA-UHFFFAOYSA-N 0.000 description 1
- CILBMBUYJCWATM-PYGJLNRPSA-N vinorelbine ditartrate Chemical compound OC(=O)[C@H](O)[C@@H](O)C(O)=O.OC(=O)[C@H](O)[C@@H](O)C(O)=O.C1N(CC=2C3=CC=CC=C3NC=22)CC(CC)=C[C@H]1C[C@]2(C(=O)OC)C1=CC([C@]23[C@H]([C@@]([C@H](OC(C)=O)[C@]4(CC)C=CCN([C@H]34)CC2)(O)C(=O)OC)N2C)=C2C=C1OC CILBMBUYJCWATM-PYGJLNRPSA-N 0.000 description 1
- 230000003442 weekly effect Effects 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- 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
- A61K38/22—Hormones
- A61K38/29—Parathyroid hormone, i.e. parathormone; Parathyroid hormone-related peptides
Definitions
- the present invention is directed to methods and compositions for improved therapeutic methods that use cytotoxic agents. More specifically, the invention is directed to the use of PTHrP or analogs thereof to prevent, reduce, abrogate or otherwise reverse radiation-induced or drug-induced or antibody-induced cytotoxicity, especially to hematopoietic cells.
- a complete blood cell count and other blood chemistries are tests that are routinely performed.
- Patients that exhibit symptomatic anemia may be immediately transfused with packed red blood cells and platelets.
- Cytopenias anemia, neutropenia and thrombocytopenia
- Hematopoeitic growth factors such as G-CSF, GM-CSF and erythropoietin are given immediately following chemotherapy in an effort to protect the patients from opportunistic infections, sepsis and associated morbidity and mortality. Growth factor support is provided as needed.
- Exemplary hematopoietic factors that could be employed for hematopoietic reconstitution and for enhancing recovery of thymopoiesis and immune reconstitution include but are not limited to Erythropoietin, G-CSF, GM-CSF, M-CSF, TCGF, flt-3, thrombopoietin, thymic stromal lymphopoietin, keratinocyte growth factor, PDGF, IGF, IL 1, IL-2, IL-3, IL-4, IL-5, IL-6, IL- 7, IL-8, IL-9, IL-10, IL-1 1, IL- 12, IL-13; IL-15, IL- 16, IL- 17, stem cell factor (also known as c-kit ligand), and M-CSF.
- Erythropoietin G-CSF, GM-CSF, M-CSF, TCGF, flt-3, thrombopoie
- the present invention is directed to methods and compositions of therapy that can be used as an adjunct to chemotherapy, radiation therapy and other therapies that cause myelosuppression.
- the methods of the invention use PTHrP or analogues thereof to prevent, reduce, abrogate or otherwise reverse radiation-induced or drug-induced or antibody-induced cytotoxicity, and are especially applicable to protection of hematopoietic cells against radiation and/or chemotherapy-induced cytotoxicity.
- cytopenia including but not limited to anemia, neutropenia and thrombocytopenia
- the administration of the composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof may be carried out prior to, after or subsequent to the chemotherapy and/or radiation therapy.
- the administration of the composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is carried out prior to the chemotherapy and/or radiation therapy.
- the composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered to the subject between 4-7 days prior to initiation of cytotoxic chemotherapy and/or radiation therapy.
- the PTHrP, a PTHrP fragment, a PTHrP analogue, or a derivative thereof is advantageously employed as a prophylactic composition.
- the methods of the invention also may be used to decrease myelosuppression in a subject comprising administering to the subject a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof.
- the myelosuppression is produced by chemotherapy or radiation therapy or immunosuppressive therapy received during organ transplantation.
- the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is preferably administered prior to chemotherapy or the radiation therapy.
- Another embodiment of the invention contemplates a method of preventing or decreasing the level of reduction in circulating blood cells in a subject receiving radiation therapy and/or chemotherapy, comprising administering to the subject a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof prior to administration of the radiation therapy and/or chemotherapy, wherein administration of the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof prevents or produces a lower level of reduction of circulating blood cells in the subject as compared to a subject that does not receive the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof.
- the methods of the invention also may be used to treat a subject that is at risk of having a decrease in the number of cells of the hematopoietic system, comprising the step of administration to the subject an effective amount of PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof, wherein the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered before, after, or during the chemotherapy and/or radiation therapy.
- the cells the hematopoietic system may be selected from the group consisting of, including but not limited to T cells, B cells, granulocytes, macrophages, mesenchymal cells, osteoblasts, osteoclasts and multipotent adult progenitor cells, or combinations thereof.
- the decrease in the number of cells of the hematopoietic system is associated with chemo- and/or radiotherapy and/or removal of blood progenitor cells. For example, this may occur as a result of the chemotherapy and/or radiotherapy and/or removal of blood progenitor cells being administered to treat cancer.
- Also contemplated is a method for protecting hematopoeitic cells from cell death in response to a chemotherapeutic and/or radiation comprising contacting the cells with a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof prior to contacting the cells with the chemotherapeutic agent and/or radiation.
- the invention further is directed to a method for treating a cytotoxic agent-induced hematopoietic or myeloid toxicity in a human patient which comprises administering prior to, simultaneous with or subsequent to the administration of the cytotoxic agent, a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof in an amount effective to prevent, mitigate or reverse such hematopoietic or myeloid toxicity.
- the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof may, but need not necessarily be administered in combination with one or more hematopoietic factors selected from the group consisting of Erythropoietin, G-CSF, GM-CSF, M-CSF, TCGF, flt-3, thrombopoietin, thymic stromal lymphopoietin, keratinocyte growth factor, PDGF, IGF, IL 1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-IO, IL-11, IL-12, IL-13; IL-15, IL-16, IL-17, stem cell factor (c-kit ligand), and M-CSF (including PEGylated derivatives and biologically fragments thereof) is administered in an amount effective to prevent, mitigate or reverse hematopoietic or mye
- the one or more hematopoietic factors administered in the combination adjunct therapy immediately above may be administered prior to, concurrently with or subsequent to the administration of PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof.
- a method of treating cancer therapy in a mammalian patient which comprises administering to the patient a combination of a cytotoxic agent in an amount effective to treat the cancer and PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof in an amount effective to prevent, mitigate or reverse hematopoietic or myeloid toxicity, wherein the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered prior to, simultaneously with or subsequent to the administration of the cytotoxic agent, wherein administration of the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof reduces the dose of the cytotoxic agent needed to produce a therapeutic effect in the patient.
- the cytotoxic agent is a therapeutic agent, including but not limited to radiation therapy or chemotherapy.
- Also taught herein is a method treating cancer therapy in a mammalian patient which comprises administering to the patient a combination of a cytotoxic agent in an amount effective to treat the cancer and a combination of PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof and at least one hematopoietic growth factor in an amount effective to prevent, mitigate or reverse hematopoietic or myeloid toxicity, wherein the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered prior to the administration of the cytotoxic agent.
- the methods of treating cancer may be used in a mammalian patient is suffering from a cancer selected from the group consisting of Hodgkins lymphoma, NonHodgkins lymphoma, pancreatic cancer, melanoma, breast cancer, prostate cancer, bladder cancer, ovarian cancer, endometrial cancer, lung cancer, Kaposi's sarcoma, leukemia, lymphoma, gastric cancer, colon cancer, colorectal cancer, esophageal cancer, renal cancer, and head and neck cancer.
- a cancer selected from the group consisting of Hodgkins lymphoma, NonHodgkins lymphoma, pancreatic cancer, melanoma, breast cancer, prostate cancer, bladder cancer, ovarian cancer, endometrial cancer, lung cancer, Kaposi's sarcoma, leukemia, lymphoma, gastric cancer, colon cancer, colorectal cancer, esophageal cancer, renal cancer, and head and neck cancer.
- the hematopoietic growth factor may be selected from the group consisting of Erythropoietin, G-CSF, GM-CSF, M-CSF, TCGF, flt-3, thrombopoietin, thymic stromal lymphopoietin, keratinocyte growth factor, PDGF, IGF, IL 1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-IO, IL-I l, IL-12, IL-13; IL- 15, IL- 16, IL- 17, stem cell factor (c-kit ligand), and M-CSF (including PEGylated derivatives and biologically active fragments thereof).
- Also taught herein is a method for preventing or mitigating myelosuppression in a human patient undergoing therapy for cancer or tissue or organ transplantation, which comprises administering to the patient a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof in an amount effective to treat myeloid toxicity, wherein the cytokine is administered prior to, simultaneously with or subsequent to the therapy.
- the invention may be used as a method for preventing or mitigating hematopoietic cell depression in a human patient undergoing therapy for cancer or tissue or organ transplantation which comprises administering to the patient a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof in an amount effective to treat myeloid toxicity, wherein the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered prior to, simultaneously with or subsequent to the administration of the therapy.
- such a cytotoxic agent may include a radionuclide, including but not limited to Iodine- 131, Strontiun-89, Yttrium-90, Rhenium 186, Rheniuml88, Cobalt 60; Cesium 137; Iridium 192, and Radium 226.
- the cytotoxic agent is conjugated to an antibody or a bone-seeking chemical.
- Exemplary bone-seeking chemicals include orthophosphate or diphosphonate.
- Also provided herein is a method of alleviating a physical symptom caused by hematopoietic or myeloid toxicity in a human patient undergoing therapy with a cytotoxic agent, wherein the patient is administered prior to, simultaneous with or subsequent to the administration of the cytotoxic agent, a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof in an amount effective to prevent, mitigate or reverse such hematopoietic or myeloid toxicity.
- the physical symptom may be bone pain associated with bone cancer or bone metastasis.
- an exemplary patient is a cancer patient.
- the subject has a cancer selected from the group consisting of Hodgkins lymphoma, Non-Hodgkins lymphoma, pancreatic cancer, melanoma, breast cancer, prostate cancer, bladder cancer, ovarian cancer, endometrial cancer, lung cancer, Kaposi's sarcoma, leukemia, lymphoma, gastric cancer, colon cancer, colorectal cancer, esophageal cancer, renal cancer, and head and neck cancer.
- the patient is a patient receiving an organ transplant and receiving immunosuppressive therapy to kill T-cells.
- the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof may be administered through any typical route of administration including, but not limited to administration via subcutaneous, intravenous, intradermal, intraarterial, intramusclar, intramammary, intraperitoneal, intrathecal, intraocular, retrobulbar, intrapulmonary (e.g., term release), aerosol, sublingual, nasal, anal, vaginal, oral, or transdermal delivery, or by surgical implantation, or a combination thereof.
- the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered in a single dose. In other embodiments, the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered in multiple doses or as a continuous infusion during therapy. In specific aspects of the invention, the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered in a dose of about 0.5 ⁇ g/kg body weight to about lO ⁇ g /kg body weight of subject/day.
- the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof may be administered on each of 2, 3, 4, 5, 6 or 7 days prior to the administration of chemotherapy or radiation therapy.
- the methods contemplate that the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof may be administered in combination with one or more hematopoietic factors.
- the hematopoietic factor may be selected from the group consisting of Erythropoietin, G-CSF, GM-CSF, M-CSF, TCGF, flt-3, thrombopoietin, thymic stromal lymphopoietin, keratinocyte growth factor, PDGF, IGF, IL 1, IL-2, 1L-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-IO, IL-11, IL-I2, IL-13; IL-15, IL-16, IL-17, stem cell factor (c-kit ligand), and M-CSF or combinations thereof.
- the hematopoietic factor may be a biologically active fragment of such a factor.
- the hematopoietic factor or fragment thereof also may be PEGylated.
- the composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof comprises PTHrP 1-34, or an analogue thereof.
- An exemplary PTHrP 1-34 analogue is semparatide.
- Figures 1, 2 and 3 depict the hematoprotective effect of Semparatide administered to mice for 14 days prior to administration of a high dose of 5-fluorouricil, showing higher levels and faster recovery post chemotherapy of circulating WBCs, lymphocytes and neutrophils respectively.
- Bone marrow stroma and local osteoblasts permit hematopoiesis by providing environmental cues, physical connections and the appropriate growth factor support. Osteoblasts and stromal cells are able to support hematopoietic progenitors and allow them to expand. The interaction between these cells has been called a niche. Without intending to be bound by theory, it is believed that stimuli that increase the number of osteoblasts, increase the availability of hematopoietic stem cell niches, which would increase the number of hematopoietic stem cells available for release into the systemic circulation following stimulation (injection of G-CSF or administration of chemotherapy).
- osteoblasts secrete cytokines such as G-CSF, GM-CSF, ILl, IL6 and LIF all of which have been shown to modulate hematopoiesis.
- cytokines such as G-CSF, GM-CSF, ILl, IL6 and LIF all of which have been shown to modulate hematopoiesis.
- Activation of osteoblasts within the stem cell niche of bone marrow allows for the proliferation and migration of stem cells to that niche.
- these cells Upon entry into the niche, these cells are tethered and following this physical attachment and through the activation of membrane protein ligand and receptor, Notch and Jagged, remain in the G 0 resting state and are thus not sensitive to the cytotoxic effects of chemotherapy.
- membrane protein ligand and receptor Notch and Jagged
- Upon activation of the stem cell niche either by the injection of G-CSF or administration of chemotherapy, many more cells are available to enter into the differentiation pathway. Thus, greater numbers of mature WBCs and neutrophils are released into the peripheral circulation, thus decreasing the duration and extent of neutropenia.
- PTHrP e.g. Semparatide
- the therapeutic methods of the invention are useful for reconstitution of levels of all blood cell types after such cytotoxic therapy.
- PTHrP, and fragments, and analogs thereof may be used to avoid the cytopenia observed in patients following chemo- or radiation therapy. Due to the unique property of this peptide to stimulate an intracellular process in hematopoietic progenitor cells that provides them with the capacity to avoid programmed cell death (apoptosis), in certain specific embodiments, it is proposed that the subject may be pretreated with the PTHrP or analog thereof prior to the initiation of the cytotoxic chemotherapy or radiation therapy in order to avoid the myelosuppression and associated cytopenias that limit the dose and duration of chemotherapy treatment.
- Prophylactic treatment with PTHrP will also be useful in that the period of time during which the patient is at risk for developing opportunistic infections and sepsis can be greatly reduced.
- the use of PTHrP or analogs thereof as described herein will also advantageously avoid the need for transfusions of RBC and platelets. It is envisioned that this prophylactic therapy would also avoid the need for expensive treatments with blood cell growth factors such as erythropoietin and G-CSF.
- the use of PTHrP, fragments or analogs thereof is used to improve the therapeutic efficacy of anticancer, antimicrobial and autoimmune disease, and anti-organ rejection therapy in that the PTHrP, fragments, or analog thereof is used to prevent, mitigate or reverse adverse radiation-induced or drug-induced toxicity, especially to hematopoietic cells.
- Use of the PTHrP, fragment, or analog thereof will allow the subject to tolerate higher doses of cytotoxic agents that are administered to the subject for therapeutic purposes.
- the PTHrP, fragment, or analog thereof may also increase the duration of time during which the cytotoxic agents can be administered and the dose-limiting hematopoietic cell toxicity that is characteristic of such cytotoxic agents can be prevented, palliated or reversed using adjunct therapy with PTHrP.
- the present invention is directed to methods of treatment in which PTHrP is administered to a subject that has or is at risk of developing cytopenia.
- the subject is one in which the cytopenia will be induced as a result of cytotoxic agents such as chemotherapy, radiotherapy, antimicrobial agents and the like.
- the invention contemplates methods of treating such subjects in which PTHrP or an analog thereof or a combination of PTHrP and an analog thereof is administered to prevent, mitigate, reduce, abrogate, reduce or otherwise reverse radiation-induced or drug-induced toxicity of normal cells, and more particularly, hematopoietic cells.
- PTHrP peptide derived from amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, amino acids, or a derivative of PTHrP that has a structure based on PTHrP but has been modified to contain a moiety that increases the uptake or distribution characteristics of the PTHrP or otherwise improve the therapeutic properties of the peptide.
- PTHrP and its various permutations that may be used in the present invention are described in further detail below, however, it should be understood that any of the various forms of PTHrP, its analogs or its derivatives may be used in the present methods.
- certain embodiments of the invention are directed to methods that provide an adjunct therapy in which PTHrP (including its analogs and derivatives) is used as the adjunct that allows the administration of higher doses of cytotoxic agents due to increased tolerance of the recipient mammal.
- adjunct PTHrP therapy can prevent, palliate, or reverse dose-limiting cytotoxic effects of the cytotoxic agents on the hematopoeitic cells.
- the PTHrP also has a radioprotective effect on hematopoietic cells.
- a great deal of therapeutic administration of radioisotopes involves administration of beta emitters, alpha emitters and/or generation of radioisotope in situ by neutron activation of Boron-10 atoms (resulting in alpha emission from the unstable nuclide produced by neutron absorption.)
- the present invention contemplates administration of PTHrP (or analog or derivative thereof) to the subject either prior to, during or after application of the radiotherapy.
- the methods of the invention may be particularly useful as adjuncts to conventional anti-cancer therapies in which the subject is administered one or more tumoricidal agent, e.g., a drug and a radioisotope, or a radioisotope and a Boron- 10 agent for neutron-activated therapy, or a drug and a biological response modifier, or an antibody conjugate and a biological response modifier.
- tumoricidal agent e.g., a drug and a radioisotope, or a radioisotope and a Boron- 10 agent for neutron-activated therapy, or a drug and a biological response modifier, or an antibody conjugate and a biological response modifier.
- the PTHrP can be integrated into such anti-cancer therapeutic regimens to maximize the efficacy of each component thereof.
- certain anti-leukemic and anti-lymphoma antibodies conjugated with beta or alpha emitting radioisotopes can induce myeloid and other hematopoietic side effects when these agents are not solely directed to the tumor cells, particularly when the tumor cells are in the circulation and in the blood-forming organs.
- the methods of the invention contemplate administration of the PTHrP or related molecule prior to, and/or concomitantly with and/or subsequent to the administration of such antibody-based therapies to reduce or ameliorate the hematopoietic side effects of the anticancer agent, while allowing the beneficial anticancer effect of the agent.
- an appropriate dose of the PTHrP or related molecule prior to, and/or concomitantly with and/or subsequent to the administration of such antibody-based therapies to reduce or ameliorate the hematopoietic side effects of the anticancer agent, while allowing the beneficial anticancer effect of the agent.
- PTHrP or analog or derivative can be administered prior to, simultaneously with or subsequent to the administration of the therapeutic agent. It is desirable to maximize the cytotoxic activity of the therapeutic agent on the pathological lesion, such as cancer cells or infectious organisms, while minimizing toxicity of that agent to the myeloid and other hematopoietic cells. In some circumstances, the PTHrP adjunct therapy may be administered continuously with the cytotoxic therapeutic agent in order to achieve the most beneficial hematoprotective effects of the PTHrP.
- anticancer therapies produce a deleterious myelosuppressive effect.
- Typical anti-cancer agents include biological agents (biotherapy), chemotherapy agents, and radiotherapy agents.
- these compositions are provided in a combined amount effective to kill or inhibit proliferation of the cancer cell, for example, by killing cancer cells, inducing apoptosis in cancer cells, reducing the growth rate of cancer cells, reducing the incidence or number of metastases, reducing tumor size, inhibiting tumor growth, reducing the blood supply to a tumor or cancer cells, promoting an immune response against cancer cells or a tumor, preventing or inhibiting the progression of cancer.
- these therapies typically also have an effect on normal, non cancer cells.
- the present invention is particularly concerned with the deleterious effects of such therapies on normal hematopoeitic cells.
- the cancer cells are generally contacted with the anticancer agent and at the same time, the hematopoietic cells also come into contact with such agents.
- Myeloid cell death in response to chemotherapy and radiotherapy agents represents a major problem in clinical oncology.
- One goal of current cancer research is to find ways to improve the efficacy of chemo- and radiotherapy by combining it with agents that are myeloprotective or are able to reconsistitute myeloid cells.
- cytokines are typically administered after the chemotherapy and or radiotherapy to facilitate myeloid reconstitution.
- PTHrP and related compositions could be used in conjunction with chemotherapeutic, radiotherapeutic, immunotherapeutic or other biological intervention, in order to act as myeloprotectant and to enhance the growth of hematopoeitic cells.
- PTHrP may be administered prior to, after or during the administration of the other therapeutic agent intervals ranging from minutes to weeks.
- the other agent and the PTHrP are applied separately to the cell, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the agent and expression construct would still be able to exert an advantageously combined effect on the cell.
- the hematoprotection afforded by the PTHrP is protection against any conventional chemotherapy, including use of for example, alkylating agents (cyclophosphamide), anti metabolites (azothioprine), plant alkaloids and terpinoids (vinblastine, etoposide, and paclitaxel), topoisomerase inhibitors (topotecan) as well as hormone therapy (antiestrogen: tamoxifen; antiandrogens: bicalutamide).
- alkylating agents cyclophosphamide
- anti metabolites azothioprine
- plant alkaloids and terpinoids vinblastine, etoposide, and paclitaxel
- topoisomerase inhibitors topotecan
- hormone therapy antiestrogen: tamoxifen; antiandrogens: bicalutamide.
- the PTHrP compositions may be used in order to protect the subject from the myelosuppressive effects of exemplary agents that include but are not limited to cisplatin (CDDP), carboplatin, procarbazine, mechlorethamine, cyclophosphamide, camptothecin, ifosfamide, melphalan, chlorambucil, busulfan, nitrosurea, dactinomycin, daunorubicin, doxorubicin, bleomycin, plicomycin, mitomycin, etoposide (VP 16), tamoxifen, raloxifene, estrogen receptor binding agents, taxol, gemcitabien, navelbine, farnesyl-protein transferase inhibitors, transplatinum, 5-fluorouracil, vincristine, vinblastine and methotrexate, Temazolomide (an aqueous form of DTIC), or any analog or derivative variant of the foregoing.
- CDDP c
- hematoprotection derived from PTHrP administration can also be used to protect hematopoietic cells from other factors that cause DNA damage and have been used extensively include what are commonly known as ⁇ -rays, X-rays, and/or the directed delivery of radioisotopes to tumor cells.
- radioisotopes include, but are not limited to Iodine 131 , Strontium 89 , Yttrium 90 , Rhenium 186 , Rhenium 188 , Cobalt 60 ; Cesium 137 ; Iridium 192 , and Radium 226 .
- Other forms of DNA damaging factors are also contemplated such as microwaves and UV-irradiation. It is most likely that all of these factors effect a broad range of damage on DNA, on the precursors of DNA, on the replication and repair of DNA, and on the assembly and maintenance of chromosomes.
- Dosage ranges for X-rays range from daily doses of 50 to 200 roentgens for prolonged periods of time (3 to 4 wk), to single doses of 2000 to 6000 roentgens.
- Dosage ranges for radioisotopes vary widely, and depend on the half-life of the isotope, the strength and type of radiation emitted, and the uptake by the neoplastic cells.
- contacted and “exposed,” when applied to a cell are used herein to describe the process by which a therapeutic agent (e.g., the PTHrP or the cytokine or an expression construct that encodes such an agent) and a chemotherapeutic or radiotherapeutic agent are delivered to a target cell or are placed in direct juxtaposition with the target cell.
- a therapeutic agent e.g., the PTHrP or the cytokine or an expression construct that encodes such an agent
- chemotherapeutic or radiotherapeutic agent e.g., the a chemotherapeutic or radiotherapeutic agent
- WBC white blood cell
- RBC Red blood cell
- platelets platelets
- other blood elements in the subject.
- WBC white blood cell
- the subject's blood sample will be monitored for components, including but not limited to erythrocyte (red blood cell/RBC) count, thrombocyte (platelet) count, and including a differential WBC analysis to monitor the myloid/lymphoid series, as well as the bone marrow hematological picture during the course of therapy.
- erythrocyte red blood cell/RBC
- thrombocyte platelet
- the administration of the PTHrP can be repeated, with the reversal of the myeloid and platelet depressions occurring within about 5-20 days after the administration of the PTHrP, usually within about 7 days.
- the ordinary skilled clinician will appreciate that variations in the timing and dosage of PTHrP administration and combinations of the PTHrP with for example cytokines and dosages of these agents are a function of the agent used, the nature of the bone marrow and/or other hematopoietic element depressed, and the nature of the patient (e.g., prior toxicity affecting bone marrow status) and the cytotoxic agent and protocol.
- PTHrP compositions can be administered in order to treat bone pain in patients with bone metastases and primary bone cancers.
- radionuclide therapy has been found to be effective and safe, particularly with the introduction of Sr-89, Y-90 and Re-186 or Re-188, either alone or conjugated to an antibody or a bone-seeking chemical such as orthophosphate or diphosphonate.
- Chemotherapeutic agents e.g., 5-fluorouracil (5-FU), have also been known to control bone pain in patients with metastatic carcinoma.
- P-32-ortho-phosphate can be administered in several ways, including single doses of about 3 to 10 mCi, multiple consecutive doses of about 1.5 mCi, or multiple intermittent doses of 7 to 10 mCi as clinically required. In multiple and intermittent dose schedules, total doses can range from 5 to 20 mCi, depending on patient response and side effects.
- these doses can be increased by from about 10% to about 35%, preferably 15 to 25%, by simultaneous administration of continuous or intermittent doses of about 5 to 20 ⁇ g of IL-I, more preferably single repeated dose of about 0.5 ⁇ g PTHrP/kg body weight to about lO ⁇ g PTHrP/kg body weight of subject/day PTHrP (including full-length PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof) either alone or in combination with a cytokine, thereby extending the time over which radionuclide therapy can be tolerated by the patient.
- PTHrP including full-length PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof
- Re-186- diphosphonates can be used for bone pain palliation in single doses of about 5 to 10 mCi, repeated up to three times, in combination with administration of the PTHrP either prior to and/or, simultaneously and/or post-therapy with Re-186-diphosphonates.
- administration of the PTHrP may be in combination with a cytokine which can be administered prior to and/or simultaneously and/or post-administration of the diphosphonates and/or the PTHrP.
- the therapy with the composition comprising the PTHrP and/or the composition comprising the cytokine can be repeated several times during a 1-2 week therapy regimen.
- 1-131 is another effective radioisotope that has been used for the treatment of cancer. It has been shown to be especially useful for treating primary and metastatic, well- differentiated thyroid carcinomas.
- a dose of 150 mCi 1-131 has been successful, with most clinicians administering a dose between 100 and 200 mCi. Again however, bone marrow depression is a major complication of this therapy and severely limits the dosages of the I- 131 that can be tolerated by the subject.
- the present invention contemplates combining 1-131 therapy, using a dose of 150-250 mCi, with PTHrP. Again the PTHrP may be administered alone or in combination with a cytokine.
- PTHrP is administered 4-7 days prior to administration of the 1-131 and produces a marked decrease in the myelosuppression seen in the cancer patient in response to the 1-131 compositions.
- the PTHrP is administered during or after the 1-131 either alone or in combination with a cytokine and again produces a beneficial decrease in myelosuppression.
- the PTHrP-based therapy is continued to one, two, three or more weeks post-radioisotope therapy. Use of the PTHrP leads to the subject having a marked decrease in myelosuppression and also a marked increase in the tolerance of higher doeses of 1-131 doses.
- PTHrP therapy is initiated 4-7 days prior to radioisotope administration, and continued twice weekly for 2-3 weeks, doses of 1-131 between 200 and 300 mCi, preferably 200-250 mCi, could be well tolerated. This will therefore lead to an increase in the therapeutic anti-cancer dose of the 1-131 that can be administered.
- the PTHrP-based therapeutic methods of the invention can be combined with various methods of radionuclide therapy in order to obtain an effective treatment of cancer and other pathological conditions, as described, e.g., in Harbert, "Nuclear Medicine Therapy", New York, Thieme Medical Publishers, 1987, pp. 1-340.
- a physician experienced in these procedures will readily be able to adapt the PTHrP adjuvant therapy described herein to such procedures to mitigate the hematopoietic side effects of the radionuclides.
- therapy with cytotoxic drugs either administered alone or as antibody conjugates for more precisely targeted therapy, e.g., for treatment of cancer, infectious or autoimmune diseases, and for organ rejection therapy, is governed by analogous principles to radioisotope therapy with isotopes or radiolabeled antibodies.
- the ordinary skilled clinician will be able to adapt the description of PTHrP use to mitigate marrow suppression and other such hematopoietic side effects by administration of the PTHrP before, during and/or after drug therapy.
- the PTHrP-based hematoprotection also can be given to subjects being treated with anticancer antibodies.
- Invading microorganisms and proliferating cancer cells can be targeted with antibodies that bind specifically to antigens produced by or associated therewith.
- Such antibodies can directly induce a cytotoxic immune response, e.g., mediated by complement, or an indirect cytotoxic immune response, e.g., through stimulation and mobilization of T- cells, e.g., killer cells (ADCC).
- Certain of such antibodies also produce side effects which include compromise of elements of the hematopoietic system, and such side effects can be prevented, mitigated and/or reversed by administration of PTHrP as described herein.
- the dose and time period of administration of the PTHrP and/or other hematoprotectant agents will again be correlated to WBC, erythrocyte and platelet counts and other aspects of the status of the hematopoietic system.
- the route of administration of the therapeutic agent as well as of the PTHrP should be coordinated and optimized.
- intracavitary e.g., intraperitoneal
- administration of a radioisotope- antibody conjugate will eventually result in lowering of the blood cell count in a patient given a high dose of the cytotoxic immunoconjugate, due to eventual diffusion of the conjugate into the bloodstream and circulation through the bone marrow.
- Administration of the PTHrP can be advantageously effected through any typical route of administration that will allow access to the region of greatest hematopoietic cell compromise.
- Such routes of administration include, subcutaneous, intravenous, intradermal, intraarterial, intramuscular, intramammary, intraperitoneal, intrathecal, intraocular, retrobulbar, intrapulmonary (e.g., term release), aerosol, sublingual, nasal, anal, vaginal, oral, or transdermal delivery, or by surgical implantation at a particular site to have its maximum effect on the circulating peripheral blood cell counts.
- Intranasal forms of administration of the PTHrP are of particular interest. Cytokines and other hematoprotectants can be administered through the same routes as the PTHrP. The clinician also may determine whether the PTHrP should be administered as a single dose, in multiple doses or as a continuous infusion during the course of therapy.
- the PTHrP is administered concomitantly with the cytotoxic agent, it may be given by continuous intravenous infusion over several hours and optionally repeated on one or more days during and after completion of the cytotoxic therapy. Continuous administration of the PTHrP can be effected by any of the transdermal modes of drug administration known to the art or yet to be developed. Similar considerations and routes of administration also should be taken into account for administration of cytokines.
- Radioisotopes typically are administered by a variety of routes for cancer therapy. These include, e.g., intravenous, intraarterial, intracavitary (including intraperitoneal), intrathecal and subcutaneous injection, as well as by implantation of seeds of radioactive material at selected sites in the patient's body.
- routes for cancer therapy include, e.g., intravenous, intraarterial, intracavitary (including intraperitoneal), intrathecal and subcutaneous injection, as well as by implantation of seeds of radioactive material at selected sites in the patient's body.
- the type, extent and time frame for myeloid and other hematopoietic cell toxicity will vary for each mode of administration of the cytotoxic agent and with the type of agent itself. In general, bone marrow toxicity will be the most serious side effect of such therapeutic regimens and intravenous or intraarterial administration of the PTHrP will be the most effective preventive or palliative measure.
- Anticancer and antimicrobial, e.g., antiviral, drugs whose major side effects are hematologic toxicity also are typically administered through any of a variety of routes, including e.g., intravenous, intraarterial, intracavitary (including intraperitoneal), intrathecal and subcutaneous injection.
- These drugs, including toxins can be administered systemically without conjugation to a tumor-targeting or infectious lesion-targeting antibody.
- Marrow toxicity is a common limiting factor in the dosage which can be administered, and the method of the invention is effective in significantly extending the dosage range and duration of therapeutic administration of such agents.
- the drugs or toxins may be conjugated with antibodies to achieve better targeting of the drug to the pathological lesion in order to improve the therapeutic efficacy of the drugs or toxins. Still further improvement in the therapeutic efficacy of these agents can be achieved by administering these immunoconjugates with PTHrP as contemplated herein.
- PTHrP adjunct therapy it is possible to increase the dosage of the immunoconjugates that can be administered to the subject because the PTHrP affords an increased tolerance for the higher levels of drug, toxin, or other cytotoxic agent necessary for maximal therapeutic activity.
- Radioisotope antibody conjugates can be administered by, e.g., intravenous, intraarterial, intracavitary, intrathecal, intramuscular, or subcutaneous routes. Again, intravenous or intraarterial administrations of PTHrP, its derivative or analogs will normally minimize bone marrow toxicity.
- the beta and alpha emitters are preferred for radioimmunotherapy, since the patient can be treated in multiple doses on an outpatient basis.
- administration of PTHrP either alone or in combination with cytokines can be effected at convenient times by injection or even by transdermal administration of an appropriate level of PTHrP and cytokine.
- Boron- 10 compounds has been contemplated as a useful anticancer treatment (e.g., systemic administration of Boron- 10-containing compounds, e.g., borates, carborane compounds and the like, followed by neutron irradiation) however, the acceptance of this therapy has been hampered by the fact that it produces excessive toxicity to normal organs. Attempts to mitigate the unacceptable toxic side effects have involved targeting the boron atoms to tumor sites by conjugating them to site-specific antibodies.
- the methods of the invention can be used as an adjunct to provide a radioprotective treatment against the alpha radiation of the activated boron atoms using PTHrP. Systemic administration of the PTHrP will allow administration of the boron compound either locally at the tumor site or systemically with the PTHrP being able to prevent or reverse hematopoietic toxicity of the targeted cytotoxic neutron capture therapy.
- the methods of the invention also can be used in other applications in which cytotoxic agents, particularly those that affect the lymphoid system (and therein particularly the T-lymphocytes), are used to depress host immunity in certain autoimmune diseases, e.g., systemic lupus erythematosis, and in patients receiving organ transplants.
- cytotoxic agents particularly those that affect the lymphoid system (and therein particularly the T-lymphocytes)
- the cytotoxic drugs used in these disorders are similar to those often used in cancer chemotherapy, with the attendant myeloid and other hematopoietic side effects.
- specific antibodies against lymphoid cells particularly T-cells
- specific antibodies against lymphoid cells e.g., the anti-Tac monoclonal antibody of Uchiyama et al., J. Immunol.
- T-cell antibody can be conjugated with a beta- or alpha-emitting radioisotope, and this can be administered to the patient prior to undertaking organ transplantation and, if needed, also thereafter.
- the treatment of the T-cell killing dose of therapy can be combined with the use of PTHrP, according to the present invention.
- This method is useful for the long-term survival of many organ transplants, such as the kidney, heart, liver, etc., because such transplants often are accompanied by a critical period during which there is a risk of organ rejection.
- the PTHrP therapy can serve as an adjunct therapy for organ transplants.
- the dosage level of the PTHrP will be a function of the extent of compromise of the hematopoietic cells, correlated generally with the peripheral blood cell count in the patient. Periodic monitoring of the WBC, RBC, platelets and other blood cell counts and adjustment of the rate and frequency of infusion or the dosage of the PTHrP administered to achieve a relatively constant level of peripheral blood cell count will ensure that the patient does not sustain undue marrow toxicity from the therapy. Experience will permit anticipation of when the treatment will lower the levels of the circulating blood components and infusion of the PTHrP at a time and in an amount sufficient to substantially prevent depression to the components of the blood in response to the cytotoxic therapy.
- the present invention includes administration PTHrP either alone or in combination with one or more cytokines, preferably lymphokines, prior to, together with and/or subsequent to administration of cytotoxic radioisotopes, drugs and/or toxins, alone or in combination, as such or in the form of antibody conjugates.
- cytokines preferably lymphokines
- the guidelines provided herein will enable the ordinary skilled clinician to adapt PTHrP administration to enhance the efficacy and mitigate the adverse hematopoietic side effects of cytotoxic therapy as a function of WBC, platelet and erythrocyte counts, marrow component status and other particular diagnostic indicia peculiar to the individual patient.
- this invention is applicable to support an enhancement of efficacy of any cytotoxic therapy that has serious hematopoietic side effects that limit the therapy's efficacy.
- the subject may receive a bone marrow transplant.
- bone marrow cells, or stem cells that have been engineered to express properties of hematopoeitic stem cells are transplanted into the individual.
- the cells are such that they have been maintained and expanded outside of the patient.
- the cells may be heterologous (from a donor e.g., a relative) or they may be autologous (from the subject itself).
- the cells also may be banked peripheral hematopoietic stem cells that are reintroduced into the patient post chemo/radiation therapy.
- ABMT autologous bone marrow transplant
- the patient will serve as his/her own bone marrow donor.
- a normally lethal dose of irradiation or chemotherapeutic may be delivered to the patient to kill tumor cells, and the bone marrow repopulated with the patients own cells that have been maintained (and perhaps expanded) ex vivo. Because, bone marrow often is contaminated with tumor cells, it is desirable to purge the bone marrow of these cells, thus the cells, once harvested may be irradiated to kill the cancer cells therein and then expanded in culture ex vivo to produce an expanded population of bone marrow cells for administration to the subject.
- the PTHrP full length PTHrP, or related PTHrP polypeptide fragment, analog, or derivative thereof
- the PTHrP may be used to enhance engraftment/survival of bone marrow transplants and decreases the period of cytopenia and window of opportunity for opportunistic infections that are prevalent post chemo/radiation.
- PTHrP PTH related peptide
- the methods and compositions of the present invention relate to the use of PTH related peptide (PTHrP) for use as a hematoprotectant in any therapy where it is desirable to prevent cytotoxic cytopenia.
- PTHrP was previously known as the factor responsible for humoral hypercalcemia of malignancy and is a peptide of 138-174 amino acids (depending on alternative splicing) which binds to the PTH/PTHrP receptor.
- the compositions of the invention may employ a full length PTHrP; a truncated
- PTHrP polypeptide that is physiologically active, or a polypeptide analog of PTHrP.
- Such proteins or polypeptides may be produced by conventional automated peptide synthesis methods or by recombinant expression. General principles for designing and making proteins are well known to those of skill in the art and are discussed in further detail below.
- the N-terminal 34 amino acid sequence of PTHrP has limited sequence homology to that of parathyroid hormone. PTHrP is generally less potent and less bone anabolic than PTH.
- the sequence of hPTHrP (1-34) is as follows:
- U.S. Patent No. 6,583,114 provides compositions of PTHrP in which amino acid residues 22-31 form an amphipathic ⁇ -helix.
- U.S. Patent No. 6,849,710 shows methods and compositions for synthesizing various useful analogs of PTHrP.
- Other PTHrP compositions that may be used in the present invention are described in e.g., U.S. Patent 6,362,163 and U.S. Patent 6,147,186, which provide descriptions of PTHrP compositions having the amino- acid sequence:
- compositions that may be useful include analogs of PTHrP described in U.S. Patent 6,503,534; U.S. Patent 6,544,949; U.S. Patent 5,723,577; U.S. Patent 5,955,574; U.S. Patent 5,969,095; U.S. Patent 6,921,750 each of which provide a teaching of composition that comprise [Glu22,25, Leu23,28,31, Aib29, Lys26,30] PTHrP (1-34)NH 2 .
- U.S. Patent 5,688,760 describes polypeptides comprising an N-terminal amino acid sequence corresponding to amino acids 107-111 of PTHrP.
- the PTHrP may be provided as a fusion protein for example as a fusion protein with an Fc region as described in U.S. Patent 6,756,480.
- a truncated PTHrP polypeptide that is physiologically active is a polypeptide that has a sequence that is less than the full complement of amino acids found in full length PTHrP which, nonetheless, elicits a physiological response that is associated with PTHrP.
- Such a response may be greater or smaller in magnitude than the response seen from the same concentration of full length PTHrP.
- the truncated PTHrP need not be fully homologous with PTHrP to elicit a similar physiological response.
- the truncated PTHrP will be truncated from the C-terminus and will range from 30 to 40 residues, with PTHrP(I -32), PTHrP(l-34), PTHrP(l-36), PTHrP(I -37),and PTHrP(l-38) being preferred, but not exclusive, representatives of this group.
- the PTHrP may be the N-terminal sequences of between 30 to 50 residues of PTHrP, preferably from 1-32, 1-33, 1-34, 1-35, 1-36, 1-37 and 1-38, are particularly contemplated to be useful in the present invention.
- the PTHrP truncated sequences may be C-terminally truncated as compared to wild-type, N-terminally truncated as compared to wild-type or may be fragments of PTHrP that have been generated from the middle of the protein i.e., are both N- and C-terminally truncated as compared to wild-type.
- a "polypeptide analog of PTHrP” refers to a polypeptide having art-accepted substitutions, deletions or insertions relative to wild-type PTHrP or is substantially homologous to PTHrP such that the analog has a similar physiological activity.
- An analog as used herein is thus any variant of the native wild-type PTHrP in which one or more of the amino acids has been substituted by another amino acid or an amino acid derivative.
- the analog may be an analog of the full-length PTHrP or alternatively, the analog will be an analog of a truncated PTHrP that polypeptide that is physiologically active.
- Those of skill in the art are referred to e.g., U.S. Patent 5,874,086; U.S. Patent 5,798,225; U.S. Patent
- SemparatideTM is a synthetic analog of naturally occurring PTHrP (1-34). Semparatide comprised of a linear chain of 34 natural amino acids, and differs from PTHrP in the substitution of residues 22-31 with residues that form an amphiphilic helix. The sequence of the molecule is:
- SEQ ID NO 1 A-V-S-E-H-Q-L-L-H-D-K-G-K-S-I-Q-D-L-R-R-R-E-L-L-E-K-L-L-E-K-L-H-T-A
- the compound may be produced by recombinant means or may be synthesized by solid- phase polymer supported synthesis, using Fmoc-compatible coupling techniques, with acid labile side chain protection. Following completion of synthesis and deprotection, the compound may be cleaved from the solid support, purified by RP-HPLC and salt exchange (to the X acetate), and lyophilized (to afford the X acetate, N hydrate). The acetate is freely soluble in water.
- This compound has the formula:
- PTHrP full length proteins, PTHrP truncated proteins that are physiologically active, or the PTHrP analogs are derived from a native sequence that is a human sequence
- PTHrP compositions may be PTHrP compositions of any mammalian species, e.g., human, bovine, porcine or rabbit may be used in this invention, with human PTHrP being the preferred source.
- the dosage of the PTHrP or analogue or derivative thereof will range between about 0.01 and 10 ⁇ g/kg body weight per day, preferably from about 0.1 to about 0.5 fg/kg body weight per day.
- the daily dose of active ingredient is from about 0.5 to about 100 ⁇ gs, preferably from about 5 to about 10 ⁇ gs.
- This dosage may be delivered in a conventional pharmaceutical composition by a single administration, by multiple applications, or via controlled release, as needed to achieve the most effective results. Dosing will continue for as long as is medically indicated, and may range from a few weeks to several months. Such dosing may be intermittent or it may be daily.
- the dosage is initiated prior to administration of the cytotoxic therapy and may continue during and after the cytotoxic therapy has stopped.
- PTHrP derivatives are used to denote a PTHrP molecule that has been altered to modify properties other than its physiological properties.
- the PTHrP molecule may be derivatized to make it more accessible. It may be derivatized to have added to it an agent that increases its uptake or its targeting.
- attaching the PTHrP to a diphosphonate or orthophosphate will facilitate the targeting of the PTHrP (or related polypeptide fragment, analogue or derivative thereof) to the bone marrow as will attaching it to certain antibodies or Fc regions of certain antibodies.
- the PTHrP may be derivatized with a water-soluble polymer.
- PEG Polyethylene glycol
- PEGylation techniques for the effective modification of drugs.
- drug delivery polymers that consists of alternating polymers of PEG and tri-functional monomers such as lysine have been used by VectraMed (Plainsboro, NJ).
- the PEG chains typically 2000 daltons or less
- Such copolymers retain the desirable properties of PEG, while providing reactive pendent groups (the carboxylic acid groups of lysine) at strictly controlled and predetermined intervals along the polymer chain.
- the reactive pendent groups can be used for derivatization, cross-linking, or conjugation with other molecules.
- These polymers are useful in producing stable, long-circulating pro-drugs by varying the molecular weight of the polymer, the molecular weight of the PEG segments, and the cleavable linkage between the drug and the polymer.
- the molecular weight of the PEG segments affects the spacing of the drug/linking group complex and the amount of drug per molecular weight of conjugate (smaller PEG segments provides greater drug loading).
- increasing the overall molecular weight of the block co-polymer conjugate will increase the circulatory half-life of the conjugate. Nevertheless, the conjugate must either be readily degradable or have a molecular weight below the threshold-limiting glomular filtration (e.g., less than 45 kDa).
- linkers may be used to maintain the therapeutic agent in a pro-drug form until released from the backbone polymer by a specific trigger, typically enzyme activity in the targeted tissue.
- a specific trigger typically enzyme activity in the targeted tissue.
- tissue activated drug delivery is particularly useful where delivery to a specific site of biodistribution is required and the therapeutic agent is released at or near the site of pathology.
- Linking group libraries for use in activated drug delivery are known to those of skill in the art and may be based on enzyme kinetics, prevalence of active enzyme, and cleavage specificity of the selected disease- specific enzymes (see e.g., technologies of established by VectraMed, Plainsboro, NJ). Such linkers may be used in modifying the PTHrP proteins described herein for therapeutic delivery.
- PTHrP proteins or fragments thereof can be synthesized in solution or on a solid support in accordance with conventional techniques.
- Various automatic synthesizers are commercially available and can be used in accordance with known protocols. See, for example, Stewart and Young, Solid Phase Peptide Synthesis, 2d. ed., Pierce Chemical Co., (1984);Tam et al., J. Am. Chem. Soc, 105:6442, (1983); Merrifield, Science, 232: 341-347, (1986); and Barany and Merrifield, The Peptides, Gross and Meienhofer, eds, Academic
- the proteins can be readily synthesized and then screened in PTHrP receptor binding/activity assays to determine whether the proteins produced possess the requisite PTHrP-like activity as an initial screen.
- the proteins/fragments also may be tested in an exemplary physiological assays. Any PTHrP-derived protein that has at least some physiological effect associated with PTHrP may be used in the methods of the present invention.
- recombinant DNA technology may be employed wherein a nucleotide sequence which encodes a protein of choice is inserted into an expression vector, transformed or transfected into an appropriate host cell and cultivated under conditions suitable for expression as described herein below.
- Recombinant methods are especially preferred for producing longer polypeptides and a variety of expression vector/host systems may be utilized to contain and express the peptide or protein coding sequence. Recombinant expression of proteins is routine and well known to those of skill in the art.
- Expression may be achieved in microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid or cosmid DNA expression vectors; yeast transformed with yeast expression vectors (Giga-Hama et al., Biotechnol Appl Biochem., 30 ( Pt 3):235-44, 1999); insect cell systems infected with virus expression vectors (e.g., baculovirus, see Ghosh et al., MoI Ther.
- microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid or cosmid DNA expression vectors; yeast transformed with yeast expression vectors (Giga-Hama et al., Biotechnol Appl Biochem., 30 ( Pt 3):235-44, 1999); insect cell systems infected with virus expression vectors (e.g., baculovirus, see Ghosh et al., MoI Ther.
- virus expression vectors e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV
- bacterial expression vectors e.g., Ti or pBR322 plasmid; see e.g., Babe et al., Biotechnol Genet Eng Rev.; 17:213-52, 2000
- animal cell systems e.g., animal cell systems.
- virus expression vectors e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV
- bacterial expression vectors e.g., Ti or pBR322 plasmid; see e.g., Babe et al., Biotechnol Genet Eng Rev.; 17:213-52, 2000
- animal cell systems e.g., cowpBR322 plasmid
- Exemplary protocols for the recombinant expression of the peptide substrates or fusion polypeptides in bacteria, yeast and other invertebrates are known to those of skill in the art.
- Mammalian host systems for the expression of recombinant proteins also are well known to those of skill in the art.
- Mammalian cells that are useful in recombinant protein productions include but are not limited to VERO cells, HeLa cells, Chinese hamster ovary (CHO) cell lines, COS cells (such as COS-7), Wl 38, BHK, HepG2, 3T3, RIN, MDCK, A549, PC 12, K562 and 293 cells.
- COS cells such as COS-7
- Wl 38 BHK, HepG2, 3T3, RIN, MDCK, A549, PC 12, K562 and 293 cells.
- Those of skill in the art are aware of various techniques for optimizing mammalian expression of proteins, see e.g., Kaufman, MoI Biotechnol. 16(2):151-60, 2000; Colosimo et al., Biotechniques, 29(2):314-8, 2000.
- Host cell strains may be chosen for a particular ability to process the expressed protein or produce certain post- translation modifications that will be useful in providing protein activity. Such modifications of the polypeptide include, but are not limited to, acetylation, carboxylation, glycosylation, phosphorylation, lipidation and acylation. Post-translational processing which cleaves a "prepro" form of the protein may also be important for correct insertion, folding and/or function. Different host cells such as CHO, HeLa, MDCK, 293, WI38, and the like have specific cellular machinery and characteristic mechanisms for such post-translational activities and may be chosen to ensure the correct modification and processing of the introduced, foreign protein.
- vectors comprising polynucleotide molecules for encoding the PTHrP-derived proteins.
- Methods of preparing such vectors as well as producing host cells transformed with such vectors are well known to those skill in the art.
- Recombinant expression may employ a selectable marker and an origin of replication, for propagation in a host. These elements of the expression constructs are well known to those of skill in the art.
- the expression vectors include DNA encoding the given protein being operably linked to suitable transcriptional or translational regulatory sequences, such as those derived from a mammalian, microbial, viral, or insect genes.
- regulatory sequences include transcriptional promoters, operators, or enhancers, mRNA ribosomal binding sites, and appropriate sequences which control transcription and translation.
- expression vector expression construct
- expression cassette are used interchangeably throughout this specification and are meant to include any type of genetic construct containing a nucleic acid coding for a gene product in which part or all of the nucleic acid encoding sequence is capable of being transcribed.
- the choice of a suitable expression vector for expression of the PTHrP peptides or polypeptides of the invention will of course depend upon the specific host cell to be used, and is within the skill of the ordinary artisan. Methods for the construction of mammalian expression vectors are disclosed, for example, in Okayama and Berg (MoI.
- the expression construct may further comprise a selectable marker that allows for the detection of the expression of a peptide or polypeptide.
- a drug selection marker aids in cloning and in the selection of transformants, for example, neomycin, puromycin, hygromycin, DHFR, zeocin and histidinol.
- enzymes such as herpes simplex virus thymidine kinase (tk) (eukaryotic), ⁇ -galactosidase, luciferase, or chloramphenicol acetyltransferase (CAT) (prokaryotic) may be employed.
- Immunologic markers also can be employed.
- epitope tags such as the FLAG system (IBI, New Haven, CT), HA and the 6xHis system (Qiagen, Chatsworth, CA) may be employed.
- glutathione S-transferase (GST) system Pharmacia, Piscataway, NJ
- the maltose binding protein system NEB, Beverley, MA
- the selectable marker employed is not believed to be important, so long as it is capable of being expressed simultaneously with the nucleic acid encoding a gene product. Further examples of selectable markers are well known to one of skill in the art.
- promoter refers to a DNA sequence recognized by the synthetic machinery of the cell, or introduced synthetic machinery, required to initiate the specific transcription of a gene.
- Nucleotide sequences are operably linked when the regulatory sequence functionally relates to the DNA encoding the protein of interest (e.g., the PTHrP or any of the hematopoietic factors discussed herein, variants thereof and the like).
- a promoter nucleotide sequence is operably linked to a given DNA sequence if the promoter nucleotide sequence directs the transcription of the sequence.
- under transcriptional control means that the promoter is in the correct location and orientation in relation to the nucleic acid to control RNA polymerase initiation and expression of the gene. Any promoter that will drive the expression of the nucleic acid may be used.
- the particular promoter employed to control the expression of a nucleic acid sequence of interest is not believed to be important, so long as it is capable of directing the expression of the nucleic acid in the targeted cell.
- a human cell it is preferable to position the nucleic acid coding region adjacent to and under the control of a promoter that is capable of being expressed in a human cell.
- a promoter might include either a human or viral promoter.
- Common promoters include, e.g., the human cytomegalovirus (CMV) immediate early gene promoter, the SV40 early promoter, the Rous sarcoma virus long terminal repeat, ⁇ -actin, rat insulin promoter, the phosphoglycerol kinase promoter and glyceraldehyde-3-phosphate dehydrogenase promoter, all of which are promoters well known and readily available to those of skill in the art, can be used to obtain high-level expression of the coding sequence of interest.
- CMV human cytomegalovirus
- an enhancer Another regulatory element that is used in protein expression is an enhancer. These are genetic elements that increase transcription from a promoter located at a distant position on the same molecule of DNA. Where an expression construct employs a cDNA insert, one will typically desire to include a polyadenylation signal sequence to effect proper polyadenylation of the gene transcript. Any polyadenylation signal sequence recognized by cells of the selected transgenic animal species is suitable for the practice of the invention, such as human or bovine growth hormone and SV40 polyadenylation signals. Also contemplated as an element of the expression cassette is a terminator. These elements can serve to enhance message levels and to minimize read through from the cassette into other sequences.
- the termination region which is employed primarily will be one selected for convenience, since termination regions for the applications such as those contemplated by the present invention appear to be relatively interchangeable.
- the termination region may be native with the transcriptional initiation, may be native to the DNA sequence of interest, or may be derived for another source.
- Site-specific mutagenesis may be useful in the preparation of individual PTHrP related proteins used in the methods of the invention.
- This technique employs specific mutagenesis of the underlying DNA (that encodes the amino acid sequence that is targeted for modification).
- the technique further provides a ready ability to prepare and test sequence variants, incorporating one or more of the foregoing considerations, by introducing one or more nucleotide sequence changes into the DNA.
- Site-specific mutagenesis allows the production of mutants through the use of specific oligonucleotide sequences that encode the DNA sequence of the desired mutation, as well as a sufficient number of adjacent nucleotides, to provide a primer sequence of sufficient size and sequence complexity to form a stable duplex on both sides of the deletion junction being traversed.
- a primer of about 17 to 25 nucleotides in length is preferred, with about 5 to 10 residues on both sides of the junction of the sequence being altered.
- the technique typically employs a bacteriophage vector that exists in both a single stranded and double stranded form.
- Typical vectors useful in site-directed mutagenesis include vectors such as the M 13 phage. These phage vectors are commercially available and their use is generally well known to those skilled in the art.
- Double stranded plasmids also are routinely employed in site-directed mutagenesis, and eliminates the step of transferring the gene of interest from a phage to a plasmid.
- site-directed mutagenesis is performed by first obtaining a single-stranded vector, or melting of two strands of a double stranded vector which includes within its sequence a DNA sequence encoding the desired protein.
- An oligonucleotide primer bearing the desired mutated sequence is synthetically prepared. This primer is then annealed with the single-stranded DNA preparation, taking into account the degree of mismatch when selecting hybridization (annealing) conditions, and subjected to DNA polymerizing enzymes such as E. coli polymerase I Klenow fragment, in order to complete the synthesis of the mutation- bearing strand. Thus, a heteroduplex is formed wherein one strand encodes the original non- mutated sequence and the second strand bears the desired mutation. This heteroduplex vector is then used to transform appropriate cells, such as E. coli cells, and clones are selected that include recombinant vectors bearing the mutated sequence arrangement.
- the above described approach for site-directed mutagenesis is not the only method of generating potentially useful mutant peptide species and as such is not meant to be limiting.
- the present invention also contemplates other methods of achieving mutagenesis such as for example, treating the recombinant vectors carrying the gene of interest mutagenic agents, such as hydroxylamine, to obtain sequence variants.
- Protein purification techniques are well known to those of skill in the art. These techniques involve, at one level, the crude fractionation of the cellular milieu to polypeptide and non-polypeptide fractions. Having separated the peptides or polypeptides of the invention from other proteins, the polypeptides or peptides of interest may be further purified using chromatographic and electrophoretic techniques to achieve partial or complete purification (or purification to homogeneity). Analytical methods particularly suited to the preparation of a pure peptide include size-exclusion chromatography, ion-exchange chromatography, hydrophobic interaction chromatography, isoelectric focusing and capillary electrophoresis.
- a particularly efficient method of purifying peptides is fast protein liquid chromatography (FPLC) or even high performance liquid chromatography (HPLC).
- Certain aspects of the present invention concern the purification, and in particular embodiments, the substantial purification, of an encoded polypeptide, protein or peptide.
- the term "purified polypeptide, protein or peptide" as used herein, is intended to refer to a composition, isolated from other components, wherein the polypeptide, protein or peptide is purified to any degree relative to its naturally-obtainable state.
- a purified polypeptide, protein or peptide therefore also refers to a polypeptide, protein or peptide, free from the cellular environment in which it may naturally occur.
- purified will refer to a polypeptide, protein or peptide composition that has been subjected to fractionation to remove various other components, and which composition substantially retains its expressed biological activity. Where the term “substantially purified” is used, this designation will refer to a composition in which the polypeptide, protein or peptide forms the major component of the composition, such as constituting about 50%, about 60%, about 70%, about 80%, about 90%, about 95% or more of the proteins in the composition.
- Hematopoietic reconstitution has previously been achieved by administration of cytokine growth factors such as Erythropoietin, G-CSF, GM-CSF, M-CSF, TCGF, flt-3, thrombopoietin, thymic stromal lymphopoietin, keratinocyte growth factor, PDGF, IGF, IL 1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL- 12, IL-13; IL-15, IL- 16, IL- 17, stem cell factor (also known as c-kit ligand), and M-CSF, or PEGylated derivatives thereof, as soon as possible after the subject has been exposed to the chemotherapeutic and/or radiotherapeutic agent.
- cytokine growth factors such as Erythropoietin, G-CSF, GM-CSF, M-C
- Combination therapy contemplated herein includes administration of one or more hematopoietic factors selected from the group consisting of Erythropoietin, G-CSF, GM-CSF, M-CSF, TCGF, flt-3, thrombopoietin, thymic stromal lymphopoietin, keratinocyte growth factor, PDGF, IGF, IL 1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13; IL-15, IL-16, IL-17, stem cell factor (also known as c-kit ligand), and M-CSF, or PEGylated derivatives thereof in combinations with PTHrP (the hematopoietic factor
- Cytokines are hormone-like peptides produced by diverse cells and are capable of modulating the proliferation, maturation and functional activation of particular cell types.
- cytokines refer to a diverse array of growth factors, such as hematopoietic cell growth factors (e.g., erythropoietin, colony stimulating factors and interleukins), nervous system growth factors (e.g., glial growth factor and nerve growth factor), mostly mesenchymal growth factors (e.g., epidermal growth factor), platelet-derived growth factor, and fibroblast growth factor I, II and III and the like.
- hematopoietic cell growth factors e.g., erythropoietin, colony stimulating factors and interleukins
- nervous system growth factors e.g., glial growth factor and nerve growth factor
- mesenchymal growth factors e.g., epidermal growth factor
- platelet-derived growth factor fibroblast growth factor I, II and III and the like.
- cytokine As such, limiting the amount of cytokine to be used in treating a patient that has undergone myelosuppressive therapy is desirable.
- the combined use of PTHrP with the cytokine-based therapy will allow the use of less cytokine for treating the subject post radiation or post chemotherapy than the amount that would be needed in the absence of the PTHrP.
- cytokines for use in the method and compositions of the invention are lymphokines, i.e., those cytokines which are primarily associated with induction of cell differentiation and maturation of myeloid and possibly other hematopoietic cells.
- a preferred lymphokine is IL- 1.
- Other such lymphokines include, but are not limited to, G-CSF, M-CSF, GM-CSF, Multi- CSF (IL-3), and IL-2 (T-cell growth factor, TCGF).
- IL-I appears to have its effect mostly on myeloid cells
- IL-2 affects mostly T-cells
- IL-3 affects mutiple precursor lymphocytes
- G- CSF affects mostly granulocytes and myeloid cells
- M-CSF affects mostly macrophage cells
- GM-CSF affects both granulocytes and macrophage.
- Other growth factors affect immature platelet (thrombocyte) cells, erythroid cells, and the like.
- the cytokines may be used alone or may be provided in a combination of two or more cytokines in order to provide protection against, mitigation and/or reversal of myeloid or hematopoietic toxicity associated with cytotoxic agents when administered in combination with the PTHrP as described herein.
- Examples of possible combinations include IL-I + GC- CSF + PTHrP, IL-I + IL-3 + PTHrP, G-CSF + IL-3 + PTHrP, IL-I + platelet growth factor + PTHrP and the like. Certain combinations will be preferred, depending on the maturation state of the target cells to be affected, and the time in the course of cytotoxic action that the protective agent needs to be administered.
- a combination of IL-I + IL-3/and/or platelet growth factor in combination with PTHrP may be particularly useful, while more severe depression of the myeloid series may require such combinations as IL-I + G-CSF in combination with the PTHrP.
- Certain cytotoxic agents have greater compromising effects on particular hematopoietic elements, either because of the nature of the agent or the dosage necessary to achieve a therapeutic effect, and the appropriate choice, dosage and mode of administration of cytokine(s) will follow from such effects.
- the PTHrP may be provided in combination with other compounds or techniques for preventing, mitigating or reversing the side effects of cytotoxic agents.
- examples of such combinations include, e.g., administration of the PTHrP and a cytokine together with a second antibody for rapid clearance, as described, e.g., in Goldenberg, U.S.
- Patent 4,624,846 from 3 to 72 hours after administration of a targeted primary antibody or antibody fragment conjugate (with a radioisotope, drug or toxin as the cytotoxic component of the immunoconjugate) or of a non-conjugated drug or toxin, to enhance clearance of the conjugate, drug or toxin from the circulation and to mitigate or reverse myeloid and other hematopoietic toxicity caused by the therapeutic agent.
- a targeted primary antibody or antibody fragment conjugate with a radioisotope, drug or toxin as the cytotoxic component of the immunoconjugate
- a non-conjugated drug or toxin to enhance clearance of the conjugate, drug or toxin from the circulation and to mitigate or reverse myeloid and other hematopoietic toxicity caused by the therapeutic agent.
- it is a desired goal bring the peripheral blood cell counts and differential to within the normal levels or as close thereto as possible.
- the RBC level in terms of hemoglobin concentration which is normally 13.5-17.5 gm/dL in men and 1 1.8 - 15.5 gm/dL in women, is reduced to less than 10 gm/dL with chemotherapy or radiotherapy; WBC count which is normally in the range of 4-1 lxlO 9 /L is reduced to less than lxlO 9 /L in subjects in response to chemotherapy of radiotherapy; platelets are in a normal range of 150,000 to 400,000/ ⁇ L but are reduced to 50,000/uL in response to chemotherapy or radiotherapy.
- the methods of the invention are useful in that where the cytotoxic therapy produces such a reduction in blood cell counts, this reduction can be reversed by administration of either PTHrP alone or a combination of PTHrP with one or more cytokines and or one or more other agents.
- cytokine is administered in combination with the PTHrP to return the blood component levels to be close to normal. It is contemplated that the cytokine is administered in a dose of from about 1 ⁇ g to about 500 ⁇ g, preferably 5-100 ⁇ g, more preferably about 10 ug.
- a complete blood cell count will routinely be monitored prior to initiation of the therapeutic regimen as well as during and post-therapy follow-up. During treatment and follow-up, monitoring the patient's blood cell count is important (e.g., prior to each treatment cycle and 10-14 days after each treatment cycle).
- a complete blood count provides important information about the kinds and numbers of cells in the blood, including red blood cells, white blood cells, and platelets.
- a complete blood count typically will involve determining a white blood cell
- WBC white blood cell count. These cells protect the body against infection and the number of white cells can increase dramatically upon infection or chemo- or radio-therapeutic insult, as such, the number of white blood cells is sometimes used to identify an infection or monitor the body's response to cancer treatment. Analysis of the white blood cell types (WBC differential) in blood will be a useful indicator for the efficacy of the methods of the invention.
- the major types of white blood cells are neutrophils, lymphocytes, monocytes, eosinophils, and basophils. Immature neutrophils, called band neutrophils, are also included and counted as part of this test. Each type of cell plays a different role in protecting the body. The numbers of each one of these types of white blood cells give important information about the immune system.
- An increase or decrease in the numbers of the different types of white blood cells can help identify infection, an allergic or toxic reaction to certain medications or chemicals, and many conditions, such as leukemia.
- WBC white blood cell
- a health professional will look at both the number (WBC count) and the WBC differential. To determine whether there are too many or too few of a certain type of cell, the total count is multiplied by the percentage of that particular cell. There are normal values for the total number of each type of white cell
- the typical normal range of white blood cells in males and non-pregnant females is between 4,500-11,000 cells/microliter ( ⁇ L) 3 or 4.5-11.0 x 10 9 /Liter (SI units). It is contemplated that the PTHrP compositions administered in accordance with the present invention will maintain or return the subjects white blood cell count to within this normal range.
- the average distribution of white blood cell types 47%-77% neutrophils; 0%-3% band neutrophils; 16%-43% lymphocytes, 0.5%-10%, monocytes, 0.3%-7% eosinophils, and 0.3%-2% basophils.
- the PTHrP administration in accordance with the methods of the present invention will balance the white blood cell differential such that the white blood cell components are present in these appropriate distributions relative to each other.
- red blood cell count average value 4.6-6.2 million RBCs per microliter ( ⁇ L) or 4.6-6.2 x 10 l2 /Liter (SI units), a hematocrit (40%-54% in men; 37%-47% women; 31-41% children; 44%-64% in newborns) and/or perform hemoglobin test (14-18 g/dL or 8.7-11.2 mmol/L (SI units) in men and 12-16 g/dL or 7.4-9.9 mmol/L in women).
- ESR erythrocyte sedimentation rate
- erticulocyte count red eel distribution width
- red blood cell indices red blood cell indices.
- the platelet count also could be determined.
- the average normal platelet count is 140,000-450,000 platelets per mm 3 or 150-400 x 109/Liter (SI units). Methods for performing such tests for various blood components are known to those of skill in the art.
- compositions for administration according to the present invention can comprise at least one PTHrP-derived protein (e.g., a full length PTHrP polypeptide, or a variant or analog thereof or any other PTHrP-related protein that protects hematopoeitic cells from the cytotoxic effects of chemo- or radiotherapy).
- PTHrP-derived protein e.g., a full length PTHrP polypeptide, or a variant or analog thereof or any other PTHrP-related protein that protects hematopoeitic cells from the cytotoxic effects of chemo- or radiotherapy.
- the pharmaceutical compositions also may include other therapeutic agents.
- the present invention stems from the unique discovery of the therapeutic efficacy of PTHrP compositions in the protection of hematopoietic cells from cytotoxic chemotherapy or radiotherapy
- the PTHrP may be administered not only alone but in combination with other therapeutic regimens designed for the treatment of cytotoxic agent-induced cytopenia (e.g., anemia, neutropenia and thrombocytopenia). Any increase in the level of peripheral blood cell count or any component of the blood count in response to such therapy will be a useful therapeutic outcome.
- cytokines such as Erythropoietin, G-CSF, GM-CSF, M-CSF, TCGF, flt-3, thrombopoietin, thymic stromal lymphopoietin, keratinocyte growth factor, PDGF, IGF, IL 1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-IO, IL-I l, IL- 12, IL-13, IL- 15, IL- 16, IL- 17, stem cell factor (also known as c-kit ligand), and M-CSF, and the like may be useful in a combination therapy protocol.
- one or more of cytokines may be provided in the pharmaceutical compositions to be used in the treatment of subjects that are about to undergo chemotherapy, radiotherapy or some other therapy that will have a myelosuppressive effect
- compositions can be administered by any means that achieve their intended purposes.
- Individualized amounts and regimens for the administration of the compositions for the treatment of infertility using the methods of the present invention can be determined readily by those with ordinary skill in the art using the guidance provided herein to determine the efficacy of a dosage in an animal model and then to increase the dosage to higher mammals.
- Administration of many cytokines are known to those of skill in the art and can readily be found in the Regimens for use of PTH in other indications also are known and discussed in the Physician's Desk Reference.
- compositions within the scope of this invention include all compositions comprising at least one PTHrP-related polypeptide formulated in an amount effective to achieve its intended purpose of protecting a composition of hematopoietic cells from cell death and/or preventing or overcoming myelosuppression.
- the active agents used in the methods of the present invention may be administered by any means normally employed for such administration.
- compositions according to the present invention will depend upon the age, health and weight of the recipient, kind of concurrent treatment, if any, frequency of treatment, and the nature of the effect desired.
- the most preferred dosage can be tailored to the individual subject, as is understood and determinable by one of skill in the art, without undue experimentation. This typically involves adjustment of a standard dose, e.g., reduction of the dose if the patient has a low body weight. Peripheral blood cell counts and differentials will be a useful determinant in assisting the clinician to determine and/or adjust the dose.
- the total dose required for each treatment may be administered in multiple doses or in a single dose.
- the compositions may be administered alone or in conjunction with other therapeutics directed to the disease or directed to other symptoms thereof.
- compositions of the invention should be formulated into suitable pharmaceutical compositions, i.e., in a form appropriate for in vivo applications in a the therapeutic intervention of myelosuppression. Generally, this will entail preparing compositions that are essentially free of pyrogens, as well as other impurities that could be harmful to humans or animals, preferably for oral administration.
- the hormone formulations may be formulated akin to the currently available hormonal preparations.
- the peptide/protein formulations may be formulated similarly to any other small protein composition. All routes of administration are contemplated (e.g.
- Such routes of administration include, subcutaneous, intravenous, intradermal, intraaterial, intramuscular, intramammary, intraperitoneal, intrathecal, intraocular, retrobulbar, intrapulmonary (e.g., term release), aerosol, sublingual, nasal, anal, vaginal, oral, or transdermal delivery, or by surgical implanation).
- Transdermal patches also may be used. Intranasal administration is of particular interest.
- compositions of the invention are provided in lyophilized form to be reconstituted prior to administration. Buffers and solutions for the reconstitution of the compositions may be provided along with the pharmaceutical formulation to produce aqueous compositions of the present invention for administration.
- aqueous compositions will comprise an effective amount of each of the therapeutic agents being used, dissolved or dispersed in a pharmaceutically acceptable carrier or aqueous medium.
- Such compositions also are referred to as inocula.
- phrases "pharmaceutically or pharmacologically acceptable” refer to molecular entities and compositions that do not produce adverse, allergic, or other untoward reactions when administered to an animal or a human.
- pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the therapeutic compositions, its use in therapeutic compositions is contemplated. Supplementary active ingredients also can be incorporated into the compositions.
- compositions according to the present invention will be via any common route so long as the target tissue is available via that route.
- Conventional routes of administration include, subcutaneous, intravenous, intradermal, intraarterial, intramusclar, intramammary, intraperitoneal, intrathecal, intraocular, retrobulbar, intrapulmonary (e.g., term release), aerosol, sublingual, nasal, anal, vaginal, oral, or transdermal delivery, or by surgical implantation at a particular site.
- the treatment may consist of a single dose or a plurality of doses over a period of time.
- the route of administration is chosen for the therapeutic intervention to have maximum impact on the circulating blood cell counts.
- the active compounds may be prepared for administration as solutions of free base or pharmacologically acceptable salts in water suitably mixed with a surfactant, such as hydroxypropylcellulose.
- Dispersions also can be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
- the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
- the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
- the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
- the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
- a coating such as lecithin
- surfactants for example, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sorbic acid, thimerosal, and the like.
- isotonic agents for example, sugars or sodium chloride.
- Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
- Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
- dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
- the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
- compositions of the present invention may be formulated in a neutral or salt form.
- Pharmaceutically-acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups also can be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like. Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
- inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like.
- Salts formed with the free carboxyl groups also can be derived from inorganic bases such as, for example, sodium, potassium,
- the formulations are easily administered in a variety of dosage forms such as injectable solutions, drug release capsules and the like.
- parenteral administration in an aqueous solution for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
- aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.
- Unit dose is defined as a discrete amount of a therapeutic composition dispersed in a suitable carrier. Parenteral administration of the therapeutic compounds may be carried out with an initial bolus followed by continuous infusion to maintain therapeutic circulating levels of drug product. Those of ordinary skill in the art will readily optimize effective dosages and administration regimens as determined by good medical practice and the clinical condition of the individual patient.
- the frequency of dosing will depend on the pharmacokinetic parameters of the agents and the routes of administration.
- the optimal pharmaceutical formulation will be determined by one of skill in the art depending on the route of administration and the desired dosage. Such formulations may influence the physical state, stability, rate of in vivo release and rate of in vivo clearance of the administered agents.
- a suitable dose may be calculated according to body weight, body surface areas or organ size. Further refinement of the calculations necessary to determine the appropriate treatment dose is routinely made by those of ordinary skill in the art without undue experimentation, especially in light of the dosage information and assays disclosed herein as well as the pharmacokinetic data observed in animals or human clinical trials.
- Appropriate dosages may be ascertained through the use of established assays for determining blood levels in conjunction with relevant dose response data.
- the final dosage regimen will be determined by the attending physician, considering factors which modify the action of drugs, e.g., the drug's specific activity, severity of the damage and the responsiveness of the patient, the age, condition, body weight, sex and diet of the patient, the severity of any infection, time of administration and other clinical factors. As studies are conducted, further information will emerge regarding appropriate dosage levels and duration of treatment for specific diseases and conditions.
- the pharmaceutical compositions and treatment methods of the invention may be useful in fields of human medicine and veterinary medicine.
- the subject to be treated may be a mammal, preferably human or other animal.
- subjects include for example, farm animals including cows, sheep, pigs, horses and goats, companion animals such as dogs and cats, exotic and/or zoo animals, laboratory animals including mice rats, rabbits, guinea pigs and hamsters; and poultry such as chickens, turkeys, ducks and geese.
- kits for use in the treatment of myelosuppression include at least a first composition comprising the PTHrP-based proteins/peptides described above in a pharmaceutically acceptable carrier.
- Another component may be an agent used in treating myelosuppression (e.g., a composition that contains one or more cytokines etc.) in a pharmaceutically acceptable carrier.
- a third component may be the agent that is used for the radiotherapy or chemotherapy.
- the kits may additionally comprise solutions or buffers for effecting the delivery of the first, second and third compositions.
- the kits may further comprise catheters, syringes or other delivering devices for the delivery of one or more of the compositions used in the methods of the invention.
- the kits may further comprise instructions containing administration protocols for the therapeutic regimens.
- a patient that is about to undergo chemotherapy or radiation therapy for cancer is selected for treatment.
- the patient is treated once daily with 50 ⁇ g PTHrP or an analogue by subcutaneous injection (on each days -7 to day 0).
- the patient On day 0, the patient is treated with chemotherapy or radiation therapy. Twenty four hours after administration of the chemotherapy (day 1) and for the next 13 days (days 2-14) the patients will receive a once daily subcutaneous injection of PTHrP or analogue at the same pre-chemotherapy dose.
- Frequent blood sampling will allow the physician to terminate PTHrP therapy prior to day 14 if circulating levels of RBCs, WBCs or platelets return to pre-treatment (normal) levels.
- the patient is able to tolerate greater amounts of the chemotherapy or radiotherapy and as such the therapeutic efficacy of the chemotherapy or radiotherapy is enhanced.
- the patient to be treated is one that has metastatic colon cancer.
- exemplary treatment protocols such a patient is treated with 5-fluorouracil (5-FU) as the chemotherapeutic agent.
- 5-FU 5-fluorouracil
- Seven days prior to initiation of the 5-FU therapy 50 ⁇ g PTHrP or an analogue are administered to the patient by subcutaneous injection (on each days -7 to day 0).
- the patient is given 5-FU therapy over 5 days at a dose of 10 mg/kg/day intravenously (days 0, 1,2,3,4,5), repeated every second day during the next five days (day 7, day 9), and then twice monthly (1st and 15th day of each month) thereafter.
- toxicity usually begins to show by the 23rd day, whereby a drop in peripheral blood cell count.
- 50 ⁇ g PTHrP or an analogue is administered per day by subcutaneous injection.
- This therapy is accompanied by frequent blood sampling and the PTHrP therapy is terminated or reduced if circulating levels of RBCs, WBCs or platelets return to pre-treatment (normal) levels.
- CAT scans several months after initiation of therapy.
- the patient to be treated has breast cancer. Seven days prior to instituting this chemotherapy, she is started on a PTHrP or a PTHrP regimen in which she is given a dose of 50 ⁇ g PTHrP or an analogue thereof per day for seven day. On day 8 and she is to be administered adriamycin (30 mg/kg i.v. daily for 3 weeks) and cytoxin (10 mg/kg i.v. every week). In a typical treatment for breast cancer the dose of adriamycin and cytoxin has be reduced due to cytotoxicity of the drugs.
- a patient presenting a peritoneal spread of a colon cancer that is non-responsive to 5-FU treatment is to be treated with radiotherapy.
- the patient will be treated with a 35 mCi dose of Yttriwn-90 conjugated to a F(ab') 2 fragment of a murine monoclonal antibody against carcinoembryonic antigen (CEA), by intraperitoneal injection.
- CEA carcinoembryonic antigen
- the patient is administered PTHrP or analogue thereof at a dose of 50 ⁇ g/day over the following fourteen days. On day 2, and periodically over the next three weeks, the patient's peripheral blood cell count is taken. As long as there is no significant drop in the patient's blood cell count as compared to his normal peripheral blood cell count, the radioimmunotherapy is repeated 3 weeks later.
- the radioimmunotherapy may be accompanied by further administration of the PTHrP or analogue thereof.
- the patient is again monitored over the coming months. A further course of treatment is given 2-3 months after the initial treatment, again with administration of PTHrP or analogue thereof initiated 7 days prior to administration of the radiotherapy. Radiological evidence of some tumor and ascites reduction is monitored. Administration of the PTHrP or analogue thereof will allow the patient to tolerate higher and more frequent doses of the radioimmunotherapy.
- the patient presents with ovarian cancer manifesting as ascites and considerable distension of her abdomen.
- the patient is initiated on a prophylactic course of PTHrP or analogue thereof seven days prior (days -7 to 0) to the initiation of the cancer therapy.
- She is given 50 ⁇ g of the PTHrP or analogue thereof per day.
- At day 0 is given a course of 5 FU therapy as described above.
- the patient continues to receive PTHrP or analogue thereof over the next 14 days. It is anticipated that the treatment with the PTHrP or analogue thereof allows the patient to tolerate multiple doses of the 5 -FU and within months of the treatment there is a reduction in the abdominal distension.
- the patient to be treated is one that is to receive a kidney transplant. 7 consecutive days prior to surgery, the patient is given 50 ⁇ g/day PTHrP or analogue thereof. On the day of the surgery, the patient is given an intravenous infusion of 20 mCi Y-90 that is conjugated to an agent that targets the Y-90 to activated T-lymphocytes, in order to kill the T-cells responsible to transplant rejection. The patient is given 50 ⁇ g/day PTHrP or analogue thereof and may also receive conventional corticosteroid therapy to prevent transplant rejection. Ten days later, a second dose of 10 mCi Y-90 is given i.v. During the interim period, the patient has continued to receive 50 ⁇ g/day doses of PTHrP or analogue thereof. The patient is monitored for graft rejection and peripheral blood cell count.
- a 50 ⁇ g/day dose of PTHrP or analogue thereof is given. It should be understood that this is merely an exemplary dose and the dose may be varied to be more or less than that amount. In addition, while it may be convenient to maintain the same dose throughout the regimen, it may be desirable to increase or decrease the dose on any given day.
- the above exemplary regimens teach administration of the PTHrP or analogue thereof as a prophylactic treatment for seven days prior to the cytotoxic therapy. This seven day period is merely an exemplary treatment period and those skilled in the art will understand that the therapy may be administered for longer (e.g., 14 days or more) or shorter time periods.
- the PTHrP or analogue thereof is given on consecutive days, the therapy could, if desired by given intermittently (e.g., every alternate day.)
- the PTHrP or PTHrP analogue administration may be initiated at the same time as the cytotoxic agent therapy and need not precede the cytotoxic agent-based therapy.
- the PTHrP or analogue thereof may be administered in combination with one or more other hematopoeitic factors such as Erythropoietin, G-CSF (filgrastim), peg-G-CSF (pegfilarastim), GM-CSF (molgramostim or sargramostim), M-CSF, TCGF, flt-3, thrombopoietin, thymic stromallymphopoietin, keratinocyte growth factor, PDGF, IGF, IL 1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-IO, IL-11, IL- 12, IL-13; IL-15, IL-16, IL-17, and stem cell factor (also known as c-kit ligand).
- a Phase I/II trial design as using separatide as an adjunct hematoprotective agent with
- Neulasta ® (pegfilgrastim) is as follows:
- Any "currently active” second malignancy other than non- melanoma skin cancer. Patients are not considered to have a "currently active” malignancy, if they have completed therapy and are considered by their physician to be at least less than 30% risk of relapse over next 3 months.
- Enrollment duration is ⁇ 3 months. Treatment with semparatide begins 7 days prior to each chemotherapy cycle.
- PHASE II Efficacy
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Endocrinology (AREA)
- Pharmacology & Pharmacy (AREA)
- Epidemiology (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Immunology (AREA)
- Medicinal Chemistry (AREA)
- Zoology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Gastroenterology & Hepatology (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
The present invention is directed to methods and compositions of therapy. More particularly, the methods and compositions disclosed herein form an adjunct to chemotherapy, radiation therapy and other therapies that cause myelosuppression. The methods of the invention use PTHrP or analogs thereof to prevent, reduce, abrogate or otherwise reverse radiation-induced or drug-induced or antibody-induced cytotoxicity, and are especially applicable to protection of hematopoietic cells against radiation and/or chemotherapy-induced cytotoxicity.
Description
METHODS AND COMPOSITIONS FOR PROTECTING AGAINST CYTOTOXIC THERAPY
BACKGROUND
Field of the Invention The present invention is directed to methods and compositions for improved therapeutic methods that use cytotoxic agents. More specifically, the invention is directed to the use of PTHrP or analogs thereof to prevent, reduce, abrogate or otherwise reverse radiation-induced or drug-induced or antibody-induced cytotoxicity, especially to hematopoietic cells. Background of the Related Art
Many different tumors are treated with combination chemotherapy or radiation therapy. However, the efficacy of these therapies is greatly hindered by the fact that these therapies have a cytotoxic effect on the bone marrow and the hematopoietic cells of the body. In almost all instances, there is a dramatic reduction in circulating red and white cells and platelets. These reductions restrict the dose and limit the potential benefit of these therapeutic regimens. Further, the risk of infection is directly related to the severity of myelosuppression induced by the cytoxic treatments. Despite the use of broad spectrum antibiotics, sepsis is not infrequent and the morbidity associated with nonfatal infection is substantial. Reductions in circulating levels of red blood cells also results in dramatic fatigue and anorexia which can further complicate recovery. The severity of myelosuppression also limits the dose and duration of exposure to chemotherapy or radiation therapy which may compromise the effectiveness of the therapy's tumor killing capability.
Following completion of a chemotherapy or radiation therapy regimen, changes in the patients blood cell populations are routinely monitored to begin treatment with stimulators of red and white blood cell proliferation. During treatment and follow-up, monitoring the patient's blood cell count is important (e.g., prior to each treatment cycle and 10-14 days after each treatment cycle).
During the examination of a patient following chemo- or radiation therapy, a complete blood cell count and other blood chemistries are tests that are routinely performed. Patients that exhibit symptomatic anemia may be immediately transfused with packed red blood cells and platelets. Cytopenias (anemia, neutropenia and thrombocytopenia) are clearly of great concern post-therapy. Hematopoeitic growth factors such as G-CSF, GM-CSF and erythropoietin are given immediately following chemotherapy in an effort to protect the
patients from opportunistic infections, sepsis and associated morbidity and mortality. Growth factor support is provided as needed.
In order to effect hematopoietic reconstitution early-term and long-term, it is important to administer G-CSF, GM-CSF, or PEGylated G-CSF as soon as possible post- radiation or chemotherapy exposure. Nevertheless, the effect of supportive care and CSF therapy on long-term immune reconstitution is unknown in the context of severely irradiated animals. There is, however, a substantial clinical database showing that the multicycle, myelosuppressive chemotherapy or myeloablative conditioning prior to stem cell transplant may have deleterious effects on immune recovery. Neutropenia is common in patients with cancer who receive myelosuppressive chemotherapy and contributes to morbidity associated with cancer. Neutropenia is associated with increased risk of infections which can be life threatening and require aggressive treatment with intravenously administered antibiotics. Such antibiotic therapy necessitates hospitalization and may be associated with other complications. Active management of neutropenia is thus of substantial importance in patients undergoing cytotoxic chemotherapy for the treatment of neoplastic disease.
Many studies have shown that recombinant G-CSF increases WBC counts and decreases the duration of neutropenia, days of hospitalization and number of infections. This therapeutic intervention also results in a decrease in chemotherapy delays, dose reductions and more patients receiving the full regimen of treatment. However, treatment of patients with filigrastim prior to chemotherapy resulted in a dramatic reduction in the marrow repopulation capacity due to the large number of mitotically activate cells within the marrow that are sensitive to chemotherapy. This resulted in a dramatically reduced overall regenerative capacity of the marrow post chemotherapy. In addition, it has been previously reported that repeated cycles of therapy with G-CSF following chemotherapy as a restorative regimen after cyclic chemotherapy may seriously reduce marrow repopulating ability or stem cell capacity of the patient. Excessive stimulation of primitive stem cell proliferation by cytokines may lead to a loss of primate stem cells and premature bone marrow failure. Studies in mice have shown that administration of G-CSF to speed recovery from repeated doses of cyclophosphamide damages primitive stem cells. It is assumed that G-CSF induced damage of primitive stem cells after multiple doses of cytotoxic agents is the increased proliferation of stem cells in response to G-CSF at the expense of self-renewal. Marrow that is already compromised appears to be most damaged by multiple exposures to G-CSF.
To date, there is no effective treatment for the prolonged T cell deficiencies associated with cytotoxic therapy. There is a significant delay in regeneration of CD4+ T cells which leads to an imbalance in the ratio of CD4/CD8 cell, this, along with a limited T-cell receptor repertoire leave the patient at risk for opportunistic infections. Long term immune reconstitution requires regeneration of naϊve, thymic-dependent T-cell. Several cytokines including IL-2, IL-4, IL-7, IL- 17, c-kit ligand, flt-3, thymic stromal lymphopoietin, and keratinocyte growth factor are associated with T-cell differentiation, proliferation, and enhanced thymopoiesis and functional recovery of peripheral T cells. Exemplary hematopoietic factors that could be employed for hematopoietic reconstitution and for enhancing recovery of thymopoiesis and immune reconstitution include but are not limited to Erythropoietin, G-CSF, GM-CSF, M-CSF, TCGF, flt-3, thrombopoietin, thymic stromal lymphopoietin, keratinocyte growth factor, PDGF, IGF, IL 1, IL-2, IL-3, IL-4, IL-5, IL-6, IL- 7, IL-8, IL-9, IL-10, IL-1 1, IL- 12, IL-13; IL-15, IL- 16, IL- 17, stem cell factor (also known as c-kit ligand), and M-CSF.
Despite the use of cytokines as a post-irradiation or post-chemotherapy reconstitution of immune cells, however, there is a continued need for further methods of preventing, mitigating or reversing toxicity to myeloid and hematopoietic cells, which is a limiting side effect of treatment of various diseases in subjects with cytotoxic agents. SUMMARY OF THE INVENTION
The present invention is directed to methods and compositions of therapy that can be used as an adjunct to chemotherapy, radiation therapy and other therapies that cause myelosuppression. The methods of the invention use PTHrP or analogues thereof to prevent, reduce, abrogate or otherwise reverse radiation-induced or drug-induced or antibody-induced cytotoxicity, and are especially applicable to protection of hematopoietic cells against radiation and/or chemotherapy-induced cytotoxicity.
In certain preferred aspects of the invention there are described methods of decreasing chemotherapy and/or radiation therapy-induced cytopenia in a subject comprising administering to the subject a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof, wherein the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered in an amount effective to decrease cytopenia (including but not limited to anemia, neutropenia and thrombocytopenia) in the subject as compared to the
cytopenia induced in the absence of the administration.
The administration of the composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof may be carried out prior to, after or subsequent to the chemotherapy and/or radiation therapy. In particular embodiments, it is contemplated that the administration of the composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is carried out prior to the chemotherapy and/or radiation therapy. For example, the composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered to the subject between 4-7 days prior to initiation of cytotoxic chemotherapy and/or radiation therapy. Thus, the PTHrP, a PTHrP fragment, a PTHrP analogue, or a derivative thereof is advantageously employed as a prophylactic composition.
The methods of the invention also may be used to decrease myelosuppression in a subject comprising administering to the subject a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof. In specific embodiments, the myelosuppression is produced by chemotherapy or radiation therapy or immunosuppressive therapy received during organ transplantation. In such methods, the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is preferably administered prior to chemotherapy or the radiation therapy.
Also contemplated herein are methods of decreasing the dose or duration of chemotherapy or radiation therapy to be administered to a subject in need thereof comprising administering to the subject a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof.
Another embodiment of the invention contemplates a method of preventing or decreasing the level of reduction in circulating blood cells in a subject receiving radiation therapy and/or chemotherapy, comprising administering to the subject a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof prior to administration of the radiation therapy and/or chemotherapy, wherein administration of the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof prevents or produces a lower level of reduction of circulating blood cells in the subject as compared to a subject that does not receive the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof.
The methods of the invention also may be used to treat a subject that is at risk
of having a decrease in the number of cells of the hematopoietic system, comprising the step of administration to the subject an effective amount of PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof, wherein the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered before, after, or during the chemotherapy and/or radiation therapy. For example, the cells the hematopoietic system may be selected from the group consisting of, including but not limited to T cells, B cells, granulocytes, macrophages, mesenchymal cells, osteoblasts, osteoclasts and multipotent adult progenitor cells, or combinations thereof. In specific embodiments, the decrease in the number of cells of the hematopoietic system is associated with chemo- and/or radiotherapy and/or removal of blood progenitor cells. For example, this may occur as a result of the chemotherapy and/or radiotherapy and/or removal of blood progenitor cells being administered to treat cancer.
Also contemplated is a method for protecting hematopoeitic cells from cell death in response to a chemotherapeutic and/or radiation comprising contacting the cells with a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof prior to contacting the cells with the chemotherapeutic agent and/or radiation.
The invention further is directed to a method for treating a cytotoxic agent-induced hematopoietic or myeloid toxicity in a human patient which comprises administering prior to, simultaneous with or subsequent to the administration of the cytotoxic agent, a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof in an amount effective to prevent, mitigate or reverse such hematopoietic or myeloid toxicity. In such methods, the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof may, but need not necessarily be administered in combination with one or more hematopoietic factors selected from the group consisting of Erythropoietin, G-CSF, GM-CSF, M-CSF, TCGF, flt-3, thrombopoietin, thymic stromal lymphopoietin, keratinocyte growth factor, PDGF, IGF, IL 1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-IO, IL-11, IL-12, IL-13; IL-15, IL-16, IL-17, stem cell factor (c-kit ligand), and M-CSF (including PEGylated derivatives and biologically fragments thereof) is administered in an amount effective to prevent, mitigate or reverse hematopoietic or myeloid toxicity.
It should be noted that the one or more hematopoietic factors administered in the combination adjunct therapy immediately above may be administered prior to, concurrently with or subsequent to the administration of PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof.
Also provided is a method of treating cancer therapy in a mammalian patient which comprises administering to the patient a combination of a cytotoxic agent in an amount effective to treat the cancer and PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof in an amount effective to prevent, mitigate or reverse hematopoietic or myeloid toxicity, wherein the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered prior to, simultaneously with or subsequent to the administration of the cytotoxic agent, wherein administration of the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof reduces the dose of the cytotoxic agent needed to produce a therapeutic effect in the patient. In some embodiments, the cytotoxic agent is a therapeutic agent, including but not limited to radiation therapy or chemotherapy.
Also taught herein is a method treating cancer therapy in a mammalian patient which comprises administering to the patient a combination of a cytotoxic agent in an amount effective to treat the cancer and a combination of PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof and at least one hematopoietic growth factor in an amount effective to prevent, mitigate or reverse hematopoietic or myeloid toxicity, wherein the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered prior to the administration of the cytotoxic agent.
The methods of treating cancer may be used in a mammalian patient is suffering from a cancer selected from the group consisting of Hodgkins lymphoma, NonHodgkins lymphoma, pancreatic cancer, melanoma, breast cancer, prostate cancer, bladder cancer, ovarian cancer, endometrial cancer, lung cancer, Kaposi's sarcoma, leukemia, lymphoma, gastric cancer, colon cancer, colorectal cancer, esophageal cancer, renal cancer, and head and neck cancer. In the methods of treating cancer, the hematopoietic growth factor may be selected from the group consisting of Erythropoietin, G-CSF, GM-CSF, M-CSF, TCGF, flt-3, thrombopoietin, thymic stromal lymphopoietin, keratinocyte growth factor, PDGF, IGF, IL 1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-IO, IL-I l, IL-12, IL-13; IL- 15, IL- 16, IL- 17, stem cell factor (c-kit ligand), and M-CSF (including PEGylated derivatives and biologically active fragments thereof).
Also taught herein is a method for preventing or mitigating myelosuppression in a human patient undergoing therapy for cancer or tissue or organ transplantation, which comprises administering to the patient a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof in an amount effective to treat myeloid toxicity, wherein the cytokine is administered prior to, simultaneously with or subsequent to the
therapy.
The invention may be used as a method for preventing or mitigating hematopoietic cell depression in a human patient undergoing therapy for cancer or tissue or organ transplantation which comprises administering to the patient a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof in an amount effective to treat myeloid toxicity, wherein the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered prior to, simultaneously with or subsequent to the administration of the therapy.
Further aspects of the invention contemplate a method for treating a cytotoxic agent-induced hematopoietic or myeloid toxicity in a human patient which comprises administering prior to, simultaneous with or subsequent to the administration of the therapeutic agent, PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof and a hematopoietic factor in an amount effective to prevent, mitigate or reverse such hematopoietic ar myeloid toxicity. For example, such a cytotoxic agent may include a radionuclide, including but not limited to Iodine- 131, Strontiun-89, Yttrium-90, Rhenium 186, Rheniuml88, Cobalt 60; Cesium 137; Iridium 192, and Radium 226. In some embodiments, the cytotoxic agent is conjugated to an antibody or a bone-seeking chemical. Exemplary bone-seeking chemicals include orthophosphate or diphosphonate.
Also provided herein is a method of alleviating a physical symptom caused by hematopoietic or myeloid toxicity in a human patient undergoing therapy with a cytotoxic agent, wherein the patient is administered prior to, simultaneous with or subsequent to the administration of the cytotoxic agent, a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof in an amount effective to prevent, mitigate or reverse such hematopoietic or myeloid toxicity. For example, the physical symptom may be bone pain associated with bone cancer or bone metastasis.
In the methods of the present invention, an exemplary patient is a cancer patient. For example, the subject has a cancer selected from the group consisting of Hodgkins lymphoma, Non-Hodgkins lymphoma, pancreatic cancer, melanoma, breast cancer, prostate cancer, bladder cancer, ovarian cancer, endometrial cancer, lung cancer, Kaposi's sarcoma, leukemia, lymphoma, gastric cancer, colon cancer, colorectal cancer, esophageal cancer, renal cancer, and head and neck cancer. In some embodiments, the patient is a patient receiving an organ transplant and receiving immunosuppressive therapy to kill T-cells.
The PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof may be administered through any typical route of administration including, but not limited to administration via subcutaneous, intravenous, intradermal, intraarterial, intramusclar, intramammary, intraperitoneal, intrathecal, intraocular, retrobulbar, intrapulmonary (e.g., term release), aerosol, sublingual, nasal, anal, vaginal, oral, or transdermal delivery, or by surgical implantation, or a combination thereof.
In some embodiments, the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered in a single dose. In other embodiments, the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered in multiple doses or as a continuous infusion during therapy. In specific aspects of the invention, the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered in a dose of about 0.5 μg/kg body weight to about lOμg /kg body weight of subject/day.
The PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof may be administered on each of 2, 3, 4, 5, 6 or 7 days prior to the administration of chemotherapy or radiation therapy. As noted herein throughout, the methods contemplate that the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof may be administered in combination with one or more hematopoietic factors. The hematopoietic factor may be selected from the group consisting of Erythropoietin, G-CSF, GM-CSF, M-CSF, TCGF, flt-3, thrombopoietin, thymic stromal lymphopoietin, keratinocyte growth factor, PDGF, IGF, IL 1, IL-2, 1L-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-IO, IL-11, IL-I2, IL-13; IL-15, IL-16, IL-17, stem cell factor (c-kit ligand), and M-CSF or combinations thereof. In addition the hematopoietic factor may be a biologically active fragment of such a factor. The hematopoietic factor or fragment thereof also may be PEGylated.
In certain specific embodiments, the composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof comprises PTHrP 1-34, or an analogue thereof. An exemplary PTHrP 1-34 analogue is semparatide.
Other features and advantages of the present invention will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples, while indicating preferred embodiments of the invention, are given by way of illustration only, because various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description.
DESCRIPTION OF DRAWINGS
Figures 1, 2 and 3 depict the hematoprotective effect of Semparatide administered to mice for 14 days prior to administration of a high dose of 5-fluorouricil, showing higher levels and faster recovery post chemotherapy of circulating WBCs, lymphocytes and neutrophils respectively.
DETAILED DESCRIPTION OF INVENTION
There is a need to establish methods and compositions for use in subjects undergoing chemotherapy and/or radiation therapy in order to protect, prevent, mitigate, reduce, abrogate, or otherwise reverse cytopenia (e.g., anemia, neutropenia and thrombocytopenia) produced by the chemotherapy and/or radiation treatment, particularly a need for strategies to increase stem cell populations prior to chemotherapy and/or radiation therapy that do not result in a chemotherapy-induced and/or radiation therapy-induced reduction in overall stem cell and WBC regenerative capacity.
Administration of PTHrP, prior to or immediately after a chemotherapeutic insult will decrease the reduction in the population of hematopoietic cells and thus decrease the cytoreduction that is normally observed after such ablative therapies. By decreasing the period of vulnerability to opportunistic infections post therapy, patients can return to normal life sooner, spending less time in an intensive care unit waiting for more conventional therapies to increase the population of white and red cells and platelets. The hematopoietic stem cell niche is intertwined with osteoblasts and bone stroma.
Bone marrow stroma and local osteoblasts permit hematopoiesis by providing environmental cues, physical connections and the appropriate growth factor support. Osteoblasts and stromal cells are able to support hematopoietic progenitors and allow them to expand. The interaction between these cells has been called a niche. Without intending to be bound by theory, it is believed that stimuli that increase the number of osteoblasts, increase the availability of hematopoietic stem cell niches, which would increase the number of hematopoietic stem cells available for release into the systemic circulation following stimulation (injection of G-CSF or administration of chemotherapy). More recently, it has been shown that osteoblasts secrete cytokines such as G-CSF, GM-CSF, ILl, IL6 and LIF all of which have been shown to modulate hematopoiesis. Indeed, the intersection of a number of recent observations suggests that a physical "harbor" is created by bone marrow stroma and osteoblasts which provides a niche for hematopoietic stem cells to attach and proliferate. These cells are held in place by
cell to cell adhesion. When activated by G-CSF or other stimuli such as chemotherapy, proteases (specifically MMP-I) are activated and free the blood cell so that it may migrate to the vascular network within the marrow and move to the peripheral vascular system.
Activation of osteoblasts within the stem cell niche of bone marrow allows for the proliferation and migration of stem cells to that niche. Upon entry into the niche, these cells are tethered and following this physical attachment and through the activation of membrane protein ligand and receptor, Notch and Jagged, remain in the G0 resting state and are thus not sensitive to the cytotoxic effects of chemotherapy. Upon activation of the stem cell niche, either by the injection of G-CSF or administration of chemotherapy, many more cells are available to enter into the differentiation pathway. Thus, greater numbers of mature WBCs and neutrophils are released into the peripheral circulation, thus decreasing the duration and extent of neutropenia. In addition, due to the more physiological method of leukocytosis, no depletion of marrow regenerative capacity is experienced. PTHrP, e.g. Semparatide, is believed to increase the hematopoietic pool size by a number of mechanisms:
• strong stimulation of the growth of trabecular bone which enlarges the hematopoietic stem cell niche
• promote the attachment of stem cells by elaborating growth factors and "mooring lines" for cellular attachment • directly stimulate stem cells to differentiate by the elaboration of growth factors
• induce the expression of ligands and receptors (such as Jagged and Notch) which allow for intercellular communication and survival of hematopoietic stem cell differentiation.
The therapeutic methods of the invention are useful for reconstitution of levels of all blood cell types after such cytotoxic therapy. In the present invention, it is shown that PTHrP, and fragments, and analogs thereof, may be used to avoid the cytopenia observed in patients following chemo- or radiation therapy. Due to the unique property of this peptide to stimulate an intracellular process in hematopoietic progenitor cells that provides them with the capacity to avoid programmed cell death (apoptosis), in certain specific embodiments, it is proposed that the subject may be pretreated with the PTHrP or analog thereof prior to the initiation of the cytotoxic chemotherapy or radiation therapy in order to avoid the myelosuppression and associated cytopenias that limit the dose and duration of chemotherapy treatment.
Prophylactic treatment with PTHrP will also be useful in that the period of time during which the patient is at risk for developing opportunistic infections and sepsis can be greatly reduced. The use of PTHrP or analogs thereof as described herein will also advantageously avoid the need for transfusions of RBC and platelets. It is envisioned that this prophylactic therapy would also avoid the need for expensive treatments with blood cell growth factors such as erythropoietin and G-CSF. However, it is contemplated that in some circumstances it may be desirable to combine the treatment with PTHrP or analog thereof, with treatments that use blood cell growth factors in order to achieve a more effective recovery than that achieved in the absence of application of the PTHrP.
Thus, in preferred aspects of the invention, the use of PTHrP, fragments or analogs thereof is used to improve the therapeutic efficacy of anticancer, antimicrobial and autoimmune disease, and anti-organ rejection therapy in that the PTHrP, fragments, or analog thereof is used to prevent, mitigate or reverse adverse radiation-induced or drug-induced toxicity, especially to hematopoietic cells. Use of the PTHrP, fragment, or analog thereof will allow the subject to tolerate higher doses of cytotoxic agents that are administered to the subject for therapeutic purposes.
The PTHrP, fragment, or analog thereof may also increase the duration of time during which the cytotoxic agents can be administered and the dose-limiting hematopoietic cell toxicity that is characteristic of such cytotoxic agents can be prevented, palliated or reversed using adjunct therapy with PTHrP.
The present invention is directed to methods of treatment in which PTHrP is administered to a subject that has or is at risk of developing cytopenia. Typically, the subject is one in which the cytopenia will be induced as a result of cytotoxic agents such as chemotherapy, radiotherapy, antimicrobial agents and the like. The invention contemplates methods of treating such subjects in which PTHrP or an analog thereof or a combination of PTHrP and an analog thereof is administered to prevent, mitigate, reduce, abrogate, reduce or otherwise reverse radiation-induced or drug-induced toxicity of normal cells, and more particularly, hematopoietic cells.
Throughout the specification and the claims herein the term "PTHrP" is used. It should be understood that these methods may use natural wild-type PTHrP, a natural variant of PTHrP, an analog of PTHrP or a derivative of PTHrP that has a structure based on PTHrP but has been modified to contain a moiety that increases the uptake or distribution characteristics of the PTHrP or otherwise improve the therapeutic properties of the peptide. PTHrP and its various permutations that may be used in the present invention are described in further detail below, however, it should be understood that any of the various forms of PTHrP, its analogs or its derivatives may be used in the present methods.
In essence, certain embodiments of the invention are directed to methods that provide an adjunct therapy in which PTHrP (including its analogs and derivatives) is used as the adjunct that allows the administration of higher doses of cytotoxic agents due to increased tolerance of the recipient mammal. Moreover, adjunct PTHrP therapy can prevent, palliate, or reverse dose-limiting cytotoxic effects of the cytotoxic agents on the hematopoeitic cells.
The PTHrP also has a radioprotective effect on hematopoietic cells. A great deal of therapeutic administration of radioisotopes involves administration of beta emitters, alpha emitters and/or generation of radioisotope in situ by neutron activation of Boron-10 atoms (resulting in alpha emission from the unstable nuclide produced by neutron absorption.) The present invention contemplates administration of PTHrP (or analog or derivative thereof) to the subject either prior to, during or after application of the radiotherapy.
Prior to the present invention, it could not be predicted with accuracy that drug- or toxin-induced hematopoietic or myeloid toxicity would respond to PTHrP treatment in humans. However, given the teachings of the present invention, it is now possible to administer not only PTHrP, but also related molecules, such as PTHrP analogs or derivatives in order to achieve a radioprotective or chemoprotective effect on hematopoietic cells. The
administration of the PTHrP or related molecule may be prior to the administration to the chemotherapy, radiotherapy or other drug or toxin-based therapy, and as such, can be a prophylactic administration of the agent. Alternatively, the PTHrP or related molecule may be administered simultaneously or subsequent to the chemotherapy, radiotherapy or other drug or toxin-based therapy to mitigate or reverse myeloid or hematopoietic toxicity.
It should be understood that the methods of the invention may be particularly useful as adjuncts to conventional anti-cancer therapies in which the subject is administered one or more tumoricidal agent, e.g., a drug and a radioisotope, or a radioisotope and a Boron- 10 agent for neutron-activated therapy, or a drug and a biological response modifier, or an antibody conjugate and a biological response modifier. The PTHrP can be integrated into such anti-cancer therapeutic regimens to maximize the efficacy of each component thereof.
In some examples, it has been shown that certain anti-leukemic and anti-lymphoma antibodies conjugated with beta or alpha emitting radioisotopes can induce myeloid and other hematopoietic side effects when these agents are not solely directed to the tumor cells, particularly when the tumor cells are in the circulation and in the blood-forming organs. The methods of the invention contemplate administration of the PTHrP or related molecule prior to, and/or concomitantly with and/or subsequent to the administration of such antibody-based therapies to reduce or ameliorate the hematopoietic side effects of the anticancer agent, while allowing the beneficial anticancer effect of the agent. Thus, depending on the specific condition being treated, an appropriate dose of the
PTHrP or analog or derivative can be administered prior to, simultaneously with or subsequent to the administration of the therapeutic agent. It is desirable to maximize the cytotoxic activity of the therapeutic agent on the pathological lesion, such as cancer cells or infectious organisms, while minimizing toxicity of that agent to the myeloid and other hematopoietic cells. In some circumstances, the PTHrP adjunct therapy may be administered continuously with the cytotoxic therapeutic agent in order to achieve the most beneficial hematoprotective effects of the PTHrP.
As noted above, numerous anticancer therapies produce a deleterious myelosuppressive effect. Typical anti-cancer agents include biological agents (biotherapy), chemotherapy agents, and radiotherapy agents. Generally, these compositions are provided in a combined amount effective to kill or inhibit proliferation of the cancer cell, for example, by killing cancer cells, inducing apoptosis in cancer cells, reducing the growth rate of cancer
cells, reducing the incidence or number of metastases, reducing tumor size, inhibiting tumor growth, reducing the blood supply to a tumor or cancer cells, promoting an immune response against cancer cells or a tumor, preventing or inhibiting the progression of cancer. In doing so, these therapies typically also have an effect on normal, non cancer cells. The present invention is particularly concerned with the deleterious effects of such therapies on normal hematopoeitic cells. The cancer cells are generally contacted with the anticancer agent and at the same time, the hematopoietic cells also come into contact with such agents.
Myeloid cell death in response to chemotherapy and radiotherapy agents represents a major problem in clinical oncology. One goal of current cancer research is to find ways to improve the efficacy of chemo- and radiotherapy by combining it with agents that are myeloprotective or are able to reconsistitute myeloid cells. For example, cytokines are typically administered after the chemotherapy and or radiotherapy to facilitate myeloid reconstitution. In the context of the present invention, it is contemplated that PTHrP and related compositions could be used in conjunction with chemotherapeutic, radiotherapeutic, immunotherapeutic or other biological intervention, in order to act as myeloprotectant and to enhance the growth of hematopoeitic cells.
PTHrP may be administered prior to, after or during the administration of the other therapeutic agent intervals ranging from minutes to weeks. In embodiments where the other agent and the PTHrP are applied separately to the cell, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the agent and expression construct would still be able to exert an advantageously combined effect on the cell. In such instances, it is contemplated that one may contact the cell with both modalities within about 12-24 h of each other and, more preferably, within about 6-12 h of each other. In some situations, it may be desirable to extend the time period for treatment significantly, however, where several days (2, 3, 4, 5, 6 or 7) to several weeks (1, 2, 3, 4, 5, 6, 7 or 8) lapse between the respective administrations.
The hematoprotection afforded by the PTHrP is protection against any conventional chemotherapy, including use of for example, alkylating agents (cyclophosphamide), anti metabolites (azothioprine), plant alkaloids and terpinoids (vinblastine, etoposide, and paclitaxel), topoisomerase inhibitors (topotecan) as well as hormone therapy (antiestrogen: tamoxifen; antiandrogens: bicalutamide). Thus the PTHrP compositions may be used in order to protect the subject from the myelosuppressive effects of exemplary agents that include but are not limited to cisplatin (CDDP), carboplatin, procarbazine, mechlorethamine,
cyclophosphamide, camptothecin, ifosfamide, melphalan, chlorambucil, busulfan, nitrosurea, dactinomycin, daunorubicin, doxorubicin, bleomycin, plicomycin, mitomycin, etoposide (VP 16), tamoxifen, raloxifene, estrogen receptor binding agents, taxol, gemcitabien, navelbine, farnesyl-protein transferase inhibitors, transplatinum, 5-fluorouracil, vincristine, vinblastine and methotrexate, Temazolomide (an aqueous form of DTIC), or any analog or derivative variant of the foregoing. Those skilled in the art will understand that these are merely exemplary chemotherapeutic agents and that the PTHrP-based methods of the invention can be used to protect against the myelosuppressive effects of other such chemotherapy agents. The hematoprotection derived from PTHrP administration can also be used to protect hematopoietic cells from other factors that cause DNA damage and have been used extensively include what are commonly known as γ-rays, X-rays, and/or the directed delivery of radioisotopes to tumor cells. Such radioisotopes include, but are not limited to Iodine131, Strontium89, Yttrium90, Rhenium186, Rhenium188, Cobalt60; Cesium137; Iridium192, and Radium226. Other forms of DNA damaging factors are also contemplated such as microwaves and UV-irradiation. It is most likely that all of these factors effect a broad range of damage on DNA, on the precursors of DNA, on the replication and repair of DNA, and on the assembly and maintenance of chromosomes. Dosage ranges for X-rays range from daily doses of 50 to 200 roentgens for prolonged periods of time (3 to 4 wk), to single doses of 2000 to 6000 roentgens. Dosage ranges for radioisotopes vary widely, and depend on the half-life of the isotope, the strength and type of radiation emitted, and the uptake by the neoplastic cells.
The terms "contacted" and "exposed," when applied to a cell, are used herein to describe the process by which a therapeutic agent (e.g., the PTHrP or the cytokine or an expression construct that encodes such an agent) and a chemotherapeutic or radiotherapeutic agent are delivered to a target cell or are placed in direct juxtaposition with the target cell. To achieve effective therapy, the PTHrP and other hematoprotectant is delivered to a cell in a amount effective to protect the cell from cell death in response to chemo- or radiotherapy.
Throughout the treatment protocols, it may be useful to monitor the white blood cell (WBC) count, Red blood cell (RBC) count, platelets and other blood elements in the subject. Thus, it is contemplated that as part of the treatment regimen the subject's blood sample will be monitored for components, including but not limited to erythrocyte (red blood cell/RBC) count, thrombocyte (platelet) count, and including a differential WBC analysis to monitor the
myloid/lymphoid series, as well as the bone marrow hematological picture during the course of therapy. It will be particularly desirable to assess the subject for possible depletion of myeloid lymphoid forms, but also the status of immature erythrocytes, myelocytes, lymphocytes and thrombocytes. Monitoring these parameters will permit optimization of the PTHrP adjunct treatment with the cytotoxic agent therapy. Depending upon which hematologic element is adversely affected, the amount and duration of the PTHrP and cytotoxic agent can be tailored to each circumstance.
Correlation of the choice of the amount of PTHrP and its combination with another hematoprotectant (such as a cytokine as discussed below), and doses thereof, to hematotoxicity is important, since each therapeutic regimen involving a cytokine generally will have its effect mostly on particular hematopoietic elements. For example, if a cytotoxic agent has both severe myeloid and thrombocytic toxicity, the combination of the PTHrP with IL-I and IL-3 in a composition in which the IL-I and IL-3 are present at a 1:1 or 2:1 (or higher) ratio will be advantageous. Thus, reduction in the WBC count to a level below about 2,000 and platelets to a level below about 20,000 can be reversed by administration of
PTHrP. The administration of the PTHrP can be repeated, with the reversal of the myeloid and platelet depressions occurring within about 5-20 days after the administration of the PTHrP, usually within about 7 days. The ordinary skilled clinician will appreciate that variations in the timing and dosage of PTHrP administration and combinations of the PTHrP with for example cytokines and dosages of these agents are a function of the agent used, the nature of the bone marrow and/or other hematopoietic element depressed, and the nature of the patient (e.g., prior toxicity affecting bone marrow status) and the cytotoxic agent and protocol.
In other embodiments of the invention PTHrP compositions can be administered in order to treat bone pain in patients with bone metastases and primary bone cancers. In such cases, radionuclide therapy has been found to be effective and safe, particularly with the introduction of Sr-89, Y-90 and Re-186 or Re-188, either alone or conjugated to an antibody or a bone-seeking chemical such as orthophosphate or diphosphonate. Chemotherapeutic agents, e.g., 5-fluorouracil (5-FU), have also been known to control bone pain in patients with metastatic carcinoma. P-32-ortho-phosphate can be administered in several ways, including single doses of about 3 to 10 mCi, multiple consecutive doses of about 1.5 mCi, or multiple intermittent doses of 7 to 10 mCi as clinically required. In multiple and intermittent dose schedules, total doses can range from 5 to 20 mCi, depending on patient response and
side effects. In order to reduce the myelosuppression of this treatment and increase the effects against bone pain and possibly also inhibit tumor growth, these doses can be increased by from about 10% to about 35%, preferably 15 to 25%, by simultaneous administration of continuous or intermittent doses of about 5 to 20 μg of IL-I, more preferably single repeated dose of about 0.5 μg PTHrP/kg body weight to about lOμg PTHrP/kg body weight of subject/day PTHrP (including full-length PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof) either alone or in combination with a cytokine, thereby extending the time over which radionuclide therapy can be tolerated by the patient. Similarly, Re-186- diphosphonates can be used for bone pain palliation in single doses of about 5 to 10 mCi, repeated up to three times, in combination with administration of the PTHrP either prior to and/or, simultaneously and/or post-therapy with Re-186-diphosphonates. Again such administration of the PTHrP may be in combination with a cytokine which can be administered prior to and/or simultaneously and/or post-administration of the diphosphonates and/or the PTHrP. The therapy with the composition comprising the PTHrP and/or the composition comprising the cytokine can be repeated several times during a 1-2 week therapy regimen.
1-131 is another effective radioisotope that has been used for the treatment of cancer. It has been shown to be especially useful for treating primary and metastatic, well- differentiated thyroid carcinomas. A dose of 150 mCi 1-131 has been successful, with most clinicians administering a dose between 100 and 200 mCi. Again however, bone marrow depression is a major complication of this therapy and severely limits the dosages of the I- 131 that can be tolerated by the subject. The present invention contemplates combining 1-131 therapy, using a dose of 150-250 mCi, with PTHrP. Again the PTHrP may be administered alone or in combination with a cytokine. In exemplary embodiments, PTHrP is administered 4-7 days prior to administration of the 1-131 and produces a marked decrease in the myelosuppression seen in the cancer patient in response to the 1-131 compositions. In other embodiments, the PTHrP is administered during or after the 1-131 either alone or in combination with a cytokine and again produces a beneficial decrease in myelosuppression. In some embodiments, the PTHrP-based therapy is continued to one, two, three or more weeks post-radioisotope therapy. Use of the PTHrP leads to the subject having a marked decrease in myelosuppression and also a marked increase in the tolerance of higher doeses of 1-131 doses.
If PTHrP therapy is initiated 4-7 days prior to radioisotope administration, and continued twice weekly for 2-3 weeks, doses of 1-131 between 200 and 300 mCi, preferably 200-250 mCi, could be well tolerated. This will therefore lead to an increase in the therapeutic anti-cancer dose of the 1-131 that can be administered. The PTHrP-based therapeutic methods of the invention can be combined with various methods of radionuclide therapy in order to obtain an effective treatment of cancer and other pathological conditions, as described, e.g., in Harbert, "Nuclear Medicine Therapy", New York, Thieme Medical Publishers, 1987, pp. 1-340. A physician experienced in these procedures will readily be able to adapt the PTHrP adjuvant therapy described herein to such procedures to mitigate the hematopoietic side effects of the radionuclides. Similarly, therapy with cytotoxic drugs, either administered alone or as antibody conjugates for more precisely targeted therapy, e.g., for treatment of cancer, infectious or autoimmune diseases, and for organ rejection therapy, is governed by analogous principles to radioisotope therapy with isotopes or radiolabeled antibodies. Thus, the ordinary skilled clinician will be able to adapt the description of PTHrP use to mitigate marrow suppression and other such hematopoietic side effects by administration of the PTHrP before, during and/or after drug therapy.
In addition to being used as an adjunct therapy to chemotherapy and radiation therapy, the PTHrP-based hematoprotection also can be given to subjects being treated with anticancer antibodies. Invading microorganisms and proliferating cancer cells can be targeted with antibodies that bind specifically to antigens produced by or associated therewith. Such antibodies can directly induce a cytotoxic immune response, e.g., mediated by complement, or an indirect cytotoxic immune response, e.g., through stimulation and mobilization of T- cells, e.g., killer cells (ADCC). Certain of such antibodies also produce side effects which include compromise of elements of the hematopoietic system, and such side effects can be prevented, mitigated and/or reversed by administration of PTHrP as described herein. The dose and time period of administration of the PTHrP and/or other hematoprotectant agents (e.g., cytokines as described herein below) will again be correlated to WBC, erythrocyte and platelet counts and other aspects of the status of the hematopoietic system.
The route of administration of the therapeutic agent as well as of the PTHrP should be coordinated and optimized. For example, intracavitary, e.g., intraperitoneal, administration of a radioisotope- antibody conjugate will eventually result in lowering of the blood cell count in a patient given a high dose of the cytotoxic immunoconjugate, due to eventual diffusion of the conjugate into the bloodstream and circulation through the bone marrow.
Administration of the PTHrP can be advantageously effected through any typical route of administration that will allow access to the region of greatest hematopoietic cell compromise. Such routes of administration include, subcutaneous, intravenous, intradermal, intraarterial, intramuscular, intramammary, intraperitoneal, intrathecal, intraocular, retrobulbar, intrapulmonary (e.g., term release), aerosol, sublingual, nasal, anal, vaginal, oral, or transdermal delivery, or by surgical implantation at a particular site to have its maximum effect on the circulating peripheral blood cell counts. Intranasal forms of administration of the PTHrP are of particular interest. Cytokines and other hematoprotectants can be administered through the same routes as the PTHrP. The clinician also may determine whether the PTHrP should be administered as a single dose, in multiple doses or as a continuous infusion during the course of therapy. If the PTHrP is administered concomitantly with the cytotoxic agent, it may be given by continuous intravenous infusion over several hours and optionally repeated on one or more days during and after completion of the cytotoxic therapy. Continuous administration of the PTHrP can be effected by any of the transdermal modes of drug administration known to the art or yet to be developed. Similar considerations and routes of administration also should be taken into account for administration of cytokines.
Radioisotopes typically are administered by a variety of routes for cancer therapy. These include, e.g., intravenous, intraarterial, intracavitary (including intraperitoneal), intrathecal and subcutaneous injection, as well as by implantation of seeds of radioactive material at selected sites in the patient's body. The type, extent and time frame for myeloid and other hematopoietic cell toxicity will vary for each mode of administration of the cytotoxic agent and with the type of agent itself. In general, bone marrow toxicity will be the most serious side effect of such therapeutic regimens and intravenous or intraarterial administration of the PTHrP will be the most effective preventive or palliative measure.
Anticancer and antimicrobial, e.g., antiviral, drugs, whose major side effects are hematologic toxicity also are typically administered through any of a variety of routes, including e.g., intravenous, intraarterial, intracavitary (including intraperitoneal), intrathecal and subcutaneous injection. These drugs, including toxins, can be administered systemically without conjugation to a tumor-targeting or infectious lesion-targeting antibody. Marrow toxicity is a common limiting factor in the dosage which can be administered, and the method of the invention is effective in significantly extending the dosage range and duration of therapeutic administration of such agents.
The drugs or toxins may be conjugated with antibodies to achieve better targeting of the drug to the pathological lesion in order to improve the therapeutic efficacy of the drugs or toxins. Still further improvement in the therapeutic efficacy of these agents can be achieved by administering these immunoconjugates with PTHrP as contemplated herein. By employing the PTHrP adjunct therapy, it is possible to increase the dosage of the immunoconjugates that can be administered to the subject because the PTHrP affords an increased tolerance for the higher levels of drug, toxin, or other cytotoxic agent necessary for maximal therapeutic activity.
Radioisotope antibody conjugates can be administered by, e.g., intravenous, intraarterial, intracavitary, intrathecal, intramuscular, or subcutaneous routes. Again, intravenous or intraarterial administrations of PTHrP, its derivative or analogs will normally minimize bone marrow toxicity.
In the therapeutic methods, the beta and alpha emitters are preferred for radioimmunotherapy, since the patient can be treated in multiple doses on an outpatient basis. To the extent that the treatment results in bone marrow toxicity, administration of PTHrP either alone or in combination with cytokines can be effected at convenient times by injection or even by transdermal administration of an appropriate level of PTHrP and cytokine.
Boron- 10 compounds has been contemplated as a useful anticancer treatment (e.g., systemic administration of Boron- 10-containing compounds, e.g., borates, carborane compounds and the like, followed by neutron irradiation) however, the acceptance of this therapy has been hampered by the fact that it produces excessive toxicity to normal organs. Attempts to mitigate the unacceptable toxic side effects have involved targeting the boron atoms to tumor sites by conjugating them to site-specific antibodies. The methods of the invention can be used as an adjunct to provide a radioprotective treatment against the alpha radiation of the activated boron atoms using PTHrP. Systemic administration of the PTHrP will allow administration of the boron compound either locally at the tumor site or systemically with the PTHrP being able to prevent or reverse hematopoietic toxicity of the targeted cytotoxic neutron capture therapy.
The methods of the invention also can be used in other applications in which cytotoxic agents, particularly those that affect the lymphoid system (and therein particularly the T-lymphocytes), are used to depress host immunity in certain autoimmune diseases, e.g., systemic lupus erythematosis, and in patients receiving organ transplants. Often the cytotoxic
drugs used in these disorders are similar to those often used in cancer chemotherapy, with the attendant myeloid and other hematopoietic side effects. In addition to these drugs, specific antibodies against lymphoid cells (particularly T-cells), e.g., the anti-Tac monoclonal antibody of Uchiyama et al., J. Immunol. 126:1393 and 1398 (1981), which specifically binds to the human IL-2 receptor of activated T-cells, have been conjugated to cytotoxic agents, such as drugs, toxins or radioisotopes, to effect a relatively select killing of these cells involved in organ rejection. For example, a T-cell antibody can be conjugated with a beta- or alpha-emitting radioisotope, and this can be administered to the patient prior to undertaking organ transplantation and, if needed, also thereafter. In order to effect a high T-cell killing dose without the concomitant limiting side effects to the hematopoietic system, the treatment of the T-cell killing dose of therapy can be combined with the use of PTHrP, according to the present invention. This method is useful for the long-term survival of many organ transplants, such as the kidney, heart, liver, etc., because such transplants often are accompanied by a critical period during which there is a risk of organ rejection. The PTHrP therapy can serve as an adjunct therapy for organ transplants.
The dosage level of the PTHrP will be a function of the extent of compromise of the hematopoietic cells, correlated generally with the peripheral blood cell count in the patient. Periodic monitoring of the WBC, RBC, platelets and other blood cell counts and adjustment of the rate and frequency of infusion or the dosage of the PTHrP administered to achieve a relatively constant level of peripheral blood cell count will ensure that the patient does not sustain undue marrow toxicity from the therapy. Experience will permit anticipation of when the treatment will lower the levels of the circulating blood components and infusion of the PTHrP at a time and in an amount sufficient to substantially prevent depression to the components of the blood in response to the cytotoxic therapy.
The present invention includes administration PTHrP either alone or in combination with one or more cytokines, preferably lymphokines, prior to, together with and/or subsequent to administration of cytotoxic radioisotopes, drugs and/or toxins, alone or in combination, as such or in the form of antibody conjugates. The guidelines provided herein will enable the ordinary skilled clinician to adapt PTHrP administration to enhance the efficacy and mitigate the adverse hematopoietic side effects of cytotoxic therapy as a function of WBC, platelet and erythrocyte counts, marrow component status and other particular diagnostic indicia peculiar to the individual patient. In general, this invention is applicable to
support an enhancement of efficacy of any cytotoxic therapy that has serious hematopoietic side effects that limit the therapy's efficacy.
In a specific embodiment, it is contemplated that post chemo/radiation therapy, the subject may receive a bone marrow transplant. In such a method, bone marrow cells, or stem cells that have been engineered to express properties of hematopoeitic stem cells are transplanted into the individual. The cells are such that they have been maintained and expanded outside of the patient. The cells may be heterologous (from a donor e.g., a relative) or they may be autologous (from the subject itself). The cells also may be banked peripheral hematopoietic stem cells that are reintroduced into the patient post chemo/radiation therapy. In autologous bone marrow transplant (ABMT) the patient will serve as his/her own bone marrow donor. Thus, a normally lethal dose of irradiation or chemotherapeutic may be delivered to the patient to kill tumor cells, and the bone marrow repopulated with the patients own cells that have been maintained (and perhaps expanded) ex vivo. Because, bone marrow often is contaminated with tumor cells, it is desirable to purge the bone marrow of these cells, thus the cells, once harvested may be irradiated to kill the cancer cells therein and then expanded in culture ex vivo to produce an expanded population of bone marrow cells for administration to the subject. It is contemplated that in such bone marrow transplants, the PTHrP (full length PTHrP, or related PTHrP polypeptide fragment, analog, or derivative thereof) may be used to enhance engraftment/survival of bone marrow transplants and decreases the period of cytopenia and window of opportunity for opportunistic infections that are prevalent post chemo/radiation.
The components and compositions for the above methods are described in further detail below.
A. PTHrP, Analogs and Derivatives Thereof The methods and compositions of the present invention relate to the use of PTH related peptide (PTHrP) for use as a hematoprotectant in any therapy where it is desirable to prevent cytotoxic cytopenia. PTHrP was previously known as the factor responsible for humoral hypercalcemia of malignancy and is a peptide of 138-174 amino acids (depending on alternative splicing) which binds to the PTH/PTHrP receptor. The compositions of the invention may employ a full length PTHrP; a truncated
PTHrP polypeptide that is physiologically active, or a polypeptide analog of PTHrP. Such proteins or polypeptides may be produced by conventional automated peptide synthesis
methods or by recombinant expression. General principles for designing and making proteins are well known to those of skill in the art and are discussed in further detail below.
The N-terminal 34 amino acid sequence of PTHrP has limited sequence homology to that of parathyroid hormone. PTHrP is generally less potent and less bone anabolic than PTH. The sequence of hPTHrP (1-34) is as follows:
Ala VaI Ser GIu His GIn Leu Leu His Asp Lys GIy Lys Ser He GIn Asp Leu Arg Arg Arg Phe Phe Leu His His Leu He Ala GIu He His Thr Ala (SEQ ID NO:1).
Those of skill in the art have reported several truncated homologs and analogs of PTHrP. Analogs in which amino acid residues 22-31 of PTHrP(I -34) are replaced by an amphipathic α-helix (U.S. Pat. No. 5,589,452 and WO 97/07815) and related derivatives have been described as useful for treating osteoporosis. (Vickery et al. J. Bone & Mineral Research, 11(12):1943-1951 (1996); D. Leaffer et al. Endocrinology, 136(8):3624-3631 (1995)).
In addition, monocyclic and bicyclic analogs of PTHrP (1-34) and PTHrP(7-34) were shown to bind strongly to the PTH receptor and stimulate (or antagonise) PTH-stimulated adenyl cyclase activity in osteoblast-like cells. (Michael Chorev et al. Biochemistry, 36:3293- 3299 (1997), and "Cyclic analogs of PTH and PTHrP," WO 96/40193).
U.S. Patent No. 6,583,114 provides compositions of PTHrP in which amino acid residues 22-31 form an amphipathic α-helix. U.S. Patent No. 6,849,710, shows methods and compositions for synthesizing various useful analogs of PTHrP. Other PTHrP compositions that may be used in the present invention are described in e.g., U.S. Patent 6,362,163 and U.S. Patent 6,147,186, which provide descriptions of PTHrP compositions having the amino- acid sequence:
1-Ala VaI Ser GIu His GIn Leu Leu His Asp Lys GIy Lys Ser He GIn Asp Leu Arg Arg Arg Phe Phe Leu His His Leu He Ala GIu He His Thr Ala GIu Tyr -36 in which amino acid 23 or amino acids 5 and 23 are altered.
Other compositions that may be useful include analogs of PTHrP described in U.S. Patent 6,503,534; U.S. Patent 6,544,949; U.S. Patent 5,723,577; U.S. Patent 5,955,574; U.S. Patent 5,969,095; U.S. Patent 6,921,750 each of which provide a teaching of composition that comprise [Glu22,25, Leu23,28,31, Aib29, Lys26,30] PTHrP (1-34)NH2.
U.S. Patent 5,688,760 describes polypeptides comprising an N-terminal amino acid sequence corresponding to amino acids 107-111 of PTHrP. The PTHrP may be provided as a fusion protein for example as a fusion protein with an Fc region as described in U.S. Patent 6,756,480. U.S. Patent 6,472,505 and U.S. application 2002/0132973describes useful cyclic and acyclic analogs of hPTH (1-34) and hPTHrP (1-34), pharmaceutical compositions of such analogs and methods of using the same in treatment of diseases associated with calcium regulation with those analogs.
"A truncated PTHrP polypeptide that is physiologically active" is a polypeptide that has a sequence that is less than the full complement of amino acids found in full length PTHrP which, nonetheless, elicits a physiological response that is associated with PTHrP.
Such a response may be greater or smaller in magnitude than the response seen from the same concentration of full length PTHrP. The truncated PTHrP need not be fully homologous with PTHrP to elicit a similar physiological response. Typically, the truncated PTHrP will be truncated from the C-terminus and will range from 30 to 40 residues, with PTHrP(I -32), PTHrP(l-34), PTHrP(l-36), PTHrP(I -37),and PTHrP(l-38) being preferred, but not exclusive, representatives of this group. In other particular embodiments, the PTHrP may be the N-terminal sequences of between 30 to 50 residues of PTHrP, preferably from 1-32, 1-33, 1-34, 1-35, 1-36, 1-37 and 1-38, are particularly contemplated to be useful in the present invention. It should be noted that the PTHrP truncated sequences may be C-terminally truncated as compared to wild-type, N-terminally truncated as compared to wild-type or may be fragments of PTHrP that have been generated from the middle of the protein i.e., are both N- and C-terminally truncated as compared to wild-type.
A "polypeptide analog of PTHrP" refers to a polypeptide having art-accepted substitutions, deletions or insertions relative to wild-type PTHrP or is substantially homologous to PTHrP such that the analog has a similar physiological activity. An analog as used herein is thus any variant of the native wild-type PTHrP in which one or more of the amino acids has been substituted by another amino acid or an amino acid derivative. The analog may be an analog of the full-length PTHrP or alternatively, the analog will be an analog of a truncated PTHrP that polypeptide that is physiologically active. Those of skill in the art are referred to e.g., U.S. Patent 5,874,086; U.S. Patent 5,798,225; U.S. Patent
5,807,823; U.S. Patent 5,821,225; U.S. Patent 5,977,070; U.S. Patent 6,051,686; U.S. Patent 5,589,452; U.S. Patent 5,693,616; U.S. Patent 5,695,955; U.S. Patent 5,840,837; U.S. Patent 6,147,186; U.S. Patent 6,362,163; U.S. Patent 6,537,965; U.S. Patent 6,495,662; RE37919
Reissue of U.S. Patent 5,171,670; U.S. Patent 6,417,333; U.S. Patent 6,803,213; U.S. Patent 5,886,148; U.S. Patent 5,955,425; U.S. Patent 6,541,450; U.S. Patent 6,316,410; U.S. Patent 5,556,940; U.S. Patent 6,503,534; U.S. Patent 6,544,949; U.S. Patent 5,723,577; U.S. Patent 5,955,574; U.S. Patent 5,969,095; U.S. Patent 6,921,750; U.S. Patent 6,583,1 14; U.S. Patent 6,849,710; U.S. Patent 6,787,518 and U.S. applications 2003/0144209 2003/0203012; and 2005/0272660 which provide teachings of exemplary such analogs. Each of the foregoing patents, along with the other patents discussed above are expressly incorporated herein by reference as providing teachings of methods and composition for preparing PTHrP and PTHrP analogs that may be useful in the methods of the present invention. A particularly useful analog for use in the methods of the present invention is
Semparatide™. Semparatide is a synthetic analog of naturally occurring PTHrP (1-34). Semparatide comprised of a linear chain of 34 natural amino acids, and differs from PTHrP in the substitution of residues 22-31 with residues that form an amphiphilic helix. The sequence of the molecule is:
SEQ ID NO 1: A-V-S-E-H-Q-L-L-H-D-K-G-K-S-I-Q-D-L-R-R-R-E-L-L-E-K-L-L-E-K-L- H-T-A
The compound may be produced by recombinant means or may be synthesized by solid- phase polymer supported synthesis, using Fmoc-compatible coupling techniques, with acid labile side chain protection. Following completion of synthesis and deprotection, the compound may be cleaved from the solid support, purified by RP-HPLC and salt exchange (to the X acetate), and lyophilized (to afford the X acetate, N hydrate). The acetate is freely soluble in water. This compound has the formula:
H-Ala-Val^er-G-u-His-Gln-Leu-Leu-His-Asp-
1 2 3 4 5 6 7 8 9 10
Lys-Gly-Lys-Ser-Ile-Gln-Asp-Leu-Arg-Arg- t l 12 13 14 15 16 17 18 19 20
Arg-Glu-Leu-Leu-Glu-Lys-Leu-Leu-Glu-Lys-
21 22 23 24 25 26 27 23 29 30
Leu-Hk-Thr-Aia.NH2 • JCC2H4O2 • >«2θ 31 32 33 34
While it is preferred that the PTHrP full length proteins, PTHrP truncated proteins that are physiologically active, or the PTHrP analogs are derived from a native sequence that
is a human sequence, it should be understood that such PTHrP compositions may be PTHrP compositions of any mammalian species, e.g., human, bovine, porcine or rabbit may be used in this invention, with human PTHrP being the preferred source. One of skill in the art will recognize that substitution, deletion and insertion variants of the preferred embodiments enumerated below, according to the art-accepted principles described above, are also within the scope of the invention.
Typically, the dosage of the PTHrP or analogue or derivative thereof will range between about 0.01 and 10 μg/kg body weight per day, preferably from about 0.1 to about 0.5 fg/kg body weight per day. For a 50 kg human subject, the daily dose of active ingredient is from about 0.5 to about 100 μgs, preferably from about 5 to about 10 μgs. This dosage may be delivered in a conventional pharmaceutical composition by a single administration, by multiple applications, or via controlled release, as needed to achieve the most effective results. Dosing will continue for as long as is medically indicated, and may range from a few weeks to several months. Such dosing may be intermittent or it may be daily. Preferably the dosage is initiated prior to administration of the cytotoxic therapy and may continue during and after the cytotoxic therapy has stopped.
The invention also contemplates the use of PTHrP derivatives. As used herein the term PTHrP derivative is used to denote a PTHrP molecule that has been altered to modify properties other than its physiological properties. For example, the PTHrP molecule may be derivatized to make it more accessible. It may be derivatized to have added to it an agent that increases its uptake or its targeting. For example attaching the PTHrP to a diphosphonate or orthophosphate will facilitate the targeting of the PTHrP (or related polypeptide fragment, analogue or derivative thereof) to the bone marrow as will attaching it to certain antibodies or Fc regions of certain antibodies. The PTHrP may be derivatized with a water-soluble polymer. Various water-soluble polymers have been shown to modify biodistribution, improve the mode of cellular uptake, change the permeability through physiological barriers, and modify the rate of clearance from the body. (Greenwald et al., Crit Rev Therap Drug Carrier Syst. 2000; 17: 101-161; Kopecek et al., J Controlled Release., 74:147-158, 2001). To achieve either a targeting or sustained-release effect, water-soluble polymers have been synthesized that contain drug moieties as terminal groups, as part of the backbone, or as pendent groups on the polymer chain.
Polyethylene glycol (PEG), has been widely used as a drug carrier, given its high degree of biocompatibility and ease of modification (Harris et al., Clin Pharmacokinet.
2001;40(7):539-51). Attachment to various drugs, proteins, and liposomes has been shown to improve residence time and decrease toxicity. (Greenwald et al., Crit Rev Therap Drug Carrier Syst. 2000;17:101-161; Zalipsky et al., Bioconjug Chem. 1997;8:11 1-118). PEG can be coupled to active agents through the hydroxyl groups at the ends of the chain and via other chemical methods; however, PEG itself is limited to at most two active agents per molecule. In a different approach, copolymers of PEG and amino acids were explored as novel biomaterials which would retain the biocompatibility properties of PEG, but which would have the added advantage of numerous attachment points per molecule (providing greater drug loading), and which could be synthetically designed to suit a variety of applications (Nathan et al., Macromolecules. 1992;25:4476-4484; Nathan et al., . Bioconj Chem. 1993;4:54-62).
Those of skill in the art are aware of PEGylation techniques for the effective modification of drugs. For example, drug delivery polymers that consists of alternating polymers of PEG and tri-functional monomers such as lysine have been used by VectraMed (Plainsboro, NJ). The PEG chains (typically 2000 daltons or less) are linked to the a- and e- amino groups of lysine through stable urethane linkages. Such copolymers retain the desirable properties of PEG, while providing reactive pendent groups (the carboxylic acid groups of lysine) at strictly controlled and predetermined intervals along the polymer chain. The reactive pendent groups can be used for derivatization, cross-linking, or conjugation with other molecules. These polymers are useful in producing stable, long-circulating pro-drugs by varying the molecular weight of the polymer, the molecular weight of the PEG segments, and the cleavable linkage between the drug and the polymer. The molecular weight of the PEG segments affects the spacing of the drug/linking group complex and the amount of drug per molecular weight of conjugate (smaller PEG segments provides greater drug loading). In general, increasing the overall molecular weight of the block co-polymer conjugate will increase the circulatory half-life of the conjugate. Nevertheless, the conjugate must either be readily degradable or have a molecular weight below the threshold-limiting glomular filtration (e.g., less than 45 kDa). Thus, PEGylated proteins in the range of between 20 and 35 kDa in molecular weight will be useful. In addition, to the polymer backbone being important in maintaining circulatory half- life, and biodistribution, linkers may be used to maintain the therapeutic agent in a pro-drug form until released from the backbone polymer by a specific trigger, typically enzyme activity in the targeted tissue. For example, this type of tissue activated drug delivery is
particularly useful where delivery to a specific site of biodistribution is required and the therapeutic agent is released at or near the site of pathology. Linking group libraries for use in activated drug delivery are known to those of skill in the art and may be based on enzyme kinetics, prevalence of active enzyme, and cleavage specificity of the selected disease- specific enzymes (see e.g., technologies of established by VectraMed, Plainsboro, NJ). Such linkers may be used in modifying the PTHrP proteins described herein for therapeutic delivery.
B. Preparation and Purification of PTHrP, Analogs and Derivatives Thereof The PTHrP proteins or fragments thereof can be synthesized in solution or on a solid support in accordance with conventional techniques. Various automatic synthesizers are commercially available and can be used in accordance with known protocols. See, for example, Stewart and Young, Solid Phase Peptide Synthesis, 2d. ed., Pierce Chemical Co., (1984);Tam et al., J. Am. Chem. Soc, 105:6442, (1983); Merrifield, Science, 232: 341-347, (1986); and Barany and Merrifield, The Peptides, Gross and Meienhofer, eds, Academic
Press, New York, 1-284, (1979), each incorporated herein by reference. The proteins can be readily synthesized and then screened in PTHrP receptor binding/activity assays to determine whether the proteins produced possess the requisite PTHrP-like activity as an initial screen. The proteins/fragments also may be tested in an exemplary physiological assays. Any PTHrP-derived protein that has at least some physiological effect associated with PTHrP may be used in the methods of the present invention.
As an alternative to automated peptide synthesis, recombinant DNA technology may be employed wherein a nucleotide sequence which encodes a protein of choice is inserted into an expression vector, transformed or transfected into an appropriate host cell and cultivated under conditions suitable for expression as described herein below. Recombinant methods are especially preferred for producing longer polypeptides and a variety of expression vector/host systems may be utilized to contain and express the peptide or protein coding sequence. Recombinant expression of proteins is routine and well known to those of skill in the art. Expression may be achieved in microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid or cosmid DNA expression vectors; yeast transformed with yeast expression vectors (Giga-Hama et al., Biotechnol Appl Biochem., 30 ( Pt 3):235-44, 1999); insect cell systems infected with virus expression vectors (e.g., baculovirus, see Ghosh et al., MoI Ther. 6(1):5-1 1, 2002); plant cell systems transfected with
virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with bacterial expression vectors (e.g., Ti or pBR322 plasmid; see e.g., Babe et al., Biotechnol Genet Eng Rev.; 17:213-52, 2000); or animal cell systems. Exemplary protocols for the recombinant expression of the peptide substrates or fusion polypeptides in bacteria, yeast and other invertebrates are known to those of skill in the art.
Mammalian host systems for the expression of recombinant proteins also are well known to those of skill in the art. Mammalian cells that are useful in recombinant protein productions include but are not limited to VERO cells, HeLa cells, Chinese hamster ovary (CHO) cell lines, COS cells (such as COS-7), Wl 38, BHK, HepG2, 3T3, RIN, MDCK, A549, PC 12, K562 and 293 cells. Those of skill in the art are aware of various techniques for optimizing mammalian expression of proteins, see e.g., Kaufman, MoI Biotechnol. 16(2):151-60, 2000; Colosimo et al., Biotechniques, 29(2):314-8, 2000. Host cell strains may be chosen for a particular ability to process the expressed protein or produce certain post- translation modifications that will be useful in providing protein activity. Such modifications of the polypeptide include, but are not limited to, acetylation, carboxylation, glycosylation, phosphorylation, lipidation and acylation. Post-translational processing which cleaves a "prepro" form of the protein may also be important for correct insertion, folding and/or function. Different host cells such as CHO, HeLa, MDCK, 293, WI38, and the like have specific cellular machinery and characteristic mechanisms for such post-translational activities and may be chosen to ensure the correct modification and processing of the introduced, foreign protein.
In the recombinant production of proteins of the invention, it would be necessary to employ vectors comprising polynucleotide molecules for encoding the PTHrP-derived proteins. Methods of preparing such vectors as well as producing host cells transformed with such vectors are well known to those skill in the art. Recombinant expression may employ a selectable marker and an origin of replication, for propagation in a host. These elements of the expression constructs are well known to those of skill in the art. Generally, the expression vectors include DNA encoding the given protein being operably linked to suitable transcriptional or translational regulatory sequences, such as those derived from a mammalian, microbial, viral, or insect genes. Examples of regulatory sequences include transcriptional promoters, operators, or enhancers, mRNA ribosomal binding sites, and appropriate sequences which control transcription and translation.
The terms "expression vector," "expression construct " or "expression cassette " are used interchangeably throughout this specification and are meant to include any type of genetic construct containing a nucleic acid coding for a gene product in which part or all of the nucleic acid encoding sequence is capable of being transcribed. The choice of a suitable expression vector for expression of the PTHrP peptides or polypeptides of the invention will of course depend upon the specific host cell to be used, and is within the skill of the ordinary artisan. Methods for the construction of mammalian expression vectors are disclosed, for example, in Okayama and Berg (MoI. Cell. Biol. 3:280 (1983)); Cosman et al. (MoI. Immunol. 23:935 (1986)); Cosman et al. (Nature 312:768 (1984)); EP-A-0367566; and WO 91/18982. Other considerations for producing expression vectors are detailed in e.g., Makrides et al., Protein Expr Purif, 17(2): 183-202; 1999; Kost et al., Curr Opin Biotechnol., 10(5):428-33, 1999. Wurm et al., Curr Opin Biotechnol. 10(2): 156-9, 1999 is incorporated herein as teaching factors for consideration in the large- scale transient expression in mammalian cells for recombinant protein production. The expression construct may further comprise a selectable marker that allows for the detection of the expression of a peptide or polypeptide. Usually the inclusion of a drug selection marker aids in cloning and in the selection of transformants, for example, neomycin, puromycin, hygromycin, DHFR, zeocin and histidinol. Alternatively, enzymes such as herpes simplex virus thymidine kinase (tk) (eukaryotic), β-galactosidase, luciferase, or chloramphenicol acetyltransferase (CAT) (prokaryotic) may be employed. Immunologic markers also can be employed. For example, epitope tags such as the FLAG system (IBI, New Haven, CT), HA and the 6xHis system (Qiagen, Chatsworth, CA) may be employed. Additionally, glutathione S-transferase (GST) system (Pharmacia, Piscataway, NJ), or the maltose binding protein system (NEB, Beverley, MA) also may be used. The selectable marker employed is not believed to be important, so long as it is capable of being expressed simultaneously with the nucleic acid encoding a gene product. Further examples of selectable markers are well known to one of skill in the art.
Expression requires that appropriate signals be provided in the vectors, such as enhancers/promoters from both viral and mammalian sources that may be used to drive expression of the nucleic acids of interest in host cells. Usually, the nucleic acid being expressed is under transcriptional control of a promoter. A "promoter" refers to a DNA sequence recognized by the synthetic machinery of the cell, or introduced synthetic machinery, required to initiate the specific transcription of a gene. Nucleotide sequences are
operably linked when the regulatory sequence functionally relates to the DNA encoding the protein of interest (e.g., the PTHrP or any of the hematopoietic factors discussed herein, variants thereof and the like). Thus, a promoter nucleotide sequence is operably linked to a given DNA sequence if the promoter nucleotide sequence directs the transcription of the sequence.
Similarly, the phrase "under transcriptional control" means that the promoter is in the correct location and orientation in relation to the nucleic acid to control RNA polymerase initiation and expression of the gene. Any promoter that will drive the expression of the nucleic acid may be used. The particular promoter employed to control the expression of a nucleic acid sequence of interest is not believed to be important, so long as it is capable of directing the expression of the nucleic acid in the targeted cell. Thus, where a human cell is targeted, it is preferable to position the nucleic acid coding region adjacent to and under the control of a promoter that is capable of being expressed in a human cell. Generally speaking, such a promoter might include either a human or viral promoter. Common promoters include, e.g., the human cytomegalovirus (CMV) immediate early gene promoter, the SV40 early promoter, the Rous sarcoma virus long terminal repeat, β-actin, rat insulin promoter, the phosphoglycerol kinase promoter and glyceraldehyde-3-phosphate dehydrogenase promoter, all of which are promoters well known and readily available to those of skill in the art, can be used to obtain high-level expression of the coding sequence of interest. The use of other viral or mammalian cellular or bacterial phage promoters which are well-known in the art to achieve expression of a coding sequence of interest is contemplated as well, provided that the levels of expression are sufficient to produce a recoverable yield of protein of interest. By employing a promoter with well known properties, the level and pattern of expression of the protein of interest following transfection or transformation can be optimized. Inducible promoters also may be used.
Another regulatory element that is used in protein expression is an enhancer. These are genetic elements that increase transcription from a promoter located at a distant position on the same molecule of DNA. Where an expression construct employs a cDNA insert, one will typically desire to include a polyadenylation signal sequence to effect proper polyadenylation of the gene transcript. Any polyadenylation signal sequence recognized by cells of the selected transgenic animal species is suitable for the practice of the invention, such as human or bovine growth hormone and SV40 polyadenylation signals.
Also contemplated as an element of the expression cassette is a terminator. These elements can serve to enhance message levels and to minimize read through from the cassette into other sequences. The termination region which is employed primarily will be one selected for convenience, since termination regions for the applications such as those contemplated by the present invention appear to be relatively interchangeable. The termination region may be native with the transcriptional initiation, may be native to the DNA sequence of interest, or may be derived for another source.
Further, it has been shown that polyhistidylation of nucleic acid molecules is useful in achieving cytosolic delivery of nucleic acids, and that ionic complexes between histidylated polylysine and a pDNA are attractive for developing a nonviral gene delivery system (Midoux et al., Somat Cell MoI Genet. 2002 Nov;27(l-6):27-47).
Site-specific mutagenesis may be useful in the preparation of individual PTHrP related proteins used in the methods of the invention. This technique employs specific mutagenesis of the underlying DNA (that encodes the amino acid sequence that is targeted for modification). The technique further provides a ready ability to prepare and test sequence variants, incorporating one or more of the foregoing considerations, by introducing one or more nucleotide sequence changes into the DNA. Site-specific mutagenesis allows the production of mutants through the use of specific oligonucleotide sequences that encode the DNA sequence of the desired mutation, as well as a sufficient number of adjacent nucleotides, to provide a primer sequence of sufficient size and sequence complexity to form a stable duplex on both sides of the deletion junction being traversed. Typically, a primer of about 17 to 25 nucleotides in length is preferred, with about 5 to 10 residues on both sides of the junction of the sequence being altered.
The technique typically employs a bacteriophage vector that exists in both a single stranded and double stranded form. Typical vectors useful in site-directed mutagenesis include vectors such as the M 13 phage. These phage vectors are commercially available and their use is generally well known to those skilled in the art. Double stranded plasmids also are routinely employed in site-directed mutagenesis, and eliminates the step of transferring the gene of interest from a phage to a plasmid. In general, site-directed mutagenesis is performed by first obtaining a single-stranded vector, or melting of two strands of a double stranded vector which includes within its sequence a DNA sequence encoding the desired protein. An oligonucleotide primer bearing
the desired mutated sequence is synthetically prepared. This primer is then annealed with the single-stranded DNA preparation, taking into account the degree of mismatch when selecting hybridization (annealing) conditions, and subjected to DNA polymerizing enzymes such as E. coli polymerase I Klenow fragment, in order to complete the synthesis of the mutation- bearing strand. Thus, a heteroduplex is formed wherein one strand encodes the original non- mutated sequence and the second strand bears the desired mutation. This heteroduplex vector is then used to transform appropriate cells, such as E. coli cells, and clones are selected that include recombinant vectors bearing the mutated sequence arrangement.
Of course, the above described approach for site-directed mutagenesis is not the only method of generating potentially useful mutant peptide species and as such is not meant to be limiting. The present invention also contemplates other methods of achieving mutagenesis such as for example, treating the recombinant vectors carrying the gene of interest mutagenic agents, such as hydroxylamine, to obtain sequence variants.
It will be desirable to purify the peptides of the present invention. Protein purification techniques are well known to those of skill in the art. These techniques involve, at one level, the crude fractionation of the cellular milieu to polypeptide and non-polypeptide fractions. Having separated the peptides or polypeptides of the invention from other proteins, the polypeptides or peptides of interest may be further purified using chromatographic and electrophoretic techniques to achieve partial or complete purification (or purification to homogeneity). Analytical methods particularly suited to the preparation of a pure peptide include size-exclusion chromatography, ion-exchange chromatography, hydrophobic interaction chromatography, isoelectric focusing and capillary electrophoresis. A particularly efficient method of purifying peptides is fast protein liquid chromatography (FPLC) or even high performance liquid chromatography (HPLC). Certain aspects of the present invention concern the purification, and in particular embodiments, the substantial purification, of an encoded polypeptide, protein or peptide. The term "purified polypeptide, protein or peptide" as used herein, is intended to refer to a composition, isolated from other components, wherein the polypeptide, protein or peptide is purified to any degree relative to its naturally-obtainable state. A purified polypeptide, protein or peptide therefore also refers to a polypeptide, protein or peptide, free from the cellular environment in which it may naturally occur.
Generally, "purified" will refer to a polypeptide, protein or peptide composition that has been subjected to fractionation to remove various other components, and which composition substantially retains its expressed biological activity. Where the term "substantially purified" is used, this designation will refer to a composition in which the polypeptide, protein or peptide forms the major component of the composition, such as constituting about 50%, about 60%, about 70%, about 80%, about 90%, about 95% or more of the proteins in the composition.
Various techniques suitable for use in protein purification will be well known to those of skill in the art. These include, for example, precipitation with ammonium sulphate, PEG, antibodies and the like or by heat denaturation, followed by centrifugation; chromatography steps such as ion exchange, gel filtration, reverse phase, hydroxylapatite and affinity chromatography; isoelectric focusing; gel electrophoresis; and combinations of such and other techniques. As is generally known in the art, it is believed that the order of conducting the various purification steps may be changed, or that certain steps may be omitted, and still result in a suitable method for the preparation of a substantially purified polypeptide, protein or peptide.
C. Combination Therapy with other Hematoprotectants
Hematopoietic reconstitution has previously been achieved by administration of cytokine growth factors such as Erythropoietin, G-CSF, GM-CSF, M-CSF, TCGF, flt-3, thrombopoietin, thymic stromal lymphopoietin, keratinocyte growth factor, PDGF, IGF, IL 1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL- 12, IL-13; IL-15, IL- 16, IL- 17, stem cell factor (also known as c-kit ligand), and M-CSF, or PEGylated derivatives thereof, as soon as possible after the subject has been exposed to the chemotherapeutic and/or radiotherapeutic agent. In the present invention, in order to combat the myelosuppression observed with the chemotherapy the PTHrP-based methods may be used in combination with cytokine therapy. Combination therapy contemplated herein includes administration of one or more hematopoietic factors selected from the group consisting of Erythropoietin, G-CSF, GM-CSF, M-CSF, TCGF, flt-3, thrombopoietin, thymic stromal lymphopoietin, keratinocyte growth factor, PDGF, IGF, IL 1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13; IL-15, IL-16, IL-17, stem cell factor (also known as c-kit ligand), and M-CSF, or PEGylated derivatives thereof in combinations with PTHrP (the hematopoietic factor may be administered either concurrently with PTHrP, or prior to, and preferably subsequent to the PTHrP) is particularly contemplated.
Erythropoietin, G-CSF, GM-CSF, M-CSF, TCGF, flt-3, thrombopoietin, thymic stromal lymphopoietin, keratinocyte growth factor, PDGF, IGF, IL 1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-IO, IL-I l, IL-12, IL-13; IL-15, IL-16, IL-17, stem cell factor (also known as c-kit ligand), and M-CSF, are merely exemplary suitable immunomodulators that can be used in combination with PTHrP or an analogue thereof for adjunct therapy. U.S. patent No. 5,120,525, which is incorporated by reference, provides methods of use of such agents in adjunct immunomodulator therapy.
Cytokines, or growth factors, are hormone-like peptides produced by diverse cells and are capable of modulating the proliferation, maturation and functional activation of particular cell types. Herein, cytokines refer to a diverse array of growth factors, such as hematopoietic cell growth factors (e.g., erythropoietin, colony stimulating factors and interleukins), nervous system growth factors (e.g., glial growth factor and nerve growth factor), mostly mesenchymal growth factors (e.g., epidermal growth factor), platelet-derived growth factor, and fibroblast growth factor I, II and III and the like. Several cytokines may act in concert to induce cell differentiation and maturation, and such cytokines may have other biological functions. As such, limiting the amount of cytokine to be used in treating a patient that has undergone myelosuppressive therapy is desirable. The combined use of PTHrP with the cytokine-based therapy will allow the use of less cytokine for treating the subject post radiation or post chemotherapy than the amount that would be needed in the absence of the PTHrP.
In the case of IL-I, there may be several forms, such as IL-I -alpha and IL-I -beta, which nevertheless appear to have a similar spectrum of biological activity. Preferred cytokines for use in the method and compositions of the invention are lymphokines, i.e., those cytokines which are primarily associated with induction of cell differentiation and maturation of myeloid and possibly other hematopoietic cells. A preferred lymphokine is IL- 1. Other such lymphokines include, but are not limited to, G-CSF, M-CSF, GM-CSF, Multi- CSF (IL-3), and IL-2 (T-cell growth factor, TCGF). IL-I appears to have its effect mostly on myeloid cells, IL-2 affects mostly T-cells, IL-3 affects mutiple precursor lymphocytes, G- CSF affects mostly granulocytes and myeloid cells, M-CSF affects mostly macrophage cells, GM-CSF affects both granulocytes and macrophage. Other growth factors affect immature platelet (thrombocyte) cells, erythroid cells, and the like.
The cytokines may be used alone or may be provided in a combination of two or more cytokines in order to provide protection against, mitigation and/or reversal of myeloid or hematopoietic toxicity associated with cytotoxic agents when administered in combination with the PTHrP as described herein. Examples of possible combinations include IL-I + GC- CSF + PTHrP, IL-I + IL-3 + PTHrP, G-CSF + IL-3 + PTHrP, IL-I + platelet growth factor + PTHrP and the like. Certain combinations will be preferred, depending on the maturation state of the target cells to be affected, and the time in the course of cytotoxic action that the protective agent needs to be administered. For example, in patients with depression of several hematopoietic cell types (e.g., myeloid, lymphoid and platelet), a combination of IL-I + IL-3/and/or platelet growth factor in combination with PTHrP may be particularly useful, while more severe depression of the myeloid series may require such combinations as IL-I + G-CSF in combination with the PTHrP. Certain cytotoxic agents have greater compromising effects on particular hematopoietic elements, either because of the nature of the agent or the dosage necessary to achieve a therapeutic effect, and the appropriate choice, dosage and mode of administration of cytokine(s) will follow from such effects.
In addition to the cytokines, the PTHrP may be provided in combination with other compounds or techniques for preventing, mitigating or reversing the side effects of cytotoxic agents. Examples of such combinations include, e.g., administration of the PTHrP and a cytokine together with a second antibody for rapid clearance, as described, e.g., in Goldenberg, U.S. Patent 4,624,846, from 3 to 72 hours after administration of a targeted primary antibody or antibody fragment conjugate (with a radioisotope, drug or toxin as the cytotoxic component of the immunoconjugate) or of a non-conjugated drug or toxin, to enhance clearance of the conjugate, drug or toxin from the circulation and to mitigate or reverse myeloid and other hematopoietic toxicity caused by the therapeutic agent. In all aspects of the invention, it is a desired goal bring the peripheral blood cell counts and differential to within the normal levels or as close thereto as possible. The RBC level in terms of hemoglobin concentration, which is normally 13.5-17.5 gm/dL in men and 1 1.8 - 15.5 gm/dL in women, is reduced to less than 10 gm/dL with chemotherapy or radiotherapy; WBC count which is normally in the range of 4-1 lxlO9/L is reduced to less than lxlO9/L in subjects in response to chemotherapy of radiotherapy; platelets are in a normal range of 150,000 to 400,000/μL but are reduced to 50,000/uL in response to chemotherapy or radiotherapy. The methods of the invention are useful in that where the cytotoxic therapy produces such a reduction in blood cell counts, this reduction can be
reversed by administration of either PTHrP alone or a combination of PTHrP with one or more cytokines and or one or more other agents. In embodiments where cytokine is administered in combination with the PTHrP to return the blood component levels to be close to normal. It is contemplated that the cytokine is administered in a dose of from about 1 μg to about 500 μg, preferably 5-100 μg, more preferably about 10 ug. The clinician will appreciate that variations in the timing and dosage of cytokine administration as a function of the type of cytokine used, the extent and rate of compromise of the bone marrow and/or other components of the myeloid and/or other hematopoietic elements and the individual characteristics of the patient and the therapy protocol will be possible and often desirable. These can easily be made by the clinician using conventional monitoring techniques and dosimetric principles.
D. Normal Levels of Blood Components
Throughout the methods of the present invention, it will be desirable to monitor changes in the subjects blood cell populations. The complete blood cell count will routinely be monitored prior to initiation of the therapeutic regimen as well as during and post-therapy follow-up. During treatment and follow-up, monitoring the patient's blood cell count is important (e.g., prior to each treatment cycle and 10-14 days after each treatment cycle). A complete blood count provides important information about the kinds and numbers of cells in the blood, including red blood cells, white blood cells, and platelets. A complete blood count typically will involve determining a white blood cell
(WBC) count. These cells protect the body against infection and the number of white cells can increase dramatically upon infection or chemo- or radio-therapeutic insult, as such, the number of white blood cells is sometimes used to identify an infection or monitor the body's response to cancer treatment. Analysis of the white blood cell types (WBC differential) in blood will be a useful indicator for the efficacy of the methods of the invention. The major types of white blood cells are neutrophils, lymphocytes, monocytes, eosinophils, and basophils. Immature neutrophils, called band neutrophils, are also included and counted as part of this test. Each type of cell plays a different role in protecting the body. The numbers of each one of these types of white blood cells give important information about the immune system. An increase or decrease in the numbers of the different types of white blood cells can help identify infection, an allergic or toxic reaction to certain medications or chemicals, and many conditions, such as leukemia.
To assess white blood cell (WBC) count for the present invention, a health professional will look at both the number (WBC count) and the WBC differential. To determine whether there are too many or too few of a certain type of cell, the total count is multiplied by the percentage of that particular cell. There are normal values for the total number of each type of white cell
The typical normal range of white blood cells in males and non-pregnant females is between 4,500-11,000 cells/microliter (μL)3 or 4.5-11.0 x 109/Liter (SI units). It is contemplated that the PTHrP compositions administered in accordance with the present invention will maintain or return the subjects white blood cell count to within this normal range. The average distribution of white blood cell types 47%-77% neutrophils; 0%-3% band neutrophils; 16%-43% lymphocytes, 0.5%-10%, monocytes, 0.3%-7% eosinophils, and 0.3%-2% basophils. In further embodiments, the PTHrP administration in accordance with the methods of the present invention will balance the white blood cell differential such that the white blood cell components are present in these appropriate distributions relative to each other.
In addition to white blood cell counts, it may be desirable to obtain a red blood cell count (average value 4.6-6.2 million RBCs per microliter (μL) or 4.6-6.2 x 10l2/Liter (SI units), a hematocrit (40%-54% in men; 37%-47% women; 31-41% children; 44%-64% in newborns) and/or perform hemoglobin test (14-18 g/dL or 8.7-11.2 mmol/L (SI units) in men and 12-16 g/dL or 7.4-9.9 mmol/L in women). Other factors that can be monitored include hemoglobin levels, erythrocyte sedimentation rate (ESR), erticulocyte count, red eel distribution width, and red blood cell indices.
The platelet count also could be determined. The average normal platelet count is 140,000-450,000 platelets per mm3 or 150-400 x 109/Liter (SI units). Methods for performing such tests for various blood components are known to those of skill in the art.
£. Pharmaceutical Compositions
Pharmaceutical compositions for administration according to the present invention can comprise at least one PTHrP-derived protein (e.g., a full length PTHrP polypeptide, or a variant or analog thereof or any other PTHrP-related protein that protects hematopoeitic cells from the cytotoxic effects of chemo- or radiotherapy).
The pharmaceutical compositions also may include other therapeutic agents. For example, while the present invention stems from the unique discovery of the therapeutic efficacy of PTHrP compositions in the protection of hematopoietic cells from cytotoxic chemotherapy or radiotherapy, it is contemplated that the PTHrP may be administered not only alone but in combination with other therapeutic regimens designed for the treatment of cytotoxic agent-induced cytopenia (e.g., anemia, neutropenia and thrombocytopenia). Any increase in the level of peripheral blood cell count or any component of the blood count in response to such therapy will be a useful therapeutic outcome. As discussed above, cytokines such as Erythropoietin, G-CSF, GM-CSF, M-CSF, TCGF, flt-3, thrombopoietin, thymic stromal lymphopoietin, keratinocyte growth factor, PDGF, IGF, IL 1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-IO, IL-I l, IL- 12, IL-13, IL- 15, IL- 16, IL- 17, stem cell factor (also known as c-kit ligand), and M-CSF, and the like may be useful in a combination therapy protocol. As such, one or more of cytokines may be provided in the pharmaceutical compositions to be used in the treatment of subjects that are about to undergo chemotherapy, radiotherapy or some other therapy that will have a myelosuppressive effect
Each of the therapeutic preparations is preferably provided in a pharmaceutically acceptable form optionally combined with a pharmaceutically acceptable carrier. These compositions can be administered by any means that achieve their intended purposes. Individualized amounts and regimens for the administration of the compositions for the treatment of infertility using the methods of the present invention can be determined readily by those with ordinary skill in the art using the guidance provided herein to determine the efficacy of a dosage in an animal model and then to increase the dosage to higher mammals. Administration of many cytokines are known to those of skill in the art and can readily be found in the Regimens for use of PTH in other indications also are known and discussed in the Physician's Desk Reference. That document provides exemplary guidance as to types of formulations, routes of administration and treatment regimens that may be used in administering PTH and other hormones. Such routes teachings can be used for the administration of PTHrP. Any of the protocols, formulations, routes of administration and the like described therein can readily be modified for use in the present invention. Compositions within the scope of this invention include all compositions comprising at least one PTHrP-related polypeptide formulated in an amount effective to achieve its intended purpose of protecting a composition of hematopoietic cells from cell death and/or preventing or overcoming myelosuppression. The active agents used in the
methods of the present invention may be administered by any means normally employed for such administration.
It is understood that the suitable dose of a composition according to the present invention will depend upon the age, health and weight of the recipient, kind of concurrent treatment, if any, frequency of treatment, and the nature of the effect desired.
However, the most preferred dosage can be tailored to the individual subject, as is understood and determinable by one of skill in the art, without undue experimentation. This typically involves adjustment of a standard dose, e.g., reduction of the dose if the patient has a low body weight. Peripheral blood cell counts and differentials will be a useful determinant in assisting the clinician to determine and/or adjust the dose.
The total dose required for each treatment may be administered in multiple doses or in a single dose. The compositions may be administered alone or in conjunction with other therapeutics directed to the disease or directed to other symptoms thereof.
The compositions of the invention should be formulated into suitable pharmaceutical compositions, i.e., in a form appropriate for in vivo applications in a the therapeutic intervention of myelosuppression. Generally, this will entail preparing compositions that are essentially free of pyrogens, as well as other impurities that could be harmful to humans or animals, preferably for oral administration. The hormone formulations may be formulated akin to the currently available hormonal preparations. The peptide/protein formulations may be formulated similarly to any other small protein composition. All routes of administration are contemplated (e.g. Such routes of administration include, subcutaneous, intravenous, intradermal, intraaterial, intramuscular, intramammary, intraperitoneal, intrathecal, intraocular, retrobulbar, intrapulmonary (e.g., term release), aerosol, sublingual, nasal, anal, vaginal, oral, or transdermal delivery, or by surgical implanation). Transdermal patches also may be used. Intranasal administration is of particular interest.
One will generally desire to employ appropriate salts and buffers to render the compositions stable and allow for uptake of the compositions at the target site. Generally the protein compositions of the invention are provided in lyophilized form to be reconstituted prior to administration. Buffers and solutions for the reconstitution of the compositions may be provided along with the pharmaceutical formulation to produce aqueous compositions of the present invention for administration. Such aqueous compositions will comprise an effective amount of each of the therapeutic agents being used, dissolved or dispersed in a
pharmaceutically acceptable carrier or aqueous medium. Such compositions also are referred to as inocula. The phrase "pharmaceutically or pharmacologically acceptable" refer to molecular entities and compositions that do not produce adverse, allergic, or other untoward reactions when administered to an animal or a human. As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the therapeutic compositions, its use in therapeutic compositions is contemplated. Supplementary active ingredients also can be incorporated into the compositions.
Methods of formulating proteins and peptides for therapeutic administration also are known to those of skill in the art. Administration of these compositions according to the present invention will be via any common route so long as the target tissue is available via that route. Conventional routes of administration include, subcutaneous, intravenous, intradermal, intraarterial, intramusclar, intramammary, intraperitoneal, intrathecal, intraocular, retrobulbar, intrapulmonary (e.g., term release), aerosol, sublingual, nasal, anal, vaginal, oral, or transdermal delivery, or by surgical implantation at a particular site. The treatment may consist of a single dose or a plurality of doses over a period of time. Preferably, the route of administration is chosen for the therapeutic intervention to have maximum impact on the circulating blood cell counts.
The active compounds may be prepared for administration as solutions of free base or pharmacologically acceptable salts in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions also can be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper
fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients also can be incorporated into the compositions. The compositions of the present invention may be formulated in a neutral or salt form.
Pharmaceutically-acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups also can be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective. The formulations are easily administered in a variety of dosage forms such as injectable solutions, drug release capsules and the like. For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.
"Unit dose" is defined as a discrete amount of a therapeutic composition dispersed in a suitable carrier. Parenteral administration of the therapeutic compounds may be carried out with an initial bolus followed by continuous infusion to maintain therapeutic circulating levels of drug product. Those of ordinary skill in the art will readily optimize effective dosages and administration regimens as determined by good medical practice and the clinical condition of the individual patient.
The frequency of dosing will depend on the pharmacokinetic parameters of the agents and the routes of administration. The optimal pharmaceutical formulation will be determined by one of skill in the art depending on the route of administration and the desired dosage. Such formulations may influence the physical state, stability, rate of in vivo release and rate of in vivo clearance of the administered agents. Depending on the route of administration, a suitable dose may be calculated according to body weight, body surface areas or organ size. Further refinement of the calculations necessary to determine the appropriate treatment dose is routinely made by those of ordinary skill in the art without undue experimentation, especially in light of the dosage information and assays disclosed herein as well as the pharmacokinetic data observed in animals or human clinical trials.
Appropriate dosages may be ascertained through the use of established assays for determining blood levels in conjunction with relevant dose response data. The final dosage regimen will be determined by the attending physician, considering factors which modify the action of drugs, e.g., the drug's specific activity, severity of the damage and the responsiveness of the patient, the age, condition, body weight, sex and diet of the patient, the severity of any infection, time of administration and other clinical factors. As studies are conducted, further information will emerge regarding appropriate dosage levels and duration of treatment for specific diseases and conditions.
It will be appreciated that the pharmaceutical compositions and treatment methods of the invention may be useful in fields of human medicine and veterinary medicine. Thus the subject to be treated may be a mammal, preferably human or other animal. For veterinary purposes, subjects include for example, farm animals including cows, sheep, pigs, horses and goats, companion animals such as dogs and cats, exotic and/or zoo animals, laboratory animals including mice rats, rabbits, guinea pigs and hamsters; and poultry such as chickens, turkeys, ducks and geese.
The present invention also contemplated kits for use in the treatment of myelosuppression. Such kits include at least a first composition comprising the PTHrP-based proteins/peptides described above in a pharmaceutically acceptable carrier. Another component may be an agent used in treating myelosuppression (e.g., a composition that contains one or more cytokines etc.) in a pharmaceutically acceptable carrier. A third component may be the agent that is used for the radiotherapy or chemotherapy. The kits may additionally comprise solutions or buffers for effecting the delivery of the first, second and third compositions. The kits may further comprise catheters, syringes or other delivering devices for the delivery of one or more of the compositions used in the methods of the invention. The kits may further comprise instructions containing administration protocols for the therapeutic regimens.
Example 1 — Mouse model
14 days of pretreatment with a daily dose of semparatide enlarges the granulocyte and lymphoid compartments in mice. These cells are available for release following a challenge with a single dose of 5-fluorouracil and can be quantified by counting cells in the systemic circulation during the two weeks post chemotherapy. Semparatide induces an increase in the area under the curve of circulating white blood cells, lymphocytes and neutrophils post chemotherapy. These enhanced levels of circulating white blood cells, lymphocytes and neutrophils would be expected to improve survival in animals that would otherwise succumb to opportunistic infections during the post chemotherapeutic or post irradiation period of myelosupression. Results of pretreatment with semparatide in mice treated with 5- fluorouracil are shown in Figures 1-3. These results show that pretreatment with semparatide at a daily dose of 200/mg/kg for 14 days prior to the induction of myelosupression by a single dose of 225mg/kg of 5-fluorouracil at day 0 reduces the cytoreduction of granulocytes and
lymphocytes immediately following therapy and stimulates a significantly greater cellular response within 14 days after cytoreduction. These results are significantly better that those observed in this model even after 14 days of twice daily injections of G-CSF.
Example 2 - Clinical Trials
A patient that is about to undergo chemotherapy or radiation therapy for cancer is selected for treatment. The patient is treated once daily with 50 μg PTHrP or an analogue by subcutaneous injection (on each days -7 to day 0).
On day 0, the patient is treated with chemotherapy or radiation therapy. Twenty four hours after administration of the chemotherapy (day 1) and for the next 13 days (days 2-14) the patients will receive a once daily subcutaneous injection of PTHrP or analogue at the same pre-chemotherapy dose.
In chemotherapy or radiotherapy, there is often a drop in peripheral blood cell count. In this case, as a result of the treatment with the 50 ug PTHrP or analogue thereof, it is found that the usual drop in peripheral blood cell count is inhibited significantly, thus permitting the patient to receive more extensive chemotherapy or radiotherapy.
Frequent blood sampling will allow the physician to terminate PTHrP therapy prior to day 14 if circulating levels of RBCs, WBCs or platelets return to pre-treatment (normal) levels. As a result of administration of the PTHrP in combination with the chemotherapy or radiotherapy, the patient is able to tolerate greater amounts of the chemotherapy or radiotherapy and as such the therapeutic efficacy of the chemotherapy or radiotherapy is enhanced.
Simply, by way of example, and without intending to limit the invention, in some embodiments, the patient to be treated is one that has metastatic colon cancer. In exemplary treatment protocols such a patient is treated with 5-fluorouracil (5-FU) as the chemotherapeutic agent. Seven days prior to initiation of the 5-FU therapy 50 μg PTHrP or an analogue are administered to the patient by subcutaneous injection (on each days -7 to day 0). The patient is given 5-FU therapy over 5 days at a dose of 10 mg/kg/day intravenously (days 0, 1,2,3,4,5), repeated every second day during the next five days (day 7, day 9), and then twice monthly (1st and 15th day of each month) thereafter. In this type of therapy, toxicity usually begins to show by the 23rd day, whereby a drop in peripheral blood cell count. Throughout the therapy period, 50 μg PTHrP or an analogue is administered per day
by subcutaneous injection. This therapy is accompanied by frequent blood sampling and the PTHrP therapy is terminated or reduced if circulating levels of RBCs, WBCs or platelets return to pre-treatment (normal) levels. As a result of this combination therapy, a significant reduction in the liver metastasis in the patient is noted on CAT scans several months after initiation of therapy.
In another example, the patient to be treated has breast cancer. Seven days prior to instituting this chemotherapy, she is started on a PTHrP or a PTHrP regimen in which she is given a dose of 50 μg PTHrP or an analogue thereof per day for seven day. On day 8 and she is to be administered adriamycin (30 mg/kg i.v. daily for 3 weeks) and cytoxin (10 mg/kg i.v. every week). In a typical treatment for breast cancer the dose of adriamycin and cytoxin has be reduced due to cytotoxicity of the drugs. In the present case, however, as a result of the prophylactic administration of the PTHrP or analogue thereof, there is marginal or low drop in her peripheral blood cell count (e.g., there is only a 10-20% drop in her WBC count). Therefore, instead of subsequent courses of chemotherapy being reduced, as is conventionally practiced when hematologic side effects occur, a full course of both drugs is repeated under continued administration of PTHrP or analogue thereof. Resolution of some of the metastatic lesions is seen radiologically several months after the initiation of the chemotherapy.
In another example, a patient presenting a peritoneal spread of a colon cancer that is non-responsive to 5-FU treatment is to be treated with radiotherapy. The patient will be treated with a 35 mCi dose of Yttriwn-90 conjugated to a F(ab')2 fragment of a murine monoclonal antibody against carcinoembryonic antigen (CEA), by intraperitoneal injection. Seven days prior to initiating the therapy with the Yttrium-90, the patient begins receiving a daily dose of 50μg/day PTHrP or an analogue thereof (day-7 to day 0). On day 0 the patient is given the 35 mCi dose of Yttrium-90. The patient is administered PTHrP or analogue thereof at a dose of 50 μg/day over the following fourteen days. On day 2, and periodically over the next three weeks, the patient's peripheral blood cell count is taken. As long as there is no significant drop in the patient's blood cell count as compared to his normal peripheral blood cell count, the radioimmunotherapy is repeated 3 weeks later. The radioimmunotherapy may be accompanied by further administration of the PTHrP or analogue thereof. The patient is again monitored over the coming months. A further course of treatment is given 2-3 months after the initial treatment, again with administration of PTHrP or analogue thereof initiated 7 days prior to administration of the radiotherapy.
Radiological evidence of some tumor and ascites reduction is monitored. Administration of the PTHrP or analogue thereof will allow the patient to tolerate higher and more frequent doses of the radioimmunotherapy.
In another exemplary therapy, the patient presents with ovarian cancer manifesting as ascites and considerable distension of her abdomen. The patient is initiated on a prophylactic course of PTHrP or analogue thereof seven days prior (days -7 to 0) to the initiation of the cancer therapy. She is given 50 μg of the PTHrP or analogue thereof per day. At day 0, is given a course of 5 FU therapy as described above. The patient continues to receive PTHrP or analogue thereof over the next 14 days. It is anticipated that the treatment with the PTHrP or analogue thereof allows the patient to tolerate multiple doses of the 5 -FU and within months of the treatment there is a reduction in the abdominal distension.
In another example, the patient to be treated is one that is to receive a kidney transplant. 7 consecutive days prior to surgery, the patient is given 50 μg/day PTHrP or analogue thereof. On the day of the surgery, the patient is given an intravenous infusion of 20 mCi Y-90 that is conjugated to an agent that targets the Y-90 to activated T-lymphocytes, in order to kill the T-cells responsible to transplant rejection. The patient is given 50 μg/day PTHrP or analogue thereof and may also receive conventional corticosteroid therapy to prevent transplant rejection. Ten days later, a second dose of 10 mCi Y-90 is given i.v. During the interim period, the patient has continued to receive 50 μg/day doses of PTHrP or analogue thereof. The patient is monitored for graft rejection and peripheral blood cell count.
In each of the above example, a 50 μg/day dose of PTHrP or analogue thereof is given. It should be understood that this is merely an exemplary dose and the dose may be varied to be more or less than that amount. In addition, while it may be convenient to maintain the same dose throughout the regimen, it may be desirable to increase or decrease the dose on any given day.
In addition the above exemplary regimens teach administration of the PTHrP or analogue thereof as a prophylactic treatment for seven days prior to the cytotoxic therapy. This seven day period is merely an exemplary treatment period and those skilled in the art will understand that the therapy may be administered for longer (e.g., 14 days or more) or shorter time periods. Moreover, while it is contemplated that the PTHrP or analogue thereof is given on consecutive days, the therapy could, if desired by given intermittently (e.g., every alternate day.) Furthermore, while a prophylactic treatment with the PTHrP or analogue
thereof is contemplated, the skilled artisan will understand that the PTHrP or PTHrP analogue administration may be initiated at the same time as the cytotoxic agent therapy and need not precede the cytotoxic agent-based therapy.
As noted elsewhere, the PTHrP or analogue thereof may be administered in combination with one or more other hematopoeitic factors such as Erythropoietin, G-CSF (filgrastim), peg-G-CSF (pegfilarastim), GM-CSF (molgramostim or sargramostim), M-CSF, TCGF, flt-3, thrombopoietin, thymic stromallymphopoietin, keratinocyte growth factor, PDGF, IGF, IL 1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-IO, IL-11, IL- 12, IL-13; IL-15, IL-16, IL-17, and stem cell factor (also known as c-kit ligand). A Phase I/II trial design as using separatide as an adjunct hematoprotective agent with
Neulasta® (pegfilgrastim) is as follows:
PHASE I/II: Safety and Proof of concept
Exclusion Subjects who meet any of the following exclusion criteria prior to Criteria enrollment are not eligible to participate in the study:
1. Participation in another clinical research study within 30 days prior to consenting for this study
2. Pregnant or nursing
3. Prior treatment with chemotherapy.
4. Prior radiation therapy completed < 4 weeks prior to enrollment
5. Any "currently active" second malignancy, other than non- melanoma skin cancer. Patients are not considered to have a "currently active" malignancy, if they have completed therapy and are considered by their physician to be at least less than 30% risk of relapse over next 3 months.
6. Systolic blood pressure >160 mraHg measured on at least 2 occasions.
7. HIV
8. NYHA Class III or IV Congestive Heart Failure.
9. Myocardial infarction within the 6 months prior to the first dose of study drug.
10. Serious intercurrent infections or nonmalignant medical illnesses that are uncontrolled.
11. Active psychiatric illnesses/social situations that would limit compliance with protocol requirements.
12. Any physical or mental condition in the opinion of the investigator may interfere with the subject's ability to comply with the study procedures
Study Duration 9 month recruitment period
Enrollment duration is ~3 months. Treatment with semparatide begins 7 days prior to each chemotherapy cycle.
PHASE II: Efficacy
Claims
1. A method of decreasing chemotherapy and/or radiation therapy- induced cytopenia in a subject comprising administering to said subject a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof, wherein said PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered in an amount effective to decrease cytopenia in said subject as compared to the cytopenia induced in the absence of said administration.
2. The method of claim 1, wherein said administration of said composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is carried out prior to, after or subsequent to said chemotherapy and/or radiation therapy.
3. The method of claim 2, wherein said administration of said composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is carried out prior to said chemotherapy and/or radiation therapy.
4. The method of claim 3, wherein said composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered to said subject between 4-7 days prior to initiation of cytotoxic chemotherapy and/or radiation therapy.
5. The method of claim 1, wherein said cytopenia is selected from the group consisting of anemia, neutropenia and thrombocytopenia.
6. A method of decreasing myelosuppression in a subject comprising administering to said subject a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof.
7. The method of claim 6, wherein said myelosuppression is produced by chemotherapy or radiation therapy.
8. The method of claim 6, wherein said PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered prior to chemotherapy or said radiation therapy.
9. A method of decreasing the dose or duration of chemotherapy or radiation therapy to be administered to a subject in need thereof comprising administering to said subject a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof.
10. A method of preventing or decreasing the level of reduction in circulating blood cells in a subject receiving radiation therapy and/or chemotherapy, comprising administering to said subject a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof prior to administration of said radiation therapy and/or chemotherapy, wherein administration of said PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof prevents or produces a lower level of reduction of circulating blood cells in said subject as compared to a subject that does not receive said PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof.
11. A method of treating a subject that is at risk of having a decrease in the number of cells of the hematopoietic system, comprising the step of administration to said subject an effective amount of PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof, wherein said PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered before, after, or during said chemotherapy and/or radiation therapy.
12. The method of claim 11, wherein said cells of the hematopoietic system are selected from the group consisting of, including but not limited to T cells, B cells, granulocytes, macrophages, mesenchymal cells, osteoblasts, osteoclasts and multipotent adult progenitor cells, or combinations thereof.
13. The method of claim 11, wherein the decrease in the number of cells of the hematopoietic system is associated with chemo- and/or radiotherapy and/or removal of blood progenitor cells.
14. The method of claim 13, wherein the chemotherapy and/or radiotherapy and/or removal of blood progenitor cells is administered to treat cancer.
15. A method for protecting hematopoeitic cells from cell death in response to a chemotherapeutic and/or radiation comprising contacting said cells with a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof prior to contacting said cells with said chemotherapeutic agent and/or radiation.
16. A method for treating a cytotoxic agent-induced hematopoietic or myeloid toxicity in a human patient which comprises administering prior to, simultaneous with or subsequent to the administration of the cytotoxic agent, a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof in an amount effective to prevent, mitigate or reverse such hematopoietic or myeloid toxicity.
17. The method of claim 16, wherein said PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered in combination with one or more hematopoietic factors selected from the group consisting of Erythropoietin, G-CSF, GM- CSF, M-CSF, TCGF, flt-3, thrombopoietin, thymic stromal lymphopoietin, keratinocyte growth factor, PDGF, IGF, IL 1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-IO, IL-11, IL-12, IL-13, IL-15, IL-16, IL-17, stem cell factor (c-kit ligand), and M-CSF is administered in an amount effective to prevent, mitigate or reverse hematopoietic or myeloid toxicity.
18. The method of claim 17, wherein said one or more hematopoietic factors are administered prior to, concurrently with or subsequent to said administration of PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof.
19. A method of treating cancer therapy in a mammalian patient which comprises administering to the patient a combination of a cytotoxic agent in an amount effective to treat the cancer and PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof in an amount effective to prevent, mitigate or reverse hematopoietic or myeloid toxicity, wherein said PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered prior to, simultaneously with or subsequent to the administration of the cytotoxic agent, wherein administration of said PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof reduces the dose of said cytotoxic agent needed to produce a therapeutic effect in said patient.
20. The method of claim 19, wherein said cytotoxic agent is a therapeutic agent.
21. A method treating cancer therapy in a mammalian patient which comprises administering to the patient a combination of a cytotoxic agent in an amount effective to treat the cancer and a combination of PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof and at least one hematopoietic growth factor in an amount effective to prevent, mitigate or reverse hematopoietic or myeloid toxicity, wherein said PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered prior to the administration of the cytotoxic agent.
22. The method of claim 20, wherein said cytotoxic agent is radiation therapy, or chemotherapy.
23. The method of claim 19, wherein said mammalian patient is suffering from a cancer selected from the group consisting of Hodgkins lymphoma, Non-Hodgkins lymphoma, pancreatic cancer, melanoma, breast cancer, prostate cancer, bladder cancer, ovarian cancer, endometrial cancer, lung cancer, Kaposi's sarcoma, leukemia, lymphoma, gastric cancer, colon cancer, colorectal cancer, esophageal cancer, renal cancer, and head and neck cancer.
24. The method of claim 21, wherein said mammalian patient is suffering from a cancer selected from the group consisting of Hodgkins lymphoma, Non-Hodgkins lymphoma, pancreatic cancer, melanoma, breast cancer, prostate cancer, bladder cancer, ovarian cancer, endometrial cancer, lung cancer, Kaposi's sarcoma, leukemia, lymphoma, gastric cancer, colon cancer, colorectal cancer, esophageal cancer, renal cancer, and head and neck cancer.
25. The method of claim 21, wherein the hematopoietic growth factor is selected from the group consisting of Erythropoietin, G-CSF, GM-CSF, M-CSF, TCGF, flt-3, thrombopoietin, thymic stromal lymphopoietin, keratinocyte growth factor, PDGF, IGF, IL 1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-IO, IL-11, IL-12, IL-13; IL-15, IL-16, IL-17, stem cell factor (c-kit ligand), and M-CSF.
26. A method for preventing or mitigating myelosuppression in a human patient undergoing therapy for cancer or tissue or organ transplantation, which comprises administering to the patient a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof in an amount effective to treat myeloid toxicity, wherein said cytokine is administered prior to, simultaneously with or subsequent to said therapy.
27. A method for preventing or mitigating hematopoietic cell depression in a human patient undergoing therapy for cancer or tissue or organ transplantation which comprises administering to the patient a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof in an amount effective to treat myeloid toxicity, wherein said PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered prior to, simultaneously with or subsequent to the administration of said therapy.
28. A method for treating a cytotoxic agent-induced hematopoietic or myeloid toxicity in a human patient which comprises administering prior to, simultaneous with or subsequent to the administration of the therapeutic agent, PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof and a hematopoietic factor in an amount effective to prevent, mitigate or reverse such hematopoietic or myeloid toxicity.
29. The method of claim 28, wherein said cytotoxic agent is 1131, Sr89, Y90, ReI 86, Re 188, Cobalt 60; cesium 137; Iridium 192, and radium 226.
30. The method of claim 28, wherein said cytotoxic agent is conjugated to an antibody or a bone-seeking chemical.
31. The method of claim 30, wherein said bone-seeking chemical is orthophosphate or diphosphonate.
32. A method for alleviating a physical symptom caused by hematopoietic or myeloid toxicity in a human patient undergoing therapy with a cytotoxic agent, wherein the patient is administered prior to, simultaneous with or subsequent to the administration of the cytotoxic agent, a composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof in an amount effective to prevent, mitigate or reverse such hematopoietic or myeloid toxicity.
33. The method of claim 32, wherein the physical symptom is bone pain associated with bone cancer or bone metastasis.
34. The method of any of claims 1 to 33, wherein said subject is a cancer patient.
35. The method of claim 34, wherein said subject has a cancer selected from the group consisting of group consisting of Hodgkins lymphoma, Non-Hodgkins lymphoma, pancreatic cancer, melanoma, breast cancer, prostate cancer, bladder cancer, ovarian cancer, endometrial cancer, lung cancer, Kaposi's sarcoma, leukemia, lymphoma, gastric cancer, colon cancer, colorectal cancer, esophageal cancer, renal cancer, and head and neck cancer.
36. The method of any of claims 1 to 35, wherein said PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered intravenously, intraarterially, transdermal Iy, or intrathecally, or a combination thereof.
37. The method of any of claims 1 to 35, wherein the composition is formulated for administration via oral, topical, rectal, parenteral, local, intranasally, inhalant or intracerebral delivery, or a combination thereof.
38. The method of any of claims 1 to 35, wherein said PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered in a single dose.
39. The method of any of claims 1 to 35, wherein said PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered in multiple doses or as a continuous infusion during therapy.
40. The method of any of claims 1 to 35, wherein said PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered in a dose of about 0.5 μg/kg body weight to about lOμg /kg body weight of subject/day.
41. The method of claim 40, wherein said PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered on each of 2, 3, 4, 5, 6 or 7 days prior to the administration of chemotherapy or radiation therapy.
42. The method of any of claims 1 to 35, wherein said PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is administered in combination with one or more hematopoietic factors.
43. The method of claim 42, wherein hematopoietic factor is selected from the group consisting of Erythropoietin, G-CSF, peg-G-CSF, GM-CSF, M-CSF, TCGF, flt-3, thrombopoietin, thymic stromal lymphopoietin, keratinocyte growth factor, PDGF, IGF, IL 1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-I l, IL-12, IL-13; IL-15, IL-16, IL-17, stem cell factor (c-kit ligand).
44. The method of any of claim 1 to 35, wherein said composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof comprises PTHrP 1-34, or an analog thereof.
45. The method of any of claims 1 to 35 wherein said composition comprising PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof comprises semparatide.
46. The method of claim 43 wherein the hematopoietic factor is selected from G-CSF and peg-G-CSF.
47. The method of any of the foregoing claims wherein the subject is undergoing therapy with 5-fluorouracil.
48. The method of any of the foregoing claims wherein the PTHrP, a PTHrP fragment, a PTHrP analog, or a derivative thereof is semparatide administered to the subject for a period of 4-14 days prior to the administration of myelosuppressive levels chemotherapy or radiation therapy at a daily dose of 50-200 mg/kg.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US8109508P | 2008-07-16 | 2008-07-16 | |
US61/081,095 | 2008-07-16 |
Publications (2)
Publication Number | Publication Date |
---|---|
WO2010008561A2 true WO2010008561A2 (en) | 2010-01-21 |
WO2010008561A3 WO2010008561A3 (en) | 2011-10-06 |
Family
ID=41550914
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2009/004110 WO2010008561A2 (en) | 2008-07-16 | 2009-07-16 | Methods and compositions for protecting against cytotoxic therapy |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2010008561A2 (en) |
Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20030219366A1 (en) * | 2002-04-12 | 2003-11-27 | Horwitz E. Philip | Multicolumn selectivity inversion generator for production of ultrapure radionuclides |
US20070281889A1 (en) * | 2002-07-25 | 2007-12-06 | Scadden David T | Parathyroid hormone receptor activation and stem and progenitor cell expansion |
-
2009
- 2009-07-16 WO PCT/US2009/004110 patent/WO2010008561A2/en active Application Filing
Patent Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20030219366A1 (en) * | 2002-04-12 | 2003-11-27 | Horwitz E. Philip | Multicolumn selectivity inversion generator for production of ultrapure radionuclides |
US20070281889A1 (en) * | 2002-07-25 | 2007-12-06 | Scadden David T | Parathyroid hormone receptor activation and stem and progenitor cell expansion |
Also Published As
Publication number | Publication date |
---|---|
WO2010008561A3 (en) | 2011-10-06 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Robinson et al. | The potential and promise of IL-15 in immuno-oncogenic therapies | |
JP6905024B2 (en) | Manufacture and use of mono- and di-PEG IL10 | |
US20200283498A1 (en) | T cell receptor fusions and conjugates and methods of use thereof | |
Morstyn et al. | Treatment of chemotherapy-induced neutropenia by subcutaneously administered granulocyte colony-stimulating factor with optimization of dose and duration of therapy. | |
AU2010204552B2 (en) | New stable formulations of recombinant human albumin-human granulocyte colony stimulating factor fusion protein | |
CA2425842C (en) | Use of il-1 inhibitors and tnf antagonists, partially in combination with recombinant erythropoietins, for the treatment of anemia | |
US8545838B2 (en) | Compositions and methods for cancer immunotherapy | |
JP2005507870A (en) | Low toxicity interleukin-2 mutant | |
ZA200505681B (en) | Humanized antibody (H14.18) of the mouse 14.18 antibody binding to GD2 and its fusion with IL-2 | |
JP2012046552A (en) | Method for treating renal cell carcinoma | |
Rogers et al. | Recombinant human serum albumin fusion proteins and novel applications in drug delivery and therapy | |
US20230331858A1 (en) | Combination therapy using an il-2 receptor agonist and an immune checkpoint inhibitor | |
Castel et al. | Treatment of high-risk neuroblastoma with anti-GD2 antibodies | |
CN111944036B (en) | Mutant protein for proliferating immune cells | |
AU2019389807A1 (en) | Combined treatment of primary central nervous system lymphoma | |
CA2657946A1 (en) | Modified protein toxins targeting cytokine -receptor bearing cells and use thereof for treating disease | |
WO2010008561A2 (en) | Methods and compositions for protecting against cytotoxic therapy | |
KR20220110747A (en) | Methods of Treating Chemotherapy or Radiation Therapy Induced Neutropenia | |
KR20170123849A (en) | Fusion Protein Comprising CCL3 Variants And Use Thereof | |
US6440386B1 (en) | Stabilized radiophosphate-labeled proteins | |
Mortensen et al. | Site-Specific Chemical Modification of a Cytokine Mimic for Small Molecule-Based Tumor Targeting | |
Tong et al. | Site-specific pegylated IL2 mutein with biased IL2 receptor binding for cancer immunotherapy | |
Rokade et al. | IL-2 based cancer immunotherapies: an evolving paradigm | |
Hasan et al. | Biopharmaceutical Drug Products: An Overview of Physicochemical Properties and Their Modification | |
JP2024528917A (en) | Combination of IL-2/IL-15Rβγ agonists with antibody-drug conjugates for treating cancer - Patents.com |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 09798303 Country of ref document: EP Kind code of ref document: A2 |
|
NENP | Non-entry into the national phase in: |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 09798303 Country of ref document: EP Kind code of ref document: A2 |