WO2019232426A1 - Methods and compositions for the treatment of osteopetrosis - Google Patents

Methods and compositions for the treatment of osteopetrosis Download PDF

Info

Publication number
WO2019232426A1
WO2019232426A1 PCT/US2019/034984 US2019034984W WO2019232426A1 WO 2019232426 A1 WO2019232426 A1 WO 2019232426A1 US 2019034984 W US2019034984 W US 2019034984W WO 2019232426 A1 WO2019232426 A1 WO 2019232426A1
Authority
WO
WIPO (PCT)
Prior art keywords
mice
bone
cre
cells
subject
Prior art date
Application number
PCT/US2019/034984
Other languages
French (fr)
Inventor
Frederic Geissmann
James T. MULLER
Original Assignee
Memorial Sloan Kettering Cancer Center
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Memorial Sloan Kettering Cancer Center filed Critical Memorial Sloan Kettering Cancer Center
Priority to EP19809981.4A priority Critical patent/EP3801589A4/en
Priority to US17/058,830 priority patent/US20210205360A1/en
Publication of WO2019232426A1 publication Critical patent/WO2019232426A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/15Cells of the myeloid line, e.g. granulocytes, basophils, eosinophils, neutrophils, leucocytes, monocytes, macrophages or mast cells; Myeloid precursor cells; Antigen-presenting cells, e.g. dendritic cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4614Monocytes; Macrophages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/462Cellular immunotherapy characterized by the effect or the function of the cells
    • A61K39/4622Antigen presenting cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0645Macrophages, e.g. Kuepfer cells in the liver; Monocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0654Osteocytes, Osteoblasts, Odontocytes; Bones, Teeth
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6472Cysteine endopeptidases (3.4.22)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/31Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy

Definitions

  • the present technology generally relates to methods and compositions for treating, preventing, or ameliorating osteopetrosis. More specifically, the present technology relates to administering a composition comprising a therapeutically effective amount of engineered monocytic cells or wild-type monocytic cells from a healthy donor to a subject suffering from or at risk for osteopetrosis.
  • Osteoclasts are multinucleated giant cells that resorb bone, ensuring development and continuous remodeling of the skeleton and the bone marrow hematopoietic niche.
  • Osteopetrosis can be partially treated by bone marrow transplantation in human and mice, in accordance with osteoclasts hematopoietic origin, and studies suggesting that they develop by fusion of hematopoietic stem cell (HSC)-derived monocytic precursors in the presence of CSF1 and RANK-Ligand.
  • HSC hematopoietic stem cell
  • the present disclosure provides a method for treating or preventing osteopetrosis in a subject in need thereof, the method comprising administering a
  • composition comprising a therapeutically effective amount of monocytic cells from a healthy donor to the subject.
  • the subject is characterized by decreased expression of one or more of CA2 , CLCN7 , CTSK , CSF1R , IKBKG, ITGB3 , OSTM1 , PLEKHM1 , TCIRG1 , TNFRSF11A, and TNFSF11, as compared to the monocytic cells of the donor.
  • the osteopetrosis comprises one or more of stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia.
  • the composition is formulated for intravenous administration by injection, infusion, or transfusion.
  • the subject is a mammal.
  • the mammalian subject is a human.
  • the subject is characterized by a cathepsin K deficiency.
  • the present disclosure provides a method for treating or preventing osteopetrosis in a subject in need thereof, the method comprising administering to the subject a composition comprising a therapeutically effective amount of monocytic cells engineered to express one or more genes selected from CA2, CLCN7 , CTSK , CSF1R, IKBKG, ITGB3, OSTMl, PLEKHM1 , TCIRG1 , TNFRSF11A , and TNFSF11.
  • the osteopetrosis comprises one or more of stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia.
  • the composition is formulated for intravenous administration by injection, infusion, or transfusion.
  • the subject is a mammal.
  • the mammalian subject is a human.
  • the subject is characterized by a cathepsin K deficiency.
  • the monocytic cells are obtained from the subject.
  • the present disclosure provides a donor monocytic cell line engineered to express one or more genes selected from CA2 , CLCN7 , CTSK , CSF1R , IKBKG, ITGB3, OSTM1, PLEKHM1 , TCIRG1 , TNFRSF11A , and TNFSF11 , wherein the one or more genes is operably linked to a heterologous nucleic acid to form a chimeric nucleic acid construct.
  • the heterologous nucleic acid encodes a selectable marker.
  • the selectable marker is a bioluminescent protein, a fluorescent protein, a chemiluminescent protein, a xanthine-guanine phosphoribosyl transferase gene (gpt), or any combination thereof.
  • the heterologous nucleic acid encodes one or more control sequences suitable for directing expression of the one or more genes in a monocytic cell.
  • the one or more control sequences comprises a promoter.
  • the donor cells comprise a vector encoding one or more genes selected from CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1 ,
  • the vector is a mammalian expression vector, a lentiviral vector, or transposon vector.
  • FIGs 1A-1I HSC-derived precursors are dispensable for osteoclasts and bone development.
  • Figure 1A Top: Representative photographs of teeth of control littermates and Csflr Cre ; CsflrKA 3-4 week old mice (right) and controls and Csflr Cre ;Tnfrsfl lcfiF mice (left).
  • Bottom left Representative photographs of leg bones from controls and CsflC n ‘; Tnfrsflla 1111 mice, white arrowhead points to lack of blood cells.
  • Figure ID Osteoclast counts in femurs from Tlt3 ( r ; Tnfrsfl I a ,I , Flt3 Cre ;Cflr ⁇ ,
  • Figure IE Representative hematoxylin/TRAP staining of femur sections from 22 week old Flt3 Cre ;Tnfrsfl lcfP , 62 week old Vav° D/Cre ;CsflrFfl mice and littermates.
  • White arrowhead points to trabecular bone.
  • Figure 1G Expression and MFI of YFP in in TRAP + multinucleated cells from Csflr Cre ;Rosa26 LSL ⁇ YFP mice at embryonic day E16.5,
  • FIG. 2A-2J EMP derived osteoclasts are required for bone development.
  • Figure 2B Representative confocal microscopy pictures of frozen sections from Figure 1 A stained with fluorescent TRAP and TO-PRO-3.
  • Figure 2C Percent of TRAP+ cells expressing YFP in femur strom from E15.5
  • FIG. 2D Representative confocal microscopy picture from Figure 2C.
  • Figure 2E Representative photographs of teeth of control littermates (Cre ), Tnfrsfl I a Koh ⁇ ( n ‘;( "sflr 1111 and Tnfrsfl la Wask ⁇ Cre CsflrFfl mice (Cre + ).
  • Figure 21 Osteoclasts counts in bone sections from E18.5, P7, and 3-4 week old Infrsfl la ( re ;CsfI r l! 11 mice and littermate controls.
  • Data are mean ⁇ SD, circles on graphs represent individual mice, n indicates the number of mice per group.
  • Statistical significance was analysed with GraphPad Prism using unpaired two tailed t- tests. Significance was considered at p value (p) *p ⁇ 0.05; **p ⁇ 0.01; ***p ⁇ 0.001; ****p ⁇ 0.0001.
  • Figures 3A-3G In vivo dynamics of osteoclasts.
  • Figure 3A Parabiosis of
  • Csflr Cre ;Rosa26 LSL YFP mouse surgically paired with a Csflr Cre ;Rosa26 LSL tdTomato partner for 4 to 8 weeks.
  • Representative (n 3) confocal microscopy image of frozen sections from the femur of a Csflr Cre ;Rosa26 LSL ⁇ YFP partner stained with antibodies for tdTomato (red) and YFP (green), ELF97 (blue) and TOPRO-3 (grey).
  • Figure 3D Scatter plots represent the Mean Fluorescent Intensity (MFI) of individual TRAP+ MGC for YFP (Y axis) and tdTomato (X axis), and histograms represent the overlayed distribution of the MFI values for YFP and tdTomato in TRAP+ MGC at the indicated time-points.
  • Figure 3G A model for development and maintenance of osteoclast syncytia. Data are mean ⁇ SD, circles on graphs represent individual mice, n indicates the number of mice per group. Statistics: two-way ANOVA with Tukey’s multiple comparisons test *p ⁇ 0.05 **p ⁇ 0.005. [0013]
  • Figures 4A-G Rescue of Osteopetrosis.
  • Figure 4B Monocyte transfer:
  • histograms represent percentages of tdTomato+ cells among bone TRAP+ MGC from
  • Figure 4C Representative high power confocal microscopy image of the femur of a recipient mouse 60 days after i.v. transfer (from Figure 4B), stained with antibodies for tdTomato and YFP, and ELF97 and TOPRO-3.
  • Figure 4E Representative confocal microscopy image of femur from mice #3 in Figure 4D, stained with antibodies for YFP, ELF97 and TOPRO-3.
  • Figure 4G Histograms represent percentages of YFP+ cells among bone marrow precursors and blood leukocytes in the recipient mice (in Figure 4D) at the time of analysis.
  • FIGS 5A-G Csflr ( rc ; Trfrsfl la 1111 , Csflr ⁇ A ;Csflr Cre , and Tnfrsfl la Cre ; Csflr ⁇ A mice are osteopetrotic.
  • Figures 5A-B Representative Computed Tomography scans
  • FIG. 5D Representative photograph of inguinal lymph nodes in Csflr Cre ;Trfrsfl lcpF mice.
  • Figure 5E Representative photograph of Csflr Cre ;Trfrsfl lcfP and littermates controls.
  • Figure 5F Representative computed tomography scan
  • Figures 6A-N Bone histology and Flow cytometry analysis of bone marrow phenotypic KSL, LT-HSCs, ST-HSCs, andMPPs in mice of indicated genotypes.
  • Figure 6A Young Flt3 Clc ;Csf 1 r il/n and Flt3 Cre ; Tnfrsfl la fl/fl mice have normal long bone: H&TRAP staining of bone sections from 4 week old Flt3 Cre ;Tnfrsfl lcflJ 1 mice showing normal bone structure and bone marrow cavity.
  • Figure 6B LSK cell numbers in bone marrow
  • FIGS 6D, E Phenotypic long-term hematopoietic stem cells (LT-HSCs) are reduced in aged but not old ( Figures 6F, G) LT-HSC are reduced in in young Csfl r /_ and to a lesser extent in young Tnfrsf l 1 y lV sklC : Csfl 1 Numbers per 2 femur are shown.
  • Figure 61 H&TRAP staining of bone sections from P7
  • LT-HSC Lin- Kit+ Sca-l+; Kit+ Scal+ CD34- Flt3-.
  • Phenotypic short-term hematopoietic stem cells ST-HSC: Lin- Kit+ Sca-l+ CD34+ Flt3-.
  • Multipotent progenitors (MPP) Lin- Kit+ Sca-l+ CD34+ Flt3+.
  • Lin CD3 CD19 NK1.1 Terl l9 CDl lb Grl B220.
  • FIGS 7A-E Bone histomorphometry in old Flt3 Cre ;Tnfrsfl lc ⁇ , FltS ⁇ CsflrFfl, and Vav Cre CsflrFflmice and control littermates.
  • Figure 7A Representative Micro CT pictures , genotype is indicated.
  • Figures 7D, E Dynamic bone histomorphometry in aged Flt3 Cre ;Tnfrsf l 1 a il/n and Flt3 Cre ;Csf 1 r il/n mice using in vivo calcein labeling.
  • Representative micrographs of calcein labeling (green) of femura of mice from the indicated genotypes and ages Figure 7D). Scale bars represent 200pm (top) and 50pm (bottom).
  • FIGS 8A-E Colonization of the bone marrow by Csflr+ and Flt3+
  • Figure 8B YFP labeling efficiency in Flt3 Cre ;Rosa26 LSL ⁇ YFP mice analyzed by flow cytometry in the indicated cell populations, and by confocal microscopy on frozen bone sections at the indicated time (Right). Insets show YFP expression on individual osteoclasts.
  • YFP anti-GFP antibody
  • TRAP ELF97 fluorescent substrate
  • TO-PRO-3 nuclear stain.
  • Figure 8C YFP labeling efficiency in Csflr Cre ;Rosa26 LSL ⁇ YFP mice analyzed as in Figure 8A.
  • Data from Figures 8A, B are representative of at least 3 experiments by time point and genotypes. Circles represent individual mice. Genetic lineage tracing of osteoclasts in ossification centers using Csflr MenCreMer ; Rosa26 LSL YFP . Representative high power confocal microscopy images from embryonic femurs showing MGCs in primary ossification centers from Csflr MenCreMer ;Rosa26 LSL YFP .
  • FIGS 9A-D Tnfrs 1 la (wask)Cre knock-in mice allow deletion of target genes in fetal macrophages, but not definitive HSC and their progeny in blood and tissues, while Fav cre mice allow deletion of target genes in definitive HSC, but not fetal macrophages .
  • Figure 9A Bar graphs indicate percentages of eYFP expression by flow cytometry in cells from
  • polymorphonuclear cells mono: monocytes, T: T cells, B: B cells, PEC: peritoneal exudate cells.
  • Data are mean ⁇ SD, circles on graphs represent individual mice, n indicates the number of mice per group.
  • FIGS 10A-F Bone morphometric and dynamic histomorphometry effects of the deletion of Csflr in P21 Tnfrsfl la Cre ;CsflHfl mice . Bone volume / total vlume (BV/TV,
  • FIG 10A Bone length ( Figure 10B), connectivity density (Conn density, Figure 10C), and Trabecular Number (Tb.N., Figure 10D) was analyzed by microCT in 21 day/old mice.
  • Figures 11A-C EdU labeling of bone marrow myeloid cells.
  • FIG. 11B Long term kinetics: (l-240hrs) EdU is injected as in Figure 11 A and % of EdU+ monocytic cells in bone marrow (top) and blood (bottom) is determined by flow cytometry, showing that labeling of -50% of monocytic cells is observed for -2 days. Labeled cells are not detectable after 3 days. Circles represent determination from individual mice, data for each time point are pooled from 2-3 independent experiments see source data.
  • Figure 11C Parabiosis between Csflr Cre ;Rosa26 LSL ⁇ YFP and
  • Scatter plots represent the Mean Fluorescent Intensity (MFI) of individual TRAP + MGC for YFP (Y axis) and tdTomato (X axis), and histograms represent the overlaid distribution of the MFI values for YFP and tdTomato in TRAP + MGC at the indicated time-points.
  • Data are mean ⁇ SD, circles on graphs represent individual mice, n indicates the number of mice per group.
  • FIGS 12A-C FACS analysis of monocyte purification, and blood / bone marrow from transferred Csflr Cre ; Csflr 1111 mice.
  • Figure 12A Representative flow cytometry plots of purified bone marrow monocytes from magnetic bead based enrichment (MACS), percentage of live YFP+ monocytes is indicated. Representative flow cytometry plots from blood
  • Figure 12B and bone marrow (Figure 12C) of 14 day old mice transferred with 1E6 YFP+ monocytes on day 5,8,11, the percentage of YFP+ cells is indicated.
  • Figure 13 Rescue of osteoclasts by monocyte transfer in Csflr Cre ; Csfld y T mice. High power confocal microscopy images of frozen sections from Csflr Cre ; Csflr ⁇ T mice transferred with monocytes from Csflr Cre Rosa26 LSL ⁇ YFP and controls, stained with antibodies for YFP, TRAP substrate ELF97 and TOPRO-3 as a nuclear stain. Examples of
  • n 3 mice from independent litters. Numbers (#1,2,3) correspond to the mice in Figure 4D.
  • Figures 14A-F Csflr deletion in Tnfrsfl la Cre Csflr F/F mice results in lack of tissue macrophages and osteoclasts at birth, while definitive HSC and their progeny are present in the fetal liver and blood.
  • Figure 14A Flow cytometry analysis of F4/80+ cells in brain (microglia) and epidermis (Langerhans cells) in El 8.5 Tnfrsfl la Cre+ ;Csflr F/F embryos and littermate controls.
  • Figure 14B H&TRAP staining of bone sections from P7
  • FIG. 14C Flow cytometry plot of Fetal liver at E15.5.
  • Figure 14D LSK numbers in bone marrow of 3-4 week old Tnfrsfl la Cre+; Csflr F/F and littermate controls.
  • Figure 14E For comparison, LSK numbers in bone marrow of 3-4 week old Csflr A and littermate controls.
  • Figure 14F Blood leukocytes numbers in 4 week old
  • Tnfrsfl I a ( n ' Csflr 1 mice and littermate controls. Circles represent individual mice, results from 3 independent experiments.
  • Tnfrsfl la Cre+ ;Csflr F/ ⁇ or F/F mice were analyzed by microCT in 21 day/old mice of indicated genotype.
  • Figures 16A-B EdU labeling of bone marrow myeloid cells.
  • the term“about” and the use of ranges in general, whether or not qualified by the term about means that the number comprehended is not limited to the exact number set forth herein, and is intended to refer to ranges substantially within the quoted range while not departing from the scope of the invention.
  • “about” will be understood by persons of ordinary skill in the art and will vary to some extent on the context in which it is used. If there are uses of the term which are not clear to persons of ordinary skill in the art given the context in which it is used,“about” will mean up to plus or minus 10% of the particular term.
  • A“chimeric nucleic acid” comprises a coding sequence or fragment thereof linked to a nucleotide sequence that is different from the nucleotide sequence with which it is associated in cells in which the coding sequence occurs naturally.
  • the terms“effective amount,” or“therapeutically effective amount,” and“pharmaceutically effective amount” refer to a quantity sufficient to achieve a desired therapeutic and/or prophylactic effect, e.g ., an amount which results in the prevention of a disease, condition, and/or symptom(s) thereof.
  • the amount of a composition administered to the subject will depend on the type and severity of the disease and on the characteristics of the individual, such as general health, age, sex, body weight, and tolerance to the composition drugs. It will also depend on the degree, severity, and type of disease or condition. The skilled artisan will be able to determine appropriate dosages depending on these and other factors. In some embodiments, multiple doses are administered. Additionally or alternatively, in some embodiments, multiple therapeutic compositions or compounds are administered.
  • compositions comprising the monocytic cells of the present technology may be administered to a subject having one or more signs, symptoms, or risk factors of osteopetrosis, including, but not limited to stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia.
  • signs, symptoms, or risk factors of osteopetrosis including, but not limited to stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morph
  • a“therapeutically effective amount” of the compositions of the present technology includes levels at which the presence, frequency, or severity of one or more signs, symptoms, or risk factors of osteopetrosis are, at a minimum, ameliorated.
  • a therapeutically effective amount reduces or ameliorates the physiological effects of osteopetrosis, and/or the risk factors of osteopetrosis, and/or the likelihood of developing osteotpetrosis.
  • a therapeutically effective amount is achieved by multiple administrations.
  • a therapeutically effective amount is achieved with a single administration.
  • engineered is used herein to refer to a cell or organism that has been manipulated to be genetically altered, modified, or changed, e.g., by disruption of the genome.
  • an“engineered monocytic cell” refers to a monocytic cell that has been manipulated to be genetically altered, modified, or changed.
  • an engineered monocytic cell refers to a monocytic cell that has been transduced with a lentivirus designed to express a nucleotide sequence of interest, e.g, a cDNA coding for the wild type allele of any one or more of CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1, PLEKHM1 , TCIRG1 , TNFRSF11A , or TNFSF11 under a strong promoter.
  • a nucleotide sequence of interest e.g, a cDNA coding for the wild type allele of any one or more of CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1, PLEKHM1 , TCIRG1 , TNFRSF11A , or TNFSF11 under a strong promoter.
  • Heterologous nucleic acid refers to a nucleic acid, DNA, or RNA, which has been introduced into a cell (or the cell’s ancestor), and which is not a copy of a sequence naturally found in the cell into which it is introduced.
  • Such heterologous nucleic acid may comprise segments that are a copy of a sequence that is naturally found in the cell into which it has been introduced, or fragments thereof.
  • “prevention,”“prevent,” or“preventing” of a disorder or condition refers to, in a statistical sample, reduction in the occurrence or recurrence of the disorder or condition in treated subjects/samples relative to an untreated controls, or refers delays the onset of one or more symptoms of the disorder or condition relative to the untreated controls.
  • “subject” and“patient” are used interchangeably and refer to a mammalian subject.
  • “subject” means any animal (mammalian, human, or other) patient that can be afflicted with osteopetrosis and when thus afflicted is in need of treatment.
  • the subject is a human.
  • Treating,”“treat,”“treated,” or“treatment” of a disease or disorder includes: (i) inhibiting the disease or disorder, z.e., arresting its development; (ii) relieving the disease or disorder, z.e., causing its regression; (iii) slowing progression of the disorder; and/or (iv) inhibiting, relieving, or slowing progression of one or more symptoms of the disease or disorder.
  • compositions and methods for the treatment of osteopetrosis identify: (i) the developmental origin of osteoclasts, the cells that shape bone architecture; (ii) a mechanism that controls the maintenance of osteoclast function after birth; and (iii) a novel strategy to treat osteopetrosis and to modulate osteoclast activity in vivo.
  • the data described herein demonstrate that parabiosis or transfusion of monocytic cells results in long-term gene transfer in osteoclasts in the absence of HSC chimerism and can rescue an adult-onset osteopetrotic phenotype caused by cathepsin-K deficiency. Transfusion of monocytic cells is also sufficient to rescue bone development in early-onset autosomal recessive osteopetrosis in newborn mice.
  • Osteopetroses are a heterogeneous group of genetic disorders characterized by increased bone density due to impaired bone resorption by osteoclasts. The increased bone density places the affected individual at an increased risk for bone fracture. Normally, bone growth is a balance between osteoblasts (cells that create bone tissue) and osteoclasts (cells that destroy bone tissue). Individuals with osteopetrosis have a deficiency of osteoclasts, resulting in too little bone resorption and too much bone creation. The types of osteopetrosis are distinguished based on their pattern of inheritance: autosomal dominant, autosomal recessive, or X-linked.
  • the signs and symptoms of osteopetrosis may vary depending on the type of the disease, and mild forms of osteopetrosis may be asymptomatic.
  • the typical signs and symptoms of osteopetrosis include: stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia.
  • ADO autosomal dominant osteopetrosis
  • Albers-Schonberg disease autosomal dominant osteopetrosis
  • ARO Autsomal recessive osteopetrosis
  • ARO is often characterized by one or more of a high risk of bone fracture resulting from minor bumps or falls, pinched nerves in the head and face, impaired bone marrow function, slow growth, short stature, dental abnormalities, hepatosplenomegaly, intellectual disability, and epilepsy.
  • IAO intermediate autosomal osteopetrosis
  • a form of osteopetrosis that can have either an autosomal dominant or recessive pattern or inheritance may be characterized by one or more of a high risk of bone fracture and anemia, calcifications in the brain, intellectual disability, and renal tubular acidosis.
  • Individuals with the X-linked pattern of inheritance may be characterized by one or more of lymphedema, anhidrotic ectodermal dysplasia, and immunodeficiency.
  • a method for treating or preventing osteopetrosis in a subject in need thereof comprising administering a composition comprising a therapeutically effective amount of monocytic cells from a healthy donor to the subject.
  • the subject is characterized by decreased expression of one or more genes implicated in or potentially underlying the development of osteopetrosis, such as CA2 , CLCN7 , CTSK, CSF1R , IKBKG, ITGB3, OSIMI, PLEKHM1 , TCIRG1 , TNFRSF11A , and TNFSF11.
  • the subject is characterized by a cathepsin K deficiency.
  • a method for treating or preventing osteopetrosis in a subject in need thereof comprising administering to the subject a composition comprising a therapeutically effective amount of monocytic cells engineered to express one or more genes implicated in or potentially underlying the development of osteopetrosis, such as CA2 , CLCN7 , CTSK, CSF1R , IKBKG, ITGB3, OSIMI, PLEKHM1 , TCIRG1 ,
  • TNFPSF11A TNFPSF11A
  • TNFSF11 TNFSF11 is provided.
  • monocytic cells used for treatment are genetically modified to correct a genetic abnormality or to improve or changed cellular functioning according to known genetic engineering protocols.
  • a method of treating or preventing osteopetrosis in a subject comprises: (a) obtaining a sample of monocytic cells from the subject; (b) genetically correcting one or more mutations in the monocytic cells, (c) culturing the monocytic cells; and (d) providing the corrected monocytic cells to the subject.
  • a method for treating or preventing osteopetrosis comprises: (a) obtaining a sample of monocytic cells from the subject; (b) genetically engineering the cells to express a nucleotide sequence coding for the wild type allele of any one or more of CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3 , OSTM1 , PLEKHM1 , TCIRG1 , TNFRSF11A, and TNFSF11 with a suitable transducing vector, such as a lentiviral vector; (c) culturing the engineered monocytic cells under conditions sufficient to express the nucleotide sequence; (d) removing the viral particles from the engineered monocytic cells; and (e) providing the engineered monocytic cells to the subject.
  • a suitable transducing vector such as a lentiviral vector
  • the transducing vector encoding the one or more of CA2 , CLCN7, CTSK , CSF1R , IKBKG, ITGB3, OSTM1, PLEKHM1 , TCIRG1 , TNFRSF11A , and TNFSF11 is a mammalian expression vector.
  • the mammalian expression vector is a lentiviral vector or transposon vector.
  • Subjects suffering from osteopetrosis can be identified by any or a combination of diagnostic or prognostic assays known in the art.
  • typical symptoms of osteopetrosis include, but are not limited to, stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia.
  • a composition comprising a therapeutically effective amount of monocytic cells from a health donor and/or monocytic cells engineered to express one or more genes selected from CA2, CLCN7, CTSK , CSF1R, IKBKG, ITGB3 , OSTM1 , PLEKHM1 , TCIRG1 , TNFRSF11A, and TNFSF11 is administered to the subject.
  • the composition is administered according to any acceptable transfusion regimen.
  • the composition is administered one, two, three, four, or five times per day.
  • the composition is administered more than five times per day. Additionally or alternatively, in some embodiments, the composition is
  • the composition is administered every day, every other day, every third day, every fourth day, every fifth day, or every sixth day.
  • the composition is administered weekly, bi-weekly, tri-weekly, or monthly.
  • the composition is administered for a period of one, two, three, four, or five weeks.
  • the composition is
  • the composition is administered for six weeks or more. In some embodiments, the composition is administered for twelve weeks or more. In some embodiments, the composition is administered for a period of less than one year. In some embodiments, the composition is administered for a period of more than one year or until a desired therapeutic outcome is observed in the subject.
  • treatment of subjects diagnosed with or suspected of having osteopetrosis with one or more compositions of the present technology ameliorates or eliminates one or more of the following symptoms of osteopetrosis: stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections,
  • hepatosplenomegaly facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia.
  • the present technology provides a method for preventing or delaying the onset of osteopetrosis or one or more symptoms of osteopetrosis in a subject at risk of having or developing osteopetrosis.
  • compositions of the present technology are administered to a subject susceptible to, or otherwise at risk of for
  • osteopetrosis in an amount sufficient to eliminate or reduce the risk, or delay the onset of the disease, including biochemical, histologic and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease.
  • Administration of a prophylactic compositions can occur prior to the manifestation of symptoms characteristic of the disease or disorder, such that the disease or disorder is prevented or, alternatively, delayed in its progression.
  • mice were bred and kept under specific pathogen conditions in separated ventilated cages in the animal facility of MSKCC and the Medical Theoretical Center of the TU Dresden. All experiments with osteopetrotic mice that lack teeth were performed with mice maximal 4 weeks of age that were kept with the lactating mother or provided with DietGel 76A (Clear H 2 0, 72-07-5022) to avoid secondary effects from malnutrition. Experiments were performed in adherence to the Institutional Review Board (IACUC 15-04-006) from MSKCC and Austindiretation Dresden and were in compliance with relevant ethical regulations. Mice greater than 7 days old were sacrificed by cervical dislocation (TU Dresden), CO2 asphyxiation or anesthesia (MSKCC). To harvest embryos, pregnant females were sacrificed and embryos were collected by postmortem cesarean from the uterus and exsanguinated through decapitation in cold PBS (Fisher, 14190).
  • Genotyping PCR genotyping was performed according to protocols described previously and indicated in Table 1. The investigators were not blinded to allocation during experiments and outcome assessment.
  • Csflr U n( reUer female mice were crossed with male Rosa26 LSL ⁇ YFP/LSL ⁇ YFP mice. Cre recombination in Csflr MenCreMer ;Rosa26 LSL ⁇ YFP embryos at embryonic day E8.5 was induced with a single dose of 4-hydroxytamoxifen (4-OHT) injected intraperitoneally in pregnant mothers at a dose of 75pg/g of body weight supplemented with 37.5 pg/g of progesterone as previously described.
  • 4-hydroxytamoxifen (4-OHT) 4-hydroxytamoxifen
  • Cre-mediated recombination in Rosa.26-CreER n tamoxifen was introduced to pregnant mother by a single TAM gavage (5mg) at El 0.5, supplemented with progesterone (37.5pg/g body weight resolved in Sunflower seed oil, Sigma-Aldrich) was injected i.p. directly after gavage.
  • progesterone 37.5pg/g body weight resolved in Sunflower seed oil, Sigma-Aldrich
  • Csflr ,Cre ;Rosa26 LSL ⁇ tdTomato females were used for parabiosis.
  • Parabionts were kept on Sulfamethoxazole/Trimethoprim (Sulfatrim) diet for up to 8 weeks. Ex-parabionts were separated after 4 weeks for cellular complementation and 6 weeks for rescue of CathepsinK activity.
  • Surgical procedure, pre-operative procedure weight-matched female partner mice for parabiosis were caged together few days before surgery. One day before the surgery the fur from lateral sides of mice was carefully removed with a trimmer followed by depilatory cream (for 3 minutes) at the site of surgery, excess of fur was removed with a moist gauze pad. Left side partner is shaved on the right side and vice versa. This procedure was performed under Isofluorane inhalation anesthesia. Mice were fed with food supplemented with Sulfatrim ad libitum one day prior to surgery.
  • mice were anesthetized intraperitoneally with l50mg/kg of ketamine and l5mg/kg of xylazine.
  • Sterile eye lubricant (Paralube Vet Ointment, 17033-211-38) was applied to both eyes to prevent corneal drying during surgery.
  • mice were placed in a supine position on a surgical tray with heat support provided by a heating pad. The surgical site was cleaned 3 times with cotton swabs soaked in povidone-iodine (Betadine) then with 70% ethanol.
  • anesthetic agent bupivacaine (Marcaine 0.25-0.5% solution) was applied locally. Surgery was performed by a longitudinal skin incision on the lateral side of mice, approximately 0.5cm above the elbow to 0.5cm below the knee joint. Mice were laid side-by-side in close contact and the ligaments of the two knees and elbows were sutured together using monofilament non-absorbable suture. Then, the skin incisions was closed by apposing and clipping skin to skin of the pair with 9 mm wound clips.
  • mice were anesthetized and prepared for surgery as indicated above. Mice were separated at the site of parabiosis junction. Using scissors, the skin joining both mice is cut longitudinally. The sutures around the elbows and knees are cut and removed. The resulting wound is closed with 9 mm wound clips. Mice were injected subcutaneously with 2 mg/kg of meloxicam and 0.5mg/kg of buprenorphine and for a maximum of 48 hours postoperatively. Wound clips were removed 14 days after surgery.
  • EdU pulse labeling 12 week old C57BL/6N mice (Charles River) were injected intraperitoneally with 25 pg/g of a 2.5mg/mL solution of EdU prepared extemporaneously (Fisher Cl 0420, Click-iT EdU Alexa Fluor 488 Flow Cytometry Assay Kit). Blood, bone marrow, and bone samples were collected at 60hrs and 72hrs post injection and bone were prepared for histology of frozen sections as indicated above for adult mice.
  • Sections are incubated for 45 minutes at room temperature with lOOpL of Click-iT EdU reaction buffer then washed with PBS and stain for fluorescent TRAP (ELF 97) and TO-PRO-3 (nuclear stain). Mounting media was 75% glycerol in PBS.
  • Microscopy Images were acquired using an inverted Zeiss LSM880 laser scanning confocal microscope. Histological sections of 15pm thickness were tile-scanned at l.Opm Z stacks.
  • Bone marrow monocytes were isolated from 12-16 weeks old donor.
  • lxlO 6 total cells from Csflr ,Cre ;Rosa26 LSL tdTomato mice were transferred at day 0, day 3 and day 6 by retro-orbital injections into recipient
  • BM Monocyte Isolation Kit
  • MCS Miltenyi Biotec 130-100-629
  • Cell numbers of Ly6C + cells were calculated by determining the cell number / ml using a Neubauer chamber in combination with a staining for Ly6C analyzed by flow cytometry. Bone samples were dissected and prepared for frozen sections as described above and stained for anti-GFP, anti-RFP, fluorescent TRAP and TO- PRO-3. The percentages of YFP + or Tomato + multinuclear cells was quantified in femurs of recipients.
  • Napthol-Elher substrate solution 2% (w/v) Napthol AS-B1 Phosphate (Sigma, 70482) in 2-Ethoxy ethanol (Sigma, 256374).
  • Sodium nitrate solution 4% (w/v) Sodium nitrate (Sigma, 237213) in water.
  • Basic fuchsin solution 5% (w/v) basic fuchsin dye (Sigma, 857343) in 2N HCL (Fisher, A144-500).
  • TRAP + multinuclear cells (more than 3 nuclei per cell), associated to bone tissue, were quantified in ImageJ using the Cell Counter plugin. Numerical values were plotted using GraphPad Prism. For Static and dynamic histomorphometry. Young and aged mice were injected (i.p.) twice with 15 mg/kg body weight Calcein (Sigma) dissolved in 1.4%
  • Tartrate- resistant acid phosphatase staining was used to assess the osteoclast surface per bone surface (Oc.S/BS) and number of osteoclasts per bone surface (N.Oc/BS). Bone sections were analyzed using the Osteomeasure software (Osteometries, USA) following international standards.
  • Sections were washed 2 times with PBS for 5 minutes. Sections stained with anti-GFP biotin antibody were first incubated with Biotin/Streptavidin blocking kit (Vector laboratories, SP2002). Streptavidin blocking solution is prepared by adding 4 drops of streptavidin solution to lmL of PBS/0.25% BSA, samples were incubated for 15 min, then washed once with PBS for 5 min. Biotin blocking solution is prepared by adding 4 drops of biotin solution to lmL of PBS/0.25% BSA, samples were incubated for 15 min then washed once with PBS for 5 min. Primary and secondary antibodies used are listed in Table 2. Sections were also stained with fluorescent TRAP and nuclear stain.
  • Fate-mapping models FU3 Cre ;Rosa26 LSL YFP , Cfslr Cre ;Rosa26 LSL YFP Csfl r MeriCreMer ;Rosa26 LSL ⁇ YFP and Tnfrsflla Cre ;Rosa26 LSL YFP
  • Fluorescent TRAP staining Sections were prepared for fluorescent TRAP by incubating with TRAP incubation solution (112 mM sodium acetate, 76 mM sodium tartate, and 11 mM sodium nitrite, pH 4.1-4.3) at room temperature for 10 minutes. Buffer was removed and incubated with ELF97 substrate (Molecular Probes E6589, 2mM) at a concentration of 125mM in TRAP incubation solution for 15 min under ETV light and washed 2 times with PBS for 5 minutes. Nuclear stain used was TO-PRO-3 Iodide (Fisher T3605) 1 :4000 in PBS for 5 minutes. Mounting media was 75% glycerol in PBS.
  • mice anesthetized under Isofluorane anesthesia and placed on an imaging table containing an animal bed equipped with a nosecone for gas inhalation and body temperature stabilization.
  • a mouse bed was used and for mice 8 weeks and older a rat bed was used.
  • Whole-body imaging of mice was acquired using a NanoSPECT/CT scanner (Mediso) for non-invasive and longitudinal monitoring of the 3D skeletal structure.
  • Each CT scan averaged 15 minutes and was acquired with an exposure time of 1, 000ms and 240 projections set at a pitch of 1 degree.
  • the tube energy of the X-ray was 55KVp and 145mA.
  • the in-plane voxel size was medium generating a voxel size of 147pnr ⁇ Reconstructed images were analyzed using In Vivo Scope 2.0 (Bioscan, Inc.) software.
  • microCT For tri-dimensional X-ray imaging by micro computed tomography, mice were sacrificed and bones placed in 70% ethanol until scanning. Bone microarchitecture was analyzed using the vivaCT40 (Scanco Medical, Switzerland). Entire femora or humeri were imaged at a resolution of 10.5pm (1 slice) with an X-ray energy of 70 kVp, 114 mA, and an integration time of 200ms. The machine was routinely calibrated using hydroxyapatite phantoms for density and geometry. Trabecular bone in femora or humeri from old mice was assessed in the metaphysis 20 slices below the growth plate using 150 slices. The trabecular region within the cortical bones (P21 mice) was determined in the femoral midshaft (100 slices up, 100 slices down). Pre-defmed scripts from Scanco were used for the evaluation.
  • Blood was collected from anesthetized mice by retro- orbital venous sinus bleeding or cardiac puncture using a lmL syringe and a 26G needle rinsed with lOOmM EDTA (sigma E4884).
  • the collected blood was lysed with 3 mL of red blood cell lysis buffer (l55mM NFECl sigma A9434, lOmM NaFlCCh sigma S5761 and O. lmM EDTA sigma E4884) for 5 mins, washed with lOmL of FACS buffer (PBS, 0.5%
  • Bone marrow cells were harvested by crushing or flushing femurs and tibias using a syringe and a 26G needle with lOmL of PBS / 5% FCS or RPMI supplemented with 10% FBS. Bone marrow was dissociated by gently pipetting up and down with a lmL or lOmL pipette.
  • Spleens were gently dispersed between frosted slides and digested for 30 min at 37 °C in PBS with 5% FCS containing Collagenase Type 4 (Worthington) at a final concentration of 4.2 El/ml and 100 pg/ml DNAsel (Sigma- Aldrich). The reaction was stopped by incubation with 12.5 mM EDTA. Peritoneal cells were harvested by flushing the peritoneal cavity with lOmL PBS/5% FCS heated to 37°C.
  • the cell pellets were resuspended in FACS buffer containing purified anti-mouse CD16/32 antibody (1 : 100, Biolegend 101301), 5% normal mouse (Fisher, 015-000-120), 5% normal rat (Fisher 012-000-120) and 5% normal rabbit serum (Fisher, 011-000-120) and incubated for 10 minutes on ice or directly stained using blocking antibodies in the staining mixtures.
  • Samples were immunostained with fluorochrome-conjugated antibodies for 30 minutes on ice, and analyzed by flow cytometry using an LSRFortessa or an LSR II (BD-Bioscience). Full list of antibodies for flow cytometry is provided in Table 3. Each sample was stained with Hoechst (ThermoFisher Scientific, Hoechst 33258, 1 pg/ml) or DAPI (Applichem, l pg/25ml) moments prior to flow cytometry acquisition.
  • Hoechst ThermoFisher
  • BM Bone marrow
  • SP Spleen
  • BL Blood
  • Y Yolk sac
  • FL Fetal Liver
  • LV Liver
  • LN Lung
  • BR Brain
  • P PEC: E: Epidermis
  • K Kidney.
  • osteoclasts arise by fusion of HSC-derived precursors and require expression of Csflr and Tnfrsfl la (Rank). To probe the origin of osteoclasts in vivo ,
  • Csflr Cre Csflr 1111 ; and Csfif rc ; Tnfrsfl la 1111 mice were generated. These mice presented with an osteopetrotic phenotype similar to Csfl (op/op), Csflr , and Tnfrsfl la mutants and characterized in young mice by lack of teeth eruption, skull and skeletal deformities with shortness of long bones, increased bone density, and lack of osteoclasts and hematopoietic cells (Figure 1A; Figure 5B).
  • Vav Cre CsflrFfl mice were not osteopetrotic at birth, had normal teeth (Figure IB), and hematopoietic cell numbers in long bones at 4 week of age (Figure 1C). However, they developed a late-onset osteopetrosis similar to Flt3 Cre ;Tnfrsfl lcfT and Flt3 Cre CsflrH 1 mice: at ⁇ 60 weeks of age Vav ( n ';Csf I r /! 11 mice had decreased osteoclasts and hematopoietic cell numbers in long bones ( Figure 1C-D), enlarged trabecular bone ( Figure IE) and increased bone mass ( Figure IF, Figure 7A-E).
  • multinucleated cells are labeled with YFP in Csfh Mer ⁇ lCre ⁇ Mer ; Rosa26 LSL ⁇ YFP mice pulsed at E8.5 with a single dose of hydroxytamoxifen (4-OHT), which labels EMP but not HSC
  • Tnfrsflla is expressed by osteoclasts, but its expression is also a hallmark of EMP-derived pMacs that colonize the developing embryo.
  • Cre-mediated expression of a Rosa26 LSL ⁇ YFP fluorescent reporter is achieved with high efficiency in fetal macrophages but with low efficiency or not at all in HSC and their progeny in blood and tissues (Figure 9A-D).
  • Tnfrsflla Cre Tnfrsfl la Cre ;CsflrFA mice.
  • Tnfrsfl la Cre ;CsflrFfl lack tissue macrophages such as brain microglia and epidermal
  • HSC-derived osteoclasts are important for the maintenance of bone mass after birth and later in life, although they may partially rescue bone development in the absence of EMP-derived osteoclasts in
  • a prediction from this model is that osteopetrosis due to a recessive mutation affecting osteoclasts function may be rescued or prevented through parabiosis with a wild- type partner.
  • Parabiosis experiments between 4 week-old cathepsin K deficient mice, which develop an adult-onset form of osteopetrosis known as pycnodysostosis, and cathepsin K +/ or cathepsin K littermates and between wild type mice as control showed a reduction of bone volume in 10 weeks old cathepsin K mice paired with cathepsin K +/ littermates (Figure 4A), suggesting that circulating blood cells carrying a wild type cathepsin K allele are sufficient to reduce bone density.
  • Example 3 Ex vivo gene therapy for the treatment of a form of osteopetrosis: pycnodvsostosis
  • This example demonstrates the prophetic use of monocytic cells obtained from a subject and engineered to express a gene, such as CTSK , for the treatment of osteopetrosis in the subject.
  • Pycnodysostosis is an autosomal recessive form of osteopetrosis, due to loss of function mutations in the cathepsin K gene (CTSK), compatible with life, and characterized by short stature, deformity of the skull maxilla and phalanges, increased density of the bones, osteosclerosis, and fragility of bone. There is no treatment for this disease as the relatively comparably milder symptoms do not qualify for bone marrow transplantation.
  • CTSK cathepsin K gene
  • Subjects suspected of having or diagnosed as having cathepsin K deficiency will be selected for a gene therapy trial and will undergo GMP grade collection of peripheral blood monocytes by apheresis and/or elutriation. Purified monocytes will then be transduced with a lentivirus designed to express a cDNA coding for the wild type allele of CTSK under a strong promoter (e.g ., CMV). Transduced monocytes will then be washed to remove contaminating viral particles and rested for 12 hours followed by reinfusion to the patient. Assuming -10 9 monocytes can be collected, the number of cells transferred per kilogram of body weight would be >l0 7 . Following the auto-transfusion, the subject will be monitored at regular intervals for clinical and radiological signs of pycnodysostosis in order to determine the efficiency of the procedure and the frequency with which it should be repeated.
  • a strong promoter e.g ., CMV
  • pycnodysostosis and receiving therapeutically effective amounts of monocytic cells engineered to express CTSK will display reduced severity or elimination of one or more symptoms associated with pycnodysostosis. Accordingly, these results will show that transfusion of monocytic cells engineered to express the wild-type version of one or more genes implicated in osteopetrosis is useful in methods for treating subjects in need thereof for the treatment of osteopetrosis.
  • Example 4 Ex vivo gene therapy for the treatment of a form of osteopetrosis: severe infantile autosomal recessive osteopetrosis
  • This example demonstrates the prophetic use of monocytic cells obtained from a subject and engineered to express a gene, such as TCIRG1, TNFRSFlla, CA2, CLCN7, OSTM1, and/or PLEKHM1, for the treatment of osteopetrosis in the subject.
  • a gene such as TCIRG1, TNFRSFlla, CA2, CLCN7, OSTM1, and/or PLEKHM1, for the treatment of osteopetrosis in the subject.
  • the protocol will be carried out in a manner similar to that of Example 3, and monocytes will be transduced with a lentivirus designed to express a cDNA coding for the wild type allele of the deficient gene under a strong promoter, with a similar number of transferred cells per kg of weight. Following the auto-transfusion, subjects will be monitored at regular intervals for clinical and radiological signs, and possibly bone biopsy in order to determine the efficiency of the procedure. In addition, collection of monocytes and hematopoietic stem
  • Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation. Cell 93, 165-176 (1998).
  • a range includes each individual member.
  • a group having 1-3 cells refers to groups having 1, 2, or 3 cells.
  • a group having 1-5 cells refers to groups having 1, 2, 3, 4, or 5 cells, and so forth.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Microbiology (AREA)
  • Epidemiology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Hematology (AREA)
  • General Engineering & Computer Science (AREA)
  • Rheumatology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Biochemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Mycology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Molecular Biology (AREA)
  • Dermatology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Virology (AREA)
  • Marine Sciences & Fisheries (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

Disclosed herein are methods and compositions for treating, preventing, or ameliorating osteopetrosis. In some embodiments, the present technology relates to administering a composition comprising a therapeutically effective amount of engineered monocytic cells or wild-type monocytic cells from a healthy donor to a subject suffering from or at risk for osteopetrosis.

Description

METHODS AND COMPOSITIONS FOR THE TREATMENT OF
OSTEOPETROSIS
CROSS-REFERENCE TO RELATED APPLICATION
[0001] The present application claims priority to U.S. Provisional Patent Application No. 62/679,553, filed on June 1, 2018, the contents of which are hereby incorporated by reference in their entirety.
STATEMENT OF FEDERALLY FUNDED RESEARCH
[0002] This invention was made with government support under CA008748, AI130345 and HL 138090 awarded by the National Institutes of Health. The government has certain rights in the invention.
TECHNICAL FIELD
[0003] The present technology generally relates to methods and compositions for treating, preventing, or ameliorating osteopetrosis. More specifically, the present technology relates to administering a composition comprising a therapeutically effective amount of engineered monocytic cells or wild-type monocytic cells from a healthy donor to a subject suffering from or at risk for osteopetrosis.
BACKGROUND
[0004] The following description is provided to assist the understanding of the reader. None of the information provided or references cited is admitted to be prior art.
[0005] Osteoclasts are multinucleated giant cells that resorb bone, ensuring development and continuous remodeling of the skeleton and the bone marrow hematopoietic niche.
Defective osteoclast activity leads to osteopetrosis and bone marrow failure, while excess activity can contribute to bone loss and osteoporosis. Osteopetrosis can be partially treated by bone marrow transplantation in human and mice, in accordance with osteoclasts hematopoietic origin, and studies suggesting that they develop by fusion of hematopoietic stem cell (HSC)-derived monocytic precursors in the presence of CSF1 and RANK-Ligand. However, the developmental origin and lifespan of osteoclasts, and the mechanisms that ensure maintenance of osteoclast function throughout life in vivo remain largely unexplored. Moreover, there is a need to develop alternative therapeutic approaches to the treatment of osteopetrosis as the current treatment by bone marrow transplantation is plagued by an approximate 48% overall survival at 6 years.
SUMMARY
[0006] In one aspect, the present disclosure provides a method for treating or preventing osteopetrosis in a subject in need thereof, the method comprising administering a
composition comprising a therapeutically effective amount of monocytic cells from a healthy donor to the subject. In some embodiments, the subject is characterized by decreased expression of one or more of CA2 , CLCN7 , CTSK , CSF1R , IKBKG, ITGB3 , OSTM1 , PLEKHM1 , TCIRG1 , TNFRSF11A, and TNFSF11, as compared to the monocytic cells of the donor. In some embodiments, the osteopetrosis comprises one or more of stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia. In some embodiments, the composition is formulated for intravenous administration by injection, infusion, or transfusion. In some embodiments, the subject is a mammal. In some embodiments, the mammalian subject is a human. In some embodiments, the subject is characterized by a cathepsin K deficiency.
[0007] In one aspect, the present disclosure provides a method for treating or preventing osteopetrosis in a subject in need thereof, the method comprising administering to the subject a composition comprising a therapeutically effective amount of monocytic cells engineered to express one or more genes selected from CA2, CLCN7 , CTSK , CSF1R, IKBKG, ITGB3, OSTMl, PLEKHM1 , TCIRG1 , TNFRSF11A , and TNFSF11. In some embodiments, the osteopetrosis comprises one or more of stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia. In some embodiments, the composition is formulated for intravenous administration by injection, infusion, or transfusion. In some embodiments, the subject is a mammal. In some embodiments, the mammalian subject is a human. In some embodiments, the subject is characterized by a cathepsin K deficiency. In some embodiments, the monocytic cells are obtained from the subject.
[0008] In one aspect, the present disclosure provides a donor monocytic cell line engineered to express one or more genes selected from CA2 , CLCN7 , CTSK , CSF1R , IKBKG, ITGB3, OSTM1, PLEKHM1 , TCIRG1 , TNFRSF11A , and TNFSF11 , wherein the one or more genes is operably linked to a heterologous nucleic acid to form a chimeric nucleic acid construct. In some embodiments, the heterologous nucleic acid encodes a selectable marker. In some embodiments, the selectable marker is a bioluminescent protein, a fluorescent protein, a chemiluminescent protein, a xanthine-guanine phosphoribosyl transferase gene (gpt), or any combination thereof. In some embodiments, the heterologous nucleic acid encodes one or more control sequences suitable for directing expression of the one or more genes in a monocytic cell. In some embodiments, the one or more control sequences comprises a promoter. In some embodiments, the donor cells comprise a vector encoding one or more genes selected from CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1 ,
PLEKHM1, TCIRG1, TNFRSF11A, and TNFSF11. In some embodiments, the vector is a mammalian expression vector, a lentiviral vector, or transposon vector.
[0009] The technology described and claimed herein has many attributes and embodiments including, but not limited to, those set forth or described or referenced in this brief summary. It is not intended to be all-inclusive and the technology described and claimed herein is not limited to or by the features or embodiments identified in this brief summary, which is included for purposes of illustration only and not restriction. Additional embodiments may be disclosed in the detailed description below.
BRIEF DESCRIPTION OF THE DRAWINGS
[0010] Figures 1A-1I. HSC-derived precursors are dispensable for osteoclasts and bone development. Figure 1A: Top: Representative photographs of teeth of control littermates and CsflrCre; CsflrKA 3-4 week old mice (right) and controls and CsflrCre ;Tnfrsfl lcfiF mice (left). Bottom left: Representative photographs of leg bones from controls and CsflC n‘; Tnfrsflla1111 mice, white arrowhead points to lack of blood cells. Right: bar graphs represent osteoclast numbers in E18.5, P7, and 4-week old CsflC rc; Tnfrsfl k 11 mice and littermates (n=3/3), and representative hematoxylin/TRAP staining of femur sections; arrow indicates an osteoclast. Figure IB: Representative photographs of teeth of 3- 4 week old Flt3Cre ;Tnfrsfl IcfJ1 , Flt3Cre Csflr^A , VavCre ;Csflr^ and littermates. Figure 1C: Bone marrow CD45+ cells numbers in l'h3 ,v; Tnfrsfl I a1111 and littermates at 4 (n=4/3) and 22 week old (n=4/5) and VavCn';Csflr1111 and littermates at 4 (n=8/8) and 62 week old (n=5/8) of the indicated age. Figure ID: Osteoclast counts in femurs from Tlt3( r ; Tnfrsfl I a,I , Flt3Cre ;Cflr^ ,
Vav( r, ;CsfIr1111 mice and littermates of the indicated ages. Figure IE: Representative hematoxylin/TRAP staining of femur sections from 22 week old Flt3Cre ;Tnfrsfl lcfP , 62 week old Vav°D/Cre ;CsflrFfl mice and littermates. White arrowhead points to trabecular bone.
Figure IF: Quantitative analysis of bone volume/total volume ratio of humerus or femurs from TH3( r ; Tufrsfl I a1111 (n=4), Flt3Cre iCsflr^fl (n=5) and controls littermates (n=8), and Vavc, ;CsfIr1111 (n=5) mice and controls littermates (n=5) as determined by MicroCT and Statistical significance determined by ANOVA. Figure 1G: Expression and MFI of YFP in in TRAP+ multinucleated cells from CsflrCre ;Rosa26LSL~YFP mice at embryonic day E16.5,
E18.5, P7, and 6 months, (n=3 per time point). For MFI and percentages, at least 100 osteoclasts were quantified per time point, genotype, displayed as a box plot, min, Ql, median, Q3 and max. Figure 1H as in Figure 1G for Flt3Cre ;Rosa26LSL~YFP mice at El 6.5,
El 8.5, P7, 4 weeks (4w), 3 months, and 6 months. Data are mean ± SD, circles represent individual independent biological replicates, n indicates number of mice per group. Statistical significance was analysed with GraphPad Prism using unpaired two-tailed t-tests unless otherwise indicated. Figure II: Representative confocal microscopy image of frozen sections from mice in (Figures 1G, 1H). Statistical significance was analysed with GraphPad Prism using unpaired t-tests and two-way ANOVA with Tukey’s multiple comparisons test as indicated in the figure legends. Significance was considered at p value (p) *p < 0.05; **p <
0.01; ***p < 0.001; ****p < 0.0001.
[0011] Figures 2A-2J. EMP derived osteoclasts are required for bone development.
Figure 2A: MGC number in femur anlage’s ossification centers from El 5.5 / E l 6.5 Mylr (n=6) and littermate controls (n=3). Figure 2B: Representative confocal microscopy pictures of frozen sections from Figure 1 A stained with fluorescent TRAP and TO-PRO-3. Figure 2C: Percent of TRAP+ cells expressing YFP in femur anlage from E15.5
CsflrMerCreMer ;Rosa26LSL~YFP mice (n=8) and Cre negative controls (n=5), pulsed at E8.5 with 4-OHT. Figure 2D: Representative confocal microscopy picture from Figure 2C. Figure 2E: Representative photographs of teeth of control littermates (Cre ), Tnfrsfl I aKoh ~( n‘;( "sflr1111 and Tnfrsfl laWask~Cre CsflrFfl mice (Cre+). Figure 2F: Leg bones from Tnfrsfl 1 a1 rc;( 'sflr1111 mice (Cre+, n=6) and control littermates (Cre , n=6) at P7and 4 weeks. Arrowhead points to the color of bone. Figure 2G: Representative MicroCT scans of long bones from mice in Figure 2E (n=6 per genotype). Figure 2H: Skull length from 3 weeks old Csflr 7 (n=6) controls littermates/h= 12) and Tnfrsf llaWask~Cre CsflrdP ice (n=4) as determined by
MicroCT. Figure 21: Osteoclasts counts in bone sections from E18.5, P7, and 3-4 week old Infrsfl la( re ;CsfI rl! 11 mice and littermate controls. Figure 2J: Numbers of bone marrow CD45+ cells determined by flow cytometry in 4 weeks old littermates (Cre , n=l3,27),
Tnfrsfl laKoha~Cre Csflrfl^ (n=9) and Tnfrsfl laWask~Cre ;Csflr-fl/fl (n=23) mice (Cre+). Data are mean ± SD, circles on graphs represent individual mice, n indicates the number of mice per group. Statistical significance was analysed with GraphPad Prism using unpaired two tailed t- tests. Significance was considered at p value (p) *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001.
[0012] Figures 3A-3G. In vivo dynamics of osteoclasts. Figure 3A: Parabiosis of
CsflrCre ;Rosa26LSL YFP mouse surgically paired with a CsflrCre ;Rosa26LSL tdTomato partner for 4 to 8 weeks. Representative (n=3) confocal microscopy image of frozen sections from the femur of a CsflrCre ;Rosa26LSL~YFP partner stained with antibodies for tdTomato (red) and YFP (green), ELF97 (blue) and TOPRO-3 (grey). Figure 3B: Pie graphs represent the percentage of tdTomato positive (red), YFP positive (green) and double tdTomato/YFP positive cells (yellow) among BM mononuclear cells (MNC), Megakaryocytes (Mega) and Multinuclear Giant Cells (MGC), from parabionts paired for 1, 2, 3, 4, and 8 weeks(n=8). Figure 3C: As in Figure 3B for parabionts separated after 4 weeks and analysed 14 weeks and 24 weeks after separation (n=3). Figure 3D: Scatter plots represent the Mean Fluorescent Intensity (MFI) of individual TRAP+ MGC for YFP (Y axis) and tdTomato (X axis), and histograms represent the overlayed distribution of the MFI values for YFP and tdTomato in TRAP+ MGC at the indicated time-points. Figure 3E: Bar graph display number of nuclei per TRAP+ MGC in femurs from wild type mice at 1, 3, and 6 month of age (n=3 mice per time point). Figure 3F: Representative confocal microscopy image of an EdU-labeled nucleus in a TRAP+ osteoclast, and histogram displaying the percentages of TRAP+ osteoclast with EdU-labeled nuclei and the number of labeled nuclei per cell 72 hrs after intravenous pulse-labeling with EdU (n=5 mice). Figure 3G: A model for development and maintenance of osteoclast syncytia. Data are mean ± SD, circles on graphs represent individual mice, n indicates the number of mice per group. Statistics: two-way ANOVA with Tukey’s multiple comparisons test *p<0.05 **p<0.005. [0013] Figures 4A-G. Rescue of Osteopetrosis. Figure 4A: Bone volume over total volume (BV/TV%) of femurs from 10 week-old Cathepsin K /_ mice (n=3) after 6 weeks parabiosis with Cathepsin K+/ heterozygote mice and from positive (n=4) and negative (n=4) controls parabionts, analysed by Von Kossa staining. Figure 4B: Monocyte transfer:
histograms represent percentages of tdTomato+ cells among bone TRAP+ MGC from
CsflrCre ;Rosa26LSL YFP recipients analysed by confocal microscopy {left) 11 days (n=2) and 60 days (n=5) after i.v. transfer at age 6 week-old of 3xl06 bone marrow Ly6C+ cells from CsflrCre ;Rosa26LSL tdTomato donors, and percentages of tdTomato+ cells among bone marrow precursors and blood leukocytes analyzed by flow cytometry after 60 days (n=5, right).
Figure 4C: Representative high power confocal microscopy image of the femur of a recipient mouse 60 days after i.v. transfer (from Figure 4B), stained with antibodies for tdTomato and YFP, and ELF97 and TOPRO-3. Figure 4D: Representative photographs of teeth, and CT scan of leg bones from ( sflrcr ; Csflr^fl mice (n=3) transferred with monocytic cells from CsflrCre; Rosa26LSL YFP donors at post-natal day 5, 8, and 11, and from wt and non-transferred Csflr cre; Csflr fl controls. Arrows indicated the presences of teeth eruption (upper panel) and bone marrow cavity (lower panel). Figure 4E: Representative confocal microscopy image of femur from mice #3 in Figure 4D, stained with antibodies for YFP, ELF97 and TOPRO-3. Figure 4F: Histograms represent {Left) the number of TRAP+ osteoclasts number in bone sections from mice in Figure 4D and non-transferred controls and {Right) the percentage of TRAP+ cells that are YFP+ in transferred mice. Symbols represents individual mice, values are average for 3 sections per mice. At least n = 100 osteoclasts were quantified per mice. Figure 4G: Histograms represent percentages of YFP+ cells among bone marrow precursors and blood leukocytes in the recipient mice (in Figure 4D) at the time of analysis. Data are mean ± SD, circles on graphs represent individual mice, n indicates the number of mice per group. Statistical significance was analyzed with GraphPad Prism using ANOVA with Tukey’s multiple comparisons test ***p < 0.001; ****p < 0.0001.
[0014] Figures 5A-G. Csflr( rc; Trfrsfl la1111, Csflr^A ;CsflrCre , and Tnfrsfl laCre ; Csflr^A mice are osteopetrotic. Figures 5A-B: Representative Computed Tomography scans
(nanospect CT) of 4 week old CsflrCre ;Tnfrsfl laf1^ mice arrows point skull deformation, and absence of bone marrow cavity in mutant mice. Figure 5C: H&TRAP staining of bone sections of 4 week old CsflrCre ;Tnfrsfl lcfl^ mice showing closure of the bone marrow.
Histology of paraffin sections (5pm thickness) corroborates the phenotype from mice in Figure 5B. Figure 5D: Representative photograph of inguinal lymph nodes in CsflrCre ;Trfrsfl lcpF mice. Figure 5E: Representative photograph of CsflrCre ;Trfrsfl lcfP and littermates controls. Figure 5F: Representative computed tomography scan
reconstructions (nanospect CT) of 4 weeks-old Tnfrsfl la(Koba)Cre; CsflrTJ1 mice. Arrows point to skull deformation, but the presence of bone marrow cavity in mutant mice, as in Figures 5A, B. Figure 5G: Representative Computed Tomography scans of CsflrCre; Csflr^ mice. Arrows shows skull deformation, and absence of bone marrow cavity in mutant mice as in A,B,. CT scans and photographs are representative of >10 litters.
[0015] Figures 6A-N. Bone histology and Flow cytometry analysis of bone marrow phenotypic KSL, LT-HSCs, ST-HSCs, andMPPs in mice of indicated genotypes. Figure 6A: Young Flt3Clc;Csf 1 ril/n and Flt3 Cre; Tnfrsfl lafl/flmice have normal long bone: H&TRAP staining of bone sections from 4 week old Flt3Cre ;Tnfrsfl lcflJ1 mice showing normal bone structure and bone marrow cavity. Figure 6B: LSK cell numbers in bone marrow
Flt3Cre ;Tnfrsfl lcflF (n=3), Flt3Cre ; Tnfrsfl lcf/+ mice (n=2), and littermate controls (n=4) in 3- 4 week old mice and Flt3Cre ;Tnfrsfl la1111 (n=5), Flt3Cre ;Tnfrsfl kP mice (n=4), and littermate controls (n=4) in 22 week old mice. Figure 6C: H&TRAP staining of bone sections from 4 week old IΊΐ3( n';CsflPl l!m\ce showing normal bone structure and bone marrow cavity. Figures 6D, E: Phenotypic long-term hematopoietic stem cells (LT-HSCs) are reduced in aged but not old
Figure imgf000008_0001
(Figures 6F, G) LT-HSC are reduced in in young Csfl r /_ and to a lesser extent in young Tnfrsf l 1 ylV sklC :Csfl 1 Numbers per 2 femur are shown. Figure 6H: Flow cytometry analysis of F4/80+ cells in brain (microglia) and epidermis (Langerhans cells) in E18.5 Tnfrsfl laCre; Csfl rTJ1 embryos and littermate controls (n=3 per group). Figure 61: H&TRAP staining of bone sections from P7
Tnfrsfl laCre ;CsflrFT and littermate control showing absence of the bone marrow cavity. Figure 6J: Flow cytometry plot of Fetal liver at E15.5 (B/C representative results of 3 experiments). Figure 6K: LSK numbers in bone marrow of 3-4 week old
Figure imgf000008_0002
and littermate controls (n=20) and Tnfrsfl laCre(koba) ;CsflrFT (n=7), Tnfrsfl laCre(koba) ;CsflrT+l {n=6) and littermate controls (n=8). Figure 6L: For comparison, LSK numbers in bone marrow of 3-4 week old Csflr /_ (n=22)and littermate controls (n=2l). Figure 6M: Representative micrographs of femur sections from 4 week old Tnfrsfl laCre ;CsflrFT and littermate control stained with hematoxylin/TRAP. Figure 6N: Blood leukocytes numbers in 4 week old Tnfrsfl laCre ;CsflrFT mice (n=5),
Tnfrsfl laCre ;CsflrF+ mice (n=6) and littermate controls (n=l2). Circles represent individual mice, results from 3 independent experiments. Data are mean ± SD, circles on graphs represent individual mice, n indicates the number of mice per group. Statistical significance was analysed with GraphPad Prism using unpaired two tailed t-tests. Significance was considered at p value (p) *p < 0.05; **p < 0.005; ***p < 0.0005; ****p < 0.0001. LT-HSC: Lin- Kit+ Sca-l+; Kit+ Scal+ CD34- Flt3-. Phenotypic short-term hematopoietic stem cells (ST-HSC): Lin- Kit+ Sca-l+ CD34+ Flt3-. Multipotent progenitors (MPP): Lin- Kit+ Sca-l+ CD34+ Flt3+. Lin: CD3 CD19 NK1.1 Terl l9 CDl lb Grl B220.
[0016] Figures 7A-E. Bone histomorphometry in old Flt3Cre ;Tnfrsfl lc ^, FltS^ CsflrFfl, and VavCre CsflrFflmice and control littermates. Figure 7A: Representative Micro CT pictures , genotype is indicated. Figure 7B: Bone length, connectivity density (Conn density), Trabecular Number (Tb.N.), and Trabecular Spacing (Tb.Sp.) analyzed by microCT in aged Flt3Cre ;Tnfrsfl IcflF (n=4), l· Ί 13 ( re ; ( is// r^1 ( n =4 ) and control littermates (n=7). Figure 7C: Bone histomorphometry as in Figure 7B, in VavCre; Csflr^fl mice and control littermates (n=5). Figures 7D, E: Dynamic bone histomorphometry in aged Flt3Cre ;Tnfrsf l 1 ail/nand Flt3Cre ;Csf 1 ril/n mice using in vivo calcein labeling. Representative micrographs of calcein labeling (green) of femura of mice from the indicated genotypes and ages (Figure 7D). Scale bars represent 200pm (top) and 50pm (bottom). Quantification of calcein labeling by fluorescence microscopy of mineralized surface/bone surface (MS/BS), mineral apposition rate (MAR), and bone formation rate/bone surface (BFR/BS) in aged Flt3Cre ;Tnfrsfl lcf^ (n=5), 1Ίΐ3( re;( sfl r,l/),(n=3 ) and control littermates (h=10) Data are mean ± SD, circles on graphs represent individual mice, n indicates the number of mice per group. Statistical significance was analysed with GraphPad Prism using unpaired two tailed t-tests.
Significance was considered at p value (p) *p < 0.05; **p < 0.005; ***p < 0.0005; ****p < 0 0001
[0017] Figures 8A-E. Colonization of the bone marrow by Csflr+ and Flt3+
hematopoietic cells. Figure 8A: Representative confocal microscopy images of frozen sections from Flt3Cre ;Rosa26LSL~YFP and CsflrCre ;Rosa26LSL~YFP mice analyzed at El6.5 (n=3). Figure 8B: YFP labeling efficiency in Flt3Cre ;Rosa26LSL~YFP mice analyzed by flow cytometry in the indicated cell populations, and by confocal microscopy on frozen bone sections at the indicated time (Right). Insets show YFP expression on individual osteoclasts. YFP: anti-GFP antibody, TRAP: ELF97 fluorescent substrate, TO-PRO-3: nuclear stain. Figure 8C: YFP labeling efficiency in CsflrCre ;Rosa26LSL~YFP mice analyzed as in Figure 8A. Data from Figures 8A, B are representative of at least 3 experiments by time point and genotypes. Circles represent individual mice. Genetic lineage tracing of osteoclasts in ossification centers using CsflrMenCreMer ; Rosa26LSL YFP. Representative high power confocal microscopy images from embryonic femurs showing MGCs in primary ossification centers from CsflrMenCreMer ;Rosa26LSL YFP . Figure 8D: El 8.5 embryos pulsed with 4- hydroxytamoxifen at E8.5 showing YFP expression in MGCs after Cre recombination and quantitated as MFI (right bar graph) from Cre+ (n=8) and Cre-(n=4), and Figure 8E unpulsed controls showing the lack of YFP in Cre+ (n=4) and Cre- (n=4). Sections were labeled with antibodies against YFP, TRAP (ELF97 substrate) and TO-PRO-3 as a nuclear stain.
[0018] Figures 9A-D. Tnfrs 1 la(wask)Cre knock-in mice allow deletion of target genes in fetal macrophages, but not definitive HSC and their progeny in blood and tissues, while Favcre mice allow deletion of target genes in definitive HSC, but not fetal macrophages . Figure 9A: Bar graphs indicate percentages of eYFP expression by flow cytometry in cells from
Tnfrsfl laCre ;Rosa26LSL~YFP in the indicated cell types, organs, and time points. Data represent 3 independent experiments n numbers indicated on x axis. Figure 9B: Lineage tracing in the fetal liver of Vavcre+;tdRFPwt/kl mice n numbers indicated on x axis. Figure 9C:
Representative molecular analysis of Csflr deletion in purified bone marrow HSPC from 62 weeks old Vavcr' ;CsfI rllJl m\ce and controls (n=5). Figure 9D: Representative photograph of teeth from 3 week old R.26-CreERT2+ Csflr^ pulsed with Tamoxifen at E10.5 (n=3 mice from 3 independent litters). FL: Fetal Liver, HSPC: hematopoietic stem and progenitor cell, LT-HSC: Long-term Hematopoietic Stem Cells, MPP: Multipotent progenitors, PMN:
polymorphonuclear cells, mono: monocytes, T: T cells, B: B cells, PEC: peritoneal exudate cells. Data are mean ± SD, circles on graphs represent individual mice, n indicates the number of mice per group.
[0019] Figures 10A-F. Bone morphometric and dynamic histomorphometry effects of the deletion of Csflr in P21 Tnfrsfl laCre ;CsflHfl mice . Bone volume / total vlume (BV/TV,
Figure 10A), Bone length (Figure 10B), connectivity density (Conn density, Figure 10C), and Trabecular Number (Tb.N., Figure 10D) was analyzed by microCT in 21 day/old mice. Csflr (n=4) and control littermates (n=7), Tnfrsfl kTv sk'( re ;C.s/7/7// (n=8) and
Tnfrsfl la(wask)Cre+ ;CsflrTl+(n=l), Tnfrsfl I a'"askl( re ;Csflr,u~(n=3) and control littermates (n=5). Figure 10E: Representative micrographs of calcein labeling (green) of femur of mice from the indicated genotypes and ages (n=4). Scale bars represent 50pm. Figure 10F:
Quantification of calcein labeling by fluorescence microscopy: mineralized surface/bone surface (MS/BS), mineral apposition rate (MAR), and bone formation rate/bone surface (BFR/BS) in Tnfrsfl la(wask)Cre+ ;Csflr^1 Csflr 7 (n=4) and control littermates (n=l5). Data are mean ± SD, circles on graphs represent individual mice, n indicates the number of mice per group. For statistical analysis unpaired two-tailed t-test. *p<0.05, **p<0.005,
***p<0.0005, ****p<0.000l.
[0020] Figures 11A-C. EdU labeling of bone marrow myeloid cells. Figure 11 A: Short term kinetics: EdU (20pg/g) is injected intraperitoneally to C57B16/N mice at t=0 min. Mice are sacrificed at the indicated time points and the % of EdU+ cells (blue) and the geometric MFI of EdU+ cells (red) and determined by flow cytometry, showing rapid EdU
incorporation. % of EdU+ cells plateau at ~30 min, and gMFI plateau at ~ 75 minutes.
Following a first round of cell division, -50% of monocytic cells are labeled after 8 to 12 hrs (n=3-8 mice see source data). Figure 11B: Long term kinetics: (l-240hrs) EdU is injected as in Figure 11 A and % of EdU+ monocytic cells in bone marrow (top) and blood (bottom) is determined by flow cytometry, showing that labeling of -50% of monocytic cells is observed for -2 days. Labeled cells are not detectable after 3 days. Circles represent determination from individual mice, data for each time point are pooled from 2-3 independent experiments see source data. Figure 11C: Parabiosis between CsflrCre ;Rosa26LSL~YFP and
CsflrCre ;Rosa26LSL tdTomato pairs as described in (Figure 3), paired for 1-8 weeks, and from CsflrCre Rosa26LSL~YFP partners separated after 4 weeks of parabiosis and analyzed 4 weeks,
14 weeks and 24 weeks after separation. Scatter plots represent the Mean Fluorescent Intensity (MFI) of individual TRAP+ MGC for YFP (Y axis) and tdTomato (X axis), and histograms represent the overlaid distribution of the MFI values for YFP and tdTomato in TRAP+ MGC at the indicated time-points. Data are mean ± SD, circles on graphs represent individual mice, n indicates the number of mice per group.
[0021] Figures 12A-C. FACS analysis of monocyte purification, and blood / bone marrow from transferred CsflrCre; Csflr1111 mice. Figure 12A: Representative flow cytometry plots of purified bone marrow monocytes from magnetic bead based enrichment (MACS), percentage of live YFP+ monocytes is indicated. Representative flow cytometry plots from blood
(Figure 12B) and bone marrow (Figure 12C) of 14 day old mice transferred with 1E6 YFP+ monocytes on day 5,8,11, the percentage of YFP+ cells is indicated. Experiments A-C are representative of 3 independent experiments. [0022] Figure 13. Rescue of osteoclasts by monocyte transfer in CsflrCre; Csfld yT mice. High power confocal microscopy images of frozen sections from CsflrCre; Csflr^T mice transferred with monocytes from CsflrCre Rosa26LSL~YFP and controls, stained with antibodies for YFP, TRAP substrate ELF97 and TOPRO-3 as a nuclear stain. Examples of
multinucleated Trap+, YFP+ cells (osteoclasts) are indicated with dotted lines. n=3 mice from independent litters. Numbers (#1,2,3) correspond to the mice in Figure 4D.
[0023] Figures 14A-F. Csflr deletion in Tnfrsfl laCre CsflrF/F mice results in lack of tissue macrophages and osteoclasts at birth, while definitive HSC and their progeny are present in the fetal liver and blood. Figure 14A: Flow cytometry analysis of F4/80+ cells in brain (microglia) and epidermis (Langerhans cells) in El 8.5 Tnfrsfl laCre+ ;CsflrF/F embryos and littermate controls. Figure 14B: H&TRAP staining of bone sections from P7
Tnfrsfl laCre+ ;CsflrF/F and Tnfrsfl Ia( ''-'"Csflr1 littermate control showing absence of the bone marrow cavity. Figure 14C: Flow cytometry plot of Fetal liver at E15.5. Figure 14D LSK numbers in bone marrow of 3-4 week old Tnfrsfl laCre+;CsflrF/F and littermate controls. Figure 14E: For comparison, LSK numbers in bone marrow of 3-4 week old Csflr A and littermate controls. Figure 14F: Blood leukocytes numbers in 4 week old
Tnfrsfl I a( n' Csflr1 mice and littermate controls. Circles represent individual mice, results from 3 independent experiments.
[0024] Figures 15A-D. Bone morphometric effects of the deletion of Csflr in P21
Tnfrsfl laCre+ ;CsflrF/~ or F/Fmice. Bone volume / total vlume (BV/TV, Figure 15A), Bone length Figure 15B, connectivity density (Conn density, Figure 15C), and Trabecular Number (Tb.N., Figure 15D) was analyzed by microCT in 21 day/old mice of indicated genotype. Controls (ctrls) included Tnfrsflla(wask)Cre+:CsflrF/+ or Tnfrsfl la(koba)Cre~ ;CsflrF/F and Tnfrsfl la(koba)Cre+ ;CsflrF/+ genotypes. Circles represent 1 mouse with 3-7 mice per group. For statistical analysis unpaired two-tailed t-test. *p<0.05, **p<0.005, ***p<0.0005, ****p<0.000l.
[0025] Figures 16A-B. EdU labeling of bone marrow myeloid cells. Figure 16A: Short term kinetics: EdET (20pg/g) is injected intraperitoneally to C57B16/N mice at t=0 min. Mice are sacrificed at the indicated time points and the % of EdU+ cells (blue) and the geometric MFI of EdU+ cells (red) and determined by flow cytometry, showing rapid EdU
incorporation. % of EdU+ cells plateau at ~30 min, and gMFI plateau at ~ 75 minutes.
Following a first round of cell division, -50% of monocytic cells are labeled after 8 to 12 hrs. Figure 16B: Long term kinetics: (l-240hrs) EdU is injected as in (A) and % of EdU+ monocytic cells in bone marrow (top) and blood (bottom) is determined by flow cytometry, showing that labeling of -50% of monocytic cells is observed for -2 days. Labeled cells are not detectable after 3 days. Circles represent determination from individual mice, data for each time point are pooled from 2-3 independent experiments.
DETAILED DESCRIPTION
[0026] It is to be appreciated that certain aspects, modes, embodiments, variations and features of the present technology are described below in various levels of detail in order to provide a substantial understanding of the present technology. The definitions of certain terms as used in this specification are provided below. Unless defined otherwise, all technical and scientific terms used herein generally have the same meaning as commonly understood by one of ordinary skill in the art to which this present technology belongs.
I. Definitions
[0027] The following terms are used herein, the definitions of which are provided for guidance.
[0028] As used herein, the singular forms“a”,“an” and“the” include plural referents unless the content clearly dictates otherwise. For example, reference to“a cell” includes a combination of two or more cells, and the like.
[0029] As used herein, the term“about” and the use of ranges in general, whether or not qualified by the term about, means that the number comprehended is not limited to the exact number set forth herein, and is intended to refer to ranges substantially within the quoted range while not departing from the scope of the invention. As used herein, “about” will be understood by persons of ordinary skill in the art and will vary to some extent on the context in which it is used. If there are uses of the term which are not clear to persons of ordinary skill in the art given the context in which it is used,“about” will mean up to plus or minus 10% of the particular term.
[0030] A“chimeric nucleic acid” comprises a coding sequence or fragment thereof linked to a nucleotide sequence that is different from the nucleotide sequence with which it is associated in cells in which the coding sequence occurs naturally. [0031] As used herein, the terms“effective amount,” or“therapeutically effective amount,” and“pharmaceutically effective amount” refer to a quantity sufficient to achieve a desired therapeutic and/or prophylactic effect, e.g ., an amount which results in the prevention of a disease, condition, and/or symptom(s) thereof. In the context of therapeutic or prophylactic applications, the amount of a composition administered to the subject will depend on the type and severity of the disease and on the characteristics of the individual, such as general health, age, sex, body weight, and tolerance to the composition drugs. It will also depend on the degree, severity, and type of disease or condition. The skilled artisan will be able to determine appropriate dosages depending on these and other factors. In some embodiments, multiple doses are administered. Additionally or alternatively, in some embodiments, multiple therapeutic compositions or compounds are administered. In the methods described herein, compositions comprising the monocytic cells of the present technology, may be administered to a subject having one or more signs, symptoms, or risk factors of osteopetrosis, including, but not limited to stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia. For example, a“therapeutically effective amount” of the compositions of the present technology, includes levels at which the presence, frequency, or severity of one or more signs, symptoms, or risk factors of osteopetrosis are, at a minimum, ameliorated. In some embodiments, a therapeutically effective amount reduces or ameliorates the physiological effects of osteopetrosis, and/or the risk factors of osteopetrosis, and/or the likelihood of developing osteotpetrosis. In some embodiments, a therapeutically effective amount is achieved by multiple administrations. In some embodiments, a therapeutically effective amount is achieved with a single administration.
[0032] The term“engineered” is used herein to refer to a cell or organism that has been manipulated to be genetically altered, modified, or changed, e.g., by disruption of the genome. For example, an“engineered monocytic cell” refers to a monocytic cell that has been manipulated to be genetically altered, modified, or changed. For example, in some embodiments, an engineered monocytic cell refers to a monocytic cell that has been transduced with a lentivirus designed to express a nucleotide sequence of interest, e.g, a cDNA coding for the wild type allele of any one or more of CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1, PLEKHM1 , TCIRG1 , TNFRSF11A , or TNFSF11 under a strong promoter.
[0033] “Heterologous nucleic acid” refers to a nucleic acid, DNA, or RNA, which has been introduced into a cell (or the cell’s ancestor), and which is not a copy of a sequence naturally found in the cell into which it is introduced. Such heterologous nucleic acid may comprise segments that are a copy of a sequence that is naturally found in the cell into which it has been introduced, or fragments thereof.
[0034] As used herein,“prevention,”“prevent,” or“preventing” of a disorder or condition refers to, in a statistical sample, reduction in the occurrence or recurrence of the disorder or condition in treated subjects/samples relative to an untreated controls, or refers delays the onset of one or more symptoms of the disorder or condition relative to the untreated controls.
[0035] As used herein“subject” and“patient” are used interchangeably and refer to a mammalian subject. In some embodiments,“subject” means any animal (mammalian, human, or other) patient that can be afflicted with osteopetrosis and when thus afflicted is in need of treatment. In some embodiments, the subject is a human.
[0036] “Treating,”“treat,”“treated,” or“treatment” of a disease or disorder includes: (i) inhibiting the disease or disorder, z.e., arresting its development; (ii) relieving the disease or disorder, z.e., causing its regression; (iii) slowing progression of the disorder; and/or (iv) inhibiting, relieving, or slowing progression of one or more symptoms of the disease or disorder.
[0037] It is to be appreciated that the various modes of treatment or prevention of medical diseases and conditions as described are intended to mean“substantial,” which includes total but also less than total treatment or prevention, and wherein some biologically or medically relevant result is achieved.
II. General
[0038] Described herein are compositions and methods for the treatment of osteopetrosis. Taken together, the data described herein identify: (i) the developmental origin of osteoclasts, the cells that shape bone architecture; (ii) a mechanism that controls the maintenance of osteoclast function after birth; and (iii) a novel strategy to treat osteopetrosis and to modulate osteoclast activity in vivo. In particular, the data described herein demonstrate that parabiosis or transfusion of monocytic cells results in long-term gene transfer in osteoclasts in the absence of HSC chimerism and can rescue an adult-onset osteopetrotic phenotype caused by cathepsin-K deficiency. Transfusion of monocytic cells is also sufficient to rescue bone development in early-onset autosomal recessive osteopetrosis in newborn mice.
III. Osteopetrosis
[0039] Osteopetroses are a heterogeneous group of genetic disorders characterized by increased bone density due to impaired bone resorption by osteoclasts. The increased bone density places the affected individual at an increased risk for bone fracture. Normally, bone growth is a balance between osteoblasts (cells that create bone tissue) and osteoclasts (cells that destroy bone tissue). Individuals with osteopetrosis have a deficiency of osteoclasts, resulting in too little bone resorption and too much bone creation. The types of osteopetrosis are distinguished based on their pattern of inheritance: autosomal dominant, autosomal recessive, or X-linked.
[0040] The signs and symptoms of osteopetrosis may vary depending on the type of the disease, and mild forms of osteopetrosis may be asymptomatic. However, the typical signs and symptoms of osteopetrosis include: stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia.
[0041] The signs and symptoms of autosomal dominant osteopetrosis (ADO; also known as Albers-Schonberg disease), include multiple bone fractures, scoliosis, arthritis in the hips, and/or osteomyelitis. Autsomal recessive osteopetrosis (ARO) is often characterized by one or more of a high risk of bone fracture resulting from minor bumps or falls, pinched nerves in the head and face, impaired bone marrow function, slow growth, short stature, dental abnormalities, hepatosplenomegaly, intellectual disability, and epilepsy. Individuals diagnosed with intermediate autosomal osteopetrosis (IAO), which is a form of osteopetrosis that can have either an autosomal dominant or recessive pattern or inheritance, may be characterized by one or more of a high risk of bone fracture and anemia, calcifications in the brain, intellectual disability, and renal tubular acidosis. Individuals with the X-linked pattern of inheritance may be characterized by one or more of lymphedema, anhidrotic ectodermal dysplasia, and immunodeficiency.
[0042] Mutations in several genes have been linked to the various forms of osteopetrosis or may underlie the development of osteopetrosis. These include mutations in CA2 , CLCN7, CTSK , CSF1R , IKBKG, ITGB3, OSIMI, PLEKHM1 , TCIRG1 , TNFRSF11A (which encodes for receptor activator of NF-kB (. RANK)), and TNFSF11 (which encodes for receptor activator of NF-kB ligand (. RANKF) ). Many of the genes associated with osteopetrosis are involved in the formation, development, and function of osteoclasts.
IV. Therapeutic Applications
[0043] In some embodiments, a method for treating or preventing osteopetrosis in a subject in need thereof, comprising administering a composition comprising a therapeutically effective amount of monocytic cells from a healthy donor to the subject is provided. In some embodiments, the subject is characterized by decreased expression of one or more genes implicated in or potentially underlying the development of osteopetrosis, such as CA2 , CLCN7 , CTSK, CSF1R , IKBKG, ITGB3, OSIMI, PLEKHM1 , TCIRG1 , TNFRSF11A , and TNFSF11. In some embodiments, the subject is characterized by a cathepsin K deficiency.
[0001] [0044] In some embodiments, a method for treating or preventing osteopetrosis in a subject in need thereof, comprising administering to the subject a composition comprising a therapeutically effective amount of monocytic cells engineered to express one or more genes implicated in or potentially underlying the development of osteopetrosis, such as CA2 , CLCN7 , CTSK, CSF1R , IKBKG, ITGB3, OSIMI, PLEKHM1 , TCIRG1 ,
TNFPSF11A , and TNFSF11 is provided. In some embodiments monocytic cells used for treatment are genetically modified to correct a genetic abnormality or to improve or changed cellular functioning according to known genetic engineering protocols. In some
embodiments, a method of treating or preventing osteopetrosis in a subject comprises: (a) obtaining a sample of monocytic cells from the subject; (b) genetically correcting one or more mutations in the monocytic cells, (c) culturing the monocytic cells; and (d) providing the corrected monocytic cells to the subject. In some embodiments, a method for treating or preventing osteopetrosis comprises: (a) obtaining a sample of monocytic cells from the subject; (b) genetically engineering the cells to express a nucleotide sequence coding for the wild type allele of any one or more of CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3 , OSTM1 , PLEKHM1 , TCIRG1 , TNFRSF11A, and TNFSF11 with a suitable transducing vector, such as a lentiviral vector; (c) culturing the engineered monocytic cells under conditions sufficient to express the nucleotide sequence; (d) removing the viral particles from the engineered monocytic cells; and (e) providing the engineered monocytic cells to the subject. In some embodiments, the transducing vector encoding the one or more of CA2 , CLCN7, CTSK , CSF1R , IKBKG, ITGB3, OSTM1, PLEKHM1 , TCIRG1 , TNFRSF11A , and TNFSF11 is a mammalian expression vector. In some embodiments, the mammalian expression vector is a lentiviral vector or transposon vector.
[0045] Subjects suffering from osteopetrosis can be identified by any or a combination of diagnostic or prognostic assays known in the art. For example, typical symptoms of osteopetrosis include, but are not limited to, stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia.
[0046] For therapeutic applications, a composition comprising a therapeutically effective amount of monocytic cells from a health donor and/or monocytic cells engineered to express one or more genes selected from CA2, CLCN7, CTSK , CSF1R, IKBKG, ITGB3 , OSTM1 , PLEKHM1 , TCIRG1 , TNFRSF11A, and TNFSF11 is administered to the subject. In some embodiments, the composition is administered according to any acceptable transfusion regimen. In some embodiments, the composition is administered one, two, three, four, or five times per day. In some embodiments, the composition is administered more than five times per day. Additionally or alternatively, in some embodiments, the composition is
administered every day, every other day, every third day, every fourth day, every fifth day, or every sixth day. In some embodiments, the composition is administered weekly, bi-weekly, tri-weekly, or monthly. In some embodiments, the composition is administered for a period of one, two, three, four, or five weeks. In some embodiments, the composition is
administered for six weeks or more. In some embodiments, the composition is administered for twelve weeks or more. In some embodiments, the composition is administered for a period of less than one year. In some embodiments, the composition is administered for a period of more than one year or until a desired therapeutic outcome is observed in the subject.
[0047] In some embodiments, treatment of subjects diagnosed with or suspected of having osteopetrosis with one or more compositions of the present technology ameliorates or eliminates one or more of the following symptoms of osteopetrosis: stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections,
hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia.
Prophylactic Methods
[0048] In one aspect, the present technology provides a method for preventing or delaying the onset of osteopetrosis or one or more symptoms of osteopetrosis in a subject at risk of having or developing osteopetrosis. In prophylactic applications, compositions of the present technology are administered to a subject susceptible to, or otherwise at risk of for
osteopetrosis in an amount sufficient to eliminate or reduce the risk, or delay the onset of the disease, including biochemical, histologic and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease.
[0049] Administration of a prophylactic compositions can occur prior to the manifestation of symptoms characteristic of the disease or disorder, such that the disease or disorder is prevented or, alternatively, delayed in its progression.
EXAMPLES
[0050] The following examples are provided by way of illustration only and not by way of limitation. Those of skill in the art will readily recognize a variety of non-critical parameters that could be changed or modified to yield essentially the same or similar results. The examples should in no way be construed as limiting the scope of the present technology, as defined by the appended claims. Materials and Methods
[0051] CsflrlCre , CsflrMenCreMer , ( 'sflrllox were kindly provided by Dr Jeffrey Pollard, Csflr from Richard Stanley, Flt3Cre were kindly provided by Dr Thomas Boehm, Myb+/ mice were kindly provided by Dr John Frampton, and VavCre were kindly provided from Dr Thomas Graf. Tnfrsfl 1 a,lox mice were kindly provided by JM Penninger, Tnfrsfl iaKoba/Cre mice were kindly provided by Dr Yasuhiro Kobayashi, Ctsktml(cre)Ska mice were kindly provided by Dr. Ostrowsky (MSKCC) and Tnfrsfl ]a Wask/Cre mice were genereated in the Waskow Lab. Rosa.26-CreERn ( R26-CreERn ) were kindly provided from Drs. Pierre Chambon and Anton Bems. Rosa26LSL~YFP (stock number: 006148) and p0sa26LSL~tdTomato (stock number: 007908) reporter mice were purchased from The Jackson Laboratory.
[0052] Animal procedures. Mice were bred and kept under specific pathogen conditions in separated ventilated cages in the animal facility of MSKCC and the Medical Theoretical Center of the TU Dresden. All experiments with osteopetrotic mice that lack teeth were performed with mice maximal 4 weeks of age that were kept with the lactating mother or provided with DietGel 76A (Clear H20, 72-07-5022) to avoid secondary effects from malnutrition. Experiments were performed in adherence to the Institutional Review Board (IACUC 15-04-006) from MSKCC and Landesdirektion Dresden and were in compliance with relevant ethical regulations. Mice greater than 7 days old were sacrificed by cervical dislocation (TU Dresden), CO2 asphyxiation or anesthesia (MSKCC). To harvest embryos, pregnant females were sacrificed and embryos were collected by postmortem cesarean from the uterus and exsanguinated through decapitation in cold PBS (Fisher, 14190).
[0053] Genotyping: PCR genotyping was performed according to protocols described previously and indicated in Table 1. The investigators were not blinded to allocation during experiments and outcome assessment.
Table 1.
Figure imgf000020_0001
Figure imgf000021_0001
Figure imgf000022_0001
[0054] Fate mapping with tamoxifen inducible Cre models. For timing of embryonic development, mice were crossed at night, the following day a positive vaginal plug was considered as 0.5 days post-coitum (dpc), as previously described. Embryos were harvested as indicated below. For postnatal time point, pregnant females were monitored for the date of delivery, caesarean sections were carried out at term and neonates were fostered using lactating CD-l females.
[0055] CsflrU n( reUer female mice were crossed with male Rosa26LSL~YFP/LSL~YFP mice. Cre recombination in CsflrMenCreMer ;Rosa26LSL~YFP embryos at embryonic day E8.5 was induced with a single dose of 4-hydroxytamoxifen (4-OHT) injected intraperitoneally in pregnant mothers at a dose of 75pg/g of body weight supplemented with 37.5 pg/g of progesterone as previously described. Cre-mediated recombination in Rosa.26-CreERn tamoxifen (TAM) was introduced to pregnant mother by a single TAM gavage (5mg) at El 0.5, supplemented with progesterone (37.5pg/g body weight resolved in Sunflower seed oil, Sigma-Aldrich) was injected i.p. directly after gavage. To analyze newborn or 3 weeks old mice, caesarean sections were carried out at term and neonates were fostered using lactating CD-l females.
[0056] Parabiosis. For cellular complementation female CsflrlCre mice were crossed to male Rosa26LSL~YFP/YFP or posa26LSL~tdTomato/tdTomato mice. CsflrlCre ;Rosa26LSL~YFP and
Csflr,Cre ;Rosa26LSL~tdTomato females were used for parabiosis. For rescue of CathepsinK activity 4 week old female CtskCre/Cre and control littermate mice were used for parabiosis. Parabionts were kept on Sulfamethoxazole/Trimethoprim (Sulfatrim) diet for up to 8 weeks. Ex-parabionts were separated after 4 weeks for cellular complementation and 6 weeks for rescue of CathepsinK activity.
[0057] Surgical procedure, pre-operative procedure: weight-matched female partner mice for parabiosis were caged together few days before surgery. One day before the surgery the fur from lateral sides of mice was carefully removed with a trimmer followed by depilatory cream (for 3 minutes) at the site of surgery, excess of fur was removed with a moist gauze pad. Left side partner is shaved on the right side and vice versa. This procedure was performed under Isofluorane inhalation anesthesia. Mice were fed with food supplemented with Sulfatrim ad libitum one day prior to surgery.
[0058] Surgery: Mice were anesthetized intraperitoneally with l50mg/kg of ketamine and l5mg/kg of xylazine. Sterile eye lubricant (Paralube Vet Ointment, 17033-211-38) was applied to both eyes to prevent corneal drying during surgery. Following confirmation that a suitable anesthetic plane (no response to stimulation) has been attained, mice were placed in a supine position on a surgical tray with heat support provided by a heating pad. The surgical site was cleaned 3 times with cotton swabs soaked in povidone-iodine (Betadine) then with 70% ethanol. Before surgery a volume of 0.2ml of anesthetic agent bupivacaine (Marcaine 0.25-0.5% solution) was applied locally. Surgery was performed by a longitudinal skin incision on the lateral side of mice, approximately 0.5cm above the elbow to 0.5cm below the knee joint. Mice were laid side-by-side in close contact and the ligaments of the two knees and elbows were sutured together using monofilament non-absorbable suture. Then, the skin incisions was closed by apposing and clipping skin to skin of the pair with 9 mm wound clips.
[0059] Post-operative procedure: immediately after surgery mice were injected
subcutaneously with 2 mg/kg of meloxicam and 0.5mg/kg of buprenorphine and for a maximum of 48 hours postoperatively. Mice are provided with Sulfatrim and Hydrogel (Clear H2O, 70-01-5022) in medicups. Wound clips were removed 14 days after surgery under Isofluorane anesthesia.
[0060] Parabiosis separation procedure: Mice were anesthetized and prepared for surgery as indicated above. Mice were separated at the site of parabiosis junction. Using scissors, the skin joining both mice is cut longitudinally. The sutures around the elbows and knees are cut and removed. The resulting wound is closed with 9 mm wound clips. Mice were injected subcutaneously with 2 mg/kg of meloxicam and 0.5mg/kg of buprenorphine and for a maximum of 48 hours postoperatively. Wound clips were removed 14 days after surgery.
[0061] Analysis: Bones were prepared for histology on frozen sections as detailed below, and stained with antibodies to detect fluorescent proteins: anti-GFP biotin, anti-RFP
(Abeam), fluorescent TRAP staining and TO-PRO-3 as a nuclear stain. Histological sections of l5pm thickness were scanned by confocal microscopy at l.5pm Z stacks. Sections were quantified for the number of TRAP+ multinuclear cells (more than 3 nuclei per cell) and their YFP and Tomato expression using Imaris in 3D view and individual Z stacks. Pictures of the region of interest (area=2mm2) were then generated in Tiff format and analyzed by ImageJ software using ROI manager to calculate the mean fluorescence intensity (MFI) of YFP and Tomato for individual osteoclasts. For rescue of CathepsinK activity, dissected femurs were fixed in 10% neutral buffered formalin for 24 hours. Undecalcified bones were embedded in methyl methacrylate resin, and 7-pm sections were prepared on a rotation microtome. For mineralized bone volume over the total volume % (B V/TV%), sections were de-plastified and stained with von Kossa reagent (1% Silver nitrate/ Sodium formamide/ 5% Sodium thiosulfate) counterstained with Van Gieson solution.
[0062] EdU pulse labeling. 12 week old C57BL/6N mice (Charles River) were injected intraperitoneally with 25 pg/g of a 2.5mg/mL solution of EdU prepared extemporaneously (Fisher Cl 0420, Click-iT EdU Alexa Fluor 488 Flow Cytometry Assay Kit). Blood, bone marrow, and bone samples were collected at 60hrs and 72hrs post injection and bone were prepared for histology of frozen sections as indicated above for adult mice.
[0063] Histology of frozen sections. Frozen sections were cut at l5pm thickness using cryofilm and stained overnight with rat anti-Tubulin (Abeam, ab6l60, 1 :200) and with secondary goat anti-rat Alexa Fluor 555 (ThermoFisher Scientific A21430), as described below. Sections were washed 3 times with PBS and stained for EdU using Click-iT EdU Alexa Fluor 488 Imaging Assay Kit (Thermo Fisher Scientific, C10337). Click-it reaction: 357.5pL of IX Click-it reaction buffer, 40pL of CuS04 solution (lOOmM), 2.5pL of Alexa Fluor 488 azide solution and 100 pL of Reaction buffer additive for 500pL. Sections are incubated for 45 minutes at room temperature with lOOpL of Click-iT EdU reaction buffer then washed with PBS and stain for fluorescent TRAP (ELF 97) and TO-PRO-3 (nuclear stain). Mounting media was 75% glycerol in PBS.
[0064] Microscopy. Images were acquired using an inverted Zeiss LSM880 laser scanning confocal microscope. Histological sections of 15pm thickness were tile-scanned at l.Opm Z stacks.
[0065] Analysis. Sections were analyzed using Imaris in 3D view and individual Z stacks to quantify the percentage of EdU+ TRAP+ multinuclear cells and EdU+ nuclei per individual osteoclasts.
[0066] Adoptive transfers. Bone marrow monocytes were isolated from 12-16 weeks old donor. In fate-mapping experiments lxlO6 total cells from Csflr,Cre ;Rosa26LSL tdTomato mice were transferred at day 0, day 3 and day 6 by retro-orbital injections into recipient
Csflr,Cre ;Rosa26LSL YFP age and gender matched mice. Mice were sacrificed at day 11 and day 60 after the first transfer. For rescue of osteoclast activity lxlO6 total bone marrow monocytes from CsflrlCre ;Rosa26LSL~YFP or CsflriCre ;Rosa26LSL tdTomato were transferred intra-peritoneal at p5, p8, and pl 1. Mice were sacrificed at pl4 5 days after the last transfer. Bone marrow and blood was analyzed for the percentage of chimerism. To enrich Ly6C+ cells the Monocyte Isolation Kit (BM) for mouse (MACS Miltenyi Biotec, 130-100-629) was used as indicated by the manufacturer. Cell numbers of Ly6C+ cells were calculated by determining the cell number / ml using a Neubauer chamber in combination with a staining for Ly6C analyzed by flow cytometry. Bone samples were dissected and prepared for frozen sections as described above and stained for anti-GFP, anti-RFP, fluorescent TRAP and TO- PRO-3. The percentages of YFP+ or Tomato+ multinuclear cells was quantified in femurs of recipients.
[0067] Preparation and analysis of bone for histology of paraffin embedded samples. Bone samples were fixed in 4% formaldehyde (Fisher, 28908) in PBS for 1 day (embryo) or 3 days (post-natal mice) at 4°C then washed 3 times with PBS and decalcified (for mice older than P7) in a 14% EDTA pH7.1 solution at 4°C for 5 days to 15 days, washed 3 times with PBS and dehydrated in 70% ethanol for 1 day and processed for paraffin sections. Longitudinal sections of femurs were cut at 5 pm thickness using a Leica RM2265 paraffin microtome then place in Superfrost microscope slides let dry for 48 hours and heated in a dry incubator at 65°C for 1 hour, dewaxed and stained for TRAP and hematoxylin. [0068] Tartrate Resistant Acid Phosphatase (TRAP) staining protocol . To stain for TRAP, slides were placed in coplin jars and incubated in a 1% (v/v) Naphtol -Ether substrate solution in basic stock solution for lhr at 37°C followed by incubation in a solution containing 2% (v/v) sodium nitrate solution and 2% (v/v) basic fuchsin solution in basic stock solution for 20 mins at 37°C. Slides are rinse in 3 changes of water then stained with hematoxylin solution (Sigma, GHS332) diluted 1 :4 in water for 2 minute, washed 3 times with water then dehydrated and mounted in Entellan (Millipore, 107960). Solutions for TRAP staining:. Basic stock solution: 0.92% (w/v) anhydrous sodium acetate (Sigma, S8750), 1.14% (w/'v) dibasic dihydrate sodium tartrate (Sigma, S4797) and 0.28% (v/v) glacial acetic acid (Sigma,
537020) in distilled water, pH was adjusted between 4.7-5.0 with 5M sodium hydroxide
(Fisher, S318-1). Napthol-Elher substrate solution: 2% (w/v) Napthol AS-B1 Phosphate (Sigma, 70482) in 2-Ethoxy ethanol (Sigma, 256374). Sodium nitrate solution: 4% (w/v) Sodium nitrate (Sigma, 237213) in water. Basic fuchsin solution: 5% (w/v) basic fuchsin dye (Sigma, 857343) in 2N HCL (Fisher, A144-500).
[0069] Individual images from histological sections of 5pm thickness were acquired using a Zeiss Axio Lab.Al light microscope with a N-Achroplan 2.5X/0.07 M27 (420920-9901) or a N-Achroplan 20X/0.45 M27 (420950-9901) objective. Pictures were taken in ZEN lite software and exported as tiff files. Panoramic images were created with the photo-merge tool in Adobe Photoshop CS6. Pictures of mice were acquired with a dissecting microscope Leica M80 equipped with a Leica IC80 HD camera at 1.0X magnification. The region of interest analyzed was the methaphyseal trabecular bone 2 mm below the growth plate. The numbers of TRAP+ multinuclear cells (more than 3 nuclei per cell), associated to bone tissue, were quantified in ImageJ using the Cell Counter plugin. Numerical values were plotted using GraphPad Prism. For Static and dynamic histomorphometry. Young and aged mice were injected (i.p.) twice with 15 mg/kg body weight Calcein (Sigma) dissolved in 1.4%
NaHC03/PBS 2 and 3 days apart, respectively. Mice were sacrificed 2 days after the last calcein injection. Femora and tibiae were fixed in 4% PBS-buffered paraformaldehyde and dehydrated in an ascending ethanol series. Subsequently, bones were embedded in methacrylate and cut into 7 pm sections to assess the fluorescent calcein labels. Unstained sections were analyzed using fluorescence microscopy to determine the mineralized surface/bone surface (MS/BS), the mineral apposition rate (MAR), and the bone formation rate/bone surface (BFR/BS).To determine numbers of osteoclasts, bones were decalcified for one week using Osteosoft (Merck), dehydrated, and embedded into paraffin. Tartrate- resistant acid phosphatase staining was used to assess the osteoclast surface per bone surface (Oc.S/BS) and number of osteoclasts per bone surface (N.Oc/BS). Bone sections were analyzed using the Osteomeasure software (Osteometries, USA) following international standards.
[0070] Preparation of and analysis of bones for immunofluorescence on frozen sections. Samples were prepared as above and after decalcification and washing were soaked in 30% sucrose in PBS at 4°C for 1-2 days. Tissue samples were placed in disposable histology plastic molds and embedded in FSC22 Frozen Section Compound Clear (Leica, 3801480) and placed on a flat surface of dry ice to let freeze.
[0071] Immunofluorescence. Bones were cut at 15 pm thickness using a Cryostat Leica CM3050S with high profile microtome blades (Leica Surgipath DB80 HS) and cryofilm (Section Lab Inc.,) and let to dry for 48 hours at 4°C. Before staining with antibodies, sections were let to equilibrate at room temperature for 30 minutes, rehydrated with PBS (Fisher, 14190) 3 times for 5 min at room temperature. Sections were incubated with blocking buffer containing 0.25% BSA (Fisher BP 1600), 10% normal goat serum (Life Technologies, PCN 5000) and 0.3% triton (Sigma, T8787) in PBS for lhr at room temperature. Sections were washed 2 times with PBS for 5 minutes. Sections stained with anti-GFP biotin antibody were first incubated with Biotin/Streptavidin blocking kit (Vector laboratories, SP2002). Streptavidin blocking solution is prepared by adding 4 drops of streptavidin solution to lmL of PBS/0.25% BSA, samples were incubated for 15 min, then washed once with PBS for 5 min. Biotin blocking solution is prepared by adding 4 drops of biotin solution to lmL of PBS/0.25% BSA, samples were incubated for 15 min then washed once with PBS for 5 min. Primary and secondary antibodies used are listed in Table 2. Sections were also stained with fluorescent TRAP and nuclear stain.
Table 2.
Fate-mapping models: FU3Cre;Rosa26LSL YFP, CfslrCre;Rosa26LSL YFP Csfl rMeriCreMer;Rosa26LSL~ YFP and TnfrsfllaCre;Rosa26LSL YFP
Figure imgf000028_0001
Parabiosis
Figure imgf000028_0002
Figure imgf000028_0003
Bone marrow monocyte
transfusions and transplants
Figure imgf000028_0004
Figure imgf000028_0005
EdU
staining*
Figure imgf000029_0001
*After antibody staining sections were stained with the Click-it EdU
Alexa Fluor 488 Imaging Assay kit, as described in methods.
[0072] Fluorescent TRAP staining. Sections were prepared for fluorescent TRAP by incubating with TRAP incubation solution (112 mM sodium acetate, 76 mM sodium tartate, and 11 mM sodium nitrite, pH 4.1-4.3) at room temperature for 10 minutes. Buffer was removed and incubated with ELF97 substrate (Molecular Probes E6589, 2mM) at a concentration of 125mM in TRAP incubation solution for 15 min under ETV light and washed 2 times with PBS for 5 minutes. Nuclear stain used was TO-PRO-3 Iodide (Fisher T3605) 1 :4000 in PBS for 5 minutes. Mounting media was 75% glycerol in PBS. Images were acquired using an inverted Zeiss LSM880 laser scanning confocal microscope with Argon- ion 488nm, Diode 405-30nm, DPSS 56l-l0nm, HeNe 633nm laser lines and Plan- Apochromat 40X/1.4 N.A. DIC (UV) VIS-IR oil objective. Histological sections of l5pm thickness were tile-scanned at l .5pm Z-stacks in ZEN black and processed using ZEN lite. Sections were analyzed using Imaris in 3D view and individual Z stacks to quantify TRAP+ multinuclear cells and YFP and Tomato labeling. Pictures of the region of interest were generated in tiff format and analyzed by ImageJ software using ROI manager to calculate the mean fluorescence intensity (MFI) of individual osteoclasts. Numerical values were plotted using GraphPad Prism.
[0073] Animal imaging by computed tomography. NanoSPECT/CT: Mice were
anesthetized under Isofluorane anesthesia and placed on an imaging table containing an animal bed equipped with a nosecone for gas inhalation and body temperature stabilization. For 3 week old mice a mouse bed was used and for mice 8 weeks and older a rat bed was used. Whole-body imaging of mice was acquired using a NanoSPECT/CT scanner (Mediso) for non-invasive and longitudinal monitoring of the 3D skeletal structure. Each CT scan averaged 15 minutes and was acquired with an exposure time of 1, 000ms and 240 projections set at a pitch of 1 degree. The tube energy of the X-ray was 55KVp and 145mA. The in-plane voxel size was medium generating a voxel size of 147pnr\ Reconstructed images were analyzed using In Vivo Scope 2.0 (Bioscan, Inc.) software.
[0074] microCT. For tri-dimensional X-ray imaging by micro computed tomography, mice were sacrificed and bones placed in 70% ethanol until scanning. Bone microarchitecture was analyzed using the vivaCT40 (Scanco Medical, Switzerland). Entire femora or humeri were imaged at a resolution of 10.5pm (1 slice) with an X-ray energy of 70 kVp, 114 mA, and an integration time of 200ms. The machine was routinely calibrated using hydroxyapatite phantoms for density and geometry. Trabecular bone in femora or humeri from old mice was assessed in the metaphysis 20 slices below the growth plate using 150 slices. The trabecular region within the cortical bones (P21 mice) was determined in the femoral midshaft (100 slices up, 100 slices down). Pre-defmed scripts from Scanco were used for the evaluation.
[0075] Preparation of tissues and staining for Flow cytometry. Yolk sac from E10.5 embryos was digested for 60 min at 37°C in PBS containing 5% FCS, Collagenase Type 4 (Worthington, final 4.2 U/ml) and DNAsel (Sigma- Aldrich, final 100 pg/ml). The digestion reaction was stopped by incubation with 12.5 mM EDTA. Fetal liver was gently dissociated between the frosted ends of glass slides, and was then digested for 30 minutes using the same digestion enzyme mix as yolk sac. Blood was collected from anesthetized mice by retro- orbital venous sinus bleeding or cardiac puncture using a lmL syringe and a 26G needle rinsed with lOOmM EDTA (sigma E4884). The collected blood was lysed with 3 mL of red blood cell lysis buffer (l55mM NFECl sigma A9434, lOmM NaFlCCh sigma S5761 and O. lmM EDTA sigma E4884) for 5 mins, washed with lOmL of FACS buffer (PBS, 0.5%
BSA and 2mM EDTA) and centrifuged at 320g for 7 minutes at 4°C. All mice were perfused with lOml of PBS after blood withdrawal. Bone marrow cells were harvested by crushing or flushing femurs and tibias using a syringe and a 26G needle with lOmL of PBS / 5% FCS or RPMI supplemented with 10% FBS. Bone marrow was dissociated by gently pipetting up and down with a lmL or lOmL pipette. Spleens were gently dispersed between frosted slides and digested for 30 min at 37 °C in PBS with 5% FCS containing Collagenase Type 4 (Worthington) at a final concentration of 4.2 El/ml and 100 pg/ml DNAsel (Sigma- Aldrich). The reaction was stopped by incubation with 12.5 mM EDTA. Peritoneal cells were harvested by flushing the peritoneal cavity with lOmL PBS/5% FCS heated to 37°C. Adult brain and liver were dissected, cut into small fragments, and incubated at 37°C for 30 min in enzyme mix consisting of PBS with lmg/ml collagenase D (Sigma, 11088882001), 100 U/ml DNase I (Sigma, DN25), 2.4mg/ml of dispase (Fisher, 17105-041) and 3% FBS (ThermoFisher Scientific 10438026) or PBS containing 4.2 U/ml Collagenase type 4 (Worthington), 100 pg/ml DNAsel, 2.4mg/ml of Dispase (Gibco) and 3% FCS at 37°C for 30 min. After enzyme digestion all tissues were further dissociated by mechanical disruption using lOOpm cell strainers (Falcon, 352360) and a 3ml syringe plunger in 6 well plates containing 4mL or 5mL of cold FACS buffer or PBS / 5% FCS. Single cell suspensions were transferred to 5mL FACS tubes and pelleted by centrifugation at 320g for 7 minutes at 4°C. The cell pellets were resuspended in FACS buffer containing purified anti-mouse CD16/32 antibody (1 : 100, Biolegend 101301), 5% normal mouse (Fisher, 015-000-120), 5% normal rat (Fisher 012-000-120) and 5% normal rabbit serum (Fisher, 011-000-120) and incubated for 10 minutes on ice or directly stained using blocking antibodies in the staining mixtures. Samples were immunostained with fluorochrome-conjugated antibodies for 30 minutes on ice, and analyzed by flow cytometry using an LSRFortessa or an LSR II (BD-Bioscience). Full list of antibodies for flow cytometry is provided in Table 3. Each sample was stained with Hoechst (ThermoFisher Scientific, Hoechst 33258, 1 pg/ml) or DAPI (Applichem, l pg/25ml) moments prior to flow cytometry acquisition.
Table 3.
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
BM: Bone marrow; SP: Spleen; BL: Blood; Y: Yolk sac; FL: Fetal Liver; LV: Liver; LN: Lung; BR: Brain; P: PEC: E: Epidermis; K: Kidney.
[0076] All analysis was conducted using FlowJo (Tree Star). In all tissues single live cells were gated by exclusion of dead cells labeled positive by Hoechst or DAPI, side scatter (SSC-A) and forward scatter (FSC-A) and doublet exclusion using forward scatter width (FSC-W) against (FSC-A), as previously described. In order to calculate cell numbers per organ or per gram of tissue, organs were weighted, cell suspensions were prepared from a weighted amount (20 to 500mg) of tissue, and the number of cells per gram of tissue was determined using a cell counter (GUAVA easyCyte HT).
[0077] Statistical analysis and reproducibility. Data are shown as mean with individual values per mouse represented as circles, unless stated otherwise. Statistical significance was analyzed with GraphPad Prism using unpaired t-tests and two-way ANOVA with Tukey’s multiple comparisons test as indicated in the figure legends. Significance was considered at P value (P) *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. The n value represents biological replicates. Experiments were repeated to ensure reproducibility of the
observations. Equal variance was assumed for cell-counting experiments. No statistical methods were used to predetermine sample size. Example 1 : Osteoclast origins and maintenance
[0078] In vitro , osteoclasts arise by fusion of HSC-derived precursors and require expression of Csflr and Tnfrsfl la (Rank). To probe the origin of osteoclasts in vivo ,
CsflrCre; Csflr1111; and Csfif rc; Tnfrsfl la1111 mice were generated. These mice presented with an osteopetrotic phenotype similar to Csfl (op/op), Csflr , and Tnfrsfl la mutants and characterized in young mice by lack of teeth eruption, skull and skeletal deformities with shortness of long bones, increased bone density, and lack of osteoclasts and hematopoietic cells (Figure 1A; Figure 5B). To confirm that osteoclast differentiation requires expression of Tnfrsfl la and Csflr in HSC-derived precursors, Flt3Cre ; Tnfrsfl lcfi^ mice, 1Ή3( n‘;CsfIr/! /! mice, and VavCre; Csfir/lJlm\ce were generated. Surprisingly, all young mice had normal teeth (Figure IB) and bone morphology (Figure 6A-C), normal bone marrow cellularity (Figure 1C), and normal osteoclast numbers (Figure ID) in comparison to control littermates.
However, 1Ίΐ3( r ; Tnfrsfl I a1111 mice, Flt3Cre ;CsflrFfl mice, and VavCre; CsflrFA mice lost their osteoclasts over time (Figure ID), and by 22-60 weeks of age had increased trabecular bone density (Figure IE), decreased hematopoietic cell numbers in the long bones (Figure 1C; Figure 6D-G) and tri-dimensional X-ray imaging by micro computed tomography (micro- CT) confirmed increased bone mass while bone formation measured by calcein incorporation was similar to control(Figure IF, Figure 7A-E).
[0079] Mice lacking Csflr expression in Flt3Cre-ex pressing progenitors presented with a similar phenotype: Flt3Cre CsflrTl1 mice have normal teeth and bone morphology at 4 week of age (Figure IB, Figure 6C), but osteoclast number are decreased over time and bone mass is increased at 22 weeks of age (Figure 1D-E, Figure 7A-E). In a complementary approach to delete Csflr in HSC-derived progenitors, VavCre ;CsflrFfl mice were generated. Again,
VavCre CsflrFfl mice were not osteopetrotic at birth, had normal teeth (Figure IB), and hematopoietic cell numbers in long bones at 4 week of age (Figure 1C). However, they developed a late-onset osteopetrosis similar to Flt3Cre ;Tnfrsfl lcfT and Flt3Cre CsflrH1 mice: at ~60 weeks of age Vav( n';Csf I r/! 11 mice had decreased osteoclasts and hematopoietic cell numbers in long bones (Figure 1C-D), enlarged trabecular bone (Figure IE) and increased bone mass (Figure IF, Figure 7A-E). These data suggested that osteoclast development, tooth eruption and the development of bone and the bone marrow cavity requires precursors that express Csflr, but not Flt3Cre or VavCre although postnatal contribution of Flt3Cre / VavCre expressing cells is important for optimal osteoclast function in adults and aging mice. [0080] Fetal expression of CsflrCre and Flt3Cre was analyzed using a fluorescent reporter in CsflrCre; Rosa26LSL YFP and Flt3Cre; Rosa26LSL YFP mice (Figure 1 G-I). TRAP+
multinucleated cells that appear at embryonic day (E)l5 in ossification centers are labeled with YFP in CsflrCre;Rosa26LSL YFP mice and osteoclasts remain YFP positive throughout life (Figure 1 G,I; Figure 8A-E), but gain expression of YFP after birth in Flt3Cre ;Rosa26LSL YFP mice despite colonization of the fetal bone marrow by Flt3Cre+ YFP+ hematopoietic cells (Figure 1 H; Figure 8A-E). These data suggested that although postnatal contribution of HSC-derived cells is important for optimal osteoclast maintenance and function in adults and aging mice, osteoclast development, tooth eruption, the development of bone, and the bone marrow cavity requires precursors independent from the HSC lineage, possibly from the embryonic EMP lineage of resident macrophages because CsflrCre mice allow deletion of target genes in both the embryonic EMP lineage and in the HSC lineage, while Flt3Cre and VavCre (Figure 9A-D) are not expressed in the EMP lineage.
[0081] In support of this hypothesis, it was found that TRAP+ multinucleated cells develop in ~E 15 ossification centers from A/) i-deficient embryo, which lack HSC but still support the development of EMP-derived macrophages (Figure 2A, B). In addition, TRAP+
multinucleated cells are labeled with YFP in CsfhMer~lCre~Mer; Rosa26LSL~YFP mice pulsed at E8.5 with a single dose of hydroxytamoxifen (4-OHT), which labels EMP but not HSC
(Figure 2C, D; Figure 8A-E). Altogether, these results indicate that fetal osteoclasts arise from EMP in ossification centers.
[0082] Whether EMP are required for bone development was investigated. Tnfrsflla is expressed by osteoclasts, but its expression is also a hallmark of EMP-derived pMacs that colonize the developing embryo. In two independent lines oiTnfrsfl laCre knock-in mice, Cre- mediated expression of a Rosa26LSL~YFP fluorescent reporter is achieved with high efficiency in fetal macrophages but with low efficiency or not at all in HSC and their progeny in blood and tissues (Figure 9A-D). It was therefore hypothesized that conditional deletion of Csflr in TnfrsfllaCre would recapitulate macrophage deficiency observed in C.s/7/ -deficient mice, while leaving the HSC lineage unaffected. To test osteoclast and bone development in this model, two independent lines of Tnfrsfl laCre ;CsflrFA mice were generated (Figure 2E-J). Tnfrsfl laCre ;CsflrFfl lack tissue macrophages such as brain microglia and epidermal
Langerhans cells at birth while development of HSCs and blood cells was preserved (Figure 6A-N). They present with a severe osteopetrotic phenotype including the lack of tooth eruption (Figure 2E), misshaped skulls and short long bones (Figure 2F-H; Figure 5A-G) with few osteoclasts (Figure 21), increased bone density and initially lack a bone marrow cavity (Figure 2F; Figure 6A-N). In contrast to C.s/// -deficient mice, however, osteoclasts and hematopoietic cells progressively colonized the long bones of Tnfrsfl laCre iCsflr^ mice during the first month of life (Figure 21; Figure 5A-G; Figure 6A-N) although the mice remained toothless and skull and long bone deformity persisted (Figure 2E-H; Figure 5A- G). Calcein incorporation was similar in Tnfrsl Id r, ;CsfIr1111 mice and their littermate controls (Figure 10A-F). In a complementary approach ablation of Csflr expression in E10.5 embryos using a single dose of tamoxifen in Rosa26-creERT2+ ,-Csflr^ mice resulted in defective tooth eruption in 3 out of 4 pups at 21 days of age (Figure 9A-D). Altogether, this analysis supports a model in which EMP-derived embryonic osteoclasts are needed for teeth eruption, normal skull shape and optimal formation of long bones, and the timely
colonization of long bones by hematopoietic progenitors, while HSC-derived osteoclasts are important for the maintenance of bone mass after birth and later in life, although they may partially rescue bone development in the absence of EMP-derived osteoclasts in
Difrsl Id re;Csflr1111 mice.
Example 2: Therapeutic approach for the treatment of osteopetrosis
[0083] To probe the mechanisms that underlie the contribution of HSC-derived blood cells to osteoclast maintenance as well as the lifespan and dynamics of osteoclasts in vivo , time- course parabiosis experiments were performed (Figure 3A). After 4 to 8 weeks of shared blood circulation between CsflrCre ;Rosa26LSL~YFP and CsflrCre ;Rosa26LSL tdTomato parabionts, all osteoclasts, defined as TRAP+ multinucleated cells lining the bone surface, expressed both YFP and tdTomato within the same cells (Figure 3A, B). No other cell was found to co- express YFP and tdTomato in bones (Figure 3B). This is compatible with the presence of nuclei from both partners in individual osteoclasts. Moreover, when parabionts were separated after 4 weeks (Figure 3C), most recipient YFP+ osteoclasts retained tdTomato staining 14 weeks after separation, and two third of osteoclasts from former parabionts still expressed both YFP and tdTomato 24 weeks after separation (Figure 3C). TdTomato signal intensity per YFP osteoclast increased during the 8-week period of shared blood circulation, and decreased after separation (Figure 11A-C). Most murine osteoclasts have ~5 (3 to 7) nuclei, with a modest increase of nuclei per cell between 1 month and 6 months of age
(Figure 3E). These data therefore suggest that individual osteoclast syncytia are long lived, but acquire new nuclei one by one every 4 to 8 weeks from circulating blood cells so that it takes more than 6 months to renew all 5 nuclei from a given osteoclast.
[0084] The number and fusion-rate of HSC-derived nuclei acquired by osteoclasts in short term 5-ethynyl-2’-deoxyuridine (EdU) incorporation studies was calculated. A single intravenous pulse of EdU (20pg/g) labels mitotic nuclei, is bioavailable in the bone marrow for ~90 min, and -50% of bone marrow and blood monocytic cells are EdU+ for ~48hrs
(Figure 11A-C). It was observed that -1 to 2% of osteoclasts were labelled after 72 hrs, with only 1 EdU+ nucleus per osteoclast in 90% of positive cells (Figure 3F) suggesting acquisition by osteoclasts of single post-mitotic nuclei at a time. The proportion of osteoclasts acquiring a new nucleus can be estimated -0.5 to 2 % per day in this model, compatible with individual nuclei being replaced every - 2 months. Altogether the studies above suggested a model (Figure 3G) where: osteoclasts that control skeletal development develop in ossification centers from EPM, while their post-natal maintenance is mediated via the serial acquisition by long-lived syncytia of new nuclei from HSC-derived blood leukocytes, rather than by de novo renewal by lateral fusion or proliferation of osteoclast precursors.
[0085] A prediction from this model is that osteopetrosis due to a recessive mutation affecting osteoclasts function may be rescued or prevented through parabiosis with a wild- type partner. Parabiosis experiments between 4 week-old cathepsin K deficient mice, which develop an adult-onset form of osteopetrosis known as pycnodysostosis, and cathepsin K+/ or cathepsin K littermates and between wild type mice as control showed a reduction of bone volume in 10 weeks old cathepsin K mice paired with cathepsin K+/ littermates (Figure 4A), suggesting that circulating blood cells carrying a wild type cathepsin K allele are sufficient to reduce bone density. To confirm that expression of a donor-derived gene by recipient osteoclasts results from fusion with blood circulating monocytic cells, intravenous injections of Kit Ly6C+ cells from the bone marrow of CsflrCre;Rosa26LSL tdTomato into CsflrCre;Rosa26LSL YFP recipients were performed (Figure 4B; Figure 12A-C). This resulted in stable expression of tdTomato in 20-40% of osteoclasts 1 week and 8 weeks after transfusion, in the absence of other donor-derived blood cells or bone marrow progenitors (Figure 4B, C; Figure 12A-C). These results suggest that parabiosis or an appropriate regimen of transfusion can achieve expression of a donor-derived gene by recipient osteoclasts in the absence of HSC chimerism, and in addition that this effect can last for several months. [0086] Partial rescue of osteopetrosis occurs postnatally in Tr/rsfl Ia( r, ;CsfIr1111 mice, suggesting that transfusion of monocytic cells may also be able to rescue bone development in early-onset congenital osteopetrosis in the absence of a bone marrow transplantation. Intra- peritoneal injections of Kit Ly6C+ monocytic cells from CsflrCre;Rosa26LSL YFP into CsflrCre; CsflrF/F neonates starting from post-natal day 5, resulted in complete or partial rescue of teeth eruption (Figure 4D) and long bone development as assessed by CT-Scan, with the development of a bone marrow cavity (Figure 4D) at day 14 in infant mice from 3 different litters (Figure 4D, Figure 13). In all mice, histology of femurs indicated the presence of numerous YFP+ TRAP+ osteoclasts lining the bone (Figure 4E, F). Histology and flow cytometry analyses showed the lack of YFP+ circulating blood cells or bone marrow progenitors (Figure 4E, G), indicating absence of HSC engraftment. These data suggest that transfusion of monocytic cells can rescue bone development in early-onset autosomal recessive osteopetrosis in infant mice, in the absence of HSC transplantation.
Summary
[0087] The results described above show that osteoclasts originating from EMP are essential for normal bone development. Moreover, it is shown that osteoclasts are long-lived in adult and their function is maintained by iterative fusion of individual HSC-derived circulating cells with existing syncytia. In the absence or deficiency of EMP-derived osteoclasts, however, their timely replacement by transfusion with monocytic cells can rescue bone development in early-onset osteopetrotic mice in the absence of bone marrow transplantation. This could contribute to the treatment of early onset osteopetrosis, since the current treatment by bone marrow transplantation. This is of potential clinical relevance because bone marrow/HSC transplantation, the standard of care in early-onset osteopetrosis in mice and human, requires irradiation or chemotherapy which carry the risk of threatening infections and is frequently performed late in patients who already suffer severe
complications and is plagued by a -48% overall survival at 6-years. In addition, the original mechanism that mediates osteoclast maintenance suggests that they represent a unique target for gene transfer by cellular therapies based on transfusion of wild type or engineered monocytic cells to modulate osteoclast activity and bone remodeling in adults. Accordingly, these results demonstrate that monocytic cells may be effective in methods for treating osteopetrosis, and to modulate osteoclast activity and bone remodeling. Example 3: Ex vivo gene therapy for the treatment of a form of osteopetrosis: pycnodvsostosis
[0088] This example demonstrates the prophetic use of monocytic cells obtained from a subject and engineered to express a gene, such as CTSK , for the treatment of osteopetrosis in the subject.
[0089] Pycnodysostosis is an autosomal recessive form of osteopetrosis, due to loss of function mutations in the cathepsin K gene (CTSK), compatible with life, and characterized by short stature, deformity of the skull maxilla and phalanges, increased density of the bones, osteosclerosis, and fragility of bone. There is no treatment for this disease as the relatively comparably milder symptoms do not qualify for bone marrow transplantation.
[0090] Subjects suspected of having or diagnosed as having cathepsin K deficiency will be selected for a gene therapy trial and will undergo GMP grade collection of peripheral blood monocytes by apheresis and/or elutriation. Purified monocytes will then be transduced with a lentivirus designed to express a cDNA coding for the wild type allele of CTSK under a strong promoter ( e.g ., CMV). Transduced monocytes will then be washed to remove contaminating viral particles and rested for 12 hours followed by reinfusion to the patient. Assuming -109 monocytes can be collected, the number of cells transferred per kilogram of body weight would be >l07. Following the auto-transfusion, the subject will be monitored at regular intervals for clinical and radiological signs of pycnodysostosis in order to determine the efficiency of the procedure and the frequency with which it should be repeated.
[0091] It is predicted that subjects suspected of having or diagnosed as having
pycnodysostosis and receiving therapeutically effective amounts of monocytic cells engineered to express CTSK will display reduced severity or elimination of one or more symptoms associated with pycnodysostosis. Accordingly, these results will show that transfusion of monocytic cells engineered to express the wild-type version of one or more genes implicated in osteopetrosis is useful in methods for treating subjects in need thereof for the treatment of osteopetrosis. Example 4: Ex vivo gene therapy for the treatment of a form of osteopetrosis: severe infantile autosomal recessive osteopetrosis
[0092] This example demonstrates the prophetic use of monocytic cells obtained from a subject and engineered to express a gene, such as TCIRG1, TNFRSFlla, CA2, CLCN7, OSTM1, and/or PLEKHM1, for the treatment of osteopetrosis in the subject.
[0093] Infants diagnosed with severe infantile autosomal recessive osteopetrosis with defects in TCIRG1, TNFRSFlla , CA2 , CLCN7 , OS/M I, and/or PLEKHM1 will be eligible for an early auto-transfusion of corrected monocytes (as described in Example 3), to try to prevent early developmental complications, including blindness and skeletal deformities, and increase the success of bone marrow transplantation, by restoring a bone marrow niche. The protocol will be carried out in a manner similar to that of Example 3, and monocytes will be transduced with a lentivirus designed to express a cDNA coding for the wild type allele of the deficient gene under a strong promoter, with a similar number of transferred cells per kg of weight. Following the auto-transfusion, subjects will be monitored at regular intervals for clinical and radiological signs, and possibly bone biopsy in order to determine the efficiency of the procedure. In addition, collection of monocytes and hematopoietic stem
cells/progenitors at the same time would allow to proceed with the auto transfusion of transduced monocytes, while stem cells would undergo gene editing in view of auto transplantation and long term genetic rescue.
[0094] It is predicted that subjects suspected of having or diagnosed as having severe infantile autosomal recessive osteopetrosis and receiving therapeutically effective amounts of monocytic cells engineered to express TCIRG1 , TNFRSFlla , CA2, CLCN7 , OSTM1, and/or PLEKHM1 will display reduced severity or elimination of one or more symptoms associated with severe infantile autosomal recessive osteopetrosis. Accordingly, these results will show that transfusion of monocytic cells engineered to express the wild-type version of one or more genes implicated in osteopetrosis is useful in methods for treating subjects in need thereof for the treatment of osteopetrosis. REFERENCES
The following references are incorporated by reference.
1. Coccia, P. F. et al. Successful bone-marrow transplantation for infantile malignant osteopetrosis. The New England journal of medicine 302, 701-708,
doi: l0.l056/NEJMl 98003273021301 (1980).
2. Sorell, M. et al. Marrow transplantation for juvenile osteopetrosis. The American journal of medicine 70, 1280-1287 (1981).
3. Lacey, D. L. et al. Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation. Cell 93, 165-176 (1998).
4. Wiktor-Jedrzejczak, W. et al. Total absence of colony-stimulating factor 1 in the macrophage-deficient osteopetrotic (op/op) mouse. Proceedings of the National Academy of Sciences 87, 4828-4832, doi: 10. l073/pnas.87.12.4828 (1990).
5. Kong, Y. Y. et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature 397, 315-323, dok lO.1038/16852 (1999).
6. Dougall, W. C. et al. RANK is essential for osteoclast and lymph node development. Genes & development 13, 2412-2424 (1999).
7. Hsu, H. et al. Tumor necrosis factor receptor family member RANK mediates osteoclast differentiation and activation induced by osteoprotegerin ligand. Proceedings of the National Academy of Sciences of the ETnited States of America 96, 3540-3545 (1999).
8. Yoshida, H. et al. The murine mutation osteopetrosis is in the coding region of the macrophage colony stimulating factor gene. Nature 345, 442-444 (1990).
9. Dai, X.-M. et al. Targeted disruption of the mouse colony-stimulating factor 1 receptor gene results in osteopetrosis, mononuclear phagocyte deficiency, increased primitive progenitor cell frequencies, and reproductive defects. Blood 99, 111-120,
doi: 10.1182/blood. V99.1.111 (2002). 10. Luchin, A. et al. Genetic and physical interactions between Microphthalmia transcription factor and PU.1 are necessary for osteoclast gene expression and differentiation. The Journal of biological chemistry 276, 36703-36710, doi: l0.l074/jbc.Ml 06418200 (2001).
11. Grigoriadis, A. E. et al. c-Fos: a key regulator of osteoclast-macrophage lineage determination and bone remodeling. Science 266, 443-448 (1994).
12. Lomaga, M. A. et al. TRAF6 deficiency results in osteopetrosis and defective interleukin- 1, CD40, and LPS signaling. Genes & development 13, 1015-1024 (1999).
13. Tondravi, M. M. et al. Osteopetrosis in mice lacking haematopoietic transcription factor PU.l. Nature 386, 81-84, doi:l0. l038/38608la0 (1997).
14. Takayanagi, H. et al. Induction and activation of the transcription factor NFATcl (NFAT2) integrate RANKL signaling in terminal differentiation of osteoclasts.
Developmental cell 3, 889-901 (2002).
15. Lau, R. Y. & Guo, X. A review on current osteoporosis research: with special focus on disuse bone loss. J Osteoporos 2011, 293808, doi: 10.4061/2011/293808 (2011).
16. Walker, D. G. Bone resorption restored in osteopetrotic mice by transplants of normal bone marrow and spleen cells. Science 190, 784-785 (1975).
17. Walker, D. G. Control of bone resorption by hematopoietic tissue. The induction and reversal of congenital osteopetrosis in mice through use of bone marrow and splenic transplants. The Journal of experimental medicine 142, 651-663 (1975).
18. Ballet, J. J., Griscelli, C., Coutris, C., Milhaud, G. & Maroteaux, P. Bone-marrow transplantation in osteopetrosis. Lancet 2, 1137 (1977).
19. Nisbet, N. W., Menage, J. & Loutit, J. F. Bone-marrow transplantation in
osteopetrosis. Lancet 2, 1236 (1977).
20. Ash, P., Loutit, J. F. & Townsend, K. M. Osteoclasts derived from haematopoietic stem cells. Nature 283, 669-670 (1980).
21. Orchard, P. J. et al. Hematopoietic stem cell transplantation for infantile osteopetrosis. Blood 126, 270-276, doi: l0. H82/blood-20l5-0l-62554l (2015). 22. Xiao, Y. et al. Identification of the Common Origins of Osteoclasts, Macrophages, and Dendritic Cells in Human Hematopoiesis. Stem cell reports 4, 984-994,
doi: l0.l0l6/j.stemcr.20l5.04.012 (2015).
23. Shalhoub, V. et al. Characterization of osteoclast precursors in human blood. British journal of haematology 111, 501-512 (2000).
24. Stadtfeld, M. & Graf, T. Assessing the role of hematopoietic plasticity for endothelial and hepatocyte development by non-invasive lineage tracing. Development 132, 203-213, doi: l0.l242/dev.0l558 (2005).
25. Taniguchi, N. et al. Stage-specific secretion of HMGB1 in cartilage regulates endochondral ossification. Molecular and cellular biology 27, 5650-5663,
doi: 10.1128/MCB.00130-07 (2007).
26. Schulz, C. et al. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science 336, 86-90, doi: 10. H26/science.1219179 (2012).
27. Gomez Perdiguero, E. et al. Tissue-resident macrophages originate from yolk-sac- derived erythro-myeloid progenitors. Nature 518, 547-551, doi: l0.l038/naturel3989 (2015).
28. Mass, E. et al. Specification of tissue-resident macrophages during organogenesis. Science 353, doi: l0.H26/science.aaf4238 (2016).
29. Deng, L. et al. A novel mouse model of inflammatory bowel disease links mammalian target of rapamycin-dependent hyperproliferation of colonic epithelium to inflammation- associated tumorigenesis. Am J Pathol 176, 952-967, doi: 10.2353/ajpath.2010.090622 (2010).
30. Qian, B. Z. et al. CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature 475, 222-225, doi:l0. l038/naturel0l38 (2011).
31. Li, J., Chen, K., Zhu, L. & Pollard, J. W. Conditional deletion of the colony stimulating factor-l receptor (c-fms proto-oncogene) in mice. Genesis 44, 328-335, doi: l0.l002/dvg.202l9 (2006). 32. Dai, X. M. et al. Targeted disruption of the mouse colony-stimulating factor 1 receptor gene results in osteopetrosis, mononuclear phagocyte deficiency, increased primitive progenitor cell frequencies, and reproductive defects. Blood 99, 111-120 (2002).
33. Benz, C., Martins, V. C., Radtke, F. & Bleul, C. C. The stream of precursors that colonizes the thymus proceeds selectively through the early T lineage precursor stage of T cell development. The Journal of experimental medicine 205, 1187-1199,
doi: l0.l084/jem.20072l68 (2008).
34. Mucenski, M. L. et al. A functional c-myb gene is required for normal murine fetal hepatic hematopoiesis. Cell 65, 677-689 (1991).
35. Stadtfeld, M. & Graf, T. Assessing the role of hematopoietic plasticity for endothelial and hepatocyte development by non-invasive lineage tracing. Development 132, 203-213, doi: l0.l242/dev.0l558 (2005).
36. Hanada, R. et al. Central control of fever and female body temperature by
RANKL/RANK. Nature 462, 505-509, doi: l0.l038/nature08596 (2009).
37. Maeda, K. et al. Wnt5a-Ror2 signaling between osteoblast-lineage cells and osteoclast precursors enhances osteoclastogenesis. Nature medicine 18, 405-412, doi:l0. l038/nm.2653 (2012).
38. Hameyer, D. et al. Toxicity of ligand-dependent Cre recombinases and generation of a conditional Cre deleter mouse allowing mosaic recombination in peripheral tissues. Physiol Genomics 31, 32-41, doi: lO.H52/physiolgenomics.00019.2007 (2007).
39. Srinivas, S. et al. Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus. BMC developmental biology 1, 4 (2001).
40. Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat Neurosci 13, 133-140, doi : 10.1038/nn.2467 (2010).
41. Schulz, C. et al. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science 336, 86-90, doi: 10. H26/science.1219179 (2012).
42. Gomez Perdiguero, E. et al. Tissue-resident macrophages originate from yolk-sac- derived erythro-myeloid progenitors. Nature 518, 547-551, doi: l0.l038/naturel3989 (2015). 43. Kamran, P. et al. Parabiosis in mice: a detailed protocol. J Vis Exp,
doi: l0.3791/50556 (2013).
44. Jiang, X. et al. Histological analysis of GFP expression in murine bone. J Histochem Cytochem 53, 593-602, doi: l0. l369/jhc.4A640l.2005 (2005).
45. Filgueira, L. Fluorescence-based staining for tartrate-resistant acidic phosphatase (TRAP) in osteoclasts combined with other fluorescent dyes and protocols. J Histochem Cytochem 52, 411-414, doi: 10.1177/002215540405200312 (2004).
46. Mass, E. et al. Specification of tissue-resident macrophages during organogenesis. Science 353, doi: l0.H26/science.aaf4238 (2016).
47. Arndt, K. et al. SETD1 A protects HSCs from activation-induced functional decline in vivo. Blood 131, 1311-1324, doi: 10. H82/blood-2017-09-806844 (2018).
48. Grinenko, T. et al. Clonal expansion capacity defines two consecutive developmental stages of long-term hematopoietic stem cells. The Journal of experimental medicine 211, 209-215, doi: l0. l084/jem.20l3 l l l5 (2014).
EQUIVALENTS
[0095] The present technology is not to be limited in terms of the particular embodiments described in this application, which are intended as single illustrations of individual aspects of the present technology. Many modifications and variations of this present technology can be made without departing from its spirit and scope, as will be apparent to those skilled in the art. Functionally equivalent methods and apparatuses within the scope of the present technology, in addition to those enumerated herein, will be apparent to those skilled in the art from the foregoing descriptions. Such modifications and variations are intended to fall within the scope of the appended claims. The present technology is to be limited only by the terms of the appended claims, along with the full scope of equivalents to which such claims are entitled. It is to be understood that this present technology is not limited to particular methods, reagents, compounds compositions or biological systems, which can, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting.
[0096] In addition, where features or aspects of the disclosure are described in terms of Markush groups, those skilled in the art will recognize that the disclosure is also thereby described in terms of any individual member or subgroup of members of the Markush group.
[0097] As will be understood by one skilled in the art, for any and all purposes, particularly in terms of providing a written description, all ranges disclosed herein also encompass any and all possible subranges and combinations of subranges thereof. Any listed range can be easily recognized as sufficiently describing and enabling the same range being broken down into at least equal halves, thirds, quarters, fifths, tenths, etc. As a nonlimiting example, each range discussed herein can be readily broken down into a lower third, middle third and upper third, etc. As will also be understood by one skilled in the art all language such as“up to,”“at least,”“greater than,”“less than,” and the like, include the number recited and refer to ranges which can be subsequently broken down into subranges as discussed above. Finally, as will be understood by one skilled in the art, a range includes each individual member. Thus, for example, a group having 1-3 cells refers to groups having 1, 2, or 3 cells. Similarly, a group having 1-5 cells refers to groups having 1, 2, 3, 4, or 5 cells, and so forth. [0098] All patents, patent applications, provisional applications, and publications referred to or cited herein are incorporated by reference in their entirety, including all figures and tables, to the extent they are not inconsistent with the explicit teachings of this specification.
[0099] Other embodiments are set forth within the following claims.

Claims

CLAIMS WHAT IS CLAIMED IS:
1. A method for treating or preventing osteopetrosis in a subject in need thereof, the method comprising administering a composition comprising a therapeutically effective amount of monocytic cells from a healthy donor to the subject.
2. The method of claim 1, wherein the subject is characterized by decreased expression of one or more of CA2, CLCN7 , CTSK, CSF1R , IKBKG, ITGB3, OSTM1, PLEKHM1 , TCIRG1 , TNFRSF11A, and TNFSF11, as compared to the monocytic cells of the donor.
3. The method of claim 1, wherein the osteopetrosis comprises one or more of stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia.
4. The method of claim 1, wherein the composition is formulated for intravenous
administration by injection, infusion, or transfusion.
5. The method of claim 1, wherein the subject is a mammal.
6. The method of claim 5, wherein the mammalian subject is a human.
7. The method of claim 1, wherein the subject is characterized by a cathepsin K
deficiency.
8. A method for treating or preventing osteopetrosis in a subject in need thereof, the method comprising administering to the subject a composition comprising a therapeutically effective amount of monocytic cells engineered to express one or more genes selected from CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1 ,
PLEKHM1, TCIRG1 , TNFRSF11A , and TNFSF11.
9. The method of claim 7, wherein the osteopetrosis comprises one or more of stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia.
10. The method of claim 8, wherein the composition is formulated for intravenous
administration by injection, infusion, or transfusion.
11. The method of claim 8, wherein the subject is a mammal.
12. The method of claim 11, wherein the mammalian subject is a human.
13. The method of claim 8, wherein the subject is characterized by a cathepsin K
deficiency.
14. The method of claim 8, wherein the monocytic cells are obtained from the subject.
15. A donor monocytic cell line engineered to express one or more genes selected from CA2 , CLCN7 , CTSK, CSF1R , IKBKG, ITGB3, OSTM1, PLEKHM1 , TCIRG1 , TNFRSF11A , and TNFSF11 , wherein the one or more genes is operably linked to a heterologous nucleic acid to form a chimeric nucleic acid construct.
16. The donor monocytic cell line of claim 15, wherein the heterologous nucleic acid encodes a selectable marker.
17. The donor monocytic cell line of claim 16, wherein the selectable marker is a
bioluminescent protein, a fluorescent protein, a chemiluminescent protein, a xanthine- guanine phosphoribosyl transferase gene (gpt), or any combination thereof.
18. The donor monocytic cell line of claim 15, wherein the heterologous nucleic acid encodes one or more control sequences suitable for directing expression of the one or more genes in a monocytic cell.
19. The donor monocytic cell line of claim 18, wherein the one or more control sequences comprises a promoter.
20. The donor monocytic cell line of claim 15, wherein the cells comprise a vector
encoding one or more genes selected from CA2, CLCN7 , CTSK , CSF1R, IKBKG, ITGB3 , OS/MI, PLEKHM1, TCIRG1, TNFRSF11A, and TNFSF11.
The donor monocytic cell line of claim 20, wherein the vector is a mammalian expression vector, a lentiviral vector, or transposon vector.
PCT/US2019/034984 2018-06-01 2019-05-31 Methods and compositions for the treatment of osteopetrosis WO2019232426A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP19809981.4A EP3801589A4 (en) 2018-06-01 2019-05-31 Methods and compositions for the treatment of osteopetrosis
US17/058,830 US20210205360A1 (en) 2018-06-01 2019-05-31 Methods and compositions for the treatment of osteopetrosis

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862679553P 2018-06-01 2018-06-01
US62/679,553 2018-06-01

Publications (1)

Publication Number Publication Date
WO2019232426A1 true WO2019232426A1 (en) 2019-12-05

Family

ID=68698991

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/034984 WO2019232426A1 (en) 2018-06-01 2019-05-31 Methods and compositions for the treatment of osteopetrosis

Country Status (3)

Country Link
US (1) US20210205360A1 (en)
EP (1) EP3801589A4 (en)
WO (1) WO2019232426A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117551619B (en) * 2024-01-10 2024-04-26 广东工业大学 THP-1 cell with high osteoclast differentiation capability, osteoclast, preparation and application

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140127268A1 (en) * 2007-01-11 2014-05-08 Inserm (Institut National De La Sante Et De La Recherche Medicale) Method for Expanding Monocytes
US20150353890A1 (en) 2013-01-08 2015-12-10 Fred Hutchinson Cancer Research Center Compositions and methods for expansion of embryonic hematopoietic stem cells
US9452187B2 (en) 2010-04-12 2016-09-27 The Trustees Of The University Of Pennsylvania In vivo and ex vivo expansion of hematopoietic stem cells with a targeted combination of clinically tested, FDA approved drugs

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2015523058A (en) * 2012-05-10 2015-08-13 バイオマトセル アクチエボラグBiomatcell AB Osteogenic differentiation of mesenchymal stem cells
EP3310365B1 (en) * 2015-06-16 2023-08-23 Fondazione Città Della Speranza - Onlus Extracellular vesicles derived from osteoblastic lineage cells for therapeutic and diagnostic use

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140127268A1 (en) * 2007-01-11 2014-05-08 Inserm (Institut National De La Sante Et De La Recherche Medicale) Method for Expanding Monocytes
US9452187B2 (en) 2010-04-12 2016-09-27 The Trustees Of The University Of Pennsylvania In vivo and ex vivo expansion of hematopoietic stem cells with a targeted combination of clinically tested, FDA approved drugs
US20150353890A1 (en) 2013-01-08 2015-12-10 Fred Hutchinson Cancer Research Center Compositions and methods for expansion of embryonic hematopoietic stem cells

Non-Patent Citations (48)

* Cited by examiner, † Cited by third party
Title
"pEGFP-N1 Vector Information", PROTOCOL # PT 3027-5, 19 March 1999 (1999-03-19), pages 1 - 3, XP055658138, Retrieved from the Internet <URL:http://www.yrgene.com/documents/vector/pegfpn1.pdf> [retrieved on 20190915] *
ARNDT, K. ET AL.: "SETD1A protects HSCs from activation-induced functional decline in vivo.", BLOOD, vol. 131, 2018, pages 1311 - 1324
ASH, P.LOUTIT, J. F.TOWNSEND, K. M.: "Osteoclasts derived from haematopoietic stem cells.", NATURE, vol. 283, 1980, pages 669 - 670
BALLET, J. J.GRISCELLI, C.COUTRIS, C.MILHAUD, G.MAROTEAUX, P.: "Bone-marrow transplantation in osteopetrosis.", LANCET, vol. 2, 1977, pages 1236
BENZ, C.MARTINS, V. C.RADTKE, F.BLEUL, C. C.: "The stream of precursors that colonizes the thymus proceeds selectively through the early T lineage precursor stage of T cell development.", THE JOURNAL OF EXPERIMENTAL MEDICINE, vol. 205, 2008, pages 1187 - 1199
COCCIA, P. F. ET AL.: "Successful bone-marrow transplantation for infantile malignant osteopetrosis.", THE NEW ENGLAND JOURNAL OF MEDICINE, vol. 302, 1980, pages 701 - 708
DAI, X. M. ET AL.: "Targeted disruption of the mouse colony-stimulating factor 1 receptor gene results in osteopetrosis, mononuclear phagocyte deficiency, increased primitive progenitor cell frequencies, and reproductive defects.", BLOOD, vol. 99, 2002, pages 111 - 120, XP002505054, DOI: 10.1182/blood.V99.1.111
DENG, L. ET AL.: "A novel mouse model of inflammatory bowel disease links mammalian target of rapamycin-dependent hyperproliferation of colonic epithelium to inflammation-associated tumorigenesis.", AM J PATHOL, vol. 176, pages 952 - 967
DOUGALL ET AL.: "RANK is essential for osteoclast and lymph node development", GENES DEV, vol. 13, 15 September 1999 (1999-09-15), pages 2412 - 2424, XP002228678, DOI: 10.1101/gad.13.18.2412 *
DOUGALL, W. C. ET AL.: "RANK is essential for osteoclast and lymph node development.", GENES & DEVELOPMENT, vol. 13, 1999, pages 2412 - 2424, XP002228678, DOI: 10.1101/gad.13.18.2412
FILGUEIRA, L.: "Fluorescence-based staining for tartrate-resistant acidic phosphatase (TRAP) in osteoclasts combined with other fluorescent dyes and protocols.", J HISTOCHEM CYTOCHEM, vol. 52, 2004, pages 411 - 414, XP008033696
GOMEZ PERDIGUERO, E. ET AL.: "Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors.", NATURE, vol. 518, 2015, pages 547 - 551, XP055477968, DOI: 10.1038/nature13989
GRIGORIADIS, A. E. ET AL.: "c-Fos: a key regulator of osteoclast-macrophage lineage determination and bone remodeling.", SCIENCE, vol. 266, 1994, pages 443 - 448
GRINENKO, T. ET AL.: "Clonal expansion capacity defines two consecutive developmental stages of long-term hematopoietic stem cells.", THE JOURNAL OF EXPERIMENTAL MEDICINE, vol. 211, 2014, pages 209 - 215
GUERRINI ET AL.: "Human osteoclast-poor osteopetrosis with hypogammaglobulinemia due to TNFRSF11A (RANK) mutations", AM J HUM GENET, vol. 83, 11 July 2008 (2008-07-11), pages 64 - 76, XP055658131 *
HAMEYER, D. ET AL.: "Toxicity of ligand-dependent Cre recombinases and generation of a conditional Cre deleter mouse allowing mosaic recombination in peripheral tissues.", PHYSIOL GENOMICS, vol. 31, 2007, pages 32 - 41, XP002525189, DOI: 10.1152/PHYSIOLGENOMICS.00019.2007
HANADA, R. ET AL.: "Central control of fever and female body temperature by RANKL/RANK.", NATURE, vol. 462, 2009, pages 505 - 509
HSU, H. ET AL.: "Tumor necrosis factor receptor family member RANK mediates osteoclast differentiation and activation induced by osteoprotegerin ligand.", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 96, 1999, pages 3540 - 3545
JACOME-GALARZA ET AL.: "Developmental origin, functional maintenance and genetic rescue of osteoclasts", NATURE, vol. 568, 10 April 2019 (2019-04-10), pages 541 - 545, XP036900382, DOI: 10.1038/s41586-019-1105-7 *
JIANG, X. ET AL.: "Histological analysis of GFP expression in murine bone.", J HISTOCHEM CYTOCHEM, vol. 53, 2005, pages 593 - 602
KAMRAN, P ET AL.: "Parabiosis in mice: a detailed protocol.", J VIS EXP, 2013
KONG, Y. Y. ET AL.: "OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis.", NATURE, vol. 397, 1999, pages 315 - 323, XP003018739, DOI: 10.1038/16852
LACEY, D. L. ET AL.: "Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation.", CELL, vol. 93, 1998, pages 165 - 176, XP002122920, DOI: 10.1016/S0092-8674(00)81569-X
LAU, R. Y.GUO, X. A: "review on current osteoporosis research: with special focus on disuse bone loss.", J OSTEOPOROS, 2011, pages 293808
LI, J.CHEN, K.ZHU, L.POLLARD, J. W.: "Conditional deletion of the colony stimulating factor-1 receptor (c-fms proto-oncogene) in mice.", GENESIS, vol. 44, 2006, pages 328 - 335
LOMAGA, M. A. ET AL.: "TRAF6 deficiency results in osteopetrosis and defective interleukin-1, CD40, and LPS signaling.", GENES & DEVELOPMENT, vol. 13, 1999, pages 1015 - 1024
LUCHIN, A. ET AL.: "Genetic and physical interactions between Microphthalmia transcription factor and PU. 1 are necessary for osteoclast gene expression and differentiation.", THE JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 276, 2001, pages 36703 - 36710
MADISEN, L. ET AL.: "A robust and high-throughput Cre reporting and characterization system for the whole mouse brain.", NAT NEUROSCI, vol. 13, 2010, pages 133 - 140, XP055199562, DOI: 10.1038/nn.2467
MAEDA, K. ET AL.: "Wnt5a-Ror2 signaling between osteoblast-lineage cells and osteoclast precursors enhances osteoclastogenesis.", NATURE MEDICINE, vol. 18, 2012, pages 405 - 412
MASS, E. ET AL.: "Specification of tissue-resident macrophages during organogenesis.", SCIENCE, vol. 353, 2016
MUCENSKI, M. L. ET AL.: "A functional c-myb gene is required for normal murine fetal hepatic hematopoiesis.", CELL, vol. 65, 1991, pages 677 - 689, XP023884312, DOI: 10.1016/0092-8674(91)90099-K
ORCHARD, P. J. ET AL.: "Hematopoietic stem cell transplantation for infantile osteopetrosis.", BLOOD, vol. 126, 2015, pages 270 - 276
QIAN, B. Z. ET AL.: "CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis.", NATURE, vol. 475, 2011, pages 222 - 225
SAFTIG ET AL.: "Impaired osteoclastic bone resorption leads to osteopetrosis in cathepsin-K-deficient mice", PROC NATL ACAD SCI USA, vol. 95, 10 November 1998 (1998-11-10), pages 13453 - 13458, XP008111281 *
SCHULZ, C. ET AL.: "A lineage of myeloid cells independent of Myb and hematopoietic stem cells.", SCIENCE, vol. 336, no. 86-90, 2012, pages 1219179 - 90
See also references of EP3801589A4
SHALHOUB, V ET AL.: "Characterization of osteoclast precursors in human blood.", BRITISH JOURNAL OF HAEMATOLOGY, vol. 111, 2000, pages 501 - 512
SORELL, M. ET AL.: "Marrow transplantation for juvenile osteopetrosis.", THE AMERICAN JOURNAL OF MEDICINE, vol. 70, 1981, pages 1280 - 1287, XP026386410, DOI: 10.1016/0002-9343(81)90839-1
SRINIVAS, S. ET AL.: "Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus.", BMC DEVELOPMENTAL BIOLOGY, vol. 1, no. 4, 2001, XP021014128, DOI: 10.1186/1471-213X-1-4
STADTFELD, M.GRAF, T.: "Assessing the role of hematopoietic plasticity for endothelial and hepatocyte development by non-invasive lineage tracing.", DEVELOPMENT, vol. 132, 2005, pages 203 - 213
TAKAYANAGI, H. ET AL.: "Induction and activation of the transcription factor NFATcl (NFAT2) integrate RANKL signaling in terminal differentiation of osteoclasts.", DEVELOPMENTAL CELL, vol. 3, 2002, pages 889 - 901, XP002994026, DOI: 10.1016/S1534-5807(02)00369-6
TANIGUCHI, N. ET AL.: "Stage-specific secretion of HMGB 1 in cartilage regulates endochondral ossification.", MOLECULAR AND CELLULAR BIOLOGY, vol. 27, 2007, pages 5650 - 5663, XP055709081, DOI: 10.1128/MCB.00130-07
TONDRAVI, M. M. ET AL.: "Osteopetrosis in mice lacking haematopoietic transcription factor PU. 1.", NATURE, vol. 386, 1997, pages 81 - 84
WALKER, D. G.: "Bone resorption restored in osteopetrotic mice by transplants of normal bone marrow and spleen cells.", SCIENCE, vol. 190, 1975, pages 784 - 785, XP008081127, DOI: 10.1126/science.1105786
WALKER, D. G.: "Control of bone resorption by hematopoietic tissue. The induction and reversal of congenital osteopetrosis in mice through use of bone marrow and splenic transplants.", THE JOURNAL OF EXPERIMENTAL MEDICINE, vol. 142, 1975, pages 651 - 663, XP008081131, DOI: 10.1084/jem.142.3.651
WIKTOR-JEDRZEJCZAK, W. ET AL.: "Total absence of colony-stimulating factor 1 in the macrophage-deficient osteopetrotic (op/op) mouse.", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 87, 1990, pages 4828 - 4832, XP002414609, DOI: 10.1073/pnas.87.12.4828
XIAO, Y. ET AL.: "Identification of the Common Origins of Osteoclasts, Macrophages, and Dendritic Cells in Human Hematopoiesis.", STEM CELL REPORTS, vol. 4, 2015, pages 984 - 994
YOSHIDA, H. ET AL.: "The murine mutation osteopetrosis is in the coding region of the macrophage colony stimulating factor gene.", NATURE, vol. 345, 1990, pages 442 - 444, XP002414608, DOI: 10.1038/345442a0

Also Published As

Publication number Publication date
EP3801589A4 (en) 2022-03-02
EP3801589A1 (en) 2021-04-14
US20210205360A1 (en) 2021-07-08

Similar Documents

Publication Publication Date Title
Jacome-Galarza et al. Developmental origin, functional maintenance and genetic rescue of osteoclasts
Ke et al. Embryogenesis and adult life in the absence of intrinsic apoptosis effectors BAX, BAK, and BOK
Lunney et al. Importance of the pig as a human biomedical model
Pridans et al. Pleiotropic impacts of macrophage and microglial deficiency on development in rats with targeted mutation of the Csf1r locus
Zhang et al. Inflammasome activation has an important role in the development of spontaneous colitis
Navankasattusas et al. The netrin receptor UNC5B promotes angiogenesis in specific vascular beds
Lever et al. Parabiosis reveals leukocyte dynamics in the kidney
Xue et al. Gene editing in a Myo6 semi-dominant mouse model rescues auditory function
CN110100788A (en) Methods and applications based on gene manipulation strategy building disease model
Fujinaga et al. Evidence for an adrenergic mechanism in the control of body asymmetry
Novak et al. Osteoclasts derive predominantly from bone marrow–resident CX3CR1+ precursor cells in homeostasis, whereas circulating CX3CR1+ cells contribute to osteoclast development during fracture repair
Kousa et al. IRF6 expression in basal epithelium partially rescues Irf6 knockout mice
Yu et al. Nuclear factor of activated T cells 2 is required for osteoclast differentiation and function in vitro but not in vivo
Liu et al. Molecular and cellular mechanisms of the first social relationship: A conserved role of 5-HT from mice to monkeys, upstream of oxytocin
WO2019232426A1 (en) Methods and compositions for the treatment of osteopetrosis
Chi et al. Sexual dimorphism in skin immunity is mediated by an androgen-ILC2-dendritic cell axis
Song et al. Development of the nude rabbit model
JP6172699B2 (en) Hyperlipidemia model pig
Tondelli et al. Fetal liver cells transplanted in utero rescue the osteopetrotic phenotype in the oc/oc mouse
Fujino et al. Generation and mutational analysis of a transgenic murine model of the human MAF mutation
Liang et al. The most common human ADAR1p150 Zα domain mutation P193A is well tolerated in mice and does not activate the integrated stress response pathway
Yu et al. Induction of the Hajdu-Cheney syndrome mutation in CD19 B cells in mice alters B-cell allocation but not skeletal homeostasis
Maes et al. Mitochondrial network adaptations of microglia reveal sex-specific stress response after injury and UCP2 knockout
Gullard et al. Reduced Dentin Matrix Protein Expression in Camurati‐Engelmann Disease Transgenic Mouse Model
Kang et al. Survival function of the FADD-CASPASE-8-cFLIPL complex

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: 19809981

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2019809981

Country of ref document: EP

Effective date: 20210111