WO2008144820A1 - Treatment of chronic lung disease - Google Patents

Treatment of chronic lung disease Download PDF

Info

Publication number
WO2008144820A1
WO2008144820A1 PCT/AU2008/000753 AU2008000753W WO2008144820A1 WO 2008144820 A1 WO2008144820 A1 WO 2008144820A1 AU 2008000753 W AU2008000753 W AU 2008000753W WO 2008144820 A1 WO2008144820 A1 WO 2008144820A1
Authority
WO
WIPO (PCT)
Prior art keywords
lung
aecs
haecs
subject
condition
Prior art date
Application number
PCT/AU2008/000753
Other languages
French (fr)
Inventor
Graham Jenkin
Alan Osborne Trounson
Euan Morrison Wallace
Yuben Moodley
Ursula Manuelpillai
Sivakami Ilancheran
Original Assignee
Monash University
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
Priority claimed from AU2007902844A external-priority patent/AU2007902844A0/en
Application filed by Monash University filed Critical Monash University
Priority to JP2010509623A priority Critical patent/JP5567476B2/en
Priority to EP08748014.1A priority patent/EP2164505B1/en
Priority to AU2008255634A priority patent/AU2008255634B2/en
Priority to US12/601,570 priority patent/US20100266552A1/en
Publication of WO2008144820A1 publication Critical patent/WO2008144820A1/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
    • A61K35/48Reproductive organs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/01Hydrocarbons
    • 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
    • A61K35/48Reproductive organs
    • A61K35/50Placenta; Placental stem cells; Amniotic fluid; Amnion; Amniotic stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • 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
    • C12N5/0603Embryonic cells ; Embryoid bodies
    • C12N5/0605Cells from extra-embryonic tissues, e.g. placenta, amnion, yolk sac, Wharton's jelly

Definitions

  • the present invention relates to a method of cellular therapy for a lung disease or condition in a subject involving the administration of multipotent epithelial stem cells derived from amnion tissue.
  • the method is used for the treatment of lung diseases and conditions such as chronic lung diseases including chronic obstructive pulmonary disease (COPD), acute lung conditions such as acute respiratory distress syndrome (ARDS), and ventilator associated lung injury (VALI).
  • COPD chronic obstructive pulmonary disease
  • ARDS acute respiratory distress syndrome
  • VALI ventilator associated lung injury
  • COPD chronic obstructive pulmonary disease
  • CF cystic fibrosis
  • IPF idiopathic pulmonary fibrosis
  • COPD chronic obstructive pulmonary disease
  • CF cystic fibrosis
  • IPF idiopathic pulmonary fibrosis
  • COPD chronic obstructive pulmonary disease
  • CF cystic fibrosis
  • IPF idiopathic pulmonary fibrosis
  • bronchodilators such as ⁇ 2-agonists and theophylline, and/or anti-inflammatory corticosteroids such as prednisone, are used to improve breathing capacity
  • drugs or treatments to arrest, or preferably, reverse disease progression are greatly awaited.
  • One possibility in this regard would be to achieve restitution of the lung with appropriate stem cells that "traffic" to sites of lung injury and differentiate into endogenous lung tissue.
  • the present applicant has studied the potential of multipotent human amnion epithelial stem cells (hAECs) (reviewed in Parolini, O., et al., 2008) for use in the treatment of chronic lung diseases and demonstrated, for the first time, that such cells, when cultured in Small Airway Growth Media (SAGM), adopt a lung cell phenotype as evidenced by the expression of lung specific markers and intra-cellular lamellar bodies. Further, the present applicant has found that when the hAECs were systemically administered into a mouse model of lung fibrosis (ie bleomycin-injured mice), they were located to the lung after 4 weeks and expressed the morphology and markers of alveolar epithelial cells.
  • SAGM Small Airway Growth Media
  • the multipotent hAECs also reduced inflammation by inhibiting the recruitment of macrophages through increased expression of macrophage inhibitory migratory factor (MIF) and down-regulation of macrophage inflammatory protein (MIP).
  • MIF macrophage inhibitory migratory factor
  • MIP macrophage inflammatory protein
  • the present invention provides a method of cellular therapy for a lung disease or condition in a subject, comprising administering to said subject a therapeutically effective amount of multipotent amnion epithelial stem cells (AECs).
  • AECs multipotent amnion epithelial stem cells
  • the AECs used in the method of the invention are human amnion epithelial stem cells (hAECs). Further, the AECs used in the method of the invention are preferably AECs derived from amnion of term, or near term, placenta.
  • the lung disease or condition that may be treated with the method of the invention may be a chronic lung disease such as chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), idiopathic pulmonary fibrosis (IPF), or an acute lung condition such as acute respiratory distress syndrome (ARDS), or damage due to chemo- and/or radio-therapy, industrial accidents or exposure to toxic compounds or particulates (eg hot smoke).
  • COPD chronic obstructive pulmonary disease
  • CF cystic fibrosis
  • IPF idiopathic pulmonary fibrosis
  • ARDS acute respiratory distress syndrome
  • the lung disease or condition may be a lung condition arising from the use of a mechanical ventilator (ie VALI).
  • the present invention provides the use of multipotent amnion epithelial stem cells (AECs) in the preparation of a composition for cellular therapy for a lung disease or condition in a subject, wherein said composition comprises said AECs in combination with a pharmaceutically-acceptable carrier.
  • AECs amnion epithelial stem cells
  • FIG. 1 a shows the assessment, by quantitative polymerase chain reaction (PCR), of the expression of pluripotent markers by hAECs.
  • PCR quantitative polymerase chain reaction
  • the results shown demonstrate expression of Octamer-4 (Oct-4), Nanog, SRY-related HMG-box gene 2 (Sox-2) and thyroid transcription factor- 1 (TTF-I, also known as Nkx 2.1), the latter being the earliest known marker of the respiratory lineage during development.
  • SPC lung-specific surfactant protein C
  • AQ-5 epithelial markers aquaporin-5
  • b shows the increase in the percentage of SPC and Aquaporin-5 (AQ-5) producing cells following differentiation in SAGM.
  • hAECs cultured in SAGM also demonstrated morphological features of type II alveolar epithelial cells such as lamellar bodies (arrows), d. shows that hAECs express low levels of major histocompatability complex (MHC) class IA and II proteins both in freshly isolated cells and following in vitro differentiation in SAGM;
  • MHC major histocompatability complex
  • Figure 2. shows the immunohistochemistry of lung sections harvested from mice following injection of hAECs into the tail vein 24h following intra-nasal bleomycin treatment. The sections demonstrate the presence of hAECs within the lung. The hAECs also acquired a morphology that closely resembled both type I (flattened) alveolar epithelial cells and type II (cuboidal) alveolar epithelial cells (lower panel), b. provides graphical results showing that the injection of hAECs into mice resulted in a reduction of the inflammation and fibrosis score of the lung following bleomycin-induced lung injury, c.
  • Figure 3 shows graphical results showing that hAEC treatment (24 hr after bleomycin administration) resulted in a significant decrease in collagen deposition as compared to bleomycin treated mice alone, b. shows graphical results showing that hAEC treatment (2 weeks after bleomycin administration) also resulted in a significant decrease in the amount of deposited collagen as compared to bleomycin treated mice alone, c. shows the assessment of MMP-2 and -9 by zymography, the results demonstrating that there was a further significant increase in MMP activity in mice treated with bleomycin and hAECs. d.
  • amnion and chorion form part of the embryonically-derived inner layers of the placenta with the maternally-derived decidua comprising the outer layer. While the chorion is a trophoblastic derivative, the amnion arises from the epiblast (ie at day 8 following fertilisation in humans) and, since the epiblast is also the origin of the three germ layers of the embryo, it has been suggested that cells of the amnion epithelium may be a ready source of pluripotent/multipotent stem cells (ie stem cells capable of differentiating into a range of cell types)(Ilancheran, S., et al., 2007).
  • pluripotent/multipotent stem cells ie stem cells capable of differentiating into a range of cell types
  • the applicant first found that at least a portion of the cells of the amnion epithelium express markers of pluripotency and proved to successfully differentiate in vitro into tissue representing all germ layers. Secondly, the applicant found that those cells could positively influence functional outcomes in chronic lung disease states; in particular, it was found that a reduction of lung inflammation and fibrosis could be achieved.
  • the present invention provides a method of cellular therapy for a lung disease or condition in a subject, comprising administering to said subject a therapeutically effective amount of multipotent amnion epithelial stem cells (AECs).
  • AECs multipotent amnion epithelial stem cells
  • the AECs are at least capable of differentiating into one or more lung cell types (eg lung epithelial cells such as type II alveolar epithelial cells).
  • lung cell types eg lung epithelial cells such as type II alveolar epithelial cells.
  • the AECs used in the method of the invention are human amnion epithelial stem cells (hAECs).
  • the AECs used in the method of the invention are preferably AECs derived from amnion of term, or near term, placenta.
  • Near term human placenta can be regarded as placenta collected (eg by emergency or elective caesarean section, or near term deliveries) after about 34 weeks from conception.
  • the lung disease or condition that may be treated with the method of the invention may be selected from those characterised by lung inflammation and/or lung fibrosis.
  • the method of the invention can be used to treat a chronic lung disease such as chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF) and idiopathic pulmonary fibrosis (IPF), or an acute condition such as acute respiratory distress syndrome (ARDS), or damage due to chemo- and/or radio-therapy, industrial accidents or exposure to toxic compounds or particulates (eg hot smoke).
  • COPD chronic obstructive pulmonary disease
  • CF cystic fibrosis
  • IPF idiopathic pulmonary fibrosis
  • ARDS acute respiratory distress syndrome
  • mechanical ventilation which is commonly used in the management of critically ill subjects with respiratory failure, has been reported to be an important co-factor of ARDS and acute lung injury (ALI).
  • ALI acute lung injury
  • the method of the invention can also be used to treat a lung condition arising from the use of a mechanical ventilator (ie ventilator associated lung injury (VALI)).
  • VALI mechanical ventilator associated lung injury
  • the method of the invention can be used to treat pre-term infants lacking type II alveolar epithelial cells, as well as to treat and/or prevent foetal respiratory distress syndrome (RDS).
  • RDS foetal respiratory distress syndrome
  • the foetal lung is not fully developed until late in gestation, infants who are delivered prematurely are typically unable to breathe independently (thereby necessitating the use of a mechanical ventilator).
  • the outcome for an infant of premature birth and/or respiratory support with a mechanical ventilator can consequently be the development of inflammation in the lung, diffuse injury to the alveoli and profound changes in lung mechanics; characteristic symptoms of RDS.
  • the method of the invention can be used to treat a lung inflammation associated with smoking or industrial exposure to toxic compounds or particulates (eg hot smoke).
  • toxic compounds or particulates eg hot smoke
  • the method involves administering to the subject a therapeutically effective amount of multipotent AECs.
  • the mode of administration of the multipotent AECs will typically be by way of systemic transfusion, bronchoscopic or intra-nasal instillation, although it may also be satisfactory to implant the AECs (with or without a supporting substrate such as an implantable gel or solid scaffold material) directly into the lung at or near to a site of lung injury (eg sites of inflammation and/or fibrosis).
  • the multipotent AECs will typically be administered in combination with a pharmaceutically-acceptable carrier (eg physiological saline).
  • the tranfused multipotent AECs can migrate to the lung, particularly to a site(s) of lung injury, and present in the lung even after 4 weeks following administration. Once at the site(s) of lung injury, the multipotent AECs differentiate into lung epithelial cells to initiate repair and regeneration of lung tissue.
  • terapéuticaally effective amount is to be understood as referring to an amount of the multipotent AECs (ie a cell number) that will, at least, arrest the lung disease or condition being treated. Such an amount may vary considerably depending upon a range of factors such as the mode of administration, the age and/or body weight of the subject, and the severity of the lung disease or condition to be treated. However, typically, the amount will be in the range of about 1 x 10 5 to 1 x 10 10 , more preferably, 1 x 10 8 to 1 x 10 10 multipotent AECs.
  • the multipotent AECs may be administered to the subject in combination with one or more active agents.
  • the AECs may be administered with one or more of the drugs presently used to alleviate and/or prevent symptoms of a lung disease or condition (eg a bronchodilator or antiinflammatory corticosteroid).
  • the AECs may be administered with an agent which causes a reduction in the expression or activity of one or more of the pro-inflammatory cytokines IL-I, JL-2, IL- 6, IFN- ⁇ and TNF- ⁇ (eg a specific inhibitor or antagonist agent or specific antibody), and/or the expression or activity of the pro-fibrotic cytokine TGF- ⁇ (eg a specific TGF- ⁇ inhibitor or antagonist agent or specific anti-TGF- ⁇ antibody).
  • an agent which causes a reduction in the expression or activity of one or more of the pro-inflammatory cytokines IL-I, JL-2, IL- 6, IFN- ⁇ and TNF- ⁇ eg a specific inhibitor or antagonist agent or specific antibody
  • the pro-fibrotic cytokine TGF- ⁇ eg a specific TGF- ⁇ inhibitor or antagonist agent or specific anti-TGF- ⁇ antibody
  • the AECs may also be administered with synthetic or animal-derived lung surfactant (eg colfosceril palmitate-based products (such as Exosurf® marketed by GlaxoSmithKline pic, Brentford, Middlesex, United Kingdom) and surfactant extracted from bovine lung lavage fluid (such as Alveofact® marketed by Boehringer Ingelheim GmbH, Ingelheim, Germany)), to open unperfused areas of the lung to thereby aid access of the AECs to sites of injury.
  • synthetic or animal-derived lung surfactant eg colfosceril palmitate-based products (such as Exosurf® marketed by GlaxoSmithKline pic, Brentford, Middlesex, United Kingdom) and surfactant extracted from bovine lung lavage fluid (such as Alveofact® marketed by Boehringer Ingelheim GmbH, Ingelheim, Germany)
  • the multipotent AECs are preferably characterised by the expression of one or more markers listed in Table 1 below. More preferably, the multipotent AECs are characterised by the expression of one or more pluripotent stem cell markers. For hAECs, the pluripotent markers will typically be selected from Oct-4, Nanog and Sox-2.
  • the multipotent hAECs are preferably characterised in that they show low expression of MHC class IA and II proteins. This is preferred since it offers the potential use of the hAECs in subjects of divergent histocompatibility.
  • the multipotent hAECs used in the method of the invention may, however, be autologous (and be sourced from preserved samples such as cryopreserved samples).
  • Multipotent AECs may be derived from amnion tissue by standard methods.
  • amnion tissue stripped from term placenta can be treated with a suitable proteolytic enzyme (eg trypsin), and the purity of dispersed cells assessed by subjecting an aliquot to flow analysis (eg by FACS) for cytokeratin-7, an epithelial cytoskeletal protein found in AECs, and/or one or more of Oct-4, Nanog and Sox-2.
  • cytokeratin-7 an epithelial cytoskeletal protein found in AECs, and/or one or more of Oct-4, Nanog and Sox-2.
  • the dispersed cells may be sorted/assessed by flow analysis for one or more relevant cell surface marker (eg SSEA-4).
  • SSEA-4 relevant cell surface marker
  • the dispersed/sorted cells may also be expanded by culturing under conditions which avoid cell differentiation (eg by culturing in a basal medium such as Dulbecco's Modified Eagle's Medium (DMEM)/F12 supplemented by animal-free serum or serum substitutes).
  • a basal medium such as Dulbecco's Modified Eagle's Medium (DMEM)/F12 supplemented by animal-free serum or serum substitutes.
  • DMEM Dulbecco's Modified Eagle's Medium
  • F12 supplemented by animal-free serum or serum substitutes
  • the dispersed/sorted, and optionally expanded, cells can be prepared for immediate administration to the subject by, for example, isolating the AECs from the culture/storage media and resuspending the AECs in a pharmaceutically-acceptable carrier.
  • the multipotent AECs may be partially substituted with derivative cells; that is, cells which have been at least partially differentiated from the AECs (ie the invention may utilise "pre- differentiated" cells).
  • derivative cells may be produced, for example, by culturing AECs under suitable conditions in a commercially available SAGM (eg SAGM products marketed by Lonza Group Ltd, Basel, Switzerland).
  • SAGM eg SAGM products marketed by Lonza Group Ltd, Basel, Switzerland.
  • the invention utilises derivative cells, the cells may be administered in an AEC:derivative cell ratio in the range of about 5: 1 to about 1 :5, more preferably about 4: 1 to about 1 :1.
  • the multipotent AECs may be partially substituted with one or more other (ie non- derivative) cell types, including one or more other stem cell types.
  • the cells may be administered in an AEC:other cell ratio in the range of about 5:1 to about 1 :5, more preferably about 4: 1 to about 1 : 1.
  • the method of the invention most preferably, utilises AECs only.
  • the present invention provides the use of multipotent amnion epithelial stem cells (AECs) in the preparation of a composition for cellular therapy for a lung disease or condition in a subject, wherein said composition comprises said AECs in combination with a pharmaceutically-acceptable carrier.
  • AECs amnion epithelial stem cells
  • the multipotent AECs used in the use of the invention are human amnion epithelial stem cells (hAECs).
  • the AECs used in the method of the invention are preferably AECs derived from amnion of term, or near term, placenta.
  • composition may also comprise cells belonging to one or more other cell types including one or more other stem cell types and/or one or more wholly or partially differentiated cell types (eg derivative cells of AECs; that is, cells which have been at least partially differentiated from AECs).
  • cells belonging to one or more other cell types including one or more other stem cell types and/or one or more wholly or partially differentiated cell types (eg derivative cells of AECs; that is, cells which have been at least partially differentiated from AECs).
  • composition may also further comprise one or more active agents (eg a bronchodilator or antiinflammatory corticosteroid).
  • active agents eg a bronchodilator or antiinflammatory corticosteroid.
  • the cellular therapy of the present invention offers the possibility of reducing, or at least arresting, lung diseases or conditions that have to date only been treated by the use of drugs to alleviate and/or prevent symptoms. Since the long term use of these drugs can also lead to serious side- effects (eg long term use of corticosteroids is associated with the development of several debilitating diseases and conditions including diabetes, osteoporosis and cataract formation), the identification and development of a cellular therapy for lung diseases or conditions, such as that of the present invention, is highly desirable.
  • the cellular therapy of the present invention is able to bring about a reduction in lung inflammation, prevent fibrosis and reduce collagen concentration in the lung following lung injury.
  • the present invention provides a method of reducing inflammation in the lung of a subject, said method comprising administering to said subject a therapeutically effective amount of multipotent amnion epithelial stem cells (AECs).
  • AECs multipotent amnion epithelial stem cells
  • the present invention provides a method of preventing fibrosis in the lung of a subject, said method comprising administering to said subject a therapeutically effective amount of multipotent amnion epithelial stem cells (AECs).
  • AECs multipotent amnion epithelial stem cells
  • the present invention provides a a method of reducing collagen concentration in the lung of a subject, said method comprising administering to said subject a therapeutically effective amount of multipotent amnion epithelial stem cells (AECs).
  • AECs multipotent amnion epithelial stem cells
  • AECs administered by systemic transfusion, bronchoscopic or intra-nasal instillation to migrate to the lung provides a means or "vector" for delivering a therapeutic agent (eg a therapeutic protein or peptide, gene therapy agent or gene silencing agent) to the lung.
  • a therapeutic agent eg a therapeutic protein or peptide, gene therapy agent or gene silencing agent
  • cystic fibrosis an exogenous polynucleotide molecule encoding cystic fibrosis transmembrane conductance regulator (CFTR) could be introduced into autologous AECs (ie lacking a functional CFTR gene) and thereafter administered to a subject suffering from CF, to provide a gene therapy for CF.
  • CFTR cystic fibrosis transmembrane conductance regulator
  • the present invention extends to a method of cellular therapy for a lung disease or condition in a subject, comprising administering to said subject a therapeutically effective amount of multipotent amnion epithelial stem cells (AECs) comprising a therapeutic agent as described above, as well as to a composition therefor (ie a composition comprising AECs comprising a therapeutic agent as described above, in combination with a pharmaceutically-acceptable carrier) and, further, to the use of AECs comprising a therapeutic agent as described above in the preparation of a composition for cellular therapy for a lung disease or condition in a subject, wherein said composition comprises said AECs in combination with a pharmaceutically-acceptable carrier.
  • AECs multipotent amnion epithelial stem cells
  • Example 1 Human amnion epithelial cell abrogation of fibrosis and augmentation of lung repair
  • hAECs were isolated and characterised using methods described previously (Ilancheran, S., et al., 2007). Briefly, tissue was digested twice in 0.25% trypsin:EDTA solution (Invitrogen Corporation, Carlsbad, CA, United States of America) for 20 minutes. Trypsin was inactivated with foetal calf serum (FCS, Invitrogen) and dispersed cells washed in M199 medium (Invitrogen). Cells were then analysed by flow cytometry for cytokeratin-7 (Dako Denmark A/S, Glostrup, Denmark), an epithelial cytoskeletal protein found in hAECs.
  • FCS foetal calf serum
  • M199 medium Invitrogen
  • cytokeratin-7 positive cells were plated onto type IV collagen (Roche Diagnostics, Basel, Switzerland) coated vessels and cultured in Small Airway Epithelial Basal Medium (SABM) (Clonetics, Walkersville, MD, United States of America) and supplements (ie retinoic acid, epidermal growth factor 1 (EGF-I), epinephrine, transferrin, insulin, tri-iodothyronine, bovine pituitary extract, bovine serum albumin (BSA), fatty acid-free serum and hydrocortisone) provided. Control cultures were grown in SABM (SABM) (Clonetics, Walkersville, MD, United States of America) and supplements (ie retinoic acid, epidermal growth factor 1 (EGF-I), epinephrine, transferrin, insulin, tri-iodothyronine, bovine pituitary extract, bovine serum albumin (BSA), fatty acid-free serum and hydrocortisone) provided. Control cultures were
  • RNA was isolated from hAECs (n 6) using RNeasy columns (Qiagen, Hilden, Germany). One ⁇ g of total RNA was converted to cDNA using the High-Capacity cDNA Archive Kit (Applied Biosystems, Foster City, CA, United States of America).
  • Oct-4 is a transcription factor that is highly expressed in the cells of the inner cell mass (ICM) and pluripotent embryonic carcinoma cells lines; low expression of Oct-4 yields the growth of trophoblastic cells only, while elevated expression of this transcription factor leads to differentiation of embryonic stem (ES) cells to embryonic and extra-embryonic tissue.
  • Nanog is a homeodomain factor and is expressed in vivo in the morula and ICM of the epiblast. As such, it is a marker of pluripotent cells.
  • Sox-2 is a transcription factor that is co-expressed with Oct-4 in the ICM as well as in ES cells and is essential for the function of Oct-4.
  • Nkx-1 is also known as thyroid transcription factor- 1 (TTF-I), and represents the earliest known marker of the respiratory lineage.
  • TTF-I thyroid transcription factor- 1
  • SPC is highly specific for type II alveolar epithelial cells and Aquaporin-5 (AQ-5) is a lung (alveolar) epithelial cell markers.
  • PCR parameters were: 95°C, 7sec and 6O 0 C, 20sec for 40 cycles. Data was normalised against ⁇ -actin and expression levels, relative to the human embryonic stem cell line ES-I (for Oct-4,
  • Nanog, Sox-2, and Nkx2.1), or adult human lung was calculated using the ⁇ Ct method.
  • TEM Transmission electron microscopy
  • Flow cytometry hAECs (2.5xlO 5 ) were incubated with antibodies against caveolin (1 :100), AQ-5 (1 :50), SPC (1 :50), human leukocyte antigen (HLA)-DP 5 -DQ 5 -DR (lO ⁇ g/ml) or allophytocyanin (APC)- conjugated HLA-A 5 -B 5 -C (10 ⁇ l) 5 for 1 hour at RT. After several washes, cells were incubated with 1:10 diluted APC-conjugated donkey anti-rabbit (caveolin), donkey anti-goat (AQ-5, SP-C), or goat anti-mouse (HLA-DP,-DQ,-DR) secondary antibodies for 30 minutes at RT.
  • caveolin caveolin
  • SPC human leukocyte antigen
  • APC allophytocyanin
  • hAECs were washed to remove excess secondary antibodies and analysed by flow cytometry.
  • Primary antibodies for caveolin, AQ-5 and SPC were purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA 5 United States of America) and HLA antibodies and secondary antisera from Becton, Dickinson and Company (Franklin Lakes, NJ, United States of America).
  • Further flow cytometry analysis of the hAECs (including a popoulation (P5) that had been passaged 5 times) was undertaken, using similar methodology and monoclonal antibodies to the cell surface markers (Becton, Dickinson and Company) indicated in Table 2 below, in comparison with human bone marrow mesenchymal stem cells (hMSC). The percentage of the cells within the cell types that were "positive" for each marker was recorded.
  • Paraffin embedded tissue sections 5 ⁇ m in thickness, were dewaxed and rehydrated. Antigen retrieval was carried out by by heating in citrate buffer. After quenching endogenous peroxidase activity and blocking non-specific binding, sections were incubated with antibodies against human inner membrane mitochondria (MM; 1:10, AbD Serotec, Kidlington, Oxfordshire, United Kingdom) and human SPC (1:50, Santa Cruz Biotechnology, Inc.) for 1 hour at 37 0 C. Isotype- matched mouse IgG2a and goat IgG were applied to sections serving as negative controls.
  • human inner membrane mitochondria MM
  • human SPC 1:50, Santa Cruz Biotechnology, Inc.
  • Antibody binding was detected using horseradish peroxidase-labelled biotin-streptavidin secondary antibody (LSAB kit, Dako), and immunostaining visualised using 3,3'-diaminobenzidine chromogen. Co-localisation of IMM and SPC was assessed by examining serial tissue sections. For inflammation and fibrosis, sections were scored by the Ashcroft method (Ashcroft, T., et al., 1988).
  • RNA was extracted from lungs of SCID mice after two and four weeks of receiving saline, bleomycin and bleomycin+hAECs (n 5/group). Following DNAse treatment, 1 ⁇ g of RNA was converted to cDNA with Superscript III (Invitrogen). The cDNA (diluted 1 :20) was amplified with PCR primers against IL-I, IL-2, interleukin-4 (IL-4), IL-6, interleukin-10 (IL-10), interferon- gamma (INF ⁇ ), TNF- ⁇ , TGF- ⁇ , MIF and MIP.
  • Cycling parameters used for quantitative PCR were: denaturation 95°C, 7sec, annealing 6O 0 C, 7-15sec and extension 72°C, 15sec for 40 cycles. After normalising data to ⁇ -actin, expression relative to saline treated control mice was calculated by the ⁇ Ct method.
  • Collagen content The collagen content in lungs of mice receiving saline, bleomycin and bleomycin+hAEC
  • Reagents, primers and TaqMan probes were purchased from Applied Biosystems (#Mm00441818_ml, #Mm00441825_ml, #Mm00441826_ml, #Mm00446568_ml). After normalising data to ⁇ -actin, expression relative to saline instilled mice was calculated by the ⁇ Ct method.
  • hAECs were derived from the placenta by stripping off the membrane and growing the hAECs in cell culture. These cells demonstrated clonality and, consistent with their epiblast derivation (Chambers L, et al., 2003; and Mitsui K., et al., 2003), expressed several markers of pluripotency, namely Oct-4, Nanog, Sox-1 and c-kit ( Figure Ia and Table 1). The cells were not of vascular endothelial or haematopoetic lineage since they were "negative” for CD31, CD34 and CD45 markers (see Table 1).
  • Nkx 2.1 is a critical transcription factor for branching morphogenesis and the formation of type II alveolar epithelial cells during lung development.
  • SAGM is primarily utilised in the maintenance of airway epithelial cells in culture and contains hydrocortisone, human epidermal growth factor (hEGF-1) and retinoic acid which are also factors present during lung development.
  • HAECs cultured in SAGM for 2 and 4 weeks demonstrated an increase in the gene expression of the lung specific gene surfactant protein C (SPC) (see Figure Ia).
  • SPC lung specific gene surfactant protein C
  • Figure Ib The increase in mRNA expression was translated into elevated protein expression of SPC shown by FACs analysis ( Figure Ib).
  • Figure Ib Ultra-structural analyses revealed that hAECs grown in SAGM for 4 weeks contained a distinct cell population demonstrating cytoplasmic organelles containing lamellar bodies, lipid filled vacuoles and surface microvilli formation that were indicative of type II alveolar epithelial cell differentiation. These features were not present in hAECs cultured in Dubelcos Modified Eagle Medium (DMEM)/F12.
  • DMEM Dubelcos Modified Eagle Medium
  • Type II alveolar epithelial cells constitute one third of the surface area of the lung but make up 66% of the cells in the distal lung and are responsible for the secretion of surface tension reducing surfactant protein, hi addition, type II alveolar epithelial cells differentiate into the gas-exchanging type I alveolar epithelial cells during lung repair. Notably, hAECs demonstrate low expression of MHC antigens when freshly isolated and following in vitro differentiation, which widens the potential for the use of these cells for clinical application (see Figure Ic and Id).
  • the bleomycin-induced model of lung inflammation and fibrosis is well characterised and reflects the phases of lung injury in human subjects.
  • the present applicant had previously established the model in SCDD mice, and this allowed the evaluation of the functional role of human cells in augmenting lung repair.
  • intra-nasally administered bleomycin induced an increase in both the inflammatory and fibrosis score on histology at both the 2 and 4 week time points (see
  • hAECs hi order to treat lung injury, hAECs were injected into the tail vein 24 hours following intra-nasal administration of bleomycin and were demonstrated in the lung by anti-human inner mitochondrial membrane (IMM) protein positive staining of cells at both 2 and 4 week time points.
  • IMM inner mitochondrial membrane
  • the anti- mitochondrial staining was negative in mouse lung tissue but present only in human controls (data not shown) thereby confirming the specificity and sensitivity of the antibody for the identification of human cells in the mouse lung.
  • cells could not be detected in lung tissue 2 weeks post injection indicating that hAECs do not "traffic" to uninjured lung.
  • the cytokines IL-2, TNF- ⁇ and IFN- ⁇ were significantly elevated in response to bleomycin in mouse strains prone to develop injury (C57/B16) as compared to the resistant strains of mice such as Balb/C, thus supporting the role for these cytokines in the progression of lung fibrosis.
  • inhibition of these cytokines by hAEC administration would serve to further reduce the development of fibrosis.
  • several previous studies have demonstrated that the reduction in endogenous lung IFN ⁇ and IFN ⁇ knock-out mice are resistant to bleomycin injury.
  • TNFR-deficient mice are protective against bleomycin fibrosis by inhibiting the activation of TGF- ⁇ .
  • the significant reduction in TNF- ⁇ by HAEC administration provides even further support to the anti-fibrotic role of hAECs.
  • CDIa CD2, CD3, CD4, CD7, CD8, CD9, CDl Ia, CDl Ib, CDl Ic, CD 14, CD 18, CD 19, CD20.
  • IL-6 this cytokine is secreted by fibroblasts, epithelial cells and macrophages, and has been found to be significantly elevated during bleomycin lung injury. It has been postulated that IL-6 acts in synergy with TNF- ⁇ to increase the levels of MIP, thereby enhancing the recruitment of macrophages and perpetuating inflammation and fibrosis.
  • TGF- ⁇ is a central cytokine in the pathogenesis of lung fibrosis.
  • TGF- ⁇ is increased in macrophages, epithelial cells, fibroblasts and myofibroblasts.
  • TGF- ⁇ has a bimodal effect on fibroblast proliferation, stimulates monocyte production of cytokines and acts as a potent stimulator of collagen deposition (Kang, H.R., et al., 2007).
  • hAECs have an anti-fibrotic role.
  • hAECs reduce the cellular infiltration of the lung and reduce the pro-inflammatory cytokines IL-I, TNF- ⁇ and IL-6.
  • hAECs Further to the anti-inflammatory effects of hAECs, the role of hAECs in collagen deposition and fibrosis following bleomycin lung injury was investigated. Bleomycin induces excessive collagen deposition and fibrosis at 14 days post-injury resulting in the development of fibroblastic foci. These foci are characterised by the distortion of lung architecture, fibroblast and myofibroblast proliferation, collagen deposition and a reduction in functional lung. All of these are "generic" characteristics of fibrosis in all organs and show remarkable homology to pathology in human subjects.
  • Collagen deposition was measured by using a hydroxyproline assay. This showed an expected increase in collagen deposition in mice treated with bleomycin alone but a significant fall in the cohort subjected to bleomycin and hAEC injection (see Figures 3a and 3b). Notably, there was no influence of hAECs injected into healthy mice on collagen deposition. Further, it was found that the reduction of collagen was specific to hAECs since the injection of primary human lung fibroblasts into bleomycin exposed did not influence collagen levels. The results achieved with mice injected two weeks after treatment with bleomycin (see Figure 3b) indicates that hAECs abrogates fibrosis.
  • MMPs matrix metalloproteinases
  • ECM extracellular matrix
  • TIMPs endogenous inhibitors
  • MMP-2 is responsible for the degradation of most ECM proteins and is also implicated in alveolar regeneration. Further, studies in human subjects have previously demonstrated that the up- regulation of MMP-2 was a strong predictor of a decrease in fibrosis in subjects with hypersensitivity pneumonitis (Selman, M. and A. Pardo, 1991). As such, it is considered that the significant up-regulation of MMP-2 is responsible for the reduction in collagen concentration within the lung.
  • MMP-9 is instrumental in cleaving several ECM proteins but not directly implicated in active fibrosis since down regulation of MMP-9 did not influence collagen levels in bleomycin treated mice.
  • the up-regulation of MMP-9 may be an additive degradative molecule during collagen deposition.
  • Tissue inhibitors of MMPs are the major endogenous inhibitors of MMPs and bind these molecules in a 1:1 stoichiometry.
  • TIMP- 1-4 there are 4 homologues of TIMPs, TIMP- 1-4, each having specific functions.
  • TIMP- 1 Inhibition of TIMP- 1 augmented the inflammatory response to bleomycin but not an increase in fibrosis. Similarly, TIMP-2 and TIMP-3 are also elevated in response to bleomycin. From lung transcripts, it was demonstrated that there was a down-regulation of TIMP-I, TIMP-3 and TIMP-4 in mice treated with hAECs at day 28 post bleomycin lung injury (Figure 3d). Moreover, there is a causal relationship between TDVIP elevation and fibrosis since TIMP concentrations increased prior to the phase of collagen deposition in the bleomycin mouse model at days 14 to 28 post-injury. Also, mouse strains resistant to bleomycin fibrosis did not demonstrate an increase in TIMP-I levels following injury. Thus, the reduction in TIMP levels accompanied by the increase in MMPs indicates the existence of a micro-environment that favours ECM degradation in response to HAEC injection, thereby reducing fibrosis.
  • hAEC appears to directly augment lung repair.
  • TGF-beta 1 Transforming growth factor (TGF)-beta 1 stimulates pulmonary fibrosis and inflammation via a Bax-dependent, bid activated pathway that involves matrix metalloproteinase-12. J Biol Chem 282:7723-7732 (2007).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Developmental Biology & Embryology (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Zoology (AREA)
  • Cell Biology (AREA)
  • Reproductive Health (AREA)
  • Biotechnology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Pregnancy & Childbirth (AREA)
  • Virology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pulmonology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

A method of cellular therapy for a lung disease or condition in a subject is disclosed, wherein the method involves the administration of multipotent epithelial stem cells derived from amnion tissue (AECs). In a particular application, the method is used for the treatment of lung diseases and conditions such as chronic lung diseases including chronic obstructive pulmonary disease (COPD), acute lung conditions such as acute respiratory distress syndrome (ARDS), and ventilator associated lung injury (VALI).

Description

TREATMENT OF CHRONIC LUNG DISEASE
FIELD OF THE INVENTION
The present invention relates to a method of cellular therapy for a lung disease or condition in a subject involving the administration of multipotent epithelial stem cells derived from amnion tissue. In a particular application, the method is used for the treatment of lung diseases and conditions such as chronic lung diseases including chronic obstructive pulmonary disease (COPD), acute lung conditions such as acute respiratory distress syndrome (ARDS), and ventilator associated lung injury (VALI).
INCORPORATION BY REFERENCE This patent application claims priority from:
- AU 2007902844 titled "Treatment of chronic lung disease" filed on 28 May 2007. The entire content of this application is hereby incorporated by reference.
BACKGROUND OF THE INVENTION
Chronic lung diseases such as chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF) and idiopathic pulmonary fibrosis (IPF) are a major global problem. COPD, in particular, is predicted to feature among the five most common causes of morbidity and mortality in the human population by 2020. As a group of diseases they are characterised by one or more of the following activities; infiltration of leukocytes into the lungs, interstitial lung inflammation, increased proinflammatory cytokine production, thickening of lung alveolar septae, hemorrhagic pneumonitis, distortion of normal lung architecture and fibrosis brought about by increased collagen deposition coupled with altered protease activity. Moreover, disease progression results in significant loss of lung tissue and scarring ultimately leading to the substantial morbidity and mortality associated with these chronic lung diseases.
Presently, treatment of chronic lung diseases is limited to symptom alleviation and/or prevention (eg bronchodilators such as β2-agonists and theophylline, and/or anti-inflammatory corticosteroids such as prednisone, are used to improve breathing capacity), and accordingly, the development of drugs or treatments to arrest, or preferably, reverse disease progression are greatly awaited. One possibility in this regard, would be to achieve restitution of the lung with appropriate stem cells that "traffic" to sites of lung injury and differentiate into endogenous lung tissue. To this end, the present applicant has studied the potential of multipotent human amnion epithelial stem cells (hAECs) (reviewed in Parolini, O., et al., 2008) for use in the treatment of chronic lung diseases and demonstrated, for the first time, that such cells, when cultured in Small Airway Growth Media (SAGM), adopt a lung cell phenotype as evidenced by the expression of lung specific markers and intra-cellular lamellar bodies. Further, the present applicant has found that when the hAECs were systemically administered into a mouse model of lung fibrosis (ie bleomycin-injured mice), they were located to the lung after 4 weeks and expressed the morphology and markers of alveolar epithelial cells. Moreover, it was observed that the multipotent hAECs also reduced inflammation by inhibiting the recruitment of macrophages through increased expression of macrophage inhibitory migratory factor (MIF) and down-regulation of macrophage inflammatory protein (MIP). This resulted in reduced expression of the inflammatory cytokines interleukin-1 (IL-I), interleukin- 2 (IL-2), interleukin-6 (IL-6), tumour necrosis factor-alpha (TNF-α), inteferon-gamma (IFN-γ) and the pro-fϊbrotic transforming growth factor-beta (TGF-β). In, addition, collagen concentrations in the lungs of the treated mice were significantly reduced by increased degradation by matrix metallo-proteinases (MMP-2 and MMP-9) and down-regulation of their endogenous inhibitors; that is, the tissue inhibitors of MMPs (TIMPs). Taken together, treatment with hAECs appears to address several pathways in the pathogenesis of lung injury and thereby offers considerable potential as the basis of an effective cellular therapy of lung diseases and conditions such as chronic lung diseases.
SUMMARY OF THE INVENTION
In a first aspect, the present invention provides a method of cellular therapy for a lung disease or condition in a subject, comprising administering to said subject a therapeutically effective amount of multipotent amnion epithelial stem cells (AECs).
Preferably, the AECs used in the method of the invention are human amnion epithelial stem cells (hAECs). Further, the AECs used in the method of the invention are preferably AECs derived from amnion of term, or near term, placenta.
The lung disease or condition that may be treated with the method of the invention may be a chronic lung disease such as chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), idiopathic pulmonary fibrosis (IPF), or an acute lung condition such as acute respiratory distress syndrome (ARDS), or damage due to chemo- and/or radio-therapy, industrial accidents or exposure to toxic compounds or particulates (eg hot smoke). Alternatively, the lung disease or condition may be a lung condition arising from the use of a mechanical ventilator (ie VALI).
In a second aspect, the present invention provides the use of multipotent amnion epithelial stem cells (AECs) in the preparation of a composition for cellular therapy for a lung disease or condition in a subject, wherein said composition comprises said AECs in combination with a pharmaceutically-acceptable carrier. BRIEF DESCRIPTION OF THE FIGURES
Figure 1 a. shows the assessment, by quantitative polymerase chain reaction (PCR), of the expression of pluripotent markers by hAECs. The results shown demonstrate expression of Octamer-4 (Oct-4), Nanog, SRY-related HMG-box gene 2 (Sox-2) and thyroid transcription factor- 1 (TTF-I, also known as Nkx 2.1), the latter being the earliest known marker of the respiratory lineage during development. Following culture in SAGM, the expression of these markers decreased while there was an increase in the expression of lung-specific surfactant protein C (SPC) as well as the epithelial markers aquaporin-5 (AQ-5). b. shows the increase in the percentage of SPC and Aquaporin-5 (AQ-5) producing cells following differentiation in SAGM. c. shows that hAECs cultured in SAGM also demonstrated morphological features of type II alveolar epithelial cells such as lamellar bodies (arrows), d. shows that hAECs express low levels of major histocompatability complex (MHC) class IA and II proteins both in freshly isolated cells and following in vitro differentiation in SAGM;
Figure 2. a. shows the immunohistochemistry of lung sections harvested from mice following injection of hAECs into the tail vein 24h following intra-nasal bleomycin treatment. The sections demonstrate the presence of hAECs within the lung. The hAECs also acquired a morphology that closely resembled both type I (flattened) alveolar epithelial cells and type II (cuboidal) alveolar epithelial cells (lower panel), b. provides graphical results showing that the injection of hAECs into mice resulted in a reduction of the inflammation and fibrosis score of the lung following bleomycin-induced lung injury, c. provides graphical results showing that with the injection of hAECs into mice there was, following bleomycin-induced lung injury, a significant reduction in the expression of pro-inflammatory cytokines (eg IL-I, IL-6 and TNF-α) from lung transcripts (*p<0.05 comparing bleomycin+HAEC to bleomycin alone, by one way ANOVA); and
Figure 3. a. shows graphical results showing that hAEC treatment (24 hr after bleomycin administration) resulted in a significant decrease in collagen deposition as compared to bleomycin treated mice alone, b. shows graphical results showing that hAEC treatment (2 weeks after bleomycin administration) also resulted in a significant decrease in the amount of deposited collagen as compared to bleomycin treated mice alone, c. shows the assessment of MMP-2 and -9 by zymography, the results demonstrating that there was a further significant increase in MMP activity in mice treated with bleomycin and hAECs. d. provides graphical results showing that transcripts of lung demonstrated reduced expression of tissue inhibitors of MMPs (TIMPs), the endogenous inhibitors of MMPs, following hAEC treatment (*p<0.05 compared to healthy controls, + p<0.05 comparing bleomycin+hAEC to bleomycin alone, by one way ANOVA). DETAILED DESCRIPTION OF THE INVENTION
The amnion and chorion form part of the embryonically-derived inner layers of the placenta with the maternally-derived decidua comprising the outer layer. While the chorion is a trophoblastic derivative, the amnion arises from the epiblast (ie at day 8 following fertilisation in humans) and, since the epiblast is also the origin of the three germ layers of the embryo, it has been suggested that cells of the amnion epithelium may be a ready source of pluripotent/multipotent stem cells (ie stem cells capable of differentiating into a range of cell types)(Ilancheran, S., et al., 2007).
In work leading to the present invention, the applicant first found that at least a portion of the cells of the amnion epithelium express markers of pluripotency and proved to successfully differentiate in vitro into tissue representing all germ layers. Secondly, the applicant found that those cells could positively influence functional outcomes in chronic lung disease states; in particular, it was found that a reduction of lung inflammation and fibrosis could be achieved.
Thus, in a first aspect, the present invention provides a method of cellular therapy for a lung disease or condition in a subject, comprising administering to said subject a therapeutically effective amount of multipotent amnion epithelial stem cells (AECs).
For the purposes of the present invention, the AECs are at least capable of differentiating into one or more lung cell types (eg lung epithelial cells such as type II alveolar epithelial cells).
Preferably, the AECs used in the method of the invention are human amnion epithelial stem cells (hAECs).
Further, the AECs used in the method of the invention are preferably AECs derived from amnion of term, or near term, placenta. Near term human placenta can be regarded as placenta collected (eg by emergency or elective caesarean section, or near term deliveries) after about 34 weeks from conception.
The lung disease or condition that may be treated with the method of the invention may be selected from those characterised by lung inflammation and/or lung fibrosis.
In particular, the method of the invention can be used to treat a chronic lung disease such as chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF) and idiopathic pulmonary fibrosis (IPF), or an acute condition such as acute respiratory distress syndrome (ARDS), or damage due to chemo- and/or radio-therapy, industrial accidents or exposure to toxic compounds or particulates (eg hot smoke). Further, mechanical ventilation, which is commonly used in the management of critically ill subjects with respiratory failure, has been reported to be an important co-factor of ARDS and acute lung injury (ALI). In particular, it has been found that mechanical ventilation exacerbates lung inflammation in response to bacterial infection (Dhanireddy, S., et al., 2006). Accordingly, the method of the invention can also be used to treat a lung condition arising from the use of a mechanical ventilator (ie ventilator associated lung injury (VALI)).
Moreover, the method of the invention can be used to treat pre-term infants lacking type II alveolar epithelial cells, as well as to treat and/or prevent foetal respiratory distress syndrome (RDS). Since the foetal lung is not fully developed until late in gestation, infants who are delivered prematurely are typically unable to breathe independently (thereby necessitating the use of a mechanical ventilator). The outcome for an infant of premature birth and/or respiratory support with a mechanical ventilator can consequently be the development of inflammation in the lung, diffuse injury to the alveoli and profound changes in lung mechanics; characteristic symptoms of RDS. Presently, foetal RDS is treated and/or prevented using "surfactant therapy" wherein synthetic or animal-derived surfactant is administered upon delivery (Morley, C.J., et al., 1978). However, while such therapy has been a highly important advance in the care of preterm infants, it is considered that the method of the invention, especially when used in combination with a synthetic or animal-derived surfactant (as discussed further below), offers the potential for a greater therapeutic benefit than treatment by surfactant therapy alone. While not wishing to be bound by theory, the present applicant believes that this greater therapeutic benefit will be achieved through the known benefit of administering a synthetic or animal-derived surfactant in combination with the observed ability of AECs to bring about a reduction in lung inflammation, prevent fibrosis and differentiate into type II alveolar epithelial cells which secrete surfactant.
Also, the method of the invention can be used to treat a lung inflammation associated with smoking or industrial exposure to toxic compounds or particulates (eg hot smoke).
The method involves administering to the subject a therapeutically effective amount of multipotent AECs.
The mode of administration of the multipotent AECs will typically be by way of systemic transfusion, bronchoscopic or intra-nasal instillation, although it may also be satisfactory to implant the AECs (with or without a supporting substrate such as an implantable gel or solid scaffold material) directly into the lung at or near to a site of lung injury (eg sites of inflammation and/or fibrosis). The multipotent AECs will typically be administered in combination with a pharmaceutically-acceptable carrier (eg physiological saline).
Using systemic transfusion, it has been found that at least a portion of the tranfused multipotent AECs can migrate to the lung, particularly to a site(s) of lung injury, and present in the lung even after 4 weeks following administration. Once at the site(s) of lung injury, the multipotent AECs differentiate into lung epithelial cells to initiate repair and regeneration of lung tissue.
The term "therapeutically effective amount" is to be understood as referring to an amount of the multipotent AECs (ie a cell number) that will, at least, arrest the lung disease or condition being treated. Such an amount may vary considerably depending upon a range of factors such as the mode of administration, the age and/or body weight of the subject, and the severity of the lung disease or condition to be treated. However, typically, the amount will be in the range of about 1 x 105 to 1 x 1010, more preferably, 1 x 108 to 1 x 1010 multipotent AECs.
The multipotent AECs may be administered to the subject in combination with one or more active agents. For example, the AECs may be administered with one or more of the drugs presently used to alleviate and/or prevent symptoms of a lung disease or condition (eg a bronchodilator or antiinflammatory corticosteroid). Alternatively, the AECs may be administered with an agent which causes a reduction in the expression or activity of one or more of the pro-inflammatory cytokines IL-I, JL-2, IL- 6, IFN-γ and TNF-α (eg a specific inhibitor or antagonist agent or specific antibody), and/or the expression or activity of the pro-fibrotic cytokine TGF-β (eg a specific TGF-β inhibitor or antagonist agent or specific anti-TGF-β antibody). The AECs may also be administered with synthetic or animal-derived lung surfactant (eg colfosceril palmitate-based products (such as Exosurf® marketed by GlaxoSmithKline pic, Brentford, Middlesex, United Kingdom) and surfactant extracted from bovine lung lavage fluid (such as Alveofact® marketed by Boehringer Ingelheim GmbH, Ingelheim, Germany)), to open unperfused areas of the lung to thereby aid access of the AECs to sites of injury.
The multipotent AECs are preferably characterised by the expression of one or more markers listed in Table 1 below. More preferably, the multipotent AECs are characterised by the expression of one or more pluripotent stem cell markers. For hAECs, the pluripotent markers will typically be selected from Oct-4, Nanog and Sox-2.
Further, the multipotent hAECs are preferably characterised in that they show low expression of MHC class IA and II proteins. This is preferred since it offers the potential use of the hAECs in subjects of divergent histocompatibility. The multipotent hAECs used in the method of the invention may, however, be autologous (and be sourced from preserved samples such as cryopreserved samples).
Multipotent AECs may be derived from amnion tissue by standard methods. For example, amnion tissue stripped from term placenta, can be treated with a suitable proteolytic enzyme (eg trypsin), and the purity of dispersed cells assessed by subjecting an aliquot to flow analysis (eg by FACS) for cytokeratin-7, an epithelial cytoskeletal protein found in AECs, and/or one or more of Oct-4, Nanog and Sox-2. Alternatively, the dispersed cells may be sorted/assessed by flow analysis for one or more relevant cell surface marker (eg SSEA-4). The dispersed/sorted cells may be stored for later use in accordance with standard methods. The dispersed/sorted cells may also be expanded by culturing under conditions which avoid cell differentiation (eg by culturing in a basal medium such as Dulbecco's Modified Eagle's Medium (DMEM)/F12 supplemented by animal-free serum or serum substitutes). When required, the dispersed/sorted, and optionally expanded, cells can be prepared for immediate administration to the subject by, for example, isolating the AECs from the culture/storage media and resuspending the AECs in a pharmaceutically-acceptable carrier.
Further, the multipotent AECs may be partially substituted with derivative cells; that is, cells which have been at least partially differentiated from the AECs (ie the invention may utilise "pre- differentiated" cells). Such derivative cells may be produced, for example, by culturing AECs under suitable conditions in a commercially available SAGM (eg SAGM products marketed by Lonza Group Ltd, Basel, Switzerland). Where the invention utilises derivative cells, the cells may be administered in an AEC:derivative cell ratio in the range of about 5: 1 to about 1 :5, more preferably about 4: 1 to about 1 :1.
Moreover, the multipotent AECs may be partially substituted with one or more other (ie non- derivative) cell types, including one or more other stem cell types. Where the invention utilises other cell types, the cells may be administered in an AEC:other cell ratio in the range of about 5:1 to about 1 :5, more preferably about 4: 1 to about 1 : 1.
However, while derivative cells or other cell types can be co-administered, the method of the invention, most preferably, utilises AECs only.
In a second aspect, the present invention provides the use of multipotent amnion epithelial stem cells (AECs) in the preparation of a composition for cellular therapy for a lung disease or condition in a subject, wherein said composition comprises said AECs in combination with a pharmaceutically-acceptable carrier. Preferably, the multipotent AECs used in the use of the invention are human amnion epithelial stem cells (hAECs). Further, the AECs used in the method of the invention are preferably AECs derived from amnion of term, or near term, placenta.
In addition to the AECs, the composition may also comprise cells belonging to one or more other cell types including one or more other stem cell types and/or one or more wholly or partially differentiated cell types (eg derivative cells of AECs; that is, cells which have been at least partially differentiated from AECs).
The composition may also further comprise one or more active agents (eg a bronchodilator or antiinflammatory corticosteroid).
The cellular therapy of the present invention offers the possibility of reducing, or at least arresting, lung diseases or conditions that have to date only been treated by the use of drugs to alleviate and/or prevent symptoms. Since the long term use of these drugs can also lead to serious side- effects (eg long term use of corticosteroids is associated with the development of several debilitating diseases and conditions including diabetes, osteoporosis and cataract formation), the identification and development of a cellular therapy for lung diseases or conditions, such as that of the present invention, is highly desirable.
The cellular therapy of the present invention is able to bring about a reduction in lung inflammation, prevent fibrosis and reduce collagen concentration in the lung following lung injury.
Accordingly, in a third aspect, the present invention provides a method of reducing inflammation in the lung of a subject, said method comprising administering to said subject a therapeutically effective amount of multipotent amnion epithelial stem cells (AECs).
And, in a fourth aspect, the present invention provides a method of preventing fibrosis in the lung of a subject, said method comprising administering to said subject a therapeutically effective amount of multipotent amnion epithelial stem cells (AECs).
Further, in a fifth aspect, the present invention provides a a method of reducing collagen concentration in the lung of a subject, said method comprising administering to said subject a therapeutically effective amount of multipotent amnion epithelial stem cells (AECs).
It will be appreciated by persons skilled in the art that numerous variations and/or modifications may be made to the invention defined in the first to fifth aspects above. Further, the ability of AECs administered by systemic transfusion, bronchoscopic or intra-nasal instillation to migrate to the lung, provides a means or "vector" for delivering a therapeutic agent (eg a therapeutic protein or peptide, gene therapy agent or gene silencing agent) to the lung. For example, for the treatment of cystic fibrosis (CF), an exogenous polynucleotide molecule encoding cystic fibrosis transmembrane conductance regulator (CFTR) could be introduced into autologous AECs (ie lacking a functional CFTR gene) and thereafter administered to a subject suffering from CF, to provide a gene therapy for CF.
Accordingly, it is to be understood that the present invention extends to a method of cellular therapy for a lung disease or condition in a subject, comprising administering to said subject a therapeutically effective amount of multipotent amnion epithelial stem cells (AECs) comprising a therapeutic agent as described above, as well as to a composition therefor (ie a composition comprising AECs comprising a therapeutic agent as described above, in combination with a pharmaceutically-acceptable carrier) and, further, to the use of AECs comprising a therapeutic agent as described above in the preparation of a composition for cellular therapy for a lung disease or condition in a subject, wherein said composition comprises said AECs in combination with a pharmaceutically-acceptable carrier.
In order that the nature of the present invention may be more clearly understood, preferred forms thereof will now be described with reference to the following non-limiting examples.
EXAMPLES
Example 1 Human amnion epithelial cell abrogation of fibrosis and augmentation of lung repair
Materials and Methods
Isolation and culture of hAECs
Amnion was obtained from women undergoing elective caesarean section at term. hAECs were isolated and characterised using methods described previously (Ilancheran, S., et al., 2007). Briefly, tissue was digested twice in 0.25% trypsin:EDTA solution (Invitrogen Corporation, Carlsbad, CA, United States of America) for 20 minutes. Trypsin was inactivated with foetal calf serum (FCS, Invitrogen) and dispersed cells washed in M199 medium (Invitrogen). Cells were then analysed by flow cytometry for cytokeratin-7 (Dako Denmark A/S, Glostrup, Denmark), an epithelial cytoskeletal protein found in hAECs. Batches with >99% cytokeratin-7 positive cells were plated onto type IV collagen (Roche Diagnostics, Basel, Switzerland) coated vessels and cultured in Small Airway Epithelial Basal Medium (SABM) (Clonetics, Walkersville, MD, United States of America) and supplements (ie retinoic acid, epidermal growth factor 1 (EGF-I), epinephrine, transferrin, insulin, tri-iodothyronine, bovine pituitary extract, bovine serum albumin (BSA), fatty acid-free serum and hydrocortisone) provided. Control cultures were grown in
DMEM/F-12 medium (Invitrogen) with 10% human serum or FCS. Cultures were maintained for 4 weeks (n=12).
Quantitative RT-PCR Total RNA was isolated from hAECs (n=6) using RNeasy columns (Qiagen, Hilden, Germany). One μg of total RNA was converted to cDNA using the High-Capacity cDNA Archive Kit (Applied Biosystems, Foster City, CA, United States of America). The cDNA, diluted 1 :20, was mixed with reagents (TaqMan Universal PCR Master Mix, Applied Biosystems), PCR primers and TaqMan probes for Oct-4, Sox-2, Nanog, Nkx2.1, SPC, AQ-5 and β-actin (Applied Biosystems #Hs0189506 l_ul, #Hs00602736_sl, #Hs02387400_gl, #NM_003317.3, #Hs00161628_ml, #Hs00387048_ml, respectively).
Oct-4 is a transcription factor that is highly expressed in the cells of the inner cell mass (ICM) and pluripotent embryonic carcinoma cells lines; low expression of Oct-4 yields the growth of trophoblastic cells only, while elevated expression of this transcription factor leads to differentiation of embryonic stem (ES) cells to embryonic and extra-embryonic tissue. Nanog is a homeodomain factor and is expressed in vivo in the morula and ICM of the epiblast. As such, it is a marker of pluripotent cells. Sox-2 is a transcription factor that is co-expressed with Oct-4 in the ICM as well as in ES cells and is essential for the function of Oct-4. Nkx-1 is also known as thyroid transcription factor- 1 (TTF-I), and represents the earliest known marker of the respiratory lineage. SPC is highly specific for type II alveolar epithelial cells and Aquaporin-5 (AQ-5) is a lung (alveolar) epithelial cell markers.
The PCR parameters were: 95°C, 7sec and 6O0C, 20sec for 40 cycles. Data was normalised against β-actin and expression levels, relative to the human embryonic stem cell line ES-I (for Oct-4,
Nanog, Sox-2, and Nkx2.1), or adult human lung (SPC and AQ-5) was calculated using the ΔΔCt method.
Transmission electron microscopy (TEM) Cells were fixed in 2.5% glutaraldehyde in 0.1 M cacodylate buffer for 2 hours at room temperature (RT) and left overnight at 40C. Cells were then post-fixed in 1% osmium tetroxide, dehydrated in graded acetone, infiltrated and embedded in Spurr's resin at 6O0C for 24 hours. Ultra-thin sections (80nm) were thereafter stained with 3% uranyl acetate and Renoylds stain.
Flow cytometry hAECs (2.5xlO5) were incubated with antibodies against caveolin (1 :100), AQ-5 (1 :50), SPC (1 :50), human leukocyte antigen (HLA)-DP5-DQ5-DR (lOμg/ml) or allophytocyanin (APC)- conjugated HLA-A5-B5-C (10μl)5 for 1 hour at RT. After several washes, cells were incubated with 1:10 diluted APC-conjugated donkey anti-rabbit (caveolin), donkey anti-goat (AQ-5, SP-C), or goat anti-mouse (HLA-DP,-DQ,-DR) secondary antibodies for 30 minutes at RT. Cells were washed to remove excess secondary antibodies and analysed by flow cytometry. Primary antibodies for caveolin, AQ-5 and SPC were purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA5 United States of America) and HLA antibodies and secondary antisera from Becton, Dickinson and Company (Franklin Lakes, NJ, United States of America). Further flow cytometry analysis of the hAECs (including a popoulation (P5) that had been passaged 5 times) was undertaken, using similar methodology and monoclonal antibodies to the cell surface markers (Becton, Dickinson and Company) indicated in Table 2 below, in comparison with human bone marrow mesenchymal stem cells (hMSC). The percentage of the cells within the cell types that were "positive" for each marker was recorded.
Injection of hAECs into SCID mice
Bleomycin sulphate in saline (0.15mg) was administered intranasally under weak ether anaesthesia to induce pulmonary fibrosis in 8-week-old Severe Combined Immune Deficient (SCID) mice (n=30). Twenty four hours after bleomycin instillation, mice were injected with hAECs (1 x 106 in 0.2ml), via the tail vein, while control mice were injected with 0.2ml saline. Saline was administered intranasally to another group of mice (n=15); these mice received no further treatment. A parallel experiment where human lung fibroblast cells (1x106) were used in place of hAECs was also carried out. Further, hAECs were also injected into healthy mice (n=12). Moreover, other SCID mice were injected, via the tail, with hAECs (1 x 106 in 0.2ml saline) two weeks after bleomycin administration. In controls for these latter animals, lung fibroblasts (1 x 106 in 0.2ml saline) isolated from lung resections were injected (via the tail) into SCID mice instead of hAECs 2 weeks after bleomycin administration. Animals were sacrificed two and four weeks post treatment (nb. four weeks only for the mice injected with hAECs/lung fibroblasts two weeks after bleomycin administration). Lung tissue was snap frozen and stored at -800C and fixed in 10% formalin for paraffin embedding. Immunohistochemical analysis of lung tissue
Paraffin embedded tissue sections, 5μm in thickness, were dewaxed and rehydrated. Antigen retrieval was carried out by by heating in citrate buffer. After quenching endogenous peroxidase activity and blocking non-specific binding, sections were incubated with antibodies against human inner membrane mitochondria (MM; 1:10, AbD Serotec, Kidlington, Oxfordshire, United Kingdom) and human SPC (1:50, Santa Cruz Biotechnology, Inc.) for 1 hour at 370C. Isotype- matched mouse IgG2a and goat IgG were applied to sections serving as negative controls. Antibody binding was detected using horseradish peroxidase-labelled biotin-streptavidin secondary antibody (LSAB kit, Dako), and immunostaining visualised using 3,3'-diaminobenzidine chromogen. Co-localisation of IMM and SPC was assessed by examining serial tissue sections. For inflammation and fibrosis, sections were scored by the Ashcroft method (Ashcroft, T., et al., 1988).
Cytokine mRNA expression Total RNA was extracted from lungs of SCID mice after two and four weeks of receiving saline, bleomycin and bleomycin+hAECs (n=5/group). Following DNAse treatment, 1 μg of RNA was converted to cDNA with Superscript III (Invitrogen). The cDNA (diluted 1 :20) was amplified with PCR primers against IL-I, IL-2, interleukin-4 (IL-4), IL-6, interleukin-10 (IL-10), interferon- gamma (INFγ), TNF-α, TGF-β, MIF and MIP. Cycling parameters used for quantitative PCR were: denaturation 95°C, 7sec, annealing 6O0C, 7-15sec and extension 72°C, 15sec for 40 cycles. After normalising data to β-actin, expression relative to saline treated control mice was calculated by the ΔΔCt method.
Collagen content The collagen content in lungs of mice receiving saline, bleomycin and bleomycin+hAEC
(n=6/group) was measured using the hydroxyproline assay as described previously (Hewitson, T.D., et al., 2007). Briefly, lungs were lyophilised, hydrolysed in HCl and absorbance measured at 558nm. Collagen content was calculated by multiplying the hydroxyproline measurements by 6.94 and expressed as a proportion of dry weight of tissue (Gallop, P.M. and M.A. Paz, 1975).
MMP activity and TIMP expression
MMPs were extracted from tissue obtained from the three groups of mice (n=6/group). MMP-2 and MMP-9 activity was assessed by gelatin zymography as described previously (Woessner, J.F. Jr, 1995). Zymographs were scanned on a densitometer (Gel Scan-710, Bio-Rad Laboratories, Inc., Hercules, CA, United States of America) and gelatinase activity quantified using Quantity-One software (Bio-Rad). The data was expressed as the mean±SEM. TMP-1, TIMP-2, TIMP-3 and TMP-4 mRNA expression in lung tissue was measured by quantitative RT-PCR as described above. Reagents, primers and TaqMan probes were purchased from Applied Biosystems (#Mm00441818_ml, #Mm00441825_ml, #Mm00441826_ml, #Mm00446568_ml). After normalising data to β-actin, expression relative to saline instilled mice was calculated by the ΔΔCt method.
Statistical analyses
Analysis of treatment effects between the different groups was performed using a one-way ANOVA analysis. P<0.05 was considered to be statistically significant.
Results and Discussion
hAECs were derived from the placenta by stripping off the membrane and growing the hAECs in cell culture. These cells demonstrated clonality and, consistent with their epiblast derivation (Chambers L, et al., 2003; and Mitsui K., et al., 2003), expressed several markers of pluripotency, namely Oct-4, Nanog, Sox-1 and c-kit (Figure Ia and Table 1). The cells were not of vascular endothelial or haematopoetic lineage since they were "negative" for CD31, CD34 and CD45 markers (see Table 1). In comparative flow cytometry analysis, it was found that expression of cell surface markers by HAECs was considerably different to that of hMSC (most prominently in the CD90 marker that is characteristic of hMSCs), thereby indicating that the hAECs are of a distinct phenotype.
As part of a focus on lung differentiation and repair by hAECs, it was also demonstrated, by quantitative PCR, that the isolated hAECs expressed Nkx 2.1 (otherwise known as TTF-I), which is the earliest lineage marker of the developing lung (Figure Ia). Nkx. 2.1 is a critical transcription factor for branching morphogenesis and the formation of type II alveolar epithelial cells during lung development.
After noting the expression of Nkx 2.1 by the hAECs, the cells were then cultured in SAGM which had previously been shown to induce the differentiation of embryonic and umbilical cord blood- derived stem cells into type II alveolar epithelial cells of the lung (Berger, M. J., et al, 2006; and AH, N.N., et al., 2002). SAGM is primarily utilised in the maintenance of airway epithelial cells in culture and contains hydrocortisone, human epidermal growth factor (hEGF-1) and retinoic acid which are also factors present during lung development. HAECs cultured in SAGM for 2 and 4 weeks, demonstrated an increase in the gene expression of the lung specific gene surfactant protein C (SPC) (see Figure Ia). The increase in mRNA expression was translated into elevated protein expression of SPC shown by FACs analysis (Figure Ib). Ultra-structural analyses revealed that hAECs grown in SAGM for 4 weeks contained a distinct cell population demonstrating cytoplasmic organelles containing lamellar bodies, lipid filled vacuoles and surface microvilli formation that were indicative of type II alveolar epithelial cell differentiation. These features were not present in hAECs cultured in Dubelcos Modified Eagle Medium (DMEM)/F12. Type II alveolar epithelial cells constitute one third of the surface area of the lung but make up 66% of the cells in the distal lung and are responsible for the secretion of surface tension reducing surfactant protein, hi addition, type II alveolar epithelial cells differentiate into the gas-exchanging type I alveolar epithelial cells during lung repair. Notably, hAECs demonstrate low expression of MHC antigens when freshly isolated and following in vitro differentiation, which widens the potential for the use of these cells for clinical application (see Figure Ic and Id).
Having demonstrated the potential for hAECs to differentiate in vitro into a cell type that is central to lung repair, the ability of the hAECs to repair injured lung tissue in vivo, was then investigated.
The bleomycin-induced model of lung inflammation and fibrosis is well characterised and reflects the phases of lung injury in human subjects. The present applicant had previously established the model in SCDD mice, and this allowed the evaluation of the functional role of human cells in augmenting lung repair. In particular, intra-nasally administered bleomycin induced an increase in both the inflammatory and fibrosis score on histology at both the 2 and 4 week time points (see
Figure 2b). Further, there was an increase in the pro-inflammatory cytokines IL-I, IL-2, IL-6, IFNγ and TNF-α, as well as the pro-fibrotic cytokine TGF-β at the 2 week time-point when maximum injury and inflammation occurs.
hi order to treat lung injury, hAECs were injected into the tail vein 24 hours following intra-nasal administration of bleomycin and were demonstrated in the lung by anti-human inner mitochondrial membrane (IMM) protein positive staining of cells at both 2 and 4 week time points. The anti- mitochondrial staining was negative in mouse lung tissue but present only in human controls (data not shown) thereby confirming the specificity and sensitivity of the antibody for the identification of human cells in the mouse lung. Also, when healthy mice were injected with hAECs, cells could not be detected in lung tissue 2 weeks post injection indicating that hAECs do not "traffic" to uninjured lung. Immunohistochemistry located the hAEC cells to the fibrotic and alveolar epithelial region of the lung (1 IMM-positive cell per 100 cells on high power field, n=8) as compared to no cells identified in uninjured healthy control mice subjected injected with hAECs. Moreover, 55 ± 5 % of hAECs in the alveoli of the lung also expressed SPC as evidenced by positive staining with a human specific antibody. Since the injected hAECs did not express SPC, the data indicates that the hAECs underwent differentiation into a respiratory lineage in the lung. Importantly, and despite the expression of some pluripotent markers, the hAECs did not form teratomas or tumours in the mice up to 4 weeks post-injection.
Notably, administration of hAEC by injection, reduced the inflammatory and fibrosis score at both 2 and 4 weeks (Figure 2c). Further, it was demonstrated that there was a significant reduction in macrophage number in the lung between the bleomycin treated mice and the mice treated with bleomycin and hAECs. In looking to elucidate the mechanism by which this reduction in macrophage number is achieved, the role of MIF and MIP was investigated. These proteins are known to be central to the absence of a rejection reaction when hAECs are used to repair damaged corneal epithelium (Hori, J. et al, 2006; and Wang, M. et al., 2006).
It was found that there was increased MIF and reduced MIP expression in lung homogenates that were treated with hAECs as compared to bleomycin controls, which appears to explain the reduced recruitment of macrophages to the sites of injury. Further, the reduction in macrophage recruitment may explain the down-regulation of the cytokines IL-I, IL-2, IL-6, IFN-γ and TNF-α at 2 weeks by hAECs (Figure 2c); IL-I is elevated during bleomycin lung injury and is usually secreted by monocytes and macrophages, promoting fibrosis by increasing fibroblast proliferation and collagen production. Indeed, the decrease in IL-I by niacin and taurine during bleomycin lung injury parallels the improvement in symptoms of lung fibrosis (Gurujeyalakshmi, G., et al., 2000). As such, the inhibition of IL-I observed in this Example further points to the ability of hAECs to produce anti-fibrotic effects on the lung.
Notably, the cytokines IL-2, TNF-α and IFN-γ were significantly elevated in response to bleomycin in mouse strains prone to develop injury (C57/B16) as compared to the resistant strains of mice such as Balb/C, thus supporting the role for these cytokines in the progression of lung fibrosis. As such, inhibition of these cytokines by hAEC administration would serve to further reduce the development of fibrosis. In addition, several previous studies have demonstrated that the reduction in endogenous lung IFNγ and IFNγ knock-out mice are resistant to bleomycin injury. Moreover, TNFR-deficient mice are protective against bleomycin fibrosis by inhibiting the activation of TGF- β. As such, the significant reduction in TNF-α by HAEC administration provides even further support to the anti-fibrotic role of hAECs. Table 1 Characterisation of human amniotic epithelial stem cells
Figure imgf000017_0001
adata from flow analyses; bdata from immunocytochemistry; nd = not detected; cdata from real time qPCR
Table 2 Characterisation of human amniotic epithelial stem cells
Figure imgf000018_0001
Figure imgf000019_0001
Figure imgf000020_0001
nd = not determined; Also tested but recording 0% for hMSC and hAEC was: CDIa, CD2, CD3, CD4, CD7, CD8, CD9, CDl Ia, CDl Ib, CDl Ic, CD 14, CD 18, CD 19, CD20. CD21, CD22, CD23, CD25, CD28, CD30, CD31, CD32, CD33, CD35, CD37, CD38, CD40, CD41a, CD42a, CD43, CD49d, CD49f, CD50, CD51/61, CD54, CD57, CD61, CD62e, CD62L, CD64, CD66, CD70, CD72, CD74, CD80, CD85J, CD87, CD88, CD94, CD97, CD99, CD99R, Cdl03, CDl 10, CDl 17, CD123, CD134, CD135, CD137(2), CD140a, CD152, CD154, CD158a, CD158b, CD161, CD162, CD163, CD172b, CD182, CD200, CD201, CD206, CD220, CD221, CD226, CD244, CMRF-5, CLIP, Vb8-TCR, HLA-DR, IntegrinB7, LAIR-I, NK-G2d, NK-P46, ABC-G2, Blood Group A, Siglec-6, Siglec-7 and B5-TCR.
Turning to IL-6, this cytokine is secreted by fibroblasts, epithelial cells and macrophages, and has been found to be significantly elevated during bleomycin lung injury. It has been postulated that IL-6 acts in synergy with TNF-α to increase the levels of MIP, thereby enhancing the recruitment of macrophages and perpetuating inflammation and fibrosis.
TGF-β is a central cytokine in the pathogenesis of lung fibrosis. At day 14 following bleomycin lung injury, TGF-β is increased in macrophages, epithelial cells, fibroblasts and myofibroblasts. Indeed, several studies have demonstrated that TGF-β has a bimodal effect on fibroblast proliferation, stimulates monocyte production of cytokines and acts as a potent stimulator of collagen deposition (Kang, H.R., et al., 2007). Moreover, several studies have conclusively shown that the amelioration of bleomycin fibrosis is dependent on the inhibition of TGF-β. Accordingly, the significant down regulation of TGF-β also indicates that hAECs have an anti-fibrotic role. In this example, it has been clearly demonstrated that hAECs reduce the cellular infiltration of the lung and reduce the pro-inflammatory cytokines IL-I, TNF-α and IL-6.
Further to the anti-inflammatory effects of hAECs, the role of hAECs in collagen deposition and fibrosis following bleomycin lung injury was investigated. Bleomycin induces excessive collagen deposition and fibrosis at 14 days post-injury resulting in the development of fibroblastic foci. These foci are characterised by the distortion of lung architecture, fibroblast and myofibroblast proliferation, collagen deposition and a reduction in functional lung. All of these are "generic" characteristics of fibrosis in all organs and show remarkable homology to pathology in human subjects.
Collagen deposition was measured by using a hydroxyproline assay. This showed an expected increase in collagen deposition in mice treated with bleomycin alone but a significant fall in the cohort subjected to bleomycin and hAEC injection (see Figures 3a and 3b). Notably, there was no influence of hAECs injected into healthy mice on collagen deposition. Further, it was found that the reduction of collagen was specific to hAECs since the injection of primary human lung fibroblasts into bleomycin exposed did not influence collagen levels. The results achieved with mice injected two weeks after treatment with bleomycin (see Figure 3b) indicates that hAECs abrogates fibrosis. This is significant since the maximum level of fibrosis in the bleoycin-induced mouse model occurs between 2-4 weeks; as such, the results indicate that hAECs injected at the time of maximal fibrosis are able to reduce or reverse collagen deposition without necessarily inhibiting the preceding inflammation that occurs maximally in the first two weeks following bleomycin lung injury.
Since matrix metalloproteinases (MMPs) are principally responsible for the degradation of extracellular matrix (ECM) proteins such as collagen, the expression of MMPs and their endogenous inhibitors (TIMPs) in homogenates of lung tissue was also assessed. Zymographic analysis demonstrated that MMP-2 and MMP-9 in bleomycin and hAEC lung homogenates were significantly elevated above bleomycin alone (Figure 3c). This appears to be a unique response to lung injury since there was no elevation of MMPs in lung samples from healthy controls injected with hAECs (data not shown). In addition, there was no additional increase in MMP levels following the injection of primary lung fibroblasts as compared to bleomycin alone (data not shown). MMP-2 is responsible for the degradation of most ECM proteins and is also implicated in alveolar regeneration. Further, studies in human subjects have previously demonstrated that the up- regulation of MMP-2 was a strong predictor of a decrease in fibrosis in subjects with hypersensitivity pneumonitis (Selman, M. and A. Pardo, 1991). As such, it is considered that the significant up-regulation of MMP-2 is responsible for the reduction in collagen concentration within the lung.
In addition, there was a significant difference in MMP-9 expression between bleomycin alone versus bleomycin and HAEC treated mice. MMP-9 is instrumental in cleaving several ECM proteins but not directly implicated in active fibrosis since down regulation of MMP-9 did not influence collagen levels in bleomycin treated mice. However, the up-regulation of MMP-9 may be an additive degradative molecule during collagen deposition. Tissue inhibitors of MMPs (TIMPs) are the major endogenous inhibitors of MMPs and bind these molecules in a 1:1 stoichiometry. There are 4 homologues of TIMPs, TIMP- 1-4, each having specific functions. Inhibition of TIMP- 1 augmented the inflammatory response to bleomycin but not an increase in fibrosis. Similarly, TIMP-2 and TIMP-3 are also elevated in response to bleomycin. From lung transcripts, it was demonstrated that there was a down-regulation of TIMP-I, TIMP-3 and TIMP-4 in mice treated with hAECs at day 28 post bleomycin lung injury (Figure 3d). Moreover, there is a causal relationship between TDVIP elevation and fibrosis since TIMP concentrations increased prior to the phase of collagen deposition in the bleomycin mouse model at days 14 to 28 post-injury. Also, mouse strains resistant to bleomycin fibrosis did not demonstrate an increase in TIMP-I levels following injury. Thus, the reduction in TIMP levels accompanied by the increase in MMPs indicates the existence of a micro-environment that favours ECM degradation in response to HAEC injection, thereby reducing fibrosis.
Taken together, there are several mechanisms by which hAEC appears to directly augment lung repair. The differentiation of hAECs to type II alveolar epithelial cells, reduction in macrophage recruitment and inhibition of cytokine expression limits damage to the lung. In addition, the elevation in MMPs, with a consequent inhibition of TIMPs, abrogates fibrosis.
Throughout this specification the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated element, integer or step, or group of elements, integers or steps, but not the exclusion of any other element, integer or step, or group of elements, integers or steps.
All publications mentioned in this specification are herein incorporated by reference. Any discussion of documents, acts, materials, devices, articles or the like which has been included in the present specification is solely for the purpose of providing a context for the present invention. It is not to be taken as an admission that any or all of these matters form part of the prior art base or were common general knowledge in the field relevant to the present invention as it existed in Australia or elsewhere before the priority date of each claim of this application.
It will be appreciated by persons skilled in the art that numerous variations and/or modifications may be made to the invention as shown in the specific embodiments without departing from the spirit or scope of the invention as broadly described. The present embodiments are, therefore, to be considered in all respects as illustrative and not restrictive.
REFERENCES
1. AH, N.N., et al., Derivation of type II alveolar epithelial cells from murine embryonic stem cells. Tissue Engineering 8:541-550 (2002).
2. Ashcroft, T., et al., Simple method of estimating severity of pulmonary fibrosis on a numerical scale. J Clin Pathol 41 :467-470 (1988).
3. Berger, M.J., et al., Differentiation of umbilical cord blood-derived multilineage progenitor cells into respiratory epithelial cells. Cytotherapy 8:480-487 (2006).
4. Chambers I., et al, Functional expression cloning of Nanog, a pluripotency sustaining factor in embryonic stem cells. Cell 113(5):551-552 (2003).
5. Dhanireddy S., et al., Mechanical ventilation induces inflammation, lung injury, and extrapulmonary organ dysfunction in experimental pneumonia. Laboratory Investigation 86:790-799 (2006).
6. Gallop, P.M. and M.A. Paz, Posttranslational protein modifications, with special attention to collagen and elastin. Physiological Reviews 55:418-487 (1975).
7. Gurujeyalakshmi, G., et al., Taurine and niacin block lung injury and fibrosis by down- regulating bleomycin-induced activation of transcription nuclear factor-kappaB in mice. J Pharmacol Exp Therap 293:82-90 (2000).
8. Hewitson, T.D., et al., Endogenous relaxin is a naturally occurring modulator of experimental renal tubulointerstitial fibrosis. Endocrinology 148:660-669 (2007).
9. Hori, J., et al., Immunological characteristics of amniotic epithelium. Cornea 25:S53-58
(2006).
10. Ilancheran S., et al., Stem cells derived from human fetal membranes display multi-lineage differentiation potential. Biol Reprod 77:577-588 (2007).
1 1. Kang, H.R., et al., Transforming growth factor (TGF)-beta 1 stimulates pulmonary fibrosis and inflammation via a Bax-dependent, bid activated pathway that involves matrix metalloproteinase-12. J Biol Chem 282:7723-7732 (2007).
12. Mitsui K., et al., The homeoprotein Nanog is required for maintenance of pluripotency in mouse epiblast and ES cells. Cell 113(5):631-642 (2003). 13. Morley, CJ. , et al., The biochemistry and physiology of fetal pulmonary surfactant. In "Preterm labour". Proc. V study group of Royal College of Obstetrics and Gynaecology. Eds. Anderson, A. B., Berad, R., Brundenell, J.M. and Dunn, P.M., pp 261-272.
14. Parolini, O., et al., Isolation and characterization of cells from human term placenta: outcome of the first international workshop on placenta derived stem cells. Stem Cells
26:300-311 (2008).
15. Selman, M. and A. Pardo, Potential role of proteases in pulmonary fibrosis. Annals of the New York Academy of Sciences 624:297-306 (1991).
16. Wang, M., et al., Immunogenicity and antigenicity of allogeneic amniotic epithelial transplants grafted to the cornea, conjunctiva, and anterior chamber. Investigative
Ophthalmology & Visual Science 47:1522-1532 (2006).
17. Woessner, J. F. Jr, Quantification of matrix metaloproteinases in tissue samples. Methods in Enzymology 248:510-528 (1995).

Claims

1. A method of cellular therapy for a lung disease or condition in a subject, comprising administering to said subject a therapeutically effective amount of multipotent amnion epithelial stem cells (AECs).
2. The method of claim 1, wherein the AECs are human amnion epithelial stem cells (hAECs).
3. The method of claim 2, wherein the hAECs are characterised by the expression of one or more markers selected from Oct-4, Nanog and Sox-2.
4. The method of any one of claims 1 to 3, wherein the AECs are derived from amnion of term, or near term, placenta.
5. The method of any one of claims 1 to 4, wherein the AECs are autologous.
6. The method of any one of claims 1 to 5, wherein the lung disease or condition to be treated is chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF) or idiopathic pulmonary fibrosis (IPF), or damage due to chemo- and/or radio-therapy, industrial accidents or exposure to toxic compounds or particulates.
7. The method of any one of claims 1 to 5, wherein the lung disease or condition to be treated is acute respiratory distress syndrome (ARDS) or acute lung injury (ALI).
8. The method of any one of claims 1 to 5, wherein the lung disease or condition to be treated is ventilator associated lung injury (VALI).
9. The method of any one of claims 1 to 5, wherein the lung disease or condition to be treated is foetal respiratory distress syndrome (RDS).
10. The method of claim 9, wherein the AECs are administered in combination with a synthetic or animal-derived surfactant.
1 1. The use of multipotent amnion epithelial stem cells (AECs) in the preparation of a composition for cellular therapy for a lung disease or condition in a subject, wherein said composition comprises said AECs in combination with a pharmaceutically-acceptable carrier.
12. The use of claim 11, wherein the AECs are human amnion epithelial stem cells (hAECs).
13. The use of claim 12, wherein the hAECs are characterised by the expression of one or more markers selected from Oct-4, Nanog and Sox-2.
14. The use of any one of claims 11 to 13, wherein the AECs are derived from amnion of term, or near term, placenta.
15. The use of any one of claims 11 to 14, wherein the AECs are autologous.
16. The use of any one of claims 11 to 15, wherein the lung disease or condition to be treated is chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF) or idiopathic pulmonary fibrosis (IPF), or damage due to chemo- and/or radio-therapy, industrial accidents or exposure to toxic compounds or particulates.
17. The use of any one of claims 11 to 15, wherein the lung disease or condition to be treated is acute respiratory distress syndrome (ARDS) or acute lung injury (ALI).
18. The use of any one of claims 11 to 15, wherein the lung disease or condition to be treated is ventilator associated lung injury (VALI).
19. The use of any one of claims 11 to 15, wherein the lung disease or condition is foetal respiratory distress syndrome (RDS).
20. The use of claim 19, wherein the composition further comprises a synthetic or animal-derived surfactant.
21. A method of reducing inflammation in the lung of a subject, said method comprising administering to said subject a therapeutically effective amount of multipotent amnion epithelial stem cells (AECs).
22. A method of preventing fibrosis in the lung of a subject, said method comprising administering to said subject a therapeutically effective amount of multipotent amnion epithelial stem cells (AECs).
23. A method of reducing collagen concentration in the lung of a subject, said method comprising administering to said subject a therapeutically effective amount of multipotent amnion epithelial stem cells (AECs).
24. The method of any one of claims 21 to 23, wherein the AECs are human amnion epithelial stem cells (hAECs).
25. The method of claim 24, wherein the hAECs are characterised by the expression of one or more markers selected from Oct-4, Nanog and Sox-2.
26. The method of any one of claims 21 to 25, wherein the AECs are derived from amnion of term, or near term, placenta.
27. The method of any one of claims 21 to 26, wherein the AECs are autologous.
PCT/AU2008/000753 2007-05-28 2008-05-28 Treatment of chronic lung disease WO2008144820A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP2010509623A JP5567476B2 (en) 2007-05-28 2008-05-28 Treatment of chronic lung disease
EP08748014.1A EP2164505B1 (en) 2007-05-28 2008-05-28 Treatment of chronic lung disease
AU2008255634A AU2008255634B2 (en) 2007-05-28 2008-05-28 Treatment of chronic lung disease
US12/601,570 US20100266552A1 (en) 2007-05-28 2008-05-28 Treatment of chronic lung disease

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
AU2007902844A AU2007902844A0 (en) 2007-05-28 Treatment of chronic lung disease
AU2007902844 2007-05-28

Publications (1)

Publication Number Publication Date
WO2008144820A1 true WO2008144820A1 (en) 2008-12-04

Family

ID=40074453

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2008/000753 WO2008144820A1 (en) 2007-05-28 2008-05-28 Treatment of chronic lung disease

Country Status (5)

Country Link
US (1) US20100266552A1 (en)
EP (1) EP2164505B1 (en)
JP (1) JP5567476B2 (en)
AU (1) AU2008255634B2 (en)
WO (1) WO2008144820A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2374871A1 (en) * 2008-12-17 2011-10-12 Beijing Health & Biotech (H&B) Co., Ltd Pluripotent stem cells, method for preparation thereof and uses thereof
EP2375907A1 (en) * 2008-11-21 2011-10-19 Anthrogenesis Corporation Treatment of diseases, disorders or conditions of the lung using placental cells
US20130302294A1 (en) * 2009-02-04 2013-11-14 Yale University Tissue Engineering of Lung
WO2016197196A1 (en) 2015-06-12 2016-12-15 Prince Henry's Institute Of Medical Research Trading As The Hudson Institute Of Medical Research A method of treatment
US9828583B2 (en) 2012-01-13 2017-11-28 The General Hospital Corporation Isolated human lung progenitor cells and uses thereof
WO2018023170A1 (en) * 2016-08-04 2018-02-08 Hudson Institute of Medical Research A method of treatment
US10104880B2 (en) 2008-08-20 2018-10-23 Celularity, Inc. Cell composition and methods of making the same
CN110101873A (en) * 2018-10-27 2019-08-09 广州呼吸健康研究院 A method of utilizing the stem-cell therapy ARDS of IL-10 transgenosis
CN110101874A (en) * 2018-10-27 2019-08-09 广州呼吸健康研究院 A method of utilizing the stem-cell therapy ARDS of HGF transgenosis
WO2021226602A1 (en) * 2020-05-08 2021-11-11 Celularity Inc. Placenta-derived adherent (pda) stem cell for the treatment of adults with sars-cov-2 related acute respiratory failure and ards (covid-19)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10386368B2 (en) 2017-02-24 2019-08-20 Trustees Of Boston University Isolation of human lung progenitors derived from pluripotent stem cells
EP3593809A4 (en) * 2017-03-08 2021-01-06 Rohto Pharmaceutical Co., Ltd. Ror1-positive mesenchymal stem cell-containing pharmaceutical composition for preventing or treating disease associated with fibrosis, method for preparing same, and method for preventing or treating disease associated with fibrosis using ror1-positive mesenchymal stem cells
SG11202011927TA (en) * 2018-06-05 2020-12-30 Medipost Co Ltd Pharmaceutical composition comprising mesenchymal stem cells as effective ingredient for prevention or treatment of inflammatory disease
IT201800020722A1 (en) 2018-12-21 2020-06-21 Assunta Borzacchiello Biomaterial and its use in the treatment of lung diseases
CN111793596A (en) * 2019-12-06 2020-10-20 上海赛傲生物技术有限公司 Separation method and preparation method of human amniotic epithelial stem cells
CN113679741B (en) * 2021-09-13 2023-09-19 北京大学第一医院 Application of human amniotic epithelial stem cells in preparation of medicines for treating cisplatin-induced acute kidney injury

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040229352A1 (en) * 1999-12-07 2004-11-18 David Warburton Lung stem cells and lung regeneration
WO2005017117A2 (en) * 2003-08-14 2005-02-24 Martin Haas Multipotent amniotic fetal stem cells (mafsc) and banking of same
WO2007141657A2 (en) * 2006-03-07 2007-12-13 Geeta Shroff Compositions comprising human embryonic stem cells and their derivatives, methods of use, and methods of preparation

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000073421A2 (en) * 1999-06-02 2000-12-07 Lifebank Services, L.L.C. Methods of isolation, cryopreservation, and therapeutic use of human amniotic epithelial cells
EP2316918B1 (en) * 2001-02-14 2015-07-01 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
JP2002326943A (en) * 2001-02-28 2002-11-15 Japan Tissue Engineering:Kk Antiinflammatory agent
US8021876B2 (en) * 2001-11-15 2011-09-20 Children's Medical Center Corporation Methods of isolation, expansion and differentiation of fetal stem cells from chorionic villus, amniotic fluid, and placenta and therapeutic uses thereof
AUPS112802A0 (en) * 2002-03-15 2002-04-18 Monash University Methods of inducing differentiation of stem cells into a specific cell lineage
US20030235563A1 (en) * 2002-04-19 2003-12-25 Strom Stephen C. Placental derived stem cells and uses thereof
US20040161419A1 (en) * 2002-04-19 2004-08-19 Strom Stephen C. Placental stem cells and uses thereof
WO2008003042A2 (en) * 2006-06-28 2008-01-03 The University Of Medicine And Dentistry Of New Jersey Amnion-derived stem cells and uses thereof
DE202006010151U1 (en) * 2006-06-29 2006-10-05 Arcos Die Haarprofis Handels-Gmbh Hair extension set with securing device has fixing arrangement with adhesive surface adjacent to recess on outside of flat material and section of self-adhesive hair band for sticking onto hairs pressed onto adhesive surface
WO2008036374A2 (en) * 2006-09-21 2008-03-27 Medistem Laboratories, Inc. Allogeneic stem cell transplants in non-conditioned recipients

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040229352A1 (en) * 1999-12-07 2004-11-18 David Warburton Lung stem cells and lung regeneration
WO2005017117A2 (en) * 2003-08-14 2005-02-24 Martin Haas Multipotent amniotic fetal stem cells (mafsc) and banking of same
WO2007141657A2 (en) * 2006-03-07 2007-12-13 Geeta Shroff Compositions comprising human embryonic stem cells and their derivatives, methods of use, and methods of preparation

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
BAILO M. ET AL.: "Engraftment Potential of Human Amnion and Chorion Cells Derived from Term Placenta", TRANSPLANTATION, vol. 78, no. 10, 2004, pages 1439 - 1448, XP002568927 *
DELPLANQUE ET AL.: "Epithelial stem cell-mediated development of the human respiratory mucosa in SCID mice", JOURNAL OF CELL SCIENCE, vol. 113, 2000, pages 767 - 778, XP008125670 *
KIM C.F. ET AL.: "Identification of Bronchioalveolar Stem Cells in Normal Lung and Lung Cancer", JOURNAL OF CELL, vol. 121, 17 June 2005 (2005-06-17), pages 823 - 835, XP008131911 *
MIKI T. ET AL.: "Stem Cell Characteristics of Amniotic Epithelial Cells", STEM CELLS EXPRESS, 4 August 2005 (2005-08-04), XP002410842 *
PAROLINI O. AND SONCINI M.: "Human Placenta: a source of Progenitor/Stem Cells", JOURNAL OF REPRODUCTIVE MEDICAL ENDOCRINOLOGY, vol. 3, no. 2, 2008, pages 117 - 126, XP008131909 *
PAROLINI O. ET AL.: "Concise Review: Isolation and Characterization of Cells from Human Term Placenta: Outcome of the First International Workshop on Placenta Derived Stem Cells", STEM CELLS, vol. 26, 2008, pages 300 - 311, XP002567088 *
See also references of EP2164505A4 *

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10104880B2 (en) 2008-08-20 2018-10-23 Celularity, Inc. Cell composition and methods of making the same
EP2375907A1 (en) * 2008-11-21 2011-10-19 Anthrogenesis Corporation Treatment of diseases, disorders or conditions of the lung using placental cells
EP2375907A4 (en) * 2008-11-21 2014-11-05 Anthrogenesis Corp Treatment of diseases, disorders or conditions of the lung using placental cells
EP2374871A4 (en) * 2008-12-17 2013-02-20 Beijing Health & Biotech H & B Co Ltd Pluripotent stem cells, method for preparation thereof and uses thereof
EP2374871A1 (en) * 2008-12-17 2011-10-12 Beijing Health & Biotech (H&B) Co., Ltd Pluripotent stem cells, method for preparation thereof and uses thereof
US20130302294A1 (en) * 2009-02-04 2013-11-14 Yale University Tissue Engineering of Lung
US11648280B2 (en) 2009-02-04 2023-05-16 Yale University Tissue engineering of lung
US10188683B2 (en) 2009-02-04 2019-01-29 Yale University Tissue engineering of lung
US10273450B2 (en) 2012-01-13 2019-04-30 The General Hospital Corporation Isolated human lung progenitor cells and uses thereof
US11987807B2 (en) 2012-01-13 2024-05-21 The General Hospital Corporation Isolated human lung progenitor cells and uses thereof
US9828583B2 (en) 2012-01-13 2017-11-28 The General Hospital Corporation Isolated human lung progenitor cells and uses thereof
US11214769B2 (en) 2012-01-13 2022-01-04 The General Hospital Corporation Isolated human lung progenitor cells and uses thereof
CN108025027B (en) * 2015-06-12 2022-05-24 哈德逊医学研究中心 A method of treatment
EP3307286A4 (en) * 2015-06-12 2018-12-26 Hudson Institute of Medical Research A method of treatment
CN108025027A (en) * 2015-06-12 2018-05-11 哈德逊医学研究中心 A kind for the treatment of method
EP4218780A1 (en) * 2015-06-12 2023-08-02 Hudson Institute of Medical Research Allogeneic mammalian amniotic exosomes for treating diseases
WO2016197196A1 (en) 2015-06-12 2016-12-15 Prince Henry's Institute Of Medical Research Trading As The Hudson Institute Of Medical Research A method of treatment
US11123376B2 (en) 2015-06-12 2021-09-21 Monash University Method of treatment
CN114984054A (en) * 2015-06-12 2022-09-02 哈德逊医学研究中心 A method of treatment
AU2016275566B2 (en) * 2015-06-12 2022-02-24 Hudson Institute of Medical Research A method of treatment
CN109843307A (en) * 2016-08-04 2019-06-04 哈德逊医学研究所 The method for the treatment of
WO2018023170A1 (en) * 2016-08-04 2018-02-08 Hudson Institute of Medical Research A method of treatment
CN110101874A (en) * 2018-10-27 2019-08-09 广州呼吸健康研究院 A method of utilizing the stem-cell therapy ARDS of HGF transgenosis
CN110101873A (en) * 2018-10-27 2019-08-09 广州呼吸健康研究院 A method of utilizing the stem-cell therapy ARDS of IL-10 transgenosis
WO2021226602A1 (en) * 2020-05-08 2021-11-11 Celularity Inc. Placenta-derived adherent (pda) stem cell for the treatment of adults with sars-cov-2 related acute respiratory failure and ards (covid-19)

Also Published As

Publication number Publication date
US20100266552A1 (en) 2010-10-21
EP2164505A4 (en) 2010-12-22
JP5567476B2 (en) 2014-08-06
AU2008255634A1 (en) 2008-12-04
EP2164505B1 (en) 2014-09-17
EP2164505A1 (en) 2010-03-24
AU2008255634B2 (en) 2014-06-05
JP2010528055A (en) 2010-08-19

Similar Documents

Publication Publication Date Title
AU2008255634B2 (en) Treatment of chronic lung disease
JP2022141809A (en) Treatment of stroke using isolated placental cells
CA2677397C (en) Treatment of inflammatory diseases using placental stem cells
EP2854825B1 (en) hUTC MODULATION OF PRO-INFLAMMATORY MEDIATORS OF LUNG AND PULMONARY DISEASES AND DISORDERS
US8221741B2 (en) Methods for modulating inflammatory and/or immune responses
Harrell et al. Therapeutic potential of amniotic fluid derived mesenchymal stem cells based on their differentiation capacity and immunomodulatory properties
CN113559126A (en) Treatment of pain using placental stem cells
JP7168653B2 (en) Composition for treating thyroid eye disease containing mesenchymal stem cells
TW201632620A (en) Treatment of retinal degeneration using progenitor cells
WO2013082487A1 (en) Afsc based therapies
US20210230537A1 (en) Angiogenesis using stimulated placental stem cells
Zhao et al. Human umbilical cord mesenchymal stem cell-derived exosomes inhibit ovarian granulosa cells inflammatory response through inhibition of NF-κB signaling in polycystic ovary syndrome
JP2022522357A (en) Extracellular vesicles derived from amniotic fluid and their use for wound healing
US20190388478A1 (en) Use of placental stem cells in treatment of acute kidney injury
Cabezas et al. In vitro preconditioning of equine adipose mesenchymal stem cells with prostaglandin E2, substance P and their combination changes the cellular protein secretomics and improves their immunomodulatory competence without compromising stemness
CA3168330A1 (en) Method for treating chronic graft versus host disease
US20190314424A1 (en) Treatment of lymphedema and related conditions using placental adherent cells
US9173908B2 (en) Methods for modulating immune responses
US20120315246A1 (en) Compositions and methods for modulating inflammatory and/or immune responses
US20180264048A1 (en) Treatment of diabetic peripheral neuropathy using placental cells
Ding et al. Human Wharton's jelly‐derived mesenchymal stem cells prevent pregnancy loss in a rat by JAK/STAT‐mediated immunomodulation
JP2024516172A (en) Methods for treating acute respiratory distress syndrome (ARDS) using mesenchymal precursors or stem cells
WO2020093050A2 (en) Treatment of cachexia using fibroblast cells and products thereof

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: 08748014

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2008255634

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2008748014

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2010509623

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2008255634

Country of ref document: AU

Date of ref document: 20080528

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 12601570

Country of ref document: US