US20220193141A1 - Compositions comprising stem cells expressing mesenchymal and neuronal markers and uses thereof to treat neurological disease - Google Patents

Compositions comprising stem cells expressing mesenchymal and neuronal markers and uses thereof to treat neurological disease Download PDF

Info

Publication number
US20220193141A1
US20220193141A1 US17/562,797 US202117562797A US2022193141A1 US 20220193141 A1 US20220193141 A1 US 20220193141A1 US 202117562797 A US202117562797 A US 202117562797A US 2022193141 A1 US2022193141 A1 US 2022193141A1
Authority
US
United States
Prior art keywords
cells
stem cells
hidpsc
cell
idpscs
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US17/562,797
Inventor
Irina Kerkis
Alexandre Kerkis
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Fundacao Butantan
Avita International Ltd
Original Assignee
Fundacao Butantan
Avita International Ltd
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 US14/214,016 external-priority patent/US9790468B2/en
Application filed by Fundacao Butantan, Avita International Ltd filed Critical Fundacao Butantan
Priority to US17/562,797 priority Critical patent/US20220193141A1/en
Publication of US20220193141A1 publication Critical patent/US20220193141A1/en
Abandoned legal-status Critical Current

Links

Images

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
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0664Dental pulp stem cells, Dental follicle stem cells
    • 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
    • A61K35/30Nerves; Brain; Eyes; Corneal cells; Cerebrospinal fluid; Neuronal stem cells; Neuronal precursor cells; Glial cells; Oligodendrocytes; Schwann cells; Astroglia; Astrocytes; Choroid plexus; Spinal cord tissue
    • 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
    • A61K35/32Bones; Osteocytes; Osteoblasts; Tendons; Tenocytes; Teeth; Odontoblasts; Cartilage; Chondrocytes; Synovial membrane
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • 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
    • C12N5/0618Cells of the nervous system
    • C12N5/0619Neurons
    • 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
    • C12N5/0618Cells of the nervous system
    • C12N5/0621Eye cells, e.g. cornea, iris pigmented cells
    • 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
    • C12N5/0618Cells of the nervous system
    • C12N5/0623Stem cells
    • 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
    • C12N5/0676Pancreatic cells
    • C12N5/0678Stem cells; Progenitor cells; Precursor cells
    • 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
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/05Inorganic components
    • C12N2500/10Metals; Metal chelators
    • C12N2500/20Transition metals
    • C12N2500/24Iron; Fe chelators; Transferrin
    • C12N2500/25Insulin-transferrin; Insulin-transferrin-selenium
    • 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
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/38Vitamins
    • 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
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/70Undefined extracts
    • C12N2500/80Undefined extracts from animals
    • C12N2500/84Undefined extracts from animals from mammals
    • 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
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/90Serum-free medium, which may still contain naturally-sourced components
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/385Hormones with nuclear receptors of the family of the retinoic acid recptor, e.g. RAR, RXR; Peroxisome proliferator-activated receptor [PPAR]
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/39Steroid hormones
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/39Steroid hormones
    • C12N2501/392Sexual steroids
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/395Thyroid hormones
    • 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
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/13Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells
    • C12N2506/1346Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells from mesenchymal stem cells
    • C12N2506/1361Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells from mesenchymal stem cells from dental pulp or dental follicle stem cells
    • 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
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/70Polysaccharides
    • C12N2533/76Agarose, agar-agar

Definitions

  • This application relates to a composition of stem cells for the treatment of neurological diseases suitable for systemic administration.
  • Stem cell-based therapies are important in order to reconstruct morphological design and functional ability of neural tissue in damaged brain areas in patients. These therapies used to have a dual role: transplanted stem cells paracrine action (anti-apoptotic, anti-inflammatory, anti-scar, anti-bacterial and angiogenic actions), which stimulates local cell survival, inhibits inflammation and brain tissue regeneration through the production of bioactive molecules acting in favor of new neurons production from the intrinsic and likely from donor stem cells.
  • the brain-derived neurotrophic factor is a gene responsible for BDNF protein expression found in the brain and spinal cord. This protein promotes the survival of nerve cells (neurons) by playing a role in the growth, maturation (differentiation), and maintenance of these cells. In the brain, the BDNF protein is active at the connections between nerve cells (synapses) where cell-to-cell communication occurs.
  • the BDNF protein helps regulate synaptic plasticity, which is important for learning and memory and is found to be expressed in regions of the brain that control eating, drinking, and body weight. Thus, BDNF has additional action in modulating all these functions.
  • synaptic dysfunction is a key pathophysiological hallmark in neurodegenerative disorders, including Alzheimer's disease.
  • BDNF neurodegenerative diseases
  • BDNF neurodegenerative diseases
  • Overdoses of BDNF could induce tumor formation in the brain; on the other hand low BDNF doses could not provide an efficient treatment.
  • Stem cells after transplantation are under the control of the patient biology, which can modulate BDNF secretion by the cells efficiently for each patient.
  • NSC transplanted nerve stem cells
  • NSC is generally difficult to access and cannot be obtained in sufficient therapeutic quantities to be applied in stem cell therapy through intravenous (IV) injection.
  • IV intravenous
  • two strategies are used to increase BDNF secretion.
  • this strategy has great limitations due to the fact that stem cells produce this factor only under in vitro conditions. Consequently, when such cells are transplanted to a patient they rapidly spend the “stock” of BDNF, which prevents long term treatment of neurodegenerative disease.
  • Another approach is to produce genetically manipulated stem cells which are suitably modified to overexpress BDNF. It is important to note that even NSC need to be genetically engineered to produce therapeutically sufficient levels of BDNF.
  • gene modification has its roots in gene therapy—an approach that still has to be proven. Therefore, there is a great need for new cell types and cell culture methods which can lead to stem cells with elevated secretion of BDNF.
  • the subventricular zone is the unique brain area where new neurons are produced throughout life (Altman J and Das G D 1965) and in generating cells to function in repair through adulthood. Blood vessels immediately subjacent to the SVZ run parallel to the direction of tangential neuroblast migration, and guide migratory neuroblasts via BDNF signaling. It is now understood that the organization of the SVZ in the adult human brain differs significantly from that of any other studied vertebrates. Specifically, this region in the adult human brain contains a unique tape of astrocytes that proliferate in vivo and can function as NSC in vitro. Astrocytes in the central nervous system perform many important and diverse functions.
  • the neuro-vascular unit which is composed of a neuron, an astrocyte and a blood vessel.
  • Astrocyte processes extend to and interact with blood vessels.
  • Astrocytic endfeet are in intimate contact with the basal lamina that is a component of the vessel wall and together with endothelial cells they form the blood-brain barrier (BBB).
  • BBB blood-brain barrier
  • Dopamine (DA) is a major neurogenesis factor in the adult SVZ (Baker et al., 2004).
  • the proximity of the SVZ with the striatum makes it a neurodegeneration therapy target for striatum-neurodegeneration associated disorders such as Huntington's disease (HD) and Parkinson's disease (PD).
  • HD Huntington's disease
  • PD Parkinson's disease
  • Both pathologies are characterized by different clinical symptoms of motor dysfunction, and both are thought to involve the SVZ—striatum DA micro-circuitry path through different mechanisms.
  • the disease-generated DA innervation that occurs in HD is a natural protective feedback mechanism to compensate for the striatal internal neurons degeneration pathology caused by inherent genetic mutation (Parent M et al., 2013).
  • Dysregulation of DA receptor D2 is a sensitive measure for Huntington disease pathology in model mice (Crook et al., 2012; Chen et al., 2013).
  • PD is associated with massive degeneration of DA neurons, due to impaired neurogenesis in the nigrostriatal area and is a major cause of the pathology (Hoglinger et. al., 2004).
  • CI chronic inflammation
  • Alzheimer's disease amyloid beta and tau
  • Parkinson's disease alpha synuclein
  • Huntington's disease Huntington's disease
  • AD Alzheimer disease
  • AD chronic inflammation
  • a history of head injury and systemic infections are factors, which typically cause brain inflammation and are known to be risk factors for AD.
  • Excessive action of the brain's immune cells, which are glial cells is another hallmark of Alzheimer's disease.
  • inflammation is associated with injury and toxicity to neurons, the relationship among glial cells, neurons and amyloid plaques still remains unclear. Inflammatory mediators released by glial cells can be extremely toxic to neurons. Thus, they have been considered as mediators of neurodegeneration.
  • IL-12 and IL-23 Two closely related inflammation-promoting proteins, IL-12 and IL-23, are among those pumped out by microglia when the cells become immunologically active. The studies demonstrated that these proteins exist at elevated levels in the cerebrospinal fluid of AD patients. Blocking these inflammatory proteins in older Alzheimer's mice whose brains were already plaque-ridden reduced the levels of soluble, more toxic forms of amyloid beta and reversed the mice's cognitive deficits (Vom Berg et al., 2012; Griffin, 2013).
  • AD Alzheimer's
  • Aging may help trigger Alzheimer's by worsening common age-related problems with neurons, which become functionally deficient and lose their ability to transport and appropriately place proteins.
  • Inflammation worsens this problem by increasing the production of amyloid-beta in inflamed regions, stressing neurons, and hastening the age-related decline of their protein-transport and disposal systems.
  • Inflammation reactivates microglia into an inflammatory state and thus reduces their ability to clear up the brain (Swindell et al., 2013).
  • innate immune cell hyperactivity was detected through elevated IL-6 production in mutant mHTT expressing myeloid cells of the central (microglia) and peripheral innate immune system (monocytes and macrophages) both in HD patients and mouse models. It has also been reported that abnormally high levels of cytokines were present in the blood of people carrying the HD gene many years before the onset of symptoms (Bjorkqvist et al., 2008; Trager et al., 2014a, b). The composition of cytokines and levels of their expression, which can be measured in a blood of patients, could be useful to establish the need to initiate intervention for therapies as well as the timing of therapies.
  • Parkinson's disease is characterized by a slow and progressive degeneration of dopaminergic neurons in the substantia nigra.
  • Using animal models researchers have obtained consistent findings about involvement of both the peripheral and the central nervous system immune components in response to inflammation, which initiates an immune response in PD.
  • the presence of continuing and increasing pro-inflammatory mechanisms results in a process whereby cellular protective mechanisms are overcome and the more susceptible cells, such as the dopaminergic neurons, enter into cell death pathways, which leads to a series of events that are a crucial for the progression of PD (Doursout et al., 2013).
  • CRP C-reactive protein
  • MCP-1 monocyte chemotactic protein-1
  • Stem cells sources used for neuro-regeneration cell therapy include mesenchymal stem cells (MSC), neural progenitor cells (NP), human fetal neuronal stem cells (huNSC), and pluripotent stem cells (both embryonic (ESCs) and induced (iPSC)).
  • MSC mesenchymal stem cells
  • NP neural progenitor cells
  • huNSC human fetal neuronal stem cells
  • iPSC pluripotent stem cells
  • Most of the studies on cell therapy for neurological conditions used neuronal-like cells through major cellular manipulation and/or highly invasive methods of delivery. For example, WO 2008/132722 and US Patent Application Publication No.
  • 2013/0344041 disclose genetically manipulated stem cells to induce stem cell traits or to release neurotrophic factors; WO 2009/144718 and US Patent Applications Publication Nos. 2014/0335059 and 2014/0154222 disclose inducing the release of neurotrophic factors at levels higher than at non-induced stage via exposure to biological, natural or chemical compounds in culture; and other studies use immortalized cell line of fetal stem cells that express early markers of neuronal differentiation. In spite of the studies on stem cell therapy, no data have shown that stem cell therapy through intravenous (IV) injection can result in direct neurogenesis via BDNF secretion or D2 expression in brain compartments suffering from neurodegenerative disease.
  • IV intravenous
  • the invention is directed to a pharmaceutical composition comprising stem cell, wherein the stem cells comprising late harvest enriched from tissue of neural crest origin.
  • tissue of neural crest origin is dental pulp.
  • the stem cells enriched from tissue of neural crest origin are immature dental pulp stem cells (IDPSCs).
  • IDPSCs immature dental pulp stem cells
  • Early harvest stem cells enriched from tissue of neural crest origin comprise IDPSCs of the first fifteen or the first 25 harvest cycles whereas late harvest stem cells comprise IDPSCs from the sixty or later or the 26th or later harvest cycle.
  • the invention is directed to a pharmaceutical composition produced by a method comprising: extracting dental pulp (DP) from a tooth; culturing the DP in basal culture medium in a first container to establish a DP explant culture, wherein the DP explant culture is cultured without or with at least one extracellular matrix components selected from the group consisting of: fibronectin, collagen, laminin, vitronectin, polylysine, heparan sulfate proteoglycans, and enactin; mechanically transferring the DP to a second container to establish a second DP explant culture; repeating the step of mechanically transferring the DP until at least 15 DP explant cultures have been established; passaging the DP explant culture to produced a passaged DP culture; and combining the passaged DP culture of an early harvest population and an late harvest population to produce the pharmaceutical composition, wherein the early harvest population comprises passaged DP culture established from at least one of the first 15 DP explant cultures and the late harvest population
  • the culturing step occurs under hypoxic conditions.
  • the step of combining the passaged DP culture of the early harvest population and the late harvest population to produce the pharmaceutical composition comprises: simultaneously thawing the frozen stock of passaged DP cultures of the early harvest population and the late harvest population; and pooling the thawed passaged DP culture to produce the pharmaceutical composition.
  • culturing the DP in basal culture medium in the method of production persist for at least three days before the DP is mechanically transferred.
  • the method of production further comprise creating a frozen stock of the passaged DP culture.
  • the frozen stock of the passaged DP culture is created at the third passage of the DP explant culture.
  • the invention is directed to a pharmaceutical composition produced by a method comprising: extracting dental pulp (DP) from a tooth; culturing the DP in basal culture medium in a first container to establish a DP explant culture, wherein the stem cells comprising late harvest enriched from tissue of neural crest origin are double positive for CD44 and CD13.
  • the stem cells enriched from tissue of neural crest origin and double positive for CD44 and CD13 are immature dental pulp stem cells (IDPSCs).
  • the invention is directed to a pharmaceutical composition produced by a method comprising: extracting dental pulp (DP) from a tooth; culturing the DP in basal culture medium in a first container to establish a DP explant culture, wherein the stem cells comprising late harvest enriched from tissue of neural crest origin demonstrated increasing level of secretion of endogenous BDNF and/or other neurotrophic factors (NF3, NF4 and NF5), when compared to stem cells obtained from early harvest.
  • the stem cells enriched from tissue of neural crest origin and secreting high level of endogenous BDNF and/or other neurotrophic factors (NF3, NF4 and NF5) are immature dental pulp stem cells (IDPSCs).
  • the present invention relates to pharmaceutical compositions for systemic administration to a subject to treat a neurological condition.
  • the neurological disease or condition may be a neurodegenerative disease or condition, autism, schizophrenia, epilepsy, stroke, ischemia, a motor disorder, or a convulsive disorder.
  • Neurodegenerative disease or condition may be Parkinson's disease (PD), multiple sclerosis, epilepsy, amyotrophic lateral sclerosis (ALS), stroke, autoimmune encephalomyelitis, diabetic neuropathy, glaucomatous neuropathy, Alzheimer's disease, or Huntington's disease (HD).
  • PD Parkinson's disease
  • multiple sclerosis epilepsy
  • stroke autoimmune encephalomyelitis
  • diabetic neuropathy glaucomatous neuropathy
  • Alzheimer's disease or Huntington's disease (HD).
  • PD Parkinson's disease
  • ALS amyotrophic lateral sclerosis
  • HD Huntington's disease
  • the pharmaceutical compositions comprise stem cells from tissue of neural crest origin.
  • Tissue of the neural crest origin may be dental pulp but not limited by this example.
  • the pharmaceutical composition comprises stem cells expressing at least one safety markers selected from the group consisting of ATP-binding cassette sub-family G member 2 (ABCG2), p53, and inactive nanog.
  • Inactive nanog is expressed nanog localizing predominantly in the cytoplasm of the stem cell.
  • at least 75% of the stem cells express ABCG2, at least 75% of the stem cells express p53, or no more than 5% of the stem cells express inactive nanog.
  • Some pharmaceutical compositions may comprise stem cells that further express the safety marker SOX2. In some such embodiments, no more than 30% of the stem cells express SOX2.
  • the stem cells of the pharmaceutical composition may further secrete at least one marker selected from the group consisting of brain-derived neurotrophic factor (BDNF), neutrotrophin-3 (NT3), neutrotrophin-4 (NT4), neutrotrophin-5 (NT5), and p75.
  • BDNF brain-derived neurotrophic factor
  • NT3 neutrotrophin-3
  • NT4 neutrotrophin-4
  • NT5 neutrotrophin-5
  • p75 p75
  • the stem cells of the pharmaceutical composition express BDNF, NT3, NT4, NT5, and p75 (CD271).
  • the pharmaceutical composition comprises stem cells expressing at least one neuroepithelial stem cell marker selected from the group consisting of BDNF, NT3, NT4, NT5, and p75.
  • these pharmaceutical compositions comprise undifferentiated stem cells expressing BDNF, NT3, NT4, NT5, and p75.
  • These pharmaceutical compositions may further express at least one safety markers selected from the group consisting of ABCG2, inactive nanog, p53, and SOX2.
  • at least 75% of stem cells express the at least one marker when the at least one marker is ABCG2.
  • at least 75% of the stem cells express p53.
  • no more than 5% of the stem cells express inactive nanog.
  • no more than 30% of the stem cells express SOX2.
  • the stem cells of the pharmaceutical composition are immature dental pulp stem cells (IDPSCs).
  • IDPSCs immature dental pulp stem cells
  • the present invention also relates to methods of using the pharmaceutical composition comprising systemically administering to a subject the pharmaceutical compositions of the invention.
  • Such methods may be used for treating neurological diseases or conditions, for example by supporting the natural neuro-protective mechanism in subjects diagnosed with early HD or repairing lost DA neurons in subjects diagnosed with PD.
  • the methods may also be used as a preventive therapy for subjects at risk of HD.
  • the methods further comprise measuring the amount of DA receptor in the subject.
  • the DA receptor is receptor D2.
  • measuring the amount of DA receptor, for example receptor D2, in the subject comprises imaging the subject to detect presence of DA receptor.
  • the subject is administered a single administration or a first and second administrations of the pharmaceutical composition.
  • the repetitive (from 1 to six) administrations of the pharmaceutical composition take place at least seven days, at least 14 days, at least 21 days, or at least 30 days after the first administration, as well as one time per year, or twice per year.
  • administration of the pharmaceutical composition is through intravenous injection.
  • the administration of the pharmaceutical composition may be repeated annually, for example, repeated three times.
  • the neurological disease or condition is treated by the stem cells crossing the blood/brain barrier (BBB) and inducing neurogenesis.
  • BBB blood/brain barrier
  • the stem cells are directly transplanted into the brain parenchyma, including striatum, following crossing of the BBB.
  • the induced neurogenesis is dopamine-associated.
  • dopamine-associated neurogenesis occurs through self-differentiation of the stem cells or activation of migration and differentiation of intrinsic stem cells by the extrinsic stem cells.
  • massive dopamine-associated neurogenesis takes place in the subventricular zone (SVZ).
  • the pharmaceutical composition provides neuroprotection.
  • systemic neuroprotection is provided with the high basal level of neurotrophic and immunoprotective factors expression and release pattern of the stem cells of the pharmaceutical composition.
  • these stem cells of the pharmaceutical composition are IDPSCs.
  • the pharmaceutical composition comprises stem cells that are autologous to the subject.
  • the stem cells that are autologous to the subject are collected from the subject before disease manifestation from contingently “healthy cells”, once these diseases are genetic and age dependent.
  • the pharmaceutical composition comprises stem cells that are allogeneic to the subject regardless of whether the major histocompatibility complex expressed by the stem cells match the major histocompatibility complex expressed by subject.
  • the pharmaceutical composition may also comprise a combination of stem cells that are autologous to the subject and stem cells that are allogeneic to the subject.
  • FIG. 1 depicts a scheme for the manufacturing process of compositions of IDPSCS comprising both early and late population IDPSC suitable for the treatment of neurological diseases and conditions.
  • FIG. 2 depicts an early phase development process for CELLAVITATM (stem cells) isolation and batch formulation.
  • FIG. 3 depicts another early phase development manufacturing process for CELLAVITATM (stem cells).
  • FIG. 4 depicts a CELLAVITATM (stem cells) production process composed of several major steps.
  • FIG. 5 depicts immunophenotyping of hIDPSC from early and late harvests. Harvests 0-10 were defined as early harvests. All harvests after 10 were defined as late harvests.
  • FIG. 6 depicts IDPSC at late harvest (13) at passage 3 were immunopositive for P75 (CD271) (A, C, D), nestin (E-G), CD13 (H) and CD73 (I), and they did not react with CD146 (J) and HLA-ABC (K). Insets, demonstrate control for respective secondary antibodies.
  • FIG. 7 depicts in a) CFU-F assay performed in triplicate at T20, passage 3, demonstrating high clonogenic capacity of a LP population of IDPSCs.
  • LP of IDPSCs (batch #11) comprise approximately 80% cells that express BDNF and DARP 32 while EP is negative for these markers (data not shown) and comprises a very low number of the cells which express D2.
  • FIG. 8A depicts positive immunostaining for BrdU (B-J) in control cells with a secondary antibody.
  • FIGS. 8B and 8C depict quantification of LP IDPSC which react positively with an BrdU antibody.
  • (A) Epi-fliorescence
  • (C) FACS analysis. Magnification (A)—200 ⁇ .
  • (B, E and H) 400 ⁇ .
  • FIG. 9 depicts quantitative PCR for expression of endogenous Oct4, Nanog and Sox2 genes observed in hIDPSCs before (black color) and after reprogramming (white color) as well as in human embryonic stem cells (hESC) (striped).
  • FIG. 10 depicts quantification of expression of GFAP (glial fibrillary acidic protein) and beta-III-tubulin in EP (early population) and LP (late population) IDPSC with flow cytometry.
  • GFAP glial fibrillary acidic protein
  • beta-III-tubulin in EP (early population) and LP (late population) IDPSC with flow cytometry.
  • FIG. 11 depicts a time line of stability studies of hIDPSC during long term cryopreservation, following thawing and shipping before local of application in animal models of human diseases.
  • FIG. 12 depicts the timeline of the pilot study in the HD disease model (groups I, II, III, and IV).
  • HD was induced during the first four days, day 0 (D0) to day 4 (D4) by 3-NP.
  • D5 day 5
  • IDPSC transplantation was administered via intravenous injection. Animals were euthanized on day 9 (D9) followed by brain tissue fixation and histological analysis of lesion for detection of IDPSC biodistribution and engraftment (Vybrant+immunohistochemistry using specific antibodies).
  • Group I and III were euthanized on day 35 (D35) and groups II and IV on day 95 followed by brain tissues fixation and histological analysis of lesion for detection of IDPSC biodistribution and engraftment (Vybrant+immunohistochemistry using specific antibodies).
  • FIG. 13 presents localization of hIDPSC in rat brain tissue four days after hIDPSC administration.
  • hIDPSC cells were stained green (Vybrant) and nuclei were stained red (PI) (A1-A4).
  • Cells were localized mainly in capillaries and two morphological types were observed: neuron-like cells and pericytes. Note the different localization of pericytes in capillaries in A2, A3, and A4; in A4, hIDPSCs are localized in the axon bifurcation.
  • A5 neuron morphology in brain tissues (A2-A4);
  • DIC Digital interference contrast
  • FIG. 14 depicts the engraftment of hIDPSCs four days after IV administration.
  • Optical cut demonstrates hIDPSC stained with Vybrant (green) and positively reacted with anti-hIDPSC antibody (red). Superposition of both produces yellow color.
  • the cells demonstrate near capillary localization.
  • Two markers for MSC were used: CD73 and CD105 demonstrating positive reaction with hIDPSC (A-D).
  • FIG. 15 depicts immunohistochemical images showing positive anti-human nuclei (hNu) staining of hIDPSCs and their localization in rat brain tissue 30 days after hIDPSC administration. Note: a few hIDPSCs in the cortex (left) and a large number of hIDPSCs in the corpus striatum (right). Light microscopy. 90 ⁇ magnification. Scale bars: 5 ⁇ m and 25 ⁇ m, respectively.
  • hNu positive anti-human nuclei
  • FIG. 16 shows immunohistochemical image of rat brain tissue after 3-NP Injection and hIDPSC administration. Note: positive anti-human nuclei (hNu) immunostaining in cells. Neuron-like cells are circled in blue, and fibroblast-like cells are circled in white. Light microscopy, 90 ⁇ magnification.
  • hNu positive anti-human nuclei
  • FIG. 17 depicts Nissl Staining in the Striatum of Untreated Animals (3-NP+saline) (a-b1); Control Animals (no 3-NP or hIDPSC) (c, c1); and Treated Animals (3-NP+hIDPSC) (d-f1).
  • score 1 (a, a1, d, and d1); score 2 (b, b1, e, and e1); and score 3 (c, c1, f, and f1).
  • Area of extensive degeneration (a, a1); severe (d, d1), moderate (b, b1, e, and e1), mild (f, f1), and no (c, c1) neuron loss.
  • Insets in (b, c, e, and f) show typical Nissl-stained neuron morphology (40 ⁇ ).
  • Light microscopy (a-f).
  • FIG. 18 depicts DARPP32 immunostaining in the corpus striatum of untreated animals (3-NP+saline) (a-b), Controls (no 3-NP or hIDPSC) (c), and Treated Animals (3-NP+hIDPSC) (d-e1).
  • a no immunostaining (blue arrow, score 1)
  • score 2 few DARPP32+ cells
  • score 3 Control animals
  • d, d1 neuron loss in Treated animals (3-NP+hIDPSC), with few DARPP32-stained cells (score 2) (black arrow).
  • In (e, e1) strong anti-DARPP-32 immunostaining (score 3).
  • Insets (a, b, c, d1) show DARPP32+ neurons (black arrow) (40 ⁇ ).
  • HE hematoxylin and eosin-stained nuclei in blue. Magnification: 10 ⁇ (a, c, d, and e) and 20 ⁇ (d, e1).
  • FIG. 19 depicts neuronal growth in the striatum of rats after hIDPSC.
  • Administration of hIDPSC resulted in a neuroreparative effect in hIDPSC-treated animals by (A) Nissl staining and (B) DARPP32 expression.
  • C Number of animals showing neuron recovery after hIDPSC administration compared to Controls. Most hIDPSC-treated animals (3-NP+hIDPSC) had scores 3 and 2 (moderate and mild), whereas most Untreated animals (3 NP+saline) had scores 2 and 1 (severe and moderate).
  • FIG. 20 depicts positive DARPP32 immunostaining for neurons in CELLAVITATM (stem cells)-treated animals 30 days after hIDPSC transplantation. Untreated animals (3-NP+saline) showed no DARPP32 immunostaining in the striatum or cortex. Rat neuron production was observed in the cortex and striatum of hIDPSC-treated animals. Light microscopy. Magnification: 20 ⁇ and 90 ⁇ .
  • FIG. 21 shows Dopamine Receptor D2 expression in the striatum of rats before and 30 days after hIDPSC Administration; Scores 3 and 2: striatum of untreated animals (3-NP+saline), showing a few D2 receptors in score 2. Score 1: striatum of treated-animals (3-NP+hIDPSC), showing positive receptor D2 immunostaining. Inset: high magnification showing positive D2 immunostaining and neuron morphology.
  • FIG. 22 depicts BDNF expression in rat brain tissue after (a-f) 3-NP Injection and (g-j) CELLAVITATM (stem cells) administration (a, b) Absence of BDNF expression seven days after 3-NP injection and (c, d) low expression after 30 days (e, f). Control animals (no 3-NP or CELLAVITATM (stem cells)). BDNF expression 7 (g, h) and 30 (i, j) days after CELLAVITATM (stem cells) administration. Magnification: 10 ⁇ (a, c, e, f, g, and h) and 20 ⁇ (b, d, i, and j).
  • FIG. 23 depicts DARP32 expression in the striatum of rats 30 days after CELLAVITATM (stem cells) administration in a 3-NP model of HD. Confocal microscopy, overlapping images in A. Epifluorescence+Digital interference contrast (DIC) microscopy. B-D: Epifluorescence. Scale bar: 10 ⁇ m.
  • FIG. 24 depicts the effect of hIDPSC administration on body weight in treated (3-NP+hIDPSC) and untreated animals (3-NP+Saline). Body weight was recorded before and 4 days after 3-NP administration and after each hIDPSC administration (every 30 days). No increase in body weight was observed in 3-NP-treated animals 30, 60, and 90 days after 3-NP administration. Body weight in hIDPSC-treated animals (1 ⁇ 10 6 dose) increased after the first hIDPSC administration.
  • FIG. 25 depicts intracellular expression of Oct4, Nanog, and Sox2. Nuclei were stained with DAPI.
  • FIG. 26 depicts expression of ABCG2 and K3/12 in IDPSC grown on various types of cell culture media.
  • FIG. 27 depicts expression of Vimentin, ABCG2, and K3/12 in IDPSC grown on various types of cell culture media.
  • FIG. 28 depicts pharmacological efficacy studies of investigational product CELLAVITATM (stem cells).
  • FIG. 29 depicts safety studies of investigational product CELLAVITATM (stem cells).
  • FIG. 30 depicts a flow cytometry analysis of early phase (EP) and late phase (LP) hIDPSC. Changes in CD146 and CD13 expression were observed following in vitro DP harvesting and hIDPSC passing. For EP-hIDPSC ⁇ 33% of CD146 positive cells were observed, while for LP-hIDPSC less than 1% of the cells were positive for this marker. For EP-hIDPSC ⁇ 52% of CD13 positive cells were observed, while for LP-hIDPSC 95% of the cells were positive for this marker.
  • FIGS. 31A -A 4 depict expression of SOX1 and ⁇ -tubulin proteins in late population (LP) of hIDPSC derived neurons.
  • FIG. 31A Nucleus (white arrow) of neurons stained with DAPI;
  • FIG. 31 A 2 Positive immunostaining for SOX1 observed in the nucleus (white arrow) of neurons;
  • FIG. 31 A 3 Positive immunostaining for ⁇ -tubulin;
  • FIGS. 31B-E depict enrichment of LP of hIDPSC with neural progenitors and neurons following 15-, 30- and 45 cycles of dental pulp mechanical transfer and induction of neural differentiation.
  • FIG. 32A demonstrates a nucleus of IDPSC-neuroblasts positively immunostained with anti-BrdU antibody and neuronal bodies, which reacts positively with ⁇ -tubulin class III.
  • FIGS. 32B-C show the enrichment of differentiated IDPSC population with slightly differentiated neuroblasts following growing cycle numbers of DP mechanical transfer.
  • FIGS. 33A -A 3 depict enrichment of LP of hIDPSC with neural progenitors and neurons following 15-, 30- and 45 cycles of dental pulp mechanical transfer and induction of neural differentiation.
  • BrdU positive cells present red and SOX2 green nucleus, which are also stained with DAPI (blue).
  • FIGS. 33B-C demonstrate percentage of BrdU and SOX2 positive cells shown in relation with nucleus stained with DAPI (blue).
  • the term “high expression” in reference to the expression level (strongly immunopositive for antigen of interest) of a gene in a population of cells refers at least 75% of the population expressing the gene.
  • low expression in reference to the expression level of a gene in a population of cells refers no more than 30% of the population expressing the gene. In preferred embodiments, low expression refers no more than 25% of the population expressing the gene.
  • no expression in reference to the expression level of a gene in a population of cells refers no detectable cells that express the gene of interest in the population. No detectable expression includes an expression level that is within the realm of error for the method of measuring expression.
  • co-express refers to the simultaneous detection of two or more molecular markers, e.g., SOX1 and SOX2, and beta-3-tubulin in same cell population, preferably in/on same cell.
  • the term “subject” or “patient” refers to any vertebrate including, without limitation, humans and other primates (e.g., chimpanzees and other apes and monkey species), farm animals (e.g., cattle, sheep, pigs, goats and horses), domestic mammals (e.g., dogs and cats), laboratory animals (e.g., rodents such as mice, rats, and guinea pigs), and birds (e.g., domestic, wild and game birds such as chickens, turkeys and other gallinaceous birds, ducks, geese, and the like).
  • the subject may be a mammal. In other implementations, the subject may be a human.
  • stem cell refers immature, unspecialized cells that, under certain conditions, may differentiate into mature, functional cells.
  • neural stem cell or “NSC” refers to multipotent cells that self-renewable and able to terminally differentiate into neurons, astrocytes, and oligodendrocytes.
  • neural progenitor cells refer to undifferentiated cells further along the stage of cell differentiation than neural stem cells. Thus these cells are derived from neural stem cells and can produce progeny that are capable of differentiating into more than one cell type of central nervous system (CNS) and peripheral nervous system (PNS).
  • CNS central nervous system
  • PNS peripheral nervous system
  • neural precursor cell refers to a mixed population of cells consisting of neural stem cells and all of its undifferentiated progeny. Therefore NPCs include both NPC and NSC. The NPCs can also be categorized into neuronal NPCs and glial NPCs, which produce neurons and glial cells, respectively.
  • the term “harvest cycle” constitutes a transfer of the org ⁇ o (e.g. of neural crest origin like dental pulp) or tissue to a new cell culture container after adherence and outgrowth of the stems cells in the tissue followed by preservation (e.g. cryopreservation) and/or sub-culturing of the outgrowth of IDPSCs.
  • org ⁇ o e.g. of neural crest origin like dental pulp
  • tissue e.g. of tissue
  • preservation e.g. cryopreservation
  • sub-culturing of the outgrowth of IDPSCs e.g. cryopreservation
  • stem cells from human exfoliated deciduous teeth can not be divided in early and late population, as these cells are isolated using enzymatic method only once from dental pulp, which are descarted after SHED isolation. Thus, only one SHED cell population can be isolated. Further, following enzymatic digestion, SHED can be passed from one to other cell culture flask thus counting cell passages, which generally are performed when SHED reach semi-confluence.
  • sex determining region Y-box 1 refers to a transcription factor which is involved in early central nervous system development. SOX1 may be expressed particularly in the ventral striatum.
  • SOX2 refers to a neural progenitor and sternness marker that is a transcription factor expressed by self-renewing and multipotent stem cells of the embryonic neuroepithelium (9). SOX2 may be expressed by actively dividing neural progenitor cells in the neurogenic regions in the adult rat brain and may also be expressed by glial fibrillary acidic protein immunopositive astroglia, widely distributed in the brain parenchyma (10).
  • SOX2 is also known, in conjunction with Oct4, SOX2 as essential transcription factor for pluripotency (Ivanona et al., Nature 442:5330538 (2006); Methods of preparing induced pluripotent stem cells from mouse are also known (Takahashi and Yamanaka, 2006). Induction of iPS cells typically require the expression of or exposure to at least one member from Sox family and at least one member from Oct family.
  • class III ⁇ -tubulin As used herein, the terms “class III ⁇ -tubulin”, “beta-3-tubulin”, “microtubule-associated protein 2 (MAP-2), or neurofilament” refer to microtubule elements expressed exclusively in neurons and serves as a specific neuronal promoter, characteristic of neurons phenotypic marker of early neuronal precursors.
  • MAP-2 microtubule-associated protein 2
  • IDPSCs can be isolated as from explant culture of the dental pulp after the first adherence of DP to plastic and cells outgrowth—early population cells. At this stage dental pulp is not descarted and used for subsequent explant dental pulp cultures—late populations. Thus, IDPSC isolated from the second or later harvest cycle are late population cells. For example, IPDSCs that are isolated from the second harvest cycle or later are late population undifferentiated stem cells.
  • the term “early passage” refers to the cells from the first five passages of an explant culture.
  • late passage refers to the cells from passages after the fifth passage, e.g. in the sixth passage or later, of an explant culture.
  • the IDPSC may be from an early or late population and additionally categorized as an early or late passage.
  • the present invention relates to the discovery that a particular composition of IDPSC, the unique stem cells population, which composed by early and late stem cells populations, from tissue of neural crest origin can cross the blood/brain barrier (BBB) and induce neurogenesis.
  • Tissue of neural crest origin include, for example, dental, periodontal, and hair follicular tissue.
  • Hair follicular tissue includes follicular tissue of the vibrissa.
  • the hIDPSC was evaluated in the 3-NP (three nitropropionic acid) HD rat model.
  • the hIDPSC showed engraftment into the rat brain after 1 month following intravenous injection of 1 ⁇ 10 6 and 1 ⁇ 10 7 cell/transplant (3 ⁇ 10 6 cell/kg and 3 ⁇ 10 7 cell/kg) labeled with fluorescent protein (Vibrant), as well as, following immunohistochemistry analysis using specific anti-human antibody. Cell engraftment was observed in different brain compartments (cortex, striatum and Subventricular zone-SVZ).
  • the ability to cross the BBB enables systemic administration (e.g. IV administration) of stem cell therapy to treat neurological conditions, which provides a significant advantage over more localized methods of administration.
  • systemically administration being less invasive, leaving stem cells to migrate to locations that require aid reduces the risk of harmful cell masses developing at the site of administration.
  • intrathecal (IT) administration of stem cell therapy is commonly contemplated in preclinical and clinical studies, this method can have significant risk when the stem cells are MSCs.
  • the composition of IDPSC may be in the form of a pharmaceutical composition comprising stem cells expressing a mesenchymal and neuroepithelial stem cell immunophenotypes.
  • expressing a mesenchymal and a neuroepithelial stem cell molecular profile is the expression of markers of MSC and neuroepithelial cells/progenitor cells and genes encoding neuro-protective and immuno-protective factors.
  • Cells expressing a MSC immunophenotype include expression of CD44.
  • Prior animal models of multiple sclerosis found that NCS adhesion to inflamed endothelial cells and then trans-endothelial migration across the BBB into the inflamed CNS areas are sequentially mediated by the constitutive expression of functional cell adhesion molecules (CAM), especially CD44 (Rampon C et al., 2008).
  • CAM functional cell adhesion molecules
  • CD44 functional cell adhesion molecules
  • Perycites are known to play a critical role in the integration of endothelial and astrocyte functions at the neurovascular unit, and in the regulation of the BBB (Armulik et al., 2010; Liu et al., 2012).
  • MSC mesenchymal stem cells
  • CD44 is a ligand of E and L blood vessel endothelial selectins (Dimitroff, et al., 2001).
  • MSC present similar homing mechanisms as leukocytes.
  • the first step of leukocyte migration involves capture of leukocytes flowing freely in the blood stream, mediated by glycoproteins known as selectins.
  • P- and E-selectins are expressed by the vascular endothelium and are the principal mediators for the rolling response in leukocyte migration through blood vessels (Luster et al., 2005).
  • MSC may use this or similar mechanisms to engraft in several organs (Sackstein et al., 2008) such as the brain.
  • CD44 is considered a pivotal factor for MSC migration into the brain.
  • hIDPSC express CD44, which suggests that CD44 is also involved in hIDPSC migration towards several organs (Barros et al., 2014, Castanheira et al., 2013) including the brain after intravenous administration.
  • hIDPSC express also CD13, (aminopeptidase N) (Kerkis et al., 2006; Kerkis and Caplan, 2012), which is multifunctional protein and plays varying roles in cell migration, cell proliferation, cell differentiation (Taylor et al., 1993; Mina-Osorio et al., 2008a/b).
  • CD13 participates in angiogenesis generating and modulating angiogenic signals, in the process of capillary tube formation, and as a marker of angiogenic vessels (Bhagwat et al., 2001). This suggests its possible role in hIDPSC capacity to migrate and to target brain vasculature. In the 3-NP rat study, hIDPSC demonstrated tight association with brain capillaries.
  • the IDPSC lack expression of CD146, HLA-DR, and/or HLA-ABC. Lack of expression of these markers facilitates the use of hIDPSC as a safe, heterologous therapy.
  • the endothelium plays an important role in the exchange of molecules, but also of immune cells between blood and the underlying tissue.
  • S-Endo 1 antigen CD146
  • CD146 is preferentially located at endothelial junctions and has been claimed to support endothelial integrity.
  • MCAM CD146 is expressed in T cells (3%) in the peripheral circulation of healthy individuals.
  • MCAM positive T cells demonstrate an increased ability to bind to endothelial monolayers and these cells could represent early components of the adaptive immune response (Dagur et al., 2015). Therefore, stem cells, which express this marker may bind to BBB and not cross BBB, as well as being immune reactive.
  • the mesenchymal stem cell genotype pattern also includes low expression of pluripotent markers OCT3/4 and nanog.
  • the undifferentiated stem cells of pharmaceutical composition of the invention need not express c-Myc, KLf-4, and REX-1. In fact, these stem cells may be negative for c-Myc, KLf-4, and REX-1.
  • Stem cells expressing a neuroepithelial stem cell molecular profile express at least one, preferably two, more preferably more than two NPC- and NSC-biomarkers selected from the group consisting of vimentin, nestin, SOX2, p75, and other neurotrophic factors essential for neural cells development and survival.
  • p75 is a neurotrophic receptor marker.
  • BDNF brain-derived neurotrophic factor
  • BDNF brain-derived neurotrophic factor
  • GNDF glial cell line-derived neurotrophic factor
  • NGF nerve growth factor
  • NTs neurotrophins
  • BNDF plays a critical role in Huntington's disease (Gauthier et al.; Strand et al.) and Parkinson's disease (Mogi et al.), both of which are dopamine-associated neurodegenerative diseases.
  • NTs essential for neuronal development and survival include NT3, NT4, or NT5.
  • NT4 and NT5 are known to promote sensory and motor axon growth.
  • the stem cells comprise cells autologous to a subject in need of the pharmaceutical composition. In other implementations, the stem cells comprise cells allogeneic to the subject in need of the pharmaceutical composition. In some implementations, stem cells comprise a combination of cells autologous to and allogeneic to the subject in need of the pharmaceutical composition.
  • the composition of stem cells is isolated from tissue of neural crest origin selected from the group consisting of dental tissue, periodontal tissue, and hair follicular tissue.
  • tissue of neural crest origin is dental pulp.
  • the stem cells are from immature dental pulp, for example, human immature dental pulp stem cells (IDPSCs) as disclosed in International Application no. PCT/IB14/59850 and U.S. patent application Ser. No. 14/214,016. IDPSCs carry multiple neuronal markers and undergo robust differentiation into neurons.
  • IDPSCs human immature dental pulp stem cells
  • IDPSC immunophenotype is unexpected expression of these markers and at the same time markers typical for MSC, presenting immunophenotype in accordance with the International Society for Cellular Therapy's minimal criteria for defining multipotent mesenchymal stromal cells (Dominici et al., 2006).
  • This combination of expression by IDPSC of MSC and multiple neuronal markers is not typical for MSCs (Dominici et al., 2006) and which has not been disclosed for MSCs.
  • compositions contemplated in the invention are preferably isotonic.
  • the population of immature stem cells should be between 10 4 -10 10 cells per injection, for example, 10 4 , 10 5 , 10 6 and 10 7 cells per kg of body weight.
  • the pharmaceutical composition comprising a population of stem cells may be used in adjunction to other pharmaceutically active compounds or modalities.
  • the pharmaceutical composition may further comprise another pharmaceutically active compound or therapeutic modality.
  • MSCs are found in bone marrow, umbilical cord tissue, dental pulp and fat pads.
  • bone marrow MSCs are relatively rare, comprising only one out of every 10,000 cells, while other sources are significantly richer in these cells.
  • MSCs are responsible for tissue regeneration in cases of disease, trauma or injury throughout human life. This function of MSCs is mediated by their capacities for self-renewal and plasticity (the capacity for differentiation—production of diverse cell types).
  • MSCs can be isolated from aforementioned tissues and cultured easily in the laboratory. After obtaining a limited number of the cells from a patient, MSCs can be multiplied rapidly in vitro and cryopreserved for the future clinical applications.
  • MSCs are able to secrete a variety of bioactive molecules, such as cytokines, which provide “trophic activities” by structuring a regenerative microenvironment, and other molecules that contribute to immunomodulatory cell functions and even to transfer products as large as mitochondria to damaged cells that need help.
  • cytokines which provide “trophic activities” by structuring a regenerative microenvironment
  • MSCs in response to chemotactic stimuli can migrate to the focal injury from both local and surrounding sites. Additionally, MSCs can act to reduce chronic inflammation, to inhibit apoptosis, to provide the appearance of myofibroblasts, to inhibit scar formation and to stimulate the mitosis of tissue-intrinsic progenitors, thus remodeling damaged tissue.
  • MSCs are also called “Medicinal Signaling Cells.” They stimulate angiogenesis, the process of new blood vessel formation, which is closely linked to neurogenesis, the process by which new nerve cells are produced. Blood vessels play an important role as a framework for neuronal progenitor cells migration toward the damaged brain region The factors secreted by MSCs also reduce the destructive effects of oxidative harm. Using all these mechanisms of action MSCs can significantly improve lesioned microenvironment that leads to restoration of the damaged cells. Therefore, MSCs are believed to be “cellular paramedics”.
  • MSCs obtained from humans were labeled, in order to track them, and injected into mice that had some type of tissue damage, they migrated throughout the damaged tissues apparently evenly. These cells can or not to be present in the tissue for a substantial period of time, which depends on disease model. The continued presence of MSCs is important, but not essential, to therapeutic development because it indicates that potential positive long-term effects of a treatment might be capable of persisting.
  • MSCs suppress the immune system and reduce inflammation.
  • MSCs can be transferred between organisms demonstrating very low immune rejection, which occurs when the immune system of the organism attacks the foreign tissue, receiving the transplant. This finding makes MSCs good candidates for transplantation or injection into a host because they can avoid rejection by the host's immune system (Le Blank, Ringén, 2006; English, 2012; Miguel et al., 2012; Griffin et al., 2012; Ankrum et al., 2014).
  • the crucial question of cellular therapies is a route of MSCs delivery into the brain, which has been approached in a number of different ways.
  • Several approaches have been proposed to deliver MSCs into the brain such as, intrathecal, intravenous, an injection into the space surrounding the spinal cord and even a route through the nose.
  • the blood/brain barrier (BBB) is formed and it controls selective molecular or cells trafficking between the bloodstream and brain interstitial space.
  • the BBB present significant problems for the delivery of therapeutic agents (drugs or cells) for treating brain malignancies and neurodegenerative disorders.
  • Systemically-infused MSCs may treat acute injuries, inflammatory diseases, stroke of the central nervous system (CNS) and even brain tumors because of their regenerative capacity and ability to secrete trophic, immune modulatory, or other engineered therapeutic factors.
  • CNS central nervous system
  • MSCs possess the ability to migrate across the BBB in normal and pathological conditions remains unresolved (Liu et al., 2013).
  • MSCs can support repair neurodegeneration by secreting trophic factors, which are proteins that stimulates differentiation and survival of cells.
  • trophic factors proteins that stimulates differentiation and survival of cells.
  • the effects of these factors allow nerve cells to carry out several processes that can support survival: axon extension, growth, and cells attachment.
  • the number of cells used in these experiments varied from 10 5 , 2 ⁇ 10 5 , 4 ⁇ 10 5 , 5 ⁇ 10 5 , and up to 10 6 per hemisphere/striatum.
  • the time of administering MSCs transplantation varied significantly across the studies with the time being 1-3 days, 2-4 weeks, and 8 weeks. These cells were found in the brain after direct grafting, but direct intrabrain delivery is a highly invasive and risky procedure. Thus these studies have not demonstrated that minimally invasion methods of administering stem cell therapy, such as systemic administration by IV injection, could be used.
  • the present invention provides a method of treating neurological diseases and condition that uses a unique population of IDPSC having the immunophenotype typical for MSCs as defined by the International Society for Cellular Therapy effective even with a minimally invasive administration, for example through classic IV route of delivery.
  • Stem cell therapy is inevitable since intracellular and cellular mechanisms are involved into HD phenotype. Stem cell therapy may also accelerate the process of brain tissue regeneration. Stem cells are an important therapy, which will help to rebuild an area of the brain that was most damaged in HD. Only drugs approach will not be able to reconstruct damaged brain areas especially in late stages of HD.
  • MSCs may be obtained from extracted human teeth, both permanent and deciduous, by enzymatic digestion (Gronthos et al. 2000; Miura et al., 2003), or by organ culture followed by explant (immature dental pulp stem cells, IDPSCs) technology as disclosed in International Application no. PCT/IB14/59850 and U.S. patent application Ser. No. 14/214,016.
  • the IDPSCs are obtained from dental pulp tissue, which anatomically originated from ectomesenchymal tissue, more precisely from neural crest, which is a mass of tissue present in the early formation of an embryo. It eventually forms the hard and soft tissues of the neck and cranium.
  • IDPSCs which are of neural crest origin, are known to migrate pre-natally into various, mainly ectodermal tissues and have the capacity to self-renewal and display a developmental potential almost the same as embryonic stem (ES) cells, but without risk of formation of embryonic bodies in vitro and teratomas in vivo (Kerkis and Caplan, 2012).
  • the postmigratory stem cells of neural crest origin generate all craniofacial bones, the majority of cells and tissues of the central and peripheral nervous systems, as well as several non-neural cell types, such as smooth muscle cells of the cardiovascular system, pigment cells in the skin, cartilage, connective tissue, corneal epithelium and dental pulp among them.
  • postmigratory postnatal stem cells of neural crest origin are of restricted developmental potential, they maintain functional characteristics resembling their embryonic counterparts and an ability to differentiate into a broad spectrum of cell types (Le Douarin et al., 2004, 2007, 2008; Dupin et al., 2007; Le Douarin & Dupin, 2003, 2012).
  • IDPSCs In vitro IDPSCs undergo uniform differentiation into neurons and glial cells. In vivo transplantation of human IDPSCs showed dense engraftment in various tissues, including neurons. Neuronal fate differentiation is based upon epigenetic “memory” of orofacial bones, including dental pulp, compared with those in axial and appendicular bone (bone marrow and ileac crest) based on their different embryological origins. Maxillas, mandible, including the alveolar bone (i.e. dentine, dental pulp and periodontal ligament), are formed exclusively by neural crest cells while axial and appendicular bones develop from mesoderm. Thus IDPSCs have the potential for neural regeneration and neuroprotection.
  • DPSC( SHED) enzymatically derived from adult rats using trypsin.
  • transplantation Unless the transplant is an autograft, there is always a risk that the host's immune system will attack the transplant. Even a well-matched allograft requires immunosuppression pretreatment. This remains true for stem cell transplantation.
  • the therapeutic stem cell population should be not be immunogenic. Immunogenicity is the ability of allogeneic stem cells to provoke an immune response when facing the host immune system after transplantation (Schu S et al., 2012).
  • MHC major histocompatibility complex
  • mice with mouse hepatitis virus-induced CNS demyelination resulted in increased T cell infiltration and NPC rejection (Weinger J G et al., 2012).
  • MHC major histocompatibility complex
  • recent evidence supports the possibility that undifferentiated adult stem cells are endowed with an immunologically privileged status and are capable of escaping the normal processes of allogeneic rejection (Bifari F et al. 2010).
  • Immunologically privileged status is possible for a population of cells if the cells lack the expression of MHCs.
  • no immunogenicity in humans can occur by the population of stem cells being essentially negative for human leukocyte antigen (HLA), which is the human version of MHC. Therefore, the absence of HLA-DR, which is a quality control characteristic of IDPSCs (see Example 1), is an essential marker for cell to be used in for systemic cell therapy without need of toxic immunosuppression pre-treatment to the patient.
  • HLA human leukocyte antigen
  • Another risk of stem cell transplantation is the increased risk of tumor development, especially for undifferentiated cells, because of these cell's potential for differentiation into other cell types.
  • Pluripotent stem cells especially hESCs and iPSCs cells are able to form spheres that resemble embryoid bodies in vitro and teratomas in vivo. All currently available technologies to apply pluripotent cells hold tumorigenicity risk. Expression and lack of expression of certain genes reduces risks to enable systemic administration of a population of stem cells.
  • Nanog is transcription factor associated with the maintenance of the pluripotent cells of the inner cell mass and the formation of embryonic stem cells.
  • Nanog is a leukemia inhibitory factor (LIF) and activator of transcription-independent factor-3 (STAT-3). It is regulated by OCT4 and SOX2 and in turn positively regulates the expression of OCT4, SOX2 and itself by binding to the respective promoter gene regions (Boyer et al, 2005; Loh et al., 2006; Li, 2010). Together, these three transcription factors play an essential role in preventing differentiation of pluripotent stem cells (Boyer et al., 2005).
  • LIF leukemia inhibitory factor
  • STAT-3 transcription-independent factor-3
  • the transfection cellular nucleus with OCT3/4, SOX2, NANOG was previously to be sufficient for inducing pluripotency in adult somatic cells (the creation of iPSC) and then lead to full pattern of embryonic stem cells theoretical characteristics: differentiation into 200 types of cells in the body, unlimited expansion, renewal potential, embryonic body formation, teratoma formation. Teratoma formation is a main threat of safety in cellular therapy.
  • absence expression of nanog in nucleus is an essential safety marker to determine whether a population of stem cell is suitable for systemic administration.
  • the lack of tumorigenicity of undifferentiated stem cells in vivo requires the absence of nanog in nucleus.
  • undifferentiated stem cells expressing inactive nanog, i.e. nanog localized in the cytoplasm also lack tumorigenicity in vivo.
  • Another important safety marker for a population of stem cells suitable for systemic administration is the expression of p53. This protein is crucial in multicellular organisms, where it regulates the cell cycle and thus prevents cancer by functioning as a tumor suppressor.
  • ABCG2 protein expression is also safety marker that indicates a population of stem cells is suitable for systemic administration.
  • ATP-binding cassette (ABC) ABCG2 protein (BCRP) expression is an important determinant of the MSC undifferentiated population phenotype.
  • ABCG2 might serve as a marker for undifferentiated stem cells from various sources, as its expression is sharply downregulated with differentiation.
  • ABCG2 transporters with Alzheimer's disease (AD), actively transport AP as confirmed histopathologically in AD cases and controls.
  • Genome-wide association studies (Bertram L et al., 2007) have implicated a have identified genes the modulate AD risk, including genetic variants in ABCA7, a variant of ABC gene. It was concluded that increase in ABCA7 expression reduces AD risk, though increased ABCA7 expression during AD is insufficient to block disease progression (Jared B et al., 2014).
  • the pharmaceutical composition of the invention comprises stem cells from tissue of neural crest origin expressing at least one safety markers selected from the group consisting of ATP-binding cassette sub-family G member 2 (ABCG2), inactive nanog, p53, and SOX2.
  • the at least one safety marker is elected from the group consisting of ATP-binding cassette sub-family G member 2 (ABCG2), inactive nanog, and p53.
  • the IDPSC have high expression of ABCG2 and p53 but low expression of inactive nanog and SOX2.
  • the at least one safety marker is ABCG2 or p53, at least 75%, 80%, 85%, 90%, 95% or 98% of the stem cells of the pharmaceutical composition express the at least one safety marker.
  • the at least one safety marker is inactive nanog or SOX2, no more than 30%, 25%, 20%, 15%, 10%, 5%, or 5% of the stem cells express the at least one safety marker.
  • the stem cells of the pharmaceutical composition coexpress ABCG3, p53, inactive nanog, and SOX2, wherein at least 75% of the stem cells express ABCG2, at least 75% of the stem cells express p53, no more than 5% of the stem cells express inactive nanog, and no more than 30% of the stem cells express SOX2.
  • the pharmaceutical composition comprises stem cells from tissue of neural crest origin expressing at least one neuroepithelial stem cell marker selected from the group consisting of brain-derived neurotrophic factor (BDNF), neutrotrophin-3 (NT3), neutrotrophin-4 (NT4), neutrotrophin-5 (NT5), and p75.
  • the stem cells have high expression of the at least one neuroepithelial stem cell marker.
  • the stem cells of the pharmaceutical composition coexpress BDNF, NT3, NT4, NT5, and p75.
  • these embodiments pharmaceutical composition comprise stem cells from tissue of neural crest origin that are negative for HLA-DR.
  • the stem cells of the pharmaceutical composition may also be negative for certain MSC markers selected from the group consisting of c-Myc, KLf-4, and REX-1.
  • the stem cells of the pharmaceutical composition are negative for HLA-DR, c-Myc, KLf-4, and REX-1.
  • the pharmaceutical composition may comprise stem cells from tissue of neural crest origin expressing at least one safety markers selected ABCG2, inactive nanog, and p53 and further express at least one neuroepithelial stem cell marker selected from the group consisting of BDNF, NT3, NT4, NT5, and p75.
  • the pharmaceutical may comprise stem cells from tissue of neural crest origin expressing express at least one neuroepithelial stem cell marker selected from the group consisting of BDNF, NT3, NT4, NT5, and p75 while further expressing at least one safety markers selected ABCG2, inactive nanog, p53 and SOX2.
  • the present invention provides for methods for treating neurological diseases and conditions comprising systemically administering the pharmaceutical composition of the invention to a subject.
  • these methods of treating neurological diseases and condition promote neurogenesis and are protective in models of neurodegenerative diseases.
  • systemic administration the population of stem cells such as by IV administration, results in direct delivery of the cells to the brain.
  • neurogenesis occurs by the population of stem cells self-differentiating and/or activating intrinsic stem cells to migrate and differentiate.
  • neurogenesis is preferably dopamine-associated.
  • the neurological disease or condition is treated by the stem cells crossing the blood/brain barrier (BBB) and inducing neurogenesis.
  • BBB blood/brain barrier
  • the stem cells are directly transplanted into the brain parenchyma, including striatum, following crossing of the BBB.
  • the induced neurogenesis is dopamine-associated.
  • dopamine-associated neurogenesis occurs through self-differentiation of the stem cells or activation of migration and differentiation of intrinsic stem cells by the extrinsic stem cells.
  • massive dopamine-associated neurogenesis takes place in the subventricular zone (SVZ).
  • the methods further comprise measuring the amount of DA receptor in the subject.
  • measuring the amount of DA receptor in the subject comprises imaging the subject to detect DA receptor.
  • neurogenesis is mediated by dopamine receptor D2, thus in some embodiments, the DA receptor measured is receptor D2.
  • the pharmaceutical composition provides neuroprotection.
  • systemic neuroprotection is provided with the high basal level of neurotrophic and immunoprotective factors expression and release pattern of the stem cells of the pharmaceutical composition.
  • these stem cells of the pharmaceutical composition are IDPSCs.
  • the neurological diseases and conditions include, for example, autism, schizophrenia, epilepsy, stroke and ischemia, a neurodegenerative disease or condition, a motor disorder, or a convulsive disorder.
  • the neurodegenerative disease or condition may be, for example, Parkinson's disease, multiple sclerosis, amyotrophic lateral sclerosis (ALS), stroke, autoimmune encephalomyelitis, diabetic neuropathy, glaucomatous neuropathy, Alzheimer's disease, and Huntingdon's disease.
  • Motor disorders include, for example, Tourette syndrome, amyotrophic lateral sclerosis (ALS), progressive bulbar palsy, spinal muscular atrophy (SMA), post-polio syndrome (PPS).
  • Convulsive disorders include, for example, epilepsy.
  • the methods for treating neurological diseases and conditions support the natural neuro-protective mechanism in subjects diagnosed with early HD. In other implementations, the methods for treating neurological diseases and conditions repairs lost DA neurons in subjects diagnosed with PD.
  • the present invention also provides for methods of using the pharmaceutical composition as a preventive therapy for subjects at risk of HD.
  • the present invention is directed to a pharmaceutical composition for systemic administration to a subject to treat a neurological condition
  • a neurological condition comprising undifferentiated stem cells from tissue of neural crest origin expressing at least one safety markers selected from the group consisting of ATP-binding cassette sub-family G member 2 (ABCG2), inactive nanog, and p53.
  • ABCG2 ATP-binding cassette sub-family G member 2
  • inactive nanog is expressed nanog localizing predominantly in the cytoplasma of the undifferentiated stem cell.
  • at least 75% of the undifferentiated stem cells express the at least one marker when the at least one marker is ABCG2 or p53.
  • no more than 5% of the undifferentiated stem cells express the at least one marker when the at least one biomarker is nanog.
  • the undifferentiated stem cells express ABCG2, inactive nanog, and p53. In one aspect, at least 75% of the undifferentiated stem cells express ABCG2, at least 75% of the undifferentiated stem cells express p53, and no more than 5% of the undifferentiated stem cells express inactive nanog. In another aspect, the undifferentiated stem cells further express SOX2, and wherein no more than 30% of the undifferentiated stem cells express SOX2.
  • the undifferentiated stem cells further express at least one neuroepithelial stem cell marker selected from the group consisting of brain-derived neurotrophic factor (BDNF), neutrotrophin-3 (NT3), neutrotrophin-4 (NT4), neutrotrophin-5 (NT5), and p75.
  • BDNF brain-derived neurotrophic factor
  • NT3 neutrotrophin-3
  • NT4 neutrotrophin-4
  • NT5 neutrotrophin-5
  • p75 neuroepithelial stem cell marker selected from the group consisting of brain-derived neurotrophic factor (BDNF), neutrotrophin-3 (NT3), neutrotrophin-4 (NT4), neutrotrophin-5 (NT5), and p75.
  • the undifferentiated stem cells express BDNF, NT3, NT4, NT5, and p75.
  • the present invention is directed to a pharmaceutical composition for systemic administration to a subject to treat a neurological condition comprising undifferentiated stem cells from tissue of neural crest origin at least one neuroepithelial stem cell marker selected from the group consisting of BDNF, NT3, NT4, NT5, and p75.
  • the undifferentiated stem cells express BDNF, NT3, NT4, NT5, and p75. In other aspects, the undifferentiated stem cells further express at least one safety markers selected from the group consisting of ABCG2, inactive nanog, p53, and SOX2. In certain aspects, inactive nanog is expressed nanog localizing predominantly in the cytoplasma of the undifferentiated stem cell.
  • the undifferentiated stem cells express the at least one marker when the at least one marker is ABCG2 or p53.
  • the undifferentiated stem cells are negative for HLA-DR.
  • the tissue of neural crest origin is dental pulp.
  • the undifferentiated stem cells from tissue of neural crest origin are immature dental pulp stem cells (IDPSCs).
  • the present invention provides a method of treating a neurological disease or condition comprising systemically administering to a subject a pharmaceutical composition comprising undifferentiated stem cells from tissue of neural crest origin expressing at least one safety marker selected from the group consisting of ABCG2, inactive nestin, and p53.
  • the undifferentiated stem cells of the pharmaceutical composition further express at least one neuroepithelial stem cell marker selected from the group consisting of BDNF, NT3, NT4, NT5, and p75.
  • the present invention is directed to a method of treating a neurological disease or condition comprising systemically administering to a subject a pharmaceutical composition comprising undifferentiated stem cells from tissue of neural crest origin expressing at least one neuroepithelial stem cell marker selected from the group consisting of BDNF, NT3, NT4, NT5, and p75.
  • the undifferentiated stem cells of the pharmaceutical composition further express at least one safety marker selected from the group consisting of ABCG2, inactive nestin, p53, and SOX2.
  • the subject is intravenously administered the pharmaceutical composition.
  • the neurological disease or condition is treated by the population of undifferentiated stem cells crossing the blood/brain barrier and inducing neurogenesis.
  • the neurological disease or condition is treated by the undifferentiated stem cells inducing neurogenesis via dopamine-associated neurogenesis.
  • the dopamine-associated neurogenesis is through self-differentiation of the undifferentiated stem cells or activation of migration and differentiation of intrinsic stem cells by the undifferentiated stem cells.
  • the undifferentiated stem cells of the pharmaceutical composition provide neurotrophic factors and immunoprotective factors. In other embodiments, the undifferentiated stem cells of the pharmaceutical composition provides systemic neuroprotection. In one embodiment, the undifferentiated stem cells are autologous and/or allogeneic to the subject.
  • the neurological disease or condition is a neurodegenerative disease or condition.
  • the neurodegenerative disease or condition may be selected from the group consisting of Parkinson's disease (PD), multiple sclerosis, amyotrophic lateral sclerosis (ALS), stroke, autoimmune encephalomyelitis, diabetic neuropathy, glaucomatous neuropathy, Alzheimer's disease, and Huntington's disease (HD).
  • the method comprises systemically administering the pharmaceutical composition to the subject, wherein the subject is diagnosed with early HD, supports the natural neuroprotective mechanism in the subject. In other aspects, the method comprises systemically administering the pharmaceutical composition to the subject, wherein the subject is diagnosed with PD, repairs lost dopaminergic neurons in the subject.
  • the neurological disease or condition is selected from the group consisting of autism, schizophrenia, stroke, and ischemia. In other embodiments, the neurological disease or condition is selected from the group consisting of a motor disorder and a convulsive disorder.
  • the subject is administered a single administration of the pharmaceutical composition. In one embodiment, the subject is administered a single intravenous injection of the pharmaceutical composition. In yet other embodiments, the subject is administered a first and a second administration of the pharmaceutical composition.
  • the subject is administered a first and a second intravenous injection of the pharmaceutical composition.
  • the second administration or intravenous injection of the pharmaceutical composition takes place at least 7 days after the first administration or intravenous injection.
  • the method further comprises measuring the amount of DA receptor in the subject. In another aspect, the method comprises measuring the amount of DA receptor in the subject comprises imaging the subject to detect DA receptor. In one aspect, the DA receptor is receptor D2.
  • the CNS is originated from ectoderm, more precisely from the pool of multipotent neuroepithelial stem cells (NSCs).
  • This cell's population is composed at least by two different groups of neural precursors: a first group can grow in monolayer cultures; a second group generally form neurospheres (i.e. grow in suspension culture).
  • both of cell types demonstrate the same differentiation potential (Noble et al., 2011, Kempermann, 2011).
  • NSCs are symmetrically dividing and under appropriate conditions can be rapidly conversed into neuroblasts, asymmetrically dividing precursors, which are able to develop into neurons after a migration phase.
  • Neuroblasts are slightly more differentiated than NSCs and can produce transit amplifying cell population. The ability to divide is main difference between neuroblasts and neurons, which are postmitotic. Neuroblast differentiates and matures into neurons but not in other cell types.
  • NSCs fate is regulated through signals that emanate from surrounding tissues. NSCs are found in specific regions, so-called stem cell niches that provide the microenvironmental cues that regulate stem cell proliferation, self-renewal and differentiation.
  • fetal neuronal precursors obtained from abortion and, second, pluripotent stem cells (ES cells, iPSCs) induced to differentiated in vitro neuronal precursors.
  • ES cells pluripotent stem cells
  • iPSCs pluripotent stem cells
  • Fetal neuronal precursors raise many ethical problems, while there are certain safety risks associated with human use of ES and iPSCs that delay therapeutic applications.
  • Alternative methods of neurodegenerative therapies from other human types of stem cells are needed to exploit the uses of cell therapy for treatment of neurological diseases. Methods for transplanting stem and progenitor cells are described in U.S. Pat. Nos. 5,928,947; 5,817,773 and PCT Publication Nos. WO 01/176507.
  • These methods include expansion in cell culture and transplantation of undifferentiated neuroprotective stem cells, and/or through by transplantation of neural precursor cells, or/and fully differentiated neuronal cells.
  • Cell therapeutic interventions may involve both cell transplantation and the stimulation of endogenous neural progenitor cells.
  • Conventional methods have multiple disadvantages and limitations and new sources of cells for stem cell neuroprotective/neuroregenerative therapy are required. Also of importance is to predict neuroprotective potential of cells for stem cell therapy using molecular markers.
  • compositions of hIDPSCs which co-express SOX1, SOX2 and beta-3-tubulin markers and especially methods of treating neurological conditions. Also disclosed herein are compositions and methods of enrichment of the co-expressed markers during LT hIDPSCS cell culture therefore increasing neuroectodermal lineage commitment of such LP hIDPSCs. Disclosed herein are new methods to the treatment of neurodegenerative diseases using hIDPSCs obtained through LT harvesting cycles and enriched with specific neuronal biomarkers, such as but not limited to SOX1, and SOX2, beta-3-tubulin.
  • the contemplated embodiments provide a substantially homogenous population of cells enriched with population of cells that co-express said stemness neuronal markers (SOX1, SOX2 and beta-3-tubulin) while maintaining normal karyotype and carry no risk of teratoma formation.
  • the homogenous population is likely advantageous for treating disease in a human (mammalian).
  • Other advantages of the cell-based therapies disclosed herein include, but are not limited to, incorporation of the cells central nervous system tissue, peripheral nervous system tissue. Such incorporated cells have the potential to differentiate or develop into neuronal, glial or other cells to replace or facilitate repair of the damaged, traumatized or degenerating tissue thereby resulting in a more permanent treatment of the degenerative, acute injury, traumatized, neurological condition.
  • Oxygen is one of critical signals that can affect stem cell properties under normal tissue homeostasis. Interestingly, low oxygen levels, or hypoxia, are a hallmark of stem cell niches (Panchision, 2009).
  • IDPSCs immature dental pulp stem cells isolated from DP after 30 times of mechanical transfer/harvesting cycle (late population (LP) of IDPSC) and tested for their capacity to produce neurons in vitro demonstrated the increased ability for neural differentiation, as well as enrichment of a population pre-committed LP IDPSC with neuronal precursor when compared with IDPSC isolated after 15 or 25 cycles of mechanical transfer/harvesting (early population (EP)).
  • LP mechanical transfer/harvesting cycle
  • the number of actively proliferating neuronal precursors (neuroblasts) in pre-committed IDPSC neural precursor are related with the number of DP transfer cycles.
  • SOX1 (for sex-determining region Y-box 1) protein is a transcription factor involved in early central nervous system development.
  • SOX1 is a known marker characteristic of a developing central nervous system. The expression of SOX1 in the neural plate and tube seem to correlate with mitotically active progenitors that are not yet committed to a final stage.
  • SOX1 defines the dividing neural precursors of the embryonic CNS and expresses particularly in the ventral striatum (Pevny et al., 1998; Kempermann et al., 2003).
  • SOX2 is one of the earliest known transcription factors expressed in the developing neural tube and is expressed in certain cells of the adult brain. SOX2 is commonly known to play a critical role in the central nervous system. Recently, Terskikh and colleagues (2011) elucidate role of SOX2 in peripheral nervous system. They developed a hESC-based model in which migratory cells undergo epithelial to mesenchymal transition (EMT) to acquire properties of neural crest (NC) cells. They found that migratory NC progenitor's down-regulate SOX2, but then start re-expressing SOX2 as they differentiate to form neurogenic dorsal root ganglion (DRG)-like clusters.
  • EMT epithelial to mesenchymal transition
  • NC neural crest
  • SOX2 down regulation was sufficient to induce EMT and resulted in massive apoptosis when neuronal differentiation was induced. They also showed that SOX2 binds directly to NGN1 (neorogenin 1) and MASH1 promoters and is required for their expression. When neural crest stem cells were prevented from re-expressing SOX2, they die or can give rise to glia or smooth muscle cells. Thus, the function of SOX2 is to keep cells multipotent or pluripotent for become neurons later in development.
  • This combination of defining properties will identify the neural progenitor cell lines of the invention regardless of the method used for their isolation.
  • One method is to transplant undifferentiated stem cells that can differentiate into neural precursor cells, and then into fully differentiated neurons (including neurons, astro-glia, or oligodendrocytes).
  • Another method is to transplant undifferentiated or pre-differentiated IDPSCs that are capable to release neuroprotective factors into affected areas of the nervous system.
  • using the described hIDPSCs transplant method of neuroregeneration may be based on administering into patient pre-differentiated ex vivo neuronal precursors or fully differentiated neurons or combinations of thereof.
  • a cell culture composition comprises (co-expressing) SOX1 and SOX2 positive hIDPSC, which are neural crest stem cells.
  • the SOX1 and SOX2 may be obtained through LT cell culture. Enrichment of the transcription factors increases towards LP culture, which holds high potential to neuronal differentiation, keeps karyotypes normal, and does not carry the risk of teratoma in vivo.
  • a method for obtaining a homogenous population of hIDPSC comprises obtaining the population thorough LT mechanical/non-enzymatic harvesting cycles.
  • the number of harvesting cycles may comprise over 25 cycles, about 45 cycles, or more than 45 cycles.
  • An embodiment of the method may further comprise culturing the taken cells and passaging a plastic adherent cell from the cell culture.
  • the plastic adherent cell typically (i) self-renews, (ii) differentiates into cells of endodermal, mesodermal, or ectodermal lineage, and (iii) expresses markers SOX1, SOX2 and 3-beta-tubulin.
  • An embodiment of the method may further comprise administering the product to a patient as a pharmaceutical composition in a pharmaceutical accepted carrier/vehicle for cell therapy to treat CNS diseases.
  • a mitotically active homogeneous neural progenitor's (neuroblasts) composition comprises (co-expressing) such markers as SOX1, SOX2, and beta-3-tubulin that are not yet committed to a final stage.
  • a method for obtaining a homogeneous population of pre-differentiated IDPSCs comprises one or more of the following steps. Obtaining IDPSCs-derived neurospheres by culturing IDPSC in non-adherent conditions (suspension).
  • the cells within the rosettes are presented by (i) transit amplifying cell population of neuroblasts positive for SOX1, SOX2, and BrdU and beta-3-tubulin that are slightly more differentiated than IDPSC; (ii) neuroblasts differentiated terminally into NEURONAL LINEAGES OF CENTRAL AND PERIPHERAL NEURAL SYSTEM—(including neuronal precursors, astroglia and oligodendrocytes and ganglion cells) upon external in vitro- or in vivo-induced microenvironment; (iii) use as a pharmaceutical composition in a pharmaceutical accepted carrier/vehicle for cell therapy to treat CNS diseases by administrating to a patient; and/or (iv) use as a pharmaceutical composition in pharmaceutical accepted carrier/vehicle for anti-cancer therapies, including gene therapy, to treat neuroblastoma and retinoblastoma but not limited to these tumor types.
  • substantially homogenous LP of hIDPSCs comprise a cell that co-expresses a marker selected from, but not limited to, the group comprising SOX1, SOX2, or beta-3-tubulin.
  • contemplated disclosures are provided an undifferentiated hIDPSC wherein the cell is immunoreactive with markers for human pluripotent stem cells including Oct3/4, Nanog and SOX2, and wherein said cell may differentiate under differentiating conditions to neural cells.
  • the cells express the transcription factor SOX1; SOX2 and tubulin as demonstrated by RT-PCR.
  • the said cells maintain a stable diploid karyotype during prolonged cultivation in vitro.
  • Sox and Oct3/4 transcription factors are thought to be central to the transcriptional regulatory hierarchy that specifies ES cell identity.
  • hIDPSC were previously shown to express Oct 4 (Insert ref 2006; and Liezer et al. 2012) approximately in about of 20% of IDPSC throughout the cell culture up to LP. Therefore, the co-existence of both OCT family and SOX family markers, indicate on high stemness and pluripotency of hIDPSC. However enrichment with SOX and beta-tubulin markers indicate on strong affinity towards neuronal fate.
  • a preparation of undifferentiated hIDPSCs using LP harvesting method where enrichment with SOX1, SOX2, BrdU, beta-3-tubulin molecular markers in LP indicate on the capability of these LP to proliferation in vitro and differentiation (in vivo or ex vivo) into neural progenitor cells, neuron cells and/or glial cells and ganglion cells.
  • the undifferentiated hIDPSCs cells have the potential to differentiate into neural progenitor cells, neuron cells and/or glial cells when subjected to differentiating conditions or in vivo micro-environment.
  • CELLAVITATM (stem cells) is the bulk material prior to final formulation. CELLAVITATM (stem cells) is referred to as Drug Substance (DS). CELLAVITATM (stem cells) for IV infusion is referred to as Drug Product (DP).
  • DS Drug Substance
  • DP Drug Product
  • the Process for the CELLAVITATM(stem cells) Substance initiates at donor screening and testing and finishes prior to final formulation and cryopreservation of the cell stock. Preparation of the Drug Product involves formulation of the CELLAVITATM (stem cells) substance with additional excipients.
  • CELLAVITATM stem cells
  • stem cells is a stem cell expressing neural crest/mesenchymal stem/progenitor cell markers, such as CD13, CD105 (Endoglin), CD73, CD29 (integrin b-1), CD44, and nestin (Kerkis et al., 2009; Kerkis et al., 2006) obtained using multiharvest org ⁇ o and tissue explant culture.
  • CELLAVITATM stem cells
  • the cells are defined in accordance with minimal criteria for defining multipotent mesenchymal stromal cells established by the Mesenchymal and Tissue Stem Cell Committee of the International Society for Cellular Therapy. This definition includes being plastic-adherent when maintained in standard culture conditions, expressing CD105, CD73 and CD90, and lack expression of CD45, CD34, CD14 or CD11b, CD79alpha or CD19 and HLA-DR surface molecules, and ability to differentiate to osteoblasts, adipocytes and chondroblasts in vitro (Dominici et al., 2006).
  • the tooth Immediately after spontaneous exfoliation, the tooth will be immersed into 3 mL of sterile transporting solution composed of DMEM (Dulbecco's Modified Eagle Medium) and 500 mM Gentamycin in a 15 mL sterile centrifuge tube. The tooth will be stored at 4° C. and processed within 48-72 hours.
  • DMEM Dynamic Eagle Medium
  • Gentamycin 500 mM Gentamycin
  • a freshly exfoliated deciduous tooth from a healthy subject will be washed repeatedly in sterile solution containing 50% pen/strep solution (100 units/mL penicillin, 100 units/mL streptomycin) and 50% Phosphate Buffered Saline (PBS). Dental pulp will be removed from the tooth with the aid of a sterile needle.
  • pen/strep solution 100 units/mL penicillin, 100 units/mL streptomycin
  • PBS Phosphate Buffered Saline
  • Freshly obtained dental pulp (DP) will be washed in a solution containing 3% Pen/strep solution (100 units/mL penicillin, 100 units/mL streptomycin). Initial plating and viability testing of the dental pulp will be performed in dental pulp Maintenance Medium supplemented with 15% fetal bovine serum (FBS, Hyclone), 100 units/mL penicillin, 100 units/mL streptomycin, 2 mM L-glutamine, and 2 mM nonessential amino acids. This procedure usually takes up to one week. Once the DP will be determined as viable it will be harvested and hIDPSC will be passaged. The resulting hIDPSC will be cryopreserved under GTP conditions for future clinical research
  • FBS fetal bovine serum
  • CELLAVITATM stem cells
  • Dr. Irina's lab for obtaining patent and generating nonclinical data.
  • the technology was transferred to a semi-industrial, non-GMP facility in Israel (Sheba Hospital).
  • the CMC content in this briefing document describes the manufacturing process in Israel, and is also representative of the plan to manufacture clinical batches in a GMP facility in Brazil, to where the technology will be transferred.
  • the production process comprises the steps illustrated in FIG. 1 which demonstrates the initial process of CELLAVITATM (stem cells) isolation and batch formulation.
  • the vertical pathway shows the process of dental pulp mechanical transfer (harvest) of early population-hIDPSC (isolated from dental pulp before 5 harvests) and late population—hIDPSC (isolated from dental pulp after 5 harvests).
  • the horizontal pathway shows the traditional enzymatic method of cell cultures when cells are replaced through repetitive passages.
  • the final batch product is a sum of hIDPSC obtained from dental pulp harvests and passages (no more then 5).
  • the production process comprises the steps illustrated in FIG. 2 and/or FIG. 3 .
  • the production process includes CELLAVITATM (stem cells) isolation and batch formation.
  • the vertical pathway shows the process of dental pulp (DP) mechanical transfer (harvest) for early population-hIDPSC isolated from dental pulp before 5 harvests and late population—hIDPSC isolated from DP after 5 DP harvests;
  • the horizontal pathway shows traditional enzymatic method of cells culturing, when cells are replaced through repetitive passages.
  • Final batch—product is a sum of hIDPSC obtained from DP harvest and passages (no more then 5).
  • CELLAVITATM stem cells
  • DP When the semi-confluent colony formation of hIDPSC will be detected around the dental pulp explant, DP will be transferred into a new cell culture vessel for continued growth in DP Maintenance Medium.
  • hIDPSC will be washed with sterile PBS, removed with TrypLE solution and centrifuged. The pellet will be resuspended in DP Maintenance Medium and thereafter will be seeded in the tissue culture flask (Passage 1-P1). When the cells reach about 80% confluency they will be passed to the new flask (Passage 2-P2). Cells will be incubated in a humidified 5% CO2 incubator.
  • hIDPSC from P5 will be maintained in culture for at least 3-5 days in order to collect cell conditioned medium for sterility and mycoplasma testing.
  • the hIDPSC freezing protocol will be adapted to the standard freezing repository protocol hIDPSC from P5 will be transferred into a 2 mL cryopreservation vials containing 1 mL of freezing media, composed of: 90% FBS and 10% DMSO (GMP/US pharmaceutical grade). 1 ⁇ 106 cells per vial will be cryopreserved.
  • Cryopreservation vials will be placed inside a Nalgene Cryo 1° C. Freezing Container filled with isopropyl alcohol and will be placed at ⁇ 80° C. overnight. Thereafter, vials will be transferred to the vapor phase of a liquid nitrogen storage tank and their locations will be recorded.
  • Table 1 depicts the process, which will be used for control of materials.
  • the parents/guardians of the subjects will answer a donor eligibility questionnaire concerning the health of their child.
  • blood samples total volume of 10 mL
  • pathogens according to guidelines for eligibility screening:
  • the total amount of blood to be drawn for these tests will not exceed 0.8 mL/kg body weight for one blood draw.
  • the tooth Immediately after spontaneous exfoliation, the tooth will be immersed into 3 mL of sterile transporting solution composed of DMEM (Dulbecco's Modified Eagle Medium) and 500 mM Gentamycin in a 15 mL sterile centrifuge tube.
  • DMEM Dulbecco's Modified Eagle Medium
  • 500 mM Gentamycin 500 mM Gentamycin in a 15 mL sterile centrifuge tube.
  • the tooth will be stored at 4° C. and processed within 48-72 hours in the GTP laboratory.
  • the hIDPSC freezing protocol will be adapted to the standard freezing repository protocol hIDPSC from P5 will be transferred into a 2 mL cryopreservation vials containing 1 mL of freezing media, composed of: 90% FBS and 10% DMSO (GMP/US pharmaceutical grade). 1 ⁇ 106 cells per vial will be cryopreserved.
  • Cryopreservation vials will be placed inside a Nalgene Cryo 1° C. Freezing Container filled with isopropyl alcohol and will be placed at ⁇ 80° C. overnight. Thereafter, vials will be transferred to the vapor phase of a liquid nitrogen storage tank and their locations will be recorded.
  • CELLAVITATM Stewing and Administration of CELLAVITATM (Stem Cells)
  • CELLAVITATM stem cells
  • the cryopreserved product after thawing must be assessed for cell viability. This will be done with a kit that will be sent along with the CELLAVITATM (stem cells). The kit and the method to be performed must be followed as described below.
  • CELLAVITA′ stem cells
  • Each donor IDPSC culture will be defined as a separate batch of raw material for future stem cell expansion process.
  • hIDPSC will be individually coded and labeled, in an anonymous fashion. Cryotubes boxes will be double-packed to reduce risk of cross contamination, according to process.
  • IDPSC After mutiharvest of DP tissue and explant culture, IDPSC demonstrate capacity to form colonies ( FIG. 7 ).
  • the colony forming assay (CFU-F) assay was performed in triplicate at T20, P3 using 480 cells seeded in each plate, at day 8 multiple colonies colonies appeared and approximately 100 colonies were formed in each plate ( FIG. 7 ).
  • Colony forming capacity is one of the principal characteristics of stem cell. Therefore, we conclude that this capacity was maintained when the cells were obtained using the disclosed multiharvest organ and tissue explant method. Additionally, the proliferative activity of LP IDPSC was evaluated as shown on FIG. 8 , and these cells demonstrated a very high proliferative rate.
  • adenosine triphosphate binding cassette (ABC) transporters was also tested in the cells. ABC transporters are involved in the active transport of an extremely diverse range of substrates across biological membranes. These transporters are commonly implicated in the development of multidrug resistance and are also involved in numerous physiological and homeostatic processes, including lipid transport, cell migration and differentiation. Moreover there is evidence that ABC transporters serve as phenotypic markers and functional regulators of stem cells (Bunting 2002).
  • ABCB1 is expressed in human fetal neural stem/progenitor cells (hNSPCs).
  • BDNF brain-derived neurotrophic factor
  • the average level of BDNF secreted by 1 ⁇ 10 6 hIDPSCs is 6589 pg, which is many times higher than other types of MSCs that secrete BNDF, such as the MSCs of Gothelf et al. in 2014 (Clin Transl Med. 2014 3:21). Gothelf et al.
  • BM-MSC induced bone marrow-derived MSCs
  • BM-MSC-NTF neurotrophic factor-secreting cells
  • cAMP dibutyryl cyclic 15 AMP
  • hbFGF human Basic Fibroblast Growth Factor
  • PDGF-AA human platelet derived growth factor
  • hIDPSCs still secreted four times more BDNF than BM-MSC-NTF. Accordingly, the hIDSPCs have much greater neuroprotective potential than bone marrow-derived MSC induced to secrete neurotrophic factors.
  • MSCs generally express pluripotent markers such as Oct4, Nanog and Sox2 at low levels as described in the literature (Jiang et al., 2002; Guillot et al., 2007).
  • hIDPSC express very low levels of such markers in comparison with human embryonic stem cells and even induced pluripotent stem cells obtained from hIDPSCs (see FIG. 9 ).
  • hIDPSC express a high level of p53.
  • the tumor suppressor gene p53 is well known as a master regulator that helps keeps cancer at bay. Blocking the p53 pathway vastly improves the ease and efficiency of transforming differentiated cells into induced pluripotent stem cells (Dolgin, 2009).
  • the neuronal system consists of two classes of neural precursor cells: neuronal NPCs that differentiate into neurons and glial NPCs that differentiate into glia. Both neuronal and glial NPCs descend from the same neuroectodermal precursor.
  • a third class of neural precursor cells, neuroglioblast was also suggested. This third class of cells include radial glial cells also can act as neuronal precursors and only later, after neurogenesis, do they shift towards an exclusive generation of astrocytes.
  • the ability to distinguish whether a population of cells in cell therapy are neuronal NPSs or glial NPCs is of extreme importance for developing an efficient cell therapy strategy for treating neurodegenerative diseases, which mainly involve the damage or loss of both glia and neurons. It is possible to test known cell populations for the potential to differentiate into neurons or glia by inducing these cells to differentiate.
  • EP early population
  • LP late population
  • FIG. 10 The capacity of EP (early population) and LP (late population) IDPSC to produce neurons and glias was tested by inducing neuronal differentiation in EP and LP IDPSC at early (P2) and late passages (P7) according to the previously described protocol (Kerkis et al., 2006) ( FIG. 10 ). After 7 days, the cells were collected and analyzed by flow cytometry using GFAP (glial fibrillary acidic protein) and beta-III-tubulin antibodies, respectively. A significant difference exists in the number of cells that express these markers between EP (B-E, left) and LP (B-E, right), which were established following a dental pulp (DP) harvesting protocol.
  • GFAP glial fibrillary acidic protein
  • IDPSCs can be neuronal and glial NPCs.
  • Early DP harvesting EP IDPSC
  • LP IDPSC late DP harvesting
  • SHED which are stem cells from human deciduous teeth, cannot be categorized as an early population or late population because they are stem cells isolated from dental pulp cells without DP harvesting.
  • the single SHED population contains neuron-committed and not glial-committed progenitors.
  • Miura et al. (2003) neuronal differentiation of SHED resulted in increased expression of beta-III-tubulin, GAD, and NeuN while the expression of nestin, GFAP, CNPase, and NFM remained the same after the induction of differentiation (see FIG. 41 of Miura).
  • MSCs from different sources can respond differently to various stimuli (Fraser J K et al., 2006).
  • Culture conditions e.g., media supplemented with either human serum or fetal calf serum (FCS), or serum-free
  • FCS fetal calf serum
  • Different isolation and culture protocols used by different groups may account for the predominance of a particular MSC subpopulation with a distinct differentiation potential (Ho et al., 2008; Pevsner-Fischer et al., 2011; Rada et al., 2011).
  • SHED and IDPSCs have different methods of isolation and come from different stem cell niches. So it is unsurprising that SHED and IPSCs also have different expression of stem cell markers (see Table 5).
  • SHED To induce neuronal differentiation, SHED need EGF 20 ng/ml (BD Bioscience), FGF 40 ng/ml (BD Bioscience) and 3% rat serum. They need four weeks in neural inductive culture in order to show neural morphology and to increase expression of neuronal markers.
  • IDPSCs have advantages over SHED regarding neurogenesis.
  • SHED were injected into the dentate gyrus of the hippocampus of immunocompromised mice. The data demonstrated that SHED were able to survive for more than 10 days in mice hippocampus and to express NFM, which were expressed also by undifferentiated SHED (Miura et al., 2003).
  • pre-differentiated SHED SHED-derived spheres created by a combination of EGF and bFGF for 7 days in vitro
  • the cell suspension (200,000/ ⁇ L) was injected into 2 DA-depleted striatum sites in rats (2.5 ⁇ L per site).
  • Cyclosporine A is shown to decreases the size of the ischemic brain infarct in rats and to protects against synaptic dysfunction and cell death in rodent models of traumatic brain infarct as well as to protects striatal neurons from mitochondrial dysfunction in Huntington disease (Matsomoto et al., 1999; Albensi et al. 2000; Leventhal et al., 2003). Therefore, benefits observed in Parkinsonian rats and HI cannot be purely attributed to SHED but also to Cyclosporine A intervention.
  • hIDPSCs from Avita International LTD are advantageous over other therapeutic stem cells on the market or in clinical trial.
  • Avita International LTD's hIDPSCs have a good safety profile with low risk of immunogenicity and a low cost of production as they can be cryopreserved.
  • Table 7 depicts CELLAVITATM (stem cells) specifications.
  • Mycoplasma tests will be performed regularly during cultivation of hIDPSC with an in-house RT-PCR test (EZ-PCR Biological Industries, Israel) according to the manufacturer's protocol.
  • Karyotype analysis will be performed several times showing karyotype stability and these data are already published (Kerkis et al., 2006; Beltr ⁇ o-Braga, 2011; Lizier et al., 2012). Additionally, independent karyotype analysis will be performed in Cytogenetics.
  • Immunostaining of cell surface markers will be carried out with monoclonal antibodies against various surface antigen markers: HLA-DR-FITC, CD44-FITC, CD45-APC, CD105-PE, CD73-FITC, CD90-APC (eBioscience CA, USA), SOX2-PE, Nestin-PE, Tubulin-APC, NGF-PE (R&D systems, MN, USA). 2 ⁇ 105 cells will be used for FACS experiments. Cells will be washed twice with PBS (w/o Ca and Mg) and suspended in 50 ⁇ l PBS. Cells will be then incubated with antibodies for 15 min at room temperature. The cells will be washed twice with PBS and analyzed with a Becton-Dickinson flow cytometer. The fluorescence of PE (FL2), FITC (FL1), APC (FL4) will be detected in 575 nm, 530 nm and 600 nm emission wave lengths, respectively.
  • BDNF levels will be quantified by using a human BDNF Quantikine ELISA kit, according to the manufacturer's protocol (R&D Systems, MN, USA).
  • Table 8 depicts batch Number 001H1-30/P1-5/F analysis.
  • Avita performed non GMP, non GLP studies regarding hIDPSC stability.
  • a single master cell bank which may mitigate variability of the final batch, was established. It was composed by 5 batches, each derived from Dental Pulp of one individual. The cells were produced as described in FIG. 1 .
  • the time line of stability studies of hIDPSC is presented on FIG. 11 .
  • CELLAVITATM stem cells
  • MSC mesenchymal stem cells
  • the research group tested the neuroprotective and/or neural tissue remodeling effects of hIDPSC in a 3-NP chemical model of Huntington's disease.
  • 3-NP mitochondrial toxin 3-nitropropionic acid
  • SDH mitochondrial succinate dehydrogenase
  • 3-NP can be administered systemically, causing selective neurodegeneration of the striatum or the entire corpus striatum.
  • 3-NP administration can simulate different stages of Huntington's disease.
  • Intraperitoneal injections of two 3-NP doses lead to hyperkinetic symptoms in mice in the early stages of disease, whereas four or more doses result in hypoactivity in the late stages of disease (Beal et al., 1993; Brouillet et al., 1995; Yang et al., 2008; Borlogan et al., 1997).
  • 3-NP-treated animals has an important limitation if compared to transgenics.
  • 3-NP model the striatum lesion can regenerate spontaneously, after 10-12 days because of the presence of normal intrinsic neuronal precursors and an absence of genetic background which provides constant neurodegeneration. Therefore, difference in motor and functional improvements between experimental and control groups can be observed before spontaneous neuroregeneration.
  • hIDPSCs were detected in the cortex and corpus striatum, indicating that they were able to cross the blood-brain barrier and migrate to the site of injury ( FIG. 13 ).
  • optical cuts demonstrate at different depth of focus (A1-A4) the presence of IDPSC stained with Vybrant (green), and nuclei are stained with PI (red).
  • the cells demonstrate capillary predominant association and different morphological types: neuron-like cells and pericytes. On A2, A3, and A4 two pericytes at different locations along capillary can be observed, and both present similar morphology. On A4 embranchment of axons is shown.
  • DIC Digital Interference Contrast
  • hIDPSC-derived neuron-like cells and pericytes were also observed in the same period ( FIG. 13 ).
  • FIG. 14 the optical cut demonstrates IDPSC stained with Vybrant (green) and positively reacted with anti-IDPSC antibody (red). Superposition of both produce yellow color.
  • the cell demonstrate near capillary localization.
  • Two markers for MSC were used: CD73 and CD105 demonstrating positive reaction with IDPSC.
  • 3-NP-induced striatal lesions were determined by neuron loss using Nissl staining and DARPP32 expression ( FIG. 17 and FIG. 18 , respectively).
  • Nissl stains are used to identify neuron structures in the brain and spinal cord ( FIG. 17 )
  • DARPP32 is a cytoskeleton marker expressed in GABAergic neurons and prevalent in the striatum of healthy mammals ( FIG. 18 ).
  • neuron loss in the corpus striatum was scored as follows:
  • 3-NP-treated animals showed complete or partial neuron loss in the striatum compared to controls (no 3-NP or hIDPSC).
  • the two experimental groups presented different scores for neuronal loss in the striatum relative to controls.
  • morphometric histological analysis revealed that most hIDPSC-treated animals had scores 3 and 2, whereas most untreated animals (3-NP+saline solution) had neuron loss scores of 2 and 1 ( FIG. 18 ). No animal had visible atrophy ( FIG. 17 and FIG. 18 ).
  • FIG. 19 depicts neuronal growth in the striatum of rats after hIDPSC.
  • Administration of hIDPSC resulted in a neuroreparative effect in hIDPSC-treated animals by (A) Nissl staining and (B) DARPP32 expression.
  • C Number of animals showing neuron recovery after hIDPSC administration compared to Controls. Most hIDPSC-treated animals (3-NP+hIDPSC) had scores 3 and 2 (moderate and mild) whereas most untreated animals (3 NP+saline) had scores 2 and 1 (severe and moderate).
  • DARPP32 dopamine- and cAMP-regulated neuronal phosphoprotein
  • the regulation of the state of DARPP-32 phosphorylation provides a mechanism for integrating information arriving at dopaminoceptive neurons, in multiple brain regions, via a variety of neurotransmitters, neuromodulators, neuropeptides, and steroid hormones (Svenningsson et al., 2004).
  • HD is associated with severe striatal D1 and D2 receptor loss and taking in consideration that recently it was reported that dysregulation of dopamine receptor D2 as a sensitive measure for Huntington disease pathology in model mice (Crook et al., 2012; Chen et al., 2013), therefore we used this marker to evaluate possible effect of IDPSC in 3-NP induced rats.
  • BDNF's mRNA and protein levels have been observed in the cerebellum, caudate putamen, striatum, and cerebral cortex of HD patients (Adachi et al., 2014).
  • BDNF expression was observed in the striatum, caudate, putamen, and subventricular zone of hIDPSC-treated animals 7 and 30 days after hIDPSC administration ( FIG. 22 ).
  • BDNF expression in the subventricular zone indicates that hIDPSC promoted neurogenesis.
  • No BDNF expression was observed in untreated animals (3-NP+saline).
  • hIDPSCs Histological and immunohistochemical analyses revealed that hIDPSCs were able to cross the blood-brain barrier and reach different areas affected by HD, including the striatum and cortex. Morphometric histological analysis revealed that most hIDPSC-treated animals showed mild neuron loss in the striatum compared to untreated animals (3-NP+saline). Moreover, hIDPSCs showed neuroprotective and neuroreparative effects, as revealed by the upregulation of BDNF, DARPP32, and D2 receptor expression, which are downregulated in Huntington's disease (Van Dellen et al., 2000; Crook and Housman, 2012).
  • the primary study assessing safety of hIDPSC was the 3-nitropropionic acid (3-NP) rat model of HD study.
  • 3-NP-treated rats received a single IV injection or a total of three IV injections at one month intervals of the cells. Seven deaths occurred in 3-NP induced animals which received 3 ⁇ 106 cell/kg, and 5 deaths occurred in animals receiving 3 ⁇ 107 cell/kg.
  • placebo groups (3-NP induced without the cell transplantation) same number (12) of animals died. All rats that died presented with extremely severe disease manifestation; the deaths occurred within 5 days of 3-NP administration. No additional deaths occurred with repeated hIDPSC doses. Based on these data, probable cause of all early deaths was 3-NP toxicity. Because no deaths occurred after repetitive hIDPSC transplantation, this supports the safety of hIDPSC transplantation (Table 13).
  • HD is a clinically debilitating disease for which there is no available therapy to stop or reverse disease progression.
  • BBB blood-brain barrier
  • the BBB is a highly selective permeability barrier that separates the circulating blood from the extracellular fluid surrounding the nervous system. Treatment with cell-based therapeutics, therefore, would seem to require localized delivery bypassing the BBB (i.e. injection through the selective permeability barrier) since cells do not generally cross the BBB.
  • hIDPSCs have mesenchymal stem cell (MSC) attributes, can secrete immunomodulating and neurotropic factors.
  • MSC mesenchymal stem cell
  • histological and immunohistochemical analyses in validated HD rat model reveal that hIDPSCs are able to cross the BBB and reach different areas affected by HD, including the striatum and cortex.
  • Morphometric histological analysis reveals that most hIDPSC-treated animals show mild neuron loss in the striatum compared to untreated animals.
  • hIDPSCs show neuroprotective and neuroreparative effects by upregulating BDNF, DARPP32, and D2 receptor expression, which are downregulated in Huntington's disease (Van Dellen et al., 2000; Crook and Housman, 2012).
  • FIG. 28 summarizes the preclinical pharmacology studies, which aimed at examining the different clinical applications of the investigational product CELLAVITATM (stem cells). Although many of the studies were conducted to investigate pharmacological efficacy for various indications, they are all supportive to demonstrate the safety profile of the product as well as for the proposed intravenous route of administration.
  • IDPSC shows Oct4 nuclear localization ( FIG. 25 ). While a majority of IDPSCs have Nanog in the cytoplasm of cells ( FIG. 25 ), very rarely do cells demonstrate nuclear localization as well. The intracellular localization of Sox2 in IDPSCs is mainly nuclear ( FIG. 25 ) though several cells can show cytoplasmic localization too. Interestingly, we can observe the symmetrical division when Nanog and Sox2 expression in seen in both daughter cells, and observe asymmetric division when after division the daughter cells do not express these markers or loses the characteristic of stem cells and becomes a less potent progenitor or a differentiated cell ( FIG. 25 ).
  • Teratomas formation is an essential tool in determining the pluripotency of any pluripotent cells, such as embryonic or induced pluripotent stem cells (ES and iPS cells).
  • ES and iPS cells embryonic or induced pluripotent stem cells
  • hIDPSC induced pluripotent stem cells from hIDPSC.
  • the pluripotency of hIDPSC derived iPS cells were tested through teratoma formation, while human embryonic stem (ES) cells and hIDPSC were used as control. Routine protocol for teratomas production for ES cells was used (Hentze et al., 2009). Following this protocol 10 6 of cells of each line: hIDPSC-iPS cells, ES cells and hIDPSC were inoculated in the rear leg muscle of 4-week-old male, SCID. In animals, which were inoculated with hIDPSC-iPS cells or ES cells teratoma formation was observed after three months.
  • hIDPSCs which were used as a negative control in this study and were inoculated in 10 animals, did not produce teratomas neither after three month nor after six months of follow-up. Five of these animals were maintained alive during one year and even after this time teratomas formation was not observed.
  • teratoma formation in pluripotent stem cells requires the development of tissues derived from the three germ layers. For adult/mesenchymal stem cells, any alterations in the integrity of normal tissue at the site of transplantation were considered. After six months, animals, which were inoculated with hIDPSCs and did not produce teratomas, were killed and histological specimens of the brain, lung, kidney, spleen, and liver were analyzed of the animals. The presence of DNA from hIDPSCs was confirmed in all aforementioned organs but no tumor formation or any morphological alterations were observed. Thus, we established the safety of cell regeneration by investigational product CELLAVITATM (stem cells) regarding tumor formation and risk of immune rejection.
  • CELLAVITATM stem cells
  • FIG. 29 summarizes additional already published preclinical studies, which support the safety of investigational product CELLAVITATM (stem cells).
  • teratoma assay sensitivity and quantitativity; definitive cell number and single cell suspension production; immunophenotyping of studied cell in respect of expression on pluripotent cell markers and karyotype; co-transplantation of studied cells together with Matrigel.
  • the cells were transplanted subcutaneously (s.c) into NOD/SCID mice, which allows for simple monitoring of teratoma development.
  • Mouse embryonic stem cells b. Mouse 3T3 fibroblasts, permanent mouse cell line Balbc 3T3 cell line, clone A31 c. Human iPS-IDPSC d. Human ES cells e. Human IDPSCs
  • iPS-IDPSC immature dental pulp stem cells
  • Beltr ⁇ o-Braga et al., 2011 the human IDPSC were used as a control for iPS-IDPSCs.
  • iPS-IDPSCs formed nice teratomas with tissues originated from all three germ layers, while hIDPSC were not able to produce any type of teratomas or any other type of neoplasms.
  • iPS-IDPSCs expressed Nanog in nucleus, and hIDPSCs did not.
  • Disclosed multiharvest explant like culture used for the isolation of a population of immature dental pulp stem cells results in expression of embryonic stem cell markers Oct-4, Nanog, SSEA-3, SSEA-4, TRA-1-60 and TRA-1-81 as well as several mesenchymal stem cell markers during at least 15 passages while maintaining the normal karyotype and the rate of expansion characteristic of stem cells.
  • the expression of these markers was maintained in subclones obtained from these cells.
  • in vitro these cells can be induced to undergo uniform differentiation into smooth and skeletal muscles, neurons, cartilage, and bone under chemically defined culture conditions.
  • IDPSC although have a small size and cytoplasm poor in cell organelles differ from na ⁇ ve pluripotent cells presenting typical mesenchymal—fibroblast like morphology. Therefore IDPSC are of mesenchymal type, in contrast to ES and iPS cells, which are of epithelial type.
  • ES and iPS cells which are of epithelial type.
  • MSC and ES or iPS cells synthetize extracellular matrix and are cell junction free cells.
  • this method was validated using dog fetal stem cells from bone marrow, liver, yolk sac, allantois and amniotic liquid which also express pluripotent markers.
  • Culture conditions can affect gene expression of the cells.
  • genes include ABCG2 and Vimentin, which are two genes that can indicate suitability of the cells in culture for therapeutic use, particularly systemic cellular systems like the present invention.
  • Most suitable culture medium form tested is DMEM/F12 basal medium supplemented with 5-10-15% of FBS, antibiotics, and Glutamate, and the cells should be cultured without an adhesion layer (e.g. without extracellular matrix (ECM) or scaffold) such that the cells adhere directly to the culture dish or beads plastic.
  • ECM extracellular matrix
  • Cells that were grown with epithelial growth media conditions turn into epithelial-like cells.
  • ECM coating can be useful for scale up of current cells into 3D culture conditions including bioreactors, such as Terumo hollofiber bioreactor, Eppendorf—New Brunswick bioreactors with beads, etc.
  • FIGS. 26 and 27 The link of ABCG2 expression and undifferentiation status of cells is shown on FIGS. 26 and 27 where we demonstrated that once ABCG2 is not expressed due to changed culture medium or coating layer, then cells have clearly a more fibroblast or epithelial cells-like morphology.
  • DMEM Dulbeccos Modified Eagle Medium
  • KSFM serum knockout medium
  • AM Amniotic membrane
  • the culture medium was changed daily, and the cells were replaced every 3 days. After they reached 80% confluence, they were washed twice in sterile phosphate-buffered saline (PBS; Gibco; 0.01 M, pH 7.4), enzymatically treated with 0.25% trypsin/EDTA (Invitrogen), and seeded onto amniotic membrane previously prepared.
  • PBS sterile phosphate-buffered saline
  • trypsin/EDTA Invitrogen
  • SHEM hormonal epithelial medium
  • DMEM/F12 Dulbecco's Modified Eagle's Medium/Ham's F-12 nutrient mixture
  • Invitrogen Gibco Cell Culture, Port-land, OR; 1:1
  • 1.05 mM calcium supplemented with 5 ⁇ g/ml crystalline bovine insulin Sigma Aldrich, St.
  • the second was keratinocyte serum-free medium (KSFM) containing 0.09 mM calcium supplemented with 30 mg/ml pituitary bovine extract, 0.2 ng/ml EGF, 10% FBS, and ampicillin/streptomycin.
  • KSFM keratinocyte serum-free medium
  • Knockout media containing 0.06 mM calcium supplemented with 1% “human corneal growth supplement” (Cascade Biologics), containing 0.2% pituitary bovine extract, 5 g/ml bovine insulin, 0.18 mg/ml hydrocortisone, 5 ⁇ g/ml bovine transferrin, 0.2 ng/ml EGF
  • Mouse anti-human monoclonal antibodies ABCG2 (Chemicon) and cytoplasmic/nuclear monoclonal antibodies: mouse anti-cytokeratin 3/12 (K3/12) (RDI, Flanders, N.J., USA), reacts with human and rabbit.
  • Mouse anti-human IDPSC antibody was obtained as described (Kerkis et al., 2006) and successfully used by us in previous studies (Fonseca et al., 2009; Monteiro et al., 2009).
  • Microscope slides were mounted in antifade solution (Vectashield mounting medium, Vector Laboratories, Hercules, Calif., USA) with 4′,6-diamidino-2-phenylindole (DAPI) and analysed using a confocal microscope. Control reactions were incubated with PBS instead of primary antibody, followed by washing and incubation with respective secondary antibody. All experiments have been done in triplicate.
  • FIG. 26 depicts that IDPSC had differential response in respect of expression of studied proteins when cultured in distinct culture medium during 7 days.
  • the IDPSCs grown on plastic surfaces did not express ABCG2 when cultured in SHEM, KSFM, Epilife and DMEM/KO ( FIG. 26 ), this protein was expressed in IDPSCs only when they were cultured in basal culture medium ( FIG. 22 A 5 ).
  • IDPSCs cultured in SHEM and DMEM/KO after seven days changed their morphology fibroblast like ( FIG. 26 ) to epithelial like ( FIG. 26 ) and start to express CK3/12, while IDPSC cultured in Epilife and KSFM and DMEM/F12 did not start to express K3/12 ( FIG. 26 ).
  • IDPSCs did not react with IDPSCs cultured in DMEM/F12 e KSFM ( FIG. 27 ) and showed very weak immunopositivity with K3/12 when cultured in SHEM and DMEM/KO ( FIG. 27 ).
  • CD146 and CD13 expression were analyzed by flow cytometry in EP-hIDPSC and LP-hIDPSC.
  • the results in FIG. 30 show that CD13 was expressed in 52% of EP-hIDPSC and 95% of LP-hIDPSC.
  • IDPSC are migratory neural crest stem cells which are capable of aggregating into neurospheres. It is known that in development, SOX2 is regained only by those cells fated to become neurons. Neural crest stem cells that remain SOX2-free differentiate into other cell types, but never become neurons.
  • FIGS. 31A -A 4 demonstrate expression of SOX1 in the nucleus, while ⁇ -tubulin shows cytoplasmic localization in the neural precursors derived from LP of IDPSC, which are organized in neurospheres. More specifically, FIG. 31A depicts expression of Sox1 and ⁇ -tubulin proteins in late population (LP) of hIDPSC derived neurons. The following are also shown in FIGS. 31A -A 4 : FIG. 31A . a nucleus (white arrow) of neurons stained with DAPI; FIG. 31 A 1 . Positive immunostaining for SOX1 observed in the nucleus (white arrow) of neurons; FIG. 31 A 2 . Positive immunostaining for ⁇ -tubulin; FIG.
  • FIGS. 31A -A 2 and 31 A 4 Major magnification of inset in FIG. 31 A 3 demonstrates superposition of DAPI and Sox1 in the nucleus of neurons (white arrow).
  • FIGS. 31B-E demonstrate enrichment of LP of hIDPSC with neural progenitors and neurons following 15, 30 and 45 cycles of dental pulp mechanical transfer and induction of neural differentiation.
  • FIG. 31B a percentage of SOX1 negative cells, nucleus stained only with DAPI (blue) are shown; SOX1 positive cells are presented in green.
  • FIG. 31D a percentage of ⁇ -tubulin negative cells, nucleus stained only with DAPI (blue); ⁇ -tubulin positive cells are presented in red.
  • FIG. 32A demonstrates nucleus of IDPSC-neuroblasts positively immunostained with anti-BrdU antibody and neuronal bodies which react positively with ⁇ -tubulin class III.
  • FIGS. 32B-C show clearly the enrichment of differentiated IDPSC population with slightly differentiated neuroblasts following growing cycle numbers of DP mechanical transfer.
  • FIGS. 33A -A 3 Even more astonishing was the finding that neuronal progenitor cells derived from IDPSC at advanced passages (5-10) that already lack expression of SOX2 start re-expressing SOX2 during neurosphere formation ( FIGS. 33A -A 3 ). Efficiency of these cells' differentiation into neural cells, however, is lower than IDPSC at passages 1-3. Re-expression of SOX2 was more efficient in IDPSC obtained from DPs, which underwent a high number mechanical transfer cycles (>30) ( FIGS. 33B ,C).
  • LP IDPSC culture of post-natal cells of neural crest origin are characterized by unique properties. These unique properties are usually typical for neural precursors derived from fetal brain or from ES or iPS cells induced to neural differentiation in vitro.

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Cell Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Developmental Biology & Embryology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Immunology (AREA)
  • Virology (AREA)
  • Rheumatology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Hematology (AREA)
  • Dermatology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

The present disclosure relates to a cryopreserved pharmaceutical composition comprising immature dental pulp stem cells (IDPSCs) expressing SOX-1 and SOX-2 and methods of treating a neurological disease or condition comprising systemically administering to a subject a cryopreserved pharmaceutical composition comprising IDPSCs expressing SOX-1 and SOX-2.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • The present application is a Continuation of U.S. patent application Ser. No. 15/065,259, filed Mar. 9, 2016 (issued as U.S. Pat. No. 11,207,352), which claims priority to and is a Continuation-In-Part of U.S. patent application Ser. No. 14/214,016 filed Mar. 14, 2014 (issued as U.S. Pat. No. 9,790,468), which claims priority to U.S. Provisional Patent Application No. 61/791,594 filed Mar. 15, 2013, and to U.S. Provisional Patent Application No. 61/800,245 filed Mar. 15, 2013. U.S. patent application Ser. No. 15/065,259 also claims priority to U.S. Provisional Application Nos. 62/130,593, filed Mar. 9, 2015; 62/130,585, filed Mar. 9, 2015; and 62/220,792, filed Sep. 18, 2015. Each of the foregoing applications is hereby incorporated by reference in its entirety.
  • TECHNICAL FIELD
  • This application relates to a composition of stem cells for the treatment of neurological diseases suitable for systemic administration.
  • BACKGROUND
  • Even though the genes responsible of neurodegenerative diseases and its protein have been identified, the mechanism of pathogenesis involved in these diseases is still unknown, which precludes the development of efficient therapeutic interventions. What is currently known is that although it is ubiquitously distributed, the mutant form of Huntington protein, for example, causes neurodegeneration and selective loss of medium spiny neurons, which preferentially occurs in the striatum and in the deeper layers of the cerebral cortex during the early phases of the disease. Thus, cell therapy has been investigated as an additional or alternative treatment which may contribute positively on the course of this disease and other similar neurodegenerative diseases. Stem cells are the essential building blocks of life, and play a crucial role in the genesis and development of all higher organisms. Due to neuronal cell death caused, for example, by accumulation of the mutated huntingtin (mHTT) protein, it is unlikely that such brain damage can be treated solely by drug-based therapies. Stem cell-based therapies are important in order to reconstruct morphological design and functional ability of neural tissue in damaged brain areas in patients. These therapies used to have a dual role: transplanted stem cells paracrine action (anti-apoptotic, anti-inflammatory, anti-scar, anti-bacterial and angiogenic actions), which stimulates local cell survival, inhibits inflammation and brain tissue regeneration through the production of bioactive molecules acting in favor of new neurons production from the intrinsic and likely from donor stem cells.
  • The brain-derived neurotrophic factor (BDNF) is a gene responsible for BDNF protein expression found in the brain and spinal cord. This protein promotes the survival of nerve cells (neurons) by playing a role in the growth, maturation (differentiation), and maintenance of these cells. In the brain, the BDNF protein is active at the connections between nerve cells (synapses) where cell-to-cell communication occurs. The BDNF protein helps regulate synaptic plasticity, which is important for learning and memory and is found to be expressed in regions of the brain that control eating, drinking, and body weight. Thus, BDNF has additional action in modulating all these functions. Increasing evidence suggests that synaptic dysfunction is a key pathophysiological hallmark in neurodegenerative disorders, including Alzheimer's disease. The deficits in BDNF signaling contribute to the pathogenesis of several major diseases and disorders such as Huntington's disease and depression. Thus, manipulating BDNF pathways represents a viable treatment approach to a variety of neurological and psychiatric disorders. Administration of BDNF alone offers a viable approach to treating neurodegenerative diseases. However, it is difficult to find an ideal dose for each patient because of genetic and individual polymorphism of neurodegenerative diseases manifestation. Overdoses of BDNF could induce tumor formation in the brain; on the other hand low BDNF doses could not provide an efficient treatment. Stem cells after transplantation are under the control of the patient biology, which can modulate BDNF secretion by the cells efficiently for each patient. Additionally, the studies investigating the benefits of stem cell transplantation for treating Alzheimer disease demonstrated that transplanted nerve stem cells (NSC) support the formation of new connections between host brain cells. These studies demonstrate that strengthening these connections can reverse memory losses in Alzheimer disease mouse models. It seems that BDNF, a factor naturally secreted by NSC, can replicate the effects produced by stem cell transplantation.
  • Once NSC is generally difficult to access and cannot be obtained in sufficient therapeutic quantities to be applied in stem cell therapy through intravenous (IV) injection. Typically, two strategies are used to increase BDNF secretion. First, is the addition of growth factors into culture medium of in vitro cultured stem cells in order to induce BDNF secretion. However, this strategy has great limitations due to the fact that stem cells produce this factor only under in vitro conditions. Consequently, when such cells are transplanted to a patient they rapidly spend the “stock” of BDNF, which prevents long term treatment of neurodegenerative disease. Another approach is to produce genetically manipulated stem cells which are suitably modified to overexpress BDNF. It is important to note that even NSC need to be genetically engineered to produce therapeutically sufficient levels of BDNF. However, gene modification has its roots in gene therapy—an approach that still has to be proven. Therefore, there is a great need for new cell types and cell culture methods which can lead to stem cells with elevated secretion of BDNF.
  • The subventricular zone (SVZ) is the unique brain area where new neurons are produced throughout life (Altman J and Das G D 1965) and in generating cells to function in repair through adulthood. Blood vessels immediately subjacent to the SVZ run parallel to the direction of tangential neuroblast migration, and guide migratory neuroblasts via BDNF signaling. It is now understood that the organization of the SVZ in the adult human brain differs significantly from that of any other studied vertebrates. Specifically, this region in the adult human brain contains a unique tape of astrocytes that proliferate in vivo and can function as NSC in vitro. Astrocytes in the central nervous system perform many important and diverse functions. They are involved in formation of the neuro-vascular unit which is composed of a neuron, an astrocyte and a blood vessel. Astrocyte processes extend to and interact with blood vessels. Astrocytic endfeet are in intimate contact with the basal lamina that is a component of the vessel wall and together with endothelial cells they form the blood-brain barrier (BBB). Isolation of stem cells, which have a capacity to migrate and home in a neurogenic niche as well as around blood vessels in the adult human brain, further being able to differentiate into neurons and glial cells, is a basis for the development of novel neurodegenerative cell therapies.
  • Dopamine (DA) is a major neurogenesis factor in the adult SVZ (Baker et al., 2004). The proximity of the SVZ with the striatum makes it a neurodegeneration therapy target for striatum-neurodegeneration associated disorders such as Huntington's disease (HD) and Parkinson's disease (PD). Both pathologies are characterized by different clinical symptoms of motor dysfunction, and both are thought to involve the SVZ—striatum DA micro-circuitry path through different mechanisms. The disease-generated DA innervation that occurs in HD is a natural protective feedback mechanism to compensate for the striatal internal neurons degeneration pathology caused by inherent genetic mutation (Parent M et al., 2013). Dysregulation of DA receptor D2 is a sensitive measure for Huntington disease pathology in model mice (Crook et al., 2012; Chen et al., 2013). In contrast, PD is associated with massive degeneration of DA neurons, due to impaired neurogenesis in the nigrostriatal area and is a major cause of the pathology (Hoglinger et. al., 2004).
  • The initial inflammatory response occurs in the body to limit the invasion of foreign bacteria or viruses or parasites and to defend tissues against molecular foes which are further removed from the organism by anti-inflammatory mechanisms. However, chronic inflammation (CI) is a double-edged sword. CI is long lasting event and it continuously harms and kills healthy cells as, for example, in rheumatoid arthritis where the inflammation becomes self-sustaining.
  • In neurodegenerative diseases several molecules of the protein are tightly aggregated together inside the cell, which pathologists call an “amyloid” structure, and they are apt to clog the brain. Such proteins were found in Alzheimer's disease—amyloid beta and tau; in Parkinson's disease—alpha synuclein, and in Huntington's disease—huntingtin. These aggregates often form large insoluble deposits in the brain. However, the truly toxic ones are considered the small, soluble aggregates of these proteins. Due to the accumulation of these aggregates in the brain, chronic inflammatory reactions remained in many age-related neurodegenerative disorders among which are aforementioned diseases (Nuzzo et al., 2014).
  • Degenerated tissue, the presence of damaged neurons and neurites, highly insoluble amyloid β peptide deposits, and neurofibrillary tangles in the brains of Alzheimer disease (AD) carriers provide obvious stimuli for inflammation (Zotova et al., 2010; Schott and Revesz, 2013).
  • Many studies have suggested that the chronic inflammation observed in AD accelerates the disease process and may even be a disease trigger. A history of head injury and systemic infections are factors, which typically cause brain inflammation and are known to be risk factors for AD. Excessive action of the brain's immune cells, which are glial cells, is another hallmark of Alzheimer's disease. Although it has been suggested that inflammation is associated with injury and toxicity to neurons, the relationship among glial cells, neurons and amyloid plaques still remains unclear. Inflammatory mediators released by glial cells can be extremely toxic to neurons. Thus, they have been considered as mediators of neurodegeneration.
  • Two closely related inflammation-promoting proteins, IL-12 and IL-23, are among those pumped out by microglia when the cells become immunologically active. The studies demonstrated that these proteins exist at elevated levels in the cerebrospinal fluid of AD patients. Blocking these inflammatory proteins in older Alzheimer's mice whose brains were already plaque-ridden reduced the levels of soluble, more toxic forms of amyloid beta and reversed the mice's cognitive deficits (Vom Berg et al., 2012; Griffin, 2013).
  • More recently, other anti-inflammatory approaches such as the blocking of a protein NLRP3 and microglial protein MRP14 have been described and also seem to work well in the same Alzheimer's mouse model. These approaches reduce brain inflammation, amyloid beta deposition, and cognitive impairments. In Alzheimer's mice that were genetically engineered to lack NLRP3, microglia were reversed back towards a non-inflammatory state in which they consume much more amyloid beta and secrete neuron beneficial proteins. In another study, a microglia protein MRP14 was targeted, which also helped to reverse microglia to a non-inflammatory state (Heneka et al., 2013; Zhang et al., 2012).
  • The other factor which is critical for AD is aging. Aging may help trigger Alzheimer's by worsening common age-related problems with neurons, which become functionally deficient and lose their ability to transport and appropriately place proteins. Inflammation worsens this problem by increasing the production of amyloid-beta in inflamed regions, stressing neurons, and hastening the age-related decline of their protein-transport and disposal systems. Inflammation reactivates microglia into an inflammatory state and thus reduces their ability to clear up the brain (Swindell et al., 2013).
  • Currently, it is assumed that inflammation helps to start the AD process by increasing the production of amyloid beta. The inflammation seems to be self-sustaining in AD because it reduces the ability of microglia to remove amyloid beta. Therefore, constant deposition of amyloid beta does not allow the inflammation to resolve, which gets worse in aged AD carriers (Akiyama et al., 2000; Vom Berg et al., 2012; Zang et al., 2012; Griffin, 2013; Heneka et al., 2013; Swindell et al., 2013; Schott and Revesz, 2013).
  • The contribution of inflammation to neurodegeneration in Huntington disease (HD) is strongly suggested; however it is less studied then in AD (Soulet and Cicchetti, 2011; Ellrichmann et al., 2013). Thus, an activation of the immune system in HD was clearly proven by the elevated expression of pro-inflammatory cytokines, which are crucial to the body's immune response, such as, IL-6 and TNF-alpha. These pro-inflammatory cytokines were significantly increased in the striatum, plasma and CSF in mouse models and in symptomatic as well as presymptomatic HD patients. Additionally, innate immune cell hyperactivity was detected through elevated IL-6 production in mutant mHTT expressing myeloid cells of the central (microglia) and peripheral innate immune system (monocytes and macrophages) both in HD patients and mouse models. It has also been reported that abnormally high levels of cytokines were present in the blood of people carrying the HD gene many years before the onset of symptoms (Bjorkqvist et al., 2008; Trager et al., 2014a, b). The composition of cytokines and levels of their expression, which can be measured in a blood of patients, could be useful to establish the need to initiate intervention for therapies as well as the timing of therapies.
  • Blood cells, due to the presence of the abnormal HD protein (huntingtin) inside the cells, were hyperactive in HD patients, as well as microglia in the brain, thus suggesting that abnormal immune activation could be one of the earliest abnormalities in HD. The patient's blood signature could provide a new insight into the effects of the HD in the brain as well as markers of HD severity. Anomalous immune activation could be a target for future treatments aimed at slowing down HD (Soulet and Cicchetti, 2011, Ellrichmann et al., 2013).
  • Parkinson's disease is characterized by a slow and progressive degeneration of dopaminergic neurons in the substantia nigra. Using animal models researchers have obtained consistent findings about involvement of both the peripheral and the central nervous system immune components in response to inflammation, which initiates an immune response in PD. The presence of continuing and increasing pro-inflammatory mechanisms results in a process whereby cellular protective mechanisms are overcome and the more susceptible cells, such as the dopaminergic neurons, enter into cell death pathways, which leads to a series of events that are a crucial for the progression of PD (Doursout et al., 2013). Inflammatory responses also manifested by glial reactions, T cell infiltration, and increased expression of inflammatory cytokines, as well as other toxic mediators derived from activated glial cells, are well known features of PD. More recent in vitro studies, however, proposed that activation of microglia and subsequently astrocytes via mediators released by injured dopaminergic neurons is involved, even though they are unlikely to be a primary cause for neuronal loss (Hirsch et al., 2003). In patients the epidemiological and genetic studies support a role of neuroinflammation in the pathophysiology of PD. Post mortem studies confirm the involvement of innate as well as adaptive immunity in the affected brain regions in patients with PD. Activated microglial cells and T lymphocytes have been detected in the substantia nigra of patients concomitantly with an increased expression of pro-inflammatory mediators (Tufekci et al., 2012; Hirsch et al., 2012). Another study, which enrolled 87 Parkinson's patients between 2008 and 2012, together with 37 healthy controls measured markers of inflammation such as C-reactive protein (CRP), interleukin-6, tumor necrosis factor-alpha, eotaxin, interferon gamma-induced protein-10, monocyte chemotactic protein-1 (MCP-1) and macrophage inflammatory protein 1-beta in routine blood tests. All participants underwent physical exams as well. This study demonstrated that the degree of neuroinflammation was significantly associated with more severe depression, fatigue, and cognitive impairment even after controlling for factors such as age, gender and disease duration (Lindqvist et al., 2013). Neuroinflammatory processes might represent a target for neuroprotection, and anti-inflammatory strategies may be one of the principal approaches in the treatment of PD.
  • Multiple approaches have been tested to repair neurodegeneration-associated CNS diseases, including clinical motor dysfunction diseases (Wernig M, et al., 2011). Stem cells sources used for neuro-regeneration cell therapy include mesenchymal stem cells (MSC), neural progenitor cells (NP), human fetal neuronal stem cells (huNSC), and pluripotent stem cells (both embryonic (ESCs) and induced (iPSC)). Most of the studies on cell therapy for neurological conditions used neuronal-like cells through major cellular manipulation and/or highly invasive methods of delivery. For example, WO 2008/132722 and US Patent Application Publication No. 2013/0344041 disclose genetically manipulated stem cells to induce stem cell traits or to release neurotrophic factors; WO 2009/144718 and US Patent Applications Publication Nos. 2014/0335059 and 2014/0154222 disclose inducing the release of neurotrophic factors at levels higher than at non-induced stage via exposure to biological, natural or chemical compounds in culture; and other studies use immortalized cell line of fetal stem cells that express early markers of neuronal differentiation. In spite of the studies on stem cell therapy, no data have shown that stem cell therapy through intravenous (IV) injection can result in direct neurogenesis via BDNF secretion or D2 expression in brain compartments suffering from neurodegenerative disease.
  • SUMMARY OF THE INVENTION
  • In one embodiment, the invention is directed to a pharmaceutical composition comprising stem cell, wherein the stem cells comprising late harvest enriched from tissue of neural crest origin. In some implementations, the tissue of neural crest origin is dental pulp. In some aspects, the stem cells enriched from tissue of neural crest origin are immature dental pulp stem cells (IDPSCs). Early harvest stem cells enriched from tissue of neural crest origin comprise IDPSCs of the first fifteen or the first 25 harvest cycles whereas late harvest stem cells comprise IDPSCs from the sixty or later or the 26th or later harvest cycle.
  • In some embodiments, the invention is directed to a pharmaceutical composition produced by a method comprising: extracting dental pulp (DP) from a tooth; culturing the DP in basal culture medium in a first container to establish a DP explant culture, wherein the DP explant culture is cultured without or with at least one extracellular matrix components selected from the group consisting of: fibronectin, collagen, laminin, vitronectin, polylysine, heparan sulfate proteoglycans, and enactin; mechanically transferring the DP to a second container to establish a second DP explant culture; repeating the step of mechanically transferring the DP until at least 15 DP explant cultures have been established; passaging the DP explant culture to produced a passaged DP culture; and combining the passaged DP culture of an early harvest population and an late harvest population to produce the pharmaceutical composition, wherein the early harvest population comprises passaged DP culture established from at least one of the first 15 DP explant cultures and the late harvest population comprises passaged DP culture established from at least one of the DP explant cultures after the 15th DP explant culture. In some implementations, the culturing step occurs under hypoxic conditions. In some implementations, the step of combining the passaged DP culture of the early harvest population and the late harvest population to produce the pharmaceutical composition comprises: simultaneously thawing the frozen stock of passaged DP cultures of the early harvest population and the late harvest population; and pooling the thawed passaged DP culture to produce the pharmaceutical composition.
  • For some embodiments of the pharmaceutical invention, culturing the DP in basal culture medium in the method of production persist for at least three days before the DP is mechanically transferred. In some implementations, the method of production further comprise creating a frozen stock of the passaged DP culture. In some aspects, the frozen stock of the passaged DP culture is created at the third passage of the DP explant culture.
  • In some embodiments, the invention is directed to a pharmaceutical composition produced by a method comprising: extracting dental pulp (DP) from a tooth; culturing the DP in basal culture medium in a first container to establish a DP explant culture, wherein the stem cells comprising late harvest enriched from tissue of neural crest origin are double positive for CD44 and CD13. In some aspects, the stem cells enriched from tissue of neural crest origin and double positive for CD44 and CD13 are immature dental pulp stem cells (IDPSCs).
  • In some embodiments, the invention is directed to a pharmaceutical composition produced by a method comprising: extracting dental pulp (DP) from a tooth; culturing the DP in basal culture medium in a first container to establish a DP explant culture, wherein the stem cells comprising late harvest enriched from tissue of neural crest origin demonstrated increasing level of secretion of endogenous BDNF and/or other neurotrophic factors (NF3, NF4 and NF5), when compared to stem cells obtained from early harvest. In some aspects, the stem cells enriched from tissue of neural crest origin and secreting high level of endogenous BDNF and/or other neurotrophic factors (NF3, NF4 and NF5) are immature dental pulp stem cells (IDPSCs).
  • The present invention relates to pharmaceutical compositions for systemic administration to a subject to treat a neurological condition. The neurological disease or condition may be a neurodegenerative disease or condition, autism, schizophrenia, epilepsy, stroke, ischemia, a motor disorder, or a convulsive disorder. Neurodegenerative disease or condition may be Parkinson's disease (PD), multiple sclerosis, epilepsy, amyotrophic lateral sclerosis (ALS), stroke, autoimmune encephalomyelitis, diabetic neuropathy, glaucomatous neuropathy, Alzheimer's disease, or Huntington's disease (HD).
  • In some implementations, the pharmaceutical compositions comprise stem cells from tissue of neural crest origin. Tissue of the neural crest origin may be dental pulp but not limited by this example.
  • In one embodiment, the pharmaceutical composition comprises stem cells expressing at least one safety markers selected from the group consisting of ATP-binding cassette sub-family G member 2 (ABCG2), p53, and inactive nanog. Inactive nanog is expressed nanog localizing predominantly in the cytoplasm of the stem cell. In some aspects, at least 75% of the stem cells express ABCG2, at least 75% of the stem cells express p53, or no more than 5% of the stem cells express inactive nanog. Some pharmaceutical compositions may comprise stem cells that further express the safety marker SOX2. In some such embodiments, no more than 30% of the stem cells express SOX2.
  • The stem cells of the pharmaceutical composition may further secrete at least one marker selected from the group consisting of brain-derived neurotrophic factor (BDNF), neutrotrophin-3 (NT3), neutrotrophin-4 (NT4), neutrotrophin-5 (NT5), and p75. In some such embodiments, the stem cells of the pharmaceutical composition express BDNF, NT3, NT4, NT5, and p75 (CD271).
  • In another embodiment, the pharmaceutical composition comprises stem cells expressing at least one neuroepithelial stem cell marker selected from the group consisting of BDNF, NT3, NT4, NT5, and p75. In some aspects, these pharmaceutical compositions comprise undifferentiated stem cells expressing BDNF, NT3, NT4, NT5, and p75. These pharmaceutical compositions may further express at least one safety markers selected from the group consisting of ABCG2, inactive nanog, p53, and SOX2. In some aspects, at least 75% of stem cells express the at least one marker when the at least one marker is ABCG2. In some aspects, at least 75% of the stem cells express p53. In some aspects, no more than 5% of the stem cells express inactive nanog. In some aspects, no more than 30% of the stem cells express SOX2.
  • In some aspects, the stem cells of the pharmaceutical composition are immature dental pulp stem cells (IDPSCs).
  • The present invention also relates to methods of using the pharmaceutical composition comprising systemically administering to a subject the pharmaceutical compositions of the invention. Such methods may be used for treating neurological diseases or conditions, for example by supporting the natural neuro-protective mechanism in subjects diagnosed with early HD or repairing lost DA neurons in subjects diagnosed with PD. The methods may also be used as a preventive therapy for subjects at risk of HD.
  • In some implementations, the methods further comprise measuring the amount of DA receptor in the subject. In some aspects, the DA receptor is receptor D2. In some embodiments, measuring the amount of DA receptor, for example receptor D2, in the subject comprises imaging the subject to detect presence of DA receptor.
  • In some aspects, the subject is administered a single administration or a first and second administrations of the pharmaceutical composition. The repetitive (from 1 to six) administrations of the pharmaceutical composition take place at least seven days, at least 14 days, at least 21 days, or at least 30 days after the first administration, as well as one time per year, or twice per year. In some implementations, administration of the pharmaceutical composition is through intravenous injection. In some implementations, the administration of the pharmaceutical composition may be repeated annually, for example, repeated three times.
  • In one embodiment of the methods, the neurological disease or condition is treated by the stem cells crossing the blood/brain barrier (BBB) and inducing neurogenesis. In some aspects, the stem cells are directly transplanted into the brain parenchyma, including striatum, following crossing of the BBB. In some embodiments, the induced neurogenesis is dopamine-associated. For example, dopamine-associated neurogenesis occurs through self-differentiation of the stem cells or activation of migration and differentiation of intrinsic stem cells by the extrinsic stem cells. In some aspects, massive dopamine-associated neurogenesis takes place in the subventricular zone (SVZ).
  • In some embodiments of the methods, the pharmaceutical composition provides neuroprotection. For example, systemic neuroprotection is provided with the high basal level of neurotrophic and immunoprotective factors expression and release pattern of the stem cells of the pharmaceutical composition. In some aspects, these stem cells of the pharmaceutical composition are IDPSCs.
  • In some implementations of the methods, the pharmaceutical composition comprises stem cells that are autologous to the subject. For example, the stem cells that are autologous to the subject are collected from the subject before disease manifestation from contingently “healthy cells”, once these diseases are genetic and age dependent. In other implementations, the pharmaceutical composition comprises stem cells that are allogeneic to the subject regardless of whether the major histocompatibility complex expressed by the stem cells match the major histocompatibility complex expressed by subject. The pharmaceutical composition may also comprise a combination of stem cells that are autologous to the subject and stem cells that are allogeneic to the subject.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 depicts a scheme for the manufacturing process of compositions of IDPSCS comprising both early and late population IDPSC suitable for the treatment of neurological diseases and conditions.
  • FIG. 2 depicts an early phase development process for CELLAVITA™ (stem cells) isolation and batch formulation.
  • FIG. 3 depicts another early phase development manufacturing process for CELLAVITA™ (stem cells).
  • FIG. 4 depicts a CELLAVITA™ (stem cells) production process composed of several major steps.
  • FIG. 5 depicts immunophenotyping of hIDPSC from early and late harvests. Harvests 0-10 were defined as early harvests. All harvests after 10 were defined as late harvests.
  • FIG. 6 depicts IDPSC at late harvest (13) at passage 3 were immunopositive for P75 (CD271) (A, C, D), nestin (E-G), CD13 (H) and CD73 (I), and they did not react with CD146 (J) and HLA-ABC (K). Insets, demonstrate control for respective secondary antibodies. A-G Light Microscopy. H-L Epi-Fluorescence. Magnification: A,J-20×; C,E-G,H,I,L-10×; D,K-40×
  • FIG. 7 depicts in a) CFU-F assay performed in triplicate at T20, passage 3, demonstrating high clonogenic capacity of a LP population of IDPSCs. In b) and c) FACS analysis performed to show that LP of IDPSCs (batch #11) comprise approximately 80% cells that express BDNF and DARP 32 while EP is negative for these markers (data not shown) and comprises a very low number of the cells which express D2.
  • FIG. 8A depicts positive immunostaining for BrdU (B-J) in control cells with a secondary antibody. FIGS. 8B and 8C depict quantification of LP IDPSC which react positively with an BrdU antibody. (A)—Epi-fliorescence, (B) amd (C)—FACS analysis. Magnification (A)—200×. (B, E and H)—400×. (C,F,I,D,G and J)—1000×.
  • FIG. 9 depicts quantitative PCR for expression of endogenous Oct4, Nanog and Sox2 genes observed in hIDPSCs before (black color) and after reprogramming (white color) as well as in human embryonic stem cells (hESC) (striped).
  • FIG. 10 depicts quantification of expression of GFAP (glial fibrillary acidic protein) and beta-III-tubulin in EP (early population) and LP (late population) IDPSC with flow cytometry.
  • FIG. 11 depicts a time line of stability studies of hIDPSC during long term cryopreservation, following thawing and shipping before local of application in animal models of human diseases.
  • FIG. 12 depicts the timeline of the pilot study in the HD disease model (groups I, II, III, and IV). HD was induced during the first four days, day 0 (D0) to day 4 (D4) by 3-NP. On the fifth day (D5), IDPSC transplantation was administered via intravenous injection. Animals were euthanized on day 9 (D9) followed by brain tissue fixation and histological analysis of lesion for detection of IDPSC biodistribution and engraftment (Vybrant+immunohistochemistry using specific antibodies). Group I and III were euthanized on day 35 (D35) and groups II and IV on day 95 followed by brain tissues fixation and histological analysis of lesion for detection of IDPSC biodistribution and engraftment (Vybrant+immunohistochemistry using specific antibodies).
  • FIG. 13 presents localization of hIDPSC in rat brain tissue four days after hIDPSC administration. hIDPSC cells were stained green (Vybrant) and nuclei were stained red (PI) (A1-A4). Cells were localized mainly in capillaries and two morphological types were observed: neuron-like cells and pericytes. Note the different localization of pericytes in capillaries in A2, A3, and A4; in A4, hIDPSCs are localized in the axon bifurcation. A5: neuron morphology in brain tissues (A2-A4); A6: schematic figure of brain capillary showing pericyte localization. Confocal microscopy. Epifluorescence+Digital interference contrast (DIC) microscopy. Scale bar=10 μm.
  • FIG. 14 depicts the engraftment of hIDPSCs four days after IV administration. Optical cut demonstrates hIDPSC stained with Vybrant (green) and positively reacted with anti-hIDPSC antibody (red). Superposition of both produces yellow color. The cells demonstrate near capillary localization. Two markers for MSC were used: CD73 and CD105 demonstrating positive reaction with hIDPSC (A-D). E. Positive control hIDPSC cultured in vitro. Confocal microscope. Epifluorescence+Digital Interference contrast (DIC). Scale bar: A=5 μm; B=10 μm; C=20 μm; D=5 μm.
  • FIG. 15 depicts immunohistochemical images showing positive anti-human nuclei (hNu) staining of hIDPSCs and their localization in rat brain tissue 30 days after hIDPSC administration. Note: a few hIDPSCs in the cortex (left) and a large number of hIDPSCs in the corpus striatum (right). Light microscopy. 90× magnification. Scale bars: 5 μm and 25 μm, respectively.
  • FIG. 16 shows immunohistochemical image of rat brain tissue after 3-NP Injection and hIDPSC administration. Note: positive anti-human nuclei (hNu) immunostaining in cells. Neuron-like cells are circled in blue, and fibroblast-like cells are circled in white. Light microscopy, 90× magnification.
  • FIG. 17 depicts Nissl Staining in the Striatum of Untreated Animals (3-NP+saline) (a-b1); Control Animals (no 3-NP or hIDPSC) (c, c1); and Treated Animals (3-NP+hIDPSC) (d-f1). Different scores were observed in the experimental groups: score 1 (a, a1, d, and d1); score 2 (b, b1, e, and e1); and score 3 (c, c1, f, and f1). Area of extensive degeneration (a, a1); severe (d, d1), moderate (b, b1, e, and e1), mild (f, f1), and no (c, c1) neuron loss. Magnification: 10× (a-f) and 20× (a-f1). Insets in (b, c, e, and f) show typical Nissl-stained neuron morphology (40×). Light microscopy (a-f).
  • FIG. 18 depicts DARPP32 immunostaining in the corpus striatum of untreated animals (3-NP+saline) (a-b), Controls (no 3-NP or hIDPSC) (c), and Treated Animals (3-NP+hIDPSC) (d-e1). In (a) no immunostaining (blue arrow, score 1), (b) few DARPP32+ cells (score 2), and (c) Control animals (no 3-NP or hIDPSC) showing positive DARPP32 immunostaining (score 3). In (d, d1), neuron loss in Treated animals (3-NP+hIDPSC), with few DARPP32-stained cells (score 2) (black arrow). In (e, e1), strong anti-DARPP-32 immunostaining (score 3). Insets (a, b, c, d1) show DARPP32+ neurons (black arrow) (40×). HE (hematoxylin and eosin)-stained nuclei in blue. Magnification: 10× (a, c, d, and e) and 20× (d, e1).
  • FIG. 19 depicts neuronal growth in the striatum of rats after hIDPSC. Administration of hIDPSC resulted in a neuroreparative effect in hIDPSC-treated animals by (A) Nissl staining and (B) DARPP32 expression. (C) Number of animals showing neuron recovery after hIDPSC administration compared to Controls. Most hIDPSC-treated animals (3-NP+hIDPSC) had scores 3 and 2 (moderate and mild), whereas most Untreated animals (3 NP+saline) had scores 2 and 1 (severe and moderate).
  • FIG. 20 depicts positive DARPP32 immunostaining for neurons in CELLAVITA™ (stem cells)-treated animals 30 days after hIDPSC transplantation. Untreated animals (3-NP+saline) showed no DARPP32 immunostaining in the striatum or cortex. Rat neuron production was observed in the cortex and striatum of hIDPSC-treated animals. Light microscopy. Magnification: 20× and 90×.
  • FIG. 21 shows Dopamine Receptor D2 expression in the striatum of rats before and 30 days after hIDPSC Administration; Scores 3 and 2: striatum of untreated animals (3-NP+saline), showing a few D2 receptors in score 2. Score 1: striatum of treated-animals (3-NP+hIDPSC), showing positive receptor D2 immunostaining. Inset: high magnification showing positive D2 immunostaining and neuron morphology.
  • FIG. 22 depicts BDNF expression in rat brain tissue after (a-f) 3-NP Injection and (g-j) CELLAVITA™ (stem cells) administration (a, b) Absence of BDNF expression seven days after 3-NP injection and (c, d) low expression after 30 days (e, f). Control animals (no 3-NP or CELLAVITA™ (stem cells)). BDNF expression 7 (g, h) and 30 (i, j) days after CELLAVITA™ (stem cells) administration. Magnification: 10× (a, c, e, f, g, and h) and 20× (b, d, i, and j).
  • FIG. 23 depicts DARP32 expression in the striatum of rats 30 days after CELLAVITA™ (stem cells) administration in a 3-NP model of HD. Confocal microscopy, overlapping images in A. Epifluorescence+Digital interference contrast (DIC) microscopy. B-D: Epifluorescence. Scale bar: 10 μm.
  • FIG. 24 depicts the effect of hIDPSC administration on body weight in treated (3-NP+hIDPSC) and untreated animals (3-NP+Saline). Body weight was recorded before and 4 days after 3-NP administration and after each hIDPSC administration (every 30 days). No increase in body weight was observed in 3-NP-treated animals 30, 60, and 90 days after 3-NP administration. Body weight in hIDPSC-treated animals (1×106 dose) increased after the first hIDPSC administration.
  • FIG. 25 depicts intracellular expression of Oct4, Nanog, and Sox2. Nuclei were stained with DAPI.
  • FIG. 26 depicts expression of ABCG2 and K3/12 in IDPSC grown on various types of cell culture media.
  • FIG. 27 depicts expression of Vimentin, ABCG2, and K3/12 in IDPSC grown on various types of cell culture media.
  • FIG. 28 depicts pharmacological efficacy studies of investigational product CELLAVITA™ (stem cells).
  • FIG. 29 depicts safety studies of investigational product CELLAVITA™ (stem cells).
  • FIG. 30 depicts a flow cytometry analysis of early phase (EP) and late phase (LP) hIDPSC. Changes in CD146 and CD13 expression were observed following in vitro DP harvesting and hIDPSC passing. For EP-hIDPSC ˜33% of CD146 positive cells were observed, while for LP-hIDPSC less than 1% of the cells were positive for this marker. For EP-hIDPSC ˜52% of CD13 positive cells were observed, while for LP-hIDPSC 95% of the cells were positive for this marker.
  • FIGS. 31A-A4 depict expression of SOX1 and β-tubulin proteins in late population (LP) of hIDPSC derived neurons. FIG. 31A. Nucleus (white arrow) of neurons stained with DAPI; FIG. 31A2. Positive immunostaining for SOX1 observed in the nucleus (white arrow) of neurons; FIG. 31A3. Positive immunostaining for β-tubulin; FIG. 31A3. Merged images FIGS. 31A-A2, FIG. 31A4. Major magnification of inset in A3 demonstrates superposition of DAPI and SOX1 in the nucleus of neurons (white arrow). Epifluorescence, Scale bar=5 μm.
  • FIGS. 31B-E depict enrichment of LP of hIDPSC with neural progenitors and neurons following 15-, 30- and 45 cycles of dental pulp mechanical transfer and induction of neural differentiation. A. Percentage of SOX1 negative cells, nucleus stained only with DAPI (blue). SOX1 positive cells are presented in green. B. Percentage of β-tubulin negative cells, nucleus stained only with DAPI (blue). β-tubulin positive cells are presented in red.
  • FIG. 32A demonstrates a nucleus of IDPSC-neuroblasts positively immunostained with anti-BrdU antibody and neuronal bodies, which reacts positively with β-tubulin class III. FIGS. 32B-C show the enrichment of differentiated IDPSC population with slightly differentiated neuroblasts following growing cycle numbers of DP mechanical transfer.
  • FIGS. 33A-A3 depict enrichment of LP of hIDPSC with neural progenitors and neurons following 15-, 30- and 45 cycles of dental pulp mechanical transfer and induction of neural differentiation. BrdU positive cells present red and SOX2 green nucleus, which are also stained with DAPI (blue). FIGS. 33B-C demonstrate percentage of BrdU and SOX2 positive cells shown in relation with nucleus stained with DAPI (blue).
  • DETAILED DESCRIPTION
  • As used herein, the verb “comprise” as is used in this description and in the claims and its conjugations are used in its non-limiting sense to mean that items following the word are included, but items not specifically mentioned are not excluded. In addition, reference to an element by the indefinite article “a” or “an” does not exclude the possibility that more than one of the elements are present, unless the context clearly requires that there is one and only one of the elements. The indefinite article “a” or “an” thus usually means “at least one”.
  • As used herein, the term “high expression” in reference to the expression level (strongly immunopositive for antigen of interest) of a gene in a population of cells refers at least 75% of the population expressing the gene.
  • As used herein, the term “low expression” in reference to the expression level of a gene in a population of cells refers no more than 30% of the population expressing the gene. In preferred embodiments, low expression refers no more than 25% of the population expressing the gene.
  • As used herein, the term “no expression” in reference to the expression level of a gene in a population of cells refers no detectable cells that express the gene of interest in the population. No detectable expression includes an expression level that is within the realm of error for the method of measuring expression.
  • As used herein, the term “co-express” refers to the simultaneous detection of two or more molecular markers, e.g., SOX1 and SOX2, and beta-3-tubulin in same cell population, preferably in/on same cell.
  • As used herein, the term “subject” or “patient” refers to any vertebrate including, without limitation, humans and other primates (e.g., chimpanzees and other apes and monkey species), farm animals (e.g., cattle, sheep, pigs, goats and horses), domestic mammals (e.g., dogs and cats), laboratory animals (e.g., rodents such as mice, rats, and guinea pigs), and birds (e.g., domestic, wild and game birds such as chickens, turkeys and other gallinaceous birds, ducks, geese, and the like). In some implementations, the subject may be a mammal. In other implementations, the subject may be a human.
  • As used herein, the term, “stem cell” refers immature, unspecialized cells that, under certain conditions, may differentiate into mature, functional cells.
  • As used herein, the term, “neural stem cell” or “NSC” refers to multipotent cells that self-renewable and able to terminally differentiate into neurons, astrocytes, and oligodendrocytes.
  • As used herein, the term “neural progenitor cells” refer to undifferentiated cells further along the stage of cell differentiation than neural stem cells. Thus these cells are derived from neural stem cells and can produce progeny that are capable of differentiating into more than one cell type of central nervous system (CNS) and peripheral nervous system (PNS).
  • As used herein, the term “neural precursor cell” or “NPC” refers to a mixed population of cells consisting of neural stem cells and all of its undifferentiated progeny. Therefore NPCs include both NPC and NSC. The NPCs can also be categorized into neuronal NPCs and glial NPCs, which produce neurons and glial cells, respectively.
  • As used herein, the term “harvest cycle” constitutes a transfer of the orgão (e.g. of neural crest origin like dental pulp) or tissue to a new cell culture container after adherence and outgrowth of the stems cells in the tissue followed by preservation (e.g. cryopreservation) and/or sub-culturing of the outgrowth of IDPSCs. Stem cells isolated from the first time from organ culture (dental pulp) using explant technology are early populations, thus stem cells isolated from the first harvest cycle (first transfer of organ culture) are early population cells. For example, of stem cells isolate from dental pulp tissue, stem cells from human exfoliated deciduous teeth (SHED) can not be divided in early and late population, as these cells are isolated using enzymatic method only once from dental pulp, which are descarted after SHED isolation. Thus, only one SHED cell population can be isolated. Further, following enzymatic digestion, SHED can be passed from one to other cell culture flask thus counting cell passages, which generally are performed when SHED reach semi-confluence.
  • As used herein the term “neuronal biomarkers” SOX1 (for sex determining region Y-box 1) refers to a transcription factor which is involved in early central nervous system development. SOX1 may be expressed particularly in the ventral striatum.
  • As used herein, the term “SOX2” refers to a neural progenitor and sternness marker that is a transcription factor expressed by self-renewing and multipotent stem cells of the embryonic neuroepithelium (9). SOX2 may be expressed by actively dividing neural progenitor cells in the neurogenic regions in the adult rat brain and may also be expressed by glial fibrillary acidic protein immunopositive astroglia, widely distributed in the brain parenchyma (10). SOX2 is also known, in conjunction with Oct4, SOX2 as essential transcription factor for pluripotency (Ivanona et al., Nature 442:5330538 (2006); Methods of preparing induced pluripotent stem cells from mouse are also known (Takahashi and Yamanaka, 2006). Induction of iPS cells typically require the expression of or exposure to at least one member from Sox family and at least one member from Oct family.
  • As used herein, the terms “class III β-tubulin”, “beta-3-tubulin”, “microtubule-associated protein 2 (MAP-2), or neurofilament” refer to microtubule elements expressed exclusively in neurons and serves as a specific neuronal promoter, characteristic of neurons phenotypic marker of early neuronal precursors.
  • In contrast to SHED for immature dental pulp stem cells (IPDSCs) enzymatic method is not used. IDPSCs can be isolated as from explant culture of the dental pulp after the first adherence of DP to plastic and cells outgrowth—early population cells. At this stage dental pulp is not descarted and used for subsequent explant dental pulp cultures—late populations. Thus, IDPSC isolated from the second or later harvest cycle are late population cells. For example, IPDSCs that are isolated from the second harvest cycle or later are late population undifferentiated stem cells. As used herein, the term “early passage” refers to the cells from the first five passages of an explant culture. As used herein, the term “late passage” refers to the cells from passages after the fifth passage, e.g. in the sixth passage or later, of an explant culture. Thus, the IDPSC may be from an early or late population and additionally categorized as an early or late passage.
  • The present invention relates to the discovery that a particular composition of IDPSC, the unique stem cells population, which composed by early and late stem cells populations, from tissue of neural crest origin can cross the blood/brain barrier (BBB) and induce neurogenesis. Tissue of neural crest origin include, for example, dental, periodontal, and hair follicular tissue. Hair follicular tissue includes follicular tissue of the vibrissa.
  • The hIDPSC was evaluated in the 3-NP (three nitropropionic acid) HD rat model. The hIDPSC showed engraftment into the rat brain after 1 month following intravenous injection of 1×106 and 1×107 cell/transplant (3×106 cell/kg and 3×107 cell/kg) labeled with fluorescent protein (Vibrant), as well as, following immunohistochemistry analysis using specific anti-human antibody. Cell engraftment was observed in different brain compartments (cortex, striatum and Subventricular zone-SVZ).
  • The ability to cross the BBB enables systemic administration (e.g. IV administration) of stem cell therapy to treat neurological conditions, which provides a significant advantage over more localized methods of administration. In addition to systemically administration being less invasive, leaving stem cells to migrate to locations that require aid reduces the risk of harmful cell masses developing at the site of administration. For example, while intrathecal (IT) administration of stem cell therapy is commonly contemplated in preclinical and clinical studies, this method can have significant risk when the stem cells are MSCs. It has been reported that depending on cellular density, bone marrow-derived MSCs that were drawn into brain parenchyma (presumably in response to chemoattractant signals from this inflammation) via intracerebroventricular (ICV) formed cellular masses in 64% of severe experimental allergic encephalomyelitis animals. Karyotypically normal MSCs at early passages also induced masses in naïve animals (Grigoriadis et al., 2011). Therefore, MSCs implanted directly within the CNS may by themselves produce local pathology of yet unknown consequences (Snyder et al., 2011). The volume of these masses appeared to correlate with cellular density. Therefore cell number as well as number of application can be limited and risky factors for IT and ICV applications in contrast to IV.
  • The composition of IDPSC may be in the form of a pharmaceutical composition comprising stem cells expressing a mesenchymal and neuroepithelial stem cell immunophenotypes. In some implementations, expressing a mesenchymal and a neuroepithelial stem cell molecular profile is the expression of markers of MSC and neuroepithelial cells/progenitor cells and genes encoding neuro-protective and immuno-protective factors.
  • Cells expressing a MSC immunophenotype include expression of CD44. Prior animal models of multiple sclerosis found that NCS adhesion to inflamed endothelial cells and then trans-endothelial migration across the BBB into the inflamed CNS areas are sequentially mediated by the constitutive expression of functional cell adhesion molecules (CAM), especially CD44 (Rampon C et al., 2008). Although the exact mechanism of how hIDPSC are able to cross BBB is not clear, it is believed that these cells have this capacity because they have perycite-like characteristics (Barros et al., 2014). Perycites are known to play a critical role in the integration of endothelial and astrocyte functions at the neurovascular unit, and in the regulation of the BBB (Armulik et al., 2010; Liu et al., 2012). According to a previous study (Yilmaz et al., 2011), mesenchymal stem cells (MSC) can engraft into brain after systemic administration due to expression of CD44, which is a ligand of E and L blood vessel endothelial selectins (Dimitroff, et al., 2001). MSC present similar homing mechanisms as leukocytes. The first step of leukocyte migration involves capture of leukocytes flowing freely in the blood stream, mediated by glycoproteins known as selectins. P- and E-selectins are expressed by the vascular endothelium and are the principal mediators for the rolling response in leukocyte migration through blood vessels (Luster et al., 2005). MSC may use this or similar mechanisms to engraft in several organs (Sackstein et al., 2008) such as the brain. Thus, CD44 is considered a pivotal factor for MSC migration into the brain. Similarly to BM MSC, hIDPSC express CD44, which suggests that CD44 is also involved in hIDPSC migration towards several organs (Barros et al., 2014, Castanheira et al., 2013) including the brain after intravenous administration. Surprisingly, that, hIDPSC express also CD13, (aminopeptidase N) (Kerkis et al., 2006; Kerkis and Caplan, 2012), which is multifunctional protein and plays varying roles in cell migration, cell proliferation, cell differentiation (Taylor et al., 1993; Mina-Osorio et al., 2008a/b). CD13 participates in angiogenesis generating and modulating angiogenic signals, in the process of capillary tube formation, and as a marker of angiogenic vessels (Bhagwat et al., 2001). This suggests its possible role in hIDPSC capacity to migrate and to target brain vasculature. In the 3-NP rat study, hIDPSC demonstrated tight association with brain capillaries.
  • In some embodiments, the IDPSC lack expression of CD146, HLA-DR, and/or HLA-ABC. Lack of expression of these markers facilitates the use of hIDPSC as a safe, heterologous therapy. The endothelium plays an important role in the exchange of molecules, but also of immune cells between blood and the underlying tissue. The endothelial molecule S-Endo 1 antigen (CD146) is preferentially located at endothelial junctions and has been claimed to support endothelial integrity. Thus, in humans, MCAM (CD146) is expressed in T cells (3%) in the peripheral circulation of healthy individuals. MCAM positive T cells also demonstrate an increased ability to bind to endothelial monolayers and these cells could represent early components of the adaptive immune response (Dagur et al., 2015). Therefore, stem cells, which express this marker may bind to BBB and not cross BBB, as well as being immune reactive.
  • The mesenchymal stem cell genotype pattern also includes low expression of pluripotent markers OCT3/4 and nanog. Interesting, the undifferentiated stem cells of pharmaceutical composition of the invention need not express c-Myc, KLf-4, and REX-1. In fact, these stem cells may be negative for c-Myc, KLf-4, and REX-1.
  • Stem cells expressing a neuroepithelial stem cell molecular profile express at least one, preferably two, more preferably more than two NPC- and NSC-biomarkers selected from the group consisting of vimentin, nestin, SOX2, p75, and other neurotrophic factors essential for neural cells development and survival. p75 is a neurotrophic receptor marker. In the recent works, it was hypothesized that normalization of p75NTR and/or TrkB expression or their signaling will improve BDNF (brain-derived neurotrophic factor) neuroprotective therapies in Huntington's disease (Brito et al., 2013).
  • Exemplary neurotrophic factors essential for neuronal development and survival include BDNF (brain-derived neurotrophic factor), GNDF (glial cell line-derived neurotrophic factor), NGF (nerve growth factor), and NTs (neurotrophins). BNDF plays a critical role in Huntington's disease (Gauthier et al.; Strand et al.) and Parkinson's disease (Mogi et al.), both of which are dopamine-associated neurodegenerative diseases. Several studies demonstrate that wild-type HD-overexpressed htt protein increases BDNF expression in CNS cells, whereas the mutated htt protein leads to down-regulation of BDNF, resulting in insufficient neurotrophic support and neuronal cell death (Zuccato et al., 2001). The brains of AD patients have reduced NGF levels (Calissano et al.); however, NGF administration can partially reduce cholinergic atrophy in aged rodents (Fischer W et. al). In some embodiments, the preferred NGF is NGF-β. NTs essential for neuronal development and survival include NT3, NT4, or NT5. NT4 and NT5 are known to promote sensory and motor axon growth.
  • In some implementations, the stem cells comprise cells autologous to a subject in need of the pharmaceutical composition. In other implementations, the stem cells comprise cells allogeneic to the subject in need of the pharmaceutical composition. In some implementations, stem cells comprise a combination of cells autologous to and allogeneic to the subject in need of the pharmaceutical composition.
  • In one embodiment, the composition of stem cells is isolated from tissue of neural crest origin selected from the group consisting of dental tissue, periodontal tissue, and hair follicular tissue. In preferred embodiments, the tissue of neural crest origin is dental pulp. In a most preferred embodiment, the stem cells are from immature dental pulp, for example, human immature dental pulp stem cells (IDPSCs) as disclosed in International Application no. PCT/IB14/59850 and U.S. patent application Ser. No. 14/214,016. IDPSCs carry multiple neuronal markers and undergo robust differentiation into neurons. The novelty of IDPSC immunophenotype is unexpected expression of these markers and at the same time markers typical for MSC, presenting immunophenotype in accordance with the International Society for Cellular Therapy's minimal criteria for defining multipotent mesenchymal stromal cells (Dominici et al., 2006). This combination of expression by IDPSC of MSC and multiple neuronal markers is not typical for MSCs (Dominici et al., 2006) and which has not been disclosed for MSCs.
  • Pharmaceutical compositions contemplated in the invention are preferably isotonic. For intravenous injection, the population of immature stem cells should be between 104-1010 cells per injection, for example, 104, 105, 106 and 107 cells per kg of body weight. The pharmaceutical composition comprising a population of stem cells may be used in adjunction to other pharmaceutically active compounds or modalities. Thus in some embodiments, the pharmaceutical composition may further comprise another pharmaceutically active compound or therapeutic modality.
  • Mesenchymal Stem Cells
  • In the body, MSCs are found in bone marrow, umbilical cord tissue, dental pulp and fat pads. However, in bone marrow MSCs are relatively rare, comprising only one out of every 10,000 cells, while other sources are significantly richer in these cells. In organism MSCs are responsible for tissue regeneration in cases of disease, trauma or injury throughout human life. This function of MSCs is mediated by their capacities for self-renewal and plasticity (the capacity for differentiation—production of diverse cell types). MSCs can be isolated from aforementioned tissues and cultured easily in the laboratory. After obtaining a limited number of the cells from a patient, MSCs can be multiplied rapidly in vitro and cryopreserved for the future clinical applications.
  • MSCs are able to secrete a variety of bioactive molecules, such as cytokines, which provide “trophic activities” by structuring a regenerative microenvironment, and other molecules that contribute to immunomodulatory cell functions and even to transfer products as large as mitochondria to damaged cells that need help. When transplanted in vivo, MSCs in response to chemotactic stimuli, can migrate to the focal injury from both local and surrounding sites. Additionally, MSCs can act to reduce chronic inflammation, to inhibit apoptosis, to provide the appearance of myofibroblasts, to inhibit scar formation and to stimulate the mitosis of tissue-intrinsic progenitors, thus remodeling damaged tissue. That is why MSCs are also called “Medicinal Signaling Cells.” They stimulate angiogenesis, the process of new blood vessel formation, which is closely linked to neurogenesis, the process by which new nerve cells are produced. Blood vessels play an important role as a framework for neuronal progenitor cells migration toward the damaged brain region The factors secreted by MSCs also reduce the destructive effects of oxidative harm. Using all these mechanisms of action MSCs can significantly improve lesioned microenvironment that leads to restoration of the damaged cells. Therefore, MSCs are believed to be “cellular paramedics”.
  • When MSCs obtained from humans were labeled, in order to track them, and injected into mice that had some type of tissue damage, they migrated throughout the damaged tissues apparently evenly. These cells can or not to be present in the tissue for a substantial period of time, which depends on disease model. The continued presence of MSCs is important, but not essential, to therapeutic development because it indicates that potential positive long-term effects of a treatment might be capable of persisting.
  • It is important to understand, that temporary presence of MSCs is not a result of the host immune system action, because the experiment in injured mice, with or without functional immune systems yielded the same results. Further investigations demonstrated that MSCs suppress the immune system and reduce inflammation. In other words, MSCs can be transferred between organisms demonstrating very low immune rejection, which occurs when the immune system of the organism attacks the foreign tissue, receiving the transplant. This finding makes MSCs good candidates for transplantation or injection into a host because they can avoid rejection by the host's immune system (Le Blank, Ringdén, 2006; English, 2012; Miguel et al., 2012; Griffin et al., 2012; Ankrum et al., 2014).
  • The crucial question of cellular therapies is a route of MSCs delivery into the brain, which has been approached in a number of different ways. Several approaches have been proposed to deliver MSCs into the brain such as, intrathecal, intravenous, an injection into the space surrounding the spinal cord and even a route through the nose. In early development, as a result of complex multicellular interactions between immature endothelial cells and neural progenitors, neurons, radial glia, and pericytes, which shared similar features with MSCs, the blood/brain barrier (BBB) is formed and it controls selective molecular or cells trafficking between the bloodstream and brain interstitial space. The BBB present significant problems for the delivery of therapeutic agents (drugs or cells) for treating brain malignancies and neurodegenerative disorders. Systemically-infused MSCs may treat acute injuries, inflammatory diseases, stroke of the central nervous system (CNS) and even brain tumors because of their regenerative capacity and ability to secrete trophic, immune modulatory, or other engineered therapeutic factors. However, whether MSCs possess the ability to migrate across the BBB in normal and pathological conditions remains unresolved (Liu et al., 2013).
  • Systemic infusion (e.g. IV) of MSCs expanded in vitro is minimally invasive and convenient procedure that is used in the large number of ongoing clinical trials. Therefore, it is essential to understand if transplanted MSCs can home and engraft at ischemic and injured sites in the brain to exert their therapeutic effects. No data has yet to suggest or disclosed that systemic delivery of minimally manipulated MSCs may result in direct transplantation of cells into the brain through the BBB.
  • The simplicity with which MSCs can be obtained, cultured, as well as their unique “trophic activities” and possibility of their transfer into a host without immune rejection are the reasons why stem cell therapy with MSCs is a promising avenue to for treating neurological diseases and conditions, for example neurodegeneration. According to recent publications, MSCs can support repair neurodegeneration by secreting trophic factors, which are proteins that stimulates differentiation and survival of cells. The effects of these factors allow nerve cells to carry out several processes that can support survival: axon extension, growth, and cells attachment. Although are evidences that MSCs can promote cell growth and repair in the brain, it is not yet definitively confirmed that MSCs can become mature nerve cells with the ability to signal, or communicate with, other nerve cells.
  • Previous studies have tested the potential of MSC therapy in HD animal models (chemical models where HD is induced by QA or 3-NP and transgenic mouse lines R6/2-J2, N171-82Q, and R6/2). However, while the authors called the cells tested MSCs, these cells were not confirmed as having the immunophenotype typical for MSCs as defined by the International Society for Cellular Therapy (Dominici et al., 2006). These pre-clinical studies used allogeneic and xenogeneic primary culture and immortalized cell lines from the bone marrow, adipose tissue, and umbilical cord blood grown under normal levels of oxygen (normoxia) or under low levels of oxygen (hypoxia), as well as, mononuclear cells. Thus these studies have not established that cells considered as MSCs by the International Society for Cellular Therapy may be successfully employed to treat neurological diseases without previous special manipulation in culture.
  • The number of cells used in these experiments varied from 105, 2×105, 4×105, 5×105, and up to 106 per hemisphere/striatum. The time of administering MSCs transplantation varied significantly across the studies with the time being 1-3 days, 2-4 weeks, and 8 weeks. These cells were found in the brain after direct grafting, but direct intrabrain delivery is a highly invasive and risky procedure. Thus these studies have not demonstrated that minimally invasion methods of administering stem cell therapy, such as systemic administration by IV injection, could be used.
  • All but one of the studies used the cells at passages no higher than 10. The exclude study used mouse umbilical cord blood-derived (mUBC-derived) MSCs at passages 40 and 50. Interestingly, the study observed that expression of marker of pluripotent stem cells, such as stage specific embryonic antigen-4 (SSEA4) increased with passaging and that transplantation of high-passage mUCB-derived MSCs confer significant motor benefits unlike transplantation with low-passage mUCB-derived MSCs. Unfortunately, potential pluripotent origin and high risk of karyotype mutation due to higher passage numbers put clinical applications at risk.
  • In contrast to these studies, the present invention provides a method of treating neurological diseases and condition that uses a unique population of IDPSC having the immunophenotype typical for MSCs as defined by the International Society for Cellular Therapy effective even with a minimally invasive administration, for example through classic IV route of delivery.
  • As demonstrated by these previous studies on treating HD with supposed MSCs, one hope for treating neurodegenerative diseases is using stem cells. Unfortunately, only treatment with administering fetal donor tissue to the striatum proceeded to clinical trial, and it was only a small trial.
  • Cell therapies in HD are intended to protect neuronal populations susceptible to disease and/or replace dysfunctional or dying neurons. Thus clinical progress in HD cell therapy has been centered on establishing protocols for transplanting fetal-derived cells into the diseased striatum. This strategy is helping the development for stem cell therapy in the clinic and provides a period of several years of improvements and stability, but not permanent cure for disease (Bachoud-Levi et al., 2006). The long-term follow up over a 3- to 10-year postoperative period of the patients concludes that fetal striatal allografting in HD is safe. However, no sustained functional benefit was seen, perhaps due to the small amount of cells that was grafted in this safety study compared with other reports of more successful transplants in patients with HD (Barker et al., 2013).
  • Use of stem cells therapies is inevitable since intracellular and cellular mechanisms are involved into HD phenotype. Stem cell therapy may also accelerate the process of brain tissue regeneration. Stem cells are an important therapy, which will help to rebuild an area of the brain that was most damaged in HD. Only drugs approach will not be able to reconstruct damaged brain areas especially in late stages of HD.
  • MSCs may be obtained from extracted human teeth, both permanent and deciduous, by enzymatic digestion (Gronthos et al. 2000; Miura et al., 2003), or by organ culture followed by explant (immature dental pulp stem cells, IDPSCs) technology as disclosed in International Application no. PCT/IB14/59850 and U.S. patent application Ser. No. 14/214,016. The IDPSCs are obtained from dental pulp tissue, which anatomically originated from ectomesenchymal tissue, more precisely from neural crest, which is a mass of tissue present in the early formation of an embryo. It eventually forms the hard and soft tissues of the neck and cranium.
  • IDPSCs, which are of neural crest origin, are known to migrate pre-natally into various, mainly ectodermal tissues and have the capacity to self-renewal and display a developmental potential almost the same as embryonic stem (ES) cells, but without risk of formation of embryonic bodies in vitro and teratomas in vivo (Kerkis and Caplan, 2012). The postmigratory stem cells of neural crest origin generate all craniofacial bones, the majority of cells and tissues of the central and peripheral nervous systems, as well as several non-neural cell types, such as smooth muscle cells of the cardiovascular system, pigment cells in the skin, cartilage, connective tissue, corneal epithelium and dental pulp among them. Although postmigratory postnatal stem cells of neural crest origin are of restricted developmental potential, they maintain functional characteristics resembling their embryonic counterparts and an ability to differentiate into a broad spectrum of cell types (Le Douarin et al., 2004, 2007, 2008; Dupin et al., 2007; Le Douarin & Dupin, 2003, 2012).
  • In vitro IDPSCs undergo uniform differentiation into neurons and glial cells. In vivo transplantation of human IDPSCs showed dense engraftment in various tissues, including neurons. Neuronal fate differentiation is based upon epigenetic “memory” of orofacial bones, including dental pulp, compared with those in axial and appendicular bone (bone marrow and ileac crest) based on their different embryological origins. Maxillas, mandible, including the alveolar bone (i.e. dentine, dental pulp and periodontal ligament), are formed exclusively by neural crest cells while axial and appendicular bones develop from mesoderm. Thus IDPSCs have the potential for neural regeneration and neuroprotection.
  • Logan A et al. described that multiple NTFs (neurotrophic factors) should be produced by cells in order to result in synergistic effect on neuroprotection. Therefore it is important that hIDPSCs cells are expressing and releasing multiple NTFs. Recent publications report the evidence for a paracrine mechanism of dental pulp stem cells (DPSC) action in neural support, with the gene expression of many NTFs, such as NGF (nerve growth factor), BDNF and NT3, the results demonstrated that hIDPSC promoted significantly more neuroprotection and neurogenesis of axotomised RGC than either hBMSC (bone marrow derived MSC) or hAMSC (adipose tissue derived MSC) (Mead B et al. 2013; Mead B et al., 2014; Martens W et al., 2013). Intravitreally transplanted DPSCs were suggested as a more appropriate cell type than BMSCs for retinal therapy (Mead B et al., 2014). These studies used DPSC(=SHED) enzymatically derived from adult rats using trypsin.
  • In addition, it has recently been strengthened by the results of an study using a rodent model of spinal cord injury with transplantation of SHED by direct dura transplantation in proximity and directly to the lesion site, wherein SHED was superior to three human skin fibroblast lines in terms of neuroprotection and neuroregeneration through both cell-autonomous and paracrine neuroregenerative activities (Sakai et al., 2012). These studies used cultured DPSC enzymatically derived from immature and adult wisdom using collagenase. During transplantation of DPSCs, animals were also treated with cyclosporine for immunosuppression.
  • Safety of Systemically Administering Stem Cells
  • Unless the transplant is an autograft, there is always a risk that the host's immune system will attack the transplant. Even a well-matched allograft requires immunosuppression pretreatment. This remains true for stem cell transplantation.
  • In order to avoid the host's immune system attacking the transplanted cells, the therapeutic stem cell population should be not be immunogenic. Immunogenicity is the ability of allogeneic stem cells to provoke an immune response when facing the host immune system after transplantation (Schu S et al., 2012). The transplantation of NPCs with mismatched major histocompatibility complex (MHC) into mice with mouse hepatitis virus-induced CNS demyelination resulted in increased T cell infiltration and NPC rejection (Weinger J G et al., 2012). However, recent evidence supports the possibility that undifferentiated adult stem cells are endowed with an immunologically privileged status and are capable of escaping the normal processes of allogeneic rejection (Bifari F et al. 2010). Immunologically privileged status is possible for a population of cells if the cells lack the expression of MHCs. For example, no immunogenicity in humans can occur by the population of stem cells being essentially negative for human leukocyte antigen (HLA), which is the human version of MHC. Therefore, the absence of HLA-DR, which is a quality control characteristic of IDPSCs (see Example 1), is an essential marker for cell to be used in for systemic cell therapy without need of toxic immunosuppression pre-treatment to the patient.
  • Another risk of stem cell transplantation is the increased risk of tumor development, especially for undifferentiated cells, because of these cell's potential for differentiation into other cell types. Pluripotent stem cells, especially hESCs and iPSCs cells are able to form spheres that resemble embryoid bodies in vitro and teratomas in vivo. All currently available technologies to apply pluripotent cells hold tumorigenicity risk. Expression and lack of expression of certain genes reduces risks to enable systemic administration of a population of stem cells.
  • Nanog is transcription factor associated with the maintenance of the pluripotent cells of the inner cell mass and the formation of embryonic stem cells. Nanog is a leukemia inhibitory factor (LIF) and activator of transcription-independent factor-3 (STAT-3). It is regulated by OCT4 and SOX2 and in turn positively regulates the expression of OCT4, SOX2 and itself by binding to the respective promoter gene regions (Boyer et al, 2005; Loh et al., 2006; Li, 2010). Together, these three transcription factors play an essential role in preventing differentiation of pluripotent stem cells (Boyer et al., 2005). The transfection cellular nucleus with OCT3/4, SOX2, NANOG was previously to be sufficient for inducing pluripotency in adult somatic cells (the creation of iPSC) and then lead to full pattern of embryonic stem cells theoretical characteristics: differentiation into 200 types of cells in the body, unlimited expansion, renewal potential, embryonic body formation, teratoma formation. Teratoma formation is a main threat of safety in cellular therapy. However, absence expression of nanog in nucleus is an essential safety marker to determine whether a population of stem cell is suitable for systemic administration. The lack of tumorigenicity of undifferentiated stem cells in vivo requires the absence of nanog in nucleus. Thus undifferentiated stem cells expressing inactive nanog, i.e. nanog localized in the cytoplasm, also lack tumorigenicity in vivo.
  • Another important safety marker for a population of stem cells suitable for systemic administration is the expression of p53. This protein is crucial in multicellular organisms, where it regulates the cell cycle and thus prevents cancer by functioning as a tumor suppressor.
  • ABCG2 protein expression is also safety marker that indicates a population of stem cells is suitable for systemic administration. ATP-binding cassette (ABC), ABCG2 protein (BCRP) expression is an important determinant of the MSC undifferentiated population phenotype. ABCG2 might serve as a marker for undifferentiated stem cells from various sources, as its expression is sharply downregulated with differentiation. Notably, ABCG2 transporters with Alzheimer's disease (AD), actively transport AP as confirmed histopathologically in AD cases and controls. Genome-wide association studies (Bertram L et al., 2007) have implicated a have identified genes the modulate AD risk, including genetic variants in ABCA7, a variant of ABC gene. It was concluded that increase in ABCA7 expression reduces AD risk, though increased ABCA7 expression during AD is insufficient to block disease progression (Jared B et al., 2014).
  • Thus some embodiments of the pharmaceutical composition of the invention comprises stem cells from tissue of neural crest origin expressing at least one safety markers selected from the group consisting of ATP-binding cassette sub-family G member 2 (ABCG2), inactive nanog, p53, and SOX2. In some aspects, the at least one safety marker is elected from the group consisting of ATP-binding cassette sub-family G member 2 (ABCG2), inactive nanog, and p53. The IDPSC have high expression of ABCG2 and p53 but low expression of inactive nanog and SOX2. For example, when the at least one safety marker is ABCG2 or p53, at least 75%, 80%, 85%, 90%, 95% or 98% of the stem cells of the pharmaceutical composition express the at least one safety marker. On the other hand, if the at least one safety marker is inactive nanog or SOX2, no more than 30%, 25%, 20%, 15%, 10%, 5%, or 5% of the stem cells express the at least one safety marker. In some aspects, the stem cells of the pharmaceutical composition coexpress ABCG3, p53, inactive nanog, and SOX2, wherein at least 75% of the stem cells express ABCG2, at least 75% of the stem cells express p53, no more than 5% of the stem cells express inactive nanog, and no more than 30% of the stem cells express SOX2.
  • Another embodiment of the pharmaceutical composition comprises stem cells from tissue of neural crest origin expressing at least one neuroepithelial stem cell marker selected from the group consisting of brain-derived neurotrophic factor (BDNF), neutrotrophin-3 (NT3), neutrotrophin-4 (NT4), neutrotrophin-5 (NT5), and p75. In some aspects, the stem cells have high expression of the at least one neuroepithelial stem cell marker. For example, at least 75%, 80%, 85%, or 90% of the cells express at least one neuroepithelial stem cell marker. In some embodiments, the stem cells of the pharmaceutical composition coexpress BDNF, NT3, NT4, NT5, and p75.
  • In some aspects, these embodiments pharmaceutical composition comprise stem cells from tissue of neural crest origin that are negative for HLA-DR. The stem cells of the pharmaceutical composition may also be negative for certain MSC markers selected from the group consisting of c-Myc, KLf-4, and REX-1. In preferred embodiments, the stem cells of the pharmaceutical composition are negative for HLA-DR, c-Myc, KLf-4, and REX-1.
  • The various embodiments of the pharmaceutical composition may also be combined. For example, the pharmaceutical composition may comprise stem cells from tissue of neural crest origin expressing at least one safety markers selected ABCG2, inactive nanog, and p53 and further express at least one neuroepithelial stem cell marker selected from the group consisting of BDNF, NT3, NT4, NT5, and p75. As another example, the pharmaceutical may comprise stem cells from tissue of neural crest origin expressing express at least one neuroepithelial stem cell marker selected from the group consisting of BDNF, NT3, NT4, NT5, and p75 while further expressing at least one safety markers selected ABCG2, inactive nanog, p53 and SOX2.
  • Methods of Using the Pharmaceutical Compositions
  • The present invention provides for methods for treating neurological diseases and conditions comprising systemically administering the pharmaceutical composition of the invention to a subject. In some implementations, these methods of treating neurological diseases and condition promote neurogenesis and are protective in models of neurodegenerative diseases. In some embodiments, systemic administration the population of stem cells, such as by IV administration, results in direct delivery of the cells to the brain. In some aspects, neurogenesis occurs by the population of stem cells self-differentiating and/or activating intrinsic stem cells to migrate and differentiate. In some aspects, neurogenesis is preferably dopamine-associated.
  • In some embodiments of the methods, the neurological disease or condition is treated by the stem cells crossing the blood/brain barrier (BBB) and inducing neurogenesis. In some aspects, the stem cells are directly transplanted into the brain parenchyma, including striatum, following crossing of the BBB. In some embodiments, the induced neurogenesis is dopamine-associated. For example, dopamine-associated neurogenesis occurs through self-differentiation of the stem cells or activation of migration and differentiation of intrinsic stem cells by the extrinsic stem cells. In some aspects, massive dopamine-associated neurogenesis takes place in the subventricular zone (SVZ).
  • In some implementations, the methods further comprise measuring the amount of DA receptor in the subject. In some embodiments, measuring the amount of DA receptor in the subject comprises imaging the subject to detect DA receptor. In most preferred embodiments of the methods, neurogenesis is mediated by dopamine receptor D2, thus in some embodiments, the DA receptor measured is receptor D2.
  • In some embodiments of the methods, the pharmaceutical composition provides neuroprotection. For example, systemic neuroprotection is provided with the high basal level of neurotrophic and immunoprotective factors expression and release pattern of the stem cells of the pharmaceutical composition. In some aspects, these stem cells of the pharmaceutical composition are IDPSCs.
  • The neurological diseases and conditions include, for example, autism, schizophrenia, epilepsy, stroke and ischemia, a neurodegenerative disease or condition, a motor disorder, or a convulsive disorder. The neurodegenerative disease or condition may be, for example, Parkinson's disease, multiple sclerosis, amyotrophic lateral sclerosis (ALS), stroke, autoimmune encephalomyelitis, diabetic neuropathy, glaucomatous neuropathy, Alzheimer's disease, and Huntingdon's disease. Motor disorders include, for example, Tourette syndrome, amyotrophic lateral sclerosis (ALS), progressive bulbar palsy, spinal muscular atrophy (SMA), post-polio syndrome (PPS). Convulsive disorders include, for example, epilepsy.
  • In some implementations, the methods for treating neurological diseases and conditions support the natural neuro-protective mechanism in subjects diagnosed with early HD. In other implementations, the methods for treating neurological diseases and conditions repairs lost DA neurons in subjects diagnosed with PD.
  • The present invention also provides for methods of using the pharmaceutical composition as a preventive therapy for subjects at risk of HD.
  • In one embodiment, the present invention is directed to a pharmaceutical composition for systemic administration to a subject to treat a neurological condition comprising undifferentiated stem cells from tissue of neural crest origin expressing at least one safety markers selected from the group consisting of ATP-binding cassette sub-family G member 2 (ABCG2), inactive nanog, and p53. In certain aspects, inactive nanog is expressed nanog localizing predominantly in the cytoplasma of the undifferentiated stem cell. In another aspect, at least 75% of the undifferentiated stem cells express the at least one marker when the at least one marker is ABCG2 or p53. In yet another aspect, no more than 5% of the undifferentiated stem cells express the at least one marker when the at least one biomarker is nanog.
  • In some embodiments, the undifferentiated stem cells express ABCG2, inactive nanog, and p53. In one aspect, at least least 75% of the undifferentiated stem cells express ABCG2, at least least 75% of the undifferentiated stem cells express p53, and no more than 5% of the undifferentiated stem cells express inactive nanog. In another aspect, the undifferentiated stem cells further express SOX2, and wherein no more than 30% of the undifferentiated stem cells express SOX2. In yet another aspect, the undifferentiated stem cells further express at least one neuroepithelial stem cell marker selected from the group consisting of brain-derived neurotrophic factor (BDNF), neutrotrophin-3 (NT3), neutrotrophin-4 (NT4), neutrotrophin-5 (NT5), and p75.
  • In other embodiments, the undifferentiated stem cells express BDNF, NT3, NT4, NT5, and p75. In one embodiment, the present invention is directed to a pharmaceutical composition for systemic administration to a subject to treat a neurological condition comprising undifferentiated stem cells from tissue of neural crest origin at least one neuroepithelial stem cell marker selected from the group consisting of BDNF, NT3, NT4, NT5, and p75.
  • In certain aspects, the undifferentiated stem cells express BDNF, NT3, NT4, NT5, and p75. In other aspects, the undifferentiated stem cells further express at least one safety markers selected from the group consisting of ABCG2, inactive nanog, p53, and SOX2. In certain aspects, inactive nanog is expressed nanog localizing predominantly in the cytoplasma of the undifferentiated stem cell.
  • In yet other embodiments, at least 75% of the undifferentiated stem cells express the at least one marker when the at least one marker is ABCG2 or p53. In certain aspects, the undifferentiated stem cells are negative for HLA-DR. In one embodiment, the tissue of neural crest origin is dental pulp. In yet other aspects, the undifferentiated stem cells from tissue of neural crest origin are immature dental pulp stem cells (IDPSCs).
  • In another aspect, the present invention provides a method of treating a neurological disease or condition comprising systemically administering to a subject a pharmaceutical composition comprising undifferentiated stem cells from tissue of neural crest origin expressing at least one safety marker selected from the group consisting of ABCG2, inactive nestin, and p53. In some aspects, the undifferentiated stem cells of the pharmaceutical composition further express at least one neuroepithelial stem cell marker selected from the group consisting of BDNF, NT3, NT4, NT5, and p75.
  • In yet another embodiment, the present invention is directed to a method of treating a neurological disease or condition comprising systemically administering to a subject a pharmaceutical composition comprising undifferentiated stem cells from tissue of neural crest origin expressing at least one neuroepithelial stem cell marker selected from the group consisting of BDNF, NT3, NT4, NT5, and p75.
  • In certain embodiments, the undifferentiated stem cells of the pharmaceutical composition further express at least one safety marker selected from the group consisting of ABCG2, inactive nestin, p53, and SOX2.
  • In other aspects, the subject is intravenously administered the pharmaceutical composition. In some embodiments, the neurological disease or condition is treated by the population of undifferentiated stem cells crossing the blood/brain barrier and inducing neurogenesis. In one aspect, the neurological disease or condition is treated by the undifferentiated stem cells inducing neurogenesis via dopamine-associated neurogenesis. In another aspect, the dopamine-associated neurogenesis is through self-differentiation of the undifferentiated stem cells or activation of migration and differentiation of intrinsic stem cells by the undifferentiated stem cells.
  • In certain embodiments, the undifferentiated stem cells of the pharmaceutical composition provide neurotrophic factors and immunoprotective factors. In other embodiments, the undifferentiated stem cells of the pharmaceutical composition provides systemic neuroprotection. In one embodiment, the undifferentiated stem cells are autologous and/or allogeneic to the subject.
  • In certain aspects, the neurological disease or condition is a neurodegenerative disease or condition. The neurodegenerative disease or condition may be selected from the group consisting of Parkinson's disease (PD), multiple sclerosis, amyotrophic lateral sclerosis (ALS), stroke, autoimmune encephalomyelitis, diabetic neuropathy, glaucomatous neuropathy, Alzheimer's disease, and Huntington's disease (HD).
  • In some aspects, the method comprises systemically administering the pharmaceutical composition to the subject, wherein the subject is diagnosed with early HD, supports the natural neuroprotective mechanism in the subject. In other aspects, the method comprises systemically administering the pharmaceutical composition to the subject, wherein the subject is diagnosed with PD, repairs lost dopaminergic neurons in the subject. In another embodiment, the neurological disease or condition is selected from the group consisting of autism, schizophrenia, stroke, and ischemia. In other embodiments, the neurological disease or condition is selected from the group consisting of a motor disorder and a convulsive disorder.
  • In certain aspects, the subject is administered a single administration of the pharmaceutical composition. In one embodiment, the subject is administered a single intravenous injection of the pharmaceutical composition. In yet other embodiments, the subject is administered a first and a second administration of the pharmaceutical composition.
  • In other embodiments, the subject is administered a first and a second intravenous injection of the pharmaceutical composition. In some aspects, the second administration or intravenous injection of the pharmaceutical composition takes place at least 7 days after the first administration or intravenous injection.
  • In one aspect, the method further comprises measuring the amount of DA receptor in the subject. In another aspect, the method comprises measuring the amount of DA receptor in the subject comprises imaging the subject to detect DA receptor. In one aspect, the DA receptor is receptor D2.
  • Enrichment of Late Population of hIDPSC with Neural Progenitors and Neurons
  • Other aspects of this disclosure relate to enrichment of late population of hIDPSC with neural progenitors and neurons following 15-, 30- and 45-cycles of dental pulp mechanical transfer and induction of neural differentiation. The CNS is originated from ectoderm, more precisely from the pool of multipotent neuroepithelial stem cells (NSCs). This cell's population is composed at least by two different groups of neural precursors: a first group can grow in monolayer cultures; a second group generally form neurospheres (i.e. grow in suspension culture). In spite of this difference, both of cell types demonstrate the same differentiation potential (Noble et al., 2011, Kempermann, 2011).
  • NSCs are symmetrically dividing and under appropriate conditions can be rapidly conversed into neuroblasts, asymmetrically dividing precursors, which are able to develop into neurons after a migration phase. Neuroblasts are slightly more differentiated than NSCs and can produce transit amplifying cell population. The ability to divide is main difference between neuroblasts and neurons, which are postmitotic. Neuroblast differentiates and matures into neurons but not in other cell types.
  • NSCs fate, among other mechanisms, is regulated through signals that emanate from surrounding tissues. NSCs are found in specific regions, so-called stem cell niches that provide the microenvironmental cues that regulate stem cell proliferation, self-renewal and differentiation.
  • Known today are two potential sources of stem cell with neural potential of differentiation: first, fetal neuronal precursors obtained from abortion and, second, pluripotent stem cells (ES cells, iPSCs) induced to differentiated in vitro neuronal precursors. Fetal neuronal precursors raise many ethical problems, while there are certain safety risks associated with human use of ES and iPSCs that delay therapeutic applications. Alternative methods of neurodegenerative therapies from other human types of stem cells are needed to exploit the uses of cell therapy for treatment of neurological diseases. Methods for transplanting stem and progenitor cells are described in U.S. Pat. Nos. 5,928,947; 5,817,773 and PCT Publication Nos. WO 01/176507. These methods include expansion in cell culture and transplantation of undifferentiated neuroprotective stem cells, and/or through by transplantation of neural precursor cells, or/and fully differentiated neuronal cells. Cell therapeutic interventions may involve both cell transplantation and the stimulation of endogenous neural progenitor cells. Conventional methods have multiple disadvantages and limitations and new sources of cells for stem cell neuroprotective/neuroregenerative therapy are required. Also of importance is to predict neuroprotective potential of cells for stem cell therapy using molecular markers.
  • Stem cells from dental pulp have been isolated by Gronthos et al. 2000, Shi et al. WO 02/07679. Sharpe (WO 01/60981) claimed the production of tooth progenitor cells from embryonic stem cells or adult stem cells or tissue culture. U.S. Patent Publication 2002/0119180 claimed a method and production of a biological tooth from third molar tooth germ (Young et al. 2002). The invention disclosed in U.S. Pat. No. 8,192,987 relates generally to pluripotent stem cells, including embryonic-like pluripotent stem cells derived from teeth.
  • Aspects of the disclosures presented herein relate to a composition of hIDPSCs which co-express SOX1, SOX2 and beta-3-tubulin markers and especially methods of treating neurological conditions. Also disclosed herein are compositions and methods of enrichment of the co-expressed markers during LT hIDPSCS cell culture therefore increasing neuroectodermal lineage commitment of such LP hIDPSCs. Disclosed herein are new methods to the treatment of neurodegenerative diseases using hIDPSCs obtained through LT harvesting cycles and enriched with specific neuronal biomarkers, such as but not limited to SOX1, and SOX2, beta-3-tubulin. This method is useful to screen for markers that affect neural lineage commitment of hIDPSCs and hold higher potential to treat CNS and PNS diseases, while maintaining normal karyotype and no teratoma formation detected in vivo. Moreover, the contemplated embodiments provide a substantially homogenous population of cells enriched with population of cells that co-express said stemness neuronal markers (SOX1, SOX2 and beta-3-tubulin) while maintaining normal karyotype and carry no risk of teratoma formation. The homogenous population is likely advantageous for treating disease in a human (mammalian). Other advantages of the cell-based therapies disclosed herein include, but are not limited to, incorporation of the cells central nervous system tissue, peripheral nervous system tissue. Such incorporated cells have the potential to differentiate or develop into neuronal, glial or other cells to replace or facilitate repair of the damaged, traumatized or degenerating tissue thereby resulting in a more permanent treatment of the degenerative, acute injury, traumatized, neurological condition.
  • Incorporated into this document are also the disclosed methods of long term in vitro culturing of dental pulp (DP) tissue in order to produce unlimited number of stem cells named hIDPSCs in U.S. Provisional Patent Application 61/791,594, the contents of which are incorporated herein by reference in their entirety. Oxygen is one of critical signals that can affect stem cell properties under normal tissue homeostasis. Interestingly, low oxygen levels, or hypoxia, are a hallmark of stem cell niches (Panchision, 2009).
  • Surprisingly, it was observed on biological samples that were stored during long term/LT cell culture of hIDPSCs that during this process the dental tissue gradually shrink, thus creating hypoxia inside the pulp tissue. More surprisingly, immature dental pulp stem cells (IDPSCs) isolated from DP after 30 times of mechanical transfer/harvesting cycle (late population (LP) of IDPSC) and tested for their capacity to produce neurons in vitro demonstrated the increased ability for neural differentiation, as well as enrichment of a population pre-committed LP IDPSC with neuronal precursor when compared with IDPSC isolated after 15 or 25 cycles of mechanical transfer/harvesting (early population (EP)).
  • According to some aspects, we discovered that the number of actively proliferating neuronal precursors (neuroblasts) in pre-committed IDPSC neural precursor are related with the number of DP transfer cycles.
  • SOX1 and SOX2 BIOMARKERS
  • SOX1 (for sex-determining region Y-box 1) protein is a transcription factor involved in early central nervous system development. SOX1 is a known marker characteristic of a developing central nervous system. The expression of SOX1 in the neural plate and tube seem to correlate with mitotically active progenitors that are not yet committed to a final stage. SOX1 defines the dividing neural precursors of the embryonic CNS and expresses particularly in the ventral striatum (Pevny et al., 1998; Kempermann et al., 2003).
  • SOX2 is one of the earliest known transcription factors expressed in the developing neural tube and is expressed in certain cells of the adult brain. SOX2 is commonly known to play a critical role in the central nervous system. Recently, Terskikh and colleagues (2011) elucidate role of SOX2 in peripheral nervous system. They developed a hESC-based model in which migratory cells undergo epithelial to mesenchymal transition (EMT) to acquire properties of neural crest (NC) cells. They found that migratory NC progenitor's down-regulate SOX2, but then start re-expressing SOX2 as they differentiate to form neurogenic dorsal root ganglion (DRG)-like clusters. SOX2 down regulation was sufficient to induce EMT and resulted in massive apoptosis when neuronal differentiation was induced. They also showed that SOX2 binds directly to NGN1 (neorogenin 1) and MASH1 promoters and is required for their expression. When neural crest stem cells were prevented from re-expressing SOX2, they die or can give rise to glia or smooth muscle cells. Thus, the function of SOX2 is to keep cells multipotent or pluripotent for become neurons later in development.
  • SOX2 in Neurodegenerative Disease Models: Hundtington Disease (HD) Models
  • Molero and co-workers (2009) reviewed an HD model. In this model, progressive cognitive deficits develop at the age of 9 months, suggesting possible hippocampal dysfunction (Kandasamy et al., 2010). They observed enhance of NSC self-renewal at late embryonic stages together with high levels of SOX2, a factor that mediates NSC self-renewal, multilineage potential, and neurogenesis. Other groups showed that in hippocampal neural stem cell niche in a transgenic rat model of Huntington disease a disease-associated progressive decline in hippocampal progenitor cell proliferation accompanied by an expansion of the pool of 5-bromo-2-deoxyuridine label-retaining SOX2-positive quiescent stem cells in the transgenic animals.
  • Upon transplantation to the developing brain they incorporate extensively into the host brain, demonstrate wide spread distribution, migrate along established host brain migratory tracks, differentiate in a region specific manner into progeny of the three fundamental neural lineages, indicating their capability to respond to local cues and participate in the development and histogenesis of the living host.
  • This combination of defining properties will identify the neural progenitor cell lines of the invention regardless of the method used for their isolation. One method is to transplant undifferentiated stem cells that can differentiate into neural precursor cells, and then into fully differentiated neurons (including neurons, astro-glia, or oligodendrocytes). Another method is to transplant undifferentiated or pre-differentiated IDPSCs that are capable to release neuroprotective factors into affected areas of the nervous system. Moreover, using the described hIDPSCs transplant method of neuroregeneration may be based on administering into patient pre-differentiated ex vivo neuronal precursors or fully differentiated neurons or combinations of thereof.
  • According to some aspects, a cell culture composition comprises (co-expressing) SOX1 and SOX2 positive hIDPSC, which are neural crest stem cells. The SOX1 and SOX2 may be obtained through LT cell culture. Enrichment of the transcription factors increases towards LP culture, which holds high potential to neuronal differentiation, keeps karyotypes normal, and does not carry the risk of teratoma in vivo.
  • According to another aspect, a method for obtaining a homogenous population of hIDPSC comprises obtaining the population thorough LT mechanical/non-enzymatic harvesting cycles. The number of harvesting cycles may comprise over 25 cycles, about 45 cycles, or more than 45 cycles. An embodiment of the method may further comprise culturing the taken cells and passaging a plastic adherent cell from the cell culture. The plastic adherent cell typically (i) self-renews, (ii) differentiates into cells of endodermal, mesodermal, or ectodermal lineage, and (iii) expresses markers SOX1, SOX2 and 3-beta-tubulin. An embodiment of the method may further comprise administering the product to a patient as a pharmaceutical composition in a pharmaceutical accepted carrier/vehicle for cell therapy to treat CNS diseases.
  • According to another aspect, a mitotically active homogeneous neural progenitor's (neuroblasts) composition comprises (co-expressing) such markers as SOX1, SOX2, and beta-3-tubulin that are not yet committed to a final stage.
  • According to another aspect, a method for obtaining a homogeneous population of pre-differentiated IDPSCs comprises one or more of the following steps. Obtaining IDPSCs-derived neurospheres by culturing IDPSC in non-adherent conditions (suspension). Following adherence to a plastic dish in order to form neural rosette, the cells within the rosettes are presented by (i) transit amplifying cell population of neuroblasts positive for SOX1, SOX2, and BrdU and beta-3-tubulin that are slightly more differentiated than IDPSC; (ii) neuroblasts differentiated terminally into NEURONAL LINEAGES OF CENTRAL AND PERIPHERAL NEURAL SYSTEM—(including neuronal precursors, astroglia and oligodendrocytes and ganglion cells) upon external in vitro- or in vivo-induced microenvironment; (iii) use as a pharmaceutical composition in a pharmaceutical accepted carrier/vehicle for cell therapy to treat CNS diseases by administrating to a patient; and/or (iv) use as a pharmaceutical composition in pharmaceutical accepted carrier/vehicle for anti-cancer therapies, including gene therapy, to treat neuroblastoma and retinoblastoma but not limited to these tumor types.
  • According to another aspect, substantially homogenous LP of hIDPSCs comprise a cell that co-expresses a marker selected from, but not limited to, the group comprising SOX1, SOX2, or beta-3-tubulin.
  • According to some aspects, contemplated disclosures are provided an undifferentiated hIDPSC wherein the cell is immunoreactive with markers for human pluripotent stem cells including Oct3/4, Nanog and SOX2, and wherein said cell may differentiate under differentiating conditions to neural cells. Preferably, the cells express the transcription factor SOX1; SOX2 and tubulin as demonstrated by RT-PCR. The said cells maintain a stable diploid karyotype during prolonged cultivation in vitro. Sox and Oct3/4 transcription factors are thought to be central to the transcriptional regulatory hierarchy that specifies ES cell identity. hIDPSC were previously shown to express Oct 4 (Insert ref 2006; and Liezer et al. 2012) approximately in about of 20% of IDPSC throughout the cell culture up to LP. Therefore, the co-existence of both OCT family and SOX family markers, indicate on high stemness and pluripotency of hIDPSC. However enrichment with SOX and beta-tubulin markers indicate on strong affinity towards neuronal fate.
  • Also disclosed herein is a preparation of undifferentiated hIDPSCs using LP harvesting method where enrichment with SOX1, SOX2, BrdU, beta-3-tubulin molecular markers in LP indicate on the capability of these LP to proliferation in vitro and differentiation (in vivo or ex vivo) into neural progenitor cells, neuron cells and/or glial cells and ganglion cells. Preferably, the undifferentiated hIDPSCs cells have the potential to differentiate into neural progenitor cells, neuron cells and/or glial cells when subjected to differentiating conditions or in vivo micro-environment.
  • The present invention is further illustrated by the following examples that should not be construed as limiting. The contents of all references, patents, and published patent applications cited throughout this application, as well as the Figures, are incorporated herein by reference in their entirety for all purposes.
  • EXAMPLES Example 1. Batch Release Process for Industrial Scale-Up of Multiharvest Organ and Tissue Explant Culture of hIDPSC—CELLAVITA™ (Stem Cells) Product by Late Population Method Nomenclature
  • CELLAVITA™ (stem cells) is the bulk material prior to final formulation. CELLAVITA™ (stem cells) is referred to as Drug Substance (DS). CELLAVITA™ (stem cells) for IV infusion is referred to as Drug Product (DP). The Process for the CELLAVITA™(stem cells) Substance initiates at donor screening and testing and finishes prior to final formulation and cryopreservation of the cell stock. Preparation of the Drug Product involves formulation of the CELLAVITA™ (stem cells) substance with additional excipients.
  • General Properties
  • In one embodiment, CELLAVITA™ (stem cells) is a stem cell expressing neural crest/mesenchymal stem/progenitor cell markers, such as CD13, CD105 (Endoglin), CD73, CD29 (integrin b-1), CD44, and nestin (Kerkis et al., 2009; Kerkis et al., 2006) obtained using multiharvest orgão and tissue explant culture.
  • In another embodiment, CELLAVITA™ (stem cells) are MSC-like cells, which possess all basic properties of these cells. The cells are defined in accordance with minimal criteria for defining multipotent mesenchymal stromal cells established by the Mesenchymal and Tissue Stem Cell Committee of the International Society for Cellular Therapy. This definition includes being plastic-adherent when maintained in standard culture conditions, expressing CD105, CD73 and CD90, and lack expression of CD45, CD34, CD14 or CD11b, CD79alpha or CD19 and HLA-DR surface molecules, and ability to differentiate to osteoblasts, adipocytes and chondroblasts in vitro (Dominici et al., 2006).
  • Manufacturing Method of Investigational Product CELLAVITA™ (Stem Cells)
  • Only healthy teeth of children aged 6-12 years will be used for the cultivation of Cellavita™. Children's legal guardians will answer an eligibility questionnaire about the child's health and blood samples will be collected for serological testing to detect infectious diseases, as recommended by the European Commission guidelines for donor eligibility (COMMISSION DIRECTIVE 2006/17/EC). The mandatory testing includes tests for HIV-1 and -2 (Anti-HIV-1 and -2), HTLV-1 and -2, HBV (specifically HBsAg, Anti-HBc), HCV (specifically anti-HCV-Ab), and Treponema pallidum (syphilis) (COMMISSION DIRECTIVE 2006/17/EC).
  • Only healthy teeth without dental diseases such as dental caries will be collected after natural loss or surgical extraction. To avoid unnecessary testing, only donors whose teeth have viable pulp for cultivation (a process determined in the laboratory after two weeks of cell culture) will be asked to return to the center for the donor eligibility test (blood collection).
  • Tooth Collection, Container, Transportation
  • Immediately after spontaneous exfoliation, the tooth will be immersed into 3 mL of sterile transporting solution composed of DMEM (Dulbecco's Modified Eagle Medium) and 500 mM Gentamycin in a 15 mL sterile centrifuge tube. The tooth will be stored at 4° C. and processed within 48-72 hours.
  • Pulp Isolation and Washing Procedure
  • A freshly exfoliated deciduous tooth from a healthy subject will be washed repeatedly in sterile solution containing 50% pen/strep solution (100 units/mL penicillin, 100 units/mL streptomycin) and 50% Phosphate Buffered Saline (PBS). Dental pulp will be removed from the tooth with the aid of a sterile needle.
  • Selection of Viable Pulps as a Raw Material for hIDPSC Stem Cell Expansion
  • Freshly obtained dental pulp (DP) will be washed in a solution containing 3% Pen/strep solution (100 units/mL penicillin, 100 units/mL streptomycin). Initial plating and viability testing of the dental pulp will be performed in dental pulp Maintenance Medium supplemented with 15% fetal bovine serum (FBS, Hyclone), 100 units/mL penicillin, 100 units/mL streptomycin, 2 mM L-glutamine, and 2 mM nonessential amino acids. This procedure usually takes up to one week. Once the DP will be determined as viable it will be harvested and hIDPSC will be passaged. The resulting hIDPSC will be cryopreserved under GTP conditions for future clinical research
  • Pharmaceutical Development
  • A small scale, non-GMP batch of CELLAVITA™ (stem cells) was produced in Dr. Irina's lab for obtaining patent and generating nonclinical data. The technology was transferred to a semi-industrial, non-GMP facility in Israel (Sheba Hospital). The CMC content in this briefing document describes the manufacturing process in Israel, and is also representative of the plan to manufacture clinical batches in a GMP facility in Brazil, to where the technology will be transferred.
  • In one aspect, the production process comprises the steps illustrated in FIG. 1 which demonstrates the initial process of CELLAVITA™ (stem cells) isolation and batch formulation. The vertical pathway shows the process of dental pulp mechanical transfer (harvest) of early population-hIDPSC (isolated from dental pulp before 5 harvests) and late population—hIDPSC (isolated from dental pulp after 5 harvests). The horizontal pathway shows the traditional enzymatic method of cell cultures when cells are replaced through repetitive passages. The final batch product is a sum of hIDPSC obtained from dental pulp harvests and passages (no more then 5).
  • In another aspect, the production process comprises the steps illustrated in FIG. 2 and/or FIG. 3. The production process includes CELLAVITA™ (stem cells) isolation and batch formation. The vertical pathway shows the process of dental pulp (DP) mechanical transfer (harvest) for early population-hIDPSC isolated from dental pulp before 5 harvests and late population—hIDPSC isolated from DP after 5 DP harvests; the horizontal pathway shows traditional enzymatic method of cells culturing, when cells are replaced through repetitive passages. Final batch—product is a sum of hIDPSC obtained from DP harvest and passages (no more then 5).
  • Description of the Proposed Manufacturing Process Controls for the Clinic
  • The production of CELLAVITA™ (stem cells) will be performed in a state of the art clean room facility and the process will be performed according to GMP regulations. In one embodiment, this production process follows the steps outlined in FIG. 4.
  • hIDPSC Harvesting
  • When the semi-confluent colony formation of hIDPSC will be detected around the dental pulp explant, DP will be transferred into a new cell culture vessel for continued growth in DP Maintenance Medium.
  • hIDPSC Passaging
  • hIDPSC will be washed with sterile PBS, removed with TrypLE solution and centrifuged. The pellet will be resuspended in DP Maintenance Medium and thereafter will be seeded in the tissue culture flask (Passage 1-P1). When the cells reach about 80% confluency they will be passed to the new flask (Passage 2-P2). Cells will be incubated in a humidified 5% CO2 incubator.
  • Safety Tests Prior to Freezing
  • hIDPSC from P5 will be maintained in culture for at least 3-5 days in order to collect cell conditioned medium for sterility and mycoplasma testing.
      • Sterility test will be performed by ISO, GMP certified
      • Mycoplasma testing will be performed using an in-house RT-PCR test (EZ-PCR Biological Industries) and by ISO, GMP certified.
    Freezing and Storage
  • The hIDPSC freezing protocol will be adapted to the standard freezing repository protocol hIDPSC from P5 will be transferred into a 2 mL cryopreservation vials containing 1 mL of freezing media, composed of: 90% FBS and 10% DMSO (GMP/US pharmaceutical grade). 1×106 cells per vial will be cryopreserved.
  • Cryopreservation vials will be placed inside a Nalgene Cryo 1° C. Freezing Container filled with isopropyl alcohol and will be placed at −80° C. overnight. Thereafter, vials will be transferred to the vapor phase of a liquid nitrogen storage tank and their locations will be recorded.
  • Control of Materials
  • Table 1 depicts the process, which will be used for control of materials.
  • TABLE 1
    Control of Materials
    Concentration Source/Country Manufacturing
    Reagent at Use of Origin Manufacturer Step
    Deciduous Tooth
    1 tooth Brazil NA Raw material
    Dulbecco Modified 500 mL Beit Haemek, Biological Transporting
    Eagle Medium-F12 Israel Industries (BI) solution
    (DMEM-F12)
    Dulbecco's 500 mL Beit Haemek, Biological Sterile solution
    Phosphate Buffered Israel Industries (BI)
    Saline without
    Calcium and
    Magnesium (DPBS)
    Gentamycin 500 mM Beit Haemek, Biological Sterile solution
    Israel Industries (BI)
    Fetal Bovine Serum 90% Washington HyClone Maintenance
    medium; freezing
    media.
    L-Glutamine Solution  2 mM Beit Haemek, Biological Maintenance
    Israel Industries (BI) medium
    Penicillin- 100,000 U/mL Beit Haemek, Biological Sterile solution
    Streptomycin penicillin and Israel Industries (BI)
    Solution 100 mg/mL
    streptomycin
    Non-essential  2 mM Beit Haemek, Biological Maintenance
    amino acids Israel Industries (BI) medium
    TrypLE 1X Gibco Cell-dissociation
    enzymes
    DMSO
    10% Sigma Cryopreservation
  • Tissue (Dental) Criteria
  • Spontaneously exfoliated deciduous teeth will be collected. Only healthy teeth without dental decay will be accepted. In order to prevent unnecessary donor testing, only donors which have a viable pulp culture (as determined by the study laboratory staff after initial plating and culture establishment, stages which take approximately two weeks) will be requested to make a second study visit in order to have blood samples drawn for donor eligibility screening. The Sponsor estimates that viable pulp cultures will be established from approximately 50% of recruited donors.
  • The parents/guardians of the subjects will answer a donor eligibility questionnaire concerning the health of their child. In addition, blood samples (total volume of 10 mL) will be drawn in order to test the donors for the following pathogens according to guidelines for eligibility screening:
      • HIV 1&2 (specifically Anti-HIV-1,2)
      • HTLV I & II
      • HBV (specifically HBsAg, Anti-HBc)
      • HCV (specifically Anti-HCV-Ab)
      • Treponema pallidum (syphilis)
  • The total amount of blood to be drawn for these tests will not exceed 0.8 mL/kg body weight for one blood draw.
  • Tooth Collection, Container, Transportation
  • Immediately after spontaneous exfoliation, the tooth will be immersed into 3 mL of sterile transporting solution composed of DMEM (Dulbecco's Modified Eagle Medium) and 500 mM Gentamycin in a 15 mL sterile centrifuge tube. The tooth will be stored at 4° C. and processed within 48-72 hours in the GTP laboratory.
  • The hIDPSC freezing protocol will be adapted to the standard freezing repository protocol hIDPSC from P5 will be transferred into a 2 mL cryopreservation vials containing 1 mL of freezing media, composed of: 90% FBS and 10% DMSO (GMP/US pharmaceutical grade). 1×106 cells per vial will be cryopreserved.
  • Cryopreservation vials will be placed inside a Nalgene Cryo 1° C. Freezing Container filled with isopropyl alcohol and will be placed at −80° C. overnight. Thereafter, vials will be transferred to the vapor phase of a liquid nitrogen storage tank and their locations will be recorded.
  • Thawing and Administration of CELLAVITA™ (Stem Cells)
  • Prior to administration of CELLAVITA™ (stem cells) in patients, the cryopreserved product after thawing must be assessed for cell viability. This will be done with a kit that will be sent along with the CELLAVITA™ (stem cells). The kit and the method to be performed must be followed as described below.
  • Composition of Thawing Kit
      • Conical falcon tube containing 15 ml thawing substance;
      • Two conical 15 ml falcon tubes containing 5 ml PBS;
      • Six sterile Pasteur pipettes
    Thawing Protocol
      • The CELLAVITA™ (stem cells) product should be thawed in a laminar flow system.
      • The Cryotubes containing CELLAVITA™ (stem cells) will be transferred to a bath of water at 37° C. (2-3 minutes).
      • After thawing, the product of cryotubes must be removed with a sterile Pastuer pipette and immediately transferred to a 15 ml conical tube containing 5 ml defrosting substance. The solution should be thoroughly mixed slowly for 5 to 10 times with a Pasteur pipette. Centrifuge product for 5 minutes at 1000 rpm.
      • The supernatant should be discarded. The precipitated cells must be added to a Falcon tube with PBS solution. The cells are homogenized slowly for 5 to 10 times. Centrifuge the cells for 5 minutes at 1000 rpm. Repeat this procedure twice.
      • At the end of this procedure the cells are ready to be administered to the patient.
    Administration to the Patient
  • For administration of CELLAVITA′ (stem cells) product, enough cells/kg body weight will be transferred to a bag of 250 ml of 0.9% saline. This solution will be infused intravenously (peripheral vein) over a period of 10 to 40 minutes.
  • Example 2. Batch Numbering System
  • Each donor IDPSC culture will be defined as a separate batch of raw material for future stem cell expansion process. hIDPSC will be individually coded and labeled, in an anonymous fashion. Cryotubes boxes will be double-packed to reduce risk of cross contamination, according to process.
  • The purpose of the batch definition is to ensure consistency and traceability. The following label will be attached to cryopreservation vial:
  • Example 3. Characterization of Early and Late Harvests IDPSCs and Derivation of Neural and Glial Cells from the Early and Late Harvest IDPSCs Characterization of Early and Late Harvests IDPSCs
  • To characterize the properties of the hIDPSC, from the early (n=8) and late (n=4) harvests (Table 2), flow cytometric analyses for the mesenchymal markers CD13, CD105, CD73, CD90, CD44 was performed. FACS experiments were performed with 2×105 cells. Cells were washed twice with PBS (without calcium and magnesium) and the tested antibodies were added for 15 minutes at room temperature. The cells were then washed twice with cold PBS and analyzed with a Becton-Dickinson flow cytometer. The fluorescence of PE (FL2), FITC (FL1), APC (FL4) were detected in 575 nm, 53 nm, and 600 nm emission wavelengths, respectively.
  • TABLE 2
    List of cells used in the FACS experiments
    Batch Harvest Passage
    Number Number (H) Number (P)
    Early Harvests:
    1 0 3
    6 0 3
    10 0 3
    11 0 3
    17 0 3
    22 0 3
    24 0 3
    26 0 4
    Late Harvests:
    11 13 3
    11 16 9
    24 13 3
    26 10 2
  • Cells from both early and late harvests expressed high levels of mesenchymal markers. Both populations were negative for HLA-DR and HLA-ABC antigen expression which allows for allogenic transplantation of these populations of cells. Both populations were double immunopositive for mesenchymal stem cell markers, such as, CD13 and CD44 and others, as well as expressed nestin, P75 (CD271), neuroepithelial stem cell markers, and nerve growth factor (NGF) (see FIGS. 5 and 6A-6L and Table 3) and they were negative for CD146 and HLA-ABC.
  • After mutiharvest of DP tissue and explant culture, IDPSC demonstrate capacity to form colonies (FIG. 7). The colony forming assay (CFU-F) assay was performed in triplicate at T20, P3 using 480 cells seeded in each plate, at day 8 multiple colonies colonies appeared and approximately 100 colonies were formed in each plate (FIG. 7). Colony forming capacity is one of the principal characteristics of stem cell. Therefore, we conclude that this capacity was maintained when the cells were obtained using the disclosed multiharvest organ and tissue explant method. Additionally, the proliferative activity of LP IDPSC was evaluated as shown on FIG. 8, and these cells demonstrated a very high proliferative rate.
  • TABLE 3
    Comparison of the cell marker expression from the hIDPSC
    derived from the same donor by FACS analysis.
    % of Fluorescence
    Markers H0P3 H13P3 H16P9
    CD105 99.4 99.76 99.6
    CD13 99.5 99.1 99.3
    CD73 95.1 99.88 85.3
    CD90 99.9 99.9 99.7
    CD44 99.12 99.45 96.4
    HLA-DR 0.8 3.03 0.4
    HLA-ABC 0.2 0.1 0.3
    NGF 18.7 55.96 49.5
    Nestin 54.5 66.57 30
    ABCG2 1.88 ND 8.1
    ABCB1 24.8 ND 45.6
  • Table 3 compares the cell marker expression from the different harvests of hIDPSC derived from the same donor (i.e., H0P3=first harvest; H13P3=later harvest; H16P9=last harvest). Late harvest populations had greater levels of NGF than the early harvest population. Expression of adenosine triphosphate binding cassette (ABC) transporters was also tested in the cells. ABC transporters are involved in the active transport of an extremely diverse range of substrates across biological membranes. These transporters are commonly implicated in the development of multidrug resistance and are also involved in numerous physiological and homeostatic processes, including lipid transport, cell migration and differentiation. Moreover there is evidence that ABC transporters serve as phenotypic markers and functional regulators of stem cells (Bunting 2002). Both early and late harvests populations expressed ABCB1 protein, the product of MDR1 gene, but expression was higher in the late harvest. According to Islam et al. (2005), ABCB1 is expressed in human fetal neural stem/progenitor cells (hNSPCs).
  • FACS analysis show that LP of IDPSCs (batch #11) comprises approximately 80% cells that express BDNF and DARP 32, while EP is negative for these markers (data not shown) and very low number of the cells, which express D2 (FIG. 7). To further characterize hIDPSC early and late harvests, total brain-derived neurotrophic factor (BDNF) levels represented by the amount of BNDF in medium was determined using ELISA (Table 4). BDNF levels were quantified by using a human BDNF Quantikine ELISA kit according to the manufacturer's protocol (R&D Systems, Minneapolis, Minn.). Cells (1×106) from different harvests were inoculated in 75 cm plastic flasks. The supernatants were harvested approximately 4 days after inoculation. The results were expressed as the BDNF concentration. BDNF was secreted in all cell subsets, but the levels were 4-10 folds higher in the late harvests.
  • TABLE 4
    Total BDNF levels in early and late harvests of hIDPSC
    Batch Harvest (H) and BDNF levels
    number Passage (P) Number (pg/ml)
    11 H0 P3 13
    24 H10 P2 154
    26 H13 P3 43

    Comparison with Other MSCs Shows that hIDPSCs Secrete Much More BDNF.
  • The average level of BDNF secreted by 1×106 hIDPSCs is 6589 pg, which is many times higher than other types of MSCs that secrete BNDF, such as the MSCs of Gothelf et al. in 2014 (Clin Transl Med. 2014 3:21). Gothelf et al. induced bone marrow-derived MSCs (BM-MSC) to differentiate into neurotrophic factor-secreting cells (BM-MSC-NTF) by incubating the BM-MSCs for 72 hours in medium containing 1 mM dibutyryl cyclic 15 AMP (cAMP), 20 ng/ml human Basic Fibroblast Growth Factor (hbFGF), 5 ng/ml human platelet derived growth factor (PDGF-AA), and 50 ng/ml human Heregulin β1. Although the induction medium nearly doubled BDNF secretion (827 pg BNDF/106 BM-MSC compared to 1640 pg BNDF/106 BM-MSC-NTF cells), hIDPSCs still secreted four times more BDNF than BM-MSC-NTF. Accordingly, the hIDSPCs have much greater neuroprotective potential than bone marrow-derived MSC induced to secrete neurotrophic factors.
  • Expression of Oct4, Nanog, Sox2 and p53
  • It is known that MSCs generally express pluripotent markers such as Oct4, Nanog and Sox2 at low levels as described in the literature (Jiang et al., 2002; Guillot et al., 2007). We showed that hIDPSC express very low levels of such markers in comparison with human embryonic stem cells and even induced pluripotent stem cells obtained from hIDPSCs (see FIG. 9). More importantly, we demonstrated that hIDPSC express a high level of p53. The tumor suppressor gene p53 is well known as a master regulator that helps keeps cancer at bay. Blocking the p53 pathway vastly improves the ease and efficiency of transforming differentiated cells into induced pluripotent stem cells (Dolgin, 2009).
  • Derivation of Neural and Glial Cells from the Early and Late Harvests IDPSCs
  • The neuronal system consists of two classes of neural precursor cells: neuronal NPCs that differentiate into neurons and glial NPCs that differentiate into glia. Both neuronal and glial NPCs descend from the same neuroectodermal precursor. A third class of neural precursor cells, neuroglioblast, was also suggested. This third class of cells include radial glial cells also can act as neuronal precursors and only later, after neurogenesis, do they shift towards an exclusive generation of astrocytes.
  • The ability to distinguish whether a population of cells in cell therapy are neuronal NPSs or glial NPCs is of extreme importance for developing an efficient cell therapy strategy for treating neurodegenerative diseases, which mainly involve the damage or loss of both glia and neurons. It is possible to test known cell populations for the potential to differentiate into neurons or glia by inducing these cells to differentiate.
  • The capacity of EP (early population) and LP (late population) IDPSC to produce neurons and glias was tested by inducing neuronal differentiation in EP and LP IDPSC at early (P2) and late passages (P7) according to the previously described protocol (Kerkis et al., 2006) (FIG. 10). After 7 days, the cells were collected and analyzed by flow cytometry using GFAP (glial fibrillary acidic protein) and beta-III-tubulin antibodies, respectively. A significant difference exists in the number of cells that express these markers between EP (B-E, left) and LP (B-E, right), which were established following a dental pulp (DP) harvesting protocol. On the other hand, no significant difference in expression of both proteins was detected between different passages (P2 and P7) obtained following enzymatic digestion, (FIG. 10). Surprisingly, IDPSCs can be neuronal and glial NPCs. Early DP harvesting (EP IDPSC) leads to isolation of neural progenitor cells committed mainly to glial differentiation while late DP harvesting (LP IDPSC) leads to isolation of neural progenitor cells committed mainly to neuronal differentiation.
  • Thus DP harvesting is important for establishing a population of NPCs with the potential to develop into neurons and glia. SHED, which are stem cells from human deciduous teeth, cannot be categorized as an early population or late population because they are stem cells isolated from dental pulp cells without DP harvesting. The single SHED population contains neuron-committed and not glial-committed progenitors. In Miura et al. (2003) neuronal differentiation of SHED resulted in increased expression of beta-III-tubulin, GAD, and NeuN while the expression of nestin, GFAP, CNPase, and NFM remained the same after the induction of differentiation (see FIG. 41 of Miura).
  • Example 4. Comparison of IDPSC with SHED and with Other Therapeutic Stem Cells which can be Potentially Used for the Treatment of Neurological Conditions 1. Comparison of IDPSC and SHED
  • MSCs from different sources (e.g. bone marrow and adipose tissue) can respond differently to various stimuli (Fraser J K et al., 2006). Culture conditions (e.g., media supplemented with either human serum or fetal calf serum (FCS), or serum-free) may also affect the differentiation potential of even MSCs of the same origin (Lindroos et al., 2011; Lizier et al., 2012). Different isolation and culture protocols used by different groups may account for the predominance of a particular MSC subpopulation with a distinct differentiation potential (Ho et al., 2008; Pevsner-Fischer et al., 2011; Rada et al., 2011).
  • SHED and IDPSCs have different methods of isolation and come from different stem cell niches. So it is unsurprising that SHED and IPSCs also have different expression of stem cell markers (see Table 5). SHED originated from a perivascular environment and STRO-1 and CD146 positive cells were found to be located around blood vessels of the remnant pulp by immunohistochemical staining. Only a minor proportion (9%) of ex vivo expanded SHED stained positive for the STRO-1 antibody using FACS.
  • TABLE 5
    Differences between SHED and IDPSCs
    SHED IDPSC
    Perivascular niche Perivascular niche
    Nerve plexus
    Subodontoblastic plexus niche
    Cell-free and cell-rich zones
    whole dental pulp (DP) minced pulp
    Enzymatic digestion: 1 hour Organ culture, explant method,
    stem cell migration from stem
    cell niches
    Can be isolated one time from DP is preserved and in vitro
    the same DP, which is descarted transferrable up to 60 times to
    after enzymatic digestion result in 60 stem cell isolations
    Culture medium: Alpha Culture medium: Dulbecco's
    modification of Eagle's medium modified Eagle's medium (DMEM)/
    (GIBCO/BRL) supplemented with: Ham's F12 (1:1) supplemented with:
    20% FCS 15% fetal bovine serum
    100 μM l-ascorbic acid 2- 100 U/ml penicillin
    phosphate 100 g/ml streptomycin
    2 mM l-glutamine 2 mM L-glutamine
    100 units/ml penicillin 2 mM nonessential amino acids
    100 μg/ml streptomycin
    Single-cell suspension Outgrowth
    Use of cell strainer
    Assess only to outer layer of Assess to outer and inner part of DP
    DP and very close layers
    Principal markers: Principal markers:
    Perivascular Mesenchymal stem cells (MSC)
    Embryonic stem cells (ES cells)
    Neuronal precursors
    Perivascular
    Osteogenic differentiation Not required
    required BMP-4
    Neurogenic differentiation Not required
    required EGF, FGF, and rat serum
    Chondrogenic differentiation Not required
    required TGF-β3 and bFGF or
    TGF-β
    High passages are needed in DP multiple transfer ensure sufficient
    order to obtain number of SHED IDPSC number at low passages
    sufficient for cell therapy
  • The requirements for inducing differentiation are also different between SHED and IDPSCs. For example, to induce neuronal differentiation, SHED need EGF 20 ng/ml (BD Bioscience), FGF 40 ng/ml (BD Bioscience) and 3% rat serum. They need four weeks in neural inductive culture in order to show neural morphology and to increase expression of neuronal markers.
  • In part because of these differences, IDPSCs have advantages over SHED regarding neurogenesis. In one study, SHED were injected into the dentate gyrus of the hippocampus of immunocompromised mice. The data demonstrated that SHED were able to survive for more than 10 days in mice hippocampus and to express NFM, which were expressed also by undifferentiated SHED (Miura et al., 2003). In another study, pre-differentiated SHED (SHED-derived spheres created by a combination of EGF and bFGF for 7 days in vitro) were transplanted into Parkinsonian rats. The cell suspension (200,000/μL) was injected into 2 DA-depleted striatum sites in rats (2.5 μL per site). Modest differentiation into DA neurons was observed (Wang et al, 2010). In a third study, SHED were injected in injured brain of postnatal day 5 mice, which were induced with in perinatal hypoxia-ischemia (HI) that has high rates of neurological deficits and mortality. Cyclosporine A was used to protect engrafted cells from the xenogeneic host immune response, nevertheless eight weeks after transplantation the engrafted SHED, had no or few cells differentiated into neurons, oligodendrocytes, or astrocytes (Yamagata et al. 2013).
  • The common theme across all three experiments is that SHED were administrated together with Cyclosporine A. Cyclosporine A is shown to decreases the size of the ischemic brain infarct in rats and to protects against synaptic dysfunction and cell death in rodent models of traumatic brain infarct as well as to protects striatal neurons from mitochondrial dysfunction in Huntington disease (Matsomoto et al., 1999; Albensi et al. 2000; Leventhal et al., 2003). Therefore, benefits observed in Parkinsonian rats and HI cannot be purely attributed to SHED but also to Cyclosporine A intervention.
  • 2. Comparison with Other Therapeutic Stem Cells for the Treatment of Neurological Conditions
  • As shown in Table 6, hIDPSCs from Avita International LTD are advantageous over other therapeutic stem cells on the market or in clinical trial. Avita International LTD's hIDPSCs have a good safety profile with low risk of immunogenicity and a low cost of production as they can be cryopreserved.
  • TABLE 6
    Comparison of commercially produced stem cells.
    BrainStorm Cell Avita
    Therapeutics NeuralStem Kadimastem International
    Company (ticker) Inc. (BCLI) Inc. (CUR) (KDST) LTD
    Cell Source Autologous MSC Allogenic; 8- hESCs (embryo) hIDPSCs from
    from bone week-old fetal and iPSCs (adult) dental pulp
    marrow spinal cord-
    derived cells
    Modifications Induction of No Induction to No
    neurotrophic differentiate into
    factor secretion astrocyte
    precursor cells
    Cell Safety Profile Good with low Less safe with Less safe with Good with low
    risk of risk of unwanted risk of unwanted risk of
    immunogenicity differentiation differentiation immunogenicity
    (teratoma) and/or (teratoma) and/or
    risk of rejection risk of rejection
    Immunosuppression Not required Required Required Not required
    Cryopreservation Not yet - Cell can be Cells can be Cell can be
    studying the expanded and frozen in expanded and
    feasibility of frozen differentiated frozen
    cryopreservation state
    of MSCNCs
    during early
    phase expansion
    Clinical Trials Compassionate Phase I/II begins Not yet in clinical On going Phase
    Care: Phase I/II in 2014 trials I (Brazil)
    complete (Israel); (Mexico); Phase
    Phase II II ends in
    underway (US) 4Q2014 (US)
    Cost High Low Low Low
  • Example 5. Product Description and Specifications
  • Table 7 depicts CELLAVITA™ (stem cells) specifications.
  • TABLE 7
    Drug Product Release Monograph
    Method of Analysis Characteristics Specification
    Appearance- Morphology Normal fibroblast like
    Morphological morphology under
    test inverted microscope
    inspection
    Cell viability Viability >95%
    via Trypan Blue
    exclusion
    Cell doublings Cell doublings number At least doubling of
    cell number in 24 hours
    CFU Cell forming units assay >5 colonies
    Sterility Microbial contamination No growth detected
    (21CFR/EP/USP) after 14 days
    Endotoxins (LAL) Less than or equal to <0.005 EU/mg A280
    1.0 EU/mg A280 protein protein
    PCR Mycoplasma Mycoplasma detection Undetectable
    test Culture - no growth
    detected
    Impurities
    FACS analysis Allogeneic marker Negative to HLA
    class II
    FACS analysis HSC marker Negative to CD34
    FACS analysis Phenotype analysis MSC Positive to CD73,
    CD105
    FACS analysis Phenotype analysis Positive to NGF,
    neuronal factors nestin
    Assay
    ELISA assay Neuronal factors Positive to BDNF
  • Analytical Procedure Safety QC—Mycoplasma Test
  • Mycoplasma tests will be performed regularly during cultivation of hIDPSC with an in-house RT-PCR test (EZ-PCR Biological Industries, Israel) according to the manufacturer's protocol.
  • Safety Characteristics—Karyotype Analysis
  • Karyotype analysis will be performed several times showing karyotype stability and these data are already published (Kerkis et al., 2006; Beltrão-Braga, 2011; Lizier et al., 2012). Additionally, independent karyotype analysis will be performed in Cytogenetics.
  • Safety and Identity QC—Flow Cytometric Analysis of Cell Surface Antigens
  • Immunostaining of cell surface markers will be carried out with monoclonal antibodies against various surface antigen markers: HLA-DR-FITC, CD44-FITC, CD45-APC, CD105-PE, CD73-FITC, CD90-APC (eBioscience CA, USA), SOX2-PE, Nestin-PE, Tubulin-APC, NGF-PE (R&D systems, MN, USA). 2×105 cells will be used for FACS experiments. Cells will be washed twice with PBS (w/o Ca and Mg) and suspended in 50 μl PBS. Cells will be then incubated with antibodies for 15 min at room temperature. The cells will be washed twice with PBS and analyzed with a Becton-Dickinson flow cytometer. The fluorescence of PE (FL2), FITC (FL1), APC (FL4) will be detected in 575 nm, 530 nm and 600 nm emission wave lengths, respectively.
  • Activity Bioassay QC—ELISA Assay
  • BDNF levels will be quantified by using a human BDNF Quantikine ELISA kit, according to the manufacturer's protocol (R&D Systems, MN, USA).
  • 1×106 cells from different harvests will be inoculated in 75-cm2 plastic flasks. The supernatants were harvested approximately 4 days after inoculation. The results were expressed as the BDNF concentration.
  • Batch Analysis
  • Table 8 depicts batch Number 001H1-30/P1-5/F analysis.
  • TABLE 8
    Batch Number 001H1-30/P1-5/F
    Method of Analysis Characteristics Specification Result
    Characteristics
    Appearance- Morphology Normal fibroblast like Confirms
    Morphological test morphology under
    inverted microscope
    inspection
    Cell viability via Trypan Viability Confirms
    Blue exclusion
    Cell doublings Cell doublings number At least doubling of cell Confirms
    number in 24 hours
    CFU Cell forming units assay >5 colonies Confirms
    Safety
    Sterility (21CFR/EP/USP) Microbial contamination No growth detected after Confirms
    14 days
    Endotoxins (LAL) Less than or equal to 1.0 <0.005 EU/mg A280 Confirms
    EU/mg A280 protein protein
    PCR Mycoplasma test Mycoplasma detection Undetectable Confirms
    Culture - no growth
    detected
    FACS analysis Allogeneic marker Negative to HLA-DR Confirms
    FACS analysis HSC marker Negative to CD34 Confirms
    Identity: mesenchymal stem cell markers and neuronal markers
    FACS analysis Phenotype analysis MSC Positive to CD13, CD73, Confirms
    CD105
    FACS analysis Phenotype analysis Positive to NGF, nestin Confirms
    neuronal factors
    Activity Bioassay
    ELISA assay Neuronal factors Positive to BDNF Confirms
  • Stability
  • Avita performed non GMP, non GLP studies regarding hIDPSC stability. For this purpose a single master cell bank, which may mitigate variability of the final batch, was established. It was composed by 5 batches, each derived from Dental Pulp of one individual. The cells were produced as described in FIG. 1. The time line of stability studies of hIDPSC is presented on FIG. 11.
  • The expression of stem cell markers, dynamics of cell proliferation, and differentiation capacity of hIDPSC derived from four batches before cryopreservation as well as migration and biodistribution in different organs after injection into Nude mice were studied. CELLAVITA™ (stem cells) showed that under standard culture conditions these cells at passage 6 from four independent batches express surface markers of mesenchymal stem cells (MSC) such as CD105, CD73, and CD13. Nevertheless, they lack the expression of CD45, CD34, CD14, CD43, and of HLA-DR. These cells were able to undergo spontaneous and induced in vitro differentiation into osteoblasts, adipocytes and chondroblasts, muscle cells, and into neurons in vitro. After transplantation into normal mice, these cells showed significant engraftment in liver, spleen, brain and kidney, among others.
  • Stability Program Development
  • Past experience showed that both the initial cell Poll (primary cells) and its final blend (after P5 expansion and mixing of all transfers) are stable when cryopreserved at −192° C.
  • The stability program being developed by the company Cellavita Pesquisas Cientificas Ltda. Scientific Research Ltda. for CELLAVITA™ (stem cells) product will be based on:
      • Time to perform;
      • Steps to be carried out;
      • Study Conditions;
      • Analysis and specifications.
        The basic program should be completed before the start of the Phase 2 proof of concept (POC) clinical study.
    Example 6. 3-Nitropropionic Acid (3-NP) Rat Model of Huntington Disease Ethical Issue
  • All studies were approved by the ethics committee of the Nuclear and Energy Research Institute (Instituto de Pesquisas Energéticas e Nucleares—IPEN), University of Sao Paulo, Sao Paulo, Brazil. Protocols concerning the maintenance, care, and handling of experimental animals are in accordance with all Brazilian current legislation and with internationally recognized norms and protocols. All staff working with experimental animals were fully accredited as a staff researcher/technician and were properly trained in the use of animals for experimental scientific purposes in accordance with current Brazilian regulations
  • Main Goal
  • The research group tested the neuroprotective and/or neural tissue remodeling effects of hIDPSC in a 3-NP chemical model of Huntington's disease.
  • Animal Model
  • Systemic administration of the mitochondrial toxin 3-nitropropionic acid (3-NP) serves as a chemical model of Huntington's disease in rodents and non-human primates and has been used to test potential drug therapies. 3-NP is an irreversible mitochondrial succinate dehydrogenase (SDH) inhibitor that causes cell death mainly in the striatum and also in GABAergic medium spiny projection neurons and spiny interneurons. Because of its ability to cross the blood-brain barrier, 3-NP can be administered systemically, causing selective neurodegeneration of the striatum or the entire corpus striatum. Depending on the drug regimen, 3-NP administration can simulate different stages of Huntington's disease. Intraperitoneal injections of two 3-NP doses lead to hyperkinetic symptoms in mice in the early stages of disease, whereas four or more doses result in hypoactivity in the late stages of disease (Beal et al., 1993; Brouillet et al., 1995; Yang et al., 2008; Borlogan et al., 1997).
  • It should be noted that the 3-NP-treated animals (chemical model) has an important limitation if compared to transgenics. In 3-NP model, the striatum lesion can regenerate spontaneously, after 10-12 days because of the presence of normal intrinsic neuronal precursors and an absence of genetic background which provides constant neurodegeneration. Therefore, difference in motor and functional improvements between experimental and control groups can be observed before spontaneous neuroregeneration.
  • Brief Protocol of hIDPSC Transplantation
  • Lewis rats (n=124) weighing 350-450 g were injected 20 mg/kg 3-NP intraperitoneally (IP) once daily for four days. The animals were kept under a light/dark cycle for 12 h and given free access to food and water. Rats were injected IP with 3-NP to induce brain injuries. Next, they were anesthetized and injected into the caudal vein with either one or three doses of 1×106 each in 250 μl of saline solution or 1×107 in 300 μl of saline solution hIDPSCs per animal, which corresponds to 0.35×106 and to 3.5×106 per kg, respectively. Multiple doses were administered 30 days apart (FIG. 12).
  • Each treatment group was paired with a control group that received saline solution only (‘untreated’). Thus, animals were grouped into five groups as shown in Table 9.
  • TABLE 9
    Groups and number of the animals used in the present study.
    Animals
    Total composed
    Groups Treatment number Deaths this study
    1 (n = 40) Treated = 1 × 106 hIDPSCs in a single 19 2 17
    administration (n = 19)
    Untreated = saline solution (n = 20) 21 0 20
    2 (n = 40) Treated = 1 × 106 hIDPSCs in three 19 5 14
    administrations (n = 19)
    Untreated = saline solution (n = 21) 21 9 12
    3 (n = 21) Treated = 1 × 107 hIDPSCs in a single 10 0 10
    administration (n = 10)
    Untreated = saline solution (n = 11) 11 0 11
    4 (n = 23) Treated = 1 × 107 hIDPSCs in three 14 5 9
    administrations (n = 14)
    Untreated = saline solution (n = 9) 9 3 6
    5 (n = 10) Control = no administration of 3-NP, 10 0 10
    saline solution, or hIDPSCs (n = 10)
  • Functional Analysis Semi-Quantitative Neurological Scale
  • Ambulatory abilities were assessed twice a week by one blinded observer for each experimental group using the quantitative neurological scale adapted from Ludolph et al., (1991). This scale measures the ambulatory behavior (scored 0-4) of rats on a flat wooden surface as follows: 0: normal behavior; 1: general slowness; 2: incoordination and gait abnormalities; 3: upper limb paralysis or impairment, inability to move; and 4: inability to leave the lying position.
  • At baseline, all rats (30) in both groups exhibited normal behavior with no marked gait abnormality before treatment with 3NP, thus receiving a score 0. At the end 3NP administration (4 days) end of the of 3NP induction, a total of the 76 rats presented general slowness (score 1); 2 rats presented difficult to move to move (score2); 20 rats exhibited incapacity to move resulting from forelimb and hindlimbs impairment (score 3); 24 rats presented recumbence and consequently death (score 4). After 24 hours HIDPSC transplantation, the rats treated with 3NP+hIDPSC (both doses) performed better compared to rats treated with 3NP. However, 5-days after hIDPSC transplantation the rats exhibited better improvement after HDPSC transplantation. Total of 27 rats presented normal score (score 0). Furthermore 20 rats presented score 1, 3 score 2 and 2 score 3 and no rats presented score 4 after HIDPSC transplantation (Table 11). While control group (3NP+saline solution) 38 rats presented score 1, 1 score 2 and 5 score 3 and 1 rat presented score 4 (Table 10).
  • TABLE 10
    Neurological rating scale scores after 5 days
    end 3NP treatment (4 days). Scores are for rats
    in the control group (3NP + saline solution).
    Group Score 0 Score 1 Score 2 Score 3 Score 4
    GI 0 16 0 0 0
    GII 0 9 0 3 0
    GIII 0 10 0 1 0
    GIV 0 3 1 1 1
    TOTAL 4 38 1 5 1
  • TABLE 11
    Neurological rating scale scores after 5 days end 3NP treatment (4
    days). Scores are for rats in the treatment group (3NP + hIDPSC).
    Group Score 0 Score 1 Score 2 Score 3 Score 4
    GI 8 11 0 0 0
    GII 10 3 0 1 0
    GIII 7 3 0 0 0
    GIV 2 3 3 1 0
    TOTAL 27 20 3 2 0

    Table 12 depicts neurological score of control group (3NP+saline solution) and hIDPSC group (3NP+hIDPSC transplant) after 3NP treatment (4 days of administration), 1 day and 5 days after 3NP induction and after HIDPSC transplantation. Normal behavior with no gait abnormalities (score 0); general slowness (score 1); incoordination and gait alterations (score 2); inability to move either the hind limbs or forelimbs (score 3); and inability to leave the lying position. This last group eventually died (score 4). Group 1=Treated=1×106 hIDPSCs in a single administration; Group 2=Treated=1×106 hIDPSCs in three administrations; Group 3=Treated=1×107 hIDPSCs in a single administration Group 4=Treated=1×107 hIDPSCs in three administrations.
  • TABLE 12
    Neurological scors of various groups of rats.
    neuro- neuro-
    neuro- logical logial
    dose and logical score score
    HIDPSC and score AF 1 AF 5
    Animal Treatment frequency AF 3NP DAY DAYS
    GROUP I
     1 3NP + SAL GI 1 1 1
     2 3NP + SAL GI 1 1 1
     3 3NP + SAL GI 1 1 1
     4 3NP + SAL GI 1 1 1
     5 3NP + SAL GI 1 1 1
     6 3NP + HIDPSC GI 1 1 1
     7 3NP + HIDPSC GI 1 1 1
     8 3NP + HIDPSC GI 1 1 1
     9 3NP + HIDPSC GI 1 1 1
    10 3NP + HIDPSC GI 1 1 1
    11 3NP + SAL GI 1 1 1
    12 3NP + SAL GI 1 1 1
    13 3NP + SAL GI 1 1 1
    14 3NP + SAL GI 1 1 1
    15 3NP + SAL GI 1 1 1
    16 3NP + HIDPSC GI 1 1 1
    17 3NP + HIDPSC GI 1 1 1
    18 3NP + HIDPSC GI 1 1 1
    19 3NP + HIDPSC GI 1 1 1
    20 3NP + HIDPSC GI 1 1 1
    64 3NP + SAL GI 1 1 1
    65 3NP + SAL GI 1 1 1
    66 3NP + SAL GI 1 1 1
    67 3NP + SAL GI 3 3 1
    68 3NP + HIDPSC GI 1 0 0
    69 3NP + HIDPSC GI 1 0 0
    70 3NP + HIDPSC GI 1 0 0
    71 3NP + SAL GI 1 1 1
    72 3NP + SAL GI 1 1 1
    73 3NP + SAL GI 1 1 1
    74 3NP + SAL GI 1 1 1
    75 3NP + SAL GI 1 1 1
    76 3NP + HIDPSC GI 1 1 1
    77 3NP + HIDPSC GI 1 0 0
    78 3NP + HIDPSC GI 4 4 death
    79 3NP + HIDPSC GI 1 0 0
    80 3NP + HIDPSC GI 1 0 0
    81 3NP + HIDPSC GI 1 0 0
    82 3NP + HIDPSC GI 1 0 0
    83 3NP + HIDPSC GI 4 death
    GROUP II
    21 3NP + SAL GII 4 4 death
    22 3NP + SAL GII 3 3 3
    23 3NP + SAL GII 3 death
    24 3NP + SAL GII 3 4 death
    25 3NP + SAL GII 4 4 death
    26 3NP + HIDPSC GII 3 3 3
    27 3NP + HIDPSC GII 4 death
    28 3NP + HIDPSC GII 4 4 death
    29 3NP + HIDPSC GII 3 3 1
    30 3NP + HIDPSC GII 3 3 1
    31 3NP + SAL GII 4 4 3
    32 3NP + SAL GII 4 4 death
    33 3NP + SAL GII 4 4 death
    34 3NP + SAL GII 4 4 death
    35 3NP + SAL GII 3 3 death
    36 3NP + SAL GII 3 3 3
    37 3NP + HIDPSC GII 3 death
    38 3NP + HIDPSC GII 4 4 death
    39 3NP + HIDPSC GII 3 3 1
    40 3NP + HIDPSC GII 4 4 death
    21A 3NP + SAL GII 4 4 death
    22A 3NP + SAL GII 1 1 1
    23A 3NP + SAL GII 1 1 1
    24A 3NP + SAL GII 1 1 1
    25A 3NP + SAL GII 1 1 1
    26A 3NP + HIDPSC GII 1 0 0
    27A 3NP + HIDPSC GII 1 0 0
    28A 3NP + HIDPSC GII 1 0 0
    29A 3NP + HIDPSC GII 1 0 0
    30A 3NP + HIDPSC GII 1 0 0
    31A 3NP + SAL GII 1 1 1
    32A 3NP + SAL GII 1 1 1
    33A 3NP + SAL GII 1 1 1
    34A 3NP + SAL GII 1 1 1
    35A 3NP + SAL GII 1 1 1
    36A 3NP + HIDPSC GII 1 0 0
    37A 3NP + HIDPSC GII 1 0 0
    38A 3NP + HIDPSC GII 1 0 0
    39A 3NP + HIDPSC GII 1 0 0
    40A 3NP + HIDPSC GII 1 0 0
    GROUP IV
    84 3NP + SAL GIII 1 1 1
    85 3NP + SAL GIII 1 1 1
    86 3NP + SAL GIII 1 1 1
    87 3NP + SAL GIII 1 1 1
    88 3NP + SAL GIII 1 1 1
    89 3NP + HIDPSC GIII 1 0 0
    90 3NP + HIDPSC GIIl 1 0 0
    91 3NP + HIDPSC GIII 1 0 0
    92 3NP + SAL GIII 1 1 1
    93 3NP + SAL GIII 1 1 1
    94 3NP + SAL GIII 1 1 1
    95 3NP + SAL GIII 1 1 1
    104  3NP + SAL GIII 3 3 3
    96 3NP + SAL GIII 1 0 1
    97 3NP + HIDPSC GIII 1 0 1
    98 3NP + HIDPSC GIII 1 0 1
    99 3NP + HIDPSC GIII 2 2 1
    100  3NP + HIDPSC GIII 1 0 0
    101  3NP + HIDPSC GIII 1 0 0
    102  3NP + HIDPSC GIII 1 0 0
    103  3NP + HIDPSC GIII 1 0 0
    GROUP III
    41 3NP + HIDPSC GIV 4 4 death
    42 3NP + HIDPSC GIV 3 3 1
    43 3NP + SAL GIV 3 3 death
    44 3NP + HIDPSC GIV 3 3 0
    45 3NP + SAL GIV 4 4 death
    46 3NP + HIDPSC GIV 4 4 2
    47 3NP + HIDPSC GIV 3 3 1
    48 3NP + HIDPSC GIV 4 4 3
    49 3NP + HIDPSC GIV 4 4 death
    50 3NP + SAL GIV 4 4 death
    51 3NP + HIDPSC GIV 4 4 2
    52 3NP + SAL GIV 4 4 4
    53 3NP + SAL GIV 3 3 3
    54 3NP + HIDPSC GIV 4 4 death
    55 3NP + HIDPSC GIV 4 4 death
    56 3NP + HIDPSC GIV 3 3 1
    57 3NP + SAL GIV 3 3 2
    58 3NP + HIDPSC GIV 4 4 2
    59 3NP + HIDPSC GIV 3 3 death
    60 3NP + SAL GIV 1 1 1
    61 3NP + SAL GIV 1 1 1
    62 3NP + HIDPSC GIV 1 0 0
    63 3NP + SAL GIV 1 1 1
  • Histopathological and Immunohistological Analysis
  • Histopathological and immunohistological analyses were conducted 7, 30, and 90 days after hIDPSC injection; animals were perfused with 4% paraformaldehyde (prepared in PBS, 0.1 mol/L). Tissue fragments were dehydrated in a decreasing ethanol series (75, 95, and 100%) and stained using Nissl staining with 0.1% cresyl violet. Two antibodies, such as, anti-human nuclei and anti-hIDPSC (1:1000, Abcam Plc) were used to determine the presence of hIDPSC in rat brain. To evaluate the neuroprotective and neuroreparative effects of hIDPSC, anti-GABAergic medium spiny neurons DARPP32 (1:1000, Abcam Plc), dopamine D2 (1:800), and BDNF (1:500) antibodies were used.
  • hIPDSC Engraftment in Rat Brain
  • One of the most relevant findings of the study was that hIDPSCs were detected in the cortex and corpus striatum, indicating that they were able to cross the blood-brain barrier and migrate to the site of injury (FIG. 13). In FIG. 13, optical cuts demonstrate at different depth of focus (A1-A4) the presence of IDPSC stained with Vybrant (green), and nuclei are stained with PI (red). The cells demonstrate capillary predominant association and different morphological types: neuron-like cells and pericytes. On A2, A3, and A4 two pericytes at different locations along capillary can be observed, and both present similar morphology. On A4 embranchment of axons is shown. Neuron nuclei are light with nucleolus, and the difference with perycite nuclei, which are strongly stained, can be observed. Blue is the artificial color of confocal microscope. Microscopy was with epifluorescence+Digital Interference Contrast (DIC), and the scale bar=10 μm.
  • In addition, four days after hIDPSC administration, a few cells were positive for specific MSC antibodies (anti-CD73 and anti-CD105), indicating that some cells were still undifferentiated at that time (FIG. 14). Nevertheless, hIDPSC-derived neuron-like cells and pericytes (perivascular cells from microvessels) were also observed in the same period (FIG. 13).
  • In FIG. 14 the optical cut demonstrates IDPSC stained with Vybrant (green) and positively reacted with anti-IDPSC antibody (red). Superposition of both produce yellow color. The cell demonstrate near capillary localization. Two markers for MSC were used: CD73 and CD105 demonstrating positive reaction with IDPSC. A confocal microscope with epifluorescence+Digital Interference contrast (DIC) was used. Scale bar=A=5 μm; B=10 μm; C=20 μm; D=5 μm
  • Thirty days after hIDPSC transplantation, a few hIDPSCs were observed in the cortex and a large number of cells were observed in the corpus striatum, along the capillaries (FIG. 15). Serial cuts obtained from rat's brain demonstrates neuron like morphology of IDPSC localized in parenchyma (FIG. 15). Additionally, neuron-like and fibroblast-like cells were also observed, confirming that hIDPSCs undergo differentiation (FIG. 16). Unexpectedly the IDPSC were found also in Subventricular zone (SVZ), which is considered a stem cell niche of neurons in the adult brain (FIG. 15).
  • Neuroprotective and Neuroreparative Effects
  • 3-NP-induced striatal lesions were determined by neuron loss using Nissl staining and DARPP32 expression (FIG. 17 and FIG. 18, respectively). Nissl stains are used to identify neuron structures in the brain and spinal cord (FIG. 17), whereas DARPP32 is a cytoskeleton marker expressed in GABAergic neurons and prevalent in the striatum of healthy mammals (FIG. 18). Using these two markers, neuron loss in the corpus striatum was scored as follows:
      • Score 1 (severe): severe neuron loss, with areas of degradation, loss of DARPP32 immunostaining in the lateral striatum with little or no cells in the central striatum;
      • Score 2 (moderate): moderate neuron loss with “dark neurons” (dead or apoptotic neurons) and few DARPP32+ cells; and
      • Score 3 (mild): no neuron loss and intense DARPP32 immunostaining (Vis et al., 2001).
  • 3-NP-treated animals showed complete or partial neuron loss in the striatum compared to controls (no 3-NP or hIDPSC). The two experimental groups (treated and untreated) presented different scores for neuronal loss in the striatum relative to controls. However, morphometric histological analysis revealed that most hIDPSC-treated animals had scores 3 and 2, whereas most untreated animals (3-NP+saline solution) had neuron loss scores of 2 and 1 (FIG. 18). No animal had visible atrophy (FIG. 17 and FIG. 18).
  • FIG. 19 depicts neuronal growth in the striatum of rats after hIDPSC. Administration of hIDPSC resulted in a neuroreparative effect in hIDPSC-treated animals by (A) Nissl staining and (B) DARPP32 expression. (C) Number of animals showing neuron recovery after hIDPSC administration compared to Controls. Most hIDPSC-treated animals (3-NP+hIDPSC) had scores 3 and 2 (moderate and mild) whereas most untreated animals (3 NP+saline) had scores 2 and 1 (severe and moderate).
  • We also observed that DARPP32 expression was higher in hIDPSC-treated animals than in untreated animals (FIG. 20) indicating neuron regeneration. Optical cuts in FIG. 20 demonstrate that neuron positively reacted with DARPP32.
  • It is reported that dysregulation of dopamine receptor D2 is a sensitive measurement for HD pathology in model mice (Crook et al., 2012). A surprising, unexpected result was obtained in respect to a robust production of DARPP32 positive neurons in rats, which received hIDPSC transplantation. In contrast this was not observed in control groups (FIG. 18). It is important to note that dopamine- and cAMP-regulated neuronal phosphoprotein (DARPP-32), was identified initially as a major target for dopamine and protein kinase A (PKA) in striatum. The regulation of the state of DARPP-32 phosphorylation provides a mechanism for integrating information arriving at dopaminoceptive neurons, in multiple brain regions, via a variety of neurotransmitters, neuromodulators, neuropeptides, and steroid hormones (Svenningsson et al., 2004). HD is associated with severe striatal D1 and D2 receptor loss and taking in consideration that recently it was reported that dysregulation of dopamine receptor D2 as a sensitive measure for Huntington disease pathology in model mice (Crook et al., 2012; Chen et al., 2013), therefore we used this marker to evaluate possible effect of IDPSC in 3-NP induced rats.
  • Surprisingly, we observed significant differences in receptor D2 expression in rats, which received IDPSC in comparison with untreated groups, a few of expression of receptor D2 cells can be observed in the striatum of control animals (FIG. 21).
  • Reduced BDNF's mRNA and protein levels have been observed in the cerebellum, caudate putamen, striatum, and cerebral cortex of HD patients (Adachi et al., 2014). In the current study, BDNF expression was observed in the striatum, caudate, putamen, and subventricular zone of hIDPSC-treated animals 7 and 30 days after hIDPSC administration (FIG. 22). BDNF expression in the subventricular zone indicates that hIDPSC promoted neurogenesis. No BDNF expression was observed in untreated animals (3-NP+saline).
  • Co-localization of hIDPSC was observed with the motor neuron marker DARRP32, suggesting that hIDPSCs differentiate into mature neurons. DARPP32 expression was detected in the striatum of hIDPSC-treated animals 30 days after hIDPSC administration in this 3-NP model of HD (FIG. 23). These data indicate that hIDPSCs differentiate into GABAergic spiny neurons in vivo. It should be noted that integration of neurotransmitter and neuromodulator signals in the striatum plays a central role in basal ganglia functions. Moreover, DARPP32 is a key player in the integration of GABAergic medium spiny neurons in response to dopamine and glutamate (Fernandez et al., 2006).
  • Histological and immunohistochemical analyses revealed that hIDPSCs were able to cross the blood-brain barrier and reach different areas affected by HD, including the striatum and cortex. Morphometric histological analysis revealed that most hIDPSC-treated animals showed mild neuron loss in the striatum compared to untreated animals (3-NP+saline). Moreover, hIDPSCs showed neuroprotective and neuroreparative effects, as revealed by the upregulation of BDNF, DARPP32, and D2 receptor expression, which are downregulated in Huntington's disease (Van Dellen et al., 2000; Crook and Housman, 2012).
  • Safety
  • The following physiological parameters were recorded during the experimental period for treated and untreated animals: body weight and feed and water intake. Fewer deaths were observed among hIDPSC-treated animals than 3-NP-injected rats, indicating that hIDPSC administration is safe. In addition, the results suggest that hIDPSC administration improved overall survival by protecting animals from the neurotoxic effects of 3-NP (Table 13).
  • TABLE 13
    The number of survived verus dead animals.
    NUMBER OF NUMBER OF NUMBER OF NUMBER OF
    ANIMALS PER DEATHS PER ANIMALS PER DEATHS PER
    GROUP GROUP GROUP GROUP ANIMALS
    3NP + 3NP + 3NP + 3NP + HIDPSC 3NP + HIDPSC COMPOSED
    GROUP SAL GROUP SAL GROUP GROUP GROUP THIS STUDY
    GI (n = 40) 19 0 21 2 38
    GII (n = 40) 21 9 19 5 26
    GIII (n = 21) 11 0 10 0 21
    GIV (n = 23) 9 3 14 5 15
    Control Group 10 10
    GV (n = 10)
    TOTAL 60 12 64 12 110
  • The primary study assessing safety of hIDPSC was the 3-nitropropionic acid (3-NP) rat model of HD study. In this study, two different cell doses were injected: 1×106 and 1×107 cell/transplant or 3×106 cell/kg and 3×107 cell/kg, respectively. 3-NP-treated rats received a single IV injection or a total of three IV injections at one month intervals of the cells. Seven deaths occurred in 3-NP induced animals which received 3×106 cell/kg, and 5 deaths occurred in animals receiving 3×107 cell/kg. In placebo groups (3-NP induced without the cell transplantation) same number (12) of animals died. All rats that died presented with extremely severe disease manifestation; the deaths occurred within 5 days of 3-NP administration. No additional deaths occurred with repeated hIDPSC doses. Based on these data, probable cause of all early deaths was 3-NP toxicity. Because no deaths occurred after repetitive hIDPSC transplantation, this supports the safety of hIDPSC transplantation (Table 13).
  • Patients with Huntington's disease often exhibit progressive weight loss despite adequate or high-energy intake. Weight loss may be an indicator of 3-NP neurotoxicity caused by decreased energy metabolism (Saydoff et al., 2003; Colle et al., 2013). No significant weight loss was observed in 3-NP-treated animals four days after 3-NP administration. However 30 days after hDPSC transplant, the hDPSC group (1×106 cells dose) exhibited significantly weight gain when compared with untreated group (3NP). Thus, hIDPSC attenuated weight loss (p=0.01) (FIG. 24).
  • HD is a clinically debilitating disease for which there is no available therapy to stop or reverse disease progression. One major obstacle encountered by many therapeutics to treat HD, is that it is a neurodegenerative disorder and some targeted systemically delivered drugs would be unable to reach their target; passage of drugs to the brain is regulated by the blood-brain barrier (BBB). The BBB is a highly selective permeability barrier that separates the circulating blood from the extracellular fluid surrounding the nervous system. Treatment with cell-based therapeutics, therefore, would seem to require localized delivery bypassing the BBB (i.e. injection through the selective permeability barrier) since cells do not generally cross the BBB.
  • Extensive studies show that hIDPSCs have mesenchymal stem cell (MSC) attributes, can secrete immunomodulating and neurotropic factors. In addition, histological and immunohistochemical analyses in validated HD rat model reveal that hIDPSCs are able to cross the BBB and reach different areas affected by HD, including the striatum and cortex. Morphometric histological analysis reveals that most hIDPSC-treated animals show mild neuron loss in the striatum compared to untreated animals. Moreover, hIDPSCs show neuroprotective and neuroreparative effects by upregulating BDNF, DARPP32, and D2 receptor expression, which are downregulated in Huntington's disease (Van Dellen et al., 2000; Crook and Housman, 2012).
  • Finally, studies evaluating the safety profile of hIPDSCs show they do not form teratomas, they do not exhibit chromosomal aberration, and they are able to form human/mouse chimeras. Since the studies suggest hIPDSCs are safe and efficacious in the treatment of HD, we propose to use hIDPSCs to treat HD.
  • Example 7. Preclinical Pharmacology Studies
  • FIG. 28 summarizes the preclinical pharmacology studies, which aimed at examining the different clinical applications of the investigational product CELLAVITA™ (stem cells). Although many of the studies were conducted to investigate pharmacological efficacy for various indications, they are all supportive to demonstrate the safety profile of the product as well as for the proposed intravenous route of administration.
  • Example 8. Identification of the Parameters for Safe Systemic Administration of a Stem Cell Treatment
  • IDPSC shows Oct4 nuclear localization (FIG. 25). While a majority of IDPSCs have Nanog in the cytoplasm of cells (FIG. 25), very rarely do cells demonstrate nuclear localization as well. The intracellular localization of Sox2 in IDPSCs is mainly nuclear (FIG. 25) though several cells can show cytoplasmic localization too. Interestingly, we can observe the symmetrical division when Nanog and Sox2 expression in seen in both daughter cells, and observe asymmetric division when after division the daughter cells do not express these markers or loses the characteristic of stem cells and becomes a less potent progenitor or a differentiated cell (FIG. 25).
  • Our data demonstrate that in contrast to pluripotent stem cells, IDPSC are classified as MSC or adult stem cells. The intracellular localization of Nanog indicates that the protein is mostly inactive. This is a dramatic difference between pluripotent stem cells and adult stem cells, which express pluripotent stem cells markers. These cells are more immature than classic MSC and can differentiate to wider spectrum of the mature cells, but they are not able to produce teratoma due to the inactive state of Nanog. Our data on symmetric and asymmetric division clearly demonstrate that these cells mimic asymmetric neural stem cells division (FIG. 25).
  • Example 9. In Vivo Tumorigenicity
  • Teratomas formation is an essential tool in determining the pluripotency of any pluripotent cells, such as embryonic or induced pluripotent stem cells (ES and iPS cells). Established a consistent protocol for assessment of teratoma forming ability of the cells, was used in our studies, similar to protocol published recently by Gropp et al., 2012. Our and recently published methods are based on subcutaneous co-transplantation of defined numbers of undifferentiated mouse or human ES and iPS cells and Matrigel into immunodeficient mice. Our method was shown to be highly reproducible and efficient when 106 cells (different from Gropp et al., 2012, which used 105 cells) of mouse ES cells and human iPS cells were used. In 100% of cases we observed teratoma formation in a large number of animals and in long follow-up (up to 6 months). We also used these methods for bio-safety analysis of other adult MSC, such as those derived from dental pulp of deciduous teeth, umbilical cord, and adipose tissue.
  • We observed derivation of induced pluripotent stem cells from hIDPSC. The pluripotency of hIDPSC derived iPS cells were tested through teratoma formation, while human embryonic stem (ES) cells and hIDPSC were used as control. Routine protocol for teratomas production for ES cells was used (Hentze et al., 2009). Following this protocol 106 of cells of each line: hIDPSC-iPS cells, ES cells and hIDPSC were inoculated in the rear leg muscle of 4-week-old male, SCID. In animals, which were inoculated with hIDPSC-iPS cells or ES cells teratoma formation was observed after three months. However, hIDPSCs, which were used as a negative control in this study and were inoculated in 10 animals, did not produce teratomas neither after three month nor after six months of follow-up. Five of these animals were maintained alive during one year and even after this time teratomas formation was not observed.
  • Histologically, teratoma formation in pluripotent stem cells requires the development of tissues derived from the three germ layers. For adult/mesenchymal stem cells, any alterations in the integrity of normal tissue at the site of transplantation were considered. After six months, animals, which were inoculated with hIDPSCs and did not produce teratomas, were killed and histological specimens of the brain, lung, kidney, spleen, and liver were analyzed of the animals. The presence of DNA from hIDPSCs was confirmed in all aforementioned organs but no tumor formation or any morphological alterations were observed. Thus, we established the safety of cell regeneration by investigational product CELLAVITA™ (stem cells) regarding tumor formation and risk of immune rejection.
  • Additionally, as shown in the aforementioned studies using hIDPSC in animal models of spinal cord injury, cranial bone defects, total limbal stem cell deficiency (TLSCD), muscular dystrophy, and osteonecrosis of the femoral head (ONFH), no teratoma formation and/or risk of immune rejection were observed (Costa et. al., 2008; Kerkis et al., 2008; Monteiro et al., 2009; Gomes et al., 2010; Feitosa et al., 2010; Almeida et al., 2011).
  • FIG. 29 summarizes additional already published preclinical studies, which support the safety of investigational product CELLAVITA™ (stem cells).
  • Example 10 Principal Criterion for Teratoma Assay
  • We evaluated the next criterion for a teratoma assay: sensitivity and quantitativity; definitive cell number and single cell suspension production; immunophenotyping of studied cell in respect of expression on pluripotent cell markers and karyotype; co-transplantation of studied cells together with Matrigel. The cells were transplanted subcutaneously (s.c) into NOD/SCID mice, which allows for simple monitoring of teratoma development.
  • The development of tumors was monitored from 4 month (˜16 weeks). Histological criteria for teratomas is the differentiation of pluripotent cells into the cells derived from three germ layers. Such study usually was performed by pathologist.
  • For adult/mesenchymal stem cells any type or any changes on normal tissue integrity in the site of cell injection were taken in consideration.
  • Application of the Teratoma Criterion A. The Experimental System(s):
  • a. Mouse embryonic stem cells
    b. Mouse 3T3 fibroblasts, permanent mouse cell line Balbc 3T3 cell line, clone A31
    c. Human iPS-IDPSC
    d. Human ES cells
    e. Human IDPSCs
  • We used aforementioned method in diverse studies to characterize different mouse ES cell lines pluripotency established by us as well as to confirm ES cells pluripotency at high 25 or more passages and for characterization of sub-clones obtained from mouse ES cell lines (Sukoyan et al., 2002; Carta et al., 2006; Kerkis et al., 2007; Lavagnolli et al., 2007; Hayshi et al., 2010).
  • Additionally, this method was used to characterize the pluripotency of iPS cells derived from immature dental pulp stem cells (IDPSC) in more recent publication of our group (Beltrão-Braga et al., 2011). In this publication the human IDPSC were used as a control for iPS-IDPSCs. We showed that iPS-IDPSCs formed nice teratomas with tissues originated from all three germ layers, while hIDPSC were not able to produce any type of teratomas or any other type of neoplasms. In addition, iPS-IDPSCs expressed Nanog in nucleus, and hIDPSCs did not.
  • Results
  • Disclosed multiharvest explant like culture used for the isolation of a population of immature dental pulp stem cells (IDPSC), results in expression of embryonic stem cell markers Oct-4, Nanog, SSEA-3, SSEA-4, TRA-1-60 and TRA-1-81 as well as several mesenchymal stem cell markers during at least 15 passages while maintaining the normal karyotype and the rate of expansion characteristic of stem cells. The expression of these markers was maintained in subclones obtained from these cells. Moreover, in vitro these cells can be induced to undergo uniform differentiation into smooth and skeletal muscles, neurons, cartilage, and bone under chemically defined culture conditions. It is important to mentioned that IDPSC although have a small size and cytoplasm poor in cell organelles differ from naïve pluripotent cells presenting typical mesenchymal—fibroblast like morphology. Therefore IDPSC are of mesenchymal type, in contrast to ES and iPS cells, which are of epithelial type. The principle difference between MSC and ES or iPS cells that MSC are migrating and plastic anchoring, they synthetize extracellular matrix and are cell junction free cells.
  • B. The Experimental System(s):
  • a. IDPSC three different primary cultures at early (n=10) and late passages (n=10)
    b. Human primary fibroblast
  • In addition, this method was validated using dog fetal stem cells from bone marrow, liver, yolk sac, allantois and amniotic liquid which also express pluripotent markers.
  • The IDPSC are composed by population of MSC with a variable number of stem cells expressing pluripotent markers (1-25% of cells) (Lizier et al., 2012). These cells were transplanted into NOD/SCID mice (n=20) and the development of tumors was monitored from 4 month (˜16 weeks). Any type of changes on normal tissue integrity in the site of cell injection were taken in consideration. This protocol was adapted for population of IDPSC, especially in respect of cell number used, which was calculated on the basis that 20% of IDPSC express pluripotent markers. In our previous tests with ES and iPS cells we used 106 cells, while in to test IDPSC and control cells teratogenicity 5×106 cells were used. After 4 month, even if macroscopically the tumors were not observed, the mice were sacrificed and frozen cuts were obtained from diverse organs, such as brain, lung, kidney, spleen, liver and were analyzed by pathologist.
  • Although presence of DNA of IDPSC within all studied organs were found, no tumor formation or any morphological changes were observed.
  • Example 11. Cell Culture Conditions to Establish Proper Population of Stem Cells
  • Culture conditions, such as culture medium and adhesive surface, can affect gene expression of the cells. Such genes include ABCG2 and Vimentin, which are two genes that can indicate suitability of the cells in culture for therapeutic use, particularly systemic cellular systems like the present invention. Most suitable culture medium form tested is DMEM/F12 basal medium supplemented with 5-10-15% of FBS, antibiotics, and Glutamate, and the cells should be cultured without an adhesion layer (e.g. without extracellular matrix (ECM) or scaffold) such that the cells adhere directly to the culture dish or beads plastic. Cells that were grown with epithelial growth media conditions turn into epithelial-like cells. Using various xeno-free medium will require growth factors supplementation and selection of appropriate ECM coating can be useful for scale up of current cells into 3D culture conditions including bioreactors, such as Terumo hollofiber bioreactor, Eppendorf—New Brunswick bioreactors with beads, etc.
  • The link of ABCG2 expression and undifferentiation status of cells is shown on FIGS. 26 and 27 where we demonstrated that once ABCG2 is not expressed due to changed culture medium or coating layer, then cells have clearly a more fibroblast or epithelial cells-like morphology. Another surprising finding was that a typical medium routinely used in the prior art for maintenance or induction of embryonic stem cells, namely Dulbeccos Modified Eagle Medium (DMEM) serum knockout medium KOSR (KSFM), is not advantageous for generating or maintaining pluripotent cells derived from hDPSC. As demonstrated in FIG. 26, use of this medium with and without fibronectin containing scaffold/ECM matrix coating or scaffold resulted in the differentiation of hIDPSCs into fibroblast-like or epithelial like cells (FIG. 26). In contrast to the use of Dulbeccos Modified Eagle Medium (DMEM) alone, it was found that Dulbeccos Modified Eagle Medium (DMEM) or Neurobasal medium (NB) if supplemented with B27 and, optionally, supplemented with FGF and/or EGF, leads to the formation of neuronal like lineages. Exemplary protocols for differentiation into cells of the neural lineages have also been described in previous patent applications, for example, International Application no. PCT/IB14/59850 and U.S. patent application Ser. No. 14/214,016.
  • Differentiation into Corneal Cells
  • Material and Methods
  • De-Epithelialization of Amniotic Membrane as a Potential Fibronectin—Containing Scaffold for hIDPSC Cell Culture
  • Amniotic membrane (AM) was obtained from placenta of donor and stored at −8° C. (Covre J L at al 2011). Prior AM use, it was thawed at room temperature and washed in three times in PBS. Next, AM was removed from nitrocellulose membrane and washed again. In order to remove the epithelia, AM was incubated with EDTA for two hours. Then, the epithelia were removed mechanically. The AM becomes transparent following the epithelia removal. Completely transparent AM was transferred on inserts (Covre J L at al 2011 and Melo G B at al 2007).
  • IDPSC Culture
  • Human IDPSC, (2n=46, XX) were isolated from dental pulp of deciduous teeth and characterized previously (Kerkis et al. 2006). hIDPSC were maintained in Dulbecco's-modified Eagle's medium (DMEM)/Ham's F12 (1:1; Invitrogen, Carls-bad, CA), supplemented with 15% fetal bovine serum (FBS; Hyclone, Logan, Utah), 100 units/mL penicillin (Gibco, Grand Island, N.Y.), 100 μg/mL streptomycin (Gibco), 2 mM L-glutamine (Gibco), and 2 mM nonessential amino acids (Gibco). The culture medium was changed daily, and the cells were replaced every 3 days. After they reached 80% confluence, they were washed twice in sterile phosphate-buffered saline (PBS; Gibco; 0.01 M, pH 7.4), enzymatically treated with 0.25% trypsin/EDTA (Invitrogen), and seeded onto amniotic membrane previously prepared.
  • Culture Media
  • To select the best culture media for cultivate IDPSC on AM, we tested the following culture media: A) The first was supplemental hormonal epithelial medium (SHEM), a combination of Dulbecco's Modified Eagle's Medium/Ham's F-12 nutrient mixture (DMEM/F12; Invitrogen, Gibco Cell Culture, Port-land, OR; 1:1) containing 1.05 mM calcium supplemented with 5 μg/ml crystalline bovine insulin (Sigma Aldrich, St. Louis, Mo.), 30 ng/ml cholera-toxin (Calbiochem, San Diego, Calif.), 2 ng/ml epidermal growth factor (EGF, R & D Systems, Inc., Minneapolis, Minn.), 0.5% dimethyl sulfoxide (DMSO, Sigma Aldrich), 0.5 μg/ml hydrocortisone, 5 ng/ml sodium selenite, and 5 μg/ml apo-transferrin, and supplemented with 10% fetal bovine serum (FBS). All reagents were obtained from Invitrogen Corporation (Grand Island, N.Y.), except those indicated in the text. B) The second was keratinocyte serum-free medium (KSFM) containing 0.09 mM calcium supplemented with 30 mg/ml pituitary bovine extract, 0.2 ng/ml EGF, 10% FBS, and ampicillin/streptomycin. C) The third was Epilife medium (Cascade Biologics, Portland, Oreg.), containing 0.06 mM calcium supplemented with 1% “human corneal growth supplement” (Cascade Biologics), containing 0.2% pituitary bovine extract, 5 g/ml bovine insulin, 0.18 mg/ml hydrocortisone, 5 μg/ml bovine transferrin, 0.2 ng/ml EGF, added 1% penicillin G sodium (Penicillin G sodium 10,000 g/ml, streptomycin sulfate 25 mg/ml, amphotericin B in 0.85% NaCl), and 5% FBS. D) Knockout media
  • Antibodies
  • Mouse anti-human monoclonal antibodies: ABCG2 (Chemicon) and cytoplasmic/nuclear monoclonal antibodies: mouse anti-cytokeratin 3/12 (K3/12) (RDI, Flanders, N.J., USA), reacts with human and rabbit. Mouse anti-human IDPSC antibody was obtained as described (Kerkis et al., 2006) and successfully used by us in previous studies (Fonseca et al., 2009; Monteiro et al., 2009).
  • Immunofluorescence Staining
  • Cells were grown on glass cover-slips up to 70% confluence and also, were grown on AM, washed in PBS (Gibco) and fixed overnight with 4% paraformaldehyde (Sigma). Coverslips were washed three times in tris buffered saline (TBS), containing 20 mm Tris-HCl pH 7.4 (Vetec, Duque de Caxias, RJ, Brazil), 0.15 m NaCl (Dinamica Reagent, Sao Paulo, SP, Brazil), and 0.05% Tween-20 (Sigma). Permeabilization was performed using 0.1% Triton X-100 for 15 min (Santa Cruz Biotechnology). Cells were washed three times and incubated for 30 min in 5% bovine serum albumin (Sigma) in PBS pH 7.4 (Gibco). Primary antibodies were added for 1 h on each slide at different dilutions (ABCG2 and K3/12 (1:100), and anti-hIDPSC (1:1000)), which were incubated at room temperature. Following washing in TBS (three times), cells were incubated in the dark for 1 h with secondary anti-mouse antibody-conjugated fluorescein isothiocyanate (FITC) at a dilution of 1:500. Microscope slides were mounted in antifade solution (Vectashield mounting medium, Vector Laboratories, Hercules, Calif., USA) with 4′,6-diamidino-2-phenylindole (DAPI) and analysed using a confocal microscope. Control reactions were incubated with PBS instead of primary antibody, followed by washing and incubation with respective secondary antibody. All experiments have been done in triplicate.
  • Results Expression of Undifferentiated LSCs and Differentiated Corneal Cells Proteins in IDPSCs Grown in Different Culture Media on Plastic Substrate
  • The expression pattern of ABCG2 protein (ATP-binding cassette sub-family G member 2), which are commonly used for LSCs characterization and CK3 (cytokeratin 3) and cytokeratin 12 that encodes the type I intermediate filament chain and both expressed in corneal epithelia were analyzed. FIG. 26 depicts that IDPSC had differential response in respect of expression of studied proteins when cultured in distinct culture medium during 7 days. The IDPSCs grown on plastic surfaces did not express ABCG2 when cultured in SHEM, KSFM, Epilife and DMEM/KO (FIG. 26), this protein was expressed in IDPSCs only when they were cultured in basal culture medium (FIG. 22 A5). Interestingly, that IDPSCs cultured in SHEM and DMEM/KO, after seven days changed their morphology fibroblast like (FIG. 26) to epithelial like (FIG. 26) and start to express CK3/12, while IDPSC cultured in Epilife and KSFM and DMEM/F12 did not start to express K3/12 (FIG. 26).
  • Expression of Undifferentiated LSCs and Differentiated Vimentin Markers in IDPSCs Grown in Different Culture Media on AM
  • Next, the expression of these markers and additionally vimentin was verified in IDPSC grown on AM during 7 days (FIG. 27). Epilife was excluded from this study due to very low survival of the cells (less then 50%), when grown in this medium and low adherence of the cells on AM in combination with Epilife. Vimentin expression was observed in all samples (FIG. 27), it was positive in IDPSCs cultured in DMEM/F12 and SHEM (FIG. 27) and showed weak positivity with IDPSCs cultured in KSFM and DMEM/KO (FIG. 27). ABCG2 antibody showed strong immunopositivity with IDPSC cultured in SHEM and DMEM/F12 (FIG. 27) and did not express in IDPSC cultured in KSFM (FIG. 27) and showed weak immunoreactivity with IDPSC cultured in DMEM/KO (FIG. 27). IDPSCs did not react with IDPSCs cultured in DMEM/F12 e KSFM (FIG. 27) and showed very weak immunopositivity with K3/12 when cultured in SHEM and DMEM/KO (FIG. 27).
  • Example 12. Expression of CD146 and CD13 in Early Phase (EP) and Late Phase (LP) hIDPSC
  • CD146 and CD13 expression were analyzed by flow cytometry in EP-hIDPSC and LP-hIDPSC. The results in FIG. 30 show that CD13 was expressed in 52% of EP-hIDPSC and 95% of LP-hIDPSC. These results demonstrate that in vitro DP harvesting and hIDPSC passing produce increased quantities of hIDPSC expressing CD13 and lacking expression of CD146.
  • Example 13. Neuronal Progenitor Cells—Neuroblasts Derived from IDPSC
  • IDPSC are migratory neural crest stem cells which are capable of aggregating into neurospheres. It is known that in development, SOX2 is regained only by those cells fated to become neurons. Neural crest stem cells that remain SOX2-free differentiate into other cell types, but never become neurons.
  • According to some aspects, it was verified how long term mechanical transfer of DP will affect capacity of IDPSC to express SOX2 and differentiate to neurons. Neural differentiation of IDPSC was induced following protocol described herein. First, it is verified that expression of SOX1 protein correlates with mitotically active progenitors that are not yet committed to a final stage and also that β-tubulin class III is a microtubule element expressed exclusively in early neurons. This marker in combination with BrdU labeling was used as evidence of a correlation between a number of DP transfer cycles and enrichment of IDPSC population with the cells strongly committed to neural differentiation.
  • FIGS. 31A-A4 demonstrate expression of SOX1 in the nucleus, while β-tubulin shows cytoplasmic localization in the neural precursors derived from LP of IDPSC, which are organized in neurospheres. More specifically, FIG. 31A depicts expression of Sox1 and β-tubulin proteins in late population (LP) of hIDPSC derived neurons. The following are also shown in FIGS. 31A-A4: FIG. 31A. a nucleus (white arrow) of neurons stained with DAPI; FIG. 31A1. Positive immunostaining for SOX1 observed in the nucleus (white arrow) of neurons; FIG. 31A2. Positive immunostaining for β-tubulin; FIG. 31A3. Merged images FIGS. 31A-A2 and 31A4. Major magnification of inset in FIG. 31A3 demonstrates superposition of DAPI and Sox1 in the nucleus of neurons (white arrow). FIGS. 31B-E demonstrate enrichment of LP of hIDPSC with neural progenitors and neurons following 15, 30 and 45 cycles of dental pulp mechanical transfer and induction of neural differentiation. In FIG. 31B, a percentage of SOX1 negative cells, nucleus stained only with DAPI (blue) are shown; SOX1 positive cells are presented in green. In FIG. 31D, a percentage of β-tubulin negative cells, nucleus stained only with DAPI (blue); β-tubulin positive cells are presented in red.
  • The proliferative potential of early neuroblasts obtained following differentiation of IDPSC toward neurons was also confirmed. FIG. 32A demonstrates nucleus of IDPSC-neuroblasts positively immunostained with anti-BrdU antibody and neuronal bodies which react positively with β-tubulin class III. FIGS. 32B-C show clearly the enrichment of differentiated IDPSC population with slightly differentiated neuroblasts following growing cycle numbers of DP mechanical transfer.
  • Surprisingly, transit amplifying IDPSC population of slightly differentiated neuroblasts was obtained. This was confirmed by concomitant expression of BrdU, Sox1 and β-tubulin, which has never been demonstrated before for neither adult stem cells nor for embryonic.
  • Even more astonishing was the finding that neuronal progenitor cells derived from IDPSC at advanced passages (5-10) that already lack expression of SOX2 start re-expressing SOX2 during neurosphere formation (FIGS. 33A-A3). Efficiency of these cells' differentiation into neural cells, however, is lower than IDPSC at passages 1-3. Re-expression of SOX2 was more efficient in IDPSC obtained from DPs, which underwent a high number mechanical transfer cycles (>30) (FIGS. 33B,C).
  • These findings provide strong evidence about the identity between IDPSC and neural crest stem cells that can be isolated at early and late embryonic stages. Furthermore, the efficiency of neural commitment increased following DP mechanical transfers supporting our previous observation about the importance of low oxygen atmosphere for IDPSC isolation and maintaining of their self-renewal and differentiation potential (Lizier et al., 2012). Therefore, LP IDPSC has an advantage over EP IDPSC in respect of neuronal commitment, which is an important conclusion for the future of stem cell use in neurodegenerative diseases.
  • Immunophenotype of hIDPSCs derived neuroblasts SOX1, SOX2, beta-3-tubulin, and early neurons: CD271+, Beta-3-tubulin+, NeuN+, Dynein+, Lis1+, NDEL+, myelin P2+, 04+, GFAP+, Synaptophysin+ and CD146+. Surprisingly, it was discovered that LP IDPSC culture of post-natal cells of neural crest origin are characterized by unique properties. These unique properties are usually typical for neural precursors derived from fetal brain or from ES or iPS cells induced to neural differentiation in vitro.
  • REFERENCES
    • 1. Altman J and Das G D. Post-natal origin of microneurones in the rat brain. Nature 1965; 207: 953-956
    • 2. Bachoud-Levi A C, Gaura V, Brugieres P, et al. Effect of fetal neural transplants in patients with Huntington's disease 6 years after surgery: a long-term follow-up study. Lancet Neurol 2006; 5: 303-09.
    • 3. Baker S A, Baker K A and Hagg T. Dopaminergic nigrostriatal projections regulate neural precursor proliferation in the adult mouse subventricular zone. Eur J Neurosci 2004; 20: 575-579
    • 4. Brito et al., Imbalance of p75NTR/TrkB protein expression in Huntington's disease: implication for neuroprotective therapies. Cell Death and Disease (2013) 4, e595; doi:10.1038/cddis.2013.116
    • 5. Lescaudron L, Unni D, Dunbar G L. Autologous adult bone marrow stem cell transplantation in an animal model of huntington's disease: behavioral and morphological outcomes. Int J Neurosci 2003; 113:945-956.
    • 6. Vatzey E M, Chen K, Hughes S M, Connor B: Transplanted adult neural progenitor cells survive, differentiate and reduce motor function impairment in a rodent model of Huntington's disease. Exp Neurol 2006; 199:384-396.
    • 7. C. Uboldi, A. Doring, C. Alt, P. Estess, M. Siegelman, and B. Engelhardt, “L-Selectin-deficient SJL and C57BL/6 mice are not resistant to experimental autoimmune encephalomyelitis,” European Journal of Immunology, vol. 38, no. 8, pp. 2156-2167, 2008.
    • 8. B. Engelhardt, “Immune cell entry into the central nervous system: involvement of adhesion molecules and chemokines,” Journal of the Neurological Sciences, vol. 274, no. 1-2, pp. 23-26, 2008.
    • 9. S. Kim, K. A. Chang, J. Kim et al., “The preventive and therapeutic effects of intravenous human adipose-derived stem cells in Alzheimer's disease mice,” PLoS ONE, vol. 7, no. 9, Article ID e45757, 2012.
    • 10. D. Jeon, K. Chu, S. Lee et al., “A cell-free extract from human adipose stemcells protects mice against epilepsy,” Epilepsia, vol. 52, no. 9, pp. 1617-1626, 2011.
    • 11. Mullen R J1, Buck C R, Smith A M. NeuN, a neuronal specific nuclear protein in vertebrates. Development. 1992 September; 116(1):201-11.
    • 12. Shuo Liu, Dritan Agalliu, Chuanhui Yu and Mark Fisher. The Role of Pericytes in Blood-Brain Barrier Function and Stroke. Current Pharmaceutical Design, 2012, 18, 3653-3662.
    • 13. De Miguel M P, Fuentes-Julián S, Blázquez-Martinez A, Pascual C Y, Aller M A, Arias J, Arnalich-Montiel F. Immunosuppressive properties of mesenchymal stem cells: advances and applications. Curr Mol Med. 2012 June; 12(5):574-91.
    • 14. Le Blanc K, Ringden O. Mesenchymal stem cells: properties and role in clinical bone marrow transplantation. Curr Opin Immunol. 2006 October; 18(5):586-91. Epub 2006 Aug. 1.
    • 15. Karen English Mechanisms of mesenchymal stromal cell immunomodulation. Immunol Cell Biol 91: 19-26; advance online publication, Oct. 23, 2012; doi:10.1038/icb.2012.56
    • 16. Matthew D Griffin, Aideen E Ryan, Senthilkumar Alagesan, Paul Lohan, Oliver Treacy and Thomas Ritter Anti-donor immune responses elicited by allogeneic mesenchymal stem cells: what have we learned so far? Immunol Cell Biol 91: 40-51; advance online publication, Dec. 4, 2012; doi:10.1038/icb.2012.67
    • 17. Svenningsson P, Nishi A, Fisone G, Girault J A, Nairn A C, Greengard P. DARPP-32: an integrator of neurotransmission. Annu Rev Pharmacol Toxicol. 2004; 44:269-96.
    • 18. Crook Z R, Housman D E. Dysregulation of dopamine receptor D2 as a sensitive measure for Huntington disease pathology in model mice. Proc Natl Acad Sci USA. 2012 May 8; 109(19):7487-92. doi: 10.1073/pnas.1204542109. Epub 2012 Apr. 23.
    • 19. Chen J Y, Wang E A, Cepeda C, Levine M S. Dopamine imbalance in Huntington's disease: a mechanism for the lack of behavioral flexibility. Front Neurosci. 2013 Jul. 4; 7:114. doi: 10.3389/fnins.2013.00114. eCollection 2013.
    • 20. Parent M, Bedard C, Pourcher E. Am J Neurodegener Dis. 2013 Sep. 18; 2(3):221-7.
    • 21. Hoglinger G U, Rizk P, Muriel M P, Duyckaerts C, Oertel W H, Caille I and Hirsch E C. Nat Neurosci 2004; 7: 726-735).
    • 22. Wernig M, et al. Proc Natl Acad Sci USA. 2008; 105:5856-5861; Caiazzo M, et al. Nature. 2011; 476:224-227;
    • 23. Lindvall, O. & Kokaia, Z., J Clin Invest., 2010 120, 29.
    • 24. Calissano P, Matrone C, Amadoro G: Nerve growth factor as a paradigm of neurotrophins related to Alzheimer's disease. Dev Neurobiol 2010, 70:372-383.
    • 25. M, Togari A, Kondo T, Mizuno Y, Komure O, Kuno S, Ichinose H, Nagatsu T: Brain-derived growth factor and nerve growth factor concentrations are decreased in the substantia nigra in Parkinson's disease. Neurosci Lett 1999, 270:45-48;
    • 26. Gauthier L R, Charrin B C, Borrell-Pages M, Dompierre J P, Rangone H, Cordelieres F P, De Mey J, MacDonald M E, Lessmann V, Humbert S, Saudou F: Huntingtin controls neurotrophic support and survival of neurons by enhancing BDNF vesicular transport along microtubules. Cell 2004, 118:127-138;
    • 27. Strand A D, Baguet Z C, Aragaki A K, Holmans P, Yang L, Cleren C, Beal M F, Jones L, Kooperberg C, Olson J M, Jones K R: Expression profiling of Huntington's disease models suggests that brain-derived neurotrophic factor depletion plays a major role in striatal degeneration. J Neurosci 2007, 27:11758-11768;
    • 28. Wictorin K, Bjorklund A, Williams L R, Varon S, Gage F H: Amelioration of cholinergic neuron atrophy and spatial memory impairment in aged rats by nerve growth factor Nature 1987, 329:65-68
    • 29. Sukoyan M A, Kerkis A Y, Mello M R, Kerkis I E, Visintin J A, Pereira L V.
  • Establishment of new murine embryonic stem cell lines for the generation of mouse models of human genetic diseases. Braz J Med Biol Res. 2002 May; 35(5):535-42.
    • 30. Carta L, Pereira L, Arteaga-Solis E, Lee-Arteaga S Y, Lenart B, Starcher B, Merkel C A, Sukoyan M, Kerkis A, Hazeki N, Keene D R, Sakai L Y, Ramirez F. Fibrillins 1 and 2 perform partially overlapping functions during aortic development. J Biol Chem. 2006 Mar. 24; 281(12):8016-23
    • 31. Kerkis A, Fonseca S A, Serafim R C, Lavagnolli T M, Abdelmassih S, Abdelmassih R, Kerkis I. In vitro differentiation of male mouse embryonic stem cells into both presumptive sperm cells and oocytes. Cloning Stem Cells. 2007 Winter; 9(4):535-48.
    • 32. Lavagnolli T M, Fonseca S A, Serafim R C, Pereira V S, Santos E J, Abdelmassih S, Kerkis A, Kerkis I. Presumptive germ cells derived from mouse pluripotent somatic cell hybrids. Differentiation. 2009 September-October; 78(2-3):124-30
    • 33. Hayashi M A, Guerreiro J R, Cassola A C, Lizier N F, Kerkis A, Camargo A C, Kerkis I. Long-term culture of mouse embryonic stem cell-derived adherent neurospheres and functional neurons. Tissue Eng Part C Methods. 2010 December; 16(6):1493-502.
    • 34. Lima B L, Santos E J, Fernandes G R, Merkel C, Mello M R, Gomes J P, Soukoyan M, Kerkis A, Massironi S M, Visintin J A, Pereira L V A new mouse model for marfan syndrome presents phenotypic variability associated with the genetic background and overall levels of Fbn1 expression. PLoS One. 2010 Nov. 30; 5(11):e14136.
    • 35. Beltrão-Braga P C, Pignatari G C, Maiorka P C, Oliveira N A, Lizier N F, Wenceslau C V, Miglino M A, Muotri A R, Kerkis I. Feeder-free derivation of induced pluripotent stem cells from human immature dental pulp stem cells. Cell Transplant. 2011; 20(11-12):1707-19.
    • 36. Bunting K D ABC transporters as phenotypic markers and functional regulators of stem cells. Stem Cells. 2002; 20(1):11-20.
    • 37. Islam M O et al. Characterization of ABC transporter ABCB1 expressed in human neural stem/progenitor cells. FEBS letters. 2005 579(17): 3473-3480.
    • 38. Mead B, Logan A, Berry M, Leadbeater W, Scheven B A. Intravitreally transplanted dental pulp stem cells promote neuroprotection and axon regeneration of retinal ganglion cells after optic nerve injury. Invest Ophthalmol Vis Sci. 2013; 54:7544-7556.
    • 39. Mead B, Logan A, Berry M, Leadbeater W, Scheven B A. Paracrine-mediated neuroprotection and neuritogenesis of axotomised retinal ganglion cells by human dental pulp stem cells: comparison with human bone marrow and adipose-derived mesenchymal stem cells. PLoS One. 2014 Oct. 7; 9(10).
    • 40. Martens W, Sanen K, Georgiou M, Struys T, Bronckaers A, et al. Human dental pulp stem cells can differentiate into Schwann cells and promote and guide neurite outgrowth in an aligned tissue-engineered collagen construct in vitro. (2013) The FASEB Journal.
    • 41. Sakai K, Yamamoto A, Matsubara K, Nakamura S, Naruse M, Yamagata M, Sakamoto K, Tauchi R, Wakao N, Imagama S, Hibi H, Kadomatsu K, Ishiguro N, Ueda M. Human dental pulp-derived stem cells promote locomotor recovery after complete transection of the rat spinal cord by multiple neuro-regenerative mechanisms. J Clin Invest. 2012; 122: 80-90.
    • 42. Rampon C, Weiss N, Deboux C, Chaverot N, Miller F, Buchet D, Tricoire-Leignel H, Cazaubon S, Baron-Van Evercooren A, Couraud P O. Molecular mechanism of systemic delivery of neural precursor cells to the brain: assembly of brain endothelial apical cups and control of transmigration by CD44. Stem Cells. 2008 July; 26(7):1673-82.
    • 43. Schu S, Nosov M, O'Flynn L, Shaw G, Treacy O, Barry F, Murphy M, O'Brien T, Ritter T. Immunogenicity of allogeneic mesenchymal stem cells. J Cell Mol Med. 2012; 16:2094-2103.
    • 44. Weinger J G, Weist B M, Plaisted W C, Klaus S M, Walsh C M, Lane T E. MHC mismatch results in neural progenitor cell rejection following spinal cord transplantation in a model of viral-induced demyelination. Stem Cells. 2012 November; 30(11):2584-950
    • 45. Bifari F, Pacelli L, Krampera M. Immunological properties of embryonic and adult stem cells. World J Stem Cells. 2010; 2:50-60.
    • 46. Bertram L, McQueen M B, Mullin K, Blacker D, Tanzi R E. (2007) “Systematic meta-analyses of Alzheimer disease genetic association studies: the AlzGene database.” Nat Genet 39(1): 17-23.
    • 47. Jared B. Vasquez,a David W. Fardo,b and Steven Estusa,* ABCA7 expression is associated with Alzheimer's disease polymorphism and disease status. Neurosci Lett. Nov. 27, 2014
    • 48. Yamagata et al. Human Dental Pulp-Derived Stem Cells Protect Against Hypoxic-Ischemic Brain Injury in Neonatal Mice. Stroke. 2013; 44:551-554.
    • 49. Albensi, B. C., Sullivan, P. G., Thompson, M. B., Scheff, S. W. & Mattson, M. P. Cyclosporine ameliorates traumatic brain injury-induced alterations of hippocampal synaptic plasticity. Exp. Neurol. 162, 385-389 (2000).
    • 50. Matsumoto, S., Friberg, H., Ferrand-Drake, M. & Wieloch, T. Blockade of the mitochondrial permeability transition pore diminishes infarct size in the rat after transient middle cerebral artery occlusion. J. Cereb. Blood Flow Metab. 19, 736-741 (1999).
    • 51. Leventhal L, Jeffrey H. Kordower Cyclosporine A Protects Striatal Neurons from Mitochondrial Dysfunction. Immunosuppressant Analogs in Neuroprotection. 2003, pp 159-174.
    • 52. Lee, R. H., Pulin, A. A., Seo, M. J., Kota, D. J., Ylostalo, J., Larson, B. L., Semprun-Prieto, L., Delafontaine, P., Prockop, D. J., 2009. Intravenous hMSCs improve myocardial infarction in mice because cells embolized in lung are activated to secrete the anti-inflammatory protein TSG-6. Cell Stem Cell 5 (1), 54-63.
    • 53. GRIGORIADIS N, LOURBOPOULOS A, LAGOUDAKI R, FRISCHER J M, POLYZOIDOU E, TOULOUMI O, SIMEONIDOU C, DERETZI G, KOUNTOURAS J, SPANDOU E, KOTTA K, KARKAVELAS G, TASCOS N, LASSMANN H (2011) Variable behavior and complications of autologous bone marrow mesenchymal stem cells transplanted in experimental autoimmune encephalomyelitis. Experimental neurology 230: 78-89.
    • 54. E. Y. Snyder. The risk of putting something where it does not belong: Mesenchymal stem cells produce masses in the brain Experimental Neurology 230 (2011) 75-77.
    • 55. Dagur P K, McCoy J P Jr. Endothelial-binding, proinflammatory T cells identified by MCAM (CD146) expression: Characterization and role in human autoimmune diseases. Autoimmun Rev. 2015 May; 14(5):415-22.

Claims (20)

We claim:
1. A cryopreserved pharmaceutical composition comprising immature dental pulp stem cells (IDPSCs), wherein the IDPSCs express SOX-1 and SOX-2.
2. The pharmaceutical composition of claim 1, wherein the IDPSCs further express beta-3-tubulin.
3. The pharmaceutical composition of claim 2, wherein the IDPSCs further express at least one marker selected from Oct3/4 and Nanog.
4. The pharmaceutical composition of claim 1, wherein the IDPSCs are negative for HLA-ABC and HLA-DR major histocompatibility (MHC) antigens.
5. The pharmaceutical composition of claim 1, wherein the IDPSCs are human IDPSCs.
6. The pharmaceutical composition of claim 1, wherein the pharmaceutical composition is formulated for intravenous (IV) injection.
7. The pharmaceutical composition of claim 1, wherein the IDPSCs are cryopreserved with dimethyl sulfoxide (DMSO).
8. A method of treating a neurological disease or condition comprising systemically administering to a subject in need thereof a cryopreserved pharmaceutical composition comprising immature dental pulp stem cells (IDPSCs), wherein the IDPSCs express SOX-1 and SOX-2.
9. The method of claim 8, wherein the wherein the IDPSCs further express beta-3-tubulin.
10. The method of claim 9, wherein the IDPSCs further express at least one marker selected from Oct3/4 and Nanog.
11. The method of claim 8, wherein the IDPSCs are negative for HLA-ABC and HLA-DR major histocompatibility (MHC) antigens.
12. The method of claim 8, wherein the pharmaceutical composition is intravenously administered to the subject.
13. The method of claim 8, wherein the IDPSCs are autologous, allogeneic or both to the subject.
14. The method of claim 8, wherein the neurological disease or condition is selected from the group consisting of Parkinson's disease (PD), multiple sclerosis, amyotrophic lateral sclerosis (ALS), stroke, autoimmune encephalomyelitis, diabetic neuropathy, glaucomatous neuropathy, Alzheimer's disease, and Huntington's disease (HD).
15. The method of claim 14, wherein the neurological disease or condition is Huntington's disease (HD).
16. The method of claim 15, wherein the subject is diagnosed with early HD and administering the hIDPSCs to the subject supports a neuroprotective mechanism in the subject.
17. The method of claim 14, wherein the neurological disease or condition is Parkinson's disease (PD).
18. The method of claim 17, wherein the subject is diagnosed with PD and administering the hIDPSCs to the subject repairs lost dopaminergic neurons in the subject.
19. The method of claim 8, wherein the neurological disease or condition is selected from the group consisting of autism, schizophrenia, stroke, ischemia, a motor disorder, and a convulsive disorder.
20. The method of claim 8, wherein the neurological disease or condition is selected from the group consisting of neuroblastoma and retinoblastoma.
US17/562,797 2013-03-15 2021-12-27 Compositions comprising stem cells expressing mesenchymal and neuronal markers and uses thereof to treat neurological disease Abandoned US20220193141A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/562,797 US20220193141A1 (en) 2013-03-15 2021-12-27 Compositions comprising stem cells expressing mesenchymal and neuronal markers and uses thereof to treat neurological disease

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US201361791594P 2013-03-15 2013-03-15
US201361800245P 2013-03-15 2013-03-15
US14/214,016 US9790468B2 (en) 2013-03-15 2014-03-14 Multifunctional immature dental pulp stem cells and therapeutic applications
US201562130593P 2015-03-09 2015-03-09
US201562130585P 2015-03-09 2015-03-09
US201562220792P 2015-09-18 2015-09-18
US15/065,259 US11207352B2 (en) 2013-03-15 2016-03-09 Compositions comprising stem cells expressing mesenchymal and neuronal markers and uses thereof to treat neurological disease
US17/562,797 US20220193141A1 (en) 2013-03-15 2021-12-27 Compositions comprising stem cells expressing mesenchymal and neuronal markers and uses thereof to treat neurological disease

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/065,259 Continuation US11207352B2 (en) 2013-03-15 2016-03-09 Compositions comprising stem cells expressing mesenchymal and neuronal markers and uses thereof to treat neurological disease

Publications (1)

Publication Number Publication Date
US20220193141A1 true US20220193141A1 (en) 2022-06-23

Family

ID=56162995

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/065,259 Active 2034-12-09 US11207352B2 (en) 2013-03-15 2016-03-09 Compositions comprising stem cells expressing mesenchymal and neuronal markers and uses thereof to treat neurological disease
US17/562,797 Abandoned US20220193141A1 (en) 2013-03-15 2021-12-27 Compositions comprising stem cells expressing mesenchymal and neuronal markers and uses thereof to treat neurological disease

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US15/065,259 Active 2034-12-09 US11207352B2 (en) 2013-03-15 2016-03-09 Compositions comprising stem cells expressing mesenchymal and neuronal markers and uses thereof to treat neurological disease

Country Status (1)

Country Link
US (2) US11207352B2 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10493105B2 (en) * 2014-09-11 2019-12-03 Taiwan Mitochondrion Applied Technology Co., Ltd Isolated adipose-derived mesenchymal stem cells treated with angelica extract or butylidenephthalide, and wherein the cells have an increased mitochondrial membrane potential and a decreased level of IL-8, and methods for treating parkinson's disease

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120201786A1 (en) * 2010-07-15 2012-08-09 Nikolai Tankovich Methods for the use of stem cells and stem cell factors in the treatment of skin conditions
US20130029292A1 (en) * 2011-07-27 2013-01-31 Kwei Mar Method of implanting mesenchymal stem cells for natural tooth regeneration in surgically prepared extraction socket and compositions thereof

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7597885B2 (en) 2004-03-26 2009-10-06 Aderans Research Institute, Inc. Tissue engineered biomimetic hair follicle graft
ATE431846T1 (en) 2005-03-23 2009-06-15 Stiftung Caesar NEURAL STEM CELLS
US20070128174A1 (en) 2005-09-21 2007-06-07 Kleinsek Donald A Methods and compositions for organ and tissue functionality
US20110059050A1 (en) 2006-01-27 2011-03-10 Kyle Cetrulo Methods and compositions relating to stem cell transplantation
WO2008080200A1 (en) 2006-12-29 2008-07-10 Irina Kerkis Process for obtaining stem cells
WO2009072527A1 (en) 2007-12-05 2009-06-11 National University Corporation Nagoya University Composition for autotransplantation or allotransplantation using dental pulp stem cell, and use of the composition
US20120219531A1 (en) 2008-03-17 2012-08-30 Agency For Science, Technology And Research Microcarriers for Stem Cell Culture
ITRM20080342A1 (en) 2008-06-26 2009-12-27 Univ Degli Studi Udine DOLLS-SIMILAR PULP CELLS, INSULATION AND USE METHODS.
US7847353B2 (en) 2008-12-05 2010-12-07 Bae Systems Information And Electronic Systems Integration Inc. Multi-thickness semiconductor with fully depleted devices and photonic integration
US20100297094A1 (en) 2009-01-30 2010-11-25 Harlan John M Therapy or prevention of diseases with cells or cell supernatant
US20120009601A1 (en) 2010-07-08 2012-01-12 The General Hospital Corporation Methods and compositions for reprogramming cells
US9938496B2 (en) 2010-08-13 2018-04-10 Kyoto University Method of inducing differentiation from pluripotent stem cells to germ cells
US9790468B2 (en) 2013-03-15 2017-10-17 Avita Iinternational Ltd. Multifunctional immature dental pulp stem cells and therapeutic applications
BR102013021202B1 (en) 2013-08-20 2022-09-06 Ccb - Centro De Criogenia Brasil Ltda PROCESS OF PRODUCTION OF MULTIPOTENTS AND PROGENITOR STEM CELLS

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120201786A1 (en) * 2010-07-15 2012-08-09 Nikolai Tankovich Methods for the use of stem cells and stem cell factors in the treatment of skin conditions
US20130029292A1 (en) * 2011-07-27 2013-01-31 Kwei Mar Method of implanting mesenchymal stem cells for natural tooth regeneration in surgically prepared extraction socket and compositions thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Atari et al. "Dental pulp of the third molar: a new source of pluipotent-like stem cells" 2012, J of Cell Science, vol. 14: 3343-3356. (Year: 2012) *
Kerkis et al. "Isolation and Charcterization of a Population of Immature Dental Pulp Stem Cells Expressing OCT-4 and Other Embryonic Stem Cell Markers" Feb. 2006: Cells Tissues Organs, vol. 184: 105-116 (Year: 2006) *

Also Published As

Publication number Publication date
US20160184366A1 (en) 2016-06-30
US11207352B2 (en) 2021-12-28

Similar Documents

Publication Publication Date Title
EP3426267B1 (en) Uses of dental pulp stem cells expressing mesenchymal and neuronal markers and compositions thereof to treat neurological disease
JP2022078245A (en) Method for differentiating pluripotent cell
KR102108245B1 (en) Mesenchymal stromal cells and uses related thereto
Zhang et al. Intrastriatal transplantation of stem cells from human exfoliated deciduous teeth reduces motor defects in Parkinsonian rats
JP6539385B2 (en) Use of stem cells to prevent axonal retraction of neurons
WO2008080200A1 (en) Process for obtaining stem cells
US20220193141A1 (en) Compositions comprising stem cells expressing mesenchymal and neuronal markers and uses thereof to treat neurological disease
US20220211765A1 (en) Use of stem cells expressing mesenchymal and neuronal markers and compositions thereof to treat neurological disease
US11306290B2 (en) Stem cells expressing mesenchymal and neuronal markers, compositions thereof, and methods of preparation thereof
KR101895648B1 (en) Pharmaceutical composition for treatment of neurological diseases comprsing dental pulp stem cell and method for preparing the same
US20230220336A1 (en) Fibroblasts as a regenerative cellular source for the treatment of blindness
Tsupykov et al. Protective effects of adipose-derived multipotent mesenchymal stromal cells of mice on periventricular leukomalacia model in vitro
Juntunen Development of Adipose-Derived Stromal/stem Cell–Based Therapies: Effects of donor weight and culture conditions on cell characteristics and testing the efficacy in stroke models
Şenkal Derivation of functional Mesenchymal stem cells (MSCs) from embryonic stem (ES) cell derived neuromesodermal progenitors (NMPs)
Weiss VERMONT STEM CELL CONFERENCE
Alviano et al. Cell Populations Isolated from Amnion, Chorion, and Wharton’s Jelly of Human Placenta
Dandapat et al. Mouse model to understand the role of Dux4 in FSHD

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION