[go: up one dir, main page]

WO2025097051A1 - Polymères thermoréversibles à stabilité améliorée et procédés et utilisations associés - Google Patents

Polymères thermoréversibles à stabilité améliorée et procédés et utilisations associés Download PDF

Info

Publication number
WO2025097051A1
WO2025097051A1 PCT/US2024/054259 US2024054259W WO2025097051A1 WO 2025097051 A1 WO2025097051 A1 WO 2025097051A1 US 2024054259 W US2024054259 W US 2024054259W WO 2025097051 A1 WO2025097051 A1 WO 2025097051A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
cell
days
hydrogel
inhibitor
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.)
Pending
Application number
PCT/US2024/054259
Other languages
English (en)
Inventor
Rocio G. SAMPAYO
Hunter J. Johnson
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.)
Axent Biosciences Inc
Original Assignee
Axent Biosciences Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Axent Biosciences Inc filed Critical Axent Biosciences Inc
Priority to AU2024343386A priority Critical patent/AU2024343386B2/en
Publication of WO2025097051A1 publication Critical patent/WO2025097051A1/fr
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/48Reproductive organs
    • A61K35/54Ovaries; Ova; Ovules; Embryos; Foetal cells; Germ cells
    • A61K35/545Embryonic stem cells; Pluripotent stem cells; Induced pluripotent stem cells; Uncharacterised 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/119Other fibroblast growth factors, e.g. FGF-4, FGF-8, FGF-10
    • 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
    • C12N2501/10Growth factors
    • C12N2501/13Nerve growth factor [NGF]; Brain-derived neurotrophic factor [BDNF]; Cilliary neurotrophic factor [CNTF]; Glial-derived neurotrophic factor [GDNF]; Neurotrophins [NT]; Neuregulins
    • 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
    • C12N2501/10Growth factors
    • C12N2501/15Transforming growth factor beta (TGF-β)
    • 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
    • C12N2501/70Enzymes
    • 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
    • C12N2501/70Enzymes
    • C12N2501/72Transferases [EC 2.]
    • C12N2501/727Kinases (EC 2.7.)
    • 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
    • C12N2506/02Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from embryonic 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
    • C12N2513/003D culture

Definitions

  • hPSCs Human pluripotent stem cells
  • hPSCs have the potential to differentiate into any cell type in the body and therefore have broad applications for regenerative medicine.
  • expansion and differentiation of hPSCs at large scale remains to be a major challenge in the field, preventing these transformative therapies from reaching the large patient population in need.
  • thermoreversible hydrogel that allows for the growth and differentiation of hPSCs into sensitive cell types at large scale.
  • the improved hydrogel formulation allows for a lower viscosity gel that enables encapsulation of sensitive cell types utilizing a diverse range of liquid handling systems, thanks to these unique properties: formulation and molecular weight (MW) of polymer renders a low viscosity hydrogel ideal for hPSC culture and differentiation into all 3 germ layers. This minimizes shear during a process to encapsulate cells for large scale bioreactor systems. This formulation and MW also allows for faster flow rates during encapsulation and higher control of bead size/geometry. MW of polymer and PEG length is optimal to render stable hydrogels that maintain structure through lengthy differentiation processes and render stiffness range that supports expansion and differentiation of hPSCs into cells in all 3 germ layers.
  • thermoreversible hydrogel we were able to expand hPSCs at large scale and differentiate into several cell fates including midbrain dopaminergic neurons, cortical interneurons, pancreatic endoderm progenitor cells and hematopoietic stem cells.
  • cINs post-mitotic inhibitory GABAergic cortical interneurons
  • human stem cells e.g., human pluripotent or multipotent cells
  • the pluripotent cell is a human pluripotent stem cell (hPSC) such as a human embryonic stem cell or a human induced pluripotent stem cell (iPSC).
  • hPSC human pluripotent stem cell
  • iPSC human induced pluripotent stem cell
  • the method is xeno-free and the hydrogel does not comprise extra cellular matrix (ECM) proteins.
  • ECM extra cellular matrix
  • the three-dimensional hydrogel for use in the methods is a thermoresponsive (e.g., thermoreversible) hydrogel.
  • the three- dimensional hydrogel is a polyethylene glycol-Poly(N-isopropylacrylamide) (PEG-PNIPAAM) based hydrogel.
  • the thermoresponsive hydrogel has a lower critical solution temperature (LCST) below about 30° C, below about 29° C, below about 28° C, below about 27° C, or below about 26° C.
  • thermoresponsive hydrogel has one or more of the following properties: (a) an LCST of about 12° C to 32° C, preferably between about 20° C and 24° C, more preferably of about 22° C; (b) a stiffness of about 100 to 8000 Pascal (Pa), preferably of about 800 to about 1000 Pa; (c) liquid phase viscosity of about 200 to about 4000 cP, preferably of about 200-1000 cP
  • thermoreversible hydrogels useful in the methods described herein.
  • compositions comprising a population of cells produced by the methods described herein, wherein the population of cells is enriched for post-mitotic inhibitory GABAergic cortical interneurons.
  • the composition is ECM-free and xeno- free.
  • the neurological disorder is epilepsy.
  • FIG. 1 is a schematic illustrating two-step synthesis of a thermoreversible poly(NIPAAm-co-Bam)-b-PEG graft copolymer of Formula (III). Mole ratios denoted by lowercase between intermediate reactions. AIBN refers to Azobisisobutyronitrile. Butyl amine is depicted; however, other lower alkyl amines can be substituted in the reaction.
  • FIG. 2 illustrates a comparison of properties of acrylate-based thermoreversible polymers vs. acrylamide-based thermoreversible polymers.
  • LCST lower critical solution temperature.
  • FIG. 3 illustrates a comparison of properties of low MW acrylate-based thermoreversible polymers vs. high MW acrylate-based thermoreversible polymers.
  • LCST lower critical solution temperature.
  • FIG. 4 illustrates a comparison of the effects of PEG molecular weight on properties of acrylate-based thermoreversible polymers.
  • FIGS. 5A-D illustrate a comparison of the effects of PEG:PNIPAAM polymer wt/wt% on properties of acrylate-based thermoreversible polymers.
  • Figs. 5C and 5D show hPSC viability throughout scalable encapsulation processes utilizing different hydrogel formulations.
  • Figure 5C Varying PEG:PNIPAAm ratios allowed to increase flow rates to meet the minimum required for scale-up of 2mL/min.
  • the lower shear of the 1:3 formulation maximizes cell viability upon encapsulation (second plot, measured 24hr after encapsulation). Maximal yields are also obtained with the 1:3 formulation.
  • Figure 5D 3D hydrogel core/shell design, consisting of a hydrogel+cell core and an outer layer of a-cellular shell. Representative image of encapsulated hPSC aggregates in 3D hydrogel of formula III is shown on the right.
  • FIG. 6 illustrates the effect of isobutyl vs n-butyl at position R 2 of formula III on properties of acrylate-based thermoreversible polymers.
  • FIGS. 6.5A-C illustrates the effect of -H (hydrogel) vs -CH3 (methyl) at position R 4 of formula III on properties of acrylate-based thermoreversible polymers.
  • 6.5 A illustrates the effect on material properties.
  • 6.5C illustrates the effect on hydrogel encapsulation.
  • FIG. 7 illustrates pendant functionalization of the PEG group (R 1 of formula III) with inert structure (methoxy and hydroxyl) and functional structures (acrylate, biotin and DBCO).
  • FIG. 8 illustrates backbone functionalization (R 3 of formula III) of the polymer with methacrylate, maleimide and DBCO.
  • FIG. 9 illustrates protein (FGF and Heparin) presentation and release from the polymer.
  • FIGS. 10A-B illustrate the effect of various groups (-NH2, -OCH3, and -AC) at the pendant PEG position R 1 of formula III and different solvents on polymer synthesis reaction parameters.
  • FIGS. 11 A-E illustrates a comparison of the generation of MGE progenitor cells from human pluripotent stem cells (hPSCs) using various neural induction protocols, performed in 2- dimensional cell culture and in 3-dimensional thermoreversible hydrogel, as assessed by expression of FOXG1 and DLX1 (qPCR, Figures 11B and 11C) (fold-change relative to undifferentiated hPSCs) and FOXG1 and NKX2-1 (FC, Flow Cytometry , Figures 1 ID and 1 IE).
  • hPSCs human pluripotent stem cells
  • FIG. 12A-F illustrates a comparison of the generation of cortical interneurons from human pluripotent stem cells (hPSCs) using various neural induction protocols, performed in 2- dimensional cell culture and in 3 -dimensional thermoreversible hydrogel, as assessed by expression of FOXG1, CALB1 and GAD1 (qPCR, Figures 12B-D) (fold-change relative to undifferentiated hPSCs) and FOXG1 and NKX2-1 (FC, Flow Cytometry, Figures 12E-F).
  • hPSCs human pluripotent stem cells
  • FIG. 13 Thermoreversible hydrogel for the generation of hPSC-derived cortical interneurons in three-dimensional environments a) Fully defined, synthetic PNIPAM-based hydrogel (is liquid at cold temperatures and quickly gels when heated at 37°C. b,c). By controlling the temperature of the system, hPSCs are mixed with the hydrogel at low temperature and extruded into warm culture media leading to the encapsulation of these cells in porous gel beads. These can be used in well plate assays (b) or easily scalable to a perfused bioreactor system (c).
  • cells can be differentiated within these beads into any cell type of interest, d) Differentiation paradigm and factors used to generate hPSC-derived cINs in 3D hydrogels, e) Evolution of marker expression throughout the differentiation process of hPSCs into cINs, and assays used to evaluate differentiation efficacy.
  • FIG. 14 Expression of lineage-specific markers of cortical interneuron differentiation
  • FIG. 15 Gene expression analysis of hPSC differentiation into cortical interneurons, a) Transcript expression analysis by qPCR in cells harvested on day 18 and day 35. Fold change (expressed as relative to gene expression in hPSCs.
  • FIG. 16 Viability and differentiation efficacy of hPSCs into cortical interneurons encapsulated in thermoreversible hydrogel beads compared to standard two-dimensional processes, a) Post-harvest viability of hPSC-derived cells after 35 days of differentiation in standard 2D cultures or 3D cultures in the novel hydrogel of formula (III) as herein described, b) Quantification of a), c) Marker expression analyzed by flow cytometry in hPSC-derived cells at different harvest timepoints shows higher differentiation efficacy in the novel hydrogel of formula (III) compared to standard 2D methods (%FoxGl+ cells, fold-change, at days 10, 18 and 35 in 2D vs 3D cultures and %Nkx2-l+ cells, fold change, at day 18). Data is represented as mean + s.d. of 2 biological replicates.
  • FIGs. 17A-C illustrates hESC and hiPSC expansion in 3D hydrogels comprising thermoreversible polymers of formula III.
  • Figure 17A Three culture scales compatible with the 3D hydrogels.
  • Positive-displacement-pipette (PDP) droplets consist of core 3D hydrogel only and allow for rapid screening of multiple culture conditions at small scale.
  • Core/shell beads (C/S beads) - static consist of encapsulated cells in 3D hydrogel beads made of a hydrogel+cell core and an outer layer of a-cellular shell and cultured in static conditions in well plates.
  • C/S beads - Spinner are encapsulated cells in 3D hydrogel core/shell beads that are grown in a spinning environment utilizing spinner flasks.
  • Figure 17B An example human embryonic stem cell line grown in 3D hydrogel for 8 continuous days achieving fold changes of 80 fold across all 3 culture scales and yields of up to 20e 6 cells/mL 3D hydrogel with high viability upon harvest.
  • Figure 17C An example of human induced pluripotent stem cell line grown in 3D hydrogel for 8 continuous days achieving fold changes of over 100-fold when grown in encapsulated C/S beads and yields of up to 25e 6 cells/mL 3D hydrogel with high viability upon harvest.
  • FIGS. 18A-C Reproducible hPSC expansion in bioreactors at different scales utilizing 3D hydrogels comprising polymers of formula III.
  • Fig. 18A Schematic of scalable bioreactor system containing 3D hydrogel beads
  • Fig. 18B Pluripotency marker expression determined by flow cytometry for each lot.
  • FIG. 18C Gene expression analysis performed by qPCR, showing strong correlation in gene expression across all 4 lots of hPSC expansion in 3D hydrogel. Pearson's correlation indexes are shown for each plot.
  • FIG. 19 3D hydrogels comprising polymers of formula III enable efficient hPSC differentiation into pancreatic endoderm cells (PE).
  • PE pancreatic endoderm cells
  • 100-mL Spinner flask contain hPSCs encapsulated in core-shell 3D hydrogel beads. Cells were expanded and differentiated in 3D hydrogel platform. Performance is compared to standard PE differentiation in suspension cultures.
  • FIGS. 20A-D 3D hydrogels comprising polymers of formula HI enable efficient hPSC differentiation into pancreatic endoderm cells (PE).
  • Fig. 20A Stages of hPSC differentiation into Pancreatic Endoderm (PE) cells.
  • Fig. 20B Differentiation efficacy was evaluated by expression of the PE marker PDX-1 (pancreatic and duodenal homebox- 1).
  • Fig. 20C Utilizing 3D hydrogel technology allows for a 45-higher production of PE cells compared to standard suspension methods.
  • Fig. 20D 3D hydrogel technology utilizing improved gel formulation allows for higher control over PE aggregate size compared to standard suspension methods, minimizing the undesired effects of nutrient limitation (such as cell death - necrotic cores - and suboptimal differentiation).
  • FIGS. 23A-C 3D hydrogels comprising polymers of formula III enable efficient HSC expansion.
  • Fig. 23 A Thaw and expansion of human cord blood-derived CD34+ HSCs in 3D hydrogel.
  • Fig. 23B Representative images of HSC expanded in hydrogel in SFEM II media (StemDiff Hematopoietic kit) supplemented with StemSpan CD34+ expansion supplement.
  • Fig. 23C Harvest analytics for HSCs expanded in improved hydrogel system for 8 days.
  • Activators refer to compounds that increase, induce, stimulate, activate, facilitate, or enhance activation the signaling function of the molecule or pathway, e.g., Wnt signaling, SHH signaling, etc.
  • a population of cells refers to a group of at least two cells.
  • a cell population can include at least about 10, at least about 100, at least about 200, at least about 300, at least about 400, at least about 500, at least about 600, at least about 700, at least about 800, at least about 900, at least about 1000 cells.
  • the population may be a pure population comprising one cell type, such as a population of dopaminergic neurons, or a population of undifferentiated stem cells.
  • the population may comprise more than one cell type, for example a mixed cell population.
  • stem cell refers to a cell with the ability to divide for indefinite periods in culture and to give rise to specialized cells.
  • embryonic stem cell and “ESC” refer to a primitive (undifferentiated) cell that is derived from preimplantation-stage embryo, capable of dividing without differentiating for a prolonged period in culture and are known to develop into cells and tissues of the three primary germ layers.
  • a human embryonic stem cell refers to an embryonic stem cell that is from a human embryo.
  • the term “human embryonic stem cell” or “hESC” refers to a type of pluripotent stem cells derived from early-stage human embryos, up to and including the blastocyst stage, that is capable of dividing without differentiating for a prolonged period in culture, and are known to develop into cells and tissues of the three primary germ layers.
  • embryonic stem cell line refers to a population of embryonic stem cells which have been cultured under in vitro conditions that allow proliferation without differentiation for up to days, months to years.
  • pluripotent refers to an ability to develop into the three developmental germ layers of the organism including endoderm, mesoderm, and ectoderm.
  • iPSC induced pluripotent stem cell
  • OCT4, SOX2, and KLF4 transgenes a type of pluripotent stem cell formed by the introduction of certain embryonic genes (such as but not limited to OCT4, SOX2, and KLF4 transgenes) (see, for example, Takahashi and Yamanaka Cell 126, 663-676 (2006), herein incorporated by reference) into a somatic cell.
  • neuron refers to a nerve cell, the principal functional units of the nervous system.
  • a neuron consists of a cell body and its processes — an axon and one or more dendrites. Neurons transmit information to other neurons or cells by releasing neurotransmitters at synapses.
  • undifferentiated refers to a cell that has not yet developed into a specialized cell type.
  • the term “differentiation” refers to a process whereby an unspecialized embryonic cell acquires the features of a specialized cell such as a neuron, heart, liver, or muscle cell. Differentiation is controlled by the interaction of a cell's genes with the physical and chemical conditions outside the cell, usually through signaling pathways involving proteins embedded in the cell surface.
  • inducing differentiation in reference to a cell refers to changing the default cell type (genotype and/or phenotype) to a non-default cell type (genotype and/or phenotype).
  • inducing differentiation in a stem cell refers to inducing the stem cell (e.g., human stem cell) to divide into progeny cells with characteristics that are different from the stem cell, such as genotype (e.g., change in gene expression as determined by genetic analysis such as a microarray) and/or phenotype (e.g., change in expression of one or more protein markers).
  • markers refers to gene or protein that identifies a particular cell or cell type.
  • a marker for a cell may not be limited to one marker, markers may refer to a “pattern” of markers such that a designated group of markers may identity a cell or cell type from another cell or cell type.
  • linker refers to a linking moiety that connects two groups and has a backbone of 100 atoms or less in length.
  • a linker or linkage may be a covalent bond that connects two groups or a chain of between 1 and 100 atoms in length, for example of 1, 2, 3, 4, 5, 6, 8, 10, 12, 14, 16, 18 or 20 carbon atoms in length, where the linker may be linear, branched, cyclic or a single atom.
  • one, two, three, four or five or more carbon atoms of a linker backbone may be optionally substituted with a sulfur, nitrogen or oxygen heteroatom.
  • the bonds between backbone atoms may be saturated or unsaturated, usually not more than one, two, or three unsaturated bonds will be present in a linker backbone.
  • the linker may include one or more substituent groups, for example with an alkyl, aryl or alkenyl group.
  • a linker may include, without limitations, polyethylene glycol); ethers, thioethers, tertiary amines, alkyls, which may be straight or branched, e.g., methyl, ethyl, n- propyl, I -methylethyl (isopropyl), n-butyl, n-pentyl, 1,1 -dimethylethyl (t-butyl), and the like.
  • the linker backbone may include a cyclic group, for example, an aryl, a heterocycle or a cycloalkyl group, where 2 or more atoms, e.g., 2, 3 or 4 atoms, of the cyclic group are included in the backbone.
  • a linker may be cleavable or non-cleavable.
  • Alkyl refers to monovalent saturated aliphatic hydrocarbyl groups having from 1 to 10 carbon atoms and such as 1 to 6 carbon atoms, or 1 to 5, or 1 to 4, or 1 to 3 carbon atoms, In some cases, a “lower alkyl” is an alkyl group having 1 to 6 carbon atoms.
  • This term includes, by way of example, linear and branched hydrocarbyl groups such as methyl (CH 3 — ), ethyl (CH 3 CH 2 — ), n-propyl (CH 3 CH 2 CH 2 — ), isopropyl ((CH 3 ) 2 CH— ), n-butyl (CH 3 CH 2 CH 2 CH 2 - ), isobutyl ((CH 3 ) 2 CHCH 2 — ), sec-butyl ((CH 3 )(CH 3 CH 2 )CH— ), t-butyl ((CH 3 ) 3 C— ), n-pentyl (CH 3 CH 2 CH 2 CH 2 CH 2 — ), and neopentyl ((CH 3 ) 3 CCH 2 -).
  • linear and branched hydrocarbyl groups such as methyl (CH 3 — ), ethyl (CH 3 CH 2 — ), n-propyl (CH 3 CH 2 CH 2 — ), isoprop
  • substituted alkyl refers to an alkyl group as defined herein wherein one or more carbon atoms in the alkyl chain have been optionally replaced with a heteroatom such as — O — , — N — , — S — , — S(O) n — (where n is 0 to 2), — NR — (where R is hydrogen or alkyl) and having from 1 to 5 substituents selected from the group consisting of alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, oxo, thioketo, carboxyl, carboxylalkyl, thioaiyloxy, thi
  • chemoselective functional group and “chemoselective tag” are used interchangeably and refer to chemoselective reactive groups that selectively react with one another to form a covalent bond.
  • Chemoselective functional groups of interest include, but are not limited to, two thiol groups, thiols and maleimide or iodoacetamide, as well as groups that can react with one another via Click chemistry, e.g., azide and alkyne groups (e.g., cyclooctyne groups).
  • Chemoselective functional groups of interest include, but are not limited to, thiols, alkyne, a cyclooctyne, an azide, a phosphine, a maleimide, an alkoxyamine, an aldehyde and protected versions thereof, and precursors thereof.
  • the chemoselective functional group is a thiol.
  • the term “terminal group” refers to group(s) which are produced as a result of any convenient method of polymerization of the subject co-monomers described herein, e.g., H, an alkyl or a substituted alkyl and/or a residual component of an initiator used during polymerization.
  • modifying agent refers to any convenient agent that provides for a desirable property of interest (e.g., a desirable physical and/or biological property) and which is capable of conjugation to the thermoreversible polymer, e.g., via a chemoselective functional group on a sidechain linker or terminal of the polymer.
  • a desirable property of interest e.g., a desirable physical and/or biological property
  • Such an agent may belong to the class of small molecule, protein, peptide, sugar, polynucleotide, etc.
  • Modifying agents of interest include, but are not limited to, a ligand, a substrate, an enzyme, a pharmaceutical agent (e.g., a chemotherapeutic agent), a plasmid, a polynucleotide, a bioactive peptide, an antibody, a biomarker, a bio-sensor, a catalyst, an element, a cell targeting agent, small drug molecules, fluorescent/radioactive/optical imaging agents, peptides/proteins/enzymes, nucleic acids (siRNA/RNA/DNA/etc.), metal based compounds/catalysts, site-specific cellular targeting agents (compounds/ligands/antibodies/etc.) and smart adjuvants, gene therapy vectors.
  • a pharmaceutical agent e.g., a chemotherapeutic agent
  • a plasmid e.g., a chemotherapeutic agent
  • a polynucleotide e.g., a chemotherapeutic
  • the modifying agent is selected from a heparin, a hyaluronic acid, a specific binding member, a peptide, a nucleic acid, gelatin, fibronectin, collagen, laminin, bFGF, EGF, insulin, progesterone, glucose, thymosin beta-4, SHH, Noggin, Activin, TGFb3, FGF8, BDNF, GDNF, NT3, PDGF-AA and IGF-1.
  • the modifying agent is a cytokine, a BMP family member (e.g., TGFbeta or activin), a neutrophin (e.g., NT3 or BDNF) or a hedgehog protein (e.g., SHH).
  • a BMP family member e.g., TGFbeta or activin
  • a neutrophin e.g., NT3 or BDNF
  • a hedgehog protein e.g., SHH.
  • any convenient methods may be utilized in conjugating a modifying agent to a thermoreversible polymer. Conjugation methods and chemistries of interest include, but are not limited to, those described by Greg Hermanson in Bioconjugate Techniques (Third edition) 2013, Academic Press.
  • the modifying agent is a protein. In certain embodiments, the modifying agent is a peptide.
  • the modifying agent is peptidic and can be conjugated to the thermoreversible polymer (e.g., via a terminal and/or a sidechain functional group) by covalent attachment to the N-terminal or C-terminal or the peptidic agent, or covalent attachment to an amino acid sidechain (e.g., an amino, thiol, hydroxyl, carboxylic acid or phenol-containing amino acid sidechain group, or a derivative thereof).
  • the modifying agent is a heparin.
  • the heparin modifying agent is linked via a thiol linkage.
  • the heparin can be linked to the subject polymers via conjugation to a carboxylic acid group of the heparin.
  • FIG. 9 depicts exemplary methods of linking thiol-heparin to an acrylate group of the polymer by Michael addition.
  • two or more modifying agents e.g., a heparin and a hyaluronic acid
  • the lower critical solution temperature refers to the critical temperature below which the components of a mixture are miscible for all compositions.
  • the word lower in the term indicates that the LCST is a lower bound to a temperature interval of partial miscibility, or miscibility for certain compositions only.
  • the present inventors have discovered that the methods described herein enable increases in production of MGE progenitor cells of at least 10-fold compared to standard 2- dimensional culture formats, with at least 4-fold higher cell viability upon harvest.
  • an in vitro method for differentiating human stem cells comprising: encapsulating human stem cells in a three dimensional synthetic hydrogel and contacting the encapsulated human stem cells with at least one inhibitor of Small Mothers against Decapentaplegic (SMAD) signaling and with at least one Wingless (Wnt) antagonist for a predetermined amount of time; and contacting the cells with at least one activator of Sonic Hedgehog (SHH) signaling for a predetermined amount of time to obtain a cell population comprising MGE progenitor cells.
  • SAD Small Mothers against Decapentaplegic
  • Wnt Wingless
  • SHH Sonic Hedgehog
  • the cell population comprising MGE progenitor cells exhibits the following features: (a) at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, or at least 86% or at least 87% of the cell population is FoxGl -positive; and (b) at least 60%, at least 65%, at least 70%, at least 75%, at least 76%, or at least 77% of the cell population is NKX2-l-positive.
  • FOXG1 -positive NKX2- 1 -positive MGE progenitor cells comprise at least at least 60%, at least 65%, at least 70%, at least 75%, or at least 80% of the cell population, e.g., as measured by flow cytometry, immunocytochemistry and/or qPCR.
  • the method further comprises contacting the produced encapsulated MGE progenitor cells with at least one neurotropic factor and optionally a notch inhibitor for a predetermined period of time to obtain a population of cells comprising FOXG1- positive differentiated inhibitory GABAergic cortical interneurons (cINs).
  • the cell population comprising cINs exhibits the following features: (a) at least 60%, at least 65%, at least 70%, at least 75%, at least 76%, or at least 77% of the cell population is FoxGl -positive;
  • At least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 81%, at least 82%, at least 83%, or at least 84% of the cell population is GABA-positive;
  • at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 81%, at least 82%, or at least 83% of the cell population is PV-positive;
  • less than 30%, less than 25%, less than 24%, or less than 23% of the cell population is NKX2- 1 -positive and/or (e) less than 5%, less than 4%, less than 3%, less than 2% or less than 1% of the cell population is ki67 -positive, as measured by e.g., flow cytometry, immunocytochemistry and/or qPCR.
  • the synthetic hydrogel for use in the methods described herein is a polyethylene glycol-poly(N-isopropylacrylamide) (PEG-PNIPAAM) based hydrogel that is solid at 37° C.
  • PEG-PNIPAAM hydrogels include those described in U.S. Patent No.
  • the hydrogel comprises a thermoreversible polymer comprising: a N-isopropylacrylamide (NIPAAM) co-monomer; a lower alkyl amine comonomer; and a poly(ethylene glycol) (PEG) co-monomer, wherein the terminal PEG monomer is substituted with alkyl, substituted alkyl, heteroalkyl, substituted heteroalkyl, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, aryl, substituted aryl, arylalkyl, substituted arylalkyl, heteroaryl, substituted heteroaryl, heteroarylalkyl, and substituted heteroaiylalkyl.
  • NIPAAM N-isopropylacrylamide
  • PEG poly(ethylene glycol)
  • the lower alkyl amine co-monomer comprises n-butyl, isobutyl, tertbutyl, n-propyl, pentyl, isopropyl, or isopentyl; and the terminal PEG monomer is substituted with an alkoxy group.
  • thermoreversible polymer comprises the formula (I): wherein: a, b, and c are molar fractions of the co-monomers, where a, b, and c are each greater than zero, preferably a > 0.8; 0.2 > b > 0; and 0.1 > c > 0; PEG n is a polyethyleneglycol polymer and n is an integer from 1 to 2500;
  • R 1 is an alkyl or a substituted alkyl, preferably C1-C6 alkyl, more preferably butyl;
  • R 2 is alkyl, substituted alkyl, heteroalkyl, substituted heteroalkyl, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, aryl, substituted aryl, arylalkyl, substituted arylalkyl, heteroaryl, substituted heteroaryL heteroatylalkyl, and substituted heteroaiylalkyl, preferably is alkoxy, more preferably methoxy; and G 1 and G 2 are each independently selected from a polymer segment, a terminal group, a linker and a linked modifying agent.
  • novel thermoreversible polymers comprising the formula III, as well as 3-dimensional hydrogels comprising the novel thermoreversible polymers and their use in the methods described herein (e.g., culturing and/or differentiating stem cells): wherein a, b, c, d represent molar fractions of the co-monomers in the polymer, wherein a, b, and c are each greater than 0.
  • R 1 if present, is any terminal or functional group not including a primary amine
  • R 2 is a lower alkyl group
  • G 1 and G 2 are each independently selected from a polymer segment, a terminal group, a linker and a linked modifying agent, and wherein the molecular weight of the polymer is greater than 50 kDa.
  • thermoreversible polymers of formula III have several advantages over the thermoreversible polymers disclosed e.g., in U.S. Patent No. 10,982,055 and U.S. Patent Publication No. 2024/0294713.
  • a thermoreversible polymer according to the present disclosure exhibits a combination of properties rendering them uniquely suitable for 3-dimensional cell culture.
  • a 3-dimensional hydrogel comprising a thermoreversible polymer of formula III exhibits, inter alia, (1) long-term stability, enabling lengthy differentiation processes to occur during cell culture (2) decreased viscosity relative to prior art polymers, enabling efficient cell encapsulation and (3) appropriate stiffness supporting cell growth.
  • thermoreversible polymer of formula III has one or more, preferably all, of the following properties: (a) LCST of 12-32° C (b) stiffness of 100-8000 Pa (c) viscosity of 100-2000 cP and (d) molecular weight of 50-500 kDa.
  • R 1 of the thermoreversible polymer of formula III is absent.
  • R 1 of the thermoreversible polymer of formula III is a functional group.
  • the functional group is a chemoselective functional group, nonlimiting examples of which include two thiol groups, thiols and maleimide or iodoacetamide, as well as groups that can react with one another via Click chemistry, e.g., azide and alkyne groups (e.g. cyloalkyne groups such as dibenzocyclooctyne (DBCO)).
  • DBCO dibenzocyclooctyne
  • Functional groups include, but are not limited to, acrylate, thiols, hydroxyl, alkoxy (e.g., methoxy), alkyne, cycloalkyne, an azide, hydrazide, a phosphine, a maleimide, a carboxylic acid, an alkoxyamine, an aldehyde, biotin, silane, 1 ,2- Distearoyl-sn-glycero-3-phosphoethanolamine (DSPE), NHS ester, toluenesulfonyl (Tos), and protected versions and precursors thereof.
  • R 1 of the thermoreversible polymer of formula III is a terminal group, nonlimiting examples of which include, without limitation, C 1 -C 6 alkoxy (e.g., methoxy, ethoxy, n-propoxy, n-butoxy isobutoxy, tert-butoxy, pentoxy or isopentoxy), alkyl, substituted alkyl, heteroalkyl, substituted heteroalkyl, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, aryl, substituted aryl, arylalkyl, substituted arylalkyl, heteroaryl, substituted heteroaryl, heteroarylalkyl, and substituted heteroarylalkyl.
  • R 1 is other than an alkyl (e.g., is other than n-butyl) or substituted alkyl.
  • R 2 of the thermoreversible polymer of formula III is a lower alkyl, optionally selected from methyl, ethyl, propyl, n-butyl, pentyl, isopropyl, isobutyl, isopentyl, tert-butyl, cyclopropyl, and cyclobutyl.
  • R 2 is n-butyl. The present inventors have discovered that n-butyl at this position exhibits higher stiffness with similar viscosity, higher gel stability and lower gel LCST compared to other conformations such as iso-butyl.
  • R 2 is a lower alkyl other than isobutyl.
  • R 3 of the thermoreversible polymer of formula III is absent.
  • R 3 of the thermoreversible polymer of formula III is a modifying agent, optionally selected from selected from a heparin, a hyaluronic acid, a specific binding member, a peptide, a nucleic acid, gelatin, fibronectin, collagen, laminin, basic fibroblast growth factor (bFGF), epidermal growth factor (EGF), insulin, progesterone, glucose, stromal cell derived factor- 1 (SDF-1), thymosin beta-4, sonic hedgehog (SHH), Noggin, Activin, transforming growth factor- ⁇ (TGF- ⁇ ), FGF8, brain-derived neurotrophic factor (BDNF), glial cell-derived neurotrophic factor (GDNF), neurotrophic factor- 3 (NT3), platelet-derived growth factor (PDGF), IL- 16, IL-2 and insulin-like growth factor- 1 (IGF-1).
  • a heparin optionally selected from
  • R 4 is a lower alkyl group, particularly where R 4 is methyl.
  • G 1 and G 2 are each independently selected from a polymer segment, a terminal group, a linker and a linked modifying agent.
  • G 1 and G 2 are each independently selected from a heparin, a hyaluronic acid, a member of a specific binding pair, a polypeptide, a nucleic acid and a carboxyl group.
  • G 1 and G 2 are each independently a modifying agent selected from gelatin, elastin, fibronectin, collagen, and laminin.
  • G 1 and G 2 are each independently selected from a chemokine, a peptide hormone, and a growth factor.
  • G 1 and G 2 are each independently selected from fibroblast growth factor, epidermal growth factor, hepatic growth factor insulin, stromal cell-derived factor- 1, thymosin beta-4, sonic hedgehog, Noggin, activin, transforming growth factor, bone morphogenic protein, brain- derived neurotrophic factor, glial cell-derived neurotrophic factor, neurotrophin-3, platelet-derived growth factor, FGF-2, FGF-8, keratinocyte growth factor, or insulin-like growth factor.
  • G 1 and G 2 are each independently selected from a chain-transfer agent non-limiting examples of which include a dithioester, dithiocarbamate, trithiocarbonate or a xanthate.
  • G 1 and G 2 are each independently selected from a chain transfer agent containing a thiolcarbonylthio group and a thermal initiator such as Azobisisobutyronitrile (AIBN).
  • thermoreversible polymer of formula III has a molecular weight (MW) of between 50-500 kDa or between 50-250 kDa. In certain embodiments, the thermoreversible polymer has a MW of at least 50 kDa, at least 60 kDa, at least 70 kDa, at least 80 kDa or at least 90 kDa.
  • the thermoreversible polymer has a MW in the range of about 50 kDa to about 250 kDa, such as about 50 kDa to about 200 kDa, about 50 kDa to about 150 kDa, about 50 kDa to about 100 kDa, or about 50 kDa to about 75 kDa. In certain embodiments, the thermoreversible polymer has a MW of about 60 kDa to about 140 kDa, about 70 kDa to about 130 kDa, about 80 kDa to about 120 kDa or about 90 kDa to about 110 kDa.
  • the MW is about 80 kDa, about 85 kDa, about 90 kDa, about 95 kDa, about 100 kDa, about 105 kDa, about 110 kDa, about 115 kDa, or about 120 kDa.
  • the thermoreversible polymer has a MW of about 50 kDa to about 250 kDa or from about 50 to about 150 kDa.
  • PEGn is a polyethylglycol polymer having a MW of about 2-20 kDa. In related embodiments, PEGn has a MW of about 2 kDa or greater, such as 2 kDa to 20 kDa, or 2 kDa to 10 kDa, or 3 kDa to 20 kDa, or 3 kDa to 10 kDa.
  • the PEGn has a MW of about 2 kDa to about 9 kDa, about 3 kDa to about 8 kDa, about 4 kDa to about 7 kDa, about 4 kDa to about 6 kDa. In some embodiments, the PEG n has a MW of about 2 kDa, about 3 kDa, about 4 kDa, about 5 kDa, about 6 kDa, about 7 kDa, about 8 kDa or about 9 kDa.
  • the PEGn group can be modified with any convenient groups, including terminal modifications..
  • a thermoreversible polymer of formula III has a weightweight (w/w) ratio of PEG to PNIPAAm copolymer above 1 :2.
  • the w/w ratio of PEG to PNIPAAm copolymer is about 1 :2.5, about 1:2.75, about 1 :3.0, about 1 :3.5, about 1 :3.75, about 1 :4.0, about 1 :4.25, or about 1 :4.5.
  • the w/w ratio of PEG to PNIPAAm copolymer is about 1:3 to about 1 :4.5, or about 1 :4.
  • the w/w ratio of PEG to PNIPAAm copolymer is measured according to the relative molecular ratio with nuclear magnetic resonance (NMR) and the total polymer molecular weight with gel permeation chromatography (GPC) (GPC-NMR analysis).
  • the weight of the PNIPAAm copolymer is defined as the weight of the copolymer backbone (i.e., prior to addition of PEG) following polymerization of the comonomers.
  • the weight of the PNIPAAm copolymer is defined as the weight of: following polymerization of the comonomers:
  • the weight of PEG is defined as the weight of prior to reaction with the copolymer backbone.
  • a thermoreversible polymer of formula II comprises a PEG:PNIPAAM weight ratio above 1 :2 and a MW between 50 kDa and 250 kDa (including any range therebetween).
  • a polymer of formula III having PEG:PNIPAAM weight ratios above 1 :2 (e.g., ratio of 1 :3) and low MW (e.g., 50kDa-250 kDa) provides several surprising advantages over polymers disclosed in, e.g., U.S. Patent No.
  • thermoreversible polymer of formula III therefore provides a scalable three-dimensional cell culture system over time periods relevant for e.g., production of functional neurons.
  • thermoreversible polymer of formula III has an LCST in the range of 12-32° C, such as 12-30° C., 15-30° C., 15-25° C. or 10-20° C. In preferred embodiments, the thermoreversible polymer of formula III has an LCST of about 20° C to about 22° C.
  • compositions comprising: a) a 3 -dimensional hydrogel comprising a thermoreversible polymer of Formula III; and b) cells encapsulated within the hydrogel.
  • the 3- dimensional hydrogel-cell composition is useful for generating a desired number of cells, by culturing the 3-dimensional hydrogel-cell composition under conditions and for a period of time sufficient to generate the desired number of cells.
  • the period of time sufficient to generate the desired number of cells is at least 14 days, at least 15 days, at least 16 days, at least 17 days, at least 18 days, at least 19 days, at least 20 days, at least 21 days, at least 22 days, at least 23 days, at least 24 days, at least 25 days, at least 26 days, at least 27 days, at least 28 days, at least 29 days, at least 30 days, at least 31 days, at least 32 days, at least 33 days, at least 34 days, at least 35 days, at least 40 days, at least 45 days, at least 50 days, at least 55 days, at least 60 days, at least 3 months, at least 4 months, at least 6 months, or more.
  • Such cells can include stem cells, differentiated cells, and the like.
  • thermoreversible polymer-cell composition of Formula III is particularly useful for differentiating cells, e.g., to generate a desired number of differentiated cells over a relatively long period of time.
  • a 3-dimensional thermoreversible polymer-cell composition of the present disclosure can be implanted into an individual in need thereof, where cells proliferate and/or differentiated within the implanted thermoreversible polymer-cell composition, and migrate out of the implanted thermoreversible polymer-cell composition.
  • the present disclosure provides a composition comprising a plurality of hydrogel capsules, wherein at least 90%, at least 95%, at least 98% or at least 99% of said hydrogel capsules in said composition comprise a cell and a hydrogel encapsulating said cell, wherein said hydrogel encapsulating said cell comprises a thermoreversible polymer of formula III.
  • at least 90% of said hydrogel capsules each comprises multiple cells, e.g., at least 100, at least 200, at least 500, at least 700, at least 800, at least 900 or more cells.
  • thermoreversible polymer of formula III provides improvements, including but not limited to, bead (i.e., capsule) homogeneity, bead circularity, bead sphericity, reduction in bead volume and diameter, flow rate to create the beads, and shape of the beads from gravity drip extrusion.
  • the conditions suitable for differentiation of the stem cells comprise culturing the stem cell or precursor cell in the hydrogel composition for a period of at least 14 days, at least 15 days, at least 16 days, at least 17 days, at least 18 days, at least 19 days, at least 20 days, at least 21 days, at least 22 days, at least 23 days, at least 24 days, at least 25 days, at least 26 days, at least 27 days, at least 28 days, at least 29 days, at least 30 days, at least 31 days, at least 32 days, at least 33 days, at least 34 days, at least 35 days, at least 40 days, at least 45 days, at least 50 days, at least 55 days, at least 60 days, at least 3 months, at least 4 months, at least 6 months, or more to produce the differentiated cell population.
  • the present disclosure provides methods for expanding stem cells, the method comprising encapsulating single cells or multicellular clusters in a three-dimensional hydrogel comprising a thermoreversible polymer of formula III and culturing the cells under suitable stem cell expansion conditions.
  • the subject thermoreversible polymers can be prepared using any convenient methods.
  • a variety of polymerization methods may be utilized in preparing a base polymeric material, e.g., including polyacrylate, polyacrylamide and mixtures thereof.
  • a variety of derivatization methods may be utilized to introduce any convenient functionality into the subject base polymeric materials.
  • a variety of chemoselective conjugation chemistries, linkers, functional groups and modifying agents may be utilized in the preparation of further derivatives and conjugates of the subject base polymeric materials and derivatives thereof.
  • stem cell refers to a cell that is totipotent or pluripotent or multipotent and is capable of differentiating into one or more different cell types.
  • the term includes without limitation as embryonic stem cells, stem cells isolated from organs, for example, skin stem cells, and induced pluripotent stem cells (iPSC).
  • iPSC induced pluripotent stem cells
  • totipotent refers to an ability of a cell to differentiate into any type of cell in a differentiated organism, as well as cell of extraembryonlc materials such as placenta.
  • iPSC induced pluripotent stem cell
  • somatic e.g., adult
  • progenitor in reference to a cell refers to an intermediate cell stage wherein said cell is no longer a pluripotent stem cell and is also not yet a fully committed cell. Progenitor cells in this disclosure are included within somatic cells.
  • multipotent refers to a cell line capable of differentiating into at least two terminally differentiated cell types.
  • Small Mothers against Decapentaplegic refers, generally, to a class of signaling molecules that are capable of modulating the directed cell differentiation of stem cells.
  • SMADs are intracellular proteins that transduce extracellular signals from transforming growth factor beta ligands to the nucleus where they activate downstream gene transcription and are members of a class of signaling molecules capable of modulating directed differentiation of stem cells.
  • inhibitors of SMAD signaling include compounds that interact with and reduce or block the activity of SMAD and/or a molecule that is associated with SMAD or other component molecules of SMAD signaling.
  • Inhibitors can bind directly to and cause a conformational change in SMAD signaling, can reduce or prevent the expression of the gene encoding SMAD or a SMAD target gene, can decrease SMAD protein levels, and/or can interfere with SMAD interactions with one or more signaling partners.
  • Inhibitors also include molecules that indirectly regulate SMAD biological activity by intercepting upstream signaling molecules (e.g., within the extracellular domain), examples of a signaling molecule and an effect include: Noggin which sequesters bone morphogenic proteins, inhibiting activation of ALK receptors 1, 2, 3, and 6, thus preventing downstream SMAD activation. Likewise, Chordin, Cerberus, Follistatin, similarly sequester extracellular activators of SMAD signaling. Bambi, a transmembrane protein, also acts as a pseudo-receptor to sequester extracellular TGF
  • an inhibitor of the present disclosure induces (changes) or alters differentiation from a default to a non-default cell type, for example, one of the methods of the present disclosure comprising at least three inhibitors that produced a non-default neural progenitor cell.
  • Inhibitors disclosed herein “alter” or “lower” or “block” default signaling in order to direct cellular differentiation towards a nondefault cell type, such as described herein for producing cortical interneurons as disclosed herein.
  • an inhibitor of the present disclosure can be a biological compound, natural or synthetic, or a small molecule for increased or decreased signal molecule activity that assists in producing for example cortical interneurons of the present disclosure.
  • Inhibitors are described in terms of competitive inhibition (binds to the active site in a manner as to exclude or reduce the binding of another known binding compound) and allosteric inhibition (binds to a protein in a manner to change the protein conformation in a manner which interferes with binding of a compound to that protein's active site) in addition to inhibition induced by binding to and affecting a molecule upstream from the named signaling molecule that in turn causes inhibition of the named molecule.
  • SMAD inhibitors that may be advantageously employed in the methods disclosed herein include those SMAD inhibitors that are well known in and are readily available to those having skill in the art. SMAD inhibitors that have been employed for the neural conversion of human ESCs and iPSCs are described in Chambers et al., Nat Biotechnol 27:275-280 (2009).
  • Exemplary SMAD inhibitors that can be used in the methods and compositions disclosed herein include the compounds designated SB431542, LDN-193189, Noggin PD 169316, SB203580, LY364947, A77-01, A-83-01, BMP4, GW788388, GW6604, SB- 505124, lerdelimumab, metelimumab, GC-I008, AP-12009, AP-11OI4, LY550410, LY580276, LY364947, LY2109761, SB-505124, E-616452 (RepSox ALK inhibitor), SD-208, SMI6, NPC- 30345, Ki26894, SB-203580, SD-093, activin-M108A, P144, soluble TBR2-Fc, DMH-1, Dorsomorphin dihydrochloride, and a derivative and/or variant thereof, wherein each derivative and/or variant thereof possesses one or more SMAD inhibitory activities.
  • TGF ⁇ /Activin/Nodal pathway includes inhibition of TGF ⁇ /Activin/Nodal pathway and the BMP pathway.
  • exemplary TGF ⁇ /activin pathway inhibitors include but are not limited to: TGF beta receptor inhibitors, inhibitors of SMAD 2/3 phosphorylation, inhibitors of the interaction of SMAD 2/3 and SMAD 4, and activators/agonists of SMAD 6 and SMAD 7.
  • TGF beta receptor inhibitors include but are not limited to: TGF beta receptor inhibitors, inhibitors of SMAD 2/3 phosphorylation, inhibitors of the interaction of SMAD 2/3 and SMAD 4, and activators/agonists of SMAD 6 and SMAD 7.
  • the categorizations described below are merely for organizational purposes and one of skill in the art would know that compounds can affect one or more points within a pathway, and thus compounds may function in more than one of the defined categories.
  • TGF beta receptor (e.g., ALK5) inhibitors can include antibodies to, dominant negative variants of, and antisense nucleic acids that suppress expression of, TGF beta receptors (e.g., ALK5).
  • Exemplary TGF ⁇ receptor/ALK5 inhibitors include, but are not limited to, SB431542 (see, e.g., Inman, et al., Molecular Pharmacology 62(l):65-74 (2002)), A-83-01, also known as 3-(6-Methyl-2-pyridinyl)-N-phenyl-4-(4-quinolinyl)- 1 H-pyrazole-1 -carbothioamide (see, e.g., Tojo, et al., Cancer Science 96(11):791-800 (2005), and commercially available from, e.g., Toicris Bioscience); 2-(3-(6-Methylpyridin-2-yl)-IH-pyrazol-4-yl
  • an ALK5 inhibitor is not intended to encompass non-specific kinase inhibitors
  • an “ALK5 10 inhibitor” should be understood to encompass inhibitors that inhibit ALK4 and/or ALK7 in addition to ALK5, such as, for example, SB-431542. See, e.g., Inman et al., J Mol Phamacol 62(l):65-74 (2002). Without intending to limit the scope of the disclosure, it is believed that ALK5 inhibitors affect the mesenchymal to epithelial conversion/transition (MET) process. TGF ⁇ /activin pathway is a driver for epithelial to mesenchymal transition (EMT). Therefore, inhibiting the TGF ⁇ /activin pathway can facilitate MET (i.e., reprogramming) process.
  • EMT epithelial to mesenchymal transition
  • inhibitors include but are not limited to SU5416; 2-(5 - benzo[l ,3]dioxol-5-yl-2-tert-butyl-3H-imidazol-4-yl)-6-methylpyridine hydrochloride (SB- 505124); lerdelimumb (CAT-152); metelimumab (CAT-192); GC-I008; ID11; AP-12009; AP- 11OI4; LY550410; LY580276; LY364947; LY2109761; SB-505124; SB-431542; SD-208; SMI6; NPC-30345; K126894; SB-203580; SD-093; Gleevec; 3,5,7,2',4'-entahydroxyflavone (Morin); activin-M108A; P144; soluble TBR2-Fc; and antisense transfected tumor cells that target TGF beta receptors.
  • SB- 505124 2-(5
  • Inhibitors of SMAD 2/3 phosphorylation can include antibodies to, dominant 5 negative variants of and antisense nucleic acids that target SMAD2 or SMAD3.
  • Specific examples of inhibitors include PD169316; SB203580; SB-431542; LY364947; A77-01; and 3,5,7,2',4'-pentahydroxyflavone (Morin).
  • Moorin 3,5,7,2',4'-pentahydroxyflavone
  • SB-431542 i.e., CAS 301836-41-9; IUPAC 4-[4-(l,3-benzodioxol-5-yl)-5-(2- pyridinyl)-lH-imidazol-2-yl]-benzamide
  • SMAD small molecule inhibitor of SMAD, which is capable of lowering or blocking transforming growth factor beta (TGFP)/Activin-Nodal signaling.
  • Inhibitors of the interaction of SMAD 2/3 and SMAD 4 can include antibodies to, dominant negative variants of and antisense nucleic acids that target SMAD2, SMAD3 and/or smad4.
  • Specific examples of inhibitors of the interaction of SMAD 2/3 and SMAD4 include but are not limited to Trx-SARA, Trx-xFoxHlb and Trx-Lefl. (See, e.g., Cui, et al., Oncogene 24:3864-3874 (2005) and Zhao, et al., Molecular Biology of the Cell, 17:3819-15 3831 (2006).)
  • Exemplary BMP pathway inhibitors include, but are not limited to: Noggin, BMP receptor inhibitors, inhibitors of SMAD 1/5/8 phosphorylation, inhibitors of the interaction of SMAD 1/5/8 and SMAD 4, and activators/agonists of SMAD 6 and SMAD 7.
  • Noggin BMP receptor inhibitors
  • inhibitors of SMAD 1/5/8 phosphorylation inhibitors of the interaction of SMAD 1/5/8 and SMAD 4
  • the categorizations described below are merely for organizational purposes and one of skill in the art would know that compounds can affect one or more points within a pathway, and thus compounds may function in more than one of the defined categories.
  • Inhibitors of SMAD 1/5/8 phosphorylation include, but are not limited to, antibodies to, dominant negative variants, antisense nucleic acids, and small molecules that target SMAD 1, SMAD 5, or SMAD 8. Specific examples of inhibitors include LDN-193189 and Dorsomorphin (commercially available from, e.g., Stemgent)
  • BMP receptor inhibitors include, but are not limited to, antibodies to, dominant negative variants of, siRNA or antisense nucleic acids, or small molecules that target BMP receptors.
  • Specific examples of inhibitors include, but are not limited to, DMH-1, Dorsomorphin dihydrochloride, and LDN-193189 (commercially available, from, e.g., Tocris Biosciences).
  • LDN193189 i.e., DM-3189, IUPAC 4-(6-(4-(piperazin-l-yl)phenyl)pyrazolo[l,5- a]pyrimidin-3-yl)quinolone
  • DM-3189 i.e., IUPAC 4-(6-(4-(piperazin-l-yl)phenyl)pyrazolo[l,5- a]pyrimidin-3-yl)quinolone
  • LDN 193189 is also a highly potent small molecule inhibitor of ALK2, ALK3, ALK6, protein tyrosine kinases (PTK), inhibiting signaling of members of the ALK1 and ALK3 families of type I TGF ⁇ receptors, resulting in the inhibition of the transmission of multiple biological signals, including the bone morphogenetic proteins (BMP) BMP2, BMP4, BMP6, BMP7, and Activin cytokine signals and subsequently SMAD phosphorylation of Smadl, Smad5, and Smad8. Yu et al., Nat Med. 14:1363-1369 (2008) and Ctmy et al., Bioorg Med Chem Lett 18: 4388-4392 (2008).
  • the method comprises the use of the BMP inhibitor LDN- 193189 at a concentration of about 50-150 nM, preferably about 100 nM for a period of about 10 days to about 20 days, preferably about 14-16 days.
  • SMAD signaling inhibitors can comprise the dual-SMAD inhibitors SB431542 and LDN- 193189, or functional derivatives and/or variants thereof.
  • LSB dual-SMAD inhibitors
  • XLSB XLSB
  • SB431542 can be contacted with the pluripotent and/or a multipotent cell at a final concentration in an in vitro culture of from about 0.1 ⁇ M to about 1 mM.
  • LDN- 193189 can be contacted with the pluripotent and/or a multipotent cell at a final concentration in an in vitro culture of from about 1 nM to about 10 ⁇ M.
  • Wnt refers to a signal pathway composed of Wnt family ligands and Wnt family receptors, such as Frizzled and LRPDerailed/RYK receptors, mediated with or without ⁇ -catenin. Wnt proteins have been implicated in oncogenesis and in several developmental processes, including regulation of cell fate and patterning during embryogenesis.
  • the Wnt pathway includes any of the proteins downstream or upstream of Wnt protein activity.
  • this could include LRPS, LRP6, Dkk, GSK-3, WntlOB, Wnt6, Wnt3 (e.g., Wnt 3A), Wntl or any of the other proteins discussed herein, and the genes that encode these proteins.
  • the Wnt pathway also includes pathways that are downstream of Wnt, such as the LRPS or HBM pathways, the Dkk pathway, the p-catenin pathway, the MAPKAPK2 pathway, the OPG/RANK pathway, and the like.
  • LRP5 pathway and IBM pathway are meant any proteins/genes including LRP5 or the HBM mutant and proteins downstream of LRPS or the HBM mutant.
  • ⁇ -catenin pathway is meant any proteins/genes including ⁇ -catenin and proteins downstream of ⁇ -catenin.
  • MAPKAPK2 pathway is meant any proteins/genes including MAPKAPK2 and proteins downstream of MAPKAPK2.
  • OPG/RANKL pathway 1 any proteins/genes including OPG/RANKL and proteins downstream of OPG and RANKL.
  • Dkk pathway is meant to include any proteins/genes involved in Dkk-1 and LRP5 and/or LRP6 interaction that is part of the Wnt pathway. Dkk-I inhibits LRP5 activity.
  • Wnt antagonist refers not only to any agent that may act by directly inhibiting the normal function of the Wnt protein, but also to any agent that inhibits the Wnt signaling pathway, and thus recapitulates the function of Wnt.
  • Wnt signaling antagonists include XAV939 (Hauang et al. Nature 461 :614-620 (2009)), vitamin A (retinoic acid), lithium, flavonoid, Dickkopfl (Dkkl), insulin-like growth factor-binding protein (IGFBP) (W02009/131166), and siRNAs against ⁇ -catenin.
  • Exemplary Wnt antagonists include, but are not limited to, XAV939, IWP-2, DKK1 (Dickkopf protein 1), and IWR1. Additional Wnt inhibitors include, but are not limited to, IWR compounds, IWP compounds, and other Wnt inhibitors described in WO09155001 and Chen et al., Nat Chem Biol 5:100-7 (2009).
  • the method comprises the use of the Wnt antagonist IWP-2 at a concentration of about 1-10 ⁇ M, preferably about 5 ⁇ M for a period of about 5 days to about 10 days, preferably about 7 or 8 days.
  • Dkk-3 Antagonistic activity of Dkk-3 has not been demonstrated.
  • Dkk proteins have distinct patterns of expression in adult and embryonic tissues and have a wide range of effects on tissue development and morphogenesis.
  • the Dkk family also includes Soggy, which is homologous to Dkk-3 but not to the other family members.
  • the sFRPs are a family of five Wnt-binding glycoproteins that resemble the membrane-bound Frizzleds. The largest family of Wnt inhibitors, they contain two groups, the first consisting of sFRP1, 2, and 5, and the second including sFRP3 and 4. All are secreted and derived from unique genes, none are alternate splice forms of the Frizzled family. Each sFRP contains an N-terminal cysteine-rich domain (CRO).
  • Other antagonists of Wnt signaling include WIF-1 (Wnt Inhibitory Factor 1), a secreted protein that binds to Wnt proteins and inhibits their activity.
  • SMAD inhibitors include, but not limited to, SB431542, LDN-193189, Noggin PD169316, SB203580, LY364947, A77-01, A-83-01, BMP4, GW788388, GW6604, SB-505124, lerdelimumb, metelimumab, GC-I008, AP-12009, AP-110I4, LY550410, LY580276, LY364947, LY2109761, SB-505124, SB-431542, SD-208, SMI6, NPC- 30345, Ki26894, SB-203580, SD-093, activin-M108A, P144, soluble TBR2-Fc, DMH-1, Dorsomorphin dihydrochloride and their derivatives.
  • Wnt antagonists include, but not limited to, XAV939, DKK1, SFRP-1, SFRP-2, SFRP-5, SFRP-3, SFRP-4, WIF-1, Soggy, IWP-2, IWR1 and their derivatives.
  • SB431542 and LDN 193189 can be used in combination to inhibit SMAD signaling pathway.
  • XAV939 can be employed to antagonize the Wnt signaling pathway.
  • the concentration of XAV939 in a cell culture can be about 0.2 ⁇ M to 20 ⁇ M; the concentration of LDN193189 in a cell culture can be from about 10 nM to about 1000 nM, and the concentration of SB431542 in a cell culture can be from about 1 ⁇ M to about 100 ⁇ M.
  • the concentrations of XAV939 can be about 2 ⁇ M, the concentration of LDN193189 can be about 100 nM, and the concentration of SB431542 can be about 10 ⁇ M.
  • forebrain progenitor cells are generated by contacting stem cells with XAV939, LDN193189, and/or SB431542 for a duration of from about 5 days to about 40 days. In related aspects, forebrain progenitor cells are generated by contacting stem cells with XAV939, LDN193189, and/or SB43I542 for a duration of from about 10 days to about 25 days.
  • Wnt signaling antagonists can be selected from the group consisting of XAV939, DKK1, DKK- 2, DKK-3, Dlck-4, SFRP-1, SFRP-2, SFRP-5, SFRP-3, SFRP-4, WIF-1, Soggy, IWP-2, IWR1, ICG-001, KY0211, Wnt-059, LGK974, IWP-L6, and a derivative and/or variant thereof, wherein each derivative and/or variant thereof possesses one or more Wnt signaling antagonist activities.
  • the Wnt signaling antagonist can comprise XAV939 or a functional derivative and/or variant thereof.
  • XAV939 can be contacted with the pluripotent and/or multipotent cell at a final concentration in an in vitro culture of from about 10 nM to about 500 ⁇ M.
  • a variety of cell culture media and supplement can be used to differentiate stem cells into forebrain progenitor cells, including KSR-medium, N2 medium (DMEM/FI2 with NaHCO3, N2B supplement (Stem Cell Technologies), and Neurobasal media with B27 (Gibco) and N2 supplements (Invitrogen), Kriks et al., Nature 480:547-551 (2011).
  • the cells are maintained on mouse embryonic fibroblasts (MEFs) as described previously and dissociated with Accutase (Innovative Cell Technologies) for differentiation or dispase for passaging (Chambers et al., Nat Biotechnol 27:275-280 (2009)).
  • MEFs mouse embryonic fibroblasts
  • NKX2.1 The ventral prosencephalic progenitor population transcription factor marker, NKX2.1, can be used to monitor differentiation. Sussel et al., Development 126:3359-3370 (1999) and Xu et al., JNeurosci 24:2612-2622 (2004). Inhibition of Wnt signaling enhances the production of FOXG1 and, subsequently, promotes ventralization by inducing the controlled, SHH-mediated differentiation of multipotent and pluripotent cells towards an NKX2.1+ forebrain progenitor fate.
  • neuronal cell lineages and populations may be achieved by contacting a neuronal precursor cells, such as a neuronal precursor cell generated as disclosed herein, with one or more activator of SHE signaling at a predetermined time after contacting a multipotent and/or pluripotent cell with one or more inhibitors of SMAD signaling and one or more antagonists of Wnt signaling and for a duration that is sufficient to induce the production of one or more markers of a cortical interneuron or precursor thereof
  • activator in the context of SHH refers to a compound that promotes and/or enhances SHH signaling thereby inducing a neuronal precursor cell to differentiate into a cortical interneuron or precursor thereof, a hypothalamic neuron or precursor thereof, and/or a pre-optic chorionic neuron or precursors thereof.
  • SHH signaling pathway activators useful in the present disclosure include proteins belonging to the hedgehog family (e.g., SHH), inhibitors of Pte's interaction with Smo, Smo receptor activators, Shh receptor activators (e.g., Hg — Ag, purmorphamine, etc.), substances increasing Ci/Gli family levels, inhibitors of the intracellular degradation of Ci/Gli factors, and SHH overexpression constructs or Ci/Gli overexpression constructs resulting from transfection.
  • SHH proteins belonging to the hedgehog family
  • inhibitors of Pte's interaction with Smo e.g., Smo receptor activators
  • Shh receptor activators e.g., Hg — Ag, purmorphamine, etc.
  • an SHH signaling pathway activator (e.g., SHH+ purmorphamine), is added to the cell culture for the full or partial duration of the culture.
  • the concentrations of SHH activators in cell culture are about 10 ng/mL to about 5000 ng/mL for SHH (or recombinant SHH) and about 0.1 ⁇ M to about 20 ⁇ M for purmorphamine.
  • the concentrations of SHH activators in cell culture are about 50 ng/mL to about 500 ng/mL for SHH (or recombinant SHH) and about 0.5 ⁇ M to about 4 ⁇ M for purmorphamine.
  • Purmorphamine is a commercially available small molecule with the name 9- cyclohexyl-N-[4-(morpholinyl)phenyl]-2-(1-naphthalenyloxy)-9H-purin-6-amine, and a chemical formula of C 31 H 32 N 6 O 2 .
  • SHH signaling activators can be selected from the group consisting of Smoothened agonist (SAG), SAG analog, SHH, C25-SHH, C24-SHH, purmorphamine, Hg — Ag and a derivative and/or variant thereof, wherein each derivative and/or variant thereof possesses one or more SMAD inhibitory activities.
  • the SHH signaling activator can comprise recombinant SHH and purmorphamine, or functional derivatives and/or variants thereof.
  • Recombinant SHH can be contacted with the pluripotent and/or multipotent cell at a final concentration in an in vitro culture of from about 5 ng/mL to about 5 pg/mL.
  • Purmorphamine can be contacted with the pluripotent and/or multipotent cell at a final concentration in an in vitro culture of from about 0.1 ⁇ M to about 20 ⁇ M.
  • the method comprises the use of the SSH activator SAG at a concentration of about 0.05-5 ⁇ M, preferably about 0.1 ⁇ M for a period of about 15 days to about 25 days, preferably about 21 or 22 days.
  • the difference in timing of SHH differentiation activation is crucial to trigger the generation of distinct ventral progenitors of divergent anterior-posterior identity.
  • Early activation of SHH signaling in the derivation of hESC-derived progenitors expressing markers of the hypothalamic strom requires the presence of FGF-8.
  • Neuronal precursor cells can be contacted with the one or more activator of SHH signaling after the passage of a predetermined period of time following generation of the neuronal precursor cell and/or following the contacting of the pluripotent and/or multipotent cell with the one or more inhibitors of SMAD and/or the one or more antagonists of Wnt signaling.
  • contacting the neuronal precursor cell with one or more activator of SHH signaling can be initiated from about 4 days to about 20 days or from about 8 days to about 18 days after contacting a pluripotent cell and/or a multipotent cell with one or more inhibitors of SMAD and with one or more antagonists of Wnt signaling.
  • Contacting the neuronal precursor cell with one or more activator of SHH signaling can be for a time period of from about 5 days to about 30 days or from about 8 days to about 16 days.
  • an "FGF receptor (FGFR) agonist” as used herein means a molecule that can activate FGFR (e.g. molecules that bind to FGFR and induce the dimerization of the receptor and activate the signaling P13K pathway and Ras/ERK pathway).
  • FGFR agonists include FGF2, FGF8 and SUN11602.
  • the FGFR agonist is FGF8 (e.g. recombinantly produced FGF8).
  • the cells are contacted with an FGFR agonist such as FGF8 in order to shift the balance towards rostralization.
  • the cells are contacted with FGF8 for a period of about 10 to about 20 days, preferably for a period of about 12 days to about 16 days, more preferably for about 13, 14, 15 or 16 days.
  • contact of the cells with an FGFR agonist such as FGF8 is initiated about 5 to about 10 days, preferably about 6 days, 7 days or 8 days, after initial contact of the cells with one or more SMAD inhibitors and/or after initial contact of the cells with a WNT inhibitor and/or after initial contact of the cells with an SHH activator.
  • the method comprises (i) LDN193189 (LDN) to inhibit BMP signaling (ii) SB-431542 (SB) to inhibit TGF ⁇ signaling (iii) recombinant FGF8 (iv) smoothened agonist (SAG; 3-chloro-N-[trans-4-(methylamino)cyclohexyl]-N-[3-(pyridin-4- yl)benzyl]-l-benzothiophene-2-carboxamide) to activate sonic hedgehog signaling (iv) IWP2 to inhibit WNT signaling and (v) FGF8 to shift the balance towards rostralization.
  • LDN193189 BMP signaling
  • SB-431542 SB-431542
  • TGF ⁇ signaling iii)
  • FGF8 recombinant FGF8
  • smoothened agonist SAG; 3-chloro-N-[trans-4-(methylamino)cyclohexyl]-N-[3-(pyr
  • MGE progenitor cells may be differentiated into cINs, preferably post-mitotic cINs, according to the methods described herein by contacting the cells with neurotrophic factors such as, without limitation, glial cell line derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF) for a predetermined amount of time.
  • neurotrophic factors such as, without limitation, glial cell line derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF) for a predetermined amount of time.
  • GDNF glial cell line derived neurotrophic factor
  • BDNF brain-derived neurotrophic factor
  • the predetermined amount of time is at least 10 days, at least 11 days, at least 12 days, at least 13 days, at least 14, days, at least 15 days, at least 16 days, at least 17 days, at least 18 days, at least 19 days, at least 20 days, at least 21 days, at least 22 days, at least 23 days, at least 24 days, at least 25 days, at least 26 days, at least 27 days, at least 28 days, at least 29 days, or at least 30 days.
  • marker refers to gene or protein that identifies a particular cell or cell type.
  • a marker for a cell may not be limited to one marker.
  • Markers may refer to a “pattern” of markers such that a designated group of markers may identify a cell or cell type from another cell or cell type.
  • Markers of cortical interneurons and/or of cortical interneuron precursor cells have been described and are readily available to those of skill in the art and include, for example, SST, PV, GABA, calbindin, LHX6, RAX, FOXA2, FOXG1, OLIG2, MASH1, NKX6.2, VGLUT1, MAP2, CTIP2, SATB2, TBR1, DLX2, ASCL1, and ChAT.
  • differentiation into a post-mitotic cIN precursor cell is identified by expression of one or more markers selected from FOXG1, PV, SST, Calbindin, DCX, ASCL1, TUJ1, GABA, GAD1, VGAT, VGLUT1, and GAD67.
  • a post-mitotic cIN precursor cell is identified by lack of expression of one or more makers selected from NKX2-1 and OLIG2 and optionally Ki67.
  • differentiation into a telencephalon cell is identified by expression of FoxGl and/or lack of expression of RAX.
  • differentiation into a ventral telencephalon cell is identified by expression of FoxGl and DLX2 and/or lack of expression of EMX1.
  • differentiation into an MGE progenitor cell is identified by expression of one or more markers selected from FOXG1, NKX2-1, NKX2-2, ASCL1, SIX6, OLIG2, NKX6.2, DLX1/2 and LHX6.
  • a post-mitotic cIN precursor cell is distinguished from an MGE progenitor cell by detecting expression of at least NKX2-1 and OLIG2, wherein an MGE progenitor cell is identified by expression of NKX2-1 and lack of expression of OLIG2 and a post-mitotic cIN precursor cell is identified by expression of OLIG2 and lack of expression of NKX2-1.
  • pluripotent cells and/or multipotent cells can be human cells or murine cells, which can be selected from the group consisting of embryonic stem cells, adult stem cells, neural stem cells, induced pluripotent cells, engineered pluripotent cells, primary progenitor cells, induced progenitor cells, and engineered progenitor cells.
  • Contacting with SMAD inhibitors and/or contacting with Wnt signaling antagonists can be carried out simultaneously or can be carried out sequentially. Contacting can be for a duration of from about 5 days to about 30 days.
  • Methods by the current disclosure can be used to produce cortical interneurons and their precursors, at an amount and purity that cannot be obtained by the state of the art.
  • a large number of pure, functional cortical interneurons obtained by using methods of the present disclosure in some embodiments, can be used to study seizures, schizophrenia or autism, and other neurological diseases. Similarly, these cells can also be used in cell therapy.
  • compositions comprising one or more in vitro differentiated neuronal cells that produce one or more markers of a cortical intemeuronal cell and/or of a cortical interneuron precursor cell, wherein the in vitro differentiated neuronal cells are produced by: (a) contacting a multipotent cell or a pluripotent cell encapsulated in a synthetic hydrogel with two or more inhibitors of SMAD signaling, (b) contacting the multipotent cell or a pluripotent cell with one or more inhibitors of Wnt signaling, and (c) contacting the multipotent cell or a pluripotent cell with one or more activators of SHH signaling.
  • Composition disclosed herein can comprise a mixture of two or more cells wherein cortical interneurons comprise at least about 30% of the total number of cells, or at least about 40% of the total number of cells, or at least about 50% of the total number of cells, or at least about 60% of the total number of cells, or at least about 70% of the total number of cells, or at least about 80% of the total number of cells, or at least about 90% of the total number of cells, or at least about 95% of the total number of cells.
  • Compositions can comprise a mixture of two or more cells wherein NKX2.1+/PV+ cortical interneurons comprise at least about 5%, or at least about 10%, or at least about 20%, or at least about 30%, or at least about 40%, at least about 50%, at least about 60%, at least about 70% or at least about 80% of the total number of cells in the composition.
  • Compositions can comprise a mixture of two or more cells wherein ⁇ -aminobutyric acid (GABA)-ergic inhibitory interneurons comprise at least about 5%, or at least about 10%, or at least about 20%, or at least about 30%, or at least about 40%, at least about 50%, at least about 60%, at least about 70% or at least about 80% of the total number of cortical interneurons in the composition.
  • GABA ⁇ -aminobutyric acid
  • Cortical interneuron or interneuron precursor cells can be modified with a transgene expressing a detectable marker such as, for example, CT-2 or green fluorescence protein (GFP). It will be understood that these detectable markers can be interchanged with other detectable markers without deviating from this aspect of the present disclosure.
  • the cortical interneuron precursor cells of the present invention give rise to functional interneurons, exhibiting the morphological, neurochemical, and electrophysiological properties of mature interneurons.
  • the immature interneuron precursor preparation can mature in vitro under controlled culture conditions mimicking their in vivo neuronal environment.
  • the immature interneuron precursor cells mature following transplantation and migration within the cerebral cortex of a mammalian subject (e.g., a human subject).
  • the immature interneuron precursor cells of the present invention can migrate extensively, in a non-radial (i.e., tangential) fashion upon transplantation into the cerebral cortex.
  • the cortical interneuron precursor cells can mature into parvalbumin and Kv3.1 expressing interneurons that exhibit fast spiking action potential discharge patterns.
  • the cortical interneuron precursor cells of the present invention can mature into somatostatin expressing interneurons, exhibiting the characteristic rebound, adapting, non-fast spiking firing patterns of this sub-group of interneurons. These somatostatin expressing interneurons may further express neuropeptide Y.
  • the cortical interneuron precursor cells can mature into interneurons having an average resting membrane potential of about - 40 mV to about -70 mV. Overtime, the average resting membrane potential becomes more hyperpolarized, ranging from about -55 mV to about -70 mV.
  • Conditions suitable for treatment in accordance with this method of the present invention include, without limitation seizure disorders, such as epilepsy or infantile spasms; neuropsychiatric disorders, such as autism, schizophrenia, an anxiety disorder, and an eating disorder; neurodevelopmental disorders, such as holoprosencephaly or microcephaly; and Parkinson's disease.
  • seizure disorders such as epilepsy or infantile spasms
  • neuropsychiatric disorders such as autism, schizophrenia, an anxiety disorder, and an eating disorder
  • neurodevelopmental disorders such as holoprosencephaly or microcephaly
  • Parkinson's disease Parkinson's disease.
  • hPSCs Early studies using hPSCs have been primarily geared towards neurodegenerative disorders, which are known to affect specific neuron types such as midbrain dopamine neurons in Parkinson's disease (PD; (Kriks et al., Nature 480:547-551 (2011); Soldner et al., Cell 136:964-977 (2009); and Soldner et al., Cell 146:318-331 (2011)) or motor neurons in amyotrophic lateral sclerosis (ALS; Dimos et al., Science 321:1218-1221 (2008)) and spinal muscular atrophy (SMA; Ebert et al., Nature 457:277-280 (2009)).
  • PD Parkinson's disease
  • ALS amyotrophic lateral sclerosis
  • SMA spinal muscular atrophy
  • the cells of the present disclosure can be delivered via intraparenchymal or intraventricular transplantation as described in U.S. Pat. Nos. 5,082,670 and 5,650,148 to Gage et al., and U.S. Patent Publication No. 20060141622 to Johe et al., which are hereby incorporated by reference in their entirety.
  • Intraparenchymal transplantation can be achieved by injecting the immature interneuron precursor cells within the host brain parenchyma or by preparing a cavity by surgical means to expose the host brain parenchyma and then depositing the cell graft into the cavity.
  • Both methods provide parenchymal apposition between the grafted cells and host brain tissue at the time of grafting, and both facilitate anatomical integration between the graft and host brain tissue.
  • the graft may be placed in a ventricle, e.g., a cerebral ventricle or subdurally, e.g., on the surface of the host brain where it is separated from the host brain parenchyma by the intervening pia mater or arachnoid and pia mater.
  • Grafting to the ventricle may be accomplished by injection of the donor cells or by growing the cells in a substrate such as 30% collagen to form a plug of solid tissue which may then be implanted into the ventricle to prevent dislocation of the graft.
  • the cells may be injected around the surface of the brain after making a slit in the dura. This is of importance if it is required that the graft become an integral part of the host brain and to survive for the life of the host.
  • transplantations of neuronal cells and precursors described by the present disclosure result in the successful grafting of large numbers of cells that can be studied after their maturation in vivo.
  • the present disclosure provides methods for the treatment of disorders and diseases that are associated with neurodegeneration including, for example, seizure disorders, Parkinson's disease (PD) and Alzheimer's disease (AD), which methods comprise the in vivo administration of cortical interneurons to a patient afflicted with a seizure disorder, PD or AD, which cortical interneurons are generated by the methods disclosed herein.
  • PD Parkinson's disease
  • AD Alzheimer's disease
  • the present disclosure provides methods for the treatment of psychiatric disorders and diseases including, for example, schizophrenia and the autism-related disorders.
  • inhibitory neurons such as cortical interneurons may have a particularly important role in schizophrenia or autism.
  • Bennett Nature 468:187-193 (2010) and Lewis et al., Nat Rev Neurosci 6:312-324 (2005).
  • the present disclosure provides purified populations of mature cortical interneurons that may be used as models for human psychiatric disorders and diseases and in therapeutic regimen for the treatment of such psychiatric disorders and diseases. Moreover, the data presented herein demonstrate that highly efficient derivation of cortical interneurons is possible following timed exposure to developmental cues.
  • hESC-derived GAB Aergic interneurons receive synaptic inputs from other human interneurons and from excitatory mouse projection neurons.
  • Cells exhibiting the neurochemical properties of cortical interneurons adopt fairly mature physiological properties within 30 days of plating on mouse cortex cultures. While the mechanisms of accelerated zTi vitro maturation of the NKX2.1 :GFP+ neurons on mouse cortical cultures are currently unknown, the involvement of species-specific timing factors derive from the data presented herein.
  • the presently disclosed data further demonstrate that synaptically active cortical interneurons can be derived in vitro and can be useful for modeling and treatment of cortical interneuron pathologies in psychiatric disorders including, but not limited to, schizophrenia and autism.
  • the present disclosure provides the generation of enriched cortical interneuron subgroups, such as somatostatin+ and PV+ cells.
  • MGE progenitors may be under control of SHH signaling, with high SHH signaling levels promoting the generation of somatostatin+ cells and lower SHH signaling levels promoting the generation of PV+ neurons.
  • Any methods known in the art for measuring gene expression may be used, in particular, quantitative methods such as, real time quantitative PCR or microarrays, or methods using gene reporter expression or qualitative methods such as immunostaining or cell sorting methods identifying cells exhibiting specific biomarkers, including cell surface markers.
  • thermoreversible polymer with improved stability over time comprising the formula (III): wherein a, b, c, d represent mole fractions of the polymer;
  • PEG n is a polyethylene glycol polymer and n is an integer from 1 to 2500;
  • R 1 if present, is any terminal group other than a primary amine
  • R 2 is a lower alkyl group
  • R 3 if present, is a functional group or linked modifying agent
  • G 1 and G 2 are each independently selected from a polymer segment, a terminal group, a tinker and a linked modifying agent.
  • Item 2 The thermoreversible polymer according to Item 1 , wherein the molecular weight (MW) of the polymer is from about 0 to about 250 kilodaltons (kDa) or about 50 to about 200 kDa.
  • Item 3 The thermoreversible polymer of Item 1 or 2, wherein R 1 is a C 1 -C 6 alkoxy selected from methoxy, ethoxy, n-propoxy, n-butoxy, isobutoxy, tert-butoxy, pentoxy and isopentoxy.
  • Item 4 The thermoreversible polymer of Item 3, wherein R 1 is methoxy.
  • Item 5 The thermoreversible polymer of any one of Items 1 to 4, wherein R 2 is selected from the group consisting of methyl, ethyl, propyl, butyl, pentyl, isopropyl, isobutyl, isopentyl, tert-butyl, cyclopropyl, and cyclobutyl.
  • Item 6 The thermoreversible polymer of Item 5, wherein R 2 is butyl.
  • Item 7 The thermoreversible polymer of any one of Items 1 to 6, wherein R 3 is not present.
  • Item 8 The thermoreversible polymer of Item 1, wherein R 1 is methoxy, R 2 is butyl, and R 3 is not present and wherein the MW of the polymer is from about 0 to about 250 kDa.
  • Item 9 The thermoreversible polymer of any one of Items 1 to 8, wherein the MW of PEGn is from about 1 to about 50 kilodaltons (kDa).
  • Item 10 The thermoreversible polymer of any one of Items 1 to 9, wherein R 3 is a chemoselective functional group selected from a thiol, an alkyne, a cyclooctyne, an azide, a phosphine, a maleimide, an alkoxyamine, an aldehyde and protected versions or precursors thereof.
  • R 3 is a chemoselective functional group selected from a thiol, an alkyne, a cyclooctyne, an azide, a phosphine, a maleimide, an alkoxyamine, an aldehyde and protected versions or precursors thereof.
  • Item 11 The thermoreversible polymer of any one of Items 1 to 9, wherein R 3 is a modifying agent selected from a heparin, a hyaluronic acid, a specific binding member, a peptide, a nucleic acid, gelatin, fibronectin, collagen, laminin, basic fibroblast growth factor (bFGF), epidermal growth factor (EGF), insulin, progesterone, glucose, stromal cell derived factor- 1 (SDF-1), thymosin beta-4, sonic hedgehog (SHH), Noggin, Activin, transforming growth factor- ⁇ (TGF- ⁇ ), FGF8, brain-derived neurotrophic factor (BDNF), glial cell-derived neurotrophic factor (GDNF), neurotrophic factor-3 (NT3), platelet-derived growth factor (PDGF), IL-16, IL-2 and insulin-like growth factor- 1 (IGF-1).
  • R 3 is a modifying agent selected from a heparin, a hyaluronic
  • thermoreversible polymer of any one of Items 1 to 11, wherein the thermoreversible polymer has one or more of the following properties: (a) an LCST of about 12- 32 °C, preferably of about 19-23 °C; (b) a stiffness of about 100 to 8000 Pa, preferably of about 100 to 3000 Pa; and a (c) a viscosity of about 100-1500 cP.
  • Item 13 A three-dimensional hydrogel comprising a thermoreversible polymer of any one of Items 1-12, and a buffered aqueous solution.
  • Item 14 An in vitro method for producing a cell population enriched for MGE progenitor cells from an initial population of human stem cells, the method comprising:
  • Item 15 The method of Item 14, wherein said human stem cells are selected from the group consisting of human embryonic stem cells, human adult stem cells, human neural stem cells, human induced pluripotent cells, human primary progenitor cells, and human induced progenitor cells.
  • Item 16 The method of Item 14, wherein said contacting with the at least one inhibitor of SMAD signaling and said contacting with the at least one Wnt antagonist are carried out simultaneously or sequentially, and each has a duration between about 5 days and about 30 days.
  • Item 17 The method of Item 16, wherein the contact of the cells with the at least one Wnt antagonist is initiated within 5 days, preferably within 4 days, 3 days, 2 days, or 1 day, from the initial contact of the cells with the at least one inhibitor of SMAD signaling, preferably wherein the contact of the cells with the at least one Wnt antagonist is initiated simultaneously with the initial contact of the cells with the at least one inhibitor of SMAD signaling.
  • Item 18 The method of Item 14, wherein said at least one inhibitor of SMAD signaling is selected from the group consisting of SB431542, LDN-193189, Noggin PD169316, SB203580, LY364947, A77-01, A-83-01, BMP4, GW788388, GW6604, SB-505124, lerdelimumab, metelimumab, GC-I008, AP-12009, AP-11014, LY550410, LY580276, LY364947, LY2109761, SB-505124, E-616452 (RepSox ALK inhibitor), SD-208, SMI6, NPC- 30345, KI26894, SB-203580, SD-093, activin-M108A, P144, soluble TBR2-Fc, DMH-1, Dorsomorphin dihydrochloride, derivatives thereof, and combinations thereof. .
  • Item 19 The method of Item 18, wherein the at least one inhibitor of SMAD signaling comprises SB431542 and LDN-193189.
  • Item 20 The method of Item 14, wherein the at least one Wnt antagonist is selected from the group consisting of XAV939, DKK1, DKK-2, DKK-3, Dkk-4, SFRP-1, SFRP-2, SFRP-5, SFRP-3, SFRP-4, WIF-1, Soggy, IWP-2, IWR1, ICG-001, KY0211, Wnt-059, LGK974, IWP-L6, derivatives thereof, and combinations thereof, preferably wherein the at least one Wnt antagonist comprises IWP-2.
  • the at least one Wnt antagonist comprises IWP-2.
  • Item 21 The method of Item 14, wherein said at least one activator of SHH signaling is selected from the group consisting of Smoothened agonist (SAG), SAG analog, SHH, C25- SHH, C24-SHH, purmorphamine, Hg — Ag, derivatives thereof, and combinations thereof.
  • SAG Smoothened agonist
  • SHH SAG analog
  • SHH C25- SHH
  • C24-SHH purmorphamine
  • Hg — Ag Hg — Ag
  • derivatives thereof derivatives thereof, and combinations thereof.
  • Item 22 The method of Item 21, wherein (i) the contact of the cells with the at least one activator of SHH signaling is concluded from about 5 days to about 30 days from its initiation, preferably about 18 to 23 days from its initiation, more preferably about 19 to 22 days from its initiation, even more preferably about 20 or 21 days from its initiation; (ii) the initial contact of the cells with the at least one activator of SHH signaling is between about 0 days and about 10 days, preferably is about 0 days, from the initial contact of the cells with the at least one inhibitor of SMAD signaling and from the initial contact of the cells with the at least one inhibitor of WNT signaling; (iii) the initial contact of the cells with the at least one inhibitor of SMAD signaling is between 0 to 4 days from the initial contact of the cells with the at least one Wnt antagonist; (iv) the contact of the cells with the at least one inhibitor of SMAD signaling is concluded between 6 and 14 days from its initiation and/or (v)
  • Item 23 The method of Item 14, wherein said at least one additional marker is selected from the group consisting of NKX2-1, NKX2-2, ASCL1, SIX6, OLIG2, NKX6.2, DLX1/2 and LXH6.
  • Item 24 The method of Item 14, wherein at least about 60%, at least 65%, at least 70%, at least 75%, or at least 80% of the obtained cell population expresses FOXG1 and NKX2- 1.
  • Item 25 The method of Item 14, wherein at least about 60%, at least about 65%, at least about 70%, at least about 75%, or at least about 80% of the obtained cell population comprises MGE progenitor cells.
  • Item 26 The method of Item 14, further comprising (c) contacting the cells, after a predetermined amount of time, with at least one neurotrophic factor (e.g., GDNF, BDNF) and optionally a notch inhibitor (e.g., DAPT) to produce a cell population enriched for differentiated inhibitory GABAergic cortical interneurons (cINs) expressing FOXG1 and at least one additional marker indicating a cIN cell.
  • at least one neurotrophic factor e.g., GDNF, BDNF
  • a notch inhibitor e.g., DAPT
  • Item 27 The method of Item 26, comprising contacting the cells with at least one neurotrophic factor and a notch inhibitor.
  • Item 28 The method of Item 27, comprising contacting the cells with GDNF, BDNF and DAPT.
  • Item 29 The method of any one of Items 26-28, wherein contacting the cells with at least one neurotropic factor and optionally a notch inhibitor occurs upon conclusion of contacting the cells with the at least one inhibitor of SMAD signaling and at least one Wnt antagonist and at least one activator of SHH signaling.
  • Item 30 The method of any one of Items 26-29, wherein contacting the cells with at least one neurotropic factor and optionally a notch inhibitor is concluded between 7 days and 30 days after initiation and/or contacting the cells with at least one neurotropic factor and optionally a notch inhibitor is concluded at least about 10, at least about 12, or at least about 14 days after initiation.
  • Item 31 The method of Item 26, wherein said at least one additional marker is selected from the group consisting of PV, SST, Calbindin, DCX, ASCL1, TUJ1, GABA, GAD1, VGAT, vGLUT1, and GAD67.
  • Item 32 The method of any one of Items 26-31 , wherein at least about 50%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, or at least about 80% of the obtained cIN cell population expresses parvalbumin (PV).
  • PV parvalbumin
  • Item 33 The method of any one of Items 26-32 wherein less than about 5% of the obtained cIN cell population expresses Ki67,
  • Item 34 A composition comprising a population of cells produced by the method of any one of Items 14-26, wherein at least 50% of the cells, or at least 60% of the cells, or at least 70% of the cells, or at least 80% of the cells, or at least 90% of the cells, or at least 95% of the cells are MGE progenitor cells, preferably wherein the method does not comprise a step to purify or further enrich MGE progenitor cells following step (b).
  • Item 35 A composition comprising a population of cells produced by the method of any one of Items 17-33, wherein at least 50% of the cells, or at least 60% of the cells, or at least 70% of the cells, or at least 80% of the cells, or at least 90% of the cells, or at least 95% of the cells are cINs, preferably wherein the method does not comprise a step to purify or further enrich cIN cells following step (c).
  • Item 36 The composition of Item 35, wherein at least 50% of the cells, or at least 60% of the cells, or at least 70% of the cells, or at least 80% of the cells, or at least 90% of the cells, or at least 95% of the cells are NKX2.1-/PV + .
  • Item 37 The use of a composition according to any one of Items 34-36 in the treatment of a neurological disorder.
  • Item 38 The use according to Item 37, wherein the neurological disorder is a seizure disorder.
  • Item 39 An in vitro method for producing a cell population enriched for MGE progenitor cells from an initial population of human stem cells, the method comprising:
  • Item 40 The method of Item 39, wherein said human stem cells are selected from the group consisting of human embryonic stem cells, human adult stem cells, human neural stem cells, human induced pluripotent cells, human primary progenitor cells, and human induced progenitor cells.
  • Item 41 The method of Item 39, wherein said contacting with the at least one inhibitor of SMAD signaling and said contacting with the at least one Wnt antagonist are carried out simultaneously or sequentially, and each has a duration between about 5 days and about 30 days.
  • Item 42 The method of Item 41 , wherein the contact of the cells with the at least one Wnt antagonist is initiated within 5 days, preferably within 4 days, 3 days, 2 days, or 1 day, from the initial contact of the cells with the at least one inhibitor of SMAD signaling, preferably wherein the contact of the cells with the at least one Wnt antagonist is initiated simultaneously with the initial contact of the cells with the at least one inhibitor of SMAD signaling.
  • Item 43 The method of Item 39, wherein said at least one inhibitor of SMAD signaling is selected from the group consisting of SB431542, LDN-193189, Noggin PD169316, SB203580, LY364947, A77-01, A-83-01, BMP4, GW788388, GW6604, SB-505124, lerdelimumab, metelimumab, GC-I008, AP-12009, AP-110I4, LY550410, LY580276,
  • Item 44 The method of Item 43 , wherein the at least one inhibitor of SMAD signaling comprises SB431542 and LDN-193189.
  • Item 45 The method of Item 39, wherein the at least one Wnt antagonist is selected from the group consisting of XAV939, DKK1, DKK-2, DKK-3, Dkk-4, SFRP-1, SFRP-2, SFRP-5, SFRP-3, SFRP-4, WIF-1, Soggy, IWP-2, IWR1, ICG-001, KY0211, Wnt-059, LGK974, IWP-L6, derivatives thereof, and combinations thereof, preferably wherein the at least one Wnt antagonist comprises IWP-2.
  • Item 46 The method of Item 39, wherein said at least one activator of SHH signaling is selected from the group consisting of Smoothened agonist (SAG), SAG analog, SHH, C25- SHH, C24-SHH, purmorphamine, Hg — Ag, derivatives thereof, and combinations thereof.
  • SAG Smoothened agonist
  • SHH SAG analog
  • SHH C25- SHH
  • C24-SHH purmorphamine
  • Hg — Ag Hg — Ag
  • derivatives thereof derivatives thereof, and combinations thereof.
  • Item 47 The method of Item 46, wherein (i) the contact of the cells with the at least one activator of SHH signaling is concluded from about 5 days to about 30 days from its initiation, preferably about 18 to 23 days from its initiation, more preferably about 19 to 22 days from its initiation, even more preferably about 20 or 21 days from its initiation; (ii) the initial contact of the cells with the at least one activator of SHH signaling is between about 0 days and about 10 days, preferably is about 0 days, from the initial contact of the cells with the at least one inhibitor of SMAD signaling and from the initial contact of the cells with the at least one inhibitor of WNT signaling; (iii) the initial contact of the cells with the at least one inhibitor of SMAD signaling is between 0 to 4 days from the initial contact of the cells with the at least one Wnt antagonist; (iv) the contact of the cells with the at least one inhibitor of SMAD signaling is concluded between 6 and 14 days from its initiation and/or (v
  • Item 48 The method of Item 39, wherein said at least one additional marker is selected from the group consisting of NKX2-1, NKX2-2, ASCL1, SIX6, OLIG2, NKX6.2, DLX1/2 and LXH6.
  • Item 49 The method of Item 39, wherein at least about 60%, at least 65%, at least 70%, at least 75%, or at least 80% of the obtained cell population expresses FOXG1 and NKX2- 1.
  • Item 50 The method of Item 39, wherein at least about 60%, at least about 65%, at least about 70%, at least about 75%, or at least about 80% of the obtained cell population comprises MGE progenitor cells.
  • Item 51 The method of Item 39, further comprising (c) contacting the cells, after a predetermined amount of time, with at least one neurotrophic factor (e.g., GDNF, BDNF) and optionally a notch inhibitor (e.g., DAFT) to produce a cell population enriched for differentiated inhibitory GABAergic cortical interneurons (cINs) expressing FOXG1 and at least one additional marker indicating a cIN cell.
  • at least one neurotrophic factor e.g., GDNF, BDNF
  • a notch inhibitor e.g., DAFT
  • Item 52 The method of Item 51, comprising contacting the cells with at least one neurotrophic factor and a notch inhibitor.
  • Item 53 The method of Item 52, comprising contacting the cells with GDNF, BDNF and DAFT.
  • Item 54 The method of any one of Items 51-53, wherein contacting the cells with at least one neurotropic factor and optionally a notch inhibitor occurs upon conclusion of contacting the cells with the at least one inhibitor of SMAD signaling and at least one Wnt antagonist and at least one activator of SHH signaling.
  • Item 55 The method of any one of Items 51 -54, wherein contacting the cells with at least one neurotropic factor and optionally a notch inhibitor is concluded between 7 days and 30 days after initiation and/or contacting the cells with at least one neurotropic factor and optionally a notch inhibitor is concluded at least about 10, at least about 12, or at least about 14 days after initiation.
  • Item 56 The method of Item 51 , wherein said at least one additional marker is selected from the group consisting of PV, SST, Calbindin, DCX, ASCL1, TUJ1, GABA, GAD1, VGAT, vGLUTl, and GAD67.
  • Item 57 The method of any one of Items 51-56, wherein at least about 50%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, or at least about 80% of the obtained cIN cell population expresses parvalbumin (PV).
  • PV parvalbumin
  • Item 58 The method of any one of Items 51-57 wherein less than about 5% of the obtained cIN cell population expresses KI67.
  • Item 60 A composition comprising a population of cells produced by the method of any one of Items 51-58, wherein at least 50% of the cells, or at least 60% of the cells, or at least 70% of the cells, or at least 80% of the cells, or at least 90% of the cells, or at least 95% of the cells are cINs, preferably wherein the method does not comprise a step to purify or further enrich cIN cells following step (c).
  • Item 61 The composition of Item 60, wherein at least 50% of the cells, or at least 60% of the cells, or at least 70% of the cells, or at least 80% of the cells, or at least 90% of the cells, or at least 95% of the cells are NKX2.1-/PV + .
  • Item 62 The use of a composition according to any one of Items 59-61 in the treatment of a neurological disorder.
  • Item 63 The use according to Item 62, wherein the neurological disorder is a seizure disorder.
  • Hydrogels disclosed in U.S. Patent Publication No. 2024/0294713 comprising acrylamide polymers of the following structure were initially tested for, inter alia, stability during the duration of cIN production from human stem cells:
  • hydrogels comprising such acrylamide polymers because the hydrogel was not stable over the full 35-day differentiation period, at any molecular weight tested (from 0 to 500 kDa).
  • These hydrogels also did not have a suitable peak stiffness range for stability ( ⁇ 1200 Pa), exhibited high viscosity (>2000 cP), had low stability/high swell ratio in aqueous environments(>1.5), and high LCSTs (>25).
  • These gels did not hold structure in aqueous environments over time, and did not hold structure with the shear stress of mixing in spinner flasks or bioreactors. Finally, these gels did not extrude and encapsulate cells well, with low sphericity ( ⁇ 0.7), large bead diameters (>4mm), and large fraction of tail formation (>0.5).
  • Additional improvements when the weight ratio of PEG to PNIPAAm is above 1 :2 include improvements to encapsulation and cell performance, such as faster flow rates with lower shear stress, higher immediate cell viability, higher 7 day cell viability and higher cell yield. See Figure 5C.
  • Polymers of formula III as described herein comprise a combination of features rendering them uniquely useful for encapsulating and suspending cells in three-dimensional hydrogel bioreactors for scale-up production in tanks for lengthy cell culture/differentiation protocols such as production of functional neurons from stem cells.
  • the present hydrogels (comprising encapsulated differentiated cells such as neurons) can be used for therapeutic purposes in humans.
  • the present systems maintain the viability of cells (e.g., stem cells) and avoid harsh methods of recovery of the cells from the system.
  • the pendant PEG group (at position R 1 ) was found to significantly affect synthesis of the polymer.
  • groups at position R 1 other than primary amines enables higher reaction v/v%, higher yield per volume of solvent, and more reproducible synthesis.
  • Fig. 10A Producing an amine terminated R 1 requires the use of a bifunctional diaminoPEG monomer in the synthesis, which has the potential for unintended covalent crosslinking. This can greatly affect polymer performance, reproducibility, and viscosity, and may prevent the polymer from reliquifying.
  • reaction 1 the solvent choice for reaction 1 was found to be critical for successful polymer synthesis and incorporation of each monomer at the intended molar ratio and for precise control of the molecular weights.
  • Certain solvents with the ideal solubility properties for all monomers exhibit high reaction efficiencies with complete incorporation (A), while some solvents only polymerize monomers that are soluble, not all monomers (B), and some solvents inhibit complete polymerization of the reaction (C-D), and some polymers are not suitable for any reaction (E-F).
  • Solvent tested ranged from polar protic solvents, polar aprotic solvents, and nonpolar solvents in standard and anhydrous forms in various structural conformations.
  • Tested solvents include Acetone, Acetonitrile, Benzene, Chloroform, Dichloromethane, Dimethylformamide, Dimethyl sulfoxide, Dioxane, Ethyl acetate, Pyridine, Ethanol, Methanol, Tetrahydrofuran, Toluene, and Water. See Fig. 10B
  • Example 3 [0274] Proof-of-concept studies were performed utilizing a low molecular weight, acrylate backbone 3D hydrogel to demonstrate robust derivation of encapsulated hPSCs into post-mitotic cINs with therapeutic potential to treat neurological disorders such as epilepsy.
  • thermoreversible graft copolymer ( Figure 1), where the PEG represents the hydrophilic block, the PNIPAAm represents the hydrophobic block, and the alkyl pendant group (here described as butyl chains but could encompass any alkyl chain) serves as the temperature shifting moiety.
  • NIPAAm N-acryloxysuccinimide
  • NMI N-acryloxysuccinimide
  • alkyl-chain methacrylate was first copolymerized via standard radical polymerization.
  • the resulting functionalizable copolymer after reprecipitation and drying, was then mixed with a monoamine-terminated PEG block.
  • the amine-terminated group attached to the PNIPAAm-co-PNASI-co-MA backbone via the amine and N-hydroxysuccinimide (NHS) amidation reaction.
  • NHS N-hydroxysuccinimide
  • Human PSC cells H9 human embryonic stem cells (WA09, WiCell, Madison, WI, passage 53-58)
  • Matrigel BD, San Jose, CA
  • E8 media Gibco, Billings, MT
  • Versene Thermo Fisher, Waltham, MA
  • hPSCs were dissociated with Accutase (Stem Cell Technologies, Vancouver, BC, Canada) and encapsulated using PEG-PNIPAAM hydrogel at a 10%wt/v final, using a concentration of 250,000 cells/ml of gel. Hydrogel and cells were mixed on ice and 50 ul droplets were subsequently generated and extruded into well plates as shown in Figure 13b, for a total of 5 gel droplets per well. Plates were incubated at 37°C for 15 min to allow for gelification, forming dome-shaped beads.
  • LDN193189 100 nM, Stemgent, Cambridge, MA
  • SB431542 10 ⁇ M, Tocris, Minneapolis, MN
  • IWP2 5 ⁇ M, Selleck Chem
  • SAG 0.1 ⁇ M, XcessBio, Chicago, IL
  • FGF8 lOOng/ml, Peprotech
  • DMEM F/12 Stem Cell Technologies
  • lOng/ml GDNF R&D Systems Minneapolis, MN
  • lOng/ml BDNF R&D
  • 2.5 ⁇ M DAPT Tocris
  • Imaging was performed using a Cytation 5 imaging system (Agilent) and images were analyzed using FIJI image analysis software (Schindelin, J. et al. Fiji: An open-source platform for biological-image analysis. Nature Methods vol. 9 Preprint at https://doi.org/10.1038/nmeth.2019 (2012)).
  • RT2 First Strand kit Qiagen
  • real-time PCR analyses were performed using the RT2 SYBR Green qPCR Mastermixes (Qiagen) and an AriaMX Real Time PCR System (Agilent). Primers were designed using the Integrated DNA Technologies PrimerQuest tools (Coralville, IA).
  • the copolymer hydrogel utilized herein is based on hydrophilic poly(ethylene glycol) (PEG) and temperature-sensitive poly(N-isopropylacrylamide) (PNIPAAm).
  • PEG poly(ethylene glycol)
  • PNIPAAm temperature-sensitive poly(N-isopropylacrylamide)
  • LCST lower critical solution temperature
  • the PNIPAAm component becomes increasingly hydrophobic and enables micelle formation, essentially physical “crosslinking” the PEG- PNIPAAm polymer ( Figure 13a).
  • This hydrogel can be used to encapsulate and differentiate hPSCs in scale down (well plate) models as well as in stirring cultures including perfused stirred- tank bioreactors, by simply extruding hydrogel droplets containing single cells or small cell clusters into warm culture media, leading to the formation of gel capsules containing these cells ( Figures 13b and c).
  • This encapsulation method allows for a fully defined, synthetic, xeno-free and scalable platform for manufacturing of clinically relevant cell types.
  • a ROCK inhibitor permits survival of dissociated human embryonic stem cells. Nat Biotechnol 25, (2007)).
  • the ALK2/3 inhibitor LDN193189 and ALK5/7 inhibitor SB431542 were used, as described previously (Kim, T. G. et al. Efficient specification of interneurons from human pluripotent stem cells by dorsoventral and rostrocaudal modulation. Stem Cells 32, (2014)).
  • a chemical inhibitor of Wnt pathway, IWP2 was used to promote rostralization of early neuroectoderm and subsequently to inhibit dorsalization of neuroectoderm (Kim, T. G., 2014).
  • SHH Sonic Hedgehog pathway
  • hPSC-derived forebrain interneurons requires an extended timeline and mimics human neural development.
  • Thermoreversible hydrogel culture platform renders higher viability and differentiation efficacy of hPSCs into GABAergic cortical interneurons compared to standard two-dimensional culture methods
  • hPSCs can be robustly differentiated into MGE progenitor cells and cINs when encapsulated in three-dimensional hydrogels, rendering over 80% expression of MGE progenitor marker FoxGl and over 75% expression of Nkx2-1 by day 18, followed by over 75% FoxGl expression by day 35 with high percentage of cells expressing cIN markers PV, SST, GAD1 and GABA, together with a strong downregulation in Nkx2-1 expression to only ⁇ 22%. This, together with nearly absent ki67 expression, indicates that a 3D thermoreversible hydrogel platform allows for efficient generation of mature post-mitotic cINs.
  • the data present herein demonstrates robust differentiation of cINs from hPSCs in 3D synthetic hydrogels such as the novel hydrogel of formula (III), with substantially higher viability and differentiation efficacy compared to standard 2D cultures, as well as accelerated differentiation period compared to the maturation timeframe reported using standard 2D cultures or hybrid 2D-suspension systems.
  • thermoreversible graft copolymer ( Figure 1), where the PEG represents the hydrophilic block, the PNIPAAm represents the hydrophobic block, and the alkyl pendant group (here described as butyl chains but could encompass any alkyl chain) serves as the temperature shifting moiety.
  • NIPAAm N-acryloxysuccinimide
  • NMI N-acryloxysuccinimide
  • alkyl-chain methacrylate was first copoiymerized via standard radical polymerization.
  • the resulting functionalizable copolymer after reprecipitation and drying, was then mixed with a monoamine-terminated PEG block.
  • the amine-terminated group attached to the PNIPAAm-co-PNASI-co-MA backbone via the amine and N-hydroxysuccinimide (NHS) amidation reaction.
  • NHS N-hydroxysuccinimide
  • Human PSC cells H9 human embryonic stem cells (WA09, WiCell, Madison, WI, passage 53-58)
  • Matrigel BD, San Jose, CA
  • E8 media Gibco, Billings, MT
  • Versene Thermo Fisher, Waltham, MA
  • ReleSR Stem Cell Technologies, Vancouver, BC, Canada
  • hPSCs were dissociated with Accutase (Stem Cell Technologies, Vancouver, BC, Canada) and encapsulated using PEG-NIP AAM hydrogel at 2.5-10%wt/v final, using a concentration of 250,000 cells/ml of gel. Hydrogel and cells were mixed , encapsulated and extruded into vessels of different sizes (lOOmL to IL). Complete E8 media heated to 37°C supplemented with Rock Inhibitor Y-27632 (Selleck Chemicals, Houston, TX) was subsequently added and cells were incubated at 37°C with 5% CO2. At the end of the expansion process (7 days) aggregates were dissociated to single cells in the presence of Accumax (Innovative Cell Technologies, San Diego, CA) and TrypLE (Thermo Fisher) in a rotating platform.
  • Accumax Innovative Cell Technologies, San Diego, CA
  • TrypLE TrypLE
  • RT2 First Strand kit Qiagen
  • real-time PCR analyses were performed using the RT2 SYBR Green qPCR Mastermixes (Qiagen) and an AriaMX Real Time PCR System (Agilent). Primers were designed using the Integrated DNA Technologies PrimerQuest tools (Coralville, IA).
  • This gel formulation allowed for the generation of beads comprised of core and shell geometries that allow for maximal cell retention upon scale up (Fig 5D). 3 different culture scales are used to quickly iterate through different conditions in scale down models that closely predict the performance in bioreactors (positive displacement pipettes - PDP - static and spinners, Fig 17A). Utilizing this improved formulation, we demonstrated high viability upon encapsulation. High-viability throughout the encapsulation process allowed for scalable hPSC expansion, with encapsulation flow rates of a minimum of 2 ml gel/min.
  • Example expansion processes utilizing this formulation are shown on Fig 18B, showing reproducible, high-quality, hPSC expansion across 4 different bioreactor scales (Fig 18A), from 100mL to IL vessel volumes, as validated by flow cytometry (Fig 18B) and qPCR (18C).
  • Fig 18A human embryonic stem cell line
  • Fig 18B human induced pluripotent stem cell line
  • 17B human induced pluripotent stem cell
  • Human PSC cells H9 human embryonic stem cells (WA09, WiCell, Madison, WI, passage 53-58)
  • Matrigel BD, San Jose, CA
  • E8 media Gibco, Billings, MT
  • Versene Thermo Fisher, Waltham, MA
  • ReleSR Stem Cell Technologies, Vancouver, BC, Canada
  • hPSCs were dissociated with Accutase (Stem Cell Technologies, Vancouver, BC, Canada) and encapsulated using PEG-PNIPAAM hydrogel at a 2.5-I0%wt/v final, using a concentration of 250,000 cells/ml of gel. Hydrogel and cells were mixed , encapsulated and extruded into lOOmL spinners. Spinners were incubated at 37°C for 15 min to allow for gelification. Complete E8 media heated to 37°C supplemented with Rock Inhibitor Y- 27632 (Selleck Chemicals, Houston, TX) was subsequently added and cells were incubated at 37°C with 5% CO2.
  • hydrogels comprising a thermoreversible polymer of formula III has broad implications in clinical manufacturing as aggregates surpassing this size can exhibit necrotic cores due to nutrient limitation and/or suboptimal differentiation efficacy due to heterogeneous response to differentiation cues throughout the aggregate.
  • Human PSC cells H9 human embryonic stem cells (WA09, WiCell, Madison, WI, passage 53-58)
  • Matrigel BD, San Jose, CA
  • E8 media Gibco, Billings, MT
  • Versene Thermo Fisher, Waltham, MA
  • ReieSR Stem Cell Technologies, Vancouver, BC, Canada
  • hPSCs were dissociated with Accutase (Stem Cell Technologies, Vancouver, BC, Canada) and encapsulated using PEG-PNIPAAM hydrogel at a 2.5-I0%wt/v final, using a concentration of 500,000 cells/ml of gel. Hydrogel and cells were mixed and plated with positive displacement pipettes (“PDP”) in plates or encapsulated and extruded into lOOmL spinners. Plates were incubated at 37°C for 15 min to allow for gelification, forming domeshaped beads.
  • PDP positive displacement pipettes
  • Human PSC cells H9 human embryonic stem cells (WA09, WiCell, Madison, WI, passage 53-58)
  • Matrigel BD, San Jose, CA
  • E8 media Gibco, Billings, MT
  • Versene Thermo Fisher, Waltham, MA
  • ReleSR Stem Cell Technologies, Vancouver, BC, Canada
  • HPSCs were dissociated with Accutase (Stem Cell Technologies, Vancouver, BC, Canada) and encapsulated using PEG-PNIPAAM hydrogel at a 2.5-10%wt/v final, using a concentration of 500,000 cells/ml of gel. Hydrogel and cells were mixed and plated with positive displacement pipettees (“PDF”) in plates. Plates were incubated at 37°C for 15 min to allow for gelification, forming dome-shaped beads. Complete E8 media heated to 37°C supplemented with Rock Inhibitor Y-27632 (Selleck Chemicals, Houston, TX) was subsequently added and cells were incubated at 37°C with 5% CO2.
  • PDF positive displacement pipettees
  • hPSCs were differentiated into HSCs utilizing the improved 3D hydrogel system.
  • human cord blood CD34+ HSCs were also successfully expanded in this 3D hydrogel system (Fig 23B and C). This data indicates that the improved hydrogel formulation is compatible with HSC expansion and differentiation from hPSCs, with high gel stability throughout lengthy differentiation processes, protecting cells from shear stress produced by bioreactor mixing upon scale up.
  • HSCs are a promising cell therapy candidate with broad therapeutic applications from sickle cell disease and b thalassemia to blood cancers.

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biomedical Technology (AREA)
  • Cell Biology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Reproductive Health (AREA)
  • Organic Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Neurology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Neurosurgery (AREA)
  • Medicinal Chemistry (AREA)
  • Virology (AREA)
  • Microbiology (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Gynecology & Obstetrics (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

L'invention concerne des polymères thermoréversibles ayant une stabilité améliorée, des compositions d'hydrogel comprenant les polymères thermoréversibles ainsi qu'un procédé in vitro pour produire une population de cellules enrichie pour des interneurones corticaux GABAergiques inhibiteurs issus de cellules pluripotentes humaines utilisant les compositions d'hydrogel.
PCT/US2024/054259 2023-11-03 2024-11-01 Polymères thermoréversibles à stabilité améliorée et procédés et utilisations associés Pending WO2025097051A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2024343386A AU2024343386B2 (en) 2023-11-03 2024-11-01 Thermoreversible polymers with improved stability and methods and uses thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202363595847P 2023-11-03 2023-11-03
US202363595841P 2023-11-03 2023-11-03
US63/595,841 2023-11-03
US63/595,847 2023-11-03

Publications (1)

Publication Number Publication Date
WO2025097051A1 true WO2025097051A1 (fr) 2025-05-08

Family

ID=93566372

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2024/054259 Pending WO2025097051A1 (fr) 2023-11-03 2024-11-01 Polymères thermoréversibles à stabilité améliorée et procédés et utilisations associés

Country Status (1)

Country Link
WO (1) WO2025097051A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN120789281A (zh) * 2025-09-12 2025-10-17 温州医科大学附属第一医院 一种桑色素纳米颗粒、pH响应性水凝胶及应用

Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5082670A (en) 1988-12-15 1992-01-21 The Regents Of The University Of California Method of grafting genetically modified cells to treat defects, disease or damage or the central nervous system
US5650148A (en) 1988-12-15 1997-07-22 The Regents Of The University Of California Method of grafting genetically modified cells to treat defects, disease or damage of the central nervous system
US6897064B2 (en) 2001-07-13 2005-05-24 Mebiol Inc. Cell or tissue-culturing carrier, and culturing method
WO2006019366A1 (fr) 2003-03-28 2006-02-23 Wisconsin Alumni Research Foundation Conditions physicochimiques de culture pour des cellules souches embryonnaires
WO2006029198A2 (fr) 2004-09-08 2006-03-16 Wisconsin Alumni Research Foundation Culture de cellules souches embryonnaires humaines
WO2006029297A2 (fr) 2004-09-10 2006-03-16 Hoftberg Steven Systeme d'etablissement de priorites theoriques des jeux pour reseau ad hoc mobiles permettant une deference hierarchique
US20060141622A1 (en) 2004-11-17 2006-06-29 Johe Karl K Transplantation of human neural cells for treatment of neurodegenerative conditions
WO2008006583A1 (fr) 2006-07-14 2008-01-17 Novartis Ag Dérivés de la pyrimidine en tant qu'inhibiteurs d'alk-5
WO2008089351A1 (fr) 2007-01-17 2008-07-24 Wisconsin Alumni Research Foundation Culture améliorée de cellules souches
WO2008094597A2 (fr) 2007-01-30 2008-08-07 University Of Georgia Research Foundation, Inc. Cellules mésodermiques précoces, une population stable de cellules mésendodermiques qui a une utilité pour la génération de lignées endodermiques et mésodermiques et de cellules migratoires multipotentes (mmc)
EP1992360A1 (fr) 2006-02-01 2008-11-19 The University of Tokyo Utilisation en association d'un inhibiteur de signal tgf-beta et d'un agent antitumeur
WO2009131166A1 (fr) 2008-04-25 2009-10-29 国立大学法人 千葉大学 Inhibiteur de la signalisation de wnt comprenant une protéine de liaison au facteur de croissance de type insuline
WO2009155001A2 (fr) 2008-05-27 2009-12-23 The Board Of Regents Of The University Of Texas System Inhibiteurs de signalisation de la protéine wnt
WO2020219696A1 (fr) * 2019-04-26 2020-10-29 The Regents Of The University Of California Dispositifs et procédés de génération de cellules progénitrices d'oligodendrocytes
US10982055B2 (en) 2015-10-05 2021-04-20 The Regents Of The University Of California Thermoreversible polymers and methods of use thereof
WO2022251137A1 (fr) * 2021-05-24 2022-12-01 The Regents Of The University Of California Polymères thermoréversibles et leurs procédés d'utilisation

Patent Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5650148A (en) 1988-12-15 1997-07-22 The Regents Of The University Of California Method of grafting genetically modified cells to treat defects, disease or damage of the central nervous system
US5082670A (en) 1988-12-15 1992-01-21 The Regents Of The University Of California Method of grafting genetically modified cells to treat defects, disease or damage or the central nervous system
US6897064B2 (en) 2001-07-13 2005-05-24 Mebiol Inc. Cell or tissue-culturing carrier, and culturing method
WO2006019366A1 (fr) 2003-03-28 2006-02-23 Wisconsin Alumni Research Foundation Conditions physicochimiques de culture pour des cellules souches embryonnaires
WO2006029198A2 (fr) 2004-09-08 2006-03-16 Wisconsin Alumni Research Foundation Culture de cellules souches embryonnaires humaines
WO2006029297A2 (fr) 2004-09-10 2006-03-16 Hoftberg Steven Systeme d'etablissement de priorites theoriques des jeux pour reseau ad hoc mobiles permettant une deference hierarchique
US20060141622A1 (en) 2004-11-17 2006-06-29 Johe Karl K Transplantation of human neural cells for treatment of neurodegenerative conditions
EP1992360A1 (fr) 2006-02-01 2008-11-19 The University of Tokyo Utilisation en association d'un inhibiteur de signal tgf-beta et d'un agent antitumeur
WO2008006583A1 (fr) 2006-07-14 2008-01-17 Novartis Ag Dérivés de la pyrimidine en tant qu'inhibiteurs d'alk-5
WO2008089351A1 (fr) 2007-01-17 2008-07-24 Wisconsin Alumni Research Foundation Culture améliorée de cellules souches
WO2008094597A2 (fr) 2007-01-30 2008-08-07 University Of Georgia Research Foundation, Inc. Cellules mésodermiques précoces, une population stable de cellules mésendodermiques qui a une utilité pour la génération de lignées endodermiques et mésodermiques et de cellules migratoires multipotentes (mmc)
WO2009131166A1 (fr) 2008-04-25 2009-10-29 国立大学法人 千葉大学 Inhibiteur de la signalisation de wnt comprenant une protéine de liaison au facteur de croissance de type insuline
WO2009155001A2 (fr) 2008-05-27 2009-12-23 The Board Of Regents Of The University Of Texas System Inhibiteurs de signalisation de la protéine wnt
US10982055B2 (en) 2015-10-05 2021-04-20 The Regents Of The University Of California Thermoreversible polymers and methods of use thereof
WO2020219696A1 (fr) * 2019-04-26 2020-10-29 The Regents Of The University Of California Dispositifs et procédés de génération de cellules progénitrices d'oligodendrocytes
WO2022251137A1 (fr) * 2021-05-24 2022-12-01 The Regents Of The University Of California Polymères thermoréversibles et leurs procédés d'utilisation
US20240294713A1 (en) 2021-05-24 2024-09-05 The Regents Of The University Of California Thermoreversible polymers and methods of use thereof

Non-Patent Citations (48)

* Cited by examiner, † Cited by third party
Title
ADIL ET AL., SCI REP, vol. 7, 2017, pages 40573
ANDERSON ET AL., NEUROSCIENCE, vol. 67, 1995, pages 7 - 22
BEASLEYREYNOLDS, SCHIZOPHR RES, vol. 24, 1997, pages 349 - 355
BRENNAND ET AL., NATURE, vol. 473, 2011, pages 221 - 225
CHAMBERS ET AL., NAT BIOTECHNOL, vol. 27, 2009, pages 275 - 280
CHANG ET AL., FRONTIERS IN BIOSCIENCE, vol. 12, 2007, pages 4393 - 4401
CHEN ET AL., NAT CHEM BIOL, vol. 5, 2009, pages 100 - 7
CHEUNG ET AL., HUM MOL GENET, vol. 20, 2011, pages 2103 - 2115
CHIANG ET AL., MOLPSYCHIATRY, vol. 16, pages 358 - 360
CTMY ET AL., BIOORG MED CHEM LETT, vol. 18, 2008, pages 4388 - 4392
CUI ET AL., ONCOGENE, vol. 24, no. 301836-41-9, 2005, pages 3864 - 3874
CUNNINGHAM, M. ET AL.: "hPSC-derived maturing GABAergic interneurons ameliorate seizures and abnormal behavior in epileptic mice", CELL STEM CELL, vol. 15, 2014
DACOSTA ET AL., MOLECULAR PHARMACOLOGY, vol. 65, no. 3, 2004, pages 744 - 752
DIMOS ET AL., SCIENCE, vol. 321, 2008, pages 1218 - 1221
ESPUNY-CAMACHO ET AL., NEURON, vol. 77, 2013, pages 440 - 456
FAZZARI ET AL., NATURE, vol. 464, 2010, pages 1376 - 1380
GELLIBERT ET AL., JOURNAL OF MEDICINAL CHEMISTRY, vol. 49, no. 7, 2006, pages 2210 - 2221
GOUVILLE ET AL., DRUG NEWS PERSPECTIVE, vol. 19, no. 2, 2006, pages 85 - 90
HAUANG ET AL., NATURE, vol. 457, 2009, pages 277 - 280
INMAN ET AL., J MOL PHAMACOL, vol. 62, no. 1, 2002, pages 65 - 74
INMAN ET AL., MOLECULAR PHARMACOLOGY, vol. 62, no. 1, 2002, pages 65 - 74
KAMINSKA ET AL., ACTA BIOCHIMICA POLONICA, vol. 52, no. 2, 2005, pages 329 - 337
KIM ET AL., XENOBIOTICA, vol. 38, no. 3, 2008, pages 325 - 339
KIM, T. G. ET AL.: "Efficient specification of interneurons from human pluripotent stem cells by dorsoventral and rostrocaudal modulation", STEM CELLS, vol. 32, no. 7, 2014, pages 1789 - 1804
LEWIS ET AL., NAT REV NEUROSCI, vol. 6, 2005, pages 312 - 324
MARCHETTO ET AL., CELL, vol. 143, 2010, pages 527 - 539
MARIN, O.: "Human cortical interneurons take their time", CELL STEM CELL, vol. 12
MAROOF, A. M. ET AL.: "Directed differentiation and functional maturation of cortical interneurons from human embryonic stem cells", CELL STEM CELL, vol. 12, 2013, XP055283863, DOI: 10.1016/j.stem.2013.04.008
NICHOLAS ET AL.: "Human cortical interneurons take their time", CELL STEM CELL, vol. 12, no. 5, 2013, pages 573 - 586, Retrieved from the Internet <URL:https://doi.org/10.1016/j.stem.2013.04.017>
NICHOLAS, C. R. ET AL.: "Functional maturation of hPSC-derived forebrain interneurons requires an extended timeline and mimics human neural development.", CELL STEM CELL, vol. 12, 2013, XP055741910, DOI: 10.1016/j.stem.2013.04.005
PASCA ET AL., NAT MED, vol. 17, 2011, pages 1657 - 1662
SCHINDELIN, J. ET AL.: "Fiji: An open-source platform for biological-image analysis", NATURE METHODS, vol. 9, 2012, XP055343835, DOI: 10.1038/nmeth.2019
SHI ET AL., NAT NEUROSCI, vol. 15, 2012, pages 477 - 486
SHIMANUKI ET AL., ONCOGENE, vol. 26, 2007, pages 3311 - 3320
SMAD8. YU ET AL., NAT MED, vol. 14, 2008, pages 1363 - 1369
SOLDNER ET AL., CELL, vol. 136, 2009, pages 964 - 977
SOLDNER ET AL., CELL, vol. 146, 2011, pages 318 - 331
SUSSEL ET AL., DEVELOPMENT, vol. 126, pages 3359 - 3370
SUZUKI ET AL., CANCER RESEARCH, vol. 67, no. 5, 2007, pages 2351 - 2359
TAKAHASHIYAMANAKA, CELL, vol. 126, 2006, pages 663 - 676
TOJO ET AL., ANCER SCIENCE, vol. 96, no. 11, 2005, pages 791 - 800
WATANABE, K. ET AL.: "A ROCK inhibitor permits survival of dissociated human embryonic stem cells", NAT BIOTECHNOL, vol. 25, 2007, XP002478043, DOI: 10.1038/nbt1310
WOO ET AL., AM J PSYCHIATRY, vol. 154, 1997, pages 1013 - 1015
WRZESINSKI ET AL., CLINICAL CANCER RESEARCH, vol. 13, no. 18, 2007, pages 5262 - 5270
XU ET AL., J NEUROSCI, vol. 24, 2004, pages 2612 - 2622
XU ET AL., NEURON, vol. 65, 2010, pages 328 - 340
ZHAO ET AL., MOLECULAR BIOLOGY OF THE CELL, vol. 17, 2006
ZHU, Q. ET AL.: "Human cortical interneurons optimized for grafting specifically integrate, abort seizures, and display prolonged efficacy without over-inhibition", NEURON, 2023

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN120789281A (zh) * 2025-09-12 2025-10-17 温州医科大学附属第一医院 一种桑色素纳米颗粒、pH响应性水凝胶及应用

Also Published As

Publication number Publication date
AU2024343386A1 (en) 2025-05-22

Similar Documents

Publication Publication Date Title
JP7023820B2 (ja) 多分化能細胞および多能性細胞の分化を方向付けることによって発生させる皮質介在ニューロンおよびその他のニューロン細胞
Mullen et al. TGF-β family signaling in embryonic and somatic stem-cell renewal and differentiation
EP3047017B1 (fr) Procédés de production de cellules souches rétiniennes de mammifère et leurs utilisations
CN112899230B (zh) 一种膀胱癌类器官培养基及制备方法及应用
CN112585262B (zh) 肠神经前体细胞的制造方法
JP2018531011A6 (ja) 中脳オルガノイドを作製するための手段及び方法
WO2013187416A1 (fr) PROCÉDÉ D&#39;AMPLIFICATION D&#39;UNE CELLULE iPS APPROPRIÉE POUR UNE DIFFÉRENCIATION NEURALE ET PROCÉDÉ DESTINÉ À L&#39;INDUCTION D&#39;UNE CELLULE SOUCHE NEURALE
CN106536718B (zh) 胰芽细胞的制造方法及含有胰芽细胞的胰疾病治疗剂
CN107109361A (zh) 制备小脑祖先组织的方法
CN112601814A (zh) 包含嗅神经细胞或其前体细胞的细胞团块及其制备方法
JPWO2016167372A1 (ja) 大脳皮質ニューロンの誘導方法
CN111492052A (zh) 细胞的培养方法
WO2025097051A1 (fr) Polymères thermoréversibles à stabilité améliorée et procédés et utilisations associés
WO2020235319A1 (fr) Procédé de culture d&#39;expansion pour cellules précurseurs de cartilage ou d&#39;os
AU2024343386B2 (en) Thermoreversible polymers with improved stability and methods and uses thereof
US12268715B2 (en) Method for producing biological tissue-like structure
US20250215397A1 (en) Two-dimensional culture method having clear chemical composition for culturing three-dimensional intestinal organoid-derived intestinal stem cell aggregate
CN119948151A (zh) 胰腺内胚层细胞的制造方法
WO2023017848A1 (fr) Procédé de production de cellules progénitrices interstitielles rénales, cellules produisant de l&#39;érythropoïétine et procédé de production de cellules produisant de la rénine
TW202246489A (zh) 成熟化劑
JP7072756B2 (ja) 多能性幹細胞から中胚葉前駆細胞および血液血管前駆細胞への分化誘導法
WO2025231435A1 (fr) Développement de la différenciation des hépatocytes humains et procédé de détection par l&#39;utilisation de cellules ipsc
WO2025127102A1 (fr) Procédé de production de cellules souches hépatiques
EA047959B1 (ru) Способ получения энтеральных нейральных клеток-предшественников
CN117836404A (zh) 肾间质前体细胞的制造方法及促红细胞生成素产生细胞和肾素产生细胞的制造方法

Legal Events

Date Code Title Description
ENP Entry into the national phase

Ref document number: 2024343386

Country of ref document: AU

Date of ref document: 20241101

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2025531786

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2025531786

Country of ref document: JP

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 24809158

Country of ref document: EP

Kind code of ref document: A1