Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
TREATMENT OF RETINAL VASCULAR DISEASE USING PROGENITOR CELLS
Document Type and Number:
WIPO Patent Application WO/2018/009385
Kind Code:
A1
Abstract:
Methods and compositions for treating ophthalmic disease, reducing retinal neovascularization and retinal vascular leakage using progenitor cells, such as postpartum-derived cells, and conditioned media from the cells, are disclosed.

Inventors:
HARRIS IAN (US)
DEJNEKA NADINE SOPHIA (US)
Application Number:
PCT/US2017/039660
Publication Date:
January 11, 2018
Filing Date:
June 28, 2017
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
JANSSEN BIOTECH INC (US)
International Classes:
A61K35/51; A61P27/02
Domestic Patent References:
WO2003025149A22003-03-27
WO2003025149A22003-03-27
Foreign References:
US20050037491A12005-02-17
US20100272803A12010-10-28
US20050037491A12005-02-17
US7524489B22009-04-28
US7510873B22009-03-31
US20050058631A12005-03-17
US5718922A1998-02-17
US5869079A1999-02-09
US6375972B12002-04-23
US5902598A1999-05-11
US6331313B12001-12-18
US5707643A1998-01-13
US5466233A1995-11-14
US6251090B12001-06-26
US6355699B12002-03-12
US20020022676A12002-02-21
US6555374B12003-04-29
Other References:
KIMIO TAKEUCHI ET AL: "Human Umbilical Tissue-derived Cells (hUTC) Ameliorate Retinal Vascular Leakage In A Diabetic Rat", ARVO ANNUAL MEETING ABSTRACT MARCH 2012, 1 March 2012 (2012-03-01), pages 1 - 2, XP055401671, Retrieved from the Internet [retrieved on 20170828]
JING CAO ET AL: "Human umbilical tissue-derived cells rescue retinal pigment epithelium dysfunction in retinal degeneration : Translational and Clinical Research", STEM CELLS., vol. 34, no. 2, 26 November 2015 (2015-11-26), US, pages 367 - 379, XP055401658, ISSN: 1066-5099, DOI: 10.1002/stem.2239
LUND, R. D. ET AL., PROGRESS IN RETINAL AND EYE RESEARCH, vol. 20, 2001, pages 415 - 449
PURVES, D ET AL.: "Neuroscience 4th edition,", 2008
TRESCO, P. A. ET AL., ADVANCED DRUG DELIVERY REVIEWS, vol. 42, 2000, pages 2 - 37
FENG W. ET AL., J BIOL CHEM., vol. 277, no. 19, 10 December 2001 (2001-12-10), pages 17016 - 17022
TAKAHASHI ET AL., CELL, vol. 126, no. 4, 2006, pages 663 - 676
TAKAHASHI ET AL., CELL, vol. 131, 2007, pages 1 - 12
DOYLE ET AL.,: "CELL & TISSUE CULTURE: LABORATORY PROCEDURES", 1995, JOHN WILEY & SONS
HO AND WANG: "ANIMAL CELL BIOREACTORS", 1991, BUTTERWORTH-HEINEMANN
FRESHNEY: "CULTURE OF ANIMAL CELLS: A MANUAL OF BASIC TECHNIQUES, 3rd ed.,", 1994, WILEY-LISS, INC.
LANG, K. J. D. ET AL., J. NEUROSCI. RES., vol. 76, 2004, pages 184 - 192
JOHE, K. K. ET AL., GENES DEVEL., vol. 10, 1996, pages 3129 - 3140
GOTTLEIB, D., ANN. REV. NEUROSCI., vol. 25, 2002, pages 381 - 407
MOMBAERTS ET AL., PROC. NAT. ACAD. SCI. U.S.A, vol. 88, 1991, pages 3084 - 3087
L. G. DAVIS ET AL.: "BASIC METHODS IN MOLECULAR BIOLOGY, 2nd ed.,", 1994, APPLETON & LANGE
P. A. TRESCO ET AL., ADV. DRUG DELIVERY REV., vol. 42, 2000, pages 3 - 27
A. DOMB ET AL., POLYMERS FOR ADVANCED TECHNOLOGIES, vol. 3, 1992, pages 279 - 291
D. W. HUTMACHER, J. BIOMATER. SCI. POLYMER EDN, vol. 12, 2001, pages 107 - 174
ANSETH, K. S. ET AL., J. CONTROLLED RELEASE, vol. 78, 2002, pages 199 - 209
WANG, D. ET AL., BIOMATERIALS, vol. 24, 2003, pages 3969 - 3980
MARX, W. F. ET AL., AM. J. NEURORADIOL, vol. 22, 2001, pages 323 - 333
"Age-Related Eye Disease Study Research Group, NEI, NIH, AREDS Report No.8", ARCH. OPHTHALMOL., vol. 119, 2001, pages 1417 - 1436
TRICHONAS, INVEST. OPHTHALMOL. VIS. SCI., vol. 51, 2010, pages 1677 - 1682
MESSINA ET AL., EXPER. NEUROL., vol. 184, 2003, pages 816 - 829
TUSHER, V. G. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 98, 2001, pages 5116 - 5121
ROSEN ET AL., CIBA FOUND. SYMP., vol. 212, 1997, pages 215 - 26
SALCEDO ET AL., BLOOD, vol. 96, 2000, pages 34 - 40
LI ET AL., J. IMMUNOL., vol. 170, 2003, pages 3369 - 76
HUGHES ET AL., ANN. THORAC. SURG, vol. 77, 2004, pages 812 - 8
CHENG ET AL., DEV. BIOL., vol. 258, 2003, pages 319 - 33
PAXINOS, G.; WATSON, C., THE RAT BRAIN IN STEREOTAXIC COORDINATES, 1997
LIU, K ET AL., PNAS, vol. 96, 1999, pages 5147 - 5152
PLAIA ET AL., STEM CELLS, vol. 24, no. 3, 2006, pages 531 - 546
Attorney, Agent or Firm:
SHIRTZ, Joseph F. et al. (US)
Download PDF:
Claims:
WE CLAIM:

1. A composition for use in inhibiting or reducing retinal neovascularization in retinopathy comprising a homogenous population of human umbilical cord tissue-derived cells, wherein the cell population is isolated from human umbilical cord tissue substantially free of blood, is capable of self-renewal and expansion in culture, expresses CD13, CD90 and HLA-ABC, and does not express CD31, CD34, CD45 and CD117.

2. The composition of claim 1, wherein the retinopathy is diabetic retinopathy.

3. The composition of claim 1, wherein the cell population further has the following characteristics:

a) potential for 40 population doublings in culture;

b) expresses CD 10, CD44 and CD73; and

c) does not express CD 141.

4. The composition of claim 1, wherein the cell population has increased expression of genes encoding interleukin 8 and reticulon 1 relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an iliac crest bone marrow cell.

5. The composition of claim 1, wherein the cell population is 1,000 to 20,000 cells.

6. The composition of claim 1, wherein the cell population is 4,000 to 20,000 cells.

7. The composition of claim 1, wherein the cell population is 1,000 to 4,000 cells.

8. A composition for use in inhibiting or reducing vascular leakage in retinopathy comprising a homogenous population of human umbilical cord tissue-derived cells, wherein the cell population is isolated from human umbilical cord tissue substantially free of blood, is capable of self-renewal and expansion in culture, expresses CD13, CD90 and HLA-ABC, and does not express CD31, CD34, CD45 and CD117..

9. The composition of claim 9, wherein the retinopathy is diabetic retinopathy.

10. The composition of claim 9, wherein the cell population further has the following characteristics:

a) potential for 40 population doublings in culture;

b) expresses CD 10, CD44 and CD73; and

c) does not express CD 141.

11. The composition of claim 9, wherein the cell population has increased expression of genes encoding interleukin 8 and reticulon 1 relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an iliac crest bone marrow cell.

12. The composition of claim 9, wherein the cell population is 1,000,000 to 30,000,000 cells.

13. The composition of claim 1, wherein the cell population is 3 x 107 cells.

14. The composition of any of claims 1 - 13, wherein the composition is wherein the composition is a pharmaceutical composition.

15. The composition of claim 14, wherein the pharmaceutical composition comprises a pharmaceutical carrier.

16. A population of postpartum-derived cells for use in inhibiting or reducing retinal neovascularization in retinopathy comprising a homogenous population of human umbilical cord tissue-derived cells, wherein the cell population is isolated from human umbilical cord tissue substantially free of blood, is capable of self-renewal and expansion in culture, expresses CD13, CD90 and HLA-ABC, and does not express CD31, CD34, CD45 and CD 117.

17. A population of postpartum-derived cells for use in inhibiting or reducing vascular leakage in retinopathy comprising a homogenous population of human umbilical cord tissue-derived cells, wherein the cell population is isolated from human umbilical cord tissue substantially free of blood, is capable of self-renewal and expansion in culture, expresses CD13, CD90 and HLA-ABC, and does not express CD31, CD34, CD45 and CD117..

Description:
TREATMENT OF RETINAL VASCULAR DISEASE

USING PROGENITOR CELLS

CROSS REFERENCE TO RELATED APPLICATIONS

[0001] This application claims benefit of U.S. Provisional Application Serial No.

62/358,389, filed July 5, 2016, the entire contents of which is incorporated by reference herein.

FIELD OF THE INVENTION

[0002] This invention relates to the field of cell-based or regenerative therapy for ophthalmic diseases and disorders, particularly ocular conditions, such as retinal degenerative conditions. The invention provides methods and compositions for the regeneration or repair of ocular cells and tissue using progenitor cells, such as umbilical cord tissue-derived cells, placenta tissue-derived cells, and conditioned media prepared from those cells.

BACKGROUND

[0003] As a complex and sensitive organ of the body, the eye can experience numerous diseases and other deleterious conditions that affect its ability to function normally. Many of these conditions are associated with damage or degeneration of specific ocular cells, and tissues made up of those cells. As one example, diseases and degenerative conditions of the optic nerve and retina are the leading causes of blindness throughout the world. Damage or degeneration of the cornea, lens and associated ocular tissues represent another significant cause of vision loss worldwide.

[0004] The retina contains seven layers of alternating cells and processes that convert a light signal into a neural signal. The retinal photoreceptors and adjacent retinal pigment epithelium (RPE) form a functional unit that, in many disorders, becomes unbalanced due to genetic mutations or environmental conditions (including age). This results in loss of photoreceptors through apoptosis or secondary degeneration, which leads to progressive deterioration of vision and, in some instances, to blindness (for a review, see, e.g., Lund, R. D. et al., Progress in Retinal and Eye Research, 2001; 20:415-449). Two classes of ocular disorders that fall into this pattern are age-related macular degeneration (AMD) and retinitis pigmentosa (RP). [0005] AMD is the most common cause of vision loss in the United States in those people whose ages are 50 or older, and its prevalence increases with age. The primary disorder in AMD appears to be due to RPE dysfunction and changes in Bruch's membranes, characterized by, among other things, lipid deposition, protein cross-linking and decreased permeability to nutrients (see Lund et al, 2001 supra). A variety of elements may contribute to macular degeneration, including genetic makeup, age, nutrition, smoking and exposure to sunlight. The nonexudative, or "dry" form of AMD accounts for 90% of AMD cases; the other 10%) being the exudative-neovascular form ("wet" AMD). In dry- AMD patients, there is a gradual disappearance of the retinal pigment epithelium (RPE), resulting in

circumscribed areas of atrophy. Since photoreceptor loss follows the disappearance of RPE, the affected retinal areas have little or no visual function.

[0006] Current therapies for AMD involve procedures, such as, for example, laser therapy and pharmacological intervention. By transferring thermal energy, the laser beam destroys the leaky blood vessels under the macula, slowing the rate of vision loss. A disadvantage of laser therapy is that the high thermal energy delivered by the beam also destroys healthy tissue nearby. Neuroscience 4 th edition, (Purves, D, et al. 2008) states "[c]urrently there is no treatment for dry AMD."

[0007] Other less common, but nonetheless debilitating retinopathies can also involve progressive cellular degeneration leading to vision loss and blindness. These include, for example, diabetic retinopathy and choroidal neovascular membrane (CNVM). Diabetic retinopathy is a major complication of Type 1 and Type 2 diabetes mellitus, being observed in most patients after 10 years of diabetes, and increasing the risk of blindness 25-fold above normal. The natural history of clinically demonstrable retinopathy has been carefully documented, and important stages have been identified: vascular occlusion, formation of capillary microaneurysms, excessive vascular permeability, proliferation of new vessels and fibrous tissue, and contraction of the fibrovascular proliferations.

[0008] The advent of stem cell-based therapy for cell and tissue repair and regeneration provides promising treatments for a number of aforementioned cell-degenerative pathologies and other retinal conditions. Stem cells are capable of self-renewal and differentiation to generate a variety of mature cell lineages. Transplantation of such cells can be utilized as a clinical tool for reconstituting a target tissue, thereby restoring physiologic and anatomic functionality. The application of stem cell technology is wide-ranging, including tissue engineering, gene therapy delivery, and cell therapeutics, i.e., delivery of biotherapeutic agents to a target location via exogenously supplied living cells or cellular components that produce or contain those agents. (For a review, see, for example, Tresco, P. A. et al,

Advanced Drug Delivery Reviews, 2000, 42: 2-37).

[0009] It has been shown that postpartum-derived cells ameliorate retinal degeneration (US 2010/0272803). The Royal College of Surgeons (RCS) rat presents with a tyrosine receptor kinase (Mertk) defect affecting outer segment phagocytosis, leading to photoreceptor cell death. (Feng W. et al., J Biol Chem., 2002, 10: 277 (19): 17016-17022). Transplantation of retinal pigment epithelial (RPE) cells into the subretinal space of RCS rats was found to limit the progress of photoreceptor loss and preserve visual function. It also has been demonstrated that postpartum-derived cells can be used to promote photoreceptor rescue and thus preserve photoreceptors in the RCS model. (US 2010/0272803). Injection of human umbilical cord tissue-derived cells (hUTCs) subretinally into RCS rat eye improved visual acuity and ameliorated retinal degeneration. Moreover, treatment with conditioned medium (CM) derived from hUTC restored phagocytosis of ROS in dystrophic RPE cells in vitro. (US 2010/0272803). The utilization of hUTC for improving vision is further demonstrated here.

SUMMARY

[00010] This invention provides compositions and methods applicable to cell-based or regenerative therapy for ophthalmic diseases and disorders. In particular, the invention features methods and compositions, including pharmaceutical compositions, for treating an ophthalmic disease or condition, including the regeneration or repair of ocular cells and tissue, using progenitor cells such as postpartum-derived cells, and conditioned media generated from those cells. The postpartum-derived cells may be umbilical cord tissue- derived cells (UTCs) or placental tissue-derived cells (PDCs).

[00011] One aspect of the invention is a method of treating an ophthalmic disease, such as retinopathy, by inhibiting or reducing retinal neovascularization comprising administering to the eye of a subject a population of progenitor cells or a conditioned medium prepared from a population of progenitor cells, or a composition comprising a population of progenitor cells or a conditioned medium prepared from a population of progenitor cells. In particular embodiments of the invention, the progenitor cells are postpartum-derived cells. In embodiments of the invention, the postpartum-derived cells are isolated from human umbilical cord tissue or placental tissue substantially free of blood.

[00012] A further aspect of the invention is a method for inhibiting or reducing retinal neovascularization in retinopathy, the method comprising administering to the eye of a subject a population of progenitor cells or a conditioned medium prepared from a population of progenitor cells, or a composition comprising a population of progenitor cells or a conditioned medium prepared from a population of progenitor cells in an amount effective to inhibit or reduce retinal neovascularization. In particular embodiments of the invention, the progenitor cells are postpartum-derived cells. In embodiments of the invention, the postpartum-derived cells are isolated from human umbilical cord tissue or placental tissue substantially free of blood.

[00013] In the embodiments described above, administration to the eye is to the interior of the eye. In the embodiments, the cells are administered by injection, such as intravitreal injection or subretinal injection. In embodiments, the cell population is administered with about 1,000 to about 20,000 cells. In particular embodiments, the cell population is administered by intravitreal injection with about 4,000 to about 20,000 cells. In some embodiments, the cell population is administered by subretinal injection with about 1,000 to about 4,000 cells. In particular embodiments, about 4,000 cells are administered.

[00014] Another aspect of the invention is a method of inhibiting or reducing vascular leakage in retinopathy comprising administering a population of progenitor cells or a conditioned medium prepared from a population of progenitor cells to the eye of a subject in an amount effective to inhibit or reduce vascular leakage. In particular embodiments, the progenitor cells are postpartum-derived cells. In embodiments, the postpartum-derived cells are isolated from human umbilical cord tissue or placental tissue substantially free of blood. In embodiments, the cells are administration to the interior of the eye. In some embodiments, the cell population is administered by injection, such as intravitreal injection or subretinal injection. In some embodiments, the cell population is administered at about 1 million to about 30 million cells, particularly at about 2 million to about 30 million, more particularly at about 10 million to about 30 million. In particular embodiments, the cell population is administered by subretinal injection at about 30 million cells.

[00015] A further embodiment of the invention is a composition for use in treating an ophthalmic disease, such as retinopathy, by inhibiting or reducing retinal neovascularization comprising a population of progenitor cells or a conditioned medium prepared from a population of progenitor cells. In embodiments, the progenitor cells are postpartum-derived cells. In embodiments of the invention, the postpartum-derived cells are isolated from human umbilical cord tissue or placental tissue substantially free of blood. In embodiments, the cell population has about about 1,000 to about 20,000 cells, particularly at about 4,000 to about 20,000 cells. In particular embodiments, the cell population is about 4,000 cells.

[00016] Another embodiment is a composition for use in a method of inhibiting or reducing retinal neovascularization in retinopathy, comprising a population of progenitor cells or a conditioned medium prepared from a population of progenitor cells. In

embodiments, the progenitor cells are postpartum-derived cells. In embodiments, the postpartum-derived cells are isolated from human umbilical cord tissue or placental tissue substantially free of blood. In embodiments, the cell population has about about 1,000 to about 20,000 cells, particularly at about 4,000 to about 20,000 cells. In particular

embodiments, the cell population is about 4,000 cells.

[00017] Yet another embodiment is a composition for use in inhibiting or reducing vascular leakage, comprising a population of progenitor cells or a conditioned medium prepared from a population of progenitor cells. In the embodiments, the composition is effective to inhibit or reduce vascular leakage. In embodiments, the progenitor cells are postpartum-derived cells. In embodiments, the postpartum-derived cells are isolated from human umbilical cord tissue or placental tissue substantially free of blood. In embodiments, the cell population has about 1 million to about 30 million cells, particularly at about 2 million to about 30 million, more particularly at about 10 million to about 30 million. In particular embodiments, the cell population is administered by subretinal injection at about 30 million cells.

[00018] Other embodiments relate to a population of progenitor cells for use in treating retinopathy. One embodiment is a population of progenitor cells for use in inhibiting or reducing retinal neovascularization. Another embodiment is a population of progenitor cells for use in inhibiting or reducing vascular leakage. Additional embodiments include use of a population of progenitor cells or a composition comprising a population of progenitor cells for treating a retinopathy by inhibiting or reducing retinal neovascularization, use of a population of progenitor cells or a composition comprising a population of progenitor cells for inhibiting or reducing retinal neovascularization in retinopathy, and use of a population of progenitor cells or a composition comprising a population of progenitor cells for inhibiting or reducing vascular leakage. In the embodiments, the composition comprises a population of progenitor cells or a conditioned medium prepared from a population of progenitor cells. In embodiments, the progenitor cells are postpartum-derived cells. In embodiments, the postpartum-derived cells are isolated from human umbilical cord tissue or placental tissue substantially free of blood. In the embodiments, the composition or cell population is administered to the eye of a subject by injection, such as intravitreal injection or subretinal injection. In embodiments, the cell population is about 1,000 to about 30 million cells. In particular embodiments, the composition or cell population is administered by intravitreal injection and the cell population comprises about 4,000 to about 20,000 cells. In some embodiments, the composition or cell population is administered by subretinal injection and the cell population comprises about 1,000 to about 4,000 cells. In particular embodiments, cell population comprises about 4,000 cells. In some embodiments, the cell population comprises about 1 million to about 30 million cells, particularly at about 2 million to about 30 million, more particularly at about 10 million to about 30 million. In particular embodiments, the composition or cell population is administered by subretinal injection and the cell population comprises at about 30 million cells.

[00019] In the embodiments of the invention, the postpartum-derived cell is derived from human umbilical cord tissue or placental tissue substantially free of blood. In embodiments, the cell is capable of expansion in culture and has the potential to differentiate into a cell of a neural phenotype; wherein the cell requires L-valine for growth and is capable of growth in at least about 5% oxygen. The cell further comprises one or more of the following

characteristics: (a) potential for at least about 40 doublings in culture; (b) attachment and expansion on a coated or uncoated tissue culture vessel, wherein the coated tissue culture vessel comprises a coating of gelatin, laminin, collagen, polyomithine, vitronectin, or fibronectin; (c) production of at least one of tissue factor, vimentin, and alpha-smooth muscle actin; (d) production of at least one of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD- L2 and HLA-A,B,C; (e) lack of production of at least one of CD31, CD34, CD45, CD80, CD86, CD117, CD141, CD178, B7-H2, HLA-G, and HLA-DR,DP,DQ, as detected by flow cytometry; (f) expression of a gene, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an iliac crest bone marrow cell, is increased for at least one of a gene encoding: interleukin 8; reticulon 1; chemokine (C--X--C motif) ligand 1 (melonoma growth stimulating activity, alpha); chemokine (C--X--C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C--X--C motif) ligand 3; tumor necrosis factor, alpha- induced protein 3; C-type lectin superfamily member 2; Wilms tumor 1; aldehyde

dehydrogenase 1 family member A2; renin; oxidized low density lipoprotein receptor 1; Homo sapiens clone EVIAGE:4179671; protein kinase C zeta; hypothetical protein DKFZp564F013; downregulated in ovarian cancer 1; and Homo sapiens gene from clone DKFZp547kl 113; (g) expression of a gene, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an iliac crest bone marrow cell, is reduced for at least one of a gene encoding: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C--X--C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeo box 2 (growth arrest-specific homeo box); sine oculis homeobox homolog 1 (Drosophila); crystallin, alpha B; disheveled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1; tetranectin

(plasminogen binding protein); src homology three (SH3) and cysteine rich domain;

cholesterol 25-hydroxylase; runt-related transcription factor 3; interleukin 1 1 receptor, alpha; procollagen C-endopeptidase enhancer; frizzled homolog 7 (Drosophila); hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; neuroblastoma, suppression of tumorigenicity 1; insulin-like growth factor binding protein 2, 36 kDa; Homo sapiens cDNA FLJ12280 fis, clone MAMMA1001744; cytokine receptor-like factor 1;

potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, beta 7; transcriptional co-activator with PDZ-binding motif (T AZ); sine oculis homeobox homolog 2 (Drosophila); KIAA1034 protein; vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1; early growth response 3; distal-less homeo box 5; hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3 -alpha hydroxysteroid dehydrogenase, type II); biglycan;

transcriptional co-activator with PDZ-binding motif (TAZ); fibronectin 1; proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens mRNA full length insert cDNA clone EUROEVIAGE 1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA; cDNA DKFZp564B222 (from clone DKFZp564B222); BCL2/adenovirus E1B 19 kDa interacting protein 3 -like; AE binding protein 1; cytochrome c oxidase subunit Vila polypeptide 1 (muscle); similar to neuralin 1; B cell translocation gene 1; hypothetical protein FLJ23191; and DKFZp586L151; and( h) lack expression of hTERT or telomerase. In one embodiment, the umbilical cord tissue-derived cell further has the characteristics of (i) secretion of at least one of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MlPlb, 1309, MDC, RANTES, and ΊΊΜΡ1; (j) lack of secretion of at least one of TGF-beta2, MlPla, ANG2, PDGFbb, and VEGF, as detected by ELISA. In another embodiment, the placenta tissue-derived cell further has the characteristics of (i) secretion of at least one of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, HB-EGF, BDNF, TPO, MlPla, RANTES, and TIMP1; (j) lack of secretion of at least one of TGF-beta2, ANG2, PDGFbb, FGF, and VEGF, as detected by ELISA.

[00020] In specific embodiments, the postpartum-derived cell has all the identifying features of cell type UMB 022803 (P7) (ATCC Accession No. PTA-6067); cell type UMB 022803 (PI 7) (ATCC Accession No. PTA-6068), cell type PLA 071003 (P8) (ATCC

Accession No. PTA-6074); cell type PLA 071003 (PI 1) (ATCC Accession No. PTA-6075); or cell type PLA 071003 (PI 6) (ATCC Accession No. PTA-6079. In an embodiment, the postpartum-derived cell derived from umbilicus tissue has all the identifying features of cell type UMB 022803 (P7) (ATCC Accession No. PTA-6067) or cell type UMB 022803 (PI 7) (ATCC Accession No. PTA-6068). In another embodiment, the postpartum-derived cell derived from placenta tissue has all the identifying features of cell type PLA 071003 (P8) (ATCC Accession No. PTA-6074); cell type PLA 071003 (PI 1) (ATCC Accession No. PTA- 6075); or cell type PLA 071003 (PI 6) (ATCC Accession No. PTA-6079).

[00021] In certain embodiments, the postpartum-derived cells are isolated in the presence of one or more enzyme activities comprising metalloprotease activity, mucolytic activity and neutral protease activity. Preferably, the cells have a normal karyotype, which is maintained as the cells are passaged in culture.

[00022] In embodiments described herein, the cells are capable of self-renewal and expansion in culture, and have the potential to differentiate into cells of other phenotypes. In some embodiments, the cells express CD13, CD90 and HLA-ABC. In preferred

embodiments, the postpartum-derived cells express each of CD10, CD13, CD44, CD73, CD90 and HLA-ABC. In certain embodiments, the postpartum-derived cells express each of CD 10, CD 13, CD44, CD73, CD90, PDGFr-alpha, and HLA-A,B,C. In some embodiments, the cells do not express any of CD31, CD34, CD45 or CD117. In some embodiments, the postpartum-derived cells do not express any of CD31, CD34, CD45, CD117 and CD141, as detected by flow cytometry. In embodiments, the cells lack expression of hTERT or telomerase.

[00023] In embodiments of the invention, the cell population is a substantially

homogeneous population of postpartum-derived cells. In a specific embodiment, the population is a homogeneous population of postpartum-derived cells. In embodiments of the invention, the postpartum-derived cells are derived from human umbilical cord tissue or placental tissue substantially free of blood.

[00024] In certain embodiments, the population of postpartum-derived cells or a conditioned medium generated from a population of postpartum-derived cells as described above is administered with at least one other cell type, such as an astrocyte, oligodendrocyte, neuron, neural progenitor, neural stem cell, retinal epithelial stem cell, corneal epithelial stem cell, or other multipotent or pluripotent stem cell. In these embodiments, the other cell type can be administered simultaneously with, or before, or after, the cell population or the conditioned medium.

[00025] Likewise, in these and other embodiments, the the population of postpartum- derived cells or the conditioned media prepared from the population of cells as described above is administered with at least one other agent, such as a drug for ocular therapy, or another beneficial adjunctive agent such as an anti-inflammatory agent, anti-apoptotic agents, antioxidants or growth factors. In these embodiments, the other agent can be administered simultaneously with, before, or after, the cell population or the conditioned media.

[00026] In various embodiments, the population of postpartum-derived cells or conditioned media generated from postpartum-derived cells (umbilical or placental) is administered to the surface of an eye, or is administered to the interior of an eye or to a location in proximity to the eye (e.g., behind the eye). In embodiments, administration to the interior of an eye may be injection, such as subretinal injection or intravitreal injection. The population of postpartum-derived cells or the conditioned media can be administered through a cannula or from a device implanted in the patient's body within or in proximity to the eye, or may be administered by implantation of a matrix or scaffold with the postpartum-derived cell population or conditioned media.

[00027] In certain embodiments, the composition comprises at least one other cell type, such as an astrocyte, oligodendrocyte, neuron, neural progenitor, neural stem cell, retinal epithelial stem cell, corneal epithelial stem cell, or other multipotent or pluripotent stem cell. In these or other embodiments, the composition comprises at least one other agent, such as a drug for treating the ocular degenerative disorder or other beneficial adjunctive agents, e.g., anti-inflammatory agents, anti-apoptotic agents, antioxidants or growth factors.

[00028] In some embodiments, the composition is a pharmaceutical composition further comprising a pharmaceutically acceptable carrier. [00029] In certain embodiments, the pharmaceutical composition is formulated for administration to the surface of an eye. Alternatively, they can be formulated for

administration to the interior of an eye or in proximity to the eye (e.g., behind the eye). In embodiments, administration to the interior of an eye may be injection, such as subretinal injection or intravitreal injection. The compositions also can be formulated as a matrix or scaffold containing the postpartum-derived cells or conditioned media.

[00030] In embodiments described above, the umbilicus-derived cells or placental-derived cells have one or more of the following characteristics: are positive for HLA-A,B,C; are positive for CD10, CD13, CD44, CD73, CD90; are negative for HLA-DR,DP,DQ; lack production of, or are negative for CD31, CD34, CD45, CD117, and CD141. In

embodiments, the cells produce vimentin and alpha-smooth muscle actin.

[00031] In further embodiments described above, the umbilicus-derived cells have increased expression of genes encoding interleukin 8 and reticulon 1 relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an iliac crest bone marrow cell. In embodiments, the umbilicus-derived cells lack expression of hTERT or telomerase.

[00032] In the embodiments described above, the retinal degeneration or retinopathy is diabetic retinopathy and choroidal neovascular membrane (CNVM).

BRIEF DESCRIPTION OF THE DRAWINGS

[00033] Figure 1 illustrates computer-assisted image analysis method by which both vascular area and neovascular area are calculated. Irregular polygons are used to designate total retinal area (not depicted), vascular area (left panel) and neovascular area (right panel). Pixel counts are converted to mm 2 . The ratio of vascular area to total retinal area yields percent retinal vascular area.

[00034] Figure 2 shows the effect of hUTC on intra-retinal vessel growth among untreated, vehicle treatement, positive control (anti-VEGF), hUTC low, medium and high dose treatment. Statistical significance was calculated using area (mm 2 ) measurements, but the data are depicted using percent total retinal area vascularized for ease of interpretation. Error bars indicate standard error.

[00035] Figures 3A-3E show the effect of hUTC on pre-retinal neovascular growth among untreated, vehicle treatement, positive control (anti-VEGF), hUTC low and medium dose bar graph (FIG. 3 A) and scatter plot (FIG. 3B). Error bars indicate standard error. FIG. 3C shows ADPase-stained retinas from several treatment groups, images are representative of the degree of vascular pathology observed following OIR treatment and intravitreal injection of cryopreservative vehicle (A), positive control compound (B), or medium density hUTC (C). No neovascular growth in seen in the right panel. FIG. 3D shows retina 58R from the high hUTC density treatment group - the injected cells appear to organize into a sheet, extending from the optic disc to the mid-periphery in some retinal quadrants and the far periphery in others. The top panel shows this sheet on the surface of an unstained, dissected retina. The bottom left panel shows the stained retina after the sheet of cells has been peeled. FIG. 3E shows a single retina from the high hUTC density treatment group (23R) yielded both vascular area and neovascular area data.

[00036] Figures 4A-4D demonstrate the effect of hUTC with subretinal injection. FIG. 4 A demonstrates the location of hUTC in the subretinal space six days after injection. FIG. 4B shows the effect of hUTC on intra-retinal vessel growth among untreated, vehicle treatement, hUTC low and medium dose. FIG. 4C shows the effect of hUTC on pre-retinal neovascular growth. Error bars indicate standard error. FIG. 4D illustrates the degree of vascular pathology observed in three treatment groups following OIR and subretinal injection of 2x10 4 hUTC, 4x10 3 hUTC or cryopreservative vehicle, respectively. Arrows indicate the location of neovascular tufts.

[00037] Figures 5A-5F show the effects of hUTC on retinal vascular leakage. FIG. 5 A shows retinal thickness in diabetes in rodents by SD-OCT. FIGS. 5B and 5C show the effect of hUTC on vascular leakage assessed by biotin-BSA assay. FIGS. 5D-5F show expression pattern of cytokines VEGF, ICAM-1, PEDF and ZO-1 in diabetes and with treatment of hUTC.

[00038] Other features and advantages of the invention will be apparent from the detailed description and examples that follow.

DETAILED DESCRIPTION

[00039] Various patents and other publications are referred to throughout the specification. Each of these publications is incorporated by reference herein, in its entirety. In the following detailed description of the illustrative embodiments, reference is made to the accompanying drawings that form a part hereof. These embodiments are described in sufficient detail to enable those skilled in the art to practice the invention, and it is understood that other embodiments may be utilized and that logical structural, mechanical, electrical, and chemical changes may be made without departing from the spirit or scope of the invention. To avoid detail not necessary to enable those skilled in the art to practice the embodiments described herein, the description may omit certain information known to those skilled in the art. The following detailed description is, therefore, not to be taken in a limiting sense, and the scope of the illustrative embodiments are defined by the appended claims.

Definitions

[00040] Various terms used throughout the specification and claims are defined as set forth below and are intended to clarify the invention.

[00041] Stem cells are undifferentiated cells defined by the ability of a single cell both to self-renew, and to differentiate to produce progeny cells, including self-renewing progenitors, non-renewing progenitors, and terminally differentiated cells. Stem cells are also

characterized by their ability to differentiate in vitro into functional cells of various cell lineages from multiple germ layers (endoderm, mesoderm and ectoderm), as well as to give rise to tissues of multiple germ layers following transplantation, and to contribute

substantially to most, if not all, tissues following injection into blastocysts.

[00042] Stem cells are classified according to their developmental potential as: (1) totipotent; (2) pluripotent; (3) multipotent; (4) oligopotent; and (5) unipotent. Totipotent cells are able to give rise to all embryonic and extraembryonic cell types. Pluripotent cells are able to give rise to all embryonic cell types. Multipotent cells include those able to give rise to a subset of cell lineages, but all within a particular tissue, organ, or physiological system (for example, hematopoietic stem cells (HSC) can produce progeny that include HSC (self- renewal), blood cell-restricted oligopotent progenitors, and all cell types and elements (e.g., platelets) that are normal components of the blood). Cells that are oligopotent can give rise to a more restricted subset of cell lineages than multipotent stem cells; and cells that are unipotent are able to give rise to a single cell lineage (e.g., spermatogenic stem cells).

[00043] Stem cells are also categorized on the basis of the source from which they may be obtained. An adult stem cell is generally a multipotent undifferentiated cell found in tissue comprising multiple differentiated cell types. The adult stem cell can renew itself. Under normal circumstances, it can also differentiate to yield the specialized cell types of the tissue from which it originated, and possibly other tissue types. Induced pluripotent stem cells (iPS cells) are adult cells that are converted into pluripotent stem cells. (Takahashi et al, Cell, 2006; 126(4):663-676; Takahashi et al, Cell, 2007; 131 : 1-12). An embryonic stem cell is a pluripotent cell from the inner cell mass of a blastocyst-stage embryo. A fetal stem cell is one that originates from fetal tissues or membranes. A postpartum stem cell is a multipotent or pluripotent cell that originates substantially from extraembryonic tissue available after birth, namely, the placenta and the umbilical cord. These cells have been found to possess features characteristic of pluripotent stem cells, including rapid proliferation and the potential for differentiation into many cell lineages. Postpartum stem cells may be blood-derived (e.g., as are those obtained from umbilical cord blood) or non-blood-derived (e.g., as obtained from the non-blood tissues of the umbilical cord and placenta).

[00044] Embryonic tissue is typically defined as tissue originating from the embryo (which in humans refers to the period from fertilization to about six weeks of development). Fetal tissue refers to tissue originating from the fetus, which in humans refers to the period from about six weeks of development to parturition. Extraembryonic tissue is tissue associated with, but not originating from, the embryo or fetus. Extraembryonic tissues include extraembryonic membranes (chorion, amnion, yolk sac and allantois), umbilical cord and placenta (which itself forms from the chorion and the maternal decidua basalis).

[00045] Differentiation is the process by which an unspecialized ("uncommitted") or less specialized cell acquires the features of a specialized cell, such as a nerve cell or a muscle cell, for example. A differentiated cell is one that has taken on a more specialized

("committed") position within the lineage of a cell. The term committed, when applied to the process of differentiation, refers to a cell that has proceeded in the differentiation pathway to a point where, under normal circumstances, it will continue to differentiate into a specific cell type or subset of cell types, and cannot, under normal circumstances, differentiate into a different cell type or revert to a less differentiated cell type. De-differentiation refers to the process by which a cell reverts to a less specialized (or committed) position within the lineage of a cell. As used herein, the lineage of a cell defines the heredity of the cell, i.e. which cells it came from and what cells it can give rise to. The lineage of a cell places the cell within a hereditary scheme of development and differentiation.

[00046] In a broad sense, a progenitor cell is a cell that has the capacity to create progeny that are more differentiated than itself, and yet retains the capacity to replenish the pool of progenitors. By that definition, stem cells themselves are also progenitor cells, as are the more immediate precursors to terminally differentiated cells. When referring to the cells of the present invention, as described in greater detail below, this broad definition of progenitor cell may be used. In a narrower sense, a progenitor cell is often defined as a cell that is intermediate in the differentiation pathway, i.e., it arises from a stem cell and is intermediate in the production of a mature cell type or subset of cell types. This type of progenitor cell is generally not able to self-renew. Accordingly, if this type of cell is referred to herein, it will be referred to as a non-renewing progenitor cell or as an intermediate progenitor or precursor cell.

[00047] As used herein, the phrase "differentiates into an ocular lineage or phenotype" refers to a cell that becomes partially or fully committed to a specific ocular phenotype, including without limitation, retinal and corneal stem cells, pigment epithelial cells of the retina and iris, photoreceptors, retinal ganglia and other optic neural lineages (e.g., retinal glia, microglia, astrocytes, Mueller cells), cells forming the crystalline lens, and epithelial cells of the sclera, cornea, limbus and conjunctiva. The phrase "differentiates into a neural lineage or phenotype" refers to a cell that becomes partially or fully committed to a specific neural phenotype of the CNS or PNS, i.e., a neuron or a glial cell, the latter category including without limitation astrocytes, oligodendrocytes, Schwann cells and microglia.

[00048] The cells exemplified herein and preferred for use in the present invention are generally referred to as postpartum-derived cells (or PPDCs). They also may sometimes be referred to more specifically as umbilicus-derived cells or placenta-derived cells (UDCs or PDCs). In addition, the cells may be described as being stem or progenitor cells, the latter term being used in the broad sense. The term derived is used to indicate that the cells have been obtained from their biological source and grown or otherwise manipulated in vitro (e.g., cultured in a Growth Medium to expand the population and/or to produce a cell line). The in vitro manipulations of umbilical stem cells and placental stem cells and the unique features of the umbilicus-derived cells and placental-derived cells of the present invention are described in detail below. Cells isolated from postpartum placenta and umbilicus by other means is also considered suitable for use in the present invention. These other cells are referred to herein as postpartum cells (rather than postpartum-derived cells).

[00049] Various terms are used to describe cells in culture. Cell culture refers generally to cells taken from a living organism and grown under controlled conditions ("in culture" or "cultured"). A primary cell culture is a culture of cells, tissues, or organs taken directly from an organism(s) before the first subculture. Cells are expanded in culture when they are placed in a Growth Medium under conditions that facilitate cell growth and/or division, resulting in a larger population of the cells. When cells are expanded in culture, the rate of cell proliferation is sometimes measured by the amount of time needed for the cells to double in number. This is referred to as doubling time.

[00050] A cell line is a population of cells formed by one or more subcultivations of a primary cell culture. Each round of subculturing is referred to as a passage. When cells are subcultured, they are referred to as having been passaged. A specific population of cells, or a cell line, is sometimes referred to or characterized by the number of times it has been passaged. For example, a cultured cell population that has been passaged ten times may be referred to as a P10 culture. The primary culture, i.e., the first culture following the isolation of cells from tissue, is designated P0. Following the first subculture, the cells are described as a secondary culture (PI or passage 1). After the second subculture, the cells become a tertiary culture (P2 or passage 2), and so on. It will be understood by those of skill in the art that there may be many population doublings during the period of passaging; therefore the number of population doublings of a culture is greater than the passage number. The expansion of cells (i.e., the number of population doublings) during the period between passaging depends on many factors, including but not limited to the seeding density, substrate, medium, growth conditions, and time between passaging.

[00051] The term Growth Medium generally refers to a medium sufficient for the culturing of PPDCs. In particular, one presently preferred medium for the culturing of the cells of the invention in comprises Dulbecco's Modified Essential Media (also abbreviated DMEM herein). Particularly preferred is DMEM-low glucose (also DMEM-LG herein) (Invitrogen, Carlsbad, Calif). The DMEM-low glucose is preferably supplemented with 15% (v/v) fetal bovine serum (e.g. defined fetal bovine serum, Hyclone, Logan Utah),

antibiotics/antimycotics ((preferably 50-100 Units/milliliter penicillin, 50-100

microgram/milliliter streptomycin, and 0-0.25 microgram/milliliter amphotericin B;

Invitrogen, Carlsbad, Calif.)), and 0.001 % (v/v) 2-mercaptoethanol (Sigma, St. Louis Mo.). As used in the Examples below, Growth Medium refers to DMEM-low glucose with 15% fetal bovine serum and antibiotics/antimycotics (when penicillin/streptomycin are included, it is preferably at 50 U/ml and 50 microgram/ml respectively; when

penicillin/streptomycin/amphotericin are used, it is preferably at 100 U/ml, 100

microgram/ml and 0.25 microgram/ml, respectively). In some cases different growth media are used, or different supplementations are provided, and these are normally indicated in the text as supplementations to Growth Medium.

[00052] A conditioned medium is a medium in which a specific cell or population of cells has been cultured, and then removed. When cells are cultured in a medium, they may secrete cellular factors that can provide trophic support to other cells. Such trophic factors include, but are not limited to hormones, cytokines, extracellular matrix (ECM), proteins, vesicles, antibodies, and granules. The medium containing the cellular factors is the conditioned medium.

[00053] Generally, a trophic factor is defined as a substance that promotes survival, growth, differentiation, proliferation and/or maturation of a cell, or stimulates increased activity of a cell. The interaction between cells via trophic factors may occur between cells of different types. Cell interaction by way of trophic factors is found in essentially all cell types, and is a particularly significant means of communication among neural cell types. Trophic factors also can function in an autocrine fashion, i.e., a cell may produce trophic factors that affect its own survival, growth, differentiation, proliferation and/or maturation.

[00054] When referring to cultured vertebrate cells, the term senescence (also replicative senescence or cellular senescence) refers to a property attributable to finite cell cultures; namely, their inability to grow beyond a finite number of population doublings (sometimes referred to as Hayflick's limit). Although cellular senescence was first described using fibroblast-like cells, most normal human cell types that can be grown successfully in culture undergo cellular senescence. The in vitro lifespan of different cell types varies, but the maximum lifespan is typically fewer than 100 population doublings (this is the number of doublings for all the cells in the culture to become senescent and thus render the culture unable to divide). Senescence does not depend on chronological time, but rather is measured by the number of cell divisions, or population doublings, the culture has undergone.

[00055] The terms ocular, ophthalmic and optic are used interchangeably herein to define "of, or about, or related to the eye." The term ocular degenerative condition (or disorder) is an inclusive term encompassing acute and chronic conditions, disorders or diseases of the eye, inclusive of the neural connection between the eye and the brain, involving cell damage, degeneration or loss. An ocular degenerative condition may be age-related, or it may result from injury or trauma, or it may be related to a specific disease or disorder. Acute ocular degenerative conditions include, but are not limited to, conditions associated with cell death or compromise affecting the eye including conditions arising from cerebrovascular insufficiency, focal or diffuse brain trauma, diffuse brain damage, infection or inflammatory conditions of the eye, retinal tearing or detachment, intra-ocular lesions (contusion

penetration, compression, laceration) or other physical injury (e.g., physical or chemical burns). Chronic ocular degenerative conditions (including progressive conditions) include, but are not limited to, retinopathies and other retinal/macular disorders such as retinitis pigmentosa (RP), age-related macular degeneration (AMD), choroidal neovascular membrane (CNVM); retinopathies such as diabetic retinopathy, occlusive retinopathy, sickle cell retinopathy and hypertensive retinopathy, central retinal vein occlusion, stenosis of the carotid artery, optic neuropathies such as glaucoma and related syndromes; disorders of the lens and outer eye, e.g., limbal stem cell deficiency (LSCD), also referred to as limbal epithelial cell deficiency (LECD), such as occurs in chemical or thermal injury, Steven- Johnson syndrome, contact lens-induced keratopathy, ocular cicatricial pemphigoid, congenital diseases of aniridia or ectodermal dysplasia, and multiple endocrine deficiency- associated keratitis.

[00056] The term treating (or treatment of) an ocular degenerative condition refers to ameliorating the effects of, or delaying, halting or reversing the progress of, or delaying or preventing the onset of, an ocular degenerative condition as defined herein.

[00057] The term effective amount refers to a concentration or amount of a reagent or pharmaceutical composition, such as a growth factor, differentiation agent, trophic factor, cell population or other agent, that is effective for producing an intended result, including cell growth and/or differentiation in vitro or in vivo, or treatment of ocular degenerative conditions, as described herein. With respect to growth factors, an effective amount may range from about 1 nanogram/milliliter to about 1 microgram/milliliter. With respect to PPDCs as administered to a patient in vivo, an effective amount may range from as few as several hundred or fewer, to as many as several million or more. In specific embodiments, an effective amount may range from 10 3 to l l 11 , more specifically at least about 10 4 cells. It will be appreciated that the number of cells to be administered will vary depending on the specifics of the disorder to be treated, including but not limited to size or total volume/surface area to be treated, as well as proximity of the site of administration to the location of the region to be treated, among other factors familiar to the medicinal biologist.

[00058] The terms effective period (or time) and effective conditions refer to a period of time or other controllable conditions (e.g., temperature, humidity for in vitro methods), necessary or preferred for an agent or pharmaceutical composition to achieve its intended result.

[00059] The term patient or subject refers to animals, including mammals, preferably humans, who are treated with the pharmaceutical compositions or in accordance with the methods described herein.

[00060] The term pharmaceutically acceptable carrier (or medium), which may be used interchangeably with the term biologically compatible carrier or medium, refers to reagents, cells, compounds, materials, compositions, and/or dosage forms that are not only compatible with the cells and other agents to be administered therapeutically, but also are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other complication commensurate with a reasonable benefit/risk ratio.

[00061] Several terms are used herein with respect to cell replacement therapy. The terms autologous transfer, autologous transplantation, autograft and the like refer to treatments wherein the cell donor is also the recipient of the cell replacement therapy. The terms allogeneic transfer, allogeneic transplantation, allograft and the like refer to treatments wherein the cell donor is of the same species as the recipient of the cell replacement therapy, but is not the same individual. A cell transfer in which the donor's cells and have been histocompatibly matched with a recipient is sometimes referred to as a syngeneic transfer. The terms xenogeneic transfer, xenogeneic transplantation, xenograft and the like refer to treatments wherein the cell donor is of a different species than the recipient of the cell replacement therapy. Transplantation as used herein refers to the introduction of autologous, or allogeneic donor cell replacement therapy into a recipient.

[00062] As used herein, the term "about" when referring to a measurable value such as an amount, a temporal duration, and the like, is meant to encompass variations of between ± 20% and ± 0.1%, preferably ± 20% or ± 10%, more preferably ± 5%, even more preferably ± 1%), and still more preferably ± 0.1% from the specified value, as such variations are appropriate to perform the disclosed methods.

Description

[00063] Ocular degenerative conditions, which encompass acute, chronic and progressive disorders and diseases having divergent causes, have as a common feature the dysfunction or loss of a specific or vulnerable group of ocular cells. This commonality enables development of similar therapeutic approaches for the repair or regeneration of vulnerable, damaged or lost ocular tissue or cells, one of which is cell-based therapy. Development of cell therapy for ocular degenerative conditions has been limited to a comparatively few types of stem or progenitor cells, including ocular-derived stem cells themselves (e.g., retinal and corneal stem cells), embryonic stem cells and a few types of adult stem or progenitor cells (e.g., neural, mucosal epithelial and bone marrow stem cells). Cells isolated from the postpartum umbilical cord and placenta have been identified as a significant new source of progenitor cells for this purpose. (US 2005-0037491 and US 2010-0272803). Moreover, conditioned media generated from cells isolated from the postpartum placenta and umbilical cord tissue provides another new source for treating ocular degenerative conditions. Accordingly, in its various embodiments described herein, the present invention features methods and pharmaceutical compositions for (repair and regeneration of ocular tissues), which use conditioned media from progenitor cells, such as cells isolated from postpartum umbilical cord or placenta. The invention is applicable to ocular degenerative conditions, but is expected to be particularly suitable for a number of ocular disorders for which treatment or cure has been difficult or unavailable. These include, without limitation, age-related macular degeneration, retinitis pigmentosa, diabetic and other retinopathies.

[00064] Conditioned media derived from progenitor cells, such as cells isolated from postpartum umbilical cord or placenta, in accordance with any method known in the art is expected to be suitable for use in the present invention. In one embodiment, however, the invention uses conditioned media derived from umbilical cord tissue-derived cells (hUTCs) or placental-tissue derived cells (PDCs) as defined above, which are derived from umbilical cord tissue or placenta that has been rendered substantially free of blood, preferably in accordance with the method set forth below. The hUTCs or PDCs are capable of expansion in culture and have the potential to differentiate into cells of other phenotypes. Certain embodiments feature conditioned media prepared from such progenitor cells, pharmaceutical compositions comprising the conditioned media, and methods of using the pharmaceutical compositions for treatment of patients with acute or chronic ocular degenerative conditions. The postpartum-derived cells of the present invention have been characterized by their growth properties in culture, by their cell surface markers, by their gene expression, by their ability to produce certain biochemical trophic factors, and by their immunological properties. The conditioned media derived from the postpartum-derived cells have been characterized by the trophic factors secreted by the cells.

Preparation of cells

[00065] The cells, cell populations and preparations comprising cell lysates, conditioned media and the like, used in the compositions and methods of the present invention are described herein, and in detail in U.S. Patent Nos. 7,524,489, and 7,510,873, and U.S. Pub. App. No. 2005/0058631, each incorporated by reference herein. According to the methods using postpartum cells, a mammalian umbilical cord and placenta are recovered upon or shortly after termination of either a full-term or pre-term pregnancy, for example, after expulsion of after-birth. The postpartum tissue may be transported from the birth site to a laboratory in a sterile container such as a flask, beaker, culture dish, or bag. The container may have a solution or medium, including but not limited to a salt solution, such as, for example, Dulbecco's Modified Eagle's Medium (DMEM) or phosphate buffered saline (PBS), or any solution used for transportation of organs used for transplantation, such as University of Wisconsin solution or perfluorochemical solution. One or more antibiotic and/or antimycotic agents, such as but not limited to penicillin, streptomycin, amphotericin B, gentamicin, and nystatin, may be added to the medium or buffer. The postpartum tissue may be rinsed with an anticoagulant solution such as heparin-containing solution. It is preferable to keep the tissue at about 4-10° C prior to extraction of PPDCs. It is even more preferable that the tissue not be frozen prior to extraction of PPDCs.

[00066] Isolation of PPDCs preferably occurs in an aseptic environment. The umbilical cord may be separated from the placenta by means known in the art. Alternatively, the umbilical cord and placenta are used without separation. Blood and debris are preferably removed from the postpartum tissue prior to isolation of PPDCs. For example, the postpartum tissue may be washed with buffer solution, such as but not limited to phosphate buffered saline. The wash buffer also may comprise one or more antimycotic and/or antibiotic agents, such as but not limited to penicillin, streptomycin, amphotericin B, gentamicin, and nystatin.

[00067] Postpartum tissue comprising a whole placenta or umbilical cord, or a fragment or section thereof is disaggregated by mechanical force (mincing or shear forces). In a presently preferred embodiment, the isolation procedure also utilizes an enzymatic digestion process. Many enzymes are known in the art to be useful for the isolation of individual cells from complex tissue matrices to facilitate growth in culture. Ranging from weakly digestive (e.g. deoxyribonucleases and the neutral protease, dispase) to strongly digestive (e.g. papain and trypsin), such enzymes are available commercially. A nonexhaustive list of enzymes compatible herewith includes mucolytic enzyme activities, metalloproteases, neutral proteases, serine proteases (such as trypsin, chymotrypsin, or elastase), and

deoxyribonucleases. Presently preferred are enzyme activities selected from metalloproteases, neutral proteases and mucolytic activities. For example, collagenases are known to be useful for isolating various cells from tissues. Deoxyribonucleases can digest singlestranded DNA and can minimize cell clumping during isolation. Preferred methods involve enzymatic treatment with for example collagenase and dispase, or collagenase, dispase, and

hyaluronidase, and such methods are provided wherein in certain preferred embodiments, a mixture of collagenase and the neutral protease dispase are used in the dissociating step. More preferred are those methods that employ digestion in the presence of at least one collagenase from Clostridium histolyticum, and either of the protease activities, dispase and thermo lysin. Still more preferred are methods employing digestion with both collagenase and dispase enzyme activities. Also preferred are methods that include digestion with a hyaluronidase activity in addition to collagenase and dispase activities. The skilled artisan will appreciate that many such enzyme treatments are known in the art for isolating cells from various tissue sources. For example, the LIB ERASE Blendzyme 3 (Roche) series of enzyme combinations are suitable for use in the instant methods. Other sources of enzymes are known, and the skilled artisan may also obtain such enzymes directly from their natural sources. The skilled artisan is also well equipped to assess new, or additional enzymes or enzyme combinations for their utility in isolating the cells of the invention. Preferred enzyme treatments are 0.5, 1, 1.5, or 2 hours long or longer. In other preferred embodiments, the tissue is incubated at 37° C during the enzyme treatment of the dissociation step.

[00068] In some embodiments of the invention, postpartum tissue is separated into sections comprising various aspects of the tissue, such as neonatal, neonatal/maternal, and maternal aspects of the placenta, for instance. The separated sections then are dissociated by mechanical and/or enzymatic dissociation according to the methods described herein. Cells of neonatal or maternal lineage may be identified by any means known in the art, for example, by karyotype analysis or in situ hybridization for a Y chromosome.

[00069] Isolated cells or postpartum tissue from which PPDCs grow out may be used to initiate, or seed, cell cultures. Isolated cells are transferred to sterile tissue culture vessels either uncoated or coated with extracellular matrix or ligands such as laminin, collagen (native, denatured or crosslinked), gelatin, fibronectin, and other extracellular matrix proteins. PPDCs are cultured in any culture medium capable of sustaining growth of the cells such as, but not limited to, DMEM (high or low glucose), advanced DMEM, DMEM/MCDB 201, Eagle's basal medium, Ham's F10 medium (F10), Ham's F-12 medium (F12), Iscove's modified Dulbecco's medium, Mesenchymal Stem Cell Growth Medium (MSCGM), DMEM/F12, RPMI 1640, and cellgro FREE™. The culture medium may be supplemented with one or more components including, for example, fetal bovine serum (FBS), preferably about 2-15% (v/v); equine serum (ES); human serum (HS); beta-mercaptoethanol (BME or 2- ME), preferably about 0.001% (v/v); one or more growth factors, for example, platelet- derived growth factor (PDGF), epidermal growth factor (EGF), fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), insulin-like growth factor- 1 (IGF-1), leukocyte inhibitory factor (LIF) and erythropoietin; amino acids, including L-valine; and one or more antibiotic and/or antimycotic agents to control microbial contamination, such as, for example, penicillin G, streptomycin sulfate, amphotericin B, gentamicin, and nystatin, either alone or in combination. The culture medium preferably comprises Growth Medium

(DMEM-low glucose, serum, BME, and an antibiotic agent).

[00070] The cells are seeded in culture vessels at a density to allow cell growth. In a preferred embodiment, the cells are cultured at about 0 to about 5 percent by volume CO 2 in air. In some preferred embodiments, the cells are cultured at about 2 to about 25 percent 0 2 in air, preferably about 5 to about 20 percent 0 2 in air. The cells preferably are cultured at about 25 to about 40° C and more preferably are cultured at 37° C. The cells are preferably cultured in an incubator. The medium in the culture vessel can be static or agitated, for example, using a bioreactor. PPDCs preferably are grown under low oxidative stress (e.g., with addition of glutathione, Vitamin C, Catalase, Vitamin E, N-Acetylcysteine). "Low oxidative stress", as used herein, refers to conditions of no or minimal free radical damage to the cultured cells.

[00071] Methods for the selection of the most appropriate culture medium, medium preparation, and cell culture techniques are well known in the art and are described in a variety of sources, including Doyle et al, (eds.), 1995, CELL & TISSUE CULTURE:

LABORATORY PROCEDURES, John Wiley & Sons, Chichester; and Ho and Wang (eds ), 1991, ANFMAL CELL BIOREACTORS, Butterworth-Heinemann, Boston, which are incorporated herein by reference.

[00072] After culturing the isolated cells or tissue fragments for a sufficient period of time, PPDCs will have grown out, either as a result of migration from the postpartum tissue or cell division, or both. In some embodiments of the invention, PPDCs are passaged, or removed to a separate culture vessel containing fresh medium of the same or a different type as that used initially, where the population of cells can be mitotically expanded. The cells of the invention may be used at any point between passage 0 and senescence. The cells preferably are passaged between about 3 and about 25 times, more preferably are passaged about 4 to about 12 times, and preferably are passaged 10 or 11 times. Cloning and/or subcloning may be performed to confirm that a clonal population of cells has been isolated.

[00073] In some aspects of the invention, the different cell types present in postpartum tissue are fractionated into subpopulations from which the PPDCs can be isolated. This may be accomplished using standard techniques for cell separation including, but not limited to, enzymatic treatment to dissociate postpartum tissue into its component cells, followed by cloning and selection of specific cell types, for example but not limited to selection based on morphological and/or biochemical markers; selective growth of desired cells (positive selection), selective destruction of unwanted cells (negative selection); separation based upon differential cell agglutinability in the mixed population as, for example, with soybean agglutinin; freeze-thaw procedures; differential adherence properties of the cells in the mixed population; filtration; conventional and zonal centrifugation; centrifugal elutriation (counter- streaming centrifugation); unit gravity separation; countercurrent distribution;

electrophoresis; and fluorescence activated cell sorting (FACS). For a review of clonal selection and cell separation techniques, see Freshney, 1994, CULTURE OF ANIMAL CELLS: A MANUAL OF BASIC TECHNIQUES, 3rd Ed., Wiley-Liss, Inc., New York, which is incorporated herein by reference.

[00074] The culture medium is changed as necessary, for example, by carefully aspirating the medium from the dish, for example, with a pipette, and replenishing with fresh medium. Incubation is continued until a sufficient number or density of cells accumulates in the dish. The original explanted tissue sections may be removed and the remaining cells trypsinized using standard techniques or using a cell scraper. After trypsinization, the cells are collected, removed to fresh medium and incubated as above. In some embodiments, the medium is changed at least once at approximately 24 hours post-trypsinization to remove any floating cells. The cells remaining in culture are considered to be PPDCs.

[00075] PPDCs may be cryopreserved. Accordingly, in a preferred embodiment described in greater detail below, PPDCs for autologous transfer (for either the mother or child) may be derived from appropriate postpartum tissues following the birth of a child, then cryopreserved so as to be available in the event they are later needed for transplantation.

Characteristics of cells

[00076] The progenitor cells of the invention, such as PPDCs, may be characterized, for example, by growth characteristics (e.g., population doubling capability, doubling time, passages to senescence), karyotype analysis (e.g., normal karyotype; maternal or neonatal lineage), flow cytometry (e.g., FACS analysis), immunohistochemistry and/or

immunocytochemistry (e.g., for detection of epitopes), gene expression profiling (e.g., gene chip arrays; polymerase chain reaction (for example, reverse transcriptase PCR, real time PCR, and conventional PCR)), protein arrays, protein secretion (e.g., by plasma clotting assay or analysis of PDC-conditioned medium, for example, by Enzyme Linked Immunosorbent Assay (ELISA)), mixed lymphocyte reaction (e.g., as measure of stimulation of PBMCs), and/or other methods known in the art.

[00077] Examples of PPDCs derived from umbilicus tissue were deposited with the American Type Culture Collection on (ATCC, 10801 University Boulevard, Manassas, VA, 20110) Jun. 10, 2004, and assigned ATCC Accession Numbers as follows: (1) strain designation UMB 022803 (P7) was assigned Accession No. PTA-6067; and (2) strain designation UMB 022803 (PI 7) was assigned Accession No. PTA-6068. Examples of PPDCs derived from placental tissue were deposited with the ATCC (Manassas, Va.) and assigned ATCC Accession Numbers as follows: (1) strain designation PLA 071003 (P8) was deposited Jun. 15, 2004 and assigned Accession No. PTA-6074; (2) strain designation PLA 071003 (PI 1) was deposited June 15, 2004 and assigned Accession No. PTA-6075; and (3) strain designation PLA 071003 (PI 6) was deposited Jun. 16, 2004 and assigned Accession No. PTA-6079.

[00078] In various embodiments, the PPDCs possess one or more of the following growth features: (1) they require L-v aline for growth in culture; (2) they are capable of growth in atmospheres containing oxygen from about 5% to at least about 20%; (3) they have the potential for at least about 40 doublings in culture before reaching senescence; and (4) they attach and expand on a coated or uncoated tissue culture vessel, wherein the coated tissue culture vessel comprises a coating of gelatin, laminin, collagen, polyomithine, vitronectin or fibronectin.

[00079] In certain embodiments the PPDCs possess a normal karyotype, which is maintained as the cells are passaged. Karyotyping is particularly useful for identifying and distinguishing neonatal from maternal cells derived from placenta. Methods for karyotyping are available and known to those of skill in the art.

[00080] In other embodiments, the PPDCs may be characterized by production of certain proteins, including: (1) production of at least one of vimentin and alpha-smooth muscle actin; and (2) production of at least one of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD- L2 and HLA-A,B,C cell surface markers, as detected by flow cytometry. In other

embodiments, the PPDCs may be characterized by lack of production of at least one of CD31, CD34, CD45, CD80, CD86, CD117, CD141, CD178, B7-H2, HLA-G, and HLA- DR,DP,DQ cell surface markers, as detected by flow cytometry. Particularly preferred are cells that produce vimentin and alpha-smooth muscle actin.

[00081] In other embodiments, the PPDCs may be characterized by gene expression, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an iliac crest bone marrow cell, is increased for a gene encoding at least one of interleukin 8; reticulon 1; chemokine (C--X--C motif) ligand 1 (melonoma growth stimulating activity, alpha);

chemokine (C--X--C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C--X-- C motif) ligand 3; tumor necrosis factor, alpha-induced protein 3; C-type lectin superfamily member 2; Wilms tumor 1; aldehyde dehydrogenase 1 family member A2; renin; oxidized low density lipoprotein receptor 1; Homo sapiens clone IMAGE:4179671; protein kinase C zeta; hypothetical protein DKFZp564F013; downregulated in ovarian cancer 1; and Homo sapiens gene from clone DKFZp547kl 113. In an embodiment, the PPDCs derived from umbilical cord tissue may be characterized by gene expression, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an iliac crest bone marrow cell, is increased for a gene encoding at least one of interleukin 8; reticulon 1; or chemokine (C--X--C motif) ligand 3. In another embodiment, the PPDCs derived from placental tissue may be characterized by gene expression, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an iliac crest bone marrow cell, is increased for a gene encoding at least one of renin or oxidized low density lipoprotein receptor 1.

[00082] In yet other embodiments, the PPDCs may be characterized by gene expression, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an iliac crest bone marrow cell, is reduced for a gene encoding at least one of: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C--X--C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeo box 2 (growth arrest-specific homeo box); sine oculis homeobox homolog 1 (Drosophila);

crystallin, alpha B; disheveled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1; tetranectin (plasminogen binding protein); src homology three (SH3) and cysteine rich domain; cholesterol 25-hydroxylase; runt-related transcription factor 3; interleukin 11 receptor, alpha; procollagen C-endopeptidase enhancer; frizzled homolog 7 (Drosophila); hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C

(hexabrachion); iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; neuroblastoma, suppression of turn origeni city 1; insulin-like growth factor binding protein 2, 36 kDa; Homo sapiens cDNA FLJ12280 fis, clone MAMMA1001744; cytokine receptor-like factor 1; potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, beta 7; transcriptional co-activator with PDZ- binding motif (TAZ); sine oculis homeobox homolog 2 (Drosophila); KIAAI034 protein; vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like

extracellular matrix protein 1; early growth response 3; distal-less homeo box 5; hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3 -alpha hydroxy steroid dehydrogenase, type II); biglycan; transcriptional co-activator with PDZ-binding motif (TAZ); fibronectin 1; proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens mRNA full length insert cDNA clone EUROFMAGE 1968422; EphA3;

KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA; cDNA

DKFZp564B222 (from clone DKFZp564B222); BCL2/adenovirus E1B 19 kDa interacting protein 3 -like; AE binding protein 1; and cytochrome c oxidase subunit Vila polypeptide 1 (muscle).

[00083] In other embodiments, the PPDCs derived from umbilical cord tissue may be characterized by secretion of trophic factors selected from thrombospondin-1,

thrombospondin-2, and thrombospondin-4. In embodiments, the PPDCs may be

characterized by secretion of at least one of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MlPlb, 1309, RANTES, MDC, and TFMPl . In some embodiments, the PPDCs derived from umbilical cord tissue may be characterized by lack of secretion of at least one of TGF-beta2, ANG2, PDGFbb, MlPla and VEGF, as detected by ELISA. In alternative embodiments, PPDCs derived from placenta tissue may be characteristics by secretion of at least one of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, HB-EGF, BDNF, TPO, MlPla, RANTES, and TFMPl, and lack of secretion of at least one of TGF-beta2, ANG2, PDGFbb, FGF, and VEGF, as detected by ELISA. In further embodiments, the PPDCs lack expression of hTERT or telomerase.

[00084] In preferred embodiments, the cell comprises two or more of the above-listed growth, protein/surface marker production, gene expression or substance-secretion characteristics. More preferred are those cells comprising, three, four, or five or more of the characteristics. Still more preferred are PPDCs comprising six, seven, or eight or more of the characteristics. Still more preferred presently are those cells comprising all of above characteristics.

[00085] In particularly preferred embodiments, the cells isolated from human umbilical cord tissue substantially free of blood, which are capable of expansion in culture, lack the production of CD117 or CD45, and do not express hTERT or telomerase. In one

embodiment, the cells lack production of CD117 and CD45 and, optionally, also do not express hTERT and telomerase. In another embodiment, the cells do not express hTERT and telomerase. In yet another embodiment, the cells are isolated from human umbilical cord tissue substantially free of blood, are capable of expansion in culture, lack the production of CD117 or CD45, and do not express hTERT or telomerase, and have one or more of the following characteristics: express CD10, CD13, CD44, CD73, and CD90; do not express CD31 or CD34; express, relative to a human fibroblast, mesenchymal stem cell, or iliac crest bone marrow cell, increased levels of interleukin 8 or reticulon 1; and have the potential to differentiate.

[00086] Among cells that are presently preferred for use with the invention in several of its aspects are postpartum cells having the characteristics described above and more particularly those wherein the cells have normal karyotypes and maintain normal karyotypes with passaging, and further wherein the cells express each of the markers CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, and HLA-A,B,C, wherein the cells produce the

immunologically-detectable proteins which correspond to the listed markers. Still more preferred are those cells which in addition to the foregoing do not produce proteins corresponding to any of the markers CD31, CD34, CD45, CD117, CD141, or HLA- DR,DP,DQ, as detected by flow cytometry. In further preferred embodiments, the cells lack expression of hTERT or telomerase.

[00087] Certain cells having the potential to differentiate along lines leading to various phenotypes are unstable and thus can spontaneously differentiate. Presently preferred for use with the invention are cells that do not spontaneously differentiate, for example along neural lines. Preferred cells, when grown in Growth Medium, are substantially stable with respect to the cell markers produced on their surface, and with respect to the expression pattern of various genes, for example as determined using an Affymetrix GE ECHIP. The cells remain substantially constant, for example in their surface marker characteristics over passaging, through multiple population doublings.

[00088] However, one feature of PPDCs is that they may be deliberately induced to differentiate into various lineage phenotypes by subjecting them to differentiation-inducing cell culture conditions. Of use in treatment of certain ocular degenerative conditions, the PPDCs may be induced to differentiate into neural phenotypes using one or more methods known in the art. For instance, as exemplified herein, PPDCs may be plated on flasks coated with laminin in Neurobasal-A medium (Invitrogen, Carlsbad, Calif.) containing B27 (B27 supplement, Invitrogen), L-glutamine and Penicillin/Streptomycin, the combination of which is referred to herein as Neural Progenitor Expansion (NPE) medium. NPE media may be further supplemented with bFGF and/or EGF. Alternatively, PPDCs may be induced to differentiate in vitro by: (1) co-culturing the PPDCs with neural progenitor cells; or (2) growing the PPDCs in neural progenitor cell-conditioned medium.

[00089] Differentiation of the PPDCs into neural phenotypes may be demonstrated by a bipolar cell morphology with extended processes. The induced cell populations may stain positive for the presence of nestin. Differentiated PPDCs may be assessed by detection of nest in, TuJl (Bill tubulin), GFAP, tyrosine hydroxylase, GABA, 04 and/or MBP. In some embodiments, PPDCs have exhibited the ability to form three-dimensional bodies characteristic of neuronal stem cell formation of neurospheres.

Cell Populations

[00090] Another aspect of the invention features populations of progenitor cells, such as postpartum-derived cells, or other progenitor cells. The postpartum-derived cells may be isolated from placental or umbilical tissue. In a preferred embodiment, the cell populations comprise the PPDCs described above, and these cell populations are described in the section below.

[00091] In some embodiments, the cell population is heterogeneous. A heterogeneous cell population of the invention may comprise at least about 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 95% of the cell. The heterogeneous cell populations of the invention may further comprise the progenitor cells (postpartum-derived cells), or other progenitor cells, such as epithelial or neural progenitor cells, or it may further comprise fully differentiated cells.

[00092] In some embodiments, the population is substantially homogeneous, i.e., comprises substantially only PPDCs (preferably at least about 96%, 97%, 98%>, 99% or more of the cells). In some embodiments, the cell population is homogeneous. In embodiments, the homogeneous cell population of the invention may comprise umbilicus- or placenta- derived cells. Homogeneous populations of umbilicus-derived cells are preferably free of cells of maternal lineage. Homogeneous populations of placenta-derived cells may be of neonatal or maternal lineage. Homogeneity of a cell population may be achieved by any method known in the art, for example, by cell sorting (e.g., flow cytometry) or by clonal expansion in accordance with known methods. Thus, preferred homogeneous PPDC populations may comprise a clonal cell line of postpartum-derived cells. Such populations are particularly useful when a cell clone with highly desirable functionality has been isolated.

[00093] Also provided herein are populations of cells incubated in the presence of one or more factors, or under conditions, that stimulate stem cell differentiation along a desired pathway (e.g., neural, epithelial). Such factors are known in the art and the skilled artisan will appreciate that determination of suitable conditions for differentiation can be accomplished with routine experimentation. Optimization of such conditions can be accomplished by statistical experimental design and analysis, for example response surface methodology allows simultaneous optimization of multiple variables, for example in a biological culture. Presently preferred factors include, but are not limited to factors, such as growth or trophic factors, demethylating agents, co-culture with neural or epithelial lineage cells or culture in neural or epithelial lineage cell-conditioned medium, as well other conditions known in the art to stimulate stem cell differentiation along these pathways (for factors useful in neural differentiation, see, e.g., Lang, K. J. D. et al, 2004, J. Neurosci. Res. 76: 184-192; Johe, K. K. et al, 1996, Genes Devel. 10: 3129-3140; Gottleib, D., 2002, Ann. Rev. Neurosci. 25: 381-407).

Conditioned Medium

[00094] In one aspect, the invention provides conditioned medium from cultured progenitor cells, such as postpartum-derived cells, or other progenitor cells, for use in vitro and in vivo as described below. Use of such conditioned medium allows the beneficial trophic factors secreted by the cells to be used allogeneically in a patient without introducing intact cells that could trigger rejection, or other adverse immunological responses.

Conditioned medium is prepared by culturing cells (such as a population of cells) in a culture medium, then removing the cells from the medium. In certain embodiments, the postpartum cells are UTCs or PDCs, more preferably hUTCs.

[00095] Conditioned medium prepared from populations of cells as described above may be used as is, further concentrated, by for example, ultrafiltration or lyophilization, or even dried, partially purified, combined with pharmaceutically-acceptable carriers or diluents as are known in the art, or combined with other compounds such as biologicals, for example pharmaceutically useful protein compositions. Conditioned medium may be used in vitro or in vivo, alone or for example, with autologous or syngeneic live cells. The conditioned medium, if introduced in vivo, may be introduced locally at a site of treatment, or remotely to provide, for example needed cellular growth or trophic factors to a patient.

[00096] Previously, it has been demonstrated that human umbilical cord tissue-derived cells improved visual function and ameliorated retinal degeneration (US 2010/0272803). It also has been demonstrated that postpartum-derived cells can be used to promote

photoreceptor rescue and thus preserve photoreceptors in a RCS model. (US 2010/0272803). Injection of hUTC subretinally into RCS rat eye improved visual acuity and ameliorated retinal degeneration. Moreover, treatment with conditioned medium (CM) derived from hUTC restored phagocytosis of ROS in dystrophic RPE cells in vitro. (US 2010/0272803). Here, embodiments of the invention disclose the positive effect of hUTCs to reduce retinal neovascularization, in particular in diabetic retinopathy.

Cell Modifications, Components and Products

[00097] Progenitor cells, such as postpartum cells, preferably PPDCs, may also be genetically modified to produce therapeutically useful gene products, or to produce antineoplastic agents for treatment of tumors. Genetic modification may be accomplished using any of a variety of vectors including, but not limited to, integrating viral vectors, e.g., retrovirus vector or adeno-associated viral vectors; non-integrating replicating vectors, e.g., papilloma virus vectors, SV40 vectors, adenoviral vectors; or replication-defective viral vectors. Other methods of introducing DNA into cells include the use of liposomes, electroporation, a particle gun, or by direct DNA injection.

[00098] Hosts cells are preferably transformed or transfected with DNA controlled by or in operative association with, one or more appropriate expression control elements such as promoter or enhancer sequences, transcription terminators, polyadenylation sites, among others, and a selectable marker. Any promoter may be used to drive the expression of the inserted gene. For example, viral promoters include, but are not limited to, the CMV promoter/enhancer, SV40, papillomavirus, Epstein-Barr virus or elastin gene promoter. In some embodiments, the control elements used to control expression of the gene of interest can allow for the regulated expression of the gene so that the product is synthesized only when needed in vivo. If transient expression is desired, constitutive promoters are preferably used in a non-integrating and/or replication-defective vector. Alternatively, inducible promoters could be used to drive the expression of the inserted gene when necessary.

Inducible promoters include, but are not limited to those associated with metallothionein and heat shock proteins.

[00099] Following the introduction of the foreign DNA, engineered cells may be allowed to grow in enriched media and then switched to selective media. The selectable marker in the foreign DNA confers resistance to the selection and allows cells to stably integrate the foreign DNA as, for example, on a plasmid, into their chromosomes and grow to form foci which, in turn, can be cloned and expanded into cell lines. This method can be

advantageously used to engineer cell lines that express the gene product.

[000100] Cells may be genetically engineered to "knock out" or "knock down" expression of factors that promote inflammation or rejection at the implant site. Negative modulatory techniques for the reduction of target gene expression levels or target gene product activity levels are discussed below. "Negative modulation," as used herein, refers to a reduction in the level and/or activity of target gene product relative to the level and/or activity of the target gene product in the absence of the modulatory treatment. The expression of a gene native to a neuron or glial cell can be reduced or knocked out using a number of techniques including, for example, inhibition of expression by inactivating the gene using the homologous recombination technique. Typically, an exon encoding an important region of the protein (or an exon 5' to that region) is interrupted by a positive selectable marker, e.g., neo, preventing the production of normal mRNA from the target gene and resulting in inactivation of the gene. A gene may also be inactivated by creating a deletion in part of a gene, or by deleting the entire gene. By using a construct with two regions of homology to the target gene that are far apart in the genome, the sequences intervening the two regions can be deleted

(Mombaerts et al, 1991, Proc. Nat. Acad. Sci. U.S.A. 88:3084-3087). Antisense,

DNAzymes, ribozymes, small interfering RNA (siRNA) and other such molecules that inhibit expression of the target gene can also be used to reduce the level of target gene activity. For example, antisense RNA molecules that inhibit the expression of major histocompatibility gene complexes (HLA) have been shown to be most versatile with respect to immune responses. Still further, triple helix molecules can be utilized in reducing the level of target gene activity. These techniques are described in detail by L. G. Davis et al. (eds), 1994, BASIC METHODS IN MOLECULAR BIOLOGY, 2nd ed., Appleton & Lange, Norwalk, CT.

[000101] In other aspects, the invention provides cell lysates and cell soluble fractions prepared from postpartum stem cells, preferably PPDCs, or heterogeneous or homogeneous cell populations comprising PPDCs, as well as PPDCs or populations thereof that have been genetically modified or that have been stimulated to differentiate along a neurogenic pathway. Such lysates and fractions thereof have many utilities. Use of the cell lysate soluble fraction (i.e., substantially free of membranes) in vivo, for example, allows the beneficial intracellular milieu to be used allogeneically in a patient without introducing an appreciable amount of the cell surface proteins most likely to trigger rejection, or other adverse immunological responses. Methods of lysing cells are well known in the art and include various means of mechanical disruption, enzymatic disruption, or chemical disruption, or combinations thereof. Such cell lysates may be prepared from cells directly in their Growth Medium and thus containing secreted growth factors and the like, or may be prepared from cells washed free of medium in, for example, PBS or other solution. Washed cells may be resuspended at concentrations greater than the original population density if preferred.

[000102] In one embodiment, whole cell lysates are prepared, e.g., by disrupting cells without subsequent separation of cell fractions. In another embodiment, a cell membrane fraction is separated from a soluble fraction of the cells by routine methods known in the art, e.g., centrifugation, filtration, or similar methods.

[000103] Cell lysates or cell soluble fractions prepared from populations of progenitor cells, such as postpartum-derived cells, may be used as is, further concentrated, by for example, ultrafiltration or lyophilization, or even dried, partially purified, combined with

pharmaceutically-acceptable carriers or diluents as are known in the art, or combined with other compounds such as biologicals, for example pharmaceutically useful protein

compositions. Cell lysates or fractions thereof may be used in vitro or in vivo, alone or for example, with autologous or syngeneic live cells. The lysates, if introduced in vivo, may be introduced locally at a site of treatment, or remotely to provide, for example needed cellular growth factors to a patient. [000104] In a further embodiment, postpartum cells, preferably PPDCs, can be cultured in vitro to produce biological products in high yield. For example, such cells, which either naturally produce a particular biological product of interest (e.g., a trophic factor), or have been genetically engineered to produce a biological product, can be clonally expanded using the culture techniques described herein. Alternatively, cells may be expanded in a medium that induces differentiation to a desired lineage. In either case, biological products produced by the cell and secreted into the medium can be readily isolated from the conditioned medium using standard separation techniques, e.g., such as differential protein precipitation, ion- exchange chromatography, gel filtration chromatography, electrophoresis, and HPLC, to name a few. A "bioreactor" may be used to take advantage of the flow method for feeding, for example, a three-dimensional culture in vitro. Essentially, as fresh media is passed through the three-dimensional culture, the biological product is washed out of the culture and may then be isolated from the outflow, as above.

[000105] Alternatively, a biological product of interest may remain within the cell and, thus, its collection may require that the cells be lysed, as described above. The biological product may then be purified using anyone or more of the above-listed techniques.

[000106] In another embodiment, an extracellular matrix (ECM) produced by culturing postpartum cells (preferably PPDCs), on liquid, solid or semi-solid substrates is prepared, collected and utilized as an alternative to implanting live cells into a subject in need of tissue repair or replacement. The cells are cultured in vitro, on a three dimensional framework as described elsewhere herein, under conditions such that a desired amount of ECM is secreted onto the framework. The cells and the framework are removed, and the ECM processed for further use, for example, as an injectable preparation. To accomplish this, cells on the framework are killed and any cellular debris removed from the framework. This process may be carried out in a number of different ways. For example, the living tissue can be flash- frozen in liquid nitrogen without a cryopreservative, or the tissue can be immersed in sterile distilled water so that the cells burst in response to osmotic pressure.

[000107] Once the cells have been killed, the cellular membranes may be disrupted and cellular debris removed by treatment with a mild detergent rinse, such as EDTA, CHAPS or a zwitterionic detergent. Alternatively, the tissue can be enzymatically digested and/or extracted with reagents that break down cellular membranes and allow removal of cell contents. Example of such enzymes include, but are not limited to, hyaluronidase, dispase, proteases, and nucleases. Examples of detergents include non-ionic detergents such as, for example, alkylaryl polyether alcohol (TRITON X-100), octylphenoxy polyethoxy-ethanol (Rohm and Haas Philadelphia, Pa.), BRIJ-35, a polyethoxyethanollauryl ether (Atlas Chemical Co., San Diego, Calif.), polysorbate 20 (TWEEN 20), a polyethoxyethanol sorbitan mono laureate (Rohm and Haas), polyethylene lauryl ether (Rohm and Haas); and ionic detergents such as, for example, sodium dodecyl sulphate, sulfated higher aliphatic alcohols, sulfonated alkanes and sulfonated alkylarenes containing 7 to 22 carbon atoms in a branched or unbranched chain.

[000108] The collection of the ECM can be accomplished in a variety of ways, depending, for example, on whether the new tissue has been formed on a three-dimensional framework that is biodegradable or non-biodegradable. For example, if the framework is nonbiodegradable, the ECM can be removed by subjecting the framework to sonication, high- pressure water jets, mechanical scraping, or mild treatment with detergents or enzymes, or any combination of the above.

[000109] If the framework is biodegradable, the ECM can be collected, for example, by allowing the framework to degrade or dissolve in solution. Alternatively, if the biodegradable framework is composed of a material that can itself be injected along with the ECM, the framework and the ECM can be processed in toto for subsequent injection. Alternatively, the ECM can be removed from the biodegradable framework by any of the methods described above for collection of ECM from a non-biodegradable framework. All collection processes are preferably designed so as not to denature the ECM.

[000110] After it has been collected, the ECM may be processed further. For example, the ECM can be homogenized to fine particles using techniques well known in the art such as by sonication, so that it can pass through a surgical needle. The components of the ECM can be crosslinked, if desired, by gamma irradiation. Preferably, the ECM can be irradiated between 0.25 to 2 mega rads to sterilize and cross link the ECM. Chemical crosslinking using agents that are toxic, such as glutaraldehyde, is possible but not generally preferred.

[000111] The amounts and/or ratios of proteins, such as the various types of collagen present in the ECM, may be adjusted by mixing the ECM produced by the cells of the invention with ECM of one or more other cell types. In addition, biologically active substances such as proteins, growth factors and/or drugs, can be incorporated into the ECM. Exemplary biologically active substances include tissue growth factors, such as TGF-beta, and the like, which promote healing and tissue repair at the site of the injection. Such additional agents may be utilized in any of the embodiments described herein above, e.g., with whole cell lysates, soluble cell fractions, or further purified components and products produced by the cells.

Pharmaceutical Compositions

[000112] In another aspect, the invention provides pharmaceutical compositions that use non-embryronic stem cells such as postpartum cells (preferably PPDCs), cell populations thereof, conditioned media produced by such cells, and cell components and products produced by such cells in various methods for treatment of ocular degenerative conditions. Certain embodiments encompass pharmaceutical compositions comprising live cells (e.g., PPDCs alone or admixed with other cell types). Other embodiments encompass

pharmaceutical compositions comprising PPDC conditioned medium. Additional embodiments may use cellular components of PPDC (e.g., cell lysates, soluble cell fractions, ECM, or components of any of the foregoing) or products (e.g., trophic and other biological factors produced naturally by the cells or through genetic modification, conditioned medium from culturing the cells). In either case, the pharmaceutical composition may further comprise other active agents, such as anti-inflammatory agents, anti-apoptotic agents, antioxidants, growth factors, neurotrophic factors or neuroregenerative, neuroprotective or ophthalmic drugs as known in the art.

[000113] Examples of other components that may be added to the pharmaceutical compositions include, but are not limited to: (1) other neuroprotective or neurobeneficial drugs; (2) selected extracellular matrix components, such as one or more types of collagen known in the art, and/or growth factors, platelet-rich plasma, and drugs (alternatively, PPDCs may be genetically engineered to express and produce growth factors); (3) anti-apoptotic agents (e.g., erythropoietin (EPO), EPO mimetibody, thrombopoietin, insulin-like growth factor (IGF)-I, IGF-II, hepatocyte growth factor, caspase inhibitors); (4) anti-inflammatory compounds (e.g., p38 MAP kinase inhibitors, TGF-beta inhibitors, statins, IL-6 and IL-1 inhibitors, PEMIROLAST, TRANILAST, REMICADE, SIROLIMUS, and non-steroidal anti-inflammatory drugs (NSAIDS) (such as TEPOXALIN, TOLMETIN, and SUPROFEN); (5) immunosuppressive or immunomodulatory agents, such as calcineurin inhibitors, mTOR inhibitors, antiproliferatives, corticosteroids and various antibodies; (6) antioxidants such as probucol, vitamins C and E, conenzyme Q-10, glutathione, L-cysteine and N-acetyl cysteine; and (6) local anesthetics, to name a few. [000114] Pharmaceutical compositions of the invention comprise progenitor cells, such as postpartum cells (preferably PPDCs), conditioned media generated from those cells, or components or products thereof, formulated with a pharmaceutically acceptable carrier or medium. Suitable pharmaceutically acceptable carriers include water, salt solution (such as Ringer's solution), alcohols, oils, gelatins, and carbohydrates, such as lactose, amylose, or starch, fatty acid esters, hydroxymethylcellulose, and polyvinyl pyrolidine. Such

preparations can be sterilized, and if desired, mixed with auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, and coloring. Typically, but not exclusively, pharmaceutical compositions comprising cellular components or products, but not live cells, are formulated as liquids. Pharmaceutical compositions comprising PPDC live cells are typically formulated as liquids, semisolids (e.g., gels) or solids (e.g., matrices, scaffolds and the like, as appropriate for ophthalmic tissue engineering).

[000115] Pharmaceutical compositions may comprise auxiliary components as would be familiar to medicinal chemists or biologists. For example, they may contain antioxidants in ranges that vary depending on the kind of antioxidant used. Reasonable ranges for commonly used antioxidants are about 0.01 % to about 0.15% weight by volume of EDTA, about 0.01 % to about 2.0% weight volume of sodium sulfite, and about 0.01 % to about 2.0% weight by volume of sodium metabisulfite. One skilled in the art may use a

concentration of about 0.1 % weight by volume for each of the above. Other representative compounds include mercaptopropionyl glycine, N-acetyl cysteine, beta-mercaptoethylamine, glutathione and similar species, although other antioxidant agents suitable for ocular administration, e.g. ascorbic acid and its salts or sulfite or sodium metabisulfite may also be employed.

[000116] A buffering agent may be used to maintain the pH of eye drop formulations in the range of about 4.0 to about 8.0; so as to minimize irritation of the eye. For direct intravitreal or intraocular injection, formulations should be at pH 7.2 to 7.5, preferably at pH 7.3-7.4. The ophthalmologic compositions may also include tonicity agents suitable for administration to the eye. Among those suitable is sodium chloride to make formulations approximately isotonic with 0.9% saline solution.

[000117] In certain embodiments, pharmaceutical compositions are formulated with viscosity enhancing agents. Exemplary agents are hydroxyethylcellulose,

hydroxypropylcellulose, methylcellulose, and polyvinylpyrrolidone. The pharmaceutical compositions may have cosolvents added if needed. Suitable cosolvents may include glycerin, polyethylene glycol (PEG), polysorbate, propylene glycol, and polyvinyl alcohol. Preservatives may also be included, e.g., benzalkonium chloride, benzethonium chloride, chlorobutanol, phenylmercuric acetate or nitrate, thimerosal, or methyl or propylparabens.

[000118] Formulations for injection are preferably designed for single-use administration and do not contain preservatives. Injectable solutions should have isotonicity equivalent to 0.9% sodium chloride solution (osmolality of 290-300 milliosmoles). This may be attained by addition of sodium chloride or other co-solvents as listed above, or excipients such as buffering agents and antioxidants, as listed above.

[000119] The tissues of the anterior chamber of the eye are bathed by the aqueous humor, while the retina is under continuous exposure to the vitreous. These fluids/gels exist in a highly reducing redox state because they contain antioxidant compounds and enzymes.

Therefore, it may be advantageous to include a reducing agent in the ophthalmologic compositions. Suitable reducing agents include N-acetylcysteine, ascorbic acid or a salt form, and sodium sulfite or metabi sulfite, with ascorbic acid and/or N-acetylcysteine or glutathione being particularly suitable for injectable solutions.

[000120] Pharmaceutical compositions comprising cells or conditioned medium, or cell components or cell products may be delivered to the eye of a patient in one or more of several delivery modes known in the art. In one embodiment that may be suitable for use in some instances, the compositions are topically delivered to the eye in eye drops or washes. In another embodiment, the compositions may be delivered to various locations within the eye via periodic intraocular injection or by infusion in an irrigating solution such as BSS or BSS PLUS (Alcon USA, Fort Worth, Tex.). Alternatively, the compositions may be applied in other ophthalmologic dosage forms known to those skilled in the art, such as pre-formed or in situ-formed gels or liposomes, for example as disclosed in U.S. Pat. No. 5,718,922 to

Herrero-Vanrell. In another embodiment, the composition may be delivered to or through the lens of an eye in need of treatment via a contact lens (e.g. Lidofilcon B, Bausch & Lomb CW79 or DELTACON (Deltafilcon A) or other object temporarily resident upon the surface of the eye. In other embodiments, supports such as a collagen corneal shield (e.g. BIO-COR dissolvable corneal shields, Summit Technology, Watertown, Mass.) can be employed. The compositions can also be administered by infusion into the eyeball, either through a cannula from an osmotic pump (ALZET, Alza Corp., Palo Alto, Calif.) or by implantation of timed- release capsules (OCCUSENT) or biodegradable disks (OCULEX, OCUSERT). These routes of administration have the advantage of providing a continuous supply of the pharmaceutical composition to the eye. This may be an advantage for local delivery to the cornea.

[000121] Pharmaceutical compositions comprising live cells in a semi-solid or solid carrier are typically formulated for surgical implantation at the site of ocular damage or distress. It will be appreciated that liquid compositions also may be administered by surgical procedures, for example conditioned media. In particular embodiments, semi-solid or solid

pharmaceutical compositions may comprise semi-permeable gels, lattices, cellular scaffolds and the like, which may be non-biodegradable or biodegradable. For example, in certain embodiments, it may be desirable or appropriate to sequester the exogenous cells from their surroundings, yet enable the cells to secrete and deliver biological molecules to surrounding cells. In these embodiments, cells may be formulated as autonomous implants comprising living PPDCs or cell population comprising PPDCs surrounded by a non-degradable, selectively permeable barrier that physically separates the transplanted cells from host tissue. Such implants are sometimes referred to as "immunoprotective," as they have the capacity to prevent immune cells and macromolecules from killing the transplanted cells in the absence of pharmacologically induced immunosuppression (for a review of such devices and methods, see, e.g., P. A. Tresco et al, 2000, Adv. Drug Delivery Rev. 42: 3-27).

[000122] In other embodiments, different varieties of degradable gels and networks are utilized for the pharmaceutical compositions of the invention. For example, degradable materials particularly suitable for sustained release formulations include biocompatible polymers, such as poly (lactic acid), poly (lactic-co-glycolic acid), methylcellulose, hyaluronic acid, collagen, and the like. The structure, selection and use of degradable polymers in drug delivery vehicles have been reviewed in several publications, including, A. Domb et al, 1992, Polymers for Advanced Technologies 3 :279-291. U.S. Pat. No. 5,869,079 to Wong et al discloses combinations of hydrophilic and hydrophobic entities in a biodegradable sustained release ocular implant. In addition, U.S. Pat. No. 6,375,972 to Guo et al, U.S. Pat. No. 5,902,598 to Chen et al, U.S. Pat. No. 6,331,313 to Wong et al, U.S. Pat. No. 5,707,643 to Ogura et al, U.S. Pat. No. 5,466,233 to Weiner et al and U.S. Pat. No. 6,251,090 to Avery et al each describes intraocular implant devices and systems that may be used to deliver pharmaceutical compositions.

[000123] In other embodiments, e.g., for repair of neural lesions, such as a damaged or severed optic nerve, it may be desirable or appropriate to deliver the cells on or in a biodegradable, preferably bioresorbable or bioabsorbable, scaffold or matrix. These typically three-dimensional biomaterials contain the living cells attached to the scaffold, dispersed within the scaffold, or incorporated in an extracellular matrix entrapped in the scaffold. Once implanted into the target region of the body, these implants become integrated with the host tissue, wherein the transplanted cells gradually become established (see, e.g., P. A. Tresco et al, 2000, supra; see also D. W. Hutmacher, 2001, J. Biomater. Sci. Polymer Edn. 12: 107- 174).

[000124] Examples of scaffold or matrix (sometimes referred to collectively as

"framework") material that may be used in the present invention include nonwoven mats, porous foams, or self-assembling peptides. Nonwoven mats may, for example, be formed using fibers comprised of a synthetic absorbable copolymer of glycolic and lactic acids (PGA/PLA), sold under the trade name VICRYL (Ethicon, Inc., Somerville, N.J). Foams, composed of, for example, poly (epsilon-caprolactone)/poly (glycolic acid) (PCL/PGA) copolymer, formed by processes such as freeze-drying, or lyophilized, as discussed in U.S. Pat. No. 6,355,699 also may be utilized. Hydrogels such as self-assembling peptides (e.g., RAD 16) may also be used. In szYw-forming degradable networks are also suitable for use in the invention (see, e.g., Anseth, K. S. et al, 2002, J. Controlled Release 78: 199-209; Wang, D. et al, 2003, Biomaterials 24: 3969-3980; U.S. Patent Publication 2002/0022676 to He et al). These materials are formulated as fluids suitable for injection, and then may be induced by a variety of means (e.g., change in temperature, pH, exposure to light) to form degradable hydrogel networks in situ or in vivo.

[000125] In another embodiment, the framework is a felt, which can be composed of a multifilament yarn made from a bioabsorbable material, e.g., PGA, PLA, PCL copolymers or blends, or hyaluronic acid. The yarn is made into a felt using standard textile processing techniques consisting of crimping, cutting, carding and needling. In another embodiment, cells are seeded onto foam scaffolds that may be composite structures.

[000126] In many of the abovementioned embodiments, the framework may be molded into a useful shape. Furthermore, it will be appreciated that PPDCs may be cultured on preformed, non-degradable surgical or implantable devices, e.g., in a manner corresponding to that used for preparing fibroblast-containing GDC endovascular coils, for instance (Marx, W. F. et al, 2001, Am. J. Neuroradiol. 22: 323-333).

[000127] The matrix, scaffold or device may be treated prior to inoculation of cells in order to enhance cell attachment. For example, prior to inoculation, nylon matrices can be treated with 0.1 molar acetic acid and incubated in polylysine, PBS, and/or collagen to coat the nylon. Polystyrene can be similarly treated using sulfuric acid. The external surfaces of a framework may also be modified to improve the attachment or growth of cells and differentiation of tissue, such as by plasma coating the framework or addition of one or more proteins (e.g., collagens, elastic fibers, reticular fibers), glycoproteins, glycosaminoglycans (e.g., heparin sulfate, chondroitin-4-sulfate, chondroitin-6-sulfate, dermatan sulfate, keratin sulfate), a cellular matrix, and/or other materials such as, but not limited to, gelatin, alginates, agar, agarose, and plant gums, among others.

[000128] Frameworks containing living cells are prepared according to methods known in the art. For example, cells can be grown freely in a culture vessel to sub-confluency or confluency, lifted from the culture and inoculated onto the framework. Growth factors may be added to the culture medium prior to, during, or subsequent to inoculation of the cells to trigger differentiation and tissue formation, if desired. Alternatively, the frameworks themselves may be modified so that the growth of cells thereon is enhanced, or so that the risk of rejection of the implant is reduced. Thus, one or more biologically active compounds, including, but not limited to, anti-inflammatory agents, immunosuppressants or growth factors, may be added to the framework for local release.

Methods of Use

[000129] Progenitor cells, such as postpartum cells (preferably hUTCs or PDCs), or cell populations thereof, or conditioned medium or other components of or products produced by such cells, may be used in a variety of ways to support and facilitate repair and regeneration of ocular cells and tissues. Such utilities encompass in vitro, ex vivo and in vivo methods. The methods set forth below are directed to PPDCs, but other progenitor cells may also be suitable for use in those methods.

In Vitro and Ex Vivo Methods

[000130] In one embodiment, progenitor cells, such as postpartum cells (preferably hUTCs or PDCs), and conditioned media generated therefrom may be used in vitro to screen a wide variety of compounds for effectiveness and cytotoxicity of pharmaceutical agents, growth factors, regulatory factors, and the like. For example, such screening may be performed on substantially homogeneous populations of PPDCs to assess the efficacy or toxicity of candidate compounds to be formulated with, or co-administered with, the PPDCs, for treatment of a an ocular condition. Alternatively, such screening may be performed on PPDCs that have been stimulated to differentiate into a cell type found in the eye, or progenitor thereof, for the purpose of evaluating the efficacy of new pharmaceutical drug candidates. In this embodiment, the PPDCs are maintained in vitro and exposed to the compound to be tested. The activity of a potentially cytotoxic compound can be measured by its ability to damage or kill cells in culture. This may readily be assessed by vital staining techniques.

[000131] As discussed above, PPDCs can be cultured in vitro to produce biological products that are either naturally produced by the cells, or produced by the cells when induced to differentiate into other lineages, or produced by the cells via genetic modification. For instance, TIMP1, TPO, KGF, HGF, FGF, HBEGF, BDNF, MlPlb, MCP1, RANTES, 1309, TARC, MDC, and IL-8 were found to be secreted from umbilicus-derived cells grown in Growth Medium. Umbilicus-derived cells also secrete thrombospondin-1,

thrombospondin-2, and thrombospondin-4. TEVIP1, TPO, KGF, HGF, HBEGF, BDNF, MlPla, MCP-1, RANTES, TARC, Eotaxin, and IL-8 were found to be secreted from placenta-derived PPDCs cultured in Growth Medium (see Examples).

[000132] In this regard, an embodiment of the invention features use of PPDCs for production of conditioned medium. Production of conditioned media from PPDCs may either be from undifferentiated PPDCs or from PPDCs incubated under conditions that stimulate differentiation. Such conditioned media are contemplated for use in in vitro or ex vivo culture of epithelial or neural precursor cells, for example, or in vivo to support transplanted cells comprising homogeneous populations of PPDCs or heterogeneous populations comprising PPDCs and other progenitors.

[000133] Cell lysates, soluble cell fractions or components from PPDCs, or ECM or components thereof, may be used for a variety of purposes. As mentioned above, some of these components may be used in pharmaceutical compositions. In other embodiments, a cell lysate or ECM is used to coat or otherwise treat substances or devices to be used surgically, or for implantation, or for ex vivo purposes, to promote healing or survival of cells or tissues contacted in the course of such treatments.

[000134] As described in Examples 12 and 13, PPDCs have demonstrated the ability to support survival, growth and differentiation of adult neural progenitor cells when grown in co-culture with those cells. Likewise, previous studies indicate that PPDCs may function to support cells of the retina via trophic mechanisms. (US 2010-0272803). Accordingly, PPDCs are used advantageously in co-cultures in vitro to provide trophic support to other cells, in particular neural cells and neural and ocular progenitors (e.g., neural stem cells and retinal or corneal epithelial stem cells). For co-culture, it may be desirable for the PPDCs and the desired other cells to be co-cultured under conditions in which the two cell types are in contact. This can be achieved, for example, by seeding the cells as a heterogeneous population of cells in culture medium or onto a suitable culture substrate. Alternatively, the PPDCs can first be grown to confluence, and then will serve as a substrate for the second desired cell type in culture. In this latter embodiment, the cells may further be physically separated, e.g., by a membrane or similar device, such that the other cell type may be removed and used separately, following the co-culture period. Use of PPDCs in co-culture to promote expansion and differentiation of neural or ocular cell types may find applicability in research and in clinical/therapeutic areas. For instance, PPDC co-culture may be utilized to facilitate growth and differentiation of such cells in culture, for basic research purposes or for use in drug screening assays, for example. PPDC co-culture may also be utilized for ex vivo expansion of neural or ocular progenitors for later administration for therapeutic purposes. For example, neural or ocular progenitor cells may be harvested from an individual, expanded ex vivo in co-culture with PPDCs, then returned to that individual (autologous transfer) or another individual (syngeneic or allogeneic transfer). In these embodiments, it will be appreciated that, following ex vivo expansion, the mixed population of cells comprising the PPDCs and progenitors could be administered to a patient in need of treatment. Alternatively, in situations where autologous transfer is appropriate or desirable, the co-cultured cell populations may be physically separated in culture, enabling removal of the autologous progenitors for administration to the patient.

In Vivo Methods

[000135] As set forth in the Examples, progenitor cells (PPDCs), or conditioned media generated from such cells, may effectively be used for treating an ocular degenerative condition. Once transplanted into a target location in the eye, progenitor cells or conditioned media from progenitor cells, such as PPDCs, provide trophic support for ocular cells, including neuronal cells in situ. [000136] Progenitor cells (PPDCs), conditioned media from progenitor cells, may be administered with other beneficial drugs, biological molecules, such as growth factors, trophic factors, conditioned medium (from progenitor or differentiated cell cultures), or other active agents, such as anti-inflammatory agents, anti-apoptotic agents, antioxidants, growth factors, neurotrophic factors or neuroregenerative or neuroprotective drugs as known in the art. When conditioned media is administered with other agents, they may be administered together in a single pharmaceutical composition, or in separate pharmaceutical compositions, simultaneously or sequentially with the other agents (either before or after administration of the other agents).

[000137] Examples of other components that may be administered with progenitor cells, such as PPDCs, and conditioned media products include, but are not limited to: (1) other neuroprotective or neurobeneficial drugs; (2) selected extracellular matrix components, such as one or more types of collagen known in the art, and/or growth factors, platelet-rich plasma, and drugs (alternatively, the cells may be genetically engineered to express and produce growth factors); (3) anti-apoptotic agents (e.g., erythropoietin (EPO), EPO mimetibody, thrombopoietin, insulin-like growth factor (IGF)-I, IGF-II, hepatocyte growth factor, caspase inhibitors); (4) anti-inflammatory compounds (e.g., p38 MAP kinase inhibitors, TGF-beta inhibitors, statins, IL-6 and IL-I inhibitors, PEMIROLAST, TRANILAST, REMICADE, SIROLEVIUS, and non-steroidal anti-inflammatory drugs (NSAIDS) (such as TEPOXALF , TOLMETIN, and SUPROFEN); (5) immunosuppressive or immunomodulatory agents, such as calcineurin inhibitors, mTOR inhibitors, antiproliferatives, corticosteroids and various antibodies; (6) antioxidants such as probucol, vitamins C and E, conenzyme Q-10, glutathione, L-cysteine and N-acetyl cysteine; and (6) local anesthetics, to name a few.

[000138] Liquid or fluid pharmaceutical compositions may be administered to a more general location in the eye (e.g., topically or intra-ocularly).

[000139] Other embodiments encompass methods of treating ocular degenerative conditions by administering pharmaceutical compositions comprising conditioned medium from progenitor cells, such as PPDCs, or trophic and other biological factors produced naturally by those cells or through genetic modification of the cells. Again, these methods may further comprise administering other active agents, such as growth factors, neurotrophic factors or neuroregenerative or neuroprotective drugs as known in the art.

[000140] Dosage forms and regimes for administering conditioned media from progenitor cells, such as PPDCs, or any of the other pharmaceutical compositions described herein are developed in accordance with good medical practice, taking into account the condition of the individual patient, e.g., nature and extent of the ocular degenerative condition, age, sex, body weight and general medical condition, and other factors known to medical practitioners. Thus, the effective amount of a pharmaceutical composition to be administered to a patient is determined by these considerations as known in the art.

[000141] It may be desirable or appropriate to pharmacologically immunosuppress a patient prior to initiating cell therapy. This may be accomplished through the use of systemic or local immunosuppressive agents, or it may be accomplished by delivering the cells in an encapsulated device, as described above. These and other means for reducing or eliminating an immune response to the transplanted cells are known in the art. As an alternative, conditioned media may be prepared from PPDCs genetically modified to reduce their immunogenicity, as mentioned above.

[000142] Survival of transplanted cells in a living patient can be determined through the use of a variety of scanning techniques, e.g., computerized axial tomography (CAT or CT) scan, magnetic resonance imaging (MRI) or positron emission tomography (PET) scans.

Determination of transplant survival can also be done post mortem by removing the tissue and examining it visually or through a microscope. Alternatively, cells can be treated with stains that are specific for neural or ocular cells or products thereof, e.g., neurotransmitters. Transplanted cells can also be identified by prior incorporation of tracer dyes such as rhodamine-or fluorescein-labeled microspheres, fast blue, ferric microparticles, bisbenzamide or genetically introduced reporter gene products, such as beta-galactosidase or beta- glucuronidase.

[000143] Functional integration of transplanted cells or conditioned medium into ocular tissue of a subject can be assessed by examining restoration of the ocular function that was damaged or diseased. For example, effectiveness in the treatment of macular degeneration or other retinopathies may be determined by improvement of visual acuity and evaluation for abnormalities and grading of stereoscopic color fundus photographs. (Age-Related Eye Disease Study Research Group, EI, NIH, AREDS Report No.8, 2001, Arch. Ophthalmol. 119: 1417-1436).

Kits and Banks

[000144] In another aspect, the invention provides kits that utilize progenitor cells, such as PPDCs, and cell populations, conditioned medium prepared from the cells, preferably from PPDCs, and components and products thereof in various methods for ocular regeneration and repair as described above. Where used for treatment of ocular degenerative conditions, or other scheduled treatment, the kits may include one or more cell populations or conditioned medium, including at least postpartum cells or conditioned medium derived from postpartum cells, and a pharmaceutically acceptable carrier (liquid, semi-solid or solid). The kits also optionally may include a means of administering the cells and conditioned medium, for example by injection. The kits further may include instructions for use of the cells and conditioned medium. Kits prepared for field hospital use, such as for military use may include full-procedure supplies including tissue scaffolds, surgical sutures, and the like, where the cells or conditioned medium are to be used in conjunction with repair of acute injuries. Kits for assays and in vitro methods as described herein may contain, for example, one or more of: (1) PPDCs or components thereof, or conditioned medium or other products of PPDCs; (2) reagents for practicing the in vitro method; (3) other cells or cell populations, as appropriate; and (4) instructions for conducting the in vitro method.

[000145] In yet another aspect, the invention also provides for banking of tissues, cells, cell populations, conditioned medium, and cellular components of the invention. As discussed above, the cells and and conditioned medium are readily cryopreserved. The invention therefore provides methods of cryopreserving the cells in a bank, wherein the cells are stored frozen and associated with a complete characterization of the cells based on immunological, biochemical and genetic properties of the cells. The frozen cells can be thawed and expanded or used directly for autologous, syngeneic, or allogeneic therapy, depending on the requirements of the procedure and the needs of the patient. Preferably, the information on each cryopreserved sample is stored in a computer, which is searchable based on the requirements of the surgeon, procedure and patient with suitable matches being made based on the characterization of the cells or populations. Preferably, the cells of the invention are grown and expanded to the desired quantity of cells and therapeutic cell compositions are prepared either separately or as co-cultures, in the presence or absence of a matrix or support. While for some applications it may be preferable to use cells freshly prepared, the remainder can be cryopreserved and banked by freezing the cells and entering the information in the computer to associate the computer entry with the samples. Even where it is not necessary to match a source or donor with a recipient of such cells, for immunological purposes, the bank system makes it easy to match, for example, desirable biochemical or genetic properties of the banked cells to the therapeutic needs. Upon matching of the desired properties with a banked sample, the sample is retrieved and prepared for therapeutic use. Cell lysates, ECM or cellular components prepared as described herein may also be cryopreserved or otherwise preserved (e.g., by lyophilization) and banked in accordance with the present invention.

[000146] The following examples are provided to describe the invention in greater detail. They are intended to illustrate, not to limit, the invention.

[000147] The following abbreviations may appear in the examples and elsewhere in the specification and claims: ANG2 (or Ang2) for angiopoietin 2; APC for antigen-presenting cells; BD F for brain-derived neurotrophic factor; bFGF for basic fibroblast growth factor; bid (BID) for "bis in die" (twice per day); CK18 for cytokeratin 18; CNS for central nervous system; CNTF for ciliary neurotrophic factor; CXC ligand 3 for chemokine receptor ligand 3; DMEM for Dulbecco's Minimal Essential Medium; DMEM:lg (or DMEM:Lg, DMEM:LG) for DMEM with low glucose; EDTA for ethylene diamine tetraacetic acid; EGF (or E) for epidermal growth factor; FACS for fluorescent activated cell sorting; FBS for fetal bovine serum; FGF (or F) for fibroblast growth factor; GBP for gabapentin; GCP-2 for granulocyte chemotactic protein-2; GDNF for glial cell-derived neurotrophic factor; GF AP for glial fibrillary acidic protein; FIB-EGF for heparin-binding epidermal growth factor; HCAEC for Human coronary artery endothelial cells; HGF for hepatocyte growth factor; hMSC for Human mesenchymal stem cells; HNF-1 alpha for hepatocyte-specific transcription factor; HVVEC for Human umbilical vein endothelial cells; 1309 for a chemokine and the ligand for the CCR8 receptor; ICAM-1 for intracellular adhesion molecule-1; IGF-1 for insulin-like growth factor 1; IL-6 for interleukin-6; IL-8 for interleukin 8;K19 for keratin 19; K8 for keratin 8; KGF for keratinocyte growth factor; LIF for leukemia inhibitory factor; MBP for myelin basic protein; MCP-1 for monocyte chemotactic protein 1; MDC for macrophage- derived chemokine; MlPlalpha for macrophage inflammatory protein 1 alpha; MlPlbeta for macrophage inflammatory protein 1 beta; MMP for matrix metalloprotease (MMP); MSC for mesenchymal stem cells; NHDF for Normal Human Dermal Fibroblasts; NPE for Neural Progenitor Expansion media; NT3 for neurotrophin 3; 04 for oligodendrocyte or glial differentiation marker 04; PBMC for Peripheral blood mononuclear cell; PBS for phosphate buffered saline; PDGF-CC for platelet derived growth factor C; PDGF-DD for platelet derived growth factor D; PDGFbb for platelet derived growth factor bb; PEDF for pigment epithelium-derived factor; PO for "per os" (by mouth); PNS for peripheral nervous system; Rantes (or RANTES) for regulated on activation, normal T cell expressed and secreted; rhGDF-5 for recombinant human growth and differentiation factor 5; SC for subcutaneously; SDF-1 alpha for stromal-derived factor 1 alpha; SiFH for sonic hedgehog; SOP for standard operating procedure; TARC for thymus and activation-regulated chemokine; TCP for Tissue culture plastic; TCPS for tissue culture polystyrene; TGFbetal for transforming growth factor betal; TGFbeta2 for transforming growth factor beta2; TGF beta-3 for transforming growth factor beta-3; TFMP1 for tissue inhibitor of matrix metalloproteinase 1; TPO for

thrombopoietin; TSP for thrombospondin; TUJ1 for Bill Tubulin; VEGF for vascular endothelial growth factor; vWF for von Willebrand factor; ZO-1 for zonula occludens protein-1; and alphaFP for alpha-fetoprotein.

[000148] The present invention is further illustrated, but not limited by, the following examples.

EXAMPLE 1

hUTC Efficacy in Rat OIR Model

[000149] The effect of hUTC on retinal neovascularization was investigated.

Materials and Methods

[000150] Human umbilical tissue derived cells (hUTC) were obtained from the methods described in Examples 4 - 16 following and in detail in U.S. Patent Nos. 7,524,489, and 7,510,873, and U.S. Pub. App. No. 2005/0058631, both incorporated by reference herein. Briefly, human umbilical cords were obtained with donor consent following live births.

Tissues were minced and enzymatically digested. After almost complete digestion with Dulbecco's modified Eagle's medium (DMEM)-low glucose (Lg) (SAFC Biosciences, Lenexa, KS) containing a mixture of 0.5U/mL Collagenase (Serva Elecrtrophoresis,

Heidelberg, Germany), 5U/mL Neutral Protease (Serva Elecrtrophoresis, Heidelberg, Germany), and 2U/mL Hyaluronidase (Cumulase; Origio a/s, Mal0v, Denmark), the cell suspension was filtered through a 70μπι filter, and the supernatant was centrifuged at 250 x g. Isolated cells were washed in DMEM-Lg several times and plated at a density of 5,000 cells/cm 2 in DMEM-Lg containing 15% (vol/vol) fetal bovine serum (FBS; SAFC

Biosciences) (5% (vol/vol) carbon dioxide, 37°C). When cells reached approximately 70% confluence (-3-4 days), they were passaged using TrypLE (Gibco, Grand Island, NY). Cells were expanded several times and banked. Cryopreserved hUTC (16-20 population doublings) were used.

[000151] Sprague Dawley rats were raised from birth through P14 in a variable oxygen atmosphere consisting of 24-hour alternating episodes of 50% and 10% oxygen. Upon removal from the oxygen exposure chamber (PI 4), rats received intravitreal injections of hUTC in one of three doses/densities (4x10 3 , 2x10 4 or lxlO 5 cells), 100 μg/ml positive control compound, or vehicle (cryopreservative solution). The treatment group distribution is shown in Table 1-1 below. All pups were sacrificed at six days postexposure (P20). Pups were enucleated and both normal, intra-retinal vascular growth and abnormal, pre-retinal neovascular growth were assessed in ADPase-stained retinal flat-mounts using widely published methods. Areas of normal and abnormal vascular growth were measured via computer-assisted image analysis using high-resolution digital images of the stained retinal flat-mounts (Figure 1).

Table 1-1

Results

[000152] Effect of hUTC on intra-retinal vascular development: Analysis of variance revealed no statistically significant differences in intra-retinal vessel growth between the vehicle treatment group and the hUTC treatment groups. In fact, the medium cell density group, which displayed the smallest retinal vascular area, also showed the smallest neovascular area. This is counter to the conventional idea that a larger region of retinal ischemia leads to increased tissue hypoxia, more growth factor production and more retinal neovascularization, and indicates that vascular area did not control neovascularization in this case. Statistical significance was calculated using area (mm 2 ) measurements, but the data are depicted using percent total retinal area vascularized for ease of interpretation. Error bars indicate standard error. (Figure 2).

[000153] Effect of hUTC on pre-retinal neovascular growth: Intravitreal injection of hUTC inhibited pre-retinal neovascular growth by 90.4% (p < 0.0002, ANOVA) at the medium cell density (2x10 4 ) and by 40.3% (p = 0.0484, ANOVA) at the low density (4x10 3 ), relative to cryopreservative vehicle-injected eyes. Retinas could not be assessed from the high cell density treatment group. The low hUTC cell density out-performed the positive control compound, though the difference between the two groups was not statistically significant (p = 0.0542, Student's t test). Data are depicted by both bar graph (FIG. 3 A) and scatter plot (FIG. 3B). Error bars indicate standard error. Examples of ADPase-stained retinas from several treatment groups are shown in FIG. 3C.

[000154] Pathologic effects of hUTC: Intravitreal injection of the highest hUTC density consistently resulted in an ocular condition reminiscent of proliferative vitreoretinopathy (PVR). This condition also was observed in some eyes receiving the medium hUTC density. In two locations, near the injection site and in the region of the optic disc, the retina displayed localized distortion, traction and detachment. The hUTC formed a contiguous sheet within the posterior vitreous at the surface of the detached retina. Removal of this sheet of cells, which was necessary for retinal staining, often led to disruption of the retinal surface that prevented assessment of pre-retinal neovascularization. FIG. 3D shows an example of this phenomenon (retina 58R) from the high hUTC density treatment group - the injected cells appeared to organize into a sheet, extending from the optic disc to the mid-periphery in some retinal quadrants and the far periphery in others. The top panel shows this sheet on the surface of an unstained, dissected retina. The bottom left panel shows the stained retina after the sheet of cells has been peeled. The central retina is distorted and folded by vitreous traction, and the tissue shows tearing artifact as a result of the peeling process, indicating that the hUTC had become associated with the retinal surface. The bottom right panel shows the sheet of cells following removal. The sheet of cells has become closely associated with the hyaloid vasculature, as indicated by the many hyaloid vessels contained in the layer after peeling. It is not clear if the hUTC proliferated during the period between injection and sacrifice or if the injected cells simply coalesced into a sheet. Table 1-2 demonstrates PVR- like tractional retinal detachment. Table 1-2

[000155] Reliable assessment of neovascularization could not be made on any retinas displaying PVR-like tractional retinal detachment. Some retinas that demonstrated tractional detachment did yield vascular area data, while others could not be evaluated because the retinal surface was torn during removal of the sheet of injected cells or because incomplete removal of the cells prohibited adequate staining. Conversely, some retinas that did not demonstrate tractional detachment still could not be evaluated, because they had strongly adherent and cell-laden posterior vitreous that caused tearing when removed or prevented staining when left in place.

[000156] In FIG. 3E, a single retina from the high hUTC density treatment group (23R) yielded both vascular area and neovascular area data. This retina demonstrated severe neovascular growth (0.8599 mm 2 NV area), meaning that the treatment exacerbated NV relative to non-injected or vehicle-injected retinas.

[000157] Statistical Summary: The neovascular area data were subjected to analysis of variance to determine whether any statistically significant differences existed, and a Dunnett's post hoc test to identify specifically how the various treatment groups compared. Analysis is presented below.

[000158] The medium hUTC density (2x10 cells) study arm displayed a statistically significant decrease in the pathologic effects (retinal neovascularization) of oxygen-induced retinopathy (p < 0.0002). At this density, hUTC outperformed all other study arms, including the positive control compound (p = 0.0282 vs. vehicle), which typically results in 50 - 60% percent inhibition. The performance of the medium hUTC density was at 90.4% inhibition of neovascularization. However, injection of hUTC at the highest density can result in tractional retinal detachment; at the medium density tractional detachment can also occur, though with less prevalence. EXAMPLE 2 hUTC Efficacy by Subretinal Injection in Rat OIR Model

[000159] The effect of subretinal injection of hUTC on retinal neovascularization was investigated.

Materials and Methods

[000160] hUTC were obtained as described above in Example 1.

[000161] Sprague Dawley rats were raised from birth through P14 in a variable oxygen atmosphere consisting of 24-hour alternating episodes of 50% and 10% oxygen. Upon removal from the oxygen exposure chamber (PI 4), rats received subretinal injections of hUTC in one of two doses/densities (4x10 3 or 2x10 4 cells), or vehicle (cryopreservative solution). The treatment group distribution is shown in Table 2-1 below. All pups were sacrificed at six days post-exposure/post-injection (P20). Pups were enucleated and both normal, intra-retinal vascular growth and abnormal, pre-retinal neovascular growth were assessed in ADPase-stained retinal flat-mounts using widely published methods. Areas of normal and abnormal vascular growth were measured via computer-assisted image analysis using high-resolution digital images of the stained retinal flat-mounts.

Table 2-1

Results

[000162] Injections: Six days after injection, cells remained in a discrete area of the subretinal space very near the point of injection (FIG. 4A). The location of the cells can be seen as a darkened area in the central subretinal space in the left panel. An outline indicates this region in the right panel. [000163] Effect of hUTC on intra-retinal vascular development: Analysis of variance revealed no statistically significant difference in intra-retinal vessel growth between the vehicle treatment group and the low hUTC cell density treatment group (2x10 4 ). (FIG. 4B). However, both vehicle and low cell density groups displayed a significantly greater percent vascular area (* p < 0.0001) than the medium cell density (4x10 3 ) group. Statistical significance was calculated using area (mm 2 ) measurements, but the data are depicted using percent total retinal area vascularized for ease of interpretation.

[000164] Effect of hUTC on pre-retinal neovascular growth: Subretinal injection of hUTC inhibited pre-retinal neovascular growth by 25.4% (p = 0.5576, ANOVA) at the medium cell density (2x10 4 ) and by 55.7% (* p = 0.0341, ANOVA) at the low density (4x10 3 ), relative to cryopreservative vehicle-injected eyes (FIG. 4C). Examples of ADPase- stained retinas from three treatment groups are shown in FIG. 4D.

[000165] Statistical Summary: The neovascular area data were subjected to analysis of variance to determine whether any statistically significant differences existed, and a Dunnett's post hoc test to identify specifically how the various treatment groups compared. Aanalysis is presented below.

[000166] The low hUTC density (4x10 3 cells) treatment arm displayed a statistically significant reduction in retinal neovascularization resulting from oxygen-induced retinopathy (p < 0.0341). This route of hUTC administration did not yield as much efficacy as that resulting from intravitreal injections, but it caused no disruption of the retina- vitreous interface - a phenomenon observed in the intravitreal injection in Example 1. The difference in efficacy between intravitreal and subretinal injection can be attributed to the distance (and potential barriers) between the site of hUTC delivery and the site of the pathological neovascular growth. Intravitreal injection of hUTC can be expected to have greater influence on pre-retinal neovascularization, while subretinal injection of hUTC is expected have greater influence on subretinal neovascularization.

[000167] The subretinal injection of hUTC resulted in a highly significant (p < 0.0001) inhibition of intra-retinal vascular development. Intra-retinal vascular development is inversely correlated with pre-retinal NV in the OIR model. That is, less intra-retinal vascular growth yields greater retinal ischemia, leading to greater retinal hypoxia, greater angiogenic growth factor induction and more pathological neovascularization. Thus, the unusual "dose- response" showing greater efficacy in the lower cell density treatment arm is consistent with this inverse correlation. The medium cell density (2x10 4 ) inhibited intra-retinal vascular growth, stimulating pre-retinal neovascular growth and counteracting efficacy in that treatment arm. Generally, this phenomenon is expected to be restricted to cases where normal retinal vascular development is underway, as in OIR rats. EXAMPLE 3

Effect of hUTC in Diabetic Retinopathy Leakage

[000168] In this example, the efficacy of subretinal administration of various doses of hUTC in ameliorating diabetic induced retinal vascular leakage was assessed using the Streptozotocin (STZ)-rat diabetic retinopathy model. STZ-diabetic rats have been the primary model for research into the pathogenesis of the vascular lesions of diabetic retinopathy and drug development for diabetic retinopathy. STZ rats exhibit similar retinal lesions: increased permeability of vasculature in the retina, pericyte and endothelial cell loss and basement membrane thickening. The early stages of retinopathy develop relatively quick in the rat; macular edema post increased capillary permeability occurs as early as 2-4 weeks, and pericyte loss and acellular capillaries are apparent after as little as two months of diabetes. hUTC secrete the anti -angiogenic factor pigment epithelium derived factor (PEDF), but do not produce vascular endothelial growth factor (VEGF-A). Based on the similarities between the retinas of STZ-diabetic rats and humans with diabetes mellitus, the STZ-diabetic rat model was used to evaluate the potential therapeutic effect of hUTC.

Materials and Methods

[000169] Animals (Long Evans Rat) were rendered diabetic via 5 daily doses of STZ (50 mg/kg i.p). Blood glucose was monitored weekly to determine the diabetic status of each animal. Animals with blood glucose of > 250mg/dL were enrolled into various treatment groups as in Table 3-1.

[000170] One month after the induction of diabetes, animals with blood glucose of >250mg/dl were enrolled into one of the various treatment groups. The cells were from master cell bank (MCB) 0000132319. MCB 0000132319 was further expanded (Lot No: LNB13610-8, 20 July 2010) and used for this study. The vehicle control (CryoStor Dlite, Biolife Solutions, NY; color-coded red) or test articles (color-coded yellow, pink, or grey) were administered at a volume of 4 μL via subretinal delivery using a 30g needle. For positive control, a high dose aspirin group was used. Rats received intra-peritoneal injection of aspirin (50 mg/kg) daily for the duration of the study starting 4 weeks after induction of diabetes. As an additional comparison, a diabetic group of rats did not receive any injection, and a group of healthy rats of the same age without intervention was also used.

[000171] Vascular retinal leakage was evaluated with fluorescein angiography (FA) and with optical coherence tomography (OCT) at 4 weeks and 8 weeks post injection. The rats were sacrificed at 8 weeks post injection and the eyes were collected to be analyzed by the biotin bovine serum albumin (BSA) assay, Western blot (WB) or immunohistochemistry (IHC). In the WB and the IHC, the expression level of PEDF, intracellular adhesion molecule- 1 (ICAM-1), VEGF, zonula occludens protein- 1 (ZO-1) and beta-actin (β-actin) was assessed.

[000172] In total, 88 Long-Evans diabetic rats and 12 healthy Long-Evans rats were used in this study. Rat utilization was as follows: 18 red (including 5 deaths), 15 yellow (including 8 deaths), 11 pink (including 5 deaths), 16 grey (including 6 deaths), 11 aspirin (including 2 deaths), 9 no injections (including 0 deaths), 6 healthy rats (including 0 deaths). Outcomes were analyzed either by the biotin-BSA assay, WB, or IHC.

[000173] Subretinal injections: Subretinal injections were performed once at 1 month post induction of diabetes, under general anesthesia with ketamine/xylazine. Briefly, a sclerotomy is performed with a 27-gauge needle and a 30-gauge needle inserted into the eye paying attention not to injure the retina or the lens. The tip of the needle was placed subretinally and 4 μΙ_, of solution were slowly injected into the subretinal space under direct observation through an operating microscope. Any eyes showing significant lens or retinal damage were excluded from the study.

[000174] Fluorescein angiography (FA): Fluorescein angiography was performed twice during the study (at 1 month and 2 months post treatment, i.e., 2 and 3 months post-diabetes induction respectively). The animals were anesthetized with a ketmaine/xylazine mixture, and eyes were dilated with instillation of phenylephrine 5%/tropicamide 0.5%. Fluorescein injections were administered intraperitoneally as 2% fluorescein sodium at a dose of 0.1 ml per 15 grams of body weight. Images were acquired with a Canon Fundus Camera specially modified for small animals.

[000175] Spectral domain OCT measurements: Retinal thickness was performed under anesthesia at 2 and 3 months after induction of diabetes. The animals were anesthetized, and eyes were dilated with instillation of phenylephrine 5%/tropicamide 0.5%. The animals were then placed in front of the imaging OCT machine by Bioptigen (non-contact) and images were acquired over 1 minute. Balanced salt solution was used to hydrate the cornea if needed. Volume analysis, centered on the optic nerve head, was performed, using 100 horizontal, raster, and consecutive B-scan lines, each one composed of 1200 A-scans. The volume size was 2.0 x 2.0 mm (rats). The software was able to generate the en face fundus image using the reflectance information obtained from the OCT sections (volume intensity projection), so that the point-to-point correlation between OCT and fundus position was possible and accurate.

[000176] Retinal vascular leakage: Measurement of retinal blood vessel leakage using biotin BSA was done after euthanasia (as described in Trichonas, Invest. Ophthalmol. Vis. Sci., 2010; 51 : 1677-1682). [000177] In summary: An injection (137 μL of 43.7mg/mL) of Biotin-BSA (Santa Cruz Biotechnology) was given to rats through the femoral vein. One hour later, a right ventricular perfusion of Ringer lactate was made for 6 minutes, at a physiological pressure of 120mmHg. Height of apparatus was adjusted to allow a flow rate of 260 mL/min. The perfusion was followed by enucleation, extraction, sonication and centrifugation of retinas. The

supernatants were kept at - 80 degrees Celsius before the ELISA was performed. When all the samples were collected, a sandwich ELISA of the retina bBSA levels was performed. A 96-well plate was coated overnight at 4 degrees C with 100 μL ννεΐΐ (5 μg/mL) of a rabbit anti-BSA antibody in PBS. The following day, the wells were washed 6 times with 0.05% Tween-PBS (PBST). To block nonspecific sites, we used a blocking agent, casein in PBST (0.5 mg/mL) for one hour at room temperature. After a second washing step with PBST, one- hundred-microliter retinal samples (150 μg protein) were added and incubated for 2 hours at room temperature. After a third washing step with PBST, 100 μL of streptavidin-HRP (1 :2000) in PBS-Tween-20 0.4% was added, and the samples were incubated for 20 minutes at room temperature and subsequently washed 6 times with 3 x PBS containing Tween-20 0.4%. Finally, the wells were incubated with tetramethylbenzidine (100 μL; Sigma-Aldrich) for 5 minutes at room temperature, and then 100 μL of stop solution (2 M H 2 S0 4 ) was added. Optical density was read at 450 nm with a spectrophotometer (model Spectra Max 190, Molecular Devices). Blood retinal barrier (BRB) breakdown, expressed as the vascular leakage rate, was calculated by dividing the bBSA (ng) by the retinal protein concentration (mg/mL) and then by multiplying the result by the lysate volume (mL). It was expressed in nanograms of bBS A/amount of protein in the retina milligrams, per hour. The retinal protein concentration was measured with a protein assay (Bradford method).

[000178] Immunofluorescence: After perfusion through the animal heart, eyes were enucleated and put into OCT compound. Frozen sections at 12 microns were obtained. Slides were blocked with 0.5% skim milk and washed x2 with 0.05% tween TBS and incubated with a mouse monoclonal VEGF antibody (1-10 μg/mL) (Abeam, Cambridge, MA), a goat polyclonal ICAM-1 antibody( 1-10 μg/mL; R&D Systems, Minneapolis, MN), a mouse monoclonal PEDF antibody (5μg/mL; LifeSpan Bioscience, Seattle, WA) or rabbit polyclonal ZO-1 antibody(l-4μg/mL; Invitrogen, Carlsbad, CA) (in 0.5% TBST ) overnight. Slides were washed x2 and incubated with Alexa 488 goat anti-mouse, Alexa 555-donkey anti-goat or Alexa 568 donkey anti-rabbit (1 :400) for 1 hour. Images were acquired with confocal microscopy. DAPI was used for nuclear staining. [000179] Western blot analysis: After enucleation, the retinas were isolated in lysis buffer (made with Complete Lysis-M reagent) in 1.5 mL tubes and sonicated. Then a centrifugation was performed at 13000rpm for 10 minutes at 4°C. The supernatant was removed and a protein assay was performed to calculate the concentrations of proteins in each sample. LDS sample buffer (Invitrogen) and 2-mercaptoethanol (Cambrex) were added to each sample. The samples were incubated at 95°C for 5 min and loaded onto a NuPAGE 4-12% Bis-Tris Gel (Invitrogen) prior to transfer onto a PVDF membrane (0.45 μιη; Millipore, Billerica, MA, USA). A blocking agent (Thermo) was used for 20 minutes. The membranes were incubated overnight with VEGF antibody, goat ICAM-1 antibody (R&D Systems, Minneapolis, MN), mouse monoclonal PEDF antibody (LifeSpan BioScience, Seattle, WA) and rabbit polyclonal ZO-1 antibody (Invitrogen, Carlsbad, CA) (1 : 1000 in 5% wt/vol BSA, Tween 20 (TTBS)) for all antibodies, at 4°C. The blotted membranes were washed 3 times (5 min) with TTBS and incubated for 20 min at room temperature with Donkey anti-rabbit secondary antibody (1 : 10,000; Jackson ImmunoResearch, West Grove, PA, USA). The membranes were washed 3 times (5 min) in TTBS, and immunoreactive bands were visualized by ECL plus and exposure onto Fuji RX film (Fujifilm, Tokyo, Japan) for approx.5 min. Most of the time, the membrane was re-blotted using a stripping agent for 15 minutes (Re-Blot Plus Strong

Solution 10X; Millipore, Temecula California), blocked for 20 minutes (Thermo) and blotted with another first antibody and the second antibody again prior to detection.

Results

[000180] STZ diabetes/hyperglycemia induction: A dose of STZ administration

(50mg/kg) given daily for 5 days resulted in 100% diabetes induction. Average blood glucose levels were > 572.70 mg/dL ( Red:>573.00, Yellow: >558.75, Pink: >581.50, Grey: >568.75, Asp: >559.55, No-injection: >587.11, Asp: >600.00) with a range of 300 mg /dL - >600 mg/dL.

[000181] Effects of diabetes induction and treatment groups on FA and SD-OCT: One month after the induction of diabetes, animals were randomized to the seven different group treatments in Table 3-1. In total, 88 Long-Evans diabetic rats and 12 healthy Long-Evans rats were employed in this study. Rat utilization was as follows: 18 red (including 5 deaths), 15 yellow (including 8 deaths), 11 pink (including 5 deaths), 16 grey (including 6 deaths), 11 aspirin (including 2 deaths), 9 no injections (including 0 deaths), 6 healthy rats (including 0 deaths). Treatment with aspirin was administered daily via IP injection. Treatment with hUTC was administered once via subretinal injection.

[000182] At one and two months after initiation of treatment, fluorescein angiography and SD-OCT were performed. Due to cataract, the number of animals that could receive FA and OCT was limited. The FA did not display any significant pathology due to diabetes. Before this study there have been no reports of analysis of retinal thickness in diabetes in rodents by SD-OCT. This study revealed no significant differences in retinal thickness. (FIG. 5A).

[000183] Effects of hUTC on retinal vascular leakage: Treatment with hUTC occurred one month after the induction of diabetes. Two months after the single administration of hUTC, vascular leakage was assessed by biotin-BSA assay (as described above). Diabetes lead to a significant increase in leakage of biotin-BSA tracer compared to healthy animals (FIG. 5B). The subretinal injection did not alter the vascular leakage seen in diabetes since there was no significant difference in the vascular leakage among diabetic animals without injection or with subretinal injection of the low and middle dose of hUTC or with subretinal injection of vehicle (FIG. 5C). The highest concentration of hUTC (grey group) lead to significant suppression of vascular leakage close to levels seen with the positive control (very high dose aspirin).

[000184] Assessment of cytokine expression in retina lysates: To assess the mode of action of hUTC, retina lysates of 5-6 animals were pooled together, and equal amounts of protein were analyzed for the expression of VEGF, ICAM-1, PEDF and ZO-1. There were no differences detected in expression of rat VEGF, rat ICAM-1 or rat ZO-1. Human PEDF could be detected in retina samples that were injected with the highest dose of cells. No effects of diabetes or hUTC treatment on the overall level of protein expression of VEGF, ICAM-1 or ZO-1 by Western blotting were detected; their localization using

immunohistochemistry was examined.

[000185] Assessment of cytokine expression in retina cross sections: To further assess the effects of diabetes and hUTC on the expression pattern of cytokines that can affect the vasculature, we analyzed their expression pattern in frozen cross section of retinas. As shown in FIGS. 5D-5F, diabetes led to alteration in the expression pattern of VEGF, ZO-1 and ICAM-1, with a more lacy appearance and increase in the staining. No PEDF was detected except in the groups of animals with the highest hUTC treatment. It also appears that the highest dose of hUTC may "normalize" the expression pattern of the cytokines. EXAMPLE 4

Derivation of Cells from Postpartum Tissue

[000186] This example describes the preparation of postpartum-derived cells from placental and umbilical cord tissues. Postpartum umbilical cords and placentae were obtained upon birth of either a full term or pre-term pregnancy. Cells were harvested from five separate donors of umbilicus and placental tissue. Different methods of cell isolation were tested for their ability to yield cells with: 1) the potential to differentiate into cells with different phenotypes, a characteristic common to stem cells; or 2) the potential to provide trophic factors useful for other cells and tissues.

Methods & Materials

[000187] Umbilical cell isolation: Umbilical cords were obtained from National Disease Research Interchange (NDRl, Philadelphia, Pa.). The tissues were obtained following normal deliveries. The cell isolation protocol was performed aseptically in a laminar flow hood. To remove blood and debris, the cord was washed in phosphate buffered saline (PBS; Invitrogen, Carlsbad, Calif.) in the presence of antimycotic and antibiotic (100 units/milliliter penicillin, 100 micrograms/milliliter streptomycin, 0.25 micrograms/milliliter amphotericin B). The tissues were then mechanically dissociated in 150 cm 2 tissue culture plates in the presence of 50 milliliters of medium (DMEM-Low glucose or DMEM-High glucose; Invitrogen), until the tissue was minced into a fine pulp. The chopped tissues were transferred to 50 milliliter conical tubes (approximately 5 grams of tissue per tube).

[000188] The tissue was then digested in either DMEM-Low glucose medium or DMEM- High glucose medium, each containing antimycotic and antibiotic as described above. In some experiments, an enzyme mixture of collagenase and dispase was used ("C:D") collagenase (Sigma, St Louis, Mo.), 500 Units/milliliter; and dispase (Invitrogen), 50

Units/milliliter in DMEM-Low glucose medium). In other experiments a mixture of collagenase, dispase and hyaluronidase ("C:D:H") was used (collagenase, 500

Units/milliliter; dispase, 50 Units/milliliter; and hyaluronidase (Sigma), 5 Units/milliliter, in DMEM-Low glucose). The conical tubes containing the tissue, medium and digestion enzymes were incubated at 37° C in an orbital shaker (Environ, Brooklyn, N.Y.) at 225 rpm for 2 hrs.

[000189] After digestion, the tissues were centrifuged at 150 x g for 5 minutes, and the supernatant was aspirated. The pellet was resuspended in 20 milliliters of Growth Medium (DMEM-Low glucose (Invitrogen), 15 percent (v/v) fetal bovine serum (FBS; defined bovine serum; Lot#A D 18475; Hyclone, Logan, Utah), 0.001% (v/v) 2-mercaptoethanol (Sigma), 1 milliliter per 100 milliliters of antibiotic/antimycotic as described above. The cell suspension was filtered through a 70-micrometer nylon cell strainer (BD Biosciences). An additional 5 milliliters rinse comprising Growth Medium was passed through the strainer. The cell suspension was then passed through a 40-micrometer nylon cell strainer (BD Biosciences) and chased with a rinse of an additional 5 milliliters of Growth Medium.

[000190] The filtrate was resuspended in Growth Medium (total volume 50 milliliters) and centrifuged at 150 x g for 5 minutes. The supernatant was aspirated and the cells were resuspended in 50 milliliters of fresh Growth Medium. This process was repeated twice more.

[000191] Upon the final centrifugation, supernatant was aspirated and the cell pellet was resuspended in 5 milliliters of fresh Growth Medium. The number of viable cells was determined using Trypan Blue staining. Cells were then cultured under standard conditions.

[000192] The cells isolated from umbilical cords were seeded at 5,000 cells/cm 2 onto gelatin-coated T-75 cm 2 flasks (Corning Inc., Corning, N.Y.) in Growth Medium with antibiotics/antimycotics as described above. After 2 days (in various experiments, cells were incubated from 2-4 days), spent medium was aspirated from the flasks. Cells were washed with PBS three times to remove debris and blood-derived cells. Cells were then replenished with Growth Medium and allowed to grow to confluence (about 10 days from passage 0) to passage 1. On subsequent passages (from passage 1 to 2 and so on), cells reached sub- confluence (75-85 percent confluence) in 4-5 days. For these subsequent passages, cells were seeded at 5000 cells/cm 2 . Cells were grown in a humidified incubator with 5 percent carbon dioxide and atmospheric oxygen, at 37° C.

[000193] Placental Cell Isolation: Placental tissue was obtained from DRI (Philadelphia, Pa.). The tissues were from a pregnancy and were obtained at the time of a normal surgical delivery. Placental cells were isolated as described for umbilical cell isolation.

[000194] The following example applies to the isolation of separate populations of maternal-derived and neonatal -derived cells from placental tissue.

[000195] The cell isolation protocol was performed aseptically in a laminar flow hood. The placental tissue was washed in phosphate buffered saline (PBS; Invitrogen, Carlsbad, Calif.) in the presence of antimycotic and antibiotic (as described above) to remove blood and debris. The placental tissue was then dissected into three sections: top-line (neonatal side or aspect), mid-line (mixed cell isolation neonatal and maternal) and bottom line (maternal side or aspect).

[000196] The separated sections were individually washed several times in PBS with antibiotic/antimycotic to further remove blood and debris. Each section was then

mechanically dissociated in 150 cm 2 tissue culture plates in the presence of 50 milliliters of DMEM-Low glucose, to a fine pulp. The pulp was transferred to 50 milliliter conical tubes. Each tube contained approximately 5 grams of tissue. The tissue was digested in either DMEM-Low glucose or DMEM-High glucose medium containing antimycotic and antibiotic (100 U/milliliter penicillin, 100 micrograms/milliliter streptomycin, 0.25

micrograms/milliliter amphotericin B) and digestion enzymes. In some experiments an enzyme mixture of collagenase and dispase ("C:D") was used containing collagenase (Sigma, St Louis, Mo.) at 500 Units/milliliter and dispase (Invitrogen) at 50 Units/milliliter in

DMEM-Low glucose medium. In other experiments a mixture of collagenase, dispase and hyaluronidase (C:D:H) was used (collagenase, 500 Units/milliliter; dispase, 50

Units/milliliter; and hyaluronidase (Sigma), 5 Units/milliliter in DMEM-Low glucose). The conical tubes containing the tissue, medium, and digestion enzymes were incubated for 2 h at 37° C in an orbital shaker (Environ, Brooklyn, N.Y.) at 225 rpm.

[000197] After digestion, the tissues were centrifuged at 150 x g for 5 minutes, the resultant supernatant was aspirated off. The pellet was resuspended in 20 milliliters of Growth

Medium with penicillin/streptomycin/amphotericin B. The cell suspension was filtered through a 70 micometer nylon cell strainer (BD Biosciences), chased by a rinse with an additional 5 milliliters of Growth Medium. The total cell suspension was passed through a 40 micometer nylon cell strainer (BD Biosciences) followed with an additional 5 milliliters of Growth Medium as a rinse.

[000198] The filtrate was resuspended in Growth Medium (total volume 50 milliliters) and centrifuged at 150 x g for 5 minutes. The supernatant was aspirated and the cell pellet was resuspended in 50 milliliters of fresh Growth Medium. This process was repeated twice more. After the final centrifugation, supernatant was aspirated and the cell pellet was resuspended in 5 milliliters of fresh Growth Medium. A cell count was determined using the Trypan Blue Exclusion test. Cells were then cultured at standard conditions.

[000199] LIBERASE Cell Isolation: Cells were isolated from umbilicus tissues in DMEM- Low glucose medium with LIB ERASE (Boehringer Mannheim Corp., Indianapolis, Ind.) (2.5 milligrams per milliliter, Blendzyme 3; Roche Applied Sciences, Indianapolis, Ind.) and hyaluronidase (5 Units/milliliter, Sigma). Digestion of the tissue and isolation of the cells was as described for other protease digestions above, using the LIBERASE/hyaluronidase mixture in place of the C:D or C:D:H enzyme mixture. Tissue digestion with LIBERASE resulted in the isolation of cell populations from postpartum tissues that expanded readily.

[000200] Cell isolation using other enzyme combinations: Procedures were compared for isolating cells from the umbilical cord using differing enzyme combinations. Enzymes compared for digestion included: i) collagenase; ii) dispase; iii) hyaluronidase; iv)

collagenase: dispase mixture (C:D); v) collagenase: hyaluronidase mixture (C:H); vi) dispase: hyaluronidase mixture (D:H); and vii) collagenase: dispase: hyaluronidase mixture (C:D:H). Differences in cell isolation utilizing these different enzyme digestion conditions were observed (Table 4-1).

[000201] Isolation of cells from residual blood in the cords: Other attempts were made to isolate pools of cells from umbilical cord by different approaches. In one instance umbilical cord was sliced and washed with Growth Medium to dislodge the blood clots and gelatinous material. The mixture of blood, gelatinous material and Growth Medium was collected and centrifuged at 150 x g. The pellet was resuspended and seeded onto gelatin-coated flasks in Growth Medium. From these experiments a cell population was isolated that readily expanded.

[000202] Isolation of cells from cord blood: Cells have also been isolated from cord blood samples attained from NDR1. The isolation protocol used here was that of International Patent Application WO 2003/025149 by Ho et al. (Ho, T. W., et al. , "Cell Populations Which Co-Express CD49C and CD90," Application No. PCT/US02/29971). Samples (50 milliliter and 10.5 milliliters, respectively) of umbilical cord blood (NDR1, Philadelphia Pa.) were mixed with lysis buffer (filter-sterilized 155 mM ammonium chloride, 10 millimolar potassium bicarbonate, 0.1 millimolar EDT A buffered to pH 7 .2 (all components from Sigma, St. Louis, Mo.)). Cells were lysed at a ratio of 1 :20 cord blood to lysis buffer. The resulting cell suspension was vortexed for 5 seconds, and incubated for 2 minutes at ambient temperature. The lysate was centrifuged (10 minutes at 200 x g). The cell pellet was resuspended in complete minimal essential medium (Gibco, Carlsbad, Calif.) containing 10 percent fetal bovine serum (Hyclone, Logan Utah), 4 millimolar glutamine (Mediatech, Herndon, Va.), 100 Units penicillin per 100 milliliters and 100 micrograms streptomycin per 100 milliliters (Gibco, Carlsbad, Calif.). The resuspended cells were centrifuged (10 minutes at 200 x g), the supernatant was aspirated, and the cell pellet was washed in complete medium. Cells were seeded directly into either T75 flasks (Corning, N. Y.), T75 laminin-coated flasks, or T 175 fibronectin-coated flasks (both Becton Dickinson, Bedford, Mass.).

[000203] Isolation of cells using different enzyme combinations and growth conditions: To determine whether cell populations could be isolated under different conditions and expanded under a variety of conditions immediately after isolation, cells were digested in Growth Medium with or without 0.001 percent (v/v) 2-mercaptoethanol (Sigma, St. Louis, Mo.), using the enzyme combination of C:D:H, according to the procedures provided above.

Placental -derived cells so isolated were seeded under a variety of conditions. All cells were grown in the presence of penicillin/streptomycin. (Table 4-2).

[000204] Isolation of cells using different enzyme combinations and growth conditions: In all conditions cells attached and expanded well between passage 0 and 1 (Table 4-2). Cells in conditions 5-8 and 13-16 were demonstrated to proliferate well up to 4 passages after seeding at which point they were cryopreserved and banked.

Results

[000205] Cell isolation using different enzyme combinations: The combination of C:D:H, provided the best cell yield following isolation, and generated cells, which expanded for many more generations in culture than the other conditions (Table 4-1). An expandable cell population was not attained using collagenase or hyaluronidase alone. No attempt was made to determine if this result is specific to the collagen that was tested.

Table 4-1: Isolation of cells from umbilical cord tissue using varying enzyme combinations

[000206] Isolation of cells using different enzyme combinations and growth conditions: Cells attached and expanded well between passage 0 and 1 under all conditions tested for enzyme digestion and growth (Table 4-2). Cells in experimental conditions 5-8 and 13-16 proliferated well up to 4 passages after seeding, at which point they were cryopreserved. All cells were cryopreserved for further investigation.

Table 4-2: Isolation and culture expansion of postpartum cells under varying conditions:

[000207] Isolation of cells from residual blood in the cords: Nucleated cells attached and grew rapidly. These cells were analyzed by flow cytometry and were similar to cells obtained by enzyme digestion.

[000208] Isolation of cells from cord blood: The preparations contained red blood cells and platelets. No nucleated cells attached and divided during the first 3 weeks. The medium was changed 3 weeks after seeding and no cells were observed to attach and grow.

[000209] Summary: Populations of cells can be derived from umbilical cord and placental tissue efficiently using the enzyme combination collagenase (a matrix metalloprotease), dispase (a neutral protease) and hyaluronidase (a mucolytic enzyme that breaks down hyaluronic acid). LIBERASE, which is a Blendzyme, may also be used. Specifically, Blendzyme 3, which is collagenase (4 Wunsch units/g) and thermolysin (1714 casein Units/g) was also used together with hyaluronidase to isolate cells. These cells expanded readily over many passages when cultured in Growth Medium on gelatin-coated plastic.

[000210] Cells were also isolated from residual blood in the cords, but not cord blood. The presence of cells in blood clots washed from the tissue that adhere and grow under the conditions used may be due to cells being released during the dissection process.

EXAMPLE 5

Karyotype Analysis of Postpartum-Derived Cells

[000211] Cell lines used in cell therapy are preferably homogeneous and free from any contaminating cell type. Cells used in cell therapy should have a normal chromosome number (46) and structure. To identify placenta-and umbilicus-derived cell lines that are

homogeneous and free from cells of non-postpartum tissue origin, karyotypes of cell samples were analyzed.

Methods & Materials

[000212] PPDCs from postpartum tissue of a male neonate were cultured in Growth Medium containing penicillin/streptomycin. Postpartum tissue from a male neonate (X,Y) was selected to allow distinction between neonatal -derived cells and maternal derived cells (X,X). Cells were seeded at 5,000 cells per square centimeter in Growth Medium in a T25 flask (Corning Inc., Corning, N.Y.) and expanded to 80% confluence. A T25 flask containing cells was filled to the neck with Growth Medium. Samples were delivered to a clinical cytogenetics laboratory by courier (estimated lab to lab transport time is one hour). Cells were analyzed during metaphase when the chromosomes are best visualized. Of twenty cells in metaphase counted, five were analyzed for normal homogeneous karyotype number (two). A cell sample was characterized as homogeneous if two karyotypes were observed. A cell sample was characterized as heterogeneous if more than two karyotypes were observed. Additional metaphase cells were counted and analyzed when a heterogeneous karyotype number (four) was identified.

Results

[000213] All cell samples sent for chromosome analysis were interpreted as exhibiting a normal appearance. Three of the 16 cell lines analyzed exhibited a heterogeneous phenotype (XX and XY) indicating the presence of cells derived from both neonatal and maternal origins (Table 5-1). Cells derived from tissue Placenta-N were isolated from the neonatal aspect of placenta. At passage zero, this cell line appeared homogeneous XY. However, at passage nine, the cell line was heterogeneous (XX/XY), indicating a previously undetected presence of cells of maternal origin.

Table 5-1. Karyotype results of PPDCs.

[000214] Summary: Chromosome analysis identified placenta-and umbilicus-derived cells whose karyotypes appeared normal as interpreted by a clinical cytogenetic laboratory.

Karyotype analysis also identified cell lines free from maternal cells, as determined by homogeneous karyotype.

EXAMPLE 6

Evaluation of Human Postpartum-Derived Cell Surface Markers by Flow Cytometry

[000215] Characterization of cell surface proteins or "markers" by flow cytometry can be used to determine a cell line's identity. The consistency of expression can be determined from multiple donors, and in cells exposed to different processing and culturing conditions.

Postpartum-derived cell (PPDC) lines isolated from the placenta and umbilicus were characterized (by flow cytometry), providing a profile for the identification of these cell lines.

Methods & Materials

[000216] Media and culture vessels: Cells were cultured in Growth Medium (Gibco Carlsbad, Calif.) with penicillin/streptomycin. Cells were cultured in plasma-treated T75, T150, and T225 tissue culture flasks (Corning Inc., Corning, N.Y.) until confluent. The growth surfaces of the flasks were coated with gelatin by incubating 2% (w/v) gelatin

(Sigma, St. Louis, Mo.) for 20 minutes at room temperature.

[000217] Antibody Staining and flow cytometry analysis: Adherent cells in flasks were washed in PBS and detached with Trypsin/EDTA. Cells were harvested, centrifuged, and resuspended in 3% (v/v) FBS in PBS at a cell concentration of lxlO 7 per milliliter. In accordance to the manufacture's specifications, antibody to the cell surface marker of interest (see below) was added to one hundred microliters of cell suspension and the mixture was incubated in the dark for 30 minutes at 4° C. After incubation, cells were washed with PBS and centrifuged to remove unbound antibody. Cells were resuspended in 500 microliter PBS and analyzed by flow cytometry. Flow cytometry analysis was performed with a

FACScalibur 111 instrument (Becton Dickinson, San Jose, Calif). Table 6-1 lists the antibodies to cell surface markers that were used. Table 6-1: Antibodies used in characterizing cell surface markers.

[000218] Placenta and umbilicus comparison: Placenta-derived cells were compared to umbilicus-derive cells at passage 8.

[000219] Passage to passage comparison: Placenta-and umbilicus-derived cells were analyzed at passages 8, 15, and 20.

[000220] Donor to donor comparison: To compare differences among donors, placenta- derived cells from different donors were compared to each other, and umbilicus-derived cells from different donors were compared to each other.

[000221] Surface coating comparison: Placenta-derived cells cultured on gelatin-coated flasks was compared to placenta-derived cells cultured on uncoated flasks. Umbilicus-derived cells cultured on gelatin-coated flasks was compared to umbilicus-derived cells cultured on uncoated flasks.

[000222] Digestion enzyme comparison: Four treatments used for isolation and preparation of cells were compared. Cells isolated from placenta by treatment with 1) collagenase; 2) collagenase/dispase; 3) collagenase/hyaluronidase; and 4) collagenase/hyaluronidase/dispase were compared.

[000223] Placental layer comparison: Cells derived from the maternal aspect of placental tissue were compared to cells derived from the villous region of placental tissue and cells derived from the neonatal fetal aspect of placenta. Results

[000224] Placenta vs. umbilicus comparison: Placenta-and umbilicus-derived cells analyzed by flow cytometry showed positive expression of CD 10, CD 13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, indicated by the increased values of fluorescence relative to the IgG control. These cells were negative for detectable expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ, indicated by fluorescence values comparable to the IgG control. Variations in fluorescence values of positive curves were accounted. The mean (i.e. CD13) and range (i.e. CD90) of the positive curves showed some variation, but the curves appeared normal, confirming a homogenous population. Both curves individually exhibited values greater than the IgG control.

[000225] Passage to passage comparison— placenta-derived cells: Placenta-derived cells at passages 8, 15, and 20 analyzed by flow cytometry all were positive for expression of CD 10, CD13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, as reflected in the increased value of fluorescence relative to the IgG control. The cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA -DR, DP, DQ having fluorescence values consistent with the IgG control.

[000226] Passage to passage comparison— umbilicus-derived cells: Umbilicus-derived cells at passage 8, 15, and 20 analyzed by flow cytometry all expressed CD10, CD13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, indicated by increased fluorescence relative to the IgG control. These cells were negative for CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ, indicated by fluorescence values consistent with the IgG control.

[000227] Donor to donor comparison— placenta-derived cells: Placenta-derived cells isolated from separate donors analyzed by flow cytometry each expressed CD10, CD13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, with increased values of fluorescence relative to the IgG control. The cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ as indicated by fluorescence value consistent with the IgG control.

[000228] Donor to donor comparison— umbilicus derived cells: Umbilicus-derived cells isolated from separate donors analyzed by flow cytometry each showed positive expression of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, reflected in the increased values of fluorescence relative to the IgG control. These cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ with fluorescence values consistent with the IgG control.

[000229] The effect of surface coating with gelatin on placenta-derived cells: Placenta- derived cells expanded on either gelatin-coated or uncoated flasks analyzed by flow cytometry all expressed of CDIO, CD13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, reflected in the increased values of fluorescence relative to the IgG control. These cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ indicated by fluorescence values consistent with the IgG control.

[000230] The effect of surface coating with gelatin on umbilicus-derived cells: Umbilicus- derived cells expanded on gelatin and uncoated flasks analyzed by flow cytometry all were positive for expression of CDIO, CD 13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, with increased values of fluorescence relative to the IgG control. These cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ, with fluorescence values consistent with the IgG control.

[000231] Effect of enzyme digestion procedure used for preparation of the cells on the cell surface marker profile: Placenta-derived cells isolated using various digestion enzymes analyzed by flow cytometry all expressed CDIO, CD 13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, as indicated by the increased values of fluorescence relative to the IgG control. These cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLADR, DP, DQ as indicated by fluorescence values consistent with the IgG control.

[000232] Placental layer comparison: Cells isolated from the maternal, villous, and neonatal layers of the placenta, respectively, analyzed by flow cytometry showed positive expression of CDIO, CD 13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, as indicated by the increased value of fluorescence relative to the IgG control. These cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ as indicated by fluorescence values consistent with the IgG control.

[000233] Summary: Analysis of placenta-and umbilicus-derived cells by flow cytometry has established of an identity of these cell lines. Placenta-and umbilicus-derived cells are positive for CDIO, CD13, CD44, CD73, CD90, PDGFr-alpha, HLA-A,B,C and negative for CD31, CD34, CD45, CD117, CD141 and HLA-DR, DP, DQ. This identity was consistent between variations in variables including the donor, passage, culture vessel surface coating, digestion enzymes, and placental layer. Some variation in individual fluorescence value histogram curve means and ranges was observed, but all positive curves under all conditions tested were normal and expressed fluorescence values greater than the IgG control, thus confirming that the cells comprise a homogenous population that has positive expression of the markers.

EXAMPLE 7

Immunohistochemical Characterization of Postpartum Tissue Phenotypes

[000234] The phenotypes of cells found within human postpartum tissues, namely umbilical cord and placenta, was analyzed by immunohistochemistry.

Methods & Materials

[000235] Tissue Preparation: Human umbilical cord and placenta tissue was harvested and immersion fixed in 4% (w/v) paraformaldehyde overnight at 4° C. Immunohistochemistry was performed using antibodies directed against the following epitopes: vimentin (1 :500; Sigma, St. Louis, Mo.), desmin (1 : 150, raised against rabbit; Sigma; or 1 :300, raised against mouse; Chemic on, Temecula, Calif), alpha-smooth muscle actin (SMA; 1 :400; Sigma), cytokeratin 18 (CK18; 1 :400; Sigma), von Willebrand Factor (vWF; 1 :200; Sigma), and CD34 (human CD34 Class III; 1 : 100; DAKOCytomation, Carpinteria, Calif). In addition, the following markers were tested: antihuman GROalpha— PE (1 : 100; Becton Dickinson, Franklin Lakes, N.J), antihuman GCP-2 (1 : 100; Santa Cruz Biotech, Santa Cruz, Calif), anti- human oxidized LDL receptor 1 (ox-LDL Rl; 1 : 100; Santa Cruz Biotech), and anti -human NOGO-A (1 : 100; Santa Cruz Biotech). Fixed specimens were trimmed with a scalpel and placed within OCT embedding compound (Tissue-Tek OCT; Sakura, Torrance, Calif) on a dry ice bath containing ethanol. Frozen blocks were then sectioned (10 μπι thick) using a standard cryostat (Leica Microsystems) and mounted onto glass slides for staining.

[000236] Immunohistochemistry: Immunohistochemistry was performed similar to previous studies (e.g., Messina, et al, 2003, Exper. Neurol. 184: 816-829). Tissue sections were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemic on, Temecula, Calif), and 0.3% (v/v) Triton (Triton X-100; Sigma) for 1 hour to access intracellular antigens. In instances where the epitope of interest would be located on the cell surface (CD34, ox-LDL Rl), Triton was omitted in all steps of the procedure in order to prevent epitope loss. Furthermore, in instances where the primary antibody was raised against goat (GCP-2, ox-LDL Rl, NOGO- A), 3% (v/v) donkey serum was used in place of goat serum throughout the procedure.

Primary antibodies, diluted in blocking solution, were then applied to the sections for a period of 4 hours at room temperature. Primary antibody solutions were removed, and cultures washed with PBS prior to application of secondary antibody solutions (1 hour at room temperature) containing block along with goat anti-mouse IgG— Texas Red (1 :250;

Molecular Probes, Eugene, Oreg.) and/or goat anti-rabbit IgG~Alexa 488 (1 :250; Molecular Probes) or donkey anti-goat IgG~FITC (1 : 150; Santa Cruz Biotech). Cultures were washed, and 10 micromolar DAPI (Molecular Probes) was applied for 10 minutes to visualize cell nuclei.

[000237] Following immunostaining, fluorescence was visualized using the appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, N. Y.). Positive staining was represented by fluorescence signal above control staining.

Representative images were captured using a digital color video camera and ImagePro software (Media Cybernetics, Carlsbad, Calif). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif).

Results

[000238] Umbilical cord characterization: Vimentin, desmin, SMA, CKI8, vWF, and CD34 markers were expressed in a subset of the cells found within umbilical cord. In particular, vWF and CD34 expression were restricted to blood vessels contained within the cord. CD34+ cells were on the innermost layer (lumen side). Vimentin expression was found throughout the matrix and blood vessels of the cord. SMA was limited to the matrix and outer walls of the artery & vein, but not contained with the vessels themselves. CK18 and desmin were observed within the vessels only, desmin being restricted to the middle and outer layers.

[000239] Placenta characterization: Vimentin, desmin, SMA, CKI8, vWF, and CD34 were all observed within the placenta and regionally specific.

[000240] GROalpha, GCP-2, ox-LDL RI, andNOGO-A Tissue Expression: None of these markers were observed within umbilical cord or placental tissue.

[000241] Summary: Vimentin, desmin, alpha-smooth muscle actin, cytokeratin 18, von Willebrand Factor, and CD34 are expressed in cells within human umbilical cord and placenta. EXAMPLE 8

Analysis of Postpartum Tissue-Derived Cells using Oligonucleotide Arrays

[000242] Affymetrix GENECHIP arrays were used to compare gene expression profiles of umbilicus-and placenta-derived cells with fibroblasts, human mesenchymal stem cells, and another cell line derived from human bone marrow. This analysis provided a characterization of the postpartum-derived cells and identified unique molecular markers for these cells.

Methods & Materials

[000243] Isolation and culture of cells: Human umbilical cords and placenta were obtained from National Disease Research Interchange (NDRI, Philadelphia, Pa.) from normal full term deliveries with patient consent. The tissues were received and cells were isolated as described in Example 6. Cells were cultured in Growth Medium (using DMEM-LG) on gelatin-coated tissue culture plastic flasks. The cultures were incubated at 37° C with 5% CO 2 .

[000244] Human dermal fibroblasts were purchased from Cambrex Incorporated

(Walkersville, Md.; Lot number 9F0844) and ATCC CRL-1501 (CCD39SK). Both lines were cultured in DMEM/F12 medium (Invitrogen, Carlsbad, Calif.) with 10% (v/v) fetal bovine serum (Hyclone) and penicillin/streptomycin (Invitrogen). The cells were grown on standard tissue-treated plastic.

[000245] Human mesenchymal stem cells (hMSC) were purchased from Cambrex

Incorporated (Walkersville, Md.; Lot numbers 2F1655, 2F1656 and 2F1657) and cultured according to the manufacturer's specifications in MSCGM Media (Cambrex). The cells were grown on standard tissue cultured plastic at 37° C with 5% CO 2 .

[000246] Human iliac crest bone marrow was received from the NDRI with patient consent. The marrow was processed according to the method outlined by Ho, et al. (W003/025149). The marrow was mixed with lysis buffer (155 mM NH 4C1, 10 mM KHCO 3 , and 0.1 mM EDTA, pH 7.2) at a ratio of 1 part bone marrow to 20 parts lysis buffer. The cell suspension was vortexed, incubated for 2 minutes at ambient temperature, and centrifuged for 10 minutes at 500 x g. The supernatant was discarded and the cell pellet was resuspended in Minimal Essential Medium-alpha (Invitrogen) supplemented with 10% (v/v) fetal bovine serum and 4 mM glutamine. The cells were centrifuged again and the cell pellet was resuspended in fresh medium. The viable mononuclear cells were counted using trypan-blue exclusion (Sigma, St. Louis, Mo.). The mononuclear cells were seeded in tissue-cultured plastic flasks at 5x10 4 cells/cm 2 . The cells were incubated at 37° C with 5% CO 2 at either standard atmospheric 0 2 or at 5% 0 2 . Cells were cultured for 5 days without a media change. Media and non-adherent cells were removed after 5 days of culture. The adherent cells were maintained in culture.

[000247] Isolation of mRNA and GENECHIP Analysis: Actively growing cultures of cells were removed from the flasks with a cell scraper in cold PBS. The cells were centrifuged for 5 minutes at 300 x g. The supernatant was removed and the cells were resuspended in fresh PBS and centrifuged again. The supernatant was removed and the cell pellet was

immediately frozen and stored at-80° C. Cellular mRNA was extracted and transcribed into cDNA, which was then transcribed into cRNA and biotin-labeled. The biotin-labeled cRNA was hybridized with HG-U133A GENECHIP oligonucleotide array (Affymetrix, Santa Clara Calif). The hybridization and data collection was performed according to the manufacturer's specifications. Analyses were performed using "Significance Analysis of Microarrays" (SAM) version 1.21 computer software (Stanford University; Tusher, V. G. et ah, 2001, Proc. Natl. Acad. Sci. USA 98: 5116-5121 ).\

Results

[000248] Fourteen different populations of cells were analyzed. The cells along with passage information, culture substrate, and culture media are listed in Table 8-1.

Table 8-1. Cells analyzed by the microarray study. The cells lines are listed by their identification code along with passage at the time of analysis, cell growth substrate, and growth media.

[000249] The data were evaluated by a Principle Component Analysis, analyzing the 290 genes that were differentially expressed in the cells. This analysis allows for a relative comparison for the similarities between the populations.

[000250] Table 8-2 shows the Euclidean distances that were calculated for the comparison of the cell pairs. The Euclidean distances were based on the comparison of the cells based on the 290 genes that were differentially expressed among the cell types. The Euclidean distance is inversely proportional to similarity between the expression of the 290 genes (i.e., the greater the distance, the less similarity exists).

Table 8-2. The Euclidean Distances for the Cell Pairs.

[000251] Tables 8-3, 8-4, and 8-5 show the expression of genes increased in placenta- derived cells (Table 8-3), increased in umbilicus-derived cells (Table 8-4), and reduced in umbilicus-and placenta-derived cells (Table 8-5). The column entitled "Probe Set ID" refers to the manufacturer's identification code for the sets of several oligonucleotide probes located on a particular site on the chip, which hybridize to the named gene (column "Gene Name"), comprising a sequence that can be found within the NCBI (GenBank) database at the specified accession number (column "NCBI Accession Number").

Table 8-3. Genes shown to have specifically increased expression in the placenta-derived cells as compared to other cell lines assayed

Table 8-4. Genes shown to have specifically increased expression in the umbilicus-derived cells as compared to other cell lines assayed

Table 8-5. Genes shown to have decreased expression in umbilicus- and placenta-derived cells as compared to other cell lines assayed

[000252] Tables 8-6, 8-7, and 8-8 show the expression of genes increased in human fibroblasts (Table 8-6), ICBM cells (Table 8-7), and MSCs (Table 8-8).

Table 8-6. Genes that were shown to have increased expression in fibroblasts as compared to the other cell lines assayed.

Table 8-7. Genes that were shown to have increased expression in the ICBM-derived cells as compared to the other cell lines assayed.

Table 8-8. Genes that were shown to have increased expression

MSC cells as compared to the other cell lines assayed.

[000253] Summary: The present examination was performed to provide a molecular characterization of the postpartum cells derived from umbilical cord and placenta. This analysis included cells derived from three different umbilical cords and three different placentas. The examination also included two different lines of dermal fibroblasts, three lines of mesenchymal stem cells, and three lines of iliac crest bone marrow cells. The mRNA that was expressed by these cells was analyzed using an oligonucleotide array that contained probes for 22,000 genes. Results showed that 290 genes are differentially expressed in these five different cell types. These genes include ten genes that are specifically increased in the placenta-derived cells and seven genes specifically increased in the umbilical cord-derived cells. Fifty-four genes were found to have specifically lower expression levels in placenta and umbilical cord, as compared with the other cell types. The expression of selected genes has been confirmed by PCR (see the example that follows). These results demonstrate that the postpartum-derived cells have a distinct gene expression profile, for example, as compared to bone marrow-derived cells and fibroblasts. EXAMPLE 9

Cell Markers in Postpartum-Derived Cells

[000254] In the preceding example, similarities and differences in cells derived from the human placenta and the human umbilical cord were assessed by comparing their gene expression profiles with those of cells derived from other sources (using an oligonucleotide array). Six "signature" genes were identified: oxidized LDL receptor 1, interleukin-8, rennin, reticulon, chemokine receptor ligand 3 (CXC ligand 3), and granulocyte chemotactic protein 2 (GCP-2). These "signature" genes were expressed at relatively high levels in postpartum- derived cells.

[000255] The procedures described in this example were conducted to verify the microarray data and find concordance/discordance between gene and protein expression, as well as to establish a series of reliable assay for detection of unique identifiers for placenta-and umbilicus-derived cells.

Methods & Materials

[000256] Cells: Placenta-derived cells (three isolates, including one isolate predominately neonatal as identified by karyotyping analysis), umbilicus-derived cells (four isolates), and Normal Human Dermal Fibroblasts (NHDF; neonatal and adult) grown in Growth Medium with penicillin/streptomycin in a gelatin-coated T75 flask. Mesechymal Stem Cells (MSCS) were grown in Mesenchymal Stem Cell Growth Medium Bullet kit (MSCGM; Cambrex, Walkerville, Md.).

[000257] For the IL-8 protocol, cells were thawed from liquid nitrogen and plated in gelatin-coated flasks at 5,000 cells/cm 2 , grown for 48 hours in Growth Medium and then grown for further 8 hours in 10 milliliters of serum starvation medium [DMEM~low glucose (Gibco, Carlsbad, Calif), penicillin/streptomycin (Gibco, Carlsbad, Calif.) and 0.1 % (w/v) Bovine Serum Albumin (BSA; Sigma, St. Louis, Mo.)]. After this treatment RNA was extracted and the supernatants were centrifuged at 150 x g for 5 minutes to remove cellular debris. Supernatants were then frozen at -80 C for ELISA analysis.

[000258] Cell culture for ELISA assay: Postpartum cells derived from placenta and umbilicus, as well as human fibroblasts derived from human neonatal foreskin were cultured in Growth Medium in gelatin-coated T75 flasks. Cells were frozen at passage 11 in liquid nitrogen. Cells were thawed and transferred to 15-milliliter centrifuge tubes. After centrifugation at 150 x g for 5 minutes, the supernatant was discarded. Cells were

resuspended in 4 milliliters culture medium and counted. Cells were grown in a 75 cm 2 flask containing 15 milliliters of Growth Medium at 375,000 cells/flask for 24 hours. The medium was changed to a serum starvation medium for 8 hours. Serum starvation medium was collected at the end of incubation, centrifuged at 14,000 x g for 5 minutes (and stored at-20° C).

[000259] To estimate the number of cells in each flask, 2 milliliters of tyrpsin/EDTA (Gibco, Carlsbad, Calif) was added each flask. After cells detached from the flask, trypsin activity was neutralized with 8 milliliters of Growth Medium. Cells were transferred to a 15 milliliters centrifuge tube and centrifuged at 150 x g for 5 minutes. Supernatant was removed and 1 milliliter Growth Medium was added to each tube to resuspend the cells. Cell number was estimated using a hemocytometer.

[000260] ELISA assay: The amount of IL-8 secreted by the cells into serum starvation medium was analyzed using ELISA assays (R&D Systems, Minneapolis, Minn.). All assays were tested according to the instructions provided by the manufacturer.

[000261] Total RNA isolation: RNA was extracted from confluent postpartum-derived cells and fibroblasts or for IL-8 expression from cells treated as described above. Cells were lysed with 350 microliters buffer RLT containing beta-mercaptoethanol (Sigma, St. Louis, Mo.) according to the manufacturer's instructions (RNeasy ® Mini Kit; Qiagen, Valencia, Calif). RNA was extracted according to the manufacturer's instructions (RNeasy ® Mini Kit; Qiagen, Valencia, Calif) and subjected to DNase treatment (2.7 U/sample) (Sigma St. Louis, Mo.). RNA was eluted with 50 microliters DEPC-treated water and stored at-80° C.

[000262] Reverse transcription: RNA was also extracted from human placenta and umbilicus. Tissue (30 milligram) was suspended in 700 microliters of buffer RLT containing 2-mercaptoethanol. Samples were mechanically homogenized and the RNA extraction proceeded according to manufacturer's specification. RNA was extracted with 50 microliters of DEPC-treated water and stored at-80° C. RNA was reversed transcribed using random hexamers with the TaqMan ® reverse transcription reagents (Applied Biosystems, Foster City, Calif.) at 25° C for 10 minutes, 37° C for 60 minutes, and 95° C for 10 minutes. Samples were stored at-20° C.

[000263] Genes identified by cDNA microarray as uniquely regulated in postpartum cells (signature genes—including oxidized LDL receptor, interleukin-8, rennin and reticulon), were further investigated using real-time and conventional PCR.

[000264] Real-time PCR: PCR was performed on cDNA samples using Assays-on- Demand ® gene expression products: oxidized LDL receptor (Hs00234028); rennin

(Hs00166915); reticulon (Hs003825 15); CXC ligand 3 (Hs00171061); GCP-2

(Hs00605742); IL-8 (Hs00174103); and GAPDH (Applied Biosystems, Foster City, Calif.) were mixed with cDNA and TaqMan ® Universal PCR master mix according to the manufacturer's instructions (Applied Biosystems, Foster City, Calif.) using a 7000 sequence detection system with ABI Prism 7000 SDS software (Applied Biosystems, Foster City, Calif). Thermal cycle conditions were initially 50° C for 2 min and 95° C for 10 min, followed by 40 cycles of 95° C for 15 sec and 60° C for 1 min. PCR data was analyzed according to manufacturer's specifications (User Bulletin #2 from Applied Biosystems for ABI Prism 7700 Sequence Detection System).

[000265] Conventional PCR: Conventional PCR was performed using an ABI PRISM 7700 (Perkin Elmer Applied Biosystems, Boston, Mass., USA) to confirm the results from realtime PCR. PCR was performed using 2 microliters of cDNA solution, lx AmpliTaq Gold universal mix PCR reaction buffer (Applied Biosystems, Foster City, Calif.) and initial denaturation at 94° C for 5 minutes. Amplification was optimized for each primer set. For IL- 8, CXC ligand 3, and reticulon (94° C for 15 seconds, 55° C for 15 seconds and 72° C for 30 seconds for 30 cycles); for rennin (94° C for 15 seconds, 53° C for 15 seconds and 72° C for 30 seconds for 38 cycles); for oxidized LDL receptor and GAPDH (94° C for 15 seconds, 55° C for 15 seconds and 72° C for 30 seconds for 33 cycles). Primers used for amplification are listed in Table 9-1. Primer concentration in the final PCR reaction was 1 micromolar except for GAPDH, which was 0.5 micromolar. GAPDH primers were the same as real-time PCR, except that the manufacturer's TaqMan ® probe was not added to the final PCR reaction. Samples were run on 2% (w/v) agarose gel and stained with ethidium bromide (Sigma, St. Louis, Mo.). Images were captured using a 667 Universal Twinpack film (VWR

International, South Plainfield, N.J.) using a focal length Polaroid camera (VWR

International, South Plainfield, N.J.). Table 9-1: Primers used

[000266] Immunofluorescence: PPDCs were fixed with cold 4% (w/v) paraformaldehyde (Sigma-Aldrich, St. Louis, Mo.) for 10 minutes at room temperature. One isolate each of umbilicus-and placenta-derived cells at passage 0 (PO) (directly after isolation) and passage 11 (P 11 ) (two isolates of placenta-derived, two isolates of umbilicus-derived cells) and fibroblasts (P 11) were used. Immunocytochemistry was performed using antibodies directed against the following epitopes: vimentin (1 :500, Sigma, St. Louis, Mo.), desmin (1 : 150; Sigma—raised against rabbit; or 1 :300; Chemicon, Temecula, Calif—raised against mouse,), alpha-smooth muscle actin (SMA; 1 :400; Sigma), cytokeratin 18 (CK18; 1 :400; Sigma), von Willebrand Factor (vWF; 1 :200; Sigma), and CD34 (human CD34 Class III; 1 : 100;

DAKOCytomation, Carpinteria, Calif). In addition, the following markers were tested on passage 11 postpartum cells: anti-human GRO alpha— PE (1 : 100; Becton Dickinson, Franklin Lakes, N.J.), anti-human GCP-2 (1 : 100; Santa Cruz Biotech, Santa Cruz, Calif), anti-human oxidized LDL receptor 1 (ox-LDL Rl; 1 : 100; Santa Cruz Biotech), and anti-human NOGA-A (1 : 100; Santa Cruz, Biotech).

[000267] Cultures were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemic on, Temecula, Calif), and 0.3% (v/v) Triton (Triton X-100; Sigma, St. Louis, Mo.) for 30 minutes to access intracellular antigens. Where the epitope of interest was located on the cell surface (CD34, ox-LDL Rl), Triton X-100 was omitted in all steps of the procedure in order to prevent epitope loss. Furthermore, in instances where the primary antibody was raised against goat (GCP-2, ox-LDL Rl, NOGO-A), 3% (v/v) donkey serum was used in place of goat serum throughout. Primary antibodies, diluted in blocking solution, were then applied to the cultures for a period of 1 hour at room temperature. The primary antibody solutions were removed and the cultures were washed with PBS prior to application of secondary antibody solutions (1 hour at room temperature) containing block along with goat anti-mouse IgG— Texas Red (1 :250; Molecular Probes, Eugene, Oreg.) and/or goat anti-rabbit IgG— Alexa 488 (1 :250; Molecular Probes) or donkey anti-goat IgG~FITC (1 : 150, Santa Cruz Biotech). Cultures were then washed and 10 micromolar DAPI (Molecular Probes) applied for 10 minutes to visualize cell nuclei.

[000268] Following immunostaining, fluorescence was visualized using an appropriate fluorescence filter on an Olympus® inverted epi-fluorescent microscope (Olympus, Melville, N. Y.). In all cases, positive staining represented fluorescence signal above control staining where the entire procedure outlined above was followed with the exception of application of a primary antibody solution. Representative images were captured using a digital color video camera and ImagePro® software (Media Cybernetics, Carlsbad, Calif). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop® software (Adobe, San Jose, Calif).

[000269] Preparation of cells for FACS analysis: Adherent cells in flasks were washed in phosphate buffered saline (PBS) (Gibco, Carlsbad, Calif) and detached with Trypsin/EDTA (Gibco, Carlsbad, Calif). Cells were harvested, centrifuged, and re-suspended 3% (v/v) FBS in PBS at a cell concentration of 1 x 10 7 per milliliter. One hundred microliter aliquots were delivered to conical tubes. Cells stained for intracellular antigens were permeabilized with Perm/Wash buffer (BD Pharmingen, San Diego, Calif). Antibody was added to aliquots as per manufactures specifications and the cells were incubated for in the dark for 30 minutes at 4° C. After incubation, cells were washed with PBS and centrifuged to remove excess antibody. Cells requiring a secondary antibody were resuspended in 100 microliters of 3% FBS. Secondary antibody was added as per manufactures specification and the cells were incubated in the dark for 30 minutes at 4° C. After incubation, cells were washed with PBS and centrifuged to remove excess secondary antibody. Washed cells were resuspended in 0.5 milliliters PBS and analyzed by flow cytometry. The following antibodies were used:

oxidized LDL receptor 1 (sc-5813; Santa Cruz, Biotech), GROa (555042; BD Pharmingen, Bedford, Mass.), Mouse IgGl kappa, (P-4685 and M-5284; Sigma), Donkey against Goat IgG (sc-3743; Santa Cruz, Biotech.). Flow cytometry analysis was performed with

FACScalibur (Becton Dickinson San Jose, Calif). Results

[000270] Results of real-time PCR for selected "signature" genes performed on cDNA from cells derived from human placentae, adult and neonatal fibroblasts and Mesenchymal Stem Cells (MSCs) indicate that both oxidized LDL receptor and rennin were expressed at higher level in the placenta-derived cells as compared to other cells. The data obtained from realtime PCR were analyzed by the AACT method and expressed on a logarithmic scale. Levels of reticulon and oxidized LDL receptor expression were higher in umbilicus-derived cells as compared to other cells. No significant difference in the expression levels of CXC ligand 3 and GCP-2 were found between postpartum-derived cells and controls. The results of realtime PCR were confirmed by conventional PCR. Sequencing of PCR products further validated these observations. No significant difference in the expression level of CXC ligand 3 was found between postpartum-derived cells and controls using conventional PCR CXC ligand 3 primers listed above in Table 9-1.

[000271] The production of the cytokine, IL-8 in postpartum was elevated in both Growth Medium-cultured and serum-starved postpartum-derived cells. All real-time PCR data was validated with conventional PCR and by sequencing PCR products.

[000272] When supernatants of cells grown in serum-free medium were examined for the presence of IL-8, the highest amounts were detected in media derived from umbilical cells and some isolates of placenta cells (Table 9-2). No IL-8 was detected in medium derived from human dermal fibroblasts.

Table 9-2: IL-8 protein expression measured by ELISA

[000273] Placenta-derived cells were also examined for the production of oxidized LDL receptor, GCP-2 and GROalpha by FACS analysis. Cells tested positive for GCP-2. Oxidized LDL receptor and GRO were not detected by this method.

[000274] Placenta-derived cells were also tested for the production of selected proteins by immunocytochemical analysis. Immediately after isolation (passage 0), cells derived from the human placenta were fixed with 4% paraformaldehyde and exposed to antibodies for six proteins: von Willebrand Factor, CD34, cytokeratin 18, desmin, alpha-smooth muscle actin, and vimentin. Cells stained positive for both alpha-smooth muscle actin and vimentin. This pattern was preserved through passage 11. Only a few cells (<5%) at passage 0 stained positive for cytokeratin 18.

[000275] Cells derived from the human umbilical cord at passage 0 were probed for the production of selected proteins by immunocytochemical analysis. Immediately after isolation (passage 0), cells were fixed with 4% paraformaldehyde and exposed to antibodies for six proteins: von Willebrand Factor, CD34, cytokeratin 18, desmin, alpha-smooth muscle actin, and vimentin. Umbilicus-derived cells were positive for alpha-smooth muscle actin and vimentin, with the staining pattern consistent through passage 11.

[000276] Placenta-derived cells at passage 11 were also investigated by

immunocytochemistry for the production of GROalpha and GCP-2. Placenta-derived cells were GCP-2 positive, but GROalpha production was not detected by this method.

[000277] The production of GROalpha, GCP-2, oxidized LDL receptor 1 and reticulon (NOGO-A) in umbilical cord-derived cells at passage 11 was investigated by

immunocytochemistry. Umbilical cord-derived cells were GCP-2 positive, but GRO alpha production was not detected by this method. Furthermore, cells were NOGO-A positive.

[000278] Summary: Concordance between gene expression levels measured by microarray and PCR (both real-time and conventional) has been established for four genes: oxidized LDL receptor 1, rennin, reticulon, and IL-8. The expression of these genes was differentially regulated at the mRNA level in PPDCs, with IL-8 also differentially regulated at the protein level. The presence of oxidized LDL receptor was not detected at the protein level by FACS analysis in cells derived from the placenta. Differential expression of GCP-2 and CXC ligand 3 was not confirmed at the mRNA level, however GCP-2 was detected at the protein level by FACS analysis in the placenta-derived cells. Although this result is not reflected by data originally obtained from the micro array experiment, this may be due to a difference in the sensitivity of the methodologies.

[000279] Immediately after isolation (passage 0), cells derived from the human placenta stained positive for both alpha-smooth muscle actin and vimentin. This pattern was also observed in cells at passage 11. Vimentin and alpha-smooth muscle actin expression may be preserved in cells with passaging, in the Growth Medium and under the conditions utilized in these procedures. Cells derived from the human umbilical cord at passage 0 were probed for the expression of alpha-smooth muscle actin and vimentin, and were positive for both. The staining pattern was preserved through passage 11.

EXAMPLE 10

Secretion of Trophic Factors by Postpartum-Derived Cells

[000280] The secretion of selected trophic factors from placenta-and umbilicus-derived cells was measured. Factors selected for detection included: (1) those known to have angiogenic activity, such as hepatocyte growth factor (HGF) (Rosen et al. (1997) Ciba Found. Symp. 212:215-26), monocyte chemotactic protein 1 (MCP-1) (Salcedo et al. (2000) Blood 96;34-40), interleukin-8 (IL-8) (Li et al. (2003) J. Immunol. 170:3369-76),

keratinocyte growth factor (KGF), basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF) (Hughes et al. (2004) Ann. Thorac. Surg. 77:812-8), matrix

metalloproteinase 1 (TIMP1), angiopoietin 2 (ANG2), platelet derived growth factor (PDGF- bb), thrombopoietin (TPO), heparin-binding epidermal growth factor (HB-EGF), stromal- derived factor 1 alpha (SDF-lalpha); (2) those known to have neurotrophic/neuroprotective activity, such as brain-derived neurotrophic factor (BD F) (Cheng et al. (2003) Dev. Biol. 258;319-33), interleukin-6 (IL-6), granulocyte chemotactic protein-2 (GCP-2), transforming growth factor beta2 (TGFbeta2) ; and (3) those known to have chemokine activity, such as macrophage inflammatory protein 1 alpha (MTPla), macrophage inflammatory protein 1 beta (MTPlb), monocyte chemoattractant-1 (MCP-1), Rantes (regulated on activation, normal T cell expressed and secreted), 1309, thymus and activation-regulated chemokine (TARe), Eotaxin, macrophage-derived chemokine (MDC), IL-8).

Methods & Materials

[000281] Cell culture: PPDCs from placenta and umbilicus as well as human fibroblasts derived from human neonatal foreskin were cultured in Growth Medium with penicillin/streptomycin on gelatin-coated T75 flasks. Cells were cryopreserved at passage 11 and stored in liquid nitrogen. After thawing of the cells, Growth Medium was added to the cells followed by transfer to a 15 milliliter centrifuge tube and centrifugation of the cells at 150 x g for 5 minutes. The supernatant was discarded. The cell pellet was resuspended in 4 milliliters Growth Medium, and cells were counted. Cells were seeded at 375,000 cells/75 cm 2 flask containing 15 milliliters of Growth Medium and cultured for 24 hours. The medium was changed to a serum-free medium (DMEM-low glucose (Gibco), 0.1% (w/v) bovine serum albumin (Sigma), penicillin/streptomycin (Gibco)) for 8 hours. Conditioned serum-free medium was collected at the end of incubation by centrifugation at 14,000 x g for 5 minutes and stored at -20° C.

[000282] To estimate the number of cells in each flask, cells were washed with PBS and detached using 2 milliliters trypsin/EDTA. Trypsin activity was inhibited by addition of 8 milliliters Growth Medium. Cells were centrifuged at 150 x g for 5 minutes. Supernatant was removed, and cells were resuspended in 1 milliliter Growth Medium. Cell number was estimated using a hemocytometer.

[000283] ELISA assay: Cells were grown at 37 C in 5% carbon dioxide and atmospheric oxygen. Placenta-derived cells (batch 101503) also were grown in 5% oxygen or beta- mercaptoethanol (BME). The amount of MCP-1, IL-6, VEGF, SDF-lalpha, GCP-2, IL-8, and TGF-beta 2 produced by each cell sample was measured by an ELISA assay (R&D Systems, Minneapolis, Minn.). All assays were performed according to the manufacturer's instructions.

[000284] SearchLight multiplexed ELISA assay: Chemokines (MIP 1 a, MIP lb, MCP- 1 , Rantes, 1309, TARC, Eotaxin, MDC, IL8), BD F, and angiogenic factors (HGF, KGF, bFGF, VEGF, TFMP1, ANG2, PDGF-bb, TPO, HB-EGF were measured using SearchLight Proteome Arrays (Pierce Biotechnology Inc.). The Proteome Arrays are multiplexed sandwich ELISAs for the quantitative measurement of two to 16 proteins per well. The arrays are produced by spotting a 2x2, 3x3, or 4x4 pattern of four to 16 different capture antibodies into each well of a 96-well plate. Following a sandwich ELISA procedure, the entire plate is imaged to capture chemiluminescent signal generated at each spot within each well of the plate. The amount of signal generated in each spot is proportional to the amount of target protein in the original standard or sample.

Results

[000285] ELISA assay: MCP-1 and IL-6 were secreted by placenta-and umbilicus-derived cells and dermal fibroblasts (Table 10-1). SDF-lalpha was secreted by placenta-derived cells cultured in 5% 0 2 and by fibroblasts. GCP-2 and IL-8 were secreted by umbilicus-derived cells and by placenta-derived cells cultured in the presence of BME or 5% 0 2 . GCP-2 also was secreted by human fibroblasts. TGF-beta2 was not detectable by ELISA assay.

Table 10-1. ELISA Results: Detection of Trophic Factors

[000286] SearchLight multiplexed ELISA assay: TIMPl, TPO, KGF, HGF, FGF, HBEGF, BDNF, MlPlb, MCP1, RANTES, 1309, TARC, MDC, and IL-8 were secreted from umbilicus-derived cells (Tables 10-2 and 10-3). TFMP1, TPO, KGF, HGF, HBEGF, BDNF, MlPla, MCP-1, RANTES, TARC, Eotaxin, and IL-8 were secreted from placenta-derived cells (Tables 10-2 and 10-3). No Ang2, VEGF, or PDGF-bb were detected.

Table 10-2. SEARCHLIGHT Multiplexed ELISA assay results

Table 10-3. SEARCHLIGHT Multiplexed ELISA assay results

EXAMPLE 11

Short-Term Neural Differentiation of Postpartum-Derived Cells

[000287] The ability of placenta-and umbilicus-derived cells (collectively postpartum - derived cells or PPDCs) to differentiate into neural lineage cells was examined.

Methods & Materials

[000288] Isolation and Expansion of Postpartum Cells: PPDCs from placental and umbilical tissues were isolated and expanded as described in Example 4.

[000289] Modified Woodbury-B lack Protocol (A): This assay was adapted from an assay originally performed to test the neural induction potential of bone marrow stromal cells (1). Umbilicus-derived cells (022803) P4 and placenta-derived cells (042203) P3 were thawed and culture expanded in Growth Media at 5,000 cells/cm 2 until sub-confluence (75%) was reached. Cells were then trypsinized and seeded at 6,000 cells per well of a Titretek II glass slide (VWR International, Bristol, Conn.). As controls, mesenchymal stem cells (P3; 1F2155; Cambrex, Walkersville, Md.), osteoblasts (P5; CC2538; Cambrex), adipose-derived cells (Artecel, U.S. Pat. No. 6,555,374 B l) (P6; Donor 2) and neonatal human dermal fibroblasts (P6; CC2509; Cambrex) were also seeded under the same conditions.

[000290] All cells were initially expanded for 4 days in DMEM/F12 medium (Invitrogen, Carlsbad, Calif.) containing 15% (v/v) fetal bovine serum (FBS; Hyclone, Logan, Utah), basic fibroblast growth factor (bFGF; 20 nanograms/milliliter; Peprotech, Rocky Hill, N.J.), epidermal growth factor (EGF; 20 nanograms/milliliter; Peprotech) and

penicillin/streptomycin (Invitrogen). After four days, cells were rinsed in phosphate-buffered saline (PBS; Invitrogen) and were subsequently cultured in DMEM/F12 medium+20%) (v/v) FBS+penicillin/streptomycin for 24 hours. After 24 hours, cells were rinsed with PBS. Cells were then cultured for 1-6 hours in an induction medium which was comprised of

DMEM/FI2 (serum-free) containing 200 mM butylated hydroxyanisole, 10 μΜ potassium chloride, 5 milligram/milliliter insulin, 10 μΜ forskolin, 4 μΜ valproic acid, and 2 μΜ hydrocortisone (all chemicals from Sigma, St. Louis, Mo.). Cells were then fixed in 100%) ice-cold methanol and immunocytochemistry was performed (see methods below) to assess human nestin protein expression.

[000291] Modified Woodbury-B lack Protocol (B): PPDCs (umbilicus (022803) P 11 ; placenta (042203) PI 1 and adult human dermal fibroblasts (1F1853, PI 1) were thawed and culture expanded in Growth Medium at 5,000 cells/cm 2 until sub-confluence (75%) was reached. Cells were then trypsinized and seeded at similar density as in (A), but onto (1) 24 well tissue culture-treated plates (TCP, Falcon brand, VWR International), (2) TCP wells+2% (w/v) gelatin adsorbed for 1 hour at room temperature, or (3) TCP wells+20 μg/milliliter adsorbed mouse laminin (adsorbed for a minimum of 2 hours at 37° C; Invitrogen).

[000292] Exactly as in (A), cells were initially expanded and media switched at the aforementioned timeframes. One set of cultures was fixed, as before, at 5 days and 6 hours, this time with ice-cold 4% (w/v) paraformaldehyde (Sigma) for 10 minutes at room temperature. In the second set of cultures, medium was removed and switched to Neural Progenitor Expansion medium (NPE) consisting of Neurobasal-A medium (Invitrogen) containing B27 (B27 supplement; Invitrogen), L-glutamine (4 mM), and

penicillin/streptomycin (Invitrogen). NPE medium was further supplemented with retinoic acid (RA; 1 μΜ; Sigma). This medium was removed 4 days later and cultures were fixed with ice-cold 4% (w/v) paraformaldehyde (Sigma) for 10 minutes at room temperature, and stained for nestin, GFAP, and TuJl protein expression (see Table 11-1).

Table 11-1. Summary of Primary Antibodies Used

[000293] Two Stage Differentiation Protocol: PPDCs (umbilicus (042203) P 11 , placenta (022803) PI 1), adult human dermal fibroblasts (PI 1; 1F1853; Cambrex) were thawed and culture expanded in Growth Medium at 5,000 cells/cm 2 until sub-confluence (75%) was reached. Cells were then trypsinized and seeded at 2,000 cells/cm 2 , but onto 24 well plates coated with laminin (BD Biosciences, Franklin Lakes, N.J.) in the presence of NPE media supplemented with bFGF (20 nanograms/milliliter; Peprotech, Rocky Hill, N.J.) and EGF (20 nanograms/milliliter; Peprotech) [whole media composition further referred to as NPE+F+E]. At the same time, adult rat neural progenitors isolated from hippocampus (P4; (062603) were also plated onto 24 welliaminin-coated plates in NPE+F+E media. All cultures were maintained in such conditions for a period of 6 days (cells were fed once during that time) at which time media was switched to the differentiation conditions listed in Table 11-2 for an additional period of 7 days. Cultures were fixed with ice-cold 4% (w/v) paraformaldehyde (Sigma) for 10 minutes at room temperature, and stained for human or rat nestin, GF AP, and TuJlprotein expression.

Table 11-2. Summary of Conditions for Two-Stage Differentiation Protocol

[000294] Multiple growth factor protocol: Umbilicus-derived cells (PI 1; (042203)) were thawed and culture expanded in Growth Medium at 5,000 cells/cm 2 until sub-confluence (75%) was reached. Cells were then trypsinized and seeded at 2,000 cells/cm 2 , onto 24 welliaminin-coated plates (BD Biosciences) in the presence of NPE+F (20

nanograms/milliliter)+E (20 nanograms/milliliter). In addition, some wells contained NPE+F+E+2% FBS or 10% FBS. After four days of "pre-differentiation" conditions, all media were removed and samples were switched to NPE medium supplemented with sonic hedgehog (SHH; 200 nanograms/milliliter; Sigma, St. Louis, Mo.), FGF8 (100

nanograms/milliliter; Peprotech), BDNF (40 nanograms/milliliter; Sigma), GDNF (20 nanograms/milliliter; Sigma), and retinoic acid (1 μΜ; Sigma). Seven days post medium change, cultures were fixed with ice-cold 4% (w/v) paraformaldehyde (Sigma) for 10 minutes at room temperature, and stained for human nestin, GFAP, TuJl, desmin, and alpha-smooth muscle actin expression.

[000295] Neural progenitor co-culture protocol: Adult rat hippocampal progenitors (062603) were plated as neurospheres or single cells (10,000 cells/well) onto laminin-coated 24 well dishes (BD Biosciences) in NPE +F (20 nanograms/milliliter) + E (20

nanograms/milliliter).

[000296] Separately, umbilicus-derived cells (042203) PI 1 and placenta-derived cells (022803) PI 1 were thawed and culture expanded in NPE +F (20 nanograms/milliliter) +E (20 nanograms/milliliter) at 5,000 cells/cm 2 for a period of 48 hours. Cells were then trypsinized and seeded at 2,500 cells/well onto existing cultures of neural progenitors. At that time, existing medium was exchanged for fresh medium. Four days later, cultures were fixed with ice-cold 4% (w/v) paraformaldehyde (Sigma) for 10 minutes at room temperature, and stained for human nuclear protein (hNuc; Chemicon) (Table 12-1 above) to identify PPDCs.

[000297] Immunocytochemistry: Immunocytochemistry was performed using the antibodies listed in Table 12-1. Cultures were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, Calif), and 0.3% (v/v) Triton (Triton X-100; Sigma) for 30 minutes to access intracellular antigens. Primary antibodies, diluted in blocking solution, were then applied to the cultures for a period of 1 hour at room temperature. Next, primary antibodies solutions were removed and cultures washed with PBS prior to application of secondary antibody solutions (1 hour at room temperature) containing blocking solution along with goat anti- mouse IgG— Texas Red (1 :250; Molecular Probes, Eugene, Oreg.) and goat anti-rabbit IgG— Alexa 488 (1 :250; Molecular Probes). Cultures were then washed and 10 micromolar DAPI (Molecular Probes) applied for 10 minutes to visualize cell nuclei.

[000298] Following immunostaining, fluorescence was visualized using the appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, N. Y.). In all cases, positive staining represented fluorescence signal above control staining where the entire procedure outlined above was followed with the exception of application of a primary antibody solution. Representative images were captured using a digital color video camera and ImagePro software (Media Cybernetics, Carlsbad, Calif). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif).

Results

[000299] Modified Woodbury-B lack Protocol (A): Upon incubation in this neural induction composition, all cell types transformed into cells with bipolar morphologies and extended processes. Other larger non-bipolar morphologies were also observed. Furthermore, the induced cell populations stained positively for nestin, a marker of multipotent neural stem and progenitor cells.

[000300] Modified Woodbury-B lack Protocol (B): When repeated on tissue culture plastic (TCP) dishes, nestin expression was not observed unless laminin was pre-adsorbed to the culture surface. To further assess whether nestin-expressing cells could then go on to generate mature neurons, PPDCs and fibroblasts were exposed to PE+RA (1 μΜ), a media composition known to induce the differentiation of neural stem and progenitor cells into such cells (2, 3, 4). Cells were stained for TuJl, a marker for immature and mature neurons, GFAP, a marker of astrocytes, and nestin. Under no conditions was TuJl detected, nor were cells with neuronal morphology observed. Furthermore, nestin and GF AP were no longer expressed by PPDCs, as determined by immunocytochemistry.

[000301] Two-stage differentiation: Umbilicus and placenta PPDC isolates (as well as human fibroblasts and rodent neural progenitors as negative and positive control cell types, respectively) were plated on laminin (neural promoting)-coated dishes and exposed to 13 different growth conditions (and two control conditions) known to promote differentiation of neural progenitors into neurons and astrocytes. In addition, two conditions were added to examine the influence of GDF5, and BMP7 on PPDC differentiation. Generally, a two-step differentiation approach was taken, where the cells were first placed in neural progenitor expansion conditions for a period of 6 days, followed by full differentiation conditions for 7 days. Morphologically, both umbilicus-and placenta-derived cells exhibited fundamental changes in cell morphology throughout the time-course of this procedure. However, neuronal or astrocytic-shaped cells were not observed except for in control, neural progenitor-plated conditions. Immunocytochemistry, negative for human nestin, TuJl, and GFAP confirmed the morphological observations.

[000302] Multiple growth factors: Following one week's exposure to a variety of neural differentiation agents, cells were stained for markers indicative of neural progenitors (human nestin), neurons (TuJl), and astrocytes (GFAP). Cells grown in the first stage in non-serum containing media had different morphologies than those cells in serum containing (2% or 10%) media, indicating potential neural differentiation. Specifically, following a two step procedure of exposing umbilicus-derived cells to EGF and bFGF, followed by SHH, FGF8, GD F, BDNF, and retinoic acid, cells showed long extended processes similar to the morphology of cultured astrocytes. When 2% FBS or 10% FBS was included in the first stage of differentiation, cell number was increased and cell morphology was unchanged from control cultures at high density. Potential neural differentiation was not evidenced by immunocytochemical analysis for human nestin, TuJl, or GFAP.

[000303] Neural progenitor and PPDC co-culture: PPDCs were plated onto cultures of rat neural progenitors seeded two days earlier in neural expansion conditions (NPE+F+E). While visual confirmation of plated PPDCs proved that these cells were plated as single cells, human-specific nuclear staining (hNuc) 4 days post-plating (6 days total) showed that they tended to ball up and avoid contact with the neural progenitors. Furthermore, where PPDCs attached, these cells spread out and appeared to be innervated by differentiated neurons that were of rat origin, suggesting that the PPDCs may have differentiated into muscle cells. This observation was based upon morphology under phase contrast microscopy. Another observation was that typically large cell bodies (larger than neural progenitors) possessed morphologies resembling neural progenitors, with thin processes spanning out in multiple directions. hNuc staining (found in one half of the cell's nucleus) showed that in some cases these human cells may have fused with rat progenitors and assumed their phenotype. Control wells containing only neural progenitors had fewer total progenitors and apparent

differentiated cells than did co-culture wells containing umbilicus or placenta PPDCs, further indicating that both umbilicus-and placenta-derived cells influenced the differentiation and behavior of neural progenitors, either by release of chemokines and cytokines, or by contact- mediated effects.

[000304] Summary: Multiple protocols were conducted to determine the short term potential of PPDCs to differentiate into neural lineage cells. These included phase contrast imaging of morphology in combination with immunocytochemistry for nestin, TuJl, and GFAP, proteins associated with multipotent neural stem and progenitor cells, immature and mature neurons, and astrocytes, respectively.

EXAMPLE 12

Long-Term Neural Differentiation of Postpartum-Derived Cells

[000305] The ability of umbilicus and placenta-derived cells (collectively postpartum - derived cells or PPDCs) to undergo long-term differentiation into neural lineage cells was evaluated.

Methods & Materials

[000306] Isolation and Expansion of PPDCs: PPDCs were isolated and expanded as described in previous Examples.

[000307] PPDC Cell Thaw and Plating: Frozen aliquots of PPDCs (umbilicus (022803) Pl l; (042203) Pl l; (071003) P12; placenta (101503) P7) previously grown in Growth Medium were thawed and plated at 5,000 cells/cm 2 in T-75 flasks coated with laminin (BD, Franklin Lakes, N.J.) in Neurobasal-A medium (Invitrogen, Carlsbad, Calif.) containing B27 (B27 supplement, Invitrogen), L-glutamine (4 mM), and Penicillin/Streptomycin (10 milliliters), the combination of which is herein referred to as Neural Progenitor Expansion (NPE) media. NPE media was further supplemented with bFGF (20 nanograms/milliliter, Peprotech, Rocky Hill, N. J.) and EGF (20 nanograms/milliliter, Peprotech, Rocky Hill, N. J.), herein referred to as NPE+bFGF+EGF.

[000308] Control Cell Plating: In addition, adult human dermal fibroblasts (Pl l, Cambrex, Walkersville, Md.) and mesenchymal stem cells (P5, Cambrex) were thawed and plated at the same cell seeding density on laminin-coated T-75 flasks in NPE+bFGF+EGF. As a further control, fibroblasts, umbilicus, and placenta PPDCs were grown in Growth Medium for the period specified for all cultures.

[000309] Cell Expansion: Media from all cultures were replaced with fresh media once a week and cells observed for expansion. In general, each culture was passaged one time over a period of one month because of limited growth in NPE+bFGF+EGF.

[000310] Immunocytochemistry: After a period of one month, all flasks were fixed with cold 4% (w/v) paraformaldehyde (Sigma) for 10 minutes at room temperature. Immunocytochemistry was performed using antibodies directed against TuJl (Bill Tubulin; 1 :500; Sigma, St. Louis, Mo.) and GFAP (glial fibrillary acidic protein; 1 :2000;

DakoCytomation, Carpinteria, Calif). Briefly, cultures were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemic on, Temecula, Calif), and 0.3% (v/v) Triton (Triton X-100; Sigma) for 30 minutes to access intracellular antigens. Primary antibodies, diluted in blocking solution, were then applied to the cultures for a period of 1 hour at room temperature. Next, primary antibodies solutions were removed and cultures washed with PBS prior to application of secondary antibody solutions (1 hour at room temperature) containing block along with goat anti-mouse IgG~Texas Red (1 :250; Molecular Probes, Eugene, Oreg.) and goat anti-rabbit IgG~Alexa 488 (1 :250; Molecular Probes). Cultures were then washed and 10 micromolar DAPI (Molecular Probes) applied for 10 minutes to visualize cell nuclei.

[000311] Following immunostaining, fluorescence was visualized using the appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, N. Y.). In all cases, positive staining represented fluorescence signal above control staining where the entire procedure outlined above was followed with the exception of application of a primary antibody solution. Representative images were captured using a digital color video camera and ImagePro software (Media Cybernetics, Carlsbad, Calif). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif).

Table 12-1. Summary of Primary Antibodies Used

Results

[000312] NPE+bFGF+EGF media slows proliferation of PPDCs and alters their morphology. Immediately following plating, a subset of PPDCs attached to the culture flasks coated with laminin. This may have been due to cell death as a function of the freeze/thaw process or because of the new growth conditions. Cells that did attach adopted morphologies different from those observed in Growth Media. [000313] Clones of umbilicus-derived cells express neuronal proteins: Cultures were fixed at one month post-thawing/plating and stained for the neuronal protein TuJl and GFAP, an intermediate filament found in astrocytes. While all control cultures grown in Growth Medium and human fibroblasts and MSCs grown in PE+bFGF+EGF medium were found to be TuJ 1-/GFAP-, TuJl was detected in the umbilicus and placenta PPDCs. Expression was observed in cells with and without neuronal-like morphologies. No expression of GFAP was observed in either culture. The percentage of cells expressing TuJl with neuronal-like morphologies was less than or equal to 1% of the total population (n=3 umbilicus-derived cell isolates tested). While not quantified, the percentage of TuJl + cells without neuronal morphologies was higher in umbilicus-derived cell cultures than placenta-derived cell cultures. These results appeared specific as age-matched controls in Growth Medium did not express TuJl .

[000314] Summary: Methods for generating differentiated neurons (based on TuJl expression and neuronal morphology) from umbilicus-derived cells were developed. While expression for TuJl was not examined earlier than one month in vitro, it is clear that at least a small population of umbilicus-derived cells can give rise to neurons either through default differentiation or through long-term induction following one month of exposure to a minimal media supplemented with L-glutamine, basic FGF, and EGF.

EXAMPLE 13

PPDC Trophic Factors for Neural Progenitor Support

[000315] The influence of umbilicus-and placenta-derived cells (collectively postpartum- derived cells or PPDCs) on adult neural stem and progenitor cell survival and differentiation through non-contact dependent (trophic) mechanisms was examined.

Methods & Materials

[000316] Adult neural stem and progenitor cell isolation: Fisher 344 adult rats were sacrificed by CO 2 asphyxiation followed by cervical dislocation. Whole brains were removed intact using bone rongeurs and hippocampus tissue dissected based on coronal incisions posterior to the motor and somatosensory regions of the brain (Paxinos, G. & Watson, C. 1997. The Rat Brain in Stereotaxic Coordinates). Tissue was washed in Neurobasal-A medium (Invitrogen, Carlsbad, Calif.) containing B27 (B27 supplement; Invitrogen), L- glutamine (4 mM; Invitrogen), and penicillin/streptomycin (Invitrogen), the combination of which is herein referred to as Neural Progenitor Expansion (NPE) medium. NPE medium was further supplemented with bFGF (20 nanograms/milliliter, Peprotech, Rocky Hill, N.J.) and EGF (20 nanograms/milliliter, Peprotech, Rocky Hill, N. J.), herein referred to as

NPE+bFGF+EGF.

[000317] Following wash, the overlying meninges were removed, and the tissue minced with a scalpel. Minced tissue was collected and trypsin/EDTA (Invitrogen) added as 75% of the total volume. DNase (100 microliters per 8 milliliters total volume, Sigma, St. Louis, Mo.) was also added. Next, the tissue/media was sequentially passed through an 18 gauge needle, 20 gauge needle, and finally a 25 gauge needle one time each (all needles from Becton Dickinson, Franklin Lakes, N.J.). The mixture was centrifuged for 3 minutes at 250 g. Supernatant was removed, fresh NPE+bFGF+EGF was added and the pellet resuspended. The resultant cell suspension was passed through a 40 micrometer cell strainer (Becton Dickinson), plated on laminin-coated T-75 flasks (Becton Dickinson) or low cluster 24-well plates (Becton Dickinson), and grown in NPE+bFGF+EGF media until sufficient cell numbers were obtained for the studies outlined.

[000318] PPDC plating: Postpartum-derived cells (umbilicus (022803) P12, (042103) P12, (071003) P12; placenta (042203) P12) previously grown in Growth Medium were plated at 5,000 cells/transwell insert (sized for 24 well plate) and grown for a period of one week in Growth Medium in inserts to achieve confluence.

[000319] Adult neural progenitor plating: Neural progenitors, grown as neurospheres or as single cells, were seeded onto laminin-coated 24 well plates at an approximate density of 2,000 cells/well in NPE+bFGF+EGF for a period of one day to promote cellular attachment. One day later, transwell inserts containing postpartum cells were added according to the following scheme: a. Transwell (umbilicus-derived cells in Growth Media, 200 microliters)+ neural progenitors (NPE+bFGF+EGF, 1 milliliter) b. Transwell (placenta-derived cells in Growth Media, 200 microliters)+ neural progenitors (NPE+bFGF+EGF, 1 milliliter) c. Transwell (adult human dermal fibroblasts [1 F 1853; Cambrex, Walkersville, Md.] P12 in Growth Media, 200 microliters)+neural progenitors (NPE+bFGF+EGF, 1 milliliter) d. Control: neural progenitors alone (NPE+bFGF+EGF, 1 milliliter) e. Control: neural progenitors alone (NPE only, 1 milliliter)

[000320] Immunocytochemistry: After 7 days in co-culture, all conditions were fixed with cold 4% (w/v) paraformaldehyde (Sigma) for a period of 10 minutes at room temperature. Immunocytochemistry was performed using antibodies directed against the epitopes listed in Table 13-1. Briefly, cultures were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemic on, Temecula, Calif.), and 0.3% (v/v) Triton (Triton X-100; Sigma) for 30 minutes to access intracellular antigens. Primary antibodies, diluted in blocking solution, were then applied to the cultures for a period of 1 hour at room temperature. Next, primary antibodies solutions were removed and cultures washed with PBS prior to application of secondary antibody solutions (1 hour at room temperature) containing blocking solution along with goat anti-mouse IgG— Texas Red (1 :250; Molecular Probes, Eugene, Oreg.) and goat anti-rabbit IgG— Alexa 488 (1 :250;

Molecular Probes). Cultures were then washed and 10 micromolar DAPI (Molecular Probes) applied for 10 minutes to visualize cell nuclei.

[000321] Following immunostaining, fluorescence was visualized using the appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, N. Y.). In all cases, positive staining represented fluorescence signal above control staining where the entire procedure outlined above was followed with the exception of application of a primary antibody solution. Representative images were captured using a digital color video camera and ImagePro software (Media Cybernetics, Carlsbad, Calif). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif).

Table 13-1. Summary of Primary Antibodies Used

[000322] Quantitative analysis of neural progenitor differentiation: Quantification of hippocampal neural progenitor differentiation was examined. A minimum of 1000 cells were counted per condition or if less, the total number of cells observed in that condition. The percentage of cells positive for a given stain was assessed by dividing the number of positive cells by the total number of cells as determined by DAPI (nuclear) staining.

[000323] Mass spectrometry analysis & 2D gel electrophoresis: In order to identify unique, secreted factors as a result of co-culture, conditioned media samples taken prior to culture fixation were frozen down at-80° C overnight. Samples were then applied to ultrafiltration spin devices (MW cutoff 30 kD). Retentate was applied to immunoaffinity chromatography (anti-Hu-albumin; IgY) (immunoaffinity did not remove albumin from the samples). Filtrate was analyzed by MALDI. The pass through was applied to Cibachron Blue affinity chromatography. Samples were analyzed by SDS-PAGE and 2D gel electrophoresis.

Results

[000324] PPDC co-culture stimulates adult neural progenitor differentiation: Following culture with umbilicus-or placenta-derived cells, co-cultured neural progenitor cells derived from adult rat hippocampus exhibited significant differentiation along all three major lineages in the central nervous system. This effect was clearly observed after five days in co-culture, with numerous cells elaborating complex processes and losing their phase bright features characteristic of dividing progenitor cells. Conversely, neural progenitors grown alone in the absence of bFGF and EGF appeared unhealthy and survival was limited.

[000325] After completion of the procedure, cultures were stained for markers indicative of undifferentiated stem and progenitor cells (nestin), immature and mature neurons (TuJl), astrocytes (GFAP), and mature oligodendrocytes (MBP). Differentiation along all three lineages was confirmed while control conditions did not exhibit significant differentiation as evidenced by retention of nestin-positive staining amongst the majority of cells. While both umbilicus-and placenta-derived cells induced cell differentiation, the degree of differentiation for all three lineages was less in co-cultures with placenta-derived cells than in co-cultures with umbilicus-derived cells.

[000326] The percentage of differentiated neural progenitors following co-culture with umbilicus-derived cells was quantified (Table 13-2). Umbilicus-derived cells significantly enhanced the number of mature oligodendrocytes (MBP) (24.0% vs. 0% in both control conditions). Furthermore, co-culture enhanced the number of GFAP + astrocytes and TuJl + neurons in culture (47.2% and 8.7% respectively). These results were confirmed by nestin staining indicating that progenitor status was lost following co-culture (13.4% vs. 71.4% in control condition 4).

[000327] Though differentiation also appeared to be influenced by adult human fibroblasts, such cells were not able to promote the differentiation of mature oligodendrocytes nor were they able to generate an appreciable quantity of neurons. Though not quantified, fibroblasts did however, appear to enhance the survival of neural progenitors.

Table 13-2. Quantification of progenitor differentiation in control vs transwell co-culture with umbilical-derived cells (E=EGF, F=bFGF)

[000328] Identification of unique compounds: Conditioned media from umbilicus-and placenta-derived co-cultures, along with the appropriate controls (NPE media±1.7% serum, media from co-culture with fibroblasts), were examined for differences. Potentially unique compounds were identified and excised from their respective 2D gels.

[000329] Summary: Co-culture of adult neural progenitor cells with umbilicus or placenta PPDCs results in differentiation of those cells. Results presented in this example indicate that the differentiation of adult neural progenitor cells following co-culture with umbilicus- derived cells is particularly profound. Specifically, a significant percentage of mature oligodendrocytes was generated in co-cultures of umbilicus-derived cells.

EXAMPLE 14

Transplantation of Postpartum-Derived Cells

[000330] Cells derived from the postpartum umbilicus and placenta are useful for regenerative therapies. The tissue produced by postpartum-derived cells (PPDCs) transplanted into SCID mice with a biodegradable material was evaluated. The materials evaluated were Vicryl non-woven, 35/65 PCL/PGA foam, and RAD 16 self-assembling peptide hydrogel.

Methods & Material

[000331] Cell Culture: Placenta-and umbilicus-derived cells were grown in Growth Medium (DMEM-Iow glucose (Gibco, Carlsbad Calif), 15% (v/v) fetal bovine serum (Cat. #SH30070.03; Hyclone, Logan, Utah), 0.001%) (v/v) betamercaptoethanol (Sigma, St. Louis, Mo.), penicillin/streptomycin (Gibco)) in a gelatin-coated flasks.

[000332] Sample Preparation: One million viable cells were seeded in 15 microliters Growth Medium onto 5 mm diameter, 2.25 mm thick Vicryl non-woven scaffolds (64.33 milligrams/cc; Lot#3547-47-1) or 5 mm diameter 35/65 PCL/PGA foam (Lot# 3415-53). Cells were allowed to attach for two hours before adding more Growth Medium to cover the scaffolds. Cells were grown on scaffolds overnight. Scaffolds without cells were also incubated in medium.

[000333] RAD 16 self-assembling peptides (3D Matrix, Cambridge, MA) was obtained as a sterile 1 % (w/v) solution in water, which was mixed 1 : 1 with 1 x 10 6 cells in 10%> (w/v) sucrose (Sigma, St Louis, Mo.), 10 mM HEPES in Dulbecco's modified medium (DMEM; Gibco) immediately before use. The final concentration of cells in RAD 16 hydrogel was 1 x 10 6 cells/100 microliters.

[000334] Test Material (N=4/Rx) a. Vicryl non-woven+ 1 x 10 6 umbilicus-derived cells b 35/65 PCL/PGA foam+ 1 x 10 6 umbilicus-derived cells c RAD 16 self-assembling peptide+ 1 x 10 6 umbilicus-derived cells d. Vicryl non-woven+ 1 x 10 6 placenta-derived cells e. 35/65 PCL/PGA foam+ 1 x 10 6 placenta-derived cells f. RAD 16 self-assembling peptide+ 1 x 10 6 placenta-derived cells g. 35/65 PCL/PGA foam h. Vicryl non- woven

[000335] Animal Preparation: The animals were handled and maintained in accordance with the current requirements of the Animal Welfare Act. Compliance with the above Public Laws were accomplished by adhering to the Animal Welfare regulations (9 CFR) and conforming to the current standards promulgated in the Guide for the Care and Use of Laboratory Animals, 7th edition.

[000336] Mice (MusMusculus)/Fox Chase SCID/Male (Harlan Sprague Daw ley, Inc., Indianapolis, Ind.), 5 weeks of age: All handling of the SCID mice took place under a hood. The mice were individually weighed and anesthetized with an intraperitoneal injection of a mixture of 60 milligrams/kg KETASET (ketamine hydrochloride, Aveco Co., Inc., Fort Dodge, Iowa) and 10 milligrams/kg ROMPUN (xylazine, Mobay Corp., Shawnee, Kans.) and saline. After induction of anesthesia, the entire back of the animal from the dorsal cervical area to the dorsal lumbosacral area was clipped free of hair using electric animal clippers. The area was then scrubbed with chlorhexidine diacetate, rinsed with alcohol, dried, and painted with an aqueous iodophor solution of 1% available iodine. Ophthalmic ointment was applied to the eyes to prevent drying of the tissue during the anesthetic period.

[000337] Subcutaneous Implantation Technique: Four skin incisions, each approximately 1.0 cm in length, were made on the dorsum of the mice. Two cranial sites were located transversely over the dorsal lateral thoracic region, about 5-mm caudal to the palpated inferior edge of the scapula, with one to the left and one to the right of the vertebral column. Another two were placed transversely over the gluteal muscle area at the caudal sacro-Iumbar level, about 5-mm caudal to the palpated iliac crest, with one on either side of the midline. Implants were randomly placed in these sites in accordance with the experimental design. The skin was separated from the underlying connective tissue to make a small pocket and the implant placed (or injected for RAD 16) about 1 -cm caudal to the incision. The appropriate test material was implanted into the subcutaneous space. The skin incision was closed with metal clips. [000338] Animal Housing: Mice were individually housed in micro isolator cages throughout the course of the study within a temperature range of 64 F - 79 F. and relative humidity of 30% to 70%, and maintained on an approximate 12 hour light/12 hour dark cycle. The temperature and relative humidity were maintained within the stated ranges to the greatest extent possible. Diet consisted of Irradiated Pico Mouse Chow 5058 (Purina Co.) and water fed ad libitum.

[000339] Mice were euthanized at their designated intervals by carbon dioxide inhalation. The subcutaneous implantation sites with their overlying skin were excised and frozen for histology.

[000340] Histology: Excised skin with implant was fixed with 10% neutral buffered formalin (Richard- Allan Kalamazoo, Mich.). Samples with overlying and adjacent tissue were centrally bisected, paraffin-processed, and embedded on cut surface using routine methods. Five-micron tissue sections were obtained by microtome and stained with hematoxylin and eosin (Poly Scientific Bay Shore, N. Y.) using routine methods.

Results

[000341] There was minimal ingrowth of tissue into foams (without cells) implanted subcutaneously in SCID mice after 30 days. In contrast there was extensive tissue fill in foams implanted with umbilical-derived cells or placenta-derived cells. Some tissue ingrowth was observed in Vicryl non-woven scaffolds. Non-woven scaffolds seeded with umbilicus-or placenta-derived cells showed increased matrix deposition and mature blood vessels.

[000342] Summary: Synthetic absorbable non-woven/foam discs (5.0 mm diameter x 1.0 mm thick) or self-assembling peptide hydrogel were seeded with either cells derived from human umbilicus or placenta and implanted subcutaneously bilaterally in the dorsal spine region of SCID mice. The results demonstrated that postpartum-derived cells could dramatically increase good quality tissue formation in biodegradable scaffolds.

EXAMPLE 15

Telomerase Expression in Umbilical Tissue-derived Cells

[000343] Telomerase functions to synthesize telomere repeats that serve to protect the integrity of chromosomes and to prolong the replicative life span of cells (Liu, K, et al, PNAS, 1999; 96:5147-5152). Telomerase consists of two components, telomerase RNA template (hTER) and telomerase reverse transcriptase (hTERT). Regulation of telomerase is determined by transcription of hTERT but not hTER. Real-time polymerase chain reaction (PCR) for hTERT mRNA thus is an accepted method for determining telomerase activity of cells.

[000344] Cell Isolation. Real-time PCR experiments were performed to determine telomerase production of human umbilical cord tissue-derived cells. Human umbilical cord tissue-derived cells were prepared in accordance the examples set forth above. Generally, umbilical cords obtained from National Disease Research Interchange (Philadelphia, Pa.) following a normal delivery were washed to remove blood and debris and mechanically dissociated. The tissue was then incubated with digestion enzymes including collagenase, dispase and hyaluronidase in culture medium at 37°C. Human umbilical cord tissue-derived cells were cultured according to the methods set forth in the examples above. Mesenchymal stem cells and normal dermal skin fibroblasts (cc-2509 lot # 9F0844) were obtained from Cambrex, Walkersville, Md. A pluripotent human testicular embryonal carcinoma (teratoma) cell line nTera-2 cells (NTERA-2 cl.Dl), (see, Plaia et al, Stem Cells, 2006; 24(3):531-546) was purchased from ATCC (Manassas, Va.) and was cultured according to the methods set forth above.

[000345] Total RNA Isolation. RNA was extracted from the cells using RNeasy® kit (Qiagen, Valencia, Ca.). RNA was eluted with 50 microliters DEPC-treated water and stored at -80°C. RNA was reverse transcribed using random hexamers with the TaqMan® reverse transcription reagents (Applied Biosystems, Foster City, Ca.) at 25°C for 10 minutes, 37°C for 60 minutes and 95°C for 10 minutes. Samples were stored at -20°C.

[000346] Real-time PCR. PCR was performed on cDNA samples using the Applied Biosystems Assays-On-Demand™ (also known as TaqMan® Gene Expression Assays) according to the manufacturer's specifications (Applied Biosystems). This commercial kit is widely used to assay for telomerase in human cells. Briefly, hTert (human telomerase gene) (HsOO 162669) and human GAPDH (an internal control) were mixed with cDNA and

TaqMan® Universal PCR master mix using a 7000 sequence detection system with ABI prism 7000 SDS software (Applied Biosystems). Thermal cycle conditions were initially 50°C for 2 minutes and 95°C for 10 minutes followed by 40 cycles of 95°C for 15 seconds and 60°C for 1 minute. PCR data was analyzed according to the manufacturer's

specifications. [000347] Human umbilical cord tissue-derived cells (ATCC Accession No. PTA-6067), fibroblasts, and mesenchymal stem cells were assayed for hTert and 18S RNA. As shown in Table 15-1, hTert, and hence telomerase, was not detected in human umbilical cord tissue- derived cells.

[000348] Human umbilical cord tissue-derived cells (isolate 022803, ATCC Accession No. PTA-6067) and nTera-2 cells were assayed and the results showed no expression of the telomerase in two lots of human umbilical cord tissue-derived cells while the teratoma cell line revealed high level of expression (Table 15-2).

[000349] Therefore, it can be concluded that the human umbilical tissue-derived cells of the present invention do not express telomerase.

[000350] Various patents and other publications are referred to throughout the specification. Each of these publications is incorporated by reference herein, in its entirety.

[000351] Although the various aspects of the invention have been illustrated above by reference to examples and preferred embodiments, it will be appreciated that the scope of the invention is defined not by the foregoing description but by the following claims properly construed under principles of patent law.