Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
A NOVEL AND EFFICIENT METHOD FOR REPROGRAMMING BLOOD TO INDUCED PLURIPOTENT STEM CELLS
Document Type and Number:
WIPO Patent Application WO/2017/219000
Kind Code:
A1
Abstract:
Described herein are methods and compositions related to generation of induced pluripotent stem cells (iPSCs). Improved techniques for establishing highly efficient, reproducible reprogramming using non-integrating episomal plasmid vectors. Using the described reprogramming protocol, one is able to consistently reprogram non-T cells with close to 100% success from non-T cell or non-B cell sources. Further advantages include use of a defined reprogramming media E7 and using defined clinically compatible substrate recombinant human L-521. Generation of iPSCs from these blood cell sources allows for recapitulation of the entire genomic repertoire, preservation of genomic fidelity and enhanced genomic stability.

Inventors:
SAREEN DHRUV (US)
ORNELAS LOREN A (US)
SVENDSEN CLIVE (US)
Application Number:
PCT/US2017/038041
Publication Date:
December 21, 2017
Filing Date:
June 16, 2017
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
CEDARS SINAI MEDICAL CENTER (US)
International Classes:
C12N5/074; C12N5/10; C12N15/09
Domestic Patent References:
WO2015188131A12015-12-10
Foreign References:
US20140134143A12014-05-15
US20140315306A12014-10-23
US20110200568A12011-08-18
US20160108369A12016-04-21
US20150175973A12015-06-25
US5585089A1996-12-17
Other References:
ALLEN ET AL.: "Remington: The Science and Practice of Pharmacy", 15 September 2012, PHARMACEUTICAL PRESS
HORNYAK ET AL.: "Introduction to Nanoscience and Nanotechnology", 2008, CRC PRESS
SINGLETONSAINSBURY: "Dictionary of Microbiology and Molecular Biology", 2006, J. WILEY & SONS
GREENFIELD: "March's Advanced Organic Chemistry Reactions, Mechanisms and Structure", 2013, COLD SPRING HARBOR PRESS
SINGLETON: "Dictionary of DNA and Genome Technology", 28 November 2012, WILEY-BLACKWELL
GREENSAMBROOK: "Molecular Cloning: A Laboratory Manual", 2012, COLD SPRING HARBOR LABORATORY PRESS
KOHLERMILSTEIN: "Derivation of specific antibody-producing tissue culture and tumor lines by cell fusion", EUR. J. IMMUNOL., vol. 6, no. 7, July 1976 (1976-07-01), pages 511 - 9, XP000654957, DOI: 10.1002/eji.1830060713
RIECHMANN ET AL.: "Reshaping human antibodies for therapy", NATURE, vol. 332, no. 6162, 24 March 1988 (1988-03-24), pages 323 - 7, XP002007067, DOI: 10.1038/332323a0
See also references of EP 3472304A4
Attorney, Agent or Firm:
CHEN, Stephen, W. et al. (US)
Download PDF:
Claims:
THE CLAIMS

1. A method of generating blood cell derived induced pluripotent stem cells, comprising:

providing a quantity of blood cells;

delivering a quantity of reprogramming factors into the blood cells; and culturing the blood cells in a reprogramming media for at least 4 days, wherein delivering the reprogramming factors, and culturing in a reprogramming media generates blood cell derived induced pluripotent stem cells.

2. The method of claim 1, wherein delivering a quantity of reprogramming factors comprises nucleofection.

3. The method of claim 1, wherein the reprogramming factors comprise one or more factors selected from the group consisting of: Oct-4, Sox-2, Klf-4, c-Myc, Lin-28, SV40 Large T Antigen ("SV40LT"), and short hairpin RNAs targeting p53 ("shRNA-p53").

4. The method of claim 3, wherein the reprogramming factors are encoded in one or more oriP/EBNAl derived vectors.

5. The method of claim 4, wherein the one or more oriP/EBNAl derived vectors comprise pEP4 E02S ET2K, pCXLE-hOCT3/4-shp53-F, pCXLE-hSK, pCXLE-hUL, and pCXWB- EBNA1.

6. The method of claim 1, comprising plating of the blood cells on a treated cell culture surface after delivering reprogramming factors into the blood cells, and culturing the blood cells in a reprogramming media on said treated cell culture surface.

7. The method of claim 6, wherein the treated cell culture surface comprises plating of mouse embryonic feeders (MEFs).

8. The method of claim 6, wherein the treated cell culture surface comprises an extracellular matrix protein.

9. The method of claim 8, wherein the extracellular matrix protein comprises laminin.

10. The method of claim 9, wherein laminin comprises L-521.

11. The method of claim 1, wherein the reprogramming media comprises embryonic stem cell (ESC) media.

12. The method of claim 11, wherein the ESC media comprises basic fibroblast growth factor (bFGF).

13. The method of claim 1, wherein the reprogramming media comprises E7 media.

14. The method of claim 13, wherein the reprogramming media comprises E7 media comprising L-Ascorbic Acid, Transferrin, Sodium Bicarbonate, Insulin, Sodium Selenite and/or bFGF.

15. The method of claim 1, wherein culturing the blood cells in a reprogramming media is for at least 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16 days.

16. The method of claim 1, wherein culturing the blood cells in a reprogramming media is for at least 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 or 31 days.

17. A cell line comprising blood cell derived induced pluripotent stem cells generated by the method of claim 1.

18. The method of claim 1, wherein the blood cells are isolated from a subject possessing a disease mutation.

19. The method of claim 18, wherein the disease mutation is associated with a

neurodegenerative disease, disorder and/or condition.

20. The method of claim 18, wherein the disease mutation is associated with an inflammatory bowel disease, disorder, and/or condition.

21. The method of claim 1, wherein blood cells are non T-cell, non B-cell mononuclear cells.

22. The method of claim 1, wherein the blood cells are a sample drawn from a human subject.

23. The method of claim 22, wherein the sample is whole blood.

24. The method of claim 22, wherein the sample is peripheral blood.

25. The method of claim 22, wherein the sample is an isolated component of non T-cell, non B-cell mononuclear cells.

26. A blood cell derived induced pluripotent stem cell line.

Description:
A NOVEL AND EFFICIENT METHOD FOR REPROGRAMMING BLOOD TO INDUCED PLURIPOTENT STEM CELLS

FIELD OF INVENTION

Described herein are methods and compositions related to regenerative medicine including the derivation of induced pluripotent stem cells (iPSCs) from whole blood and peripheral blood, such methods and compositions that provide a renewable source of transplant material. BACKGROUND

Pluripotent stem cells ("pSCs") present broad opportunities to generate therapeutic materials for use in regenerative medicine, as well as providing invaluable in vitro models for studying disease initiation and progression. One category of pSCs, induced pluripotent stem cells ("iPSCs"), possess the hallmark stem cell properties of self-renewal (i.e., immortal) and differentiation capacity into cells derived from all three embryonic germ layers (i.e., pluripotency). These cells can be obtained through "reprogramming", which involves dedifferentiation of cells from non-embryonic sources, such as adult somatic cells. The reprogramming process obviates potential ethical concerns over embryonic source material for other types such pSCs, such as embryonic stem cells ("ESCs"), while providing a further benefit of enabling potential patient-specific immunological incompatibility.

In addition to establishing robust reprogramming techniques, full realization of therapeutic goals for stem-cell regenerative medicine further requires consideration of the types of host cells that can serve as a resource for renewable regenerative material. Ideally, cells would possess not only the requisite plasticity for successful reprogramming and stability in subsequent propagation, but also provide advantages in clinical aspects, such as ease of isolation, storage, stability and maintenance. In this regard, blood cells represent an attractive choice for such uses, given their wide use medical diagnostics and as a highly accessible resource for cellular reprogramming. Whereas fibroblasts have been a widely used cellular source for many reprogramming experiments performed in the last decade, this source material but may not be the best choice for directed reprogramming. Skin biopsy to obtain fibroblasts is an invasive, non-sterile procedure requiring expansion of harvested cells before experimentation. Most importantly, skin cells harbor more mutations due to environmental insults such as UV irradiation than cells from inside the body such as blood. Given the eventual therapeutic goal of generating patient-specific, immunocompatible biological material, there is a great need in the art to establish a robust and reproducible means for reprogramming cells, along with identifying sources of therapeutic materials suitable for eventual clinical application. Such improved methods would need to possess high efficiency of reprogramming, consistent reproduction, produce cells possessing genomic stability and be readily extendible to a variety of cell types.

Described herein are improved techniques for establishing highly efficient, reproducible reprogramming using non-integrating episomal plasmid vectors, including generation of iPSCs from non-T cell, non-B cell component in blood samples. These described approaches allow for use of blood as a readily accessible resource for cellular reprogramming with superior properties in genomic and karyotype stability.

SUMMARY OF THE INVENTION

Described herein is a method of generating blood cell derived induced pluripotent stem cells, comprising providing a quantity of blood cells, delivering a quantity of reprogramming factors into the blood cells, and culturing the blood cells in a reprogramming media for at least 4 days, wherein delivering the reprogramming factors, and culturing in a reprogramming media generates blood cell derived induced pluripotent stem cells. In other embodiments, delivering a quantity of reprogramming factors comprises nucleofection. In other embodiments, the reprogramming factors comprise one or more factors selected from the group consisting of: Oct-4, Sox-2, Klf-4, c-Myc, Lin-28, SV40 Large T Antigen ("SV40LT"), and short hairpin RNAs targeting p53 ("shRNA-p53"). In other embodiments, the reprogramming factors are encoded in one or more oriP/EBNAl derived vectors. In other embodiments, the one or more oriP/EBNAl derived vectors comprise pEP4 E02S ET2K, pCXLE-hOCT3/4-shp53-F, pCXLE-hSK, pCXLE-hUL, and pCXWB-EBNAl . In other embodiments, plating of the blood cells on a treated cell culture surface after delivering reprogramming factors into the blood cells, and culturing the blood cells in a reprogramming media on said treated cell culture surface. In other embodiments, the treated cell culture surface comprises plating of mouse embryonic feeders (MEFs). In other embodiments, the treated cell culture surface comprises an extracellular matrix protein. In other embodiments, the extracellular matrix protein comprises laminin. In other embodiments, laminin comprises L-521. In other embodiments, the reprogramming media comprises embryonic stem cell (ESC) media. In other embodiments, the ESC media comprises basic fibroblast growth factor (bFGF). In other embodiments, the reprogramming media comprises E7 media. In other embodiments, the reprogramming media comprises E7 media comprising L-Ascorbic Acid, Transferrin, Sodium Bicarbonate, Insulin, Sodium Selenite and/or bFGF. In other embodiments, culturing the blood cells in a reprogramming media is for at least 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16 days. In other embodiments, culturing the blood cells in a reprogramming media is for at least 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 or 31 days. Further described herein is a cell line comprising blood cell derived induced pluripotent stem cells generated by the aforementioned method. In other embodiments, the blood cells are isolated from a subject possessing a disease mutation. In other embodiments, the disease mutation is associated with a neurodegenerative disease, disorder and/or condition. In other embodiments, the disease mutation is associated with an inflammatory bowel disease, disorder, and/or condition. In other embodiments, the blood cells are non T- cell, non B-cell mononuclear cells. In other embodiments, the blood cells are a sample drawn from a human subject. In other embodiments, the sample is whole blood. In other embodiments, the sample is peripheral blood. In other embodiments, the sample comprises an isolated component of non T-cell, non B-cell mononuclear cells. Further described herein is a blood cell derived induced pluripotent stem cell line.

Further described herein is a method for generating induced pluripotent stem cells, comprising providing a quantity of blood cells, delivering a quantity of reprogramming factors into the blood cells, plating the blood cells on a treated cell culture surface and culturing the blood cells in a reprogramming media for at least 4 days, wherein delivering the reprogramming factors, and culturing in a reprogramming media generates blood cell derived induced pluripotent stem cells. In other embodiments, delivering a quantity of reprogramming factors comprises nucleofection, the reprogramming factors comprise one or more factors selected from the group consisting of: Oct-4, Sox-2, Klf-4, c-Myc, Lin-28, SV40 Large T Antigen ("SV40LT"), and short hairpin RNAs targeting p53 ("shRNA-p53") , encoded in one or more oriP/EBNAl derived vectors. In other embodiments,the one or more oriP/EBNAl derived vectors comprise pEP4 E02S ET2K, pCXLE-hOCT3/4-shp53-F, pCXLE-hSK, pCXLE-hUL, and pCXWB-EBNAl . In other embodiments,the treated cell culture surface comprises plating of mouse embryonic feeders (MEFs). In other embodiments,the treated cell culture surface comprises an extracellular matrix protein. In other embodiments,the extracellular matrix protein comprises laminin. In other embodiments, laminin comprises L-521. In other embodiments, the reprogramming media comprises embryonic stem cell (ESC) media comprising basic fibroblast growth factor (bFGF). In other embodiments, the reprogramming media comprises E7 media comprising L-Ascorbic Acid, Transferrin, Sodium Bicarbonate, Insulin, Sodium Selenite and/or bFGF. In other embodiments, culturing the blood cells in a reprogramming media is for at least 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16 days. In other embodiments, culturing the blood cells in a reprogramming media is for at least 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 or 31 days. In other embodiments, the blood cells are a sample drawn from a human subject. In other embodiments, the sample is whole blood. In other embodiments, the sample is peripheral blood. In other embodiments, the sample comprises an isolated component of non T-cell, non B-cell mononuclear cells. Further described herein is a blood cell derived induced pluripotent stem cell line.

BRIEF DESCRIPTION OF FIGURES

Figure 1(A) to Figure 1(B). Separation of Blood Components. Figure 1(A)

Schematic showing separated components. Figure 1(B) Picture of separated components.

Figure 2(A) to Figure 2(E). Quality Control: iPS cells. Various measures of quality control for generated cells are shown including Figure 2(A) microscopy, and staining for pluripotent stem cell markers. Figure 2(B) Depiction of the pluripotency of stem cells. Figure 2(C) Karyotype analysis. Figure 2(D) RT-PCR of pluripotent and lineage specific markers for different cell lines. Figure 2(E) Pluripotent stem cells as capable of differentiating into all three embryonic lineages.

Figure 3. PBMC isolation and cryopreservation process. Process for obtaining blood cells utilized as a source for reprogramming and generation of iPSCs.

Figure 4. Peripheral blood mononuclear cells (PBMCs). Depiction of the various components of blood, including populations of PBMCs and relative population abundance.

Figure 5. T-cell Receptor Clonality Assay for iPSCs derived from Blood.

Figure 6(A) to Figure 6(B). Ideogram demonstrating the chromosomal changes observed in the iPSC lines are Cedars-Sinai iPSC Core. Each colored bar represents one chromosome change occurrence in one cell line. The cytogenetic changes are color coded; Marooon, monosomy, loss of a whole chromosome. BC-iPSCs are superior at maintaining stable karyotypes than fibroblast-derived iPSCs. Individual chromosome level ideogram representation of abnormal karyotypes from fibroblast-(Figure 6(A)) and PBMC-derived iPSCs (Figure 6(B)) in the Inventors' laboratory. Abnormal iPSC karyotypes represented in the ideograms are; (Figure 6(A)) fib-iPSCs (n: 59) of total 258 fib-iPSC cultures karyotyped, and (Figure 6(B)) BC-iPSCs (n: 4) of total BC-iPSCss karyotyped (n: 106).

Figure 7(A) to 7(B). Karyotypes. Figure 7(A) Distribution of abnormal karyotypes in relation to passage number. Figure 7(B) Distribution of abnormal karyotypes in relation to age of tissue donor Marooon, monosomy, loss of a whole chromosome;

Figure 8(A) to 8(F). Reliable epsiomal reprogramming of PBMCs into iPSCs. Figure 8(A): Schematic depicting the episomal reprogramming process and timeline of iPSC generation from PBMCs. Figure 8(B): Bright-field images of the reprogrammed iPSC colonies from PBMCs obtained two healthy volunteers, one with a non-T cell (03iCTR- NTnl) reprogramming method and another with a T-cell (80iCTR-Tnl) reprogramming method, show a high nuclear-to-cytoplasmic ratio, are alkaline phosphatase positive and immunopositive for the surface antigens, SSEA4, TRA-1-60, TRA-1-81; and nuclear pluripotency markers OCT3/4, SOX2 and NANOG. Scale bar, 75μιη. Figure 8(C): Microarray Illumina gene-chip and bioinformatics based PluriTest confirms pluripotency levels in the BC-iPSCs. (D): The PBMC- -iPSC lines maintained a normal G-band karyotype as shown from the two representative lines. Figure 8(E): non-T or T-cell clonality assay. Figure 8(F): Quantitative RT-PCR analyses of POU5F1 (OCT4), SOX2, LIN28, L-MYC, and KLF4 expression in 49iCTR and 84iSMA LCL-iPSCs relative to H9 hESCs. (G) T cell receptor gene rearrangement assay shows different sets of PCR primers were used to detect TCRG gene rearrangements occurring in the BC-iPSCs derived from the T-cell method, while PCR products in the 145-255 bp range were not detectable in the non-T cell iPSC clones. A clonal positive control was included. Figure 8(F): TaqMan hPSC Scorecard table showing the tri-lineage potential of the representative BC-iPSCs rom selected genes in four gene groups (self-renewal/pluripotency, ectoderm, endoderm, and mesoderm) comparing spontaneous in vitro EB differentiation of EBs derived from the PBMC-iPSC lines. Figure 8(G): Directed neuronal differentiation of representative fibroblast-derived (n: 26) and PBMC-derived iPSCs (n: 12) showing equivalent numbers of ectodermal (Nkx6.1+/p 3 - tubulin+ neuronal cells). Scale bar, 75μm. Figure 8(H): High percent of iPSC reprogramming success for cryopreserved PBMCs by the 5p method (n = 28 for T cells and n = 13 for non-T cells) when compared to the published 4p method (n = 7 for both T and non-T cells). Percent reprogramming success is calculated as a proportion out of 10 the total number of attempts (successful or failed) at reprogramming the PBMCs. [{T cells, 5p versus 4p (***): z-value = 3.74, two-tailed p-value = 0.00018, two-proportion z-test}; {non-T cells, 4p versus 5p (****): z-value = 7.18, two-tailed p-value < 0.0001, two-proportion z-test}]. ND: none detected.

It was further observed that there was greater reprogramming efficiency of the 5p method for both T and non-T cell populations when compared to the 4p method. Reprogramming efficiency is based on individual iPSC colonies identified and scored normalized to the total number of PBMCs nucleofected post-thaw. [{T cells, 4p versus 5p (***): two-tailed p-value = 0.0004, unpaired t-test with Welch's correction}; {non-T cells, 4p versus 5p (***): two-tailed p-value < 0.0001, unpaired t-test with Welch's correction}]. ND: none detected. Figure 8(1) & Figure 8(J): Individual chromosome level ideogram representation of abnormal karyotypes from fibroblast (top) and PBMC-derived iPSCs (bottom) in the Inventors' laboratory. Abnormal iPSC karyotypes represented in the ideograms are; (top) fib-iPSCs (n = 61) of total n = 260 fib-iPSC cultures karyotyped, and (bottom) PBMC-iPSCs (n = 4) of total n = 206 PBMC-iPSC lines karyotyped. Analysis includes examination of chromosomes in a minimum of 20 cells per culture. Only abnormalities that met the ISCN 2013 definition of clonality were included in this manuscript dataset. The letters above the translocation (navy blue bars) category are matched such that they correspond between the chromosomal locations to where each translocation abnormality occurred. The bar widths in each chromosomal ideogram were scaled in proportion to the number of samples examined for fib-iPSCs and PBMC-iPSCs. Therefore, the bars displaying abnormalities found in PBMC-iPSC lines are 1.26 times (260/206) the width of bars depicting abnormalities found in fib-iPSC lines.

Figure 9(A) to Figure 9(D): Compared to fib-iPSC, the BC-iPSCs are more stable over extended culture. Figure 9(A) Percent abnormal G-band karyotypes observed in the iPSC lines in correlation to donor age of fibroblasts and PBMCs. Figure 9(B) Percent abnormal G-band karyotypes at the first karyotype post-reprogramming and in repeat karyotypes for iPSC lines evaluated in extended cell culture. The dotted lines represent the overall average karyotype abnormality rate for fib-iPSCs (orange) and BC-iPSCs (blue). Figure 9(C) Percent abnormal at the first G-band karyotypes evaluated for the fib- and PBMC-iPSC clonal lines in relation to the passage number of the iPSCs. Figure 9(D) Percent abnormal at the second - fourth (repeat) G-band karyotypes evaluated for the fib- and PBMC-iPSC clonal lines in relation to the passage number of the iPSCs. Figure 10(A) to Figure 10(E): Comparative genomic hybridization confirms that the BC-iPSCs have relatively small and fewer submicroscopic cytogenetic aberrations compared to fib-iPSCs. (FigurelO(A)-FigurelO(B)) The average size of de novo acquired amplifications and deletions detected by aCGH in Mb of fib-iPSCs (n: 6) and PBMC- iPSCs (n:7) when compared to each iPSC line's parental cells. The iPSC lines used in aCGH had both abnormal and normal G-band karyotypes. Abnormal: fib-iPSC (n: 3) and PBMC-iPSC (n: 2). Normal: fib-iPSC (n: 3) and PBMC-iPSC (n: 5). (FigurelO(C)-FigurelO(D)) The average number of de novo acquired amplifications and deletions detected by aCGH in Mb of fib-iPSCs (n: 6) and PBMC- iPSCs (n: 7) when compared to each iPSC line's parental cells. Figure 10(E) A list of most recurrent submicroscopic amps/dels observed in the fib- or PBMC-iPSC lines observed in the Inventors' laboratory. Figure 10(F): All data represented here is based on iPSC lines derived by the Inventors' laboratory at Cedars-Sinai. The fib- iPSCs were reprogrammed by the 3p method and the PBMC-iPSCs were reprogrammed by the 5p method. Percent abnormal G-band karyotypes observed in the iPSC lines in correlation to donor age of fibroblasts and PBMCs. [{fib-iPSCs, donor age groups 21-40 versus 0-20 or 51-60 or 41-50 or 61-90: not significant (ns), two-proportion z-test}]. fib- iPSCs: n = 10/63 (0-20), n = 17/50 (21-40), n = 4/28 (41-50), n = 10/41 (51-60), n = 7/29 (61- 90). [{fib-iPSCs versus PBMC-iPSCs - age group 0-20: ns; age group 21-40 (**): z-value = 2.9, two-tailed p-value = 0.003; age group 41-50 (*): z-value = 2.4, two-tailed p-value = 0.015; age group 51-60 (***): z-value = 3.5, two-tailed p30 value = 0.0005; age group 61-90 (****): z-value = 4.02, two-tailed p-value < 0.0001; two-proportion z-test}]. PBMC-iPSCs: n = 2/34 (0-20), n = 0/19 (21-40), n = 0/39 (41-50), n = 1/56 (51-60), n = 0/62 (61-90). ND: none detected. Percent abnormal G-band karyotypes at the first karyotype post- reprogramming and in repeat karyotypes for iPSC lines evaluated in extended cell culture. The dotted lines represent the overall average karyotype abnormality rate for fib-iPSCs (black) and PBMC-1 iPSCs (grey). [{ 1st karyotype, fib-iPSCs versus PBMC-iPSCs (****): z-value = 5.59, two-tailed p-value < 0.0001, two-proportion z-test}; {repeat (2nd-4th) karyotypes, fib-iPSCs versus PBMC-iPSCs (***): z-value = 3.72, two-tailed p-value = 0.0002, two-proportion z-test}; {fib-iPSCs and PBMC-iPSCs, 1st versus repeat karyotypes tested as not significant by two-proportion z-test}]. fib-iPSCs: n = 46/205 (1 st karyotype) and n = 15/55 (repeat karyotype). PBMC-iPSCs: n = 2/143 (1st karyotype) and n = 2/63 (repeat karyotype). Percent abnormal at the first G-band karyotypes evaluated for the fib- and PBMC-iPSC clonal lines in relation to the passage number of the iPSCs. [{fib-iPSCs versus PBMC-iPSCs - passages 1-8 (****): z-value = 4.31, two-tailed p-value < 0.0001; passages 9- 23 (***)· z.value = 3 21, two tailed p-value = 0.001; two-proportion z-test}]. fib-iPSCs: n = 20/102 (passages 1-8) and n =12 26/103 (passages 9-23). PBMC-iPSCs: n = 1/99 (passages 1-8) and n = 1/42 (passages 9-23). Percent abnormal at the second - fourth (repeat) G-band karyotypes evaluated for the fib- and PBMC-iPSC clonal lines in relation to the passage number of the iPSCs. [{fib-iPSCs versus

PBMC-iPSCs - low passages 1-14: ns, z-value = 1.53, two-tailed p-value = 0.12; two- proportion z-test}]. [{fib-iPSCs versus PBMC-iPSCs - high passages > 15 (**): z-value = 2.61, two-tailed pl7 value = 0.009; two-proportion z-test}]. fib-iPSCs: n = 7/12 (low passages 1-14) and n = 8/43 (high passages > 15). PBMC-iPSCs: n = 0/2 (low passages 1-14) and n = 2/61 (high passages > 15). ND: none detected. Figure 10(G): The average size of de novo acquired amplifications and deletions (amps/dels) detected by aCGH in Mb of fib- iPSCs (n = 6) and PBMC- iPSCs (n = 8) when compared to each iPSC line's parental cells. [{fib-iPSC versus PBMC-iPSC (**): two-tailed p-value = 0.0015, unpaired t-test}]. The iPSC lines used in aCGH amps/dels size analysis had both abnormal and normal G-band karyotypes. Abnormal: fib-iPSC (n = 3) and PBMC-iPSC (n = 2). Normal: fib-iPSC (n = 3) and PBMC-iPSC (n = 6). The average number of de novo acquired amplifications and deletions detected by aCGH in Mb of fib-iPSCs (n = 6) and PBMC- iPSCs (n = 8) when compared to each iPSC line's parental cells. [{fib-iPSC versus PBMC-iPSC (*): two-tailed p- value = 0.0369, unpaired t-test}]. The iPSC lines used in number of de novo amps/dels analysis with 1 abnormal karyotypes: fib2 iPSC (n = 3) and PBMC-iPSC (n = 2) as well as normal karyotypes: fib-iPSC (n = 3) and PBMC3 iPSC (n = 6). List of most recurrent submicroscopic amps/dels observed in iPSC lines observed in the Inventors' laboratory. Figure 10(H): Frequency of the different karyotype abnormalities in fibroblast-derived iPSCs (fib-iPSCs) at individual chromosome level observed by G-band karyotyping. Rearr. - Structural Rearrangements including translocations, inversions and derivatives. Cells shaded in gray depict chromosomes where karyotype abnormalities were never observed in fib-iPSC cultures. Cells shaded in orange depict the highest ranked (top 3) percent karyotype abnormality for each of the categories of total chromosomal abnormalities, which include gain, loss or structural rearrangements of a chromosome.

Figure 11(A) to Figure 11(C): Transgene-free status of BC-iPSCs Figure 11(A) Lack of plasmid-based EBNA gene expression using a assay in the genomic DNA of the BC- iPSCs. Figure 11 (B): Relative normalized gene expression measured by quantitative RT- PCR analyses using primers detecting endogenous POU5F1 (OCT4), SOX2, LIN28, L-MYC, and KLF4 expression (coding DNA sequence, CDS). Figure 11 (C) as well as plasmid- derived expression (Pla), which is undetectable in the iPSC lines.

Figure 12(A) to Figure 12(B): Chromosomal ideograms of lymphoblastoid cell line (LCL)-iPSCs and epithelial cell-derived iPSCs. Individual chromosome level ideogram representation of abnormal karyotypes from LCL-derived Figure 12(A) and epithelial-derived iPSCs Figure 12(B) in the Inventors' laboratory. Abnormal iPSC karyotypes represented in the ideograms are; (Figure 12(A)) LCL-iPSCs (n: 15) of total 60 LCL-iPSC cultures karyotyped, and (Figure 12(B)) epithelial -iPSCs (n: 7) of total BC-iPSCs karyotyped (n: 26).

Figure 13(A) to Figure 13(B). Tri-lineage potential of PBMC-derived iPSCs. TaqMan hPSC Scorecard table showing the tri-lineage potential of the representative PBMC- derived iPSCs generated in the Inventors' laboratory. Figure 13(A): Expression of selected genes in four groups (self-renewal/pluripotency, ectoderm, endoderm, and mesoderm) is compared for spontaneous in vitro differentiation of embryoid bodies (EBs) derived from each Figure 13(B): PBMC-iPSC line between 14-18 days post-iPSC stage. Negative scores for self-renewal reflect the differentiating status of EBs, while positive scores for each of the 3 germ layers shows relatively equal propensity of a PBMC-derived iPSC line to generate the ectoderm, mesoderm and endoderm without directed differentiation.

Figure 14(A) to Figure 14(C). PBMC-iPSCs reprogrammed with episomal plasmids are transgene-free. Figure 14(A): Lack of plasmid-based EBNA1 gene presence in the genomic DNA of the PBMC-iPSCs. Figure 14(B) and Figure 14(C): Relative normalized gene expression measured by quantitative RT-PCR analyses using primers detecting endogenous POU5F1 (OCT4), SOX2, LIN28, L-MYC, and KLF4 expression (coding DNA sequence, CDS), as well as plasmid-derived expression (Pla), which is undetectable in the PBMC-iPSC lines.

Figure 15(A) to Figure 15(B). PBMC-iPSCs Reprogrammed with 5p Method are Free of SV40LT Gene Integration and Expression. Figure 15(A), Lack of plasmid-based EBNA1 and SV40LT gene presence in the genomic and episomal DNA isolated from PBMC-iPSCs, H9-hESCs, integrated iPSC control lines and a positive control (nucleofected with 5p in PBMCS for 30 days). Figure 15(B): Gene expression measured by quantitative RT-PCR analyses in the same cell lines using primers detecting expression for EBNA1, SV40LT and GAPDH. SV40LT gene expression is undetectable in the reprogrammed PBMC-iPSC lines, when compared to the nucleofected positive control. 32 PCR cycles were used for all primer sets.

Figure 16(A) to Figure 16(B). Chromosomal ideograms of lymphoblastoid cell line (LCL)-iPSCs and epithelial cell-derived iPSCs. Individual chromosome level ideogram representation of abnormal karyotypes from Figure 16(A): LCL-derived (top) and Figure 16(B): epithelial-derived iPSCs (bottom) in the Inventors' laboratory. Abnormal iPSC karyotypes represented in the ideograms are; (top) LCL-iPSCs (n =15) of total 60 LCL-iPSC cultures karyotyped, and (bottom) epithelial-iPSCs (n = 7) of total PBMC-iPSCs karyotyped (n = 26).

Figure 17. p53 analysis. Somatic variants (those appearing in only iPSC clones are identified using realigned and recalibrated sequence files (.bam) generated in the GATK toolkit and the MuTect package. The package accepts processed sequencing reads from paired normal (in this case blood derived DNA) and somatic (in this case iPSC Clone DNA) samples and removes reads with low quality sequence, then applies a Bayesian classifier to identify variants that occur in only the somatic sample before additional quality control steps identify variants as germline or somatic. Identified variants are then annotated with a large SNP database of common variation identified in the population (dbSNP).

Figure 18(A) to Figure 18(B). Additional characterization of reprogramming techniques in fibroblasts using 3P, 5P methods. Figure 18(A) Day 22 post nucleofection Figure 18(B) Day 28 post nucleofection

Figure 19(A) to Figure 19(B). Additional characterization of reprogramming techniques in fibroblasts using 3P, 5P methods. Figure 19(A) AP Stain - 3 Plasmid Condition Figure 19(B) AP Stain - 5 Plasmid Condition.

Figure 20. Frequency of the different karyotype abnormalities in fibroblast- derived iPSCs (fib-iPSCs) at individual chromosome level observed by G-band karyotyping. Structural Rearrangements including translocations, inversions and derivatives. Cells shaded in gray depict chromosomes where karyotype abnormalities were never observed in fib-iPSC cultures. Cells shaded in orange depict the highest ranked (top 3) percent karyotype abnormality for each of the categories of total chromosomal abnormalities, which include gain, loss or structural rearrangements of a chromosome. DETAILED DESCRIPTION

All references cited herein are incorporated by reference in their entirety as though fully set forth. Unless defined otherwise, technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Allen et al, Remington: The Science and Practice of Pharmacy 22 nd ed., Pharmaceutical Press (September 15, 2012); Hornyak et al, Introduction to Nanoscience and Nanotechnology, CRC Press (2008); Singleton and Sainsbury, Dictionary of Microbiology and Molecular Biology 3 rd ed., revised ed., J. Wiley & Sons (New York, NY 2006); Smith, March 's Advanced Organic Chemistry Reactions, Mechanisms and Structure 7 th ed., J. Wiley & Sons (New York, NY 2013); Singleton, Dictionary of DNA and Genome Technology 3 rd ed., Wiley -Blackwell (November 28, 2012); and Green and Sambrook, Molecular Cloning: A Laboratory Manual 4th ed., Cold Spring Harbor Laboratory Press (Cold Spring Harbor, NY 2012), provide one skilled in the art with a general guide to many of the terms used in the present application. For references on how to prepare antibodies, see Greenfield, Antibodies A Laboratory Manual 2 nd ed, Cold Spring Harbor Press (Cold Spring Harbor NY, 2013); Kohler and Milstein, Derivation of specific antibody-producing tissue culture and tumor lines by cell fusion, Eur. J. Immunol. 1976 Jul, 6(7):511-9; Queen and Selick, Humanized immunoglobulins, U. S. Patent No. 5,585,089 (1996 Dec); and Riechmann et al, Reshaping human antibodies for therapy, Nature 1988 Mar 24, 332(6162):323-7.

One skilled in the art will recognize many methods and materials similar or equivalent to those described herein, which could be used in the practice of the present invention. Indeed, the present invention is in no way limited to the methods and materials described. For purposes of the present invention, the following terms are defined below.

As used in the description herein and throughout the claims that follow, the meaning of "a," "an," and "the" includes plural reference unless the context clearly dictates otherwise. Also, as used in the description herein, the meaning of "in" includes "in" and "on" unless the context clearly dictates otherwise.

As described, patient-specific induced pluripotent stem cells ("iPSCs") hold great promise for many applications, including disease modeling to elucidate mechanisms involved in disease pathogenesis, drug screening, and ultimately regenerative medicine therapies, the utility of human iPSCs is contingent upon maintaining quality, genomic integrity and stability. Multiple laboratories have shown recurrent genomic aberrations in human iPSC lines upon long-term culture, with -10-25% rate of karyotype abnormalities derived from various tissue sources.

Maintaining genomic integrity and stability of hiPSC lines is imperative for reliable disease modeling and safe clinical applications of stem cells as a regenerative therapy. Aberrant cytogenetic errors that arise during reprogramming of somatic cells and/or during maintenance, expansion and prolonged culture of hiPSCs will impact the accuracy of in vitro disease modelling or, more critically, the in vivo utility of iPSCs for regenerative medicine. It is important that iPSCs in clinical use are free from cancer-associated genomic aberrations, especially given that several studies have reported chromosomal aneuploidy, translocations, duplications and deletions, and point mutations in iPSCs. The highly aneuploidy human embryonal carcinoma (EC) stem cells, which are the malignant analogues of normal hESCs, typically contain amplified regions of the short arm of chromosome 12 and gains of chromosomes 1, 17 and X.

Increased sub-chromosomal copy number variations (CNVs) with differences in early, intermediate or late passage numbers, deletions associated with tumor-suppressor genes, and duplications of oncogenes have all been reported in iPSCs In most of these studies, the genomic abnormalities in the iPSC lines have been generated from integrating reprogramming methods. To date, however, no systematic and well-controlled (similar reprogramming and stem cell culture methods) reports of recurrent sub-chromosomal abnormalities specific to large numbers of fibroblast- or blood-derived hiPSC lines have been described. The Inventors have developed an iPSC core facility with extensive experience producing, characterizing and differentiating human iPSC lines. This bio-repository currently has over 160 well-characterized individual donor human iPSC lines with multiple clones (2- 6) per cell line. The iPSC Core has systematically monitored the genomic stability of hiPSCs derived in the Core from 450 independent iPSC cultures over multiple passages, which have been derived from fibroblast, peripheral blood mononuclear cells, immortalized lymphoblastoid cell lines and primary epithelial cells.

A frequently used starting source of cells for reprogramming has been dermal fibroblasts isolated from skin biopsies. However, as described, iPSCs derived from skin punch biopsies are more invasive, require a prolonged 2-3 week period of expansion in culture prior to reprogramming. Most importantly, there are greater numbers of mutations associated with epidermal exposure to UV light, raising concerns over the safety of the iPSCs cells derived from skin. Issues with the common fibroblast cell source would be circumvented with peripheral blood, which can be utilized as an easily accessible source of patient tissue for reprogramming. Peripheral blood is the most accessible adult tissue and permits access to numerous frozen samples already stored at blood banks. Additionally, many repositories have stored peripheral or lymphoblast blood with specific genotypes.

Blood appears to an advantageous alternative for iPSC derivation as widely used in clinical diagnostics, involving less invasive methods of collection that is standardized and less traumatic than skin biopsy. The major components of peripheral blood (PB) are red blood cells and platelets. White blood cells are the nucleated cells in PB at concentrations of 3.6-11 x 10 6 /ml. Mature T cells and primary progenitor cells in PB can be readily expanded using established methods and are among the most successfully-used sources for reprogramming. T cells are the most abundant cells after granulocytes in PB (20-30%) and T cells can be readily expanded with IL-2 and anti-CD3/CD28 microbeads. Reprogramming of T cells into pluripotency has been reported by many labs using different approaches for the sake of developing replacements to aged T cells for immunotherapy. However, mature T cells harbor a single T cell receptor (TCR) after somatic recombination and have lost the ability to regenerate the T cell repertoire with unlimited possibilities.

Here the Inventors have developed a reliable protocol to efficiently reprogram blood cells (BCs) including peripheral mononuclear blood cells (PBMCs) into iPSCs (BC-iPSCs) and show that these iPSC lines are superior in terms of cytogenetic stability in comparison to their fibroblast-derived iPSC (Fib-iPSCs) lines obtained from public repositories or local clinics. The Inventors describe methods and cytogenetic stability for derivation of BC-iPSCs from both a lymphoid T cell and a myeloid non-T cell population. The Inventors discovered that hiPSC lines generated from PBMCs using non-integrating methods have greatly lower incidences of genomic aberrations than those generated by integrating methods. Both fib- iPSCs and BC-iPSCs derived from the same somatic cells contained comparable numbers of de novo CNVs. The Inventors' results show that freshly isolated human PBMCs can be faithfully reprogrammed to pluripotency with greater cytogenetic stability using episomal plasmids. As such, PBMCs should be a preferred somatic cell source for iPSC reprogramming to minimize any effects of acquired genomic aberrations and, further, should be considered as an ideal cell source for regenerative medicine.

In contrast to mature T or B cells, the alternative source of blood progenitors contain an intact genome. In addition, they can be expanded in culture conditions that favor the proliferation of myeloid cells or erythroid cells. Blood stem/progenitor cells express surface marker CD34 and reside in the stem cell niche. However, only about 1% stem/progenitor cells enter circulation each day and as a result, only 0.01-0.1% cells in PB are CD34+ cells. This population can be enriched by magnetic-activated cell sorting (MACS) or culture of MNCs for several days can be relied upon to expand CD34+ cells to a 5-20% purity, which can be used for reprogramming without further purification.

Other nucleated peripheral blood cells include granulocytes (mostly neutrophils), monocytes, T lymphocytes, B lymphocytes and a few progenitor cells. Focusing on these constitutes of blood can be achieved by depleting red blood cells and platelet using lysis buffer followed by multiple centrifugations. Ficoll gradient centrifugation can also be utilized to deplete both red blood cells and granulocytes, leading to the enrichment of mononuclear cells (MNCs). Against this backdrop, reprogramming with exogenously expressed factors is notoriously inefficient and requires multiple cell cycles to achieve pluripotency. As such, primary granulocytes, monocytes and B lymphocytes are among the most difficult cells to be reprogrammed due to the lack of reliable protocols to expand these cells. Primary success in this area includes Epstein-Barr virus immortalized lymphoblastoid B cells can be readily expanded in ex vivo culture and thus be reprogrammed to pluripotency.

In view of the above, of great interest is reprogramming of the non-T cell component of blood. Existing techniques are largely unable to reprogram this population from isolated peripheral blood mononuclear cells (PBMCs). More specifically, PBMCs are any peripheral blood cell having a round nucleus. This includes lymphocytes (T cells, B cells, NK cells), monocytes, dendritic cells. Lymphocytes are Small (5-10μm) and Medium (10-18 μm) and constitute 70-90% of PBMCs. Of these cells, 70 - 85% CD3+ T cells (40 -70% of PBMCs), CD4 Helper T cells (25-60% of PBMCs), typically with CD4 to CD 8 ratio of 2: 1, CD8 "Cytotoxic" compartment T cells (5-30% of PBMCs). The remaining compartment includes 5 - 20% B Cells (up to 15% of PBMCs) and 5 - 20% NK Cells (up to 15% of PBMCs). Monocytes are 16-25 μm and 10-30%) of PBMCs (macrophages). Dendritic cells: 1-2% of PBMCs

Here, the Inventors describe a new and reliable nonintegrating episomal plasmid reprogramming method for fresh (non-expanded) peripheral blood mononuclear cell (PBMC) into iPSCs (PBMC-iPSCs). The Inventors show that PBMC-iPSCs produced using this method have superior chromosome-level karyotype stability at only a 1.9% abnormality rate, and exhibit few sub-chromosomal de novo copy number variations. In contrast, episomally reprogrammed iPSC lines derived from pre-expanded dermal fibroblasts (fib-iPSCs), lymphoblastoid cell lines (LCL-iPSCs) or epithelial cells (epi-iPSCs), exhibit over 10-fold greater karyotype instability (23-27%). These findings show that iPSCs reprogrammed from non expanded PBMCs and maintained with the methods described here, have excellent cytogenetic stability and minimal genomic aberrations, suggesting that this could be a preferred method for iPSCs in research and therapeutic clinical applications.

These described approaches allow for use of peripheral blood as a readily accessible resource for cellular reprogramming with superior properties in genomic and karyotype stability avoiding environmental insults for which mutations or other forms of structural alteration would otherwise be therapeutic materials derived therein.

As described, the Inventors have established improved techniques for highly efficient, reproducible reprogramming using non-integrating episomal plasmid vectors, including generation of iPSCs from blood cells, including whole blood and peripheral blood, the resulting reprogrammed pluripotent cells described herein as "BC-iPSCs".

Generally, different approaches for non-integrative reprogramming span at least categories: 1) integration-defective viral delivery, 2) episomal delivery, 3) direct RNA delivery, 4) direct protein delivery and 5) chemical induction. As described further herein, the adoption of episomal vectors allows for generation of iPSCs substantially free of the vectors used in their production, as episomal or similar vectors do not encode sufficient viral genome sufficient to give rise to infection or a replication-competent virus. At the same time, these vectors do possess a limited degree of self-replication capacity in the beginning somatic host cells. This self-replication capacity provides a degree of persistent expression understood to be beneficial in allowing the dedifferentiation process to initiate take hold in a target host cell.

One example of a plasmid vector satisfying these criteria includes the Epstein Barr oriP/Nuclear Antigen-1 ("EBNA1") combination, which is capable of limited self-replication and known to function in mammalian cells. As containing two elements from Epstein-Barr virus, oriP and EBNA1, binding of the EBNA1 protein to the virus replicon region oriP maintains a relatively long-term episomal presence of plasmids in mammalian cells. This particular feature of the oriP/EBNAl vector makes it ideal for generation of integration-free iPSCs.

More specifically, persistent expression of reprogramming factor encoded in an oriP/EBNAl vector occurs across multiple cell divisional cycles. Sufficiently high levels of reprogramming factors across several cell divisions allows for successful reprogramming even after only one infection. While sustained expression of reprogramming factors is understood to be beneficial during initial programming stages, otherwise unlimited constitutive expression would hamper subsequent stages of the reprogramming process. For example, unabated expression of reprogramming factors would interfere with subsequent growth, development, and fate specification of the host cells.

At the same time, a further benefit is the eventual removal of the reprogramming factor transgenes, as a small portion of episomes is lost per cell cycle. This is due to the asymmetric replication capacity of the host cell genome and episomal self-replication and it is estimated that approximately 0.5% of vector is lost per generation. Gradual depletion of plasmids during each cell division is inevitable following propagation leading to a population of integration-free iPSCs. The persistent, yet eventual abrogation of reprogramming factor expression on oriP/EBNAl is highly coincident with the needs for different stages of the reprogramming process and eliminates the need for further manipulation steps for excision of the reprogramming factors, as has been attempted through use of transposons and excisable polycistronic lentiviral vector elements. Although oriP/EBNAl has been applied by others in reprogramming studies, the reported efficiencies are extremely low (as few as 3 to 6 colonies per million cells nucleofected), which may be due, in-part, to reliance on large plasmids encoding multiple reprogramming factors (e.g., more than 12 kb), negatively impacting transfection efficiency.

In addition to these choices in vector designs, the specific combinations of reprogramming factors implemented in the literature have varied. As mentioned, reprogramming factors that have been used include pluripotency-related genes Oct-4, Sox-2, Lin-28, Nanog, Sall4, Fbx-15 and Utf-1. These factors, traditionally are understood be normally expressed early during development and are involved in the maintenance of the pluripotent potential of a subset of cells that will constituting the inner cell mass of the pre- implantation embryo and post-implantation embryo proper. Their ectopic expression of is believed to allow the establishment of an embryonic-like transcriptional cascade that initiates and propagates an otherwise dormant endogenous core pluripotency program within a host cell. Certain other reprogramming determinants, such as Tert, Klf-4, c-Myc, SV40 Large T Antigen ("SV40LT") and short hairpin RNAs targeting p53 ("shRNA-p53") have been applied. There determinants may not be potency-determining factors in and of themselves, but have been reported to provide advantages in reprogramming. For example, TERT and SV40LT are understood to enhance cell proliferation to promote survival during reprogramming, while others such as short hairpin targeting of p53 inhibit or eliminate reprogramming barriers, such as senescence and apoptosis mechanisms. In each case, an increase in both the speed and efficiency of reprogramming is observed. In addition, microRNAs ("miRNAs") are also known to influence pluripotency and reprogramming, and some miRNAs from the cluster have been applied in reprogramming studies. For

example, the introduction of miR-291-3p, miR-294 or miR-295 into fibroblasts, along with pluripotency-related genes, has also been reported to increase reprogramming efficiency.

While various vectors and reprogramming factors in the art appear to present multiple ingredients capable of establishing reprogramming in cells, a high degree of complexity occurs when taking into account the stoichiometric expression levels necessary for successful reprogramming to take hold. For example, somatic cell reprogramming efficiency is reportedly fourfold higher when Oct-4 and Sox2 are encoded in a single transcript on a single vector in a 1 : 1 ratio, in contrast to delivering the two factors on separate vectors. The latter case results in a less controlled uptake ratio of the two factors, providing a negative impact on reprogramming efficiency. One approach towards addressing these obstacles is the use of polycistronic vectors, such as inclusion of an internal ribosome entry site ("IRES"), provided upstream of transgene(s) that is distal from the transcriptional promoter. This organization allows one or more transgenes to be provided in a single reprogramming vector, and various inducible or constitutive promoters can be combined together as an expression cassette to impart a more granular level of transcriptional control for the plurality of transgenes. These more specific levels of control can benefit the reprogramming process considerably, and separate expression cassettes on a vector can be designed accordingly as under the control of separate promoters.

Although there are advantages to providing such factors via a single, or small number of vectors, upper size limitations on eventual vector size do exist, which can stymie attempts to promote their delivery in a host target cell. For example, early reports on the use of polycistronic vectors were notable for extremely poor efficiency of reprogramming, sometimes occurring in less than 1% of cells, more typically less than 0.1%. These obstacles are due, in-part, to certain target host cells possessing poor tolerance for large constructs (e.g., fibroblasts), or inefficient processing of IRES sites by the host cells. Moreover, positioning of a factor in a vector expression cassette affects both its stoichiometric and temporal expression, providing an additional variable impacting reprogramming efficiency. Thus, some improved techniques can rely on multiple vectors each encoding one or more reprogramming factors in various expression cassettes. Under these designs, alteration of the amount of a particular vector for delivery provides a coarse, but relatively straightforward route for adjusting expression levels in a target cell.

A further advantage of the techniques described herein is the use of defined media conditions for the reprogramming process, including the use of ESC media and/or E7 media. While certain additives may be present to spur the reprogramming process (e.g., L-Ascorbic Acid, Transferrin, Sodium Bicarbonate, Insulin, Sodium Selenite and/or bFGF), no serum or animal components are used. In some instances, there may be further benefits in altering the chemical and/or atmospheric conditions under which reprogramming will take place. For example, as the pre-implantation embryo is not vascularized and hypoxic (similar to bone marrow stem-cell niches) reprogramming under hypoxic conditions of 5% O 2 , instead of the atmospheric 21% O 2 , may further provide an opportunity to increase the reprogramming efficiency. Similarly, chemical induction techniques have been used in combination with reprogramming, particularly histone deacetylase (HDAC) inhibitor molecule, valproic acid (VP A), which has been found wide use in different reprogramming studies. At the same time, other small molecules such as MAPK kinase (MEK)-ERK ("MEK") inhibitor PD0325901, transforming growth factor beta ("TGF-β") type I receptor ALK4, ALK5 and ALK7 inhibitor SB431542 and the glycogen synthase kinase-3 ("GSK3") inhibitor CHIR99021 have been applied for activation of differentiation-inducing pathways (e.g. BMP signaling), coupled with the modulation of other pathways (e.g. inhibition of the MAPK kinase (MEK)-ERK pathway) in order to sustain self-renewal. Other small molecules, such as Rho-associated coiled-coil-containing protein kinase ("ROCK") inhibitors, such as Y- 27632 and thiazovivin ("Tzv") have been applied in order to promote survival and reduce vulnerability of pSCs to cell death, particularly upon single-cell dissociation.

In addition to the choice of delivery vectors, reprogramming factor combinations, and conditions for reprogramming, further variations must consider the nature of the host target cell for reprogramming. To date, a wide variety of cells have served as sources for reprogramming including fibroblasts, stomach and liver cell cultures, human keratinocytes, adipose cells, and frozen human monocyte. Clearly, there is a wide and robust potential for dedifferentiation across many tissues sources. Nevertheless, it is widely understood that depending on the donor cell type, reprogramming is achieved with different efficiencies and kinetics. For example, although fibroblasts remain the most popular donor cell type for reprogramming studies, other types of cells such as human primary keratinocytes transduced with Oct-4, Sox-2, Klf-4 and c-Myc have been reported to reprogram 100 times more efficiently and two-fold faster. Additionally, some other cell types, such as cord blood cells, may only require a subset of reprogramming factors, such as Oct-4 and Sox-2 for dedifferentiation to take hold, while neural progenitor cells may only require Oct-4. Without being bound to any particular theory, it is believed that differences in reprogramming efficiency and/or reprogramming factor requirements of specific host cells result from high endogenous levels of certain reprogramming factors and/or intrinsic epigenetic states that are more amenable to reprogramming.

Although these many other sources have been used across studies for the generation of iPSCs, mononuclear cells (MNCs) from peripheral blood (PB) are a highly attractive host cell candidate due to convenience and features as an almost unlimited resource for cell reprogramming. PB cells in particular are relatively easy to isolate (e.g., blood draw) compared to isolation from other sources such as fibroblasts (e.g., skin biopsy). These cells do not require laborious culturing and propagation prior to reprogramming, thereby reducing the overall time from which reprogramming iPSCs can be obtained.

Following successful reprogramming, clonal selection allows for generation of pluripotent stem cell lines. Ideally, such cells possess requisite morphology (i.e., compact colony, high nucleus to cytoplasm ratio and prominent nucleolus), self-renewal capacity for unlimited propagation in culture (i.e., immortal), and with the capability to differentiate into all three germ layers (e.g., endoderm, mesoderm and ectoderm). Further techniques to characterize the pluripotency of a given population of cells include injection into an immunocompromised animal, such as a severe combined immunodeficient ("SCID") mouse, for formation of teratomas containing cells or tissues characteristic of all three germ layers.

As an example of the above, approximately, 5 x 10 6 cells per nucleofection of

PBMCs were nucleofected with either plasmid mixture 4p or plasmid mixture 5p using program V-024 on the Amaxa Nucleofector 2D Device with the Amaxa Human T-cell Nucleofector® Kit. Approximately 1 x 10 6 cells were then seeded into wells of a 6-well plate covered with mitomycin treated mouse embryonic feeder (MEF) layer or coated with 10 μg/ml Laminin-521 (L-521;BioLamina). Each episomal plasmid (Addgene) expressing 7 factors: OCT4, SOX2, KLF4, LMYC, LIN28, SV40LT and p53 shRNA (pEP4 E02S ET2K, pCXLE-hOCT3/4-shp53-F, pCXLEhUL, pCXLE-hSK, and pCXLE-EBNAl). This method has a significant advantage over viral transduction, because exogenously introduced genes do not integrate and are instead expressed episomally in a transient fashion. Cells were plated in 2 mL of either αβ T-cell medium (X- vivo 10 supplemented with 30U/ml IL-2 and 5ul/well Dynabeads Human T-activator CD3/CD28) or non T-cell medium (aMEM supplemented with 10% FBS, lOng/ml IL-3, lOng/ml IL-6, lOng/ml G-CSF and lOng/ml GM-CSF). Two days after nucleofection an equal amount of Primate ESC medium (ReproCell) containing 5 ng/ml bFGF (for MEF condition) or E7 medium (for L-521 condition) was added to the wells without aspirating the previous medium. Beginning on day four, the medium was gently aspirated from each well and 2ml of the appropriate fresh reprogramming media was added to each well. Medium was replaced every other day. At approximately day 18 post nucleofection, individual colonies were observed in all wells of each condition. At approximately day 25 post nucleofection, individual colonies were isolated and sub-cloned into 1 well of 12-well plate containing the appropriate substrate and medium. These nucleofected cells were plated on feeder-independent BD Matrigel™ growth factor-reduced Matrix (Corning/BD Biosciences, #354230). All cultures were maintained at 20% 02 during the reprogramming process. Individual PBMC-iPSC colonies with ES/iPSC-like morphology appeared between day 25-32 and those with best morphology were mechanically isolated, transferred onto 12-well plates with fresh Matrigel™ Matrix, and maintained in mTeSR®l medium. The iPSC clones were further expanded and scaled up for further analysis. All the fibroblasts were reprogrammed with the 3p reprogramming plasmid mixture as described.

Described herein is a composition of blood cell derived induced pluripotent stem cells ("BC-iPSCs"). In certain embodiments, the composition of blood cell derived induced pluripotent stem cells includes cells generated by providing a quantity of blood cells, delivering a quantity of reprogramming factors into the blood cells, culturing the blood cells in a reprogramming media for at least 4 days, wherein delivering the reprogramming factors, and culturing generates the blood cells derived induced pluripotent stem cells. In certain embodiments, the blood cells are T-cells. In other embodiments, the blood cells are non-T- cells. In other embodiments, the blood cells are mononuclear cells (MNCs), including for example peripheral blood mononuclear cells (PBMCs). In other embodiments, the cells are primary granulocytes, monocytes and B lymphocytes.

In certain embodiments, the reprogramming factors are Oct-4, Sox-2, Klf-4, c-Myc, Lin-28, SV40 Large T Antigen ("SV40LT"), and short hairpin RNAs targeting p53 ("shRNA-p53"). In other embodiments, these reprogramming factors are encoded in a combination of vectors including pEP4 E02S ET2K, pCXLE-hOCT3/4-shp53-F, pCXLE- hSK, pCXLE-hUL and pCXWB-EBNAl . This includes, for example, using about 0.5 - 1.0 μg pCXLE-hOCT3/4-shp53, 0.5 - 1.0 μg pCXLE-hSK, 0.5 - 1.0 μg pCXLE-UL, about 0.25 - 0.75 μg pCXWB-EBNAl and 0.5 - 1.0 μg pEP4 E02S ET2K. This includes, for example, using 0.83μg pCXLE-hOCT3/4-shp53, 0.83μg pCXLE-hSK, 0.83μg pCXLE-UL, 0.5μg pCXWB-EBNAl and 0.83 μg pEP4 E02S ET2K, wherein the stoichiometric ratio of SV40LT (encoded in pEP4 E02S ET2K) and EBNA-1 (encoded in pCXWB-EBNAl) supports the reprogramming of non-T cell component of blood, including peripheral blood mononuclear cells. In certain other embodiments, the reprogramming media includes PD0325901, CHIR99021, HA-100, and A-83-01. In other embodiments, the culturing the blood cells in a reprogramming media is for 4-30 days. In various embodiments, the blood cells are plated on a treated cell culture surface after delivering a quantity of reprogramming factors. In various embodiments, treatment includes plating of feeder cells, such as mouse embryonic fibroblasts. In other embodiments, treatment includes coating with extracellular matrix proteins. In various embodiment, extracellular matrix proteins include laminin.

In various embodiments, the BC-iPSCs are capable of serial passaging as a cell line. In various embodiments, the BC-iPSCs possess genomic stability. Genomic stability can be ascertained by various techniques known in the art. For example, G-band karyotyping can identify abnormal cells lacking genomic stability, wherein abnormal cells possess about 10% or more mosaicism, or one or more balanced translocations of greater than about 5, 6, 7, 8, 9, 10 or more Mb. Alternatively, genomic stability can be measured using comparative genomic hybridization (aCGH) microarray, comparing for example, BC-iPSCs against iPSCs from a non-blood cell source such as fibroblasts. Genomic stability can include copy number variants (CNVs), duplications/deletions, and unbalanced translocations. In various embodiments, BC-iPSCs exhibit no more than about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 15, 16, 17, 18, 19, or 20 Mb average size of amplification and deletion. In various embodiments, BC-iPSCs exhibit no more than about 20-30 Mb average size of amplification and deletion. In various embodiments, BC-iPSCs exhibit no more than about 30-40 Mb average size of amplification and deletion. In various embodiments, BC-iPSCs exhibit no more than about 40-50 Mb average size of amplification and deletion. In various embodiments, the average number of acquired de novo amplification and deletions in BC-iPSCs is less than about 5, 4, 3, 2, or 1. For example, de novo amplification and deletions in fib-iPSCs are at least two-fold greater than in PBMC-iPSCs. In different embodiments, reprogramming factors can also include one or more of following: Oct-4, Sox-2, Klf-4, c-Myc, Lin-28, SV40LT, shRNA-p53, nanog, Sall4, Fbx-15, Utf-1, Tert, or a Mir-290 cluster microRNA such as

In different embodiments, the reprogramming factors are encoded by a vector. In different embodiments, the vector can be, for example, a non-integrating episomal vector, minicircle vector, plasmid, retrovirus (integrating and non-integrating) and/or other genetic elements known to one of ordinary skill. In different embodiments, the reprogramming factors are encoded by one or more oriP/EBNAl derived vectors. In different embodiments, the vector encodes one or more reprogramming factors, and combinations of vectors can be used together to deliver one or more of Oct-4, Sox-2, Klf-4, c-Myc, Lin-28, SV40LT, shRNA-p53, nanog, Sall4, Fbx-15, Utf-1, Tert, or a Mir-290 cluster microRNA such as miRD291 D3p, miRD294 or miRD295. For example, oriP/EBNAl is an episomal vector that can encode a vector combination of multiple reprogramming factors, such as pCXLE-hUL, pCXLE-hSK, pCXLE-hOCT3/4-shp53-F, pEP4 E02S T2K and pCXWB-EBNAl .

In other embodiments, the reprogramming factors are delivered by techniques known in the art, such as nuclefection, transfection, transduction, electrofusion, electroporation, microinjection, cell fusion, among others. In other embodiments, the reprogramming factors are provided as RNA, linear DNA, peptides or proteins, or a cellular extract of a pluripotent stem cell.

In various embodiments, the reprogramming media is embryonic stem cell (ESC) media. In various embodiments, the reprogramming media includes bFGF. In various embodiments, the reprogramming media is E7 media. In various embodiments, the reprogramming E7 media includes L-Ascorbic Acid, Transferrin, Sodium Bicarbonate, Insulin, Sodium Selenite and/or bFGF In different embodiments, the reprogramming media comprises at least one small chemical induction molecule. In different embodiments, the at least one small chemical induction molecule comprises PD0325901, CHIR99021, HA- 100, A-83-01, valproic acid (VP A), SB431542, Y-27632 or thiazovivin ("Tzv"). In different embodiments, culturing the BCs in a reprogramming media is for at least 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 days.

In certain embodiments, the BC-iPSCs are derived from blood cells previously isolated from a subject, by for, example, drawing a blood sample from the subject. In other embodiments, the blood cells are isolated from a subject possessing a disease mutation. For example, subjects possessing any number of mutations, such as autosomal dominant, recessive, sex-linked, can serve as a source of blood cells to generate BC-iPSCs possessing said mutation. In other embodiments, the disease mutation is associated with a neurodegenerative disease, disorder and/or condition. In other embodiments, the disease mutation is associated with an inflammatory bowel disease, disorder, and/or condition. In various embodiments, the BC-iPSCs possess features of pluripotent stem cells. Some exemplary features of pluripotent stem cells including differentiation into cells of all three germ layers (ectoderm, endoderm, mesoderm), either in vitro or in vivo when injected into an immunodeficient animal, expression of pluripotency markers such as Oct-4, Sox-2, nanog, TRA-1-60, TRA-1-81, SSEA4, high levels of alkaline phosphatase ("AP") expression, indefinite propagation in culture, among other features recognized and appreciated by one of ordinary skill.

Other embodiments include a pharmaceutical composition including a quantity of blood cells derived induced pluripotent stem cells generated by the above described methods, and a pharmaceutically acceptable carrier.

Also described herein is an efficient method for generating induced pluripotent stem cells, including providing a quantity of cells, delivering a quantity of reprogramming factors into the cells, culturing the cells in a reprogramming media for at least 4 days, wherein delivering the reprogramming factors, and culturing generates induced pluripotent stem cells. In certain embodiments, the cells are primary culture cells. In other embodiments, the cells are blood cells (BCs). In certain embodiments, the blood cells are T-cells. In other embodiments, the blood cells are non-T-cells. In other embodiments, the cells are mononuclear cells (MNCs), including for example peripheral blood mononuclear cells (PBMCs). In other embodiments, the cells are primary granulocytes, monocytes and B lymphocytes.

In certain embodiments, the reprogramming factors are Oct-4, Sox-2, Klf-4, c-Myc, Lin-28, SV40 Large T Antigen ("SV40LT"), and short hairpin RNAs targeting p53 ("shRNA-p53"). In other embodiments, these reprogramming factors are encoded in a combination of vectors including pEP4 E02S ET2K, pCXLE-hOCT3/4-shp53-F, pCXLE- hSK, pCXLE-hUL and pCXWB-EBNAl . This includes, for example, using about 0.5 - 1.0 μg pCXLE-hOCT3/4-shp53, 0.5 - 1.0 μg pCXLE-hSK, 0.5 - 1.0 μg pCXLE-UL, about 0.25 - 0.75 μg pCXWB-EBNAl and 0.5 - 1.0 μg pEP4 E02S ET2K. This includes, for example, using 0.83μg pCXLE-hOCT3/4-shp53, 0.83μg pCXLE-hSK, 0.83μg pCXLE-UL, 0.5μg pCXWB-EBNAl and 0.83 μg pEP4 E02S ET2K, wherein the stoichiometric ratio of SV40LT (encoded in pEP4 E02S ET2K) and EBNA-1 (encoded in pCXWB-EBNAl) supports the reprogramming of non-T cell component of blood, including peripheral blood mononuclear cells. In various embodiments, the reprogramming media is embryonic stem cell (ESC) media. In various embodiments, the reprogramming media includes bFGF. In various embodiments, the reprogramming media is E7 media. In various embodiments, the reprogramming E7 media includes L-Ascorbic Acid, Transferrin, Sodium Bicarbonate, Insulin, Sodium Selenite and/or bFGF. In different embodiments, the reprogramming media comprises at least one small chemical induction molecule. In certain other embodiments, the reprogramming media includes PD0325901, CHIR99021, HA-100, and A-83-01. In other embodiments, the culturing the blood cells in a reprogramming media is for 4-30 days.

In various embodiments, the BC-iPSCs are capable of serial passaging as a cell line. In various embodiments, the BC-iPSCs possess genomic stability. Genomic stability can be ascertained by various techniques known in the art. For example, G-band karyotyping can identify abnormal cells lacking genomic stability, wherein abnormal cells possess about 10% or more mosaicism, or one or more balanced translocations of greater than about 5, 6, 7, 8, 9, 10 or more Mb. Alternatively, genomic stability can be measured using comparative genomic hybridization (aCGH) microarray, comparing for example, BC-iPSCs against iPSCs from a non-blood cell source such as fibroblasts. Genomic stability can include copy number variants (CNVs), duplications/deletions, and unbalanced translocations. In various embodiments, BC-iPSCs exhibit no more than about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 15, 16, 17, 18, 19, or 20 Mb average size of amplification and deletion. In various embodiments, BC-iPSCs exhibit no more than about 20-30 Mb average size of amplification and deletion. In various embodiments, BC-iPSCs exhibit no more than about 30-40 Mb average size of amplification and deletion. In various embodiments, BC-iPSCs exhibit no more than about 40-50 Mb average size of amplification and deletion. In various embodiments, the average number of acquired de novo amplification and deletions in BC-iPSCs is less than about 5, 4, 3, 2, or 1. For example, de novo amplification and deletions in fib-iPSCs are at least two-fold greater than in PBMC-iPSCs. In various embodiments, the methods produces iPSC cell lines collectively exhibiting about 20%, 15%, 10%, 5% or less abnormal karyotypes over 4-8, 9- 13, 13-17, 17-21, 21-25, or 29 or more passages when serially passaged as a cell line.

In different embodiments, reprogramming factors can also include one or more of following: Oct-4, Sox-2, Klf-4, c-Myc, Lin-28, SV40LT, shRNA-p53, nanog, Sall4, Fbx-15, Utf-1, Tert, or a Mir-290 cluster microRNA such as

In different embodiments, the reprogramming factors are encoded by a vector. In different embodiments, the vector can be, for example, a non-integrating episomal vector, minicircle vector, plasmid, retrovirus (integrating and non-integrating) and/or other genetic elements known to one of ordinary skill. In different embodiments, the reprogramming factors are encoded by one or more oriP/EBNAl derived vectors. In different embodiments, the vector encodes one or more reprogramming factors, and combinations of vectors can be used together to deliver one or more of Oct-4, Sox-2, Klf-4, c-Myc, Lin-28, SV40LT, shRNA-p53, nanog, Sall4, Fbx-15, Utf-1, Tert, or a Mir-290 cluster microRNA such as

For example, oriP/EBNAl is an episomal vector that can encode a

vector combination of multiple reprogramming factors, such as pCXLE-hUL, pCXLE-hSK, pCXLE-hOCT3/4-shp53-F, pEP4 E02S T2K and pCXWB-EBNAl .

In other embodiments, the reprogramming factors are delivered by techniques known in the art, such as nuclefection, transfection, transduction, electrofusion, electroporation, microinjection, cell fusion, among others. In other embodiments, the reprogramming factors are provided as RNA, linear DNA, peptides or proteins, or a cellular extract of a pluripotent stem cell. In certain embodiments, the cells are treated with sodium butyrate prior to delivery of the reprogramming factors. In other embodiments, the cells are incubated or 1, 2, 3, 4, or more days on a tissue culture surface before further culturing. This can include, for example, incubation on a Matrigel coated tissue culture surface. In other embodiments, the reprogramming conditions include application of norm-oxygen conditions, such as 5% O 2 , which is less than atmospheric 21% O 2 .

In various embodiments, the reprogramming media is embryonic stem cell (ESC) media. In various embodiments, the reprogramming media includes bFGF. In various embodiments, the reprogramming media is E7 media. In various embodiments, the reprogramming E7 media includes L-Ascorbic Acid, Transferrin, Sodium Bicarbonate, Insulin, Sodium Selenite and/or bFGF. In different embodiments, the reprogramming media comprises at least one small chemical induction molecule. In different embodiments, the at least one small chemical induction molecule comprises PD0325901, CHIR99021, HA- 100, A-83-01, valproic acid (VP A), SB431542, Y-27632 or thiazovivin ("Tzv"). In different embodiments, culturing the BCs in a reprogramming media is for at least 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 days. Efficiency of reprogramming is readily ascertained by one of many techniques readily understood by one of ordinary skill. For example, efficiency can be described by the ratio between the number of donor cells receiving the full set of reprogramming factors and the number of reprogrammed colonies generated. Measuring the number donor cells receiving reprogramming factors can be measured directly, when a reporter gene such as GFP is included in a vector encoding a reprogramming factor. Alternatively, indirect measurement of delivery efficiency can be provided by transfecting a vector encoding a reporter gene as a proxy to gaμge delivery efficiency in paired samples delivering reprogramming factor vectors. Further, the number of reprogrammed colonies generated can be measured by, for example, observing the appearance of one or more embryonic stem cell-like pluripotency characteristics such as alkaline phosphatase (AP)-positive clones, colonies with endogenous expression of transcription factors Oct or Nanog, or antibody staining of surface markers such as Tra-1-60. In another example, efficiency can be described by the kinetics of induced pluripotent stem cell generation. For example, efficiency can include producing cell lines of normal karyotype, including the method producing iPSC cell lines collectively exhibiting about 20%, 15%, 10%, 5% or less abnormal karyotypes over 4-8, 9-13, 13-17, 17-21, 21-25, or 29 or more passages when serially passaged as a cell line.

Example 1

General iPSC Reprogramming Protocol for Blood

Generally, the reprogramming method involves the following steps. Frozen peripheral blood mononuclear cells (PBMCs), or freshly isolated PBMCs can be used. Treated cell culture surfaces can be used for plating thawed or freshly isolated PBMS. This includes, for example, cell culture surfaces treated with mouse embryonic feeders (MEF) or extracellular matrix laminin L-521A. An expression plasmid mixture is provided, with the plasmid mixture encoding a combination of pluripotency factors. Plasmids are introduced to PBMCs, by using for example, nucleofection, and thereafter placed on the treated cell culture surface. After 2 days of culturing, reprogramming media is introduced, and replenished until colonies are formed. Example 2

Peripheral Blood Collection and Minimal Processing for Reprogramming

The Inventors have established processes to isolate lymphocytes from freshly collected or commercial sources of human or mammalian peripheral blood (PB). The preferred format for the collection and shipment of such samples is the CPT tubes. This format allows the supplier to centrifμge the vacutainer(s) and separate the red blood cells from the plasma components prior to shipping. The protocol provides for the isolation of PBMCs from the plasma layer of the CPT tubes.

Optional tests: Prior to opening the Vacutainer, several disease state tests can be performed, including testing for HIV, Hepatitis B and C, and syphilis. The latter test results should be made available within 1-3 working days from the collection date. If there is a positive result, the sample(s) will be properly discarded, and all laboratory equipment that was used to process the samples will be decontaminated. Example 3

Collection Procedure

(1) Red blood cells and platelets are depleted using containers. BD Vacutainer® CPT™ Tube with Sodium Citrate should be at room temperature (18-25°C) and properly labeled for patient identification. Spray the CPT Vacutainer lightly with 70% isopropanol/ethanol, wipe the alcohol off.

(2) Blood collection is into three (3) 8ml CPT tubes per sample using the standard technique for BD Vacutainer® Evacuated Blood Collection Tubes. After collection, tube is stored upright at room temperature until centrifμgation. Blood samples should be centrifμged within two hours of blood collection for best results.

(3) Tube/blood sample is centrifμged at room temperature (18-25°C) in a horizontal rotor (swing-out head) for a minimum of 20 minutes (up to 30 minutes) at 1500 to 1800 RCF (Relative Centrifμgal Force).

(4) For blood shipped in CPT tubes and reprogramming is performed within 24 hours: After centrifμgation, mononuclear cells and platelets will be in a whitish layer just under the plasma layer (see Figure 1). An alternative procedure for recovering the separated mononuclear cells is to resuspend the cells into the plasma by inverting the unopened BD Vacutainer® CPT™ Tube gently 5 to 10 times. This is the preferred method for storing or transporting the separated sample for up to 24 hours after centrifμgation. Example 4

Important Parameters

Temperature: Since the principle of separation depends on a density gradient, and the density of the components varies with temperature, the temperature of the system should be controlled between 18-25°C during separation.

Centrifμgation: Since the principle of separation depends on the movement of formed elements in the blood throμgh the separation media, the "RCF" should be controlled at 1500 RCF to 1800 RCF. The time of centrifμgation should be a minimum of 20 minutes. (As noted in the trouble shooting section, some specimens may require up to 30 minutes for optimal separation.) Centrifμgation of the BD Vacutainer® CPT™ Tube up to 30 minutes has the effect of reducing red blood cell contamination of the mononuclear cell fraction. Centrifμgation beyond 30 minutes has little additional effect. The BD Vacutainer® CPT™ Tube may be re-centrifμged if the mononuclear "band" or layer is not disturbed.

Time: Blood samples should be centrifμged or separated within two hours of blood drawing. Red blood cell contamination in the separated mononuclear cell fraction increases with longer delays in sample separation. Mononuclear cell recovery decreases with increased time delay before centrifμgation, falling to approximately 40% mononuclear cell recovery at 24 hours. Pour the contents (the plasma layer) off into an appropriately labeled 50 mL tube.

Example 5

Preprogramming Procedure Materials and Supplies

Described herein is the procedure for reprogramming separated whole blood samples (peripheral blood mononuclear cells - PBMCs) for iPSC generation. Materials for use include: Sterile 1.5ml Eppendorf tubes; Sterile pipette tips (lOOOul, 200ul, lOul); Amaxa Nucleofector™ 2b Device (Lonza); Human T-Cell Nucleofector Kit (Lonza, Cat #VPA- 1002); Prepared MEF 6-well TC Plates or L-521 Coated Plates; 0.22um SteriFlip (optional); Primate ESC Medium or E7 Medium, described below in Tables 1-4.

Reprogramming Plasmids used include (1) pCXLE-hOCT3/4-shp53; (2) pCXLE- hSK; (3) pCXLE-UL; (4) pCXWB-EBNAl; (5) pEP4 E02S ET2K. L-Ascorbic Acid 64μg/ml

Transferrin 10.7μg/ml

Sodium Bicarbonate 543μg/ml

Insulin 19.4μg/ml

Sodium Selenite 14ng/ml

bFGF lOOng/ml

Example 6

Reprogramming Procedure

Day -1 - Preparation of plates

MEF Plates - 1. Coat each well of 6-well plate with 1ml of 0.1% Gelatin. 2. Incubate plate at 37°C for a minimum of 1 hour. 3. Obtain a vial of frozen MEFs from the LN2 tank. 4. Thaw vial in a water bath by gently moving the frozen vial in a figure 8 motion in the water. 5. Collect thaw cells into a 15ml conical. 6. Slowly add MEF media to bring the volume to lOmls. 7. Centrifμge tube/MEFs at room temperature (18-25°C) at 1000 to 1200 RPM (Relative Centrifμgal Force) for 5 minutes. 8. While the cells are spinning, aspirate the gelatin from the 6-well plate and add 1ml of MEF media to each well. 9. After the centrifμged has stopped, remove the conical and aspirate the supernatant without disturbing the cell pellet. 10. Resuspend the cell pellet in enoμgh MEF media to achieve approximately 50,000cells/ml. 11. Add 1ml of cell/MEF media mixture to each well. 12. Swirl plate to ensure even distribution of MEFs and place in incubator overnight. 13. Prepare lOμg/ml of L- 521 by thawing one 1ml vial of lOOμg/ml L-521 and adding the contents to 9mls of sterile PBS. 14. Filter the mixture throμgh a 0.22um filter or SteriFlip. 15. Add 1ml of the lOμg/ml L-521 mixture to each well of a 6-well plate. 16. Wrap the plate with Parafilm and place in the 37oC incubator for 2 hours. 17. Remove plate from the incubator and place in 4oC fridge.

Day 0 - Cell Preparation of Frozen PBMCs

18. Thaw 2 frozen vials of PBMCs containing 5e6 cells each in the water bath using a "figure 8" motion until a small ball of ice remains. 19. Collect the contents of each tube into two separate sterile 15ml corneals and label the tubes 1 and 2. 20. Add sterile PBS to each tube to bring the volume to lOmls. Mix cells by inverting tube 5 times. 21. Centrifμge for 15 minutes at 1000 RPM. 22. Proceed to step 25 Cell Preparation of Freshly Isolated PBMCs

23. Place 3e6 freshly isolated PBMCs into 2 sterile 15ml corneals each. 24. Perform steps 19 to 22.

Reprogramming

If using L-521 coated plates, one allows plates to equilibrate to room-temperature for at least 1 hr. 25. Prepare a 1.5ml Eppendorf tubes of Lonza Nucleofection solution according to protocol (82ul of Nucleofection Solution + 18ul of Supplement per reaction). Label the tube NS.

NOTE: One will be performing each reaction twice; once for T-cell conditions and once for non T-cell conditions. It is okay to prepare a master mix of NS for all reactions.

26. In an 1.5ml Eppendorf tube, prepare 3.83μg of expression plasmid mixture for each reaction as follows: i. pCXLE-hOCT3/4-shp53 : 0.83μg

ii. pCXLE-hSK : 0.83μg

iii. pCXLE-UL: 0.83μg

iv. pCXWB-EBNAl : 0.5μg

v. pEP4 E02S ET2K: 0.83μg

27. Prepare a 15ml conical with 6mls T-cell media. 28. Prepare a 15ml conical with 6mls non T-cell media. 29. Aspirate the supernatant from one of the 15ml corneals containing your cells. 30. Gently flick the tube to loosen up the cell pellet. 31. Add lOOul of NS mixture to your cell pellet and gently pipette up and down 3-4 times. 32. Transfer the total volume of NS/cell mixture to a 1.5ml Eppendorf tube containing your plasmid mixture and mix by gently pipetting up and down 3-4 times. 33. Transfer the total volume of NS/cell/plasmid mixture to a glass cuvette provided with the Lonza T-cell kit. 34. Place the cuvette in the Amaxa nucleofector and run program V-024. 35. Using the plastic pipette tip provided with the Lonza T-cell kit, transfer a small amount of T-cell media to the glass cuvette then transfer the entire contents of the cuvette to the 15ml conical containing 6mls of T-cell media. 36 Repeat steps 29 - 34 for your second 15ml conical containing cells, proceed to step 37. 37. Using the plastic pipette tip provided with the Lonza T-cell kit, transfer a small amount of non T-cell media to the glass cuvette then transfer the entire contents of the cuvette to the 15ml conical containing 6mls of non T-cell media. Plate Down

MEF/ Primate ESC condition

38. Remove the MEF media from each of the wells. 39. Rinse each well with 1ml of sterile PBS. 40. Transfer the entire contents of the 15ml conical from step 35 to the top 3 wells by adding 2mls to each well. 41. Add 5ul of Dynabeads Human T-activator CD3/CD28 to each well. 42. Transfer the entire contents of the 15ml conical from step 37 to the bottom 3 wells by adding 2mls to each well. 43. Place the plate in a 37°C incubator.

L-521/ E7 Condition

44. Aspirate the L-521 from each well. 45. Transfer the entire contents of the 15ml conical from step 5.35 to the top 3 wells by adding 2mls to each well. 46. Add 5ul of Dynabeads Human T-activator CD3/CD28 to each well. 47. Transfer the entire contents of the 15ml conical from step 37 to the bottom 3 wells by adding 2mls to each well. 48. Place the plate in a 37°C incubator. Day 2

49. On day 2 post nucleofection, add 2mls of the appropriate reprogramming media (Primate ESC + bFGF or E7) to each well DO NOT ASPIRATE ANYTHING FROM THE WELLS. Day 4-25+

50. Beginning on day 4, gently aspirate the media from each well and add 2mls fresh reprogramming media to each well. This will be done every other day.

Day 25+

51. Using a pulled glass pipette, isolate individual colonies and transfer single colonies into 1 well of 12-well dish containing the appropriate substrate.

Example 7 A variety of quality control assays can be performed to confirm proper generation of pluripotent stem cells. Examples of such tests exemplifying desirable characteristics of pSC generation are presented in Table 1.

Example 8

Alkaline Phosphatase Staining

For characterization of BC-iPSCs, Alkaline Phosphatase staining can be performed using the Alkaline Phosphatase Staining Kit II (Stemgent, Cat no. 00-0055) according to the manufacturer's instructions.

Example 9

Immunohisto/cytochemistry

For further characterization of BC-iPSCs, BC-iPSCs or differentiated cells can be plated on glass coverslips or optical -bottom 96-well plates (Thermo, # 165305) and subsequently fixed in 4% paraformaldehyde. Intestinal organoids can be fixed in 4% paraformaldehyde, transferred to 30% sucrose, embedded in HistoPrep (Thermo Fisher Scientific) and cut into 20 μm sections. All cells are blocked in 510% goat or donkey serum with 0.1%) Triton X-100 and incubated with primary antibodies either for either 3hrs at room temperature or overnight at 4°C. Cells are then rinsed and incubated in species-specific AF488 or AF594-conjμgated secondary antibodies followed by Hoechst 33258 (0.5 μg/ml; Sigma) to counterstain nuclei.

Antibodies suitable for immunocytochemistry and immunoblotting include: (as listed by antigen, dilution, catalog no., isotype, and manufacturer) SSEA4, 1 :250, MAB4304, mIgG3, Millipore; TRA-1-60, 1 :250, 09-0010, IgM, Stemgent; TRA-1-81, 1 :250,09-0011, mlgM, Stemgent; OCT4, 1 :250, 09-0023, Rabbit IgG, Stemgent; NANOG, 1 :250, 09-0020, Rabbit IgG, Stemgent; SOX2,1 :500, AB5603, Rabbit IgG, Millipore; TuJl (p3-tubulin), 1 : 1000, T8535, mIgG2b, Sigma; CDX2, 1 :500, NBP1-40553, IgG, Novus; FABP2, 1 :500, AF3078, IgG, R & D systems; Collagen Type 1, 1 :500, 600-401-103-0.1, Rabbit Rockland; CD73, 1 :500, 550257, mlgGl, BD Pharmingen; NKX6.1, 1 : 100 F55A10, mlgGl, DSHB Iowa; HB9, 1 :25, 81.5C10, mlgGl, DSHB Iowa; ISELT1, 1 :250, AF1837, Goat IgG, R & D systems; SMI32, 1 : 1000, SMI-32R, mlgGl, Covance; CHAT, 1 :250, AB 144P, Goat IgG, Millipore; SMN, 1 :250, 610647, mlgGl, BD Biosciences; Cox-IV, 1 : 1000, 4850s, Rabbit Cell signaling; GAPDH, 1 : 1000, ab9484, mIgG2b, Abeam.

Example 10

Flow Cytometry

Additional characterization of can include flow cytometry. Specifically, BC-iPSCs are dissociated into a single cell suspension using Accutase (Millipore, #SCR005). Surface staining of IPSCs is carried out using SSEA4 (R&D Systems, FAB1435A).

Cells were then fixed, permeabilized and stained intracellularly for Oct3/4 (BD Pharmingen, 560186). Recommended isotypes are used according to the antibodies recommendation (R&D Systems, FABIC007 and BD Pharmingen, 562547). All samples re analyzed using a BD LSRFortessa flow cytometer using BD FACSDiva software. All images are generated using FloJo software.

Example 11

Karyotype

Karyotyping can also be performed as follows. Human BC-iPSCs are incubated in Colcemid (100 ng/mL; Life Technologies) for 30 minutes at 37°C and dissociated using TrypLE for 10 minutes. They can be washed in phosphate buffered saline (PBS) and incubated at 37°C in 5mL hypotonic solution (lg KC1, lg Na Citrate in 400mL water) for 30 minutes. The cells can be centrifμged for 2.5 minutes at 1500 RPM and resuspended in fixative (methanol: acetic acid, 3 : 1) at room temperature for 5 minutes. This is repeated twice, and finally cells were resuspended in 500 μΐ of fixative solution and submitted to the Cedars-Sinai Clinical Cytogenetics Core for G-Band karyotyping.

Example 12 PluriTest

Pluritest provides a robust measurement of pluripotency. Total RNA can be isolated using the RNeasy Mini Kit (Qiagen) and subsequently run on a Human HT-12 v4 Expression BeadChip Kit (Illumina). The raw data file (idat file) was subsequently uploaded onto the Pluritest widget online (www.pluritest.org).

Example 13

Quantitative RT-PCR

Total RNA was isolated using the RNeasy Mini Kit (Qiagen), and 1 μg of RNA was used to make cDNA using the transcription system (Promega). qRT-PCR was performed using specific primer sequences (Table 2) under standard conditions. "CDS" indicates that primers designed for the coding sequence measured expression of the total endogenous gene expression only, whereas "Pla" indicates that primers designed for the plasmid transgene expression only. Data are represented as mean ± SEM

Table 2. qRT-PCR Primer Sequences

Table 4B. Cytogenetic Stability of PBMCs

* De Novo or deletions are those newly acquired amplifications or deletions during reprogramming and/or expansion of the respective iPSC lines when compared to the parent donor tissue aCGH profile

A variety of results demonstrating superior properties of BC-iPSCs are shown, including specific cell lines. High Success rate of peripheral blood mononuclear cell (PBMC)-based episomal reprogramming on MEFs is shown in Table 5. Frequency of different types of karyotype abnormalities at individual; chromosomal level observed by G- band karyotyping in fibroblast-derived iPSCs. Rearr. - Rearrangement; Inv. - Inversion; Der. - derivatives is shown in Table 6. Success rate of blood-based reprogramming on human recombinant laminin 521 in defined E7 reprogramming media is shown in Table 7. Lack of success of blood-based episomal reprogramming on Matrigel substrate and defined E7 reprogramming media is shown in Table 8.

NP: Not performed

T: T cell reprogramming method

NT: non-T cell reprogramming method

4P/5P: reprogramming factors on 4 or 5 plasmids

NP: Not performed

T: T cell reprogramming method

NT: non-T cell reprogramming method

4P/5P: reprogramming factors on 4 or 5 plasmids

Example 16

PBMCs isolated from a blood draw are reliably reprogrammed to iPSCs Dermal fibroblasts obtained from a skin biopsy have been the most common source of iPSC reprogramming material, due largely to their inexpensive and relatively easy usage. However, compared to a blood draw, a skin punch biopsy can be between the size of 3-4 mm and is often a painful procedure that requires local anesthetics. Compared to skin biopsy- derived fibroblasts, blood is a more accessible source of patient tissue during hospital visits and, therefore, a preferred choice of source tissue for iPSC reprogramming by both patients and clinicians. Various human blood reprogramming methods have been reported with mixed efficiencies, including CD34 + cells mobilized from peripheral blood (ref). Unless isolated from cord blood, CD34 + cells are usually isolated from the peripheral blood or bone marrow of patients undergoing G-CSF mobilization for several days. In order to generate sufficient cell numbers for reprogramming, isolated CD34 + progenitor cells need to be enriched and expanded in culture with complex and expensive protocols. The Inventors were successful in reprogramming PBMCs that were freshly isolated from a peripheral blood draw without prior cell expansion or CD34 + cell isolation using episomal reprogramming plasmids. Such an approach provides the fastest and most cost effective procedure for obtaining a patient's reprogrammed iPSCs from the time of collecting a patient bio-specimen to generating their rigorously characterized iPSCs.

Reprogramming large cohorts of PBMCs from multiple subjects simultaneously as well as iPSC line generation at a much later date after a patient blood draw that allows for iPSC "future-proofing", requires a PBMC cryopreservation step to recover viable cells reliably. Additionally, this process is flexible because selective cohorts of well cryopreserved PBMCs from representative patients could be converted to iPSCs when greater patient genotype-phenotype information is available. However, when cryopreserved PBMCs from multiple individuals were reprogrammed to iPSCs with episomal plasmids expressing POU5F1, SOX2, KLF4, LIN28, L-MYC, p53 shRNA, the Inventors observed significant variability in isolating identifiable iPSC clones even after 35-40 days, regardless of PBMC cell type, ECM substrata and media). The T reprogramming method was successful with only 33% of PBMC samples, while the non-T method on cryopreserved PBMCs did not result in any clonal iPSC lines (data not shown).

Critically, the reliability of PBMC reprogramming in the Inventors' hands was improved significantly when the Inventors utilized, (a) two additional episomal plasmids containing SV40 large T antigen (SV40LT) and EBNA-1 in specific stoichiometry to minimize PBMC cell death and sustain reprogramming factor plasmid expression and (b) a defined reprogramming media to promote high surface attachment of the nucleofected PBMCs (Fig. 8A). This novel protocol resulted in successful and efficient generation of multiple adherent PBMC-iPSC clones that could be mechanically isolated and scaled up for expansion after 25-35 days post-nucleofection (Fig. 8 A). Importantly, the success rate in reprogramming multiple individual cryopreserved PBMCs from unaffected controls or diseased patients' was 90% for the T cell method and 83% for the non-T cell method (Table 5) on mouse embryonic fibroblast feeders (MEFs). Given the limitations imposed by T cell- iPSCs and the low efficiencies for PBMC-derived non-T cell reprogramming, this method resulted in reliable reprogramming of non-T cell derived BC-iPSCs.

The Inventors next tested whether this new protocol was also amenable when using reprogramming methods that are more clinically compatible. Use of recombinant human laminin 521 substrate and chemically defined reprogramming media resulted in successful PBMC-reprogramming, albeit at a slightly lower success rate at 75% and 58%, respectively. The average reprogramming efficiency for T-cell and non-T cell method was 2.2 x 10 "4 % and 1.6 x 10 "6 %, respectively (Table 5). All the PBMC-iPSC lines exhibited typical PSC characteristics, including tightly packed colonies, high cell nuclear-cytoplasmic ratio, robust alkaline phosphatase activity, and expression of pluripotency antigens (Fig. 8B). The BC- iPSCs passed pluripotency quality control metrics determined by the PluriTest assay, demonstrating that the PBMC-iPSC transcription profile was analogous to well established hESCs and fib-iPSCs, but not differentiated fibroblasts and neural progenitor cells (Fig. 8C). BC-iPSCs maintained a normal G-band karyotype (Fig. 8D) and were confirmed to be clonal derivatives of either T cells or non-T cells from the PBMCs using the T cell receptor (TCR)-β and -γ, gene rearrangement/clonality assays (Fig. 8E). The trilineage potential of BC-iPSCs was demonstrated by spontaneous embryoid body formation and by measuring germ-layer specific gene expression profile by the TaqMan Scorecard assay (Fig. 8F). The "footprint- free" status of BC-iPSCs was confirmed by demonstration of endogenous pluripotency gene expression and absence of exogenous reprogramming transgenes using RT-qPCR (Fig. 11 A). The EBNA plasmid-related latency element was eventually eliminated from the established BC-iPSCs (Fig. 11B). Example 17

BC-iPSCs have equivalent neuronal differentiation compared to fib-iPSCs

It has been reported that iPSCs, regardless of the source tissue, ultimately lose most of their gene expression and epigenetic profiles related to the original cell source. However, it remains unclear whether blood-derived iPSCs can differentiate as efficiently as fibroblast- derived iPSCs into other various cell types, possibly due to a stronger retention of epigenetic memory in blood-sourced iPSCs. The Inventors addressed this by directing blood or fibroblast-derived iPSCs, both of mesodermal origin, to a different germ layer such as ectoderm. Specifically, the Inventors performed neural ectoderm differentiation from large numbers of fibroblast (n=26) and PBMC lines (n=8), from both healthy volunteers as well as diseased patients. Based on immunocytochemistry for neural ectoderm markers of TUBB3 (β 3 -tubulin) and NKX6.1 and subsequent cell counts, neuronal differentiation was shown to occur at a similar efficiency between fibroblast and BC-iPSCs (Fig. 8G), further demonstrating the utility of BC-iPSCs.

Example 18

BC-iPSCs maintain a significantly more stable karyotype compared to fib-iPSCs Recurrent chromosomal abnormalities have been described previously for an abundant number of hESC lines. A few reports have chronicled common chromosomal aberrations for hiPSC lines, however, these did not methodically account for variability in source tissues, reprogramming methods and cell culture methods. To the Inventors' knowledge, there have been no systematic studies describing cytogenetic analysis comparing frequency between iPSCs derived from fibroblast and blood. Over the past six years the Inventors performed routine cytogenetic analysis on iPSC lines derived from 104 unique fibroblast or PBMC donors, which includes 339 independent clonal iPSC lines. All fibroblast and blood-derived iPSCs assessed were reprogrammed using the similar non-integrating episomal reprogramming and standard feeder-free matrigel/mTeSR cell culture methods.

Althoμgh fib-iPSCs and BC-iPSCs derived in the iPSC Core resemble each other with regard to their reprogramming methods, pluripotency and differentiation, an early observation of striking differences in cytogenetic stability between the two source cell types prompted the need for large-scale comparative analyses. Many laboratories and stem cell repositories perform routine quality control assays of cytogenetic changes in PSCs using G-band karyotyping by obtaining cell metaphases and Giemsa staining of the chromosomes. This method can readily identify small subpopulations of abnormal cells where reliable identification of 5% abnormal cells is readily achieved, thus allowing detection of mosaicism (>10%) and balanced translocations at low resolution (>5-10 Mb).

The Inventors performed karyotype analysis on 364 human iPSC cultures where source fibroblasts and PBMCs were collected from multiple laboratories or public repositories. The Inventors' data reveal remarkable differences in the incidence of chromosomal aberrations in fib-iPSCs and BC-iPSCs. Abnormal karyotypes with clonal aberrations were observed in 59 of 258 (22.9%) cultures from clonally independent human fib-iPSC lines, derived from 78 unique donors (Fig. 6). In stark contrast, only 4 in 106 (3.8%)) cultures of clonal human PBMC-iPSC lines derived from 32 unique donors displayed low-frequency abnormalities. It is evident from the ideograms, schematic representation of the chromosomes, that recurrent aberrations are represented in much greater degree in in fib- iPSCs compared to the BC-iPSCs (Fig. 6). Indeed, this remarkable cytogenetic stability is unique to PBMCs, as iPSCs reprogrammed from other cell origins, including lymphoblastoid cell lines (25%) and primary epithelial cells (27%) have similar high rates of karyotypes aberrations (Fig 12).

The most frequent karyotypic abnormalities in fib-iPSCs were observed in chromosomes 13 (13.2%), 1 (11.8%), 12 (11.1%), and X (10.4%) (Table 6). To a lesser extent karyotype changes were also observed in chromosomes 21, 11, and 6, in descending order of frequency. Of the aneuploidies, chromosomal gains (trisomy or duplications) were most commonly observed in 12 (21.1%), 1 (15.8%), 11 (13.2%), and 6 (10.5%) and chromosomal losses were repeatedly observed in chromosomes X (38.5%) and 21 (23.1%). Translocation rearrangements in chromosomes 1 and 13 (13.9%) and 14 (11.1%) in fib-iPSCs were also observed. BC-iPSCs had one line each display chromosome gains in 14 (trisomy) or 18 (duplication). The other two PBMC-iPSC lines had exhibited uncommon abnormalities: a translocation 46,XY,t(7; 14)(q34;ql l .2) and a mosaic deletion 46,X,del(X)(q22q26) in 19% of cells (Table 6).

Example 19

Fib- and BC-iPSCs karyotype distribution relates not to the donor age but rather to passage number

The skin, unlike the blood, is routinely exposed to external environmental elements like potential sun damage and the fibroblasts derived from the biopsies require a certain level of expansion prior to reprogramming, which is akin to further aging in culture. Given this, the Inventors posited that the donor age might be a contributing factor that impacts the cytogenetic instability of fib-iPSCs. However, with respect to the frequency of cytogenetic abnormalities in fib-iPSCs, the Inventors did not observe a significant trend or correlation with the age of the donor (Fig. 9A). Indeed, based on G band karyotype analysis, the highest percent of karyotype abnormalities (31.9%) was observed in the 21-40 age group fibroblast- iPSCs.

Pluripotent stem cells have a highly proliferative nature. As such, the Inventors next attempted to analyze how the fib- and BC-iPSCs fared over time in cell culture and whether increasing passage number led to a greater propensity for accumulating cytogenetic aberrations. About 21.6% of fibroblast-derived iPSCs were observed to have abnormal karyotypes (> 1/20 cells with clonal aberrations) in their first G-band karyotype evaluation post-iPSC generation, which was typically between passages 4-23. In stark contrast, only 2.8%) of BC-iPSCs had abnormal karyotypes in their first assessment. Any iPSC line that was determined to have an abnormal karyotype was no longer cultured or evaluated. Therefore, between 2 nd and 4 th repeat karyotypes were only evaluated on iPSC lines that were initially identified as cytogenetically normal in their first karyotype. This analysis allowed us to determine whether fib-iPSCs have an inherent level of cytogenetic instability over BC-iPSCs upon continued time in culture and passaging even after being established as normal post- iPSC generation (at their first G-band karyotype evaluation). G-band karyotyping showed that a greater proportion of fib-iPSCs (27.8%>) acquired abnormal karyotypes in culture upon repeat karyotyping when compared to the proportion at first karyotypes (P < 0.001, two way ANOVA with Bonferroni posttest), while the percent abnormal karyotypes in BC-iPSCs did not show any significant difference between first and repeat karyotypes (Fig. 9B). Upon analyzing the origin of the cytogenetic instability in fib-iPSCs, it appeared that the highest rate of abnormal karyotypes (first or repeat) occur during passages 10 - 23 in the life of the iPSC line (Fig. 9C-D), which is typically when the largest amount of iPSC expansion occurs for any given cell line. The iPSC expansion increases at passages 10 - 23 largely due to characterization and cell banking coinciding around these passage numbers. Nevertheless, this is a similar process for both fib- or BC-iPSCs, and yet the PBMC-derived cells do not display an increased disposition towards abnormal karyotypes upon extended culture or expansion. Example 20

BC-iPSCs acquire less submicroscopic amplifications and deletions G-band karyotype has the advantage that it can detect balanced translocations, inversions, <20% culture mosaicism, chromosomal position of genomic gains or losses as commonly observed in the fib-iPSC cultures (Fig. 6 and Table 5). However, since G-band karyotype is unable to detect submicroscopic genomic abnormalities (<5Mb), the Inventors decided to conduct comparative genomic hybridization (aCGH) microarray on a subset of fib- iPSC and PBMC-iPSC lines. While aCGH is unable to detect balanced translocations, inversions and < 20% culture mosaicism, it is a good supportive method to detect smaller size genomic gains and losses, copy number variants (CNVs), duplications/deletions, and unbalanced translocations in the iPSC lines. The Inventors analyzed and recorded any de novo amplifications or deletions acquired in the iPSCs upon comparison with the parental fibroblast or PBMC source bio-specimen. In this analysis only those amplifications and deletions acquired de novo in iPSCs were considered, when they were not normal CNVs upon cross referencing against the Database of Genomic Variants (DGV) that contains genomic variations observed in healthy individuals.

Including iPSC lines with abnormal and normal G-band karyotypes, the average size of the amplification and deletions detected by aCGH were significantly greater in fib-iPSC lines at 44 Mb compared to 2.1 Mb in PBMC-iPSC lines (Fig. 10A), the preponderance of which was due to fib-iPSC lines with an abnormal karyotype (Fig. 10B). Upon segregating the size analysis comparison between iPSC lines with normal karyotypes, the de novo amps/dels were on average 2.31 Mb in fib-iPSCs and 2 Mb in PBMC-iPSC lines. Supporting this data, the average number of acquired de novo total amps/dels in fib-iPSCs (3.7) were at least two-fold greater than in BC-iPSCs (1.8). Even in iPSC lines that were determined to have normal G- band karyotypes, the number of new amps/dels were greater in fib-iPSCs at 3.3 vs. BC-iPSCs at 1.8. The most commonly acquired submicroscopic (0.8-1.5 Mb) de novo amplifications or deletions in fib- or PBMC-iPSC lines detected by aCGH was amplification of chromosome 7q31.32 or deletion of chromosomes 10ql5.2-q25.1, 16pl 1.2, 21pl 1.2-pl 1.1 (Fig. 10E).

Here the Inventors report a new and more reliable method for reprogramming of the non-T cell component of blood using episomal plasmids expressing pluripotency factors. Using the described reprogramming protocol, one is able to consistently reprogram non-T cells with close to 100% success from non-T cell or non-B cell sources. Further advantages include use of a defined reprogramming media E7 and using defined clinically compatible substrate recombinant human L-521. Blood cell-derived iPSCs ("BC-iPSCs") exhibited identical characteristics to fibroblastderived iPSCs ("fib-iPSCs"), retain genotype, exhibit a normal pluripotency profile, and readily differentiate into all three germ-layer cell types. This method for reliably deriving iPSCs from patient blood samples paves the way for rapidly generate new human iPSC lines, including patient-specific lines, thus providing an enormous bioresource for disease modeling, drμg discovery, and regenerative medicine applications.

Example 21

Additional Characterization Studies

G-Band Karyotype. Human iPSCs were incubated in Colcemid (100 ng/mL; Life

Technologies) for 30 minutes at 37°C and then dissociated using TrypLE for 10 minutes. They were then washed in phosphate buffered saline (PBS) and incubated at 37°C in 5mL hypotonic solution (lg KC1, lg Na Citrate in 400mL water) for 30 minutes. The cells were centrifμged for 2.5 minutes at 1500 RPM and resuspended in fixative (methanol: acetic acid, 3 : 1) at room temperature for 5 minutes. This was repeated twice, and finally cells were resuspended in 500 μΐ of fixative solution and submitted to the Cedars-Sinai Clinical Cytogenetics Core for G-Band karyotyping. Analysis includes examination of chromosomes in a minimum of 20 cells per culture. Only abnormalities that met the ISCN 2013 definition of clonality were included in this manuscript dataset4. All normal and abnormal karyotypes were transcribed into a table format. The chromosomal ideograms were then generated by importing the table into R Bioconductor (programming language) using the IdeoViz package that plots data associated with arbitrary genomic intervals along chromosomal ideogram. This Package is available on the open source Bioconductor website. To accurately reflect the frequency of chromosomal rearrangement, the bar widths for each ideogram were scaled in proportion to the number of samples examined. As such, the bars displaying abnormalities found in PBMC-iPSC lines are 2.16 times the width of bars depicting abnormalities found in fib-iPSC lines to account for the fibroblast ideogram portraying the number of abnormalities found in a sample set that was 2.16 times larger than the PBMC set. The letters above the chromosomal translocation (navy blue bars) category are matched such that they correspond between the chromosomal locations to where the translocation abnormality occurred.

Copy Number Variations with Array Comparative Genomic Hybridization (aCGH). Array CGH is a high-resolution karyotype analysis solution for the detection of unbalanced structural and numerical chromosomal alterations with high-throμghput capabilities. High quality genomic DNA was isolated from primary dermal fibroblasts or isolated PBMCs and their reprogrammed iPSCs. Quality of genomic DNA as determined by UV spec. (NanoVue), fluorometer (Qubit) and Agarose Gel analysis. The samples were analyzed for CNVs using the Agilent 60K Standard aCGH platform suitable for human stem cell and cancer cell lines by Cell Line Genetics (Madison, WI). Comparison of iPSC lines to their parental donor cell of origin yielded de novo copy-number variation (gains/amplifications or losses/deletions) for the indicated loci. TCRG chain rearrangement assay. Genomic DNA (350ng) was harvested from all cell lines using the MasterPure DNA Purification Kit (Epicenter Biotechnologies). An embryonic stem cell line (H9) was used as a negative control. Primer sets that recognize the three framework regions in the heavy chain locus of the IgH gene were obtained from InVivoScribe Technologies (Cat no.11010010, San Diego, CA) and the PCR was carried out as per the manufacturer's protocol. Episomal plasmid related gene analysis Genomic DNA (400ng) was harvested from all cell lines and an embryonic stem cell line (H9) was used a negative control. Primers that recognize EBNA-1, along with GAPDH, which was used as a housekeeping gene, were included in this study. PCR was run for 35 cycles at 95°C for 30 sec, 60°C for 30 s, and 72°C for 30 s.

Neuronal differentiation from iPSCs. The human fibroblast- and PBMC-iPSCs were seeded in 6-well Matrigel-coated plates. The iPSCs were grown to near confluence under normal maintenance conditions before the start of the differentiation. The next day neuronal differentiation was initiated by neuroectoderm differentiation by dual SMAD and GSK-3beta inhibition using LDN193189 (0.2 μΜ, Cayman), SB431542 (10 μΜ, Cayman) and CHIR99021 (3 μΜ, Cayman) this treatment is carried on for 6 days in Iscove's modified Dulbecco's medium (EVIDM) / F12 (1 : 1) media containing nonessential amino acids (NEAA; 1%), B27 (2%), and N2 (1%). After 6 days differentiating cells were then gently lifted by accutase treatment for 5 min at 37°C. Cells at a density of 7.5 X 10 5 were subsequently placed in a 6-well plate or 1 X 104 cells were seeded in a 96-well plate in above neural differentiation medium with the addition of 0.1 μΜ all-trans retinoic acid and 1 μΜ sonic hedgehog agonist (SAG). Fresh media was changed every other day. At day 12 of differentiation media was changed to terminal differentiation (IMDM) / F12 (1 : 1) media supplemented with NEAA (1%), B27 (2%), N2 (1%), compound E (0.1 μΜ; Calbiochem), DAPT (2.5 μΜ; Cayman), retinoic acid (0.5 μΜ) all-trans, SAG (0.1 μΜ), ascorbic acid (200 ng/ml), dibutyryl cyclic adenosine monophosphate (ΙμΜ), brain-derived neurotrophic factor (10 ng/ml), and glial cell line-derived neurotrophic factor (10 ng/ml). Differentiating neurons were fixed at day 18 of differentiation and analyzed.

Example 22

Cryopreserved PBMCs Isolated from Blood are Reliably Reprogrammed to iPSCs Human iPSCs can be generated from freshly isolated non-expanded PBMCs with episomal plasmids. However, there are no descriptions for reprogramming cryopreserved PBMCs to iPSCs. When the Inventors reprogrammed cryopreserved PBMCs to iPSCs from multiple individuals with episomal plasmids expressing POU5F1, SOX2, KLF4, LIN28, L- MYC, and TP53 shRNA (termed 4p method), the Inventors were either unsuccessful or observed significant variability in isolating identifiable iPSC clones even after 35-40 days. Other attempts to generate iPSCs from whole blood have shown that in order to generate sufficient cell numbers for reprogramming, isolated CD34+ progenitor cells need to be enriched and expanded in culture with complex and expensive protocols. This highlighted the need to simplify this process and avoid expansion of somatic cells prior to reprogramming. The Inventors instead isolated and cryopreserved non-expanded PBMCs for reprogramming to iPSCs because this somatic cell source provides the fastest and most cost-effective procedure from multiple subject cohorts. Using the 4p method, the T reprogramming protocol was successful with only 14.3% of PBMC samples, while the non-T protocol on cryopreserved PBMCs did not result in any clonal iPSC lines.

To increase the efficiency of PBMC reprogramming the Inventors utilized, (a) an additional episomal plasmid containing SV40 large T antigen (SV40LT) in specific stoichiometry to minimize PBMC cell death, termed 5p method, and (b) a defined E7 reprogramming media to promote high surface attachment of the nucleofected PBMC. This novel protocol resulted in successful and efficient generation of multiple adherent PBMC- iPSC clones that could be mechanically isolated and scaled up for expansion within 25-35 days post-nucleofection. Importantly, the success rate and the efficiencies in reprogramming multiple subject cryopreserved PBMCs from unaffected controls or diseased patients was significantly greater at 85% for T cell and 92% for non-T cell types using the 5p method, in contrast to between 0 and 15% for the 4p method. Reprogramming PBMCs on mouse embryonic fibroblast feeders (MEFs) in a commercial media (primate ESC media; PESC) resulted close to 89-95% success for both T and non-T cell types with good reprogramming efficiencies. The Inventors next tested whether this 5p reprogramming protocol was also amenable with xenobiotic-free components, as this may be required for clinically compatible iPSC-based cell therapies. Use of recombinant human laminin 521 substrate and chemically defined reprogramming 1 media (E7)21 also resulted in successful T- and non-T cell reprogramming, at 80% and 64%, respectively. Collectively, these results show that the 5p method, when compared to 4p method, significantly enhanced reprogramming success and efficiency of PBMCs (both T and non-T cells) and provides a valuable protocol for reliable reprogramming of cryopreserved PBMCs. Each PBMC-iPSC line exhibited typical PSC characteristics, including tightly packed colonies, high cell nuclear-cytoplasmic ratio, robust alkaline phosphatase activity, and expression of multiple pluripotency antigens, which are all clear in representative reprogrammed iPSC colonies from PBMCs of two healthy volunteers (non-T cell source 03iCTR10 NTnl and T-cell source 80iCTR-Tnl). The PBMC-iPSCs passed pluripotency quality control metrics determined by the PluriTest assay, demonstrating that the PBMC-iPSC transcription profile was analogous to well established hESCs and fib- iPSCs, but not differentiated fibroblasts and neural progenitor cells. PBMC-iPSCs maintained a normal G-band karyotype and were confirmed to be clonal derivatives of either T cells or non-T cells from the PBMCs using the T cell receptor (TCR)-β and -γ, gene rearrangement/clonality assays. The tri-lineage potential of PBMC-iPSCs was demonstrated by spontaneous embryoid body formation and by measuring germ-layer specific gene expression profile by the TaqMan Scorecard assay. The genomic transgene-free status of PBMC iPSCs was confirmed by, (1) demonstration that the EBNA plasmid-related latency element was eventually eliminated from the established PBMC-iPSCs and not detected in the genomic DNA; and (2) the expression of endogenous pluripotency genes and absence of any exogenous reprogramming transgenes using RT-qPCR. Further, the SV40LT factor that is a component of the 5p reprogramming cocktail was undetectable at the genomic DNA level and not expressed by the reprogrammed PBMC-iPSC lines.

Example 23

PBMC-iPSCs have Equivalent Neuronal Differentiation as Fib-iPSCs

While many early reports sμggested that newly reprogrammed iPSCs retain some epigenetic memory of their parental donor cell type, recent reports sμggest that hiPSCs, regardless of the source tissue, ultimately lose most of their gene expression and epigenetic profiles related to the original cell source following expansion. However, it still remains unclear whether blood derived iPSCs can differentiate as efficiently as fibroblast-derived iPSCs into various cell types, possibly due to a stronger retention of epigenetic memory in blood-sourced iPSCs compared to other donor cells. The Inventors addressed this by directing blood or fibroblast-derived iPSCs, both of mesodermal origin, towards a different germ layer, i.e. neuroectoderm. Specifically, the Inventors performed neural ectoderm differentiation from large numbers of iPSC lines derived from fibroblast (n = 26) and PBMC lines (n = 20), of both healthy subjects as well as diseased patients. Immunocytochemistry for neural ectoderm markers of TUBB3 (β3-tubulin) and NKX6.1 and subsequent quantification demonstrated that neuronal differentiation occurred at a similar efficiency between fib-iPSC and PBMC-iPSCs, further demonstrating the utility of PBMC iPSCs. PBMC-iPSCs Reprogrammed with 5p Maintain a Significantly More Stable Karyotype Recurrent chromosomal abnormalities have been described previously for hESC lines. Some reports have also chronicled common chromosomal aberrations for hiPSC lines, however, these did not methodically account for variability in source tissues, reprogramming methods and cell culture methods. To the Inventors' knowledge, there have been no systematic studies describing cytogenetic analysis comparing frequency between iPSCs derived from fibroblast and blood. The Inventors performed in-depth cytogenetic analysis on iPSC lines derived from 171 unique donors of 16 fibroblasts (77) or PBMCs (94). With multiple clonal lines per donor, this included karyotypes from 348 independent clonal fibroblast (205) or PBMC (143) iPSC lines. All the fibroblast, lymphoblastoid cell lines, epithelial and blood-derived iPSCs assessed were reprogrammed using similar non-integrating episomal reprogramming methods. The major difference between fibroblast and PBMC-reprogramming reported here was the number of reprogramming factor plasmids used; 3p31 fibroblasts and 4p or 5p for PBMCs. After successful reprograming, all the iPSC lines analyzed and reported here were maintained using feeder-free Matrigel®/mTeSRl™ cell culture methods and passaged using mechanical StemPro® EZPassage™ tool. To establish the genetic stability of these lines, the Inventors first performed G-band karyotype analysis on 552 human iPSC cultures derived from source fibroblasts (260), PBMCs (206), epithelial cells (26) and lymphoblastoid cell lines (60) that were collected from multiple laboratories or public repositories. The Inventors' data reveal remarkably lower incidence of chromosomal aberrations in PBMC iPSCs. Abnormal karyotypes with clonal aberrations were observed in 61 of 260 (23.5%) cultures from clonally independent human fib-iPSC lines, derived from 77 unique donors. Typically, the fibroblasts had been expanded for 9-10 passages prior to reprogramming. In stark contrast, only 4 in 206 (1.9%) cultures of clonal human PBMC-iPSC lines derived from 94 unique donors displayed any karyotype abnormalities. The 4 abnormalities observed were also low-frequency. It is evident from the chromosomal ideogram schematics that recurrent aberrations are represented in a significantly greater degree 1 in fib-iPSCs compared to the PBMC-iPSCs. Indeed, this remarkable cytogenetic stability is unique to PBMCs reprogrammed with the new 5p method, as iPSCs reprogrammed from other cell origins, including lymphoblastoid cell lines (25%; 15 abnormal from 60) and primary epithelial cells (27%; 5 abnormal from 26) have similar high rates of karyotypes aberrations as those seen in fibroblasts. The most frequent karyotype abnormalities in fib-iPSCs were observed in chromosomes 13 (14.9%), 1 (11.7%), 12 (9.6%), and X (8.5%). To a lesser extent karyotype changes were also observed in chromosomes 21, 11, and 6. Interestingly, karyotype aberrations were never detected on chromosomes 2, 5 and 7 in fib-iPSCs. Of the aneuploidies, chromosomal gains (trisomy or duplications) were most commonly observed in 12 (20.0%)), 1 (15.0%)), and 13 (12.5%), and chromosomal losses were repeatedly observed in chromosomes X (38.5%) and 21 (23.1%). Structural rearrangements, including translocations, inversions and derivative chromosomes, were recurrent in chromosomes 13 (22.0%), 1 (12.2%) and 14 (9.8%) in fib-iPSCs. In stark contrast, only two PBMC-iPSC lines from 178 iPSC cultures analyzed showed chromosome gains in 14 (trisomy) or 18 (duplication). The other two PBMC-iPSC lines exhibited uncommon abnormalities: a translocation 46,XY,t(7; 14)(q34;ql 1.2) and a mosaic deletion 46,X,del(X)(q22q26) in 19% of cells.

Example 24

Enhanced Karyotype Stability of PBMC-iPSCs is Independent of Donor Age and Passage

Number

The skin, unlike the blood, is routinely exposed to external environmental elements like potential sun damage and the fibroblasts derived from the biopsies require a certain level of expansion prior to reprogramming, which is akin to further aging in culture. Given this, the Inventors posited that the donor age might be a contributing factor that impacts the cytogenetic instability of fib-iPSCs. G27 band karyotype analysis was used to assess this possibility. There was no clear trend and correlation between the increasing age of the donor and the frequency of cytogenetic abnormalities in fib-iPSCs. However, results confirmed that PBMC-iPSCs had much greater karyotype stability over fib-iPSCs in every donor age group. Apart from donor group between ages 0-20, PBMC-iPSCs were significantly more stable than fib-iPSCs for all donor groups between ages 21-90. To establish how proliferation of the iPSCs in culture may affect genetic stability, 1 the Inventors next analyzed fib- and PBMC-iPSCs over time. About 22.4% of fibroblast-derived iPSCs were observed to have abnormal karyotypes (> 1/20 cells with clonal aberrations) in their first G-band karyotype evaluation post-iPSC generation, which was typically between passages 1-23. In stark contrast, only 1.4% of PBMC-derived iPSCs had abnormal karyotypes in their first assessment (p < 0.0001). Any iPSC line that was determined to have an abnormal karyotype in the first karyotype was no longer cultured or evaluated. As such, between 2nd and 4th repeat karyotypes of an iPSC line were only evaluated on lines that were initially identified as cytogenetically normal in their first karyotype. There was a trend towards greater proportion of fib-iPSCs (27.3%)) acquiring abnormal karyotypes in culture upon repeat karyotyping when compared to the proportion at first karyotypes, and significantly greater than PBMC- iPSCs (p < 0.001). Strikingly, the percent abnormal karyotypes in PBMC-iPSCs did not show any significant differences between first and repeat karyotypes. Upon analyzing the origin of the cytogenetic instability in fib-iPSCs, it appeared that the highest rate of abnormal karyotypes (first or repeat) occurred during passages 1-23 in the life of the iPSC line. This passage range is typically when the greatest amount of iPSC expansion occurs for any given cell line largely due to characterization and cell banking coinciding around these passage numbers. Nevertheless, this is a similar process for both fib- or PBMC-iPSCs, and yet, critically, the PBMC-derived cells reprogrammed by the Inventors' new method do not display an increased disposition towards abnormal karyotypes upon extended culture or expansion. PBMC-iPSCs Acquire Less Submicroscopic Amplifications and Deletions Since G-band karyotype is unable to detect submicroscopic genomic abnormalities (<5Mb), next array comparative genomic hybridization (aCGH) microarrays were next conducted on a subset of fib-iPSC and PBMC-iPSC lines. While aCGH is unable to detect balanced translocations, inversions and < 20% culture mosaicism, it is a good supportive method to detect smaller size genomic gains and losses, copy number variants (CNVs), duplications/deletions, and unbalanced translocations in the iPSC linesl 1,30,32,33. The Inventors analyzed and recorded any de novo amplifications or deletions acquired in the iPSCs upon comparison with the parental fibroblast or PBMC source bio-specimen. In this analysis, only those amplifications and deletions acquired de novo in iPSCs were considered, when they were not normal CNVs upon cross referencing against the Database of Genomic Variants that contains genomic variations observed in healthy individuals.

Including iPSC lines with abnormal and normal G-band karyotypes, 1 the average size of the amplification and deletions (amps/dels) detected by aCGH were significantly greater in fib-iPSC lines at 44 Mb compared to 2.1 Mb in PBMC-iPSC lines, the preponderance of which was due to fib-iPSC lines with an abnormal karyotype. Upon segregating the size analysis comparison between iPSC lines with normal karyotypes, the de novo amps/dels were on average 2.31 Mb in fib-iPSCs and 2 Mb in PBMC-iPSC lines. Supporting this data, the average number of acquired de novo total amps/dels in fib-iPSCs (3.7) were significantly greater (two-fold) than in PBMC-iPSCs (1.8). Even in iPSC lines that were determined to have normal G-band karyotypes, the number of new amps/dels was greater in fib-iPSCs at 3.3 vs. PBMC-iPSCs at 1.8. The most commonly acquired submicroscopic (0.8-1.5 Mb) de novo amplifications or deletions detected by aCGH were amplification of chromosome 7q31.32 or deletion of chromosomes 10ql5.2-q25.1, 16pl 1.2, and 21pl 1.2-pl 1.1. Collectively, this data demonstrates that this new method to reprogram PBMCs results in more stable human iPSCs with regards to acquisition of chromosomal aberrations, compared to any other published method.

Example 25

Additional Characterization of 5p method

Table 9 - Greater n for PBMCs reprogrammed to iPSCs

The Inventors performed further analysis to confirm whether p53 (TP53) in PBMC-iPSC lines reprogrammed using the Inventors' method. These lines were reprogrammed on L521 substrate in defined E7 media. The Inventors have performed extensive analyses of multiple iPSC lines. Upon whole genome sequencing (subject of another manuscript in preparation), the Inventors did not identify any variants (S Ps or In-dels) in the iPSC lines in the exon (coding region) of the TP 53 gene at the genome level.

Whole genome sequencing was performed using 750ng of genomic DNA with the

Illumina TruSeq PCR free library preparation kit according to manufacturer's instructions (Illumina, San Diego, CA). Library quality was confirmed with the BioAnalyzer (Agilent Technologies, Santa Clara, CA) and libraries were sequenced to at least 40x read depth on an Illumina Hi-SeqlOX at Fulgent Therapeutics, Temple City, CA. An average of >460Million reads per sample were generated. Fastq files were aligned to the reference genome in build hg38 using the BWA-mem algorithm in the Burroμghs-Wheeler aligner, with more than 400Million reads successfully aligned for each sample providing an average of 43x read depth across the genome.

Using this pipeline, the Inventors did not identify any variants (SNPs or In-dels) that arise in only the iPSC line or their clones in the coding region (exons) of TP53. Of the multiple iPSC lines analyzed and their clonal derivatives, a single intronic de novo SNP was detected in two of the three iPSC clones derived from only one donor iPSC line; rsl2940247 (hg38 chrl7:7,664,385 G/A), which is in Intron 1 of a single (and rare) isoform of TP53. This common SNP is found in at least 36% of all populations assayed, with a minor allele (G) frequency of 0.403 in a reference population of Caucasian samples (the donor line iPSC line was from a Caucasian donor). This common variant is not implicated with any function, or annotated in typical clinical annotation databases used to assign potential impact of variants in cancer (CiVIC, ClinVar, COSMIC Cancer Consensus). There is also no evidence for this variant to act as a regulator of TP53 expression level in any healthy tissue in the Gene Tissue Expression (GTEx) dataset. The variant rs 12940247 does not intersect with any regulatory features in the Tier 1 cell lines (including a lymphoblastoid cell line GM 12878, which has been derived from PBMC). The Inventors have also used the Illumina Omni2.5M genotyping array to characterize these samples. In keeping with the results from whole genome sequencing there were no variants detected in the iPSC clones that were discordant from the genotype of the germline (blood) sample, and there were no copy number variants identified spanning this locus. Table 10 - Non-coding common variant rsl2940247 is observed as a de novo event in two clones from donor CS-002.

The location of the S P in intron 6 of one isoform of TP53 is marked with a grey line. Isoforms of TP53 are shown from GENCODE v24 Comprehensive in black (features with corresponding entry in the Protein Data bank) or dark blue (reviewed or validated transcript from either RefSeq or SwissProt). Below gene annotation are four tracks showing transcription, active promoters (none of which are observed in the genomic interval plotted) and active enhancers (tracks 3 and 4, again, none of which are observed in the genomic interval plotted). Some poised regulatory elements are observed (represented as blue peaks showing enrichment of signal from ChlP-Seq of H3K4mel) in the gene body of TP53 in HUVEC cells. There is no evidence for the observed non-coding variant occurring de novo in two clones from donor iPSC has any regulatory function of TP53.

Brief methodology: Germline variant calling was performed with the Genome Analysis Toolkit (GATK) using Best Practices for germline variant calling. Briefly, aligned sequence is pre-processing with Picard to identify duplicate reads (those originating from a single fragment of DNA, which can be generated during sample preparation or using cluster amplification during the sequencing process) by the sequence at the 5' end of the read. Base quality scores are recalibrated after local genome realignment around In-dels to improve genotype calling accuracy (a process which relies heavily on base quality scores) before variants are identified. Called single nucleotide polymorphisms (SNPs) and In-dels are then used to recalibrate variant calls in a joint genotyping module to generate variant calls with high confidence. In conclusion, based on the highly detailed and extensive analysis of multiple iPSC lines and clones at the SNP and WGS level, the Inventors do not detect any coding mutations in the TP53 gene that would disable its normal regulation.

Example 26

Discussion

In this study, the Inventors show that human iPSCs derived by a new method from non-expanded and cryopreserved PBMCs (PBMC-iPSCs) are remarkably cytogenetically stable compared to iPSCs generated from other somatic cells that require cell expansion prior to reprogramming, including skin fibroblast cultures, LCLs, epithelial cells, and adipose cells. These PBMC-iPSCs reprogrammed with the 5p method also have enhanced stability rates compared to those historically reported for other pluripotent stem cell lines such as hESCs and iPSCs derived in other laboratories. This entire study entailing episomal plasmid reprogramming of various cell types from over 200 unique donors, identical stem cell maintenance procedures, pluripotency characterization, and karyotype analysis were performed in the Inventors' laboratories and the cytogenetics core. The Inventors systematically analyzed over 200 individual clonal PBMC-iPSC lines generated in the Inventors' laboratory, with both lymphoid T cell and myeloid non-T cell populations being reliably reprogrammed, irrespective of substrate and media. Further, the Inventors demonstrated creation of stable PBMC-iPSCs on a xeno-free recombinant human L521 substrate and defined reprogramming media. Most importantly, the PBMC-iPSCs reprogrammed with the 5p method maintain karyotype stability upon long-term culture. Collectively, the combination of this improved technology and a non-expanded cell source such as PBMCs offer 1 both clinically compatible reprogramming methods and facile access to large numbers of patient samples.

Aneuploidy and rearrangements in chromosomes 1, 12, 13 and X were the most frequent abnormalities observed in fib-iPSCs generated by episomal reprogramming in the Inventors' laboratory. In contrast these aberrations were never detected in any of the PBMC- iPSCs the Inventors generated and analyzed. Some of these changes observed in fib-iPSCs are similar to recurrent abnormalities found in human ESCs where there are short arm of chromosome 12 and gains or losses of chromosomes 1, 17 and X, similar to their malignant human embryonal carcinoma (EC) stem cell counterparts. Using aCGH microarray, the PBMC-iPSCs showed half the number of de novo CNVs compared to the fib-iPSCs. These de novo CNVs were not shared between hiPSCs and their respective parental fibroblasts or PBMCs and were likely acquired during reprogramming or iPSC expansion. The average base pair size of chromosomal change in the CNVs in fib-iPSCs was also far greater when compared to PBMC-iPSC lines, further supporting the stability of this new reprogramming method.

Increased karyotype abnormalities, coding mutations, and small genomic alterations have previously been reported in iPSCs, likely reflecting the mutagenic consequences due to the reprogramming procedure itself. However, the bulk of these reported genomic abnormalities have been examined in iPSC lines generated from skin fibroblasts using integrating retroviral and lentiviral reprogramming methods. Aneuploidy rates and CNVs comparing different nonintegrating and lentiviral reprogramming methods for low-passage iPSCs have been described. They reported an aneuploidy rate around 12-13% in low-passage iPSCs using episomal and integrating reprogramming methods, althoμgh this data predominantly includes aberration rates for iPSCs derived from expanded fibroblast cultures that were not segregated by the origin of donor cells or obtained from non-expanded cells, like PBMCs. Various factors can affect genetic stability during long-term culture and expansion of PSCs including, oxygen tension, growth supplements, growth factors, passage technique, cryopreservation and the extracellular substrate on which cells are grown36. There is often substantial laboratory variation in PSC culture techniques given these aforementioned factors; however, these factors were all tightly controlled for in the Inventors' study. The Progenitor Cell Biology Consortium (www.synapse.org) identified correlations in gene expression and CNVs among key developmental and oncogenic regulators as a result of donor line stability, reprogramming technology, and cell of origin. However, karyotype abnormalities and line stability were not distinguished by cell of origin and their previous cell expansion history in this study, and only small numbers of abnormal 1 fib-iPSCs were analyzed.

Althoμgh methylation and transcription profiles of iPSCs segregated somewhat based on blood and fibroblast cell of origin, they could not be directly attributed to somatic cell epigenetic memory profiles. This group also observed a trend towards higher level of CNVs in lines generated using integrating reprogramming methods. Recently, another group revealed that blood cells and derived iPSCs were much closer to bona fide hiPSCs and ESCs in their methylation profiles (more hyper-methylated state) compared to any other parental tissue or their iPSCs that had more aberrant DNA methylation, supporting the Inventors' argument that a cell population that has not been previously expanded in tissue-culture, such as adult PBMCs or cord blood, could be a preferred choice for reprogramming and maintenance of cytogenetic stability at the iPSC stage. The mechanisms underlying the genetic stability of iPSCs derived from non-expanded PBMCs compared to iPSCs generated from expanded somatic cell types such as skin fibroblasts, LCLs, epithelial and adipose stem cells are likely to be multi-factorial. First, the bone marrow niche allows for turnover of blood more frequently (1-5 days for most cells of the PBMC lineage) than almost any other cell type of the body (BNID 101940). Therefore, a host blood may be considered far less "aged" than a resident dermal fibroblast, epithelial or adipose cell, which only renews over a period of months to years. Notably, the reprogramming of donor-derived skin fibroblasts, LCLs, epithelial and adipose cells, requires a significant period of ex vivo somatic cell expansion in tissue-culture. These factors may lead to a starting pool of cells with increased tissue culture-associated cell stress and senescence, thus making susceptible to decreased DNA repair capacities. It is also likely, therefore, that a culture of previously expanded somatic cells may start with some overall heterogeneity and inherent genotype instability, and low-level mosaicism could remain undetected by traditional G-band karyotype in a bulk heterogeneous cell culture prior to reprogramming. Subsequently, fib-iPSC, LCL-iPSC or epithelial-iPSC line may be isolated in which low-level karyotype abnormalities become magnified upon significant clonal proliferation and expansion. This risk is avoided in the Inventors' method, as isolated PBMCs were initiated for reprogramming immediately post-isolation without any period of prior cell expansion. Lastly, the PBMC 5p reprogramming protocol contains an additional episomal plasmid where one of the reprogramming factors SV40LT minimizes PBMC cell death and promote surface attachment, which may also enhance the stability of the PBMCs during the stressful reprogramming phase for a cell. It has also been previously shown that the use SV40LT as a component of the reprogramming factor cocktail is non-transforming since the transgene does not integrate into the genome of the iPSC lines derived by episomal plasmid reprogramming, which is also supported by the Inventors' data. While it is possible that pre- expanded somatic cell types like fibroblasts may obtain greater genetic stability if reprogrammed with the 5p protocol, the Inventors feel that this single difference in reprogramming between the pre-expanded cells and the nonexpanded PMBCs does not completely account for the vast differences in karyotype stability. Importantly, apart from the additional factor, all the pre-expanded cell types and non-expanded blood-derived iPSCs were reprogrammed using similar non-integrating episomal reprogramming methods and maintained with identical feeder-free Matrigel®/mTeSRl™ cell culture and the manual StemPro® EZPassage™ tool. Nonetheless, it would be a great advancement to the field if the Inventors' novel 5p protocol facilitated reprogramming of additional cell types beyond non- expanded PBMCs and ongoing studies are assessing fibroblast reprogramming using the optimized protocol. The first autologous human iPSC clinical trial for treating age-related macular degeneration patients sponsored by RIKEN institute was suspended because single nucleotide variations (SNVs) and CNVs were detected in a patient iPSC line, which were not detectable in the patient's original fibroblasts. While the CNVs were all single-gene deletions, one of the SNVs was previously identified in a cancer-related somatic mutations database6,16. This emphasizes that a stable karyotype is paramount for the success of clinical applications of human iPSCs. In addition, scale-up of iPSCs and derivatives for predictive toxicology and high-throμghput drμg screening for therapeutic discoveries requires a source of iPSCs with substantial cell stability. This optimum reprogramming method in non- expanded PBMCs will minimize effects of acquired genomic aberrations. Together, these results highlight that optimizing techniques of reprogramming as well as the quality and the period of pre-culture prior to reprogramming are crucial for long-term iPSC line stability. This data will assist in refining the safety of iPSC-derived cell products for clinical and research discovery applications, as well as aid in greater reproducibility in iPSC disease modeling studies.

The various methods and techniques described above provide a number of ways to carry out the invention. Of course, it is to be understood that not necessarily all objectives or advantages described may be achieved in accordance with any particular embodiment described herein. Thus, for example, those skilled in the art will recognize that the methods can be performed in a manner that achieves or optimizes one advantage or group of advantages as taμght herein without necessarily achieving other objectives or advantages as may be taμght or sμggested herein. A variety of advantageous and disadvantageous alternatives are mentioned herein. It is to be understood that some preferred embodiments specifically include one, another, or several advantageous features, while others specifically exclude one, another, or several disadvantageous features, while still others specifically mitigate a present disadvantageous feature by inclusion of one, another, or several advantageous features.

Furthermore, the skilled artisan will recognize the applicability of various features from different embodiments. Similarly, the various elements, features and steps discussed above, as well as other known equivalents for each such element, feature or step, can be mixed and matched by one of ordinary skill in this art to perform methods in accordance with principles described herein. Among the various elements, features, and steps some will be specifically included and others specifically excluded in diverse embodiments.

Althoμgh the invention has been disclosed in the context of certain embodiments and examples, it will be understood by those skilled in the art that the embodiments of the invention extend beyond the specifically disclosed embodiments to other alternative embodiments and/or uses and modifications and equivalents thereof.

Many variations and alternative elements have been disclosed in embodiments of the present invention. Still further variations and alternate elements will be apparent to one of skill in the art. Among these variations, without limitation, are sources of blood cells, cellular components of blood, pluripotent stem cells derived from therein, techniques and composition related to deriving pluripotent stem cells from blood cells cells, differentiating techniques and compositions, biomarkers associated with such cells, and the particular use of the products created throμgh the teachings of the invention. Various embodiments of the invention can specifically include or exclude any of these variations or elements.

In some embodiments, the numbers expressing quantities of ingredients, properties such as concentration, reaction conditions, and so forth, used to describe and claim certain embodiments of the invention are to be understood as being modified in some instances by the term "about." Accordingly, in some embodiments, the numerical parameters set forth in the written description and attached claims are approximations that can vary depending upon the desired properties soμght to be obtained by a particular embodiment. In some embodiments, the numerical parameters should be construed in light of the number of reported significant digits and by applying ordinary rounding techniques. Notwithstanding that the numerical ranges and parameters setting forth the broad scope of some embodiments of the invention are approximations, the numerical values set forth in the specific examples are reported as precisely as practicable. The numerical values presented in some embodiments of the invention may contain certain errors necessarily resulting from the standard deviation found in their respective testing measurements.

In some embodiments, the terms "a" and "an" and "the" and similar references used in the context of describing a particular embodiment of the invention (especially in the context of certain of the following claims) can be construed to cover both the singular and the plural. The recitation of ranges of values herein is merely intended to serve as a shorthand method of referring individually to each separate value falling within the range. Unless otherwise indicated herein, each individual value is incorporated into the specification as if it were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g. "such as") provided with respect to certain embodiments herein is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention otherwise claimed. No language in the specification should be construed as indicating any non-claimed element essential to the practice of the invention.

Groupings of alternative elements or embodiments of the invention disclosed herein are not to be construed as limitations. Each group member can be referred to and claimed individually or in any combination with other members of the group or other elements found herein. One or more members of a group can be included in, or deleted from, a group for reasons of convenience and/or patentability. When any such inclusion or deletion occurs, the specification is herein deemed to contain the group as modified thus fulfilling the written description of all Markush groups used in the appended claims.

Preferred embodiments of this invention are described herein, including the best mode known to the inventors for carrying out the invention. Variations on those preferred embodiments will become apparent to those of ordinary skill in the art upon reading the foregoing description. It is contemplated that skilled artisans can employ such variations as appropriate, and the invention can be practiced otherwise than specifically described herein. Accordingly, many embodiments of this invention include all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context.

Furthermore, numerous references have been made to patents and printed publications throμghout this specification. Each of the above cited references and printed publications are herein individually incorporated by reference in their entirety.

In closing, it is to be understood that the embodiments of the invention disclosed herein are illustrative of the principles of the present invention. Other modifications that can be employed can be within the scope of the invention. Thus, by way of example, but not of limitation, alternative configurations of the present invention can be utilized in accordance with the teachings herein. Accordingly, embodiments of the present invention are not limited to that precisely as shown and described.