Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHODS OF T CELL PRODUCTION
Document Type and Number:
WIPO Patent Application WO/2021/032836
Kind Code:
A1
Abstract:
This invention relates to the production of a population of TCR αβ+ T cells by a method comprising (i) differentiating a population of haematopoietic progenitor cells (HPCs) into progenitor T cells and (ii) maturing the progenitor T cells to produce a population of TCR αβ+ T cells. One or both of steps (i) and (ii) are performed in the presence of Inducible Co-stimulator ligand (ICOS-L). The presence of ICOS-L in steps (i) and/or (ii) may increase expression of αβ T cell receptors and/or increase the proportion of HPCs or progenitor T cells that mature into TCR αβ+ cells. This may be useful for example in the production of T cells for immunotherapy.

Inventors:
GUEGUEN CLAIRE (GB)
Application Number:
PCT/EP2020/073332
Publication Date:
February 25, 2021
Filing Date:
August 20, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ADAPTIMMUNE LTD (GB)
International Classes:
C12N5/0783; A61K35/17; C12N5/0789
Domestic Patent References:
WO2018135646A12018-07-26
WO2011097477A12011-08-11
WO2018176144A12018-10-04
WO2017075389A12017-05-04
Foreign References:
US20040171148A12004-09-02
Other References:
LIANG L ET AL: "The right place at the right time: novel B7 family members regulate effector T cell responses", CURRENT OPINION IN IMMUNOLOGY, ELSEVIER, OXFORD, GB, vol. 14, no. 3, 1 June 2002 (2002-06-01), pages 384 - 390, XP004350491, ISSN: 0952-7915, DOI: 10.1016/S0952-7915(02)00342-4
R. I. NURIEVA ET AL: "B7h is required for T cell activation, differentiation, and effector function", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 100, no. 24, 25 November 2003 (2003-11-25), pages 14163 - 14168, XP055565496, ISSN: 0027-8424, DOI: 10.1073/pnas.2335041100
SIMPSON T R ET AL: "Regulation of CD4 T cell activation and effector function by inducible costimulator (ICOS)", CURRENT OPINION IN IMMUNOLOGY, ELSEVIER, OXFORD, GB, vol. 22, no. 3, 1 June 2010 (2010-06-01), pages 326 - 332, XP027072474, ISSN: 0952-7915, [retrieved on 20100129], DOI: 10.1016/J.COI.2010.01.001
RICHTER G ET AL: "Tumor Necrosis Factor-alpha regulates the expression of inducible costimulator receptor ligand on CD34+ progenitor cells during differentiation into antigen presenting cells", JOURNAL OF BIOLOGICAL CHEMISTRY, AMERICAN SOCIETY FOR BIOCHEMISTRY AND MOLECULAR BIOLOGY, US, vol. 276, no. 49, 1 January 2001 (2001-01-01), pages 45686 - 45693, XP002414708, ISSN: 0021-9258, DOI: 10.1074/JBC.M108509200
MCDERMOTT ET AL., CANCER TREAT REV., vol. 40, no. 9, October 2014 (2014-10-01), pages 1056 - 64
SHARMAALLISON, CELL, vol. 161, no. 2, 9 April 2015 (2015-04-09), pages 205 - 14
ROSENBERG ET AL., NAT MED., vol. 10, no. 9, September 2004 (2004-09-01), pages 909 - 15
GAUDERNACK ET AL., J IMMUNOL METHODS, vol. 90, 1986, pages 179
RHEINHERZ ET AL., PNAS, vol. 76, 1979, pages 4061
LUM ET AL., CELL IMMUNOL, vol. 72, 1982, pages 122
YAMANAKA ET AL., NATURE, vol. 448, 2007, pages 313 - 7
KIM ET AL., NATURE, vol. 454, no. 7204, 31 July 2008 (2008-07-31), pages 646 - 50
TAKAHASHI, CELL, vol. 131, no. 5, 30 November 2007 (2007-11-30), pages 861 - 72
PARK ET AL., NATURE, vol. 451, no. 7175, 10 January 2008 (2008-01-10), pages 141 - 6
KIMET ET AL., CELL STEM CELL, vol. 4, no. 6, 5 June 2009 (2009-06-05), pages 472 - 6
VALLIER, L. ET AL., STEM CELLS, vol. 9999, no. 999A, 2009
BAGHBADERANI ET AL., STEM CELL REV., vol. 12, no. 4, August 2016 (2016-08-01), pages 394 - 420
BAGHBADERANI ET AL., STEM CELL REPORTS, vol. 5, no. 4, 2015, pages 647 - 659
VALLIER, L. ET AL., DEV. BIOL., vol. 275, 2004, pages 403 - 421
COWAN, C.A. ET AL., N. ENGL. J. MED., vol. 350, 2004, pages 1353 - 1356
JOANNIDES, A. ET AL., STEM CELLS, vol. 24, 2006, pages 230 - 235
KLIMANSKAYA, I. ET AL., LANCET, vol. 365, 2005, pages 1636 - 1641
LUDWIG, T.E. ET AL., NAT. BIOTECHNOL., vol. 24, 2006, pages 185 - 187
C. HELGASON: "Basic Cell Culture Protocols", 15 October 2004, HUMANA PRESS INC.
"Human Cell Culture Protocols (Methods in Molecular Medicine S.", 9 December 2004, HUMANA PRESS INC.
R. FRESHNEY: "Culture of Animal Cells: A Manual of Basic Technique", 2 August 2005, JOHN WILEY & SONS INC
HO WY ET AL., J IMMUNOL METHODS, vol. 310, 2006, pages 40 - 52
J. POLLARDJ. M. WALKER, BASIC CELL CULTURE PROTOCOLS, 1997
A. DOYLEJ. B. GRIFFITHS, MAMMALIAN CELL CULTURE: ESSENTIAL TECHNIQUES, 1997
A. CHIUM. RAO, HUMAN EMBRYONIC STEM CELLS, 2003
A. BONGSO, STEM CELLS: FROM BENCH TO BEDSIDE, 2005
PETERSONLORING: "Human Stem Cell Manual: A Laboratory Guide", 2012, ACADEMIC PRESS
K. TURKSEN, HUMAN EMBRYONIC STEM CELL PROTOCOLS, 2006
"NCBI", Database accession no. 2056
VALLIER ET AL., CELL SCIENCE, vol. 118, 2005, pages 4495 - 4509
RING D. B. ET AL., DIABETES, vol. 52, 2003, pages 588 - 595
SATO ET AL., NAT MED., vol. 10, no. 1, January 2004 (2004-01-01), pages 55 - 63
COGHLAN ET AL., CHEM BIOL., vol. 7, no. 10, October 2000 (2000-10-01), pages 793 - 803
PRICE ET AL., FOCUS, vol. 25, 2003, pages 3 - 6
WILLIAMS, G.M. ET AL., EXP. CELL RESEARCH, vol. 89, 1974, pages 139 - 142
MOORE, G.E.WOODS L.K.: "Tissue Culture Association Manual", vol. 3, 1976, pages: 503 - 508
MITOLA ET AL., BLOOD, vol. 116, no. 18, 2010, pages 3677 - 3680
TURUNEN ET AL., CIRC RES., vol. 105, no. 6, 11 September 2009 (2009-09-11), pages 604 - 9
ROBBINS ET AL., J IMMUNOL, vol. 180, no. 9, 2008, pages 6116
SAN MIGUEL ET AL., CANCER CELL, vol. 28, no. 3, 2015, pages 281 - 283
SCHMITT ET AL., BLOOD, vol. 122, 2013, pages 348 - 256
JIANG ET AL., CANCER DISCOVERY, vol. 5, 2015, pages 901
"Protocols in Molecular Biology", 1992, JOHN WILEY & SONS
HIRSCH ET AL., METHODS MOL BIOL, vol. 1114, 2014, pages 291 - 307
"Remington's Pharmaceutical Sciences", 1990, MACK PUBLISHING COMPANY
"Harrison's Principles of Internal Medicine", 2001, MCGRAW-HILL
ROSENBERG ET AL., NEW ENG. J. OF MED., vol. 319, 1988, pages 1676
Attorney, Agent or Firm:
MEWBURN ELLIS LLP (GB)
Download PDF:
Claims:
Claims:

1 . A method of producing a population of TCR ab+ T cells comprising;

(i) differentiating a population of haematopoietic progenitor cells (HPCs) into progenitor T cells and;

(ii) maturing the progenitor T cells to produce a population of TCR ab+ T cells, wherein one or both of (i) and (ii) are performed in the presence of Inducible Co-stimulator ligand (ICOS-L).

2. A method according to claim 1 wherein the presence of ICOS-L increases the proportion of HPCs or progenitor T cells that mature into TCR ab+ cells.

3. A method according to claim 1 or claim 2 wherein the HPCs are differentiated into progenitor T cells in the presence of ICOS-L.

4. A method according to any one of claims 1 to 3 wherein the progenitor T cells are matured in the presence of ICOS-L.

5. A method according to any one of the preceding claims wherein the HPCs and/or progenitor T cells are cultured on a surface coated with ICOS-L.

6. A method according to any one of the preceding claims wherein the HPCs have a CD34+ phenotype.

7. A method according to any one of the preceding claims wherein the population of HPCs is produced in vitro from induced pluripotent stem cells (iPSCs).

8. A method according to claim 7 wherein the method comprises providing a population of iPSCs and differentiating the iPSCs into a population of HPCs.

9. A method according to claim 7 or claim 8 wherein the iPSCs are derived from T cells obtained from a donor individual.

10. A method according to claim 9 wherein the T cells obtained from the donor individual are specific for a target antigen.

11. A method according to claim 10 wherein the target antigen is a tumour antigen.

12. A method according to claim 10 or 11 wherein the T cells obtained from the donor individual are tumour-infiltrating lymphocytes (TILs).

13. A method according to any one of the preceding claims wherein the HPCs are differentiated by a method comprising culturing the population of HPCs in a lymphoid expansion medium to produce the progenitor T cells.

14. A method according to any one of the preceding claims wherein the progenitor T cells have a CD5+, CD7+ phenotype.

15. A method according to any one of the preceding claims wherein the progenitor T cells are differentiated by a method comprising culturing the population of progenitor T cells in a T cell maturation medium to produce the TCR ab+ T cells.

16. A method according to any one of the preceding claims wherein the TCR ab+ T cells have a CD8+ CD4+ phenotype.

17. A method according to any one of the preceding claims comprising activating and expanding the TCR ab+ T cells to produce a population of T cells have a CD8+ single positive phenotype or a CD4+ single positive phenotype.

18. A method according to any one of the preceding claims wherein the TCR ab+ T cells specifically bind to cells expressing a target antigen.

19. A method according to claim 18 wherein the target antigen is a tumour antigen.

20. A method according to claim 19 wherein the TCR ab+ T cells specifically bind to cancer cells expressing the tumour antigen.

21 . A method according to any one of claims 1 to 20 wherein the method further comprises introducing heterologous nucleic acid encoding an ab TCR into the iPSCs, HPCs or progenitor T cells.

22. A method according to claim 21 wherein the heterologous nucleic acid encoding the ab TCR is comprised in an expression vector.

23. A method according to claim 22 wherein the expression vector is a lentiviral vector.

24. A method according to any one of claims 21 to 23 wherein the ab TCR is an affinity enhanced TCR.

25. A method according to one of claims 21 to 24 wherein the ab TCR binds specifically to an MHC displaying a peptide fragment of a target antigen expressed by cells or specifically binds to a target antigen or peptide thereof expressed by cells independently of MHC presentation.

26. A method according to claim 25 wherein the ab TCR binds specifically to an MHC displaying a peptide fragment of a tumour antigen expressed by the cancer cells or binds specifically to a tumour antigen or peptide fragment thereof expressed by cancer cells independently of MHC presentation.

27. A method according to any one of the preceding claims further comprising isolating or purifying the TCR ab+ T cells.

28. A method according to claim 27 wherein TCR ab+ T cells are isolated by magnetic activated cell sorting.

29. A method according to according to any one of the preceding claims comprising concentrating the population of TCR ab+ T cells.

30. A method according to according to any one of the preceding claims comprising storing the population of TCR ab+ T cells.

31 . A method according to any one of the preceding claims comprising formulating the population of TCR ab+ T cells with a pharmaceutically acceptable excipient.

32. A population of TCR ab+ T cells produced by a method according to any one of claims 1 to 31 .

33. A pharmaceutical composition comprising a population of TCR ab+ T cells produced by a method according to any one of claims 1 to 31 and a pharmaceutically acceptable excipient.

34. A population of TCR ab+ T cells produced by a method according to any one of claims 1 to 31 for use in a method of treatment.

35. A population of TCR ab+ T cells produced by a method according to any one of claims 1 to 31 for use in a method of treatment of cancer.

36. A coating composition for treating the surface of a cell culture vessel, the composition comprising ICOS-L.

37. A composition according to claim 36 further comprising a Notch signalling ligand.

38. A culture vessel for T cell culture, said vessel comprising a surface coated with ICOS-L.

39. A culture vessel according to claim 39 wherein the surface is additionally coated with a Notch signalling ligand.

Description:
Methods of T Cell Production

Field

This invention relates to the production of T cells, for example for use in immunotherapy.

Background

Immunotherapeutics are poised to transform the cancer treatment landscape with the promise of long-term survival (McDermott et al., Cancer Treat Rev. 2014 Oct; 40(9): 1056-64). There is a clear unmet medical need for new immunomodulatory drugs to expand patient population and range of tumor types. In addition, new agents are needed to enhance the magnitude and duration of anti-tumor responses. The development of these agents has been possible because of the in-depth understanding of the basic principles controlling T-cell immunity over the last two decades (Sharma and Allison, Cell. 2015 Apr 9; 161 (2): 205-14). This typically requires tumor specific CD4+ and CD8+ T-cells recognising tumor-associated peptide antigens presented by MHC molecules. Different vaccination strategies and adoptive transfer of ex vivo expanded tumor infiltrated lymphocytes have in some cases demonstrated the ability of tumor specific T-cells to treat late stage cancer (Rosenberg et al., Nat Med. 2004 Sep; 10(9): 909-15).

However, current adoptive T cell therapies are limited by a lack of suitable patient and tumor-specific T cells and there is a need for therapeutically sufficient and functional antigen-specific T cells for effective use in immunotherapy.

Summary

The present inventors have recognised that the expression of ab T cell receptors is increased when T cells are generated from antecedent cells in the presence of Inducible Co-stimulator ligand (ICOS-L). This may be useful for example in the production of T cells for immunotherapy.

A first aspect of the invention provides a method of producing a population of TCR ab+ T cells comprising;

(i) differentiating a population of haematopoietic progenitor cells (HPCs) into progenitor T cells and;

(ii) maturing the progenitor T cells to produce a population of TCR ab+ T cells, wherein one or both of (i) and (ii) are performed in the presence of Inducible Co-stimulator ligand (ICOS-L).

The presence of ICOS-L in steps (i) and/or (ii) may increase the proportion of HPCs or progenitor T cells that mature into TCR ab+ cells.

In some embodiments of the first aspect, a method of producing a population of TCR ab+T cells may comprise;

(i) differentiating a population of HPCs into T cell progenitor cells in the presence of ICOS-L; and

(ii) maturing the progenitor T cells to produce a population of TCR ab+ T cells. The presence of ICOS-L in step (i) may increase the proportion of HPCs that differentiate into TCR ab+ progenitor T cells, for example TCR ab+ CD45+CD3+ cells, and are matured into TCR ab+ T cells.

In other embodiments of the first aspect, a method of producing a population of TCR c^+ T cells may comprise;

(i) differentiating a population of HPCs into T cell progenitors; and

(ii) maturing the progenitor T cells in the presence of ICOS-L to produce a population of TCR ab+ T cells.

The presence of ICOS-L in step (ii) may increase the proportion of progenitor T cells that mature into TCR ab+ cells, for example TCR ab+ CD8+ CD4+ cells.

The cells may be differentiated or matured in a medium comprising ICOS-L or on a surface, such as a culture vessel surface, coated with ICOS-L.

Preferably, HPCs for use in the methods of the first aspect may be produced by differentiating a population of induced pluripotent stem cells (iPSCs) into HPCs.

In some embodiments of the first aspect, a method of producing a population of TCR ab+ cells may comprise;

(i) differentiating a population of iPSCs into HPCs;

(ii) differentiating the HPCs into progenitor T cells and;

(iii) maturing the progenitor T cells into a population of TCR ab+ T cells, wherein one or both of steps (ii) and (iii) are performed in the presence of Inducible Co-stimulator ligand (ICOS-L).

A method of the first aspect may further comprise activating and expanding the TCR ab+ T cells to produce a population of TCR ab+ CD8+ T cells or a population of TCR ab+ CD4+ T cells.

A second aspect of the invention provides a population of TCR ab+ T cells produced by a method of the first aspect.

A third aspect of the invention provides a pharmaceutical composition comprising a population of TCR ab+ T cells of the second aspect and a pharmaceutically acceptable excipient.

A fourth aspect of the invention provides a method of treatment comprising administering a therapeutically effect amount of a population of TCR ab+ T cells of the second aspect to an individual in need thereof.

A fifth aspect provides a coating composition for a T cell culture surface, the composition comprising ICOS-L. A sixth aspect provides a culture vessel for T cell culture, said vessel comprising a surface coated with ICOS-L.

These and other aspects and embodiments of the invention are described in more detail below.

Brief Description of Figures

Figure 1 shows an assessment of the percentage of TCRap+ T cells in CD45+ CD3+ population (raw data) at end of stage 4. CD34+ cells were untreated or treated with ICOS-L (0.5, 5, 50pg/ml). The data are shown as the percentage of TCRap+ cells in CD45+ CD3+ population. Data shows mean +/- SD (WT n=2 for untreated and 5pg/ml and n=1 for 0.5 and 50pg/ml, C3F3 n=3 for untreated and 5pg/ml and n=2 for 0.5 and 50pg/ml and C1 A12 n=3 for untreated and 5pg/ml and n=2 for 0.5 and 50pg/ml).

Figure 2 shows an assessment of the percentage of TCRap+ T cells in CD45+ CD3+ population at end of stage 4 - pooled data. CD34+ cells were untreated or treated with ICOS-L (0.5, 5, 50pg/ml). The data are shown as the percentage of TCRap+ cells in CD45+ CD3+ population. Data shows mean +/- SD (n=8, pooled data from WT, C3F3 and C1A12 n=8 for untreated and 5pg/ml and n=5 for 0.5 and 50pg/ml). Data were analysed using paired t test **p<0.01 and *** p<0.001 .

Figure 3 shows an assessment of the percentage of TCRap+ T cells in CD45+ CD8+ CD4+ and CD8+ CD4- population at end of stage 5. Cells from DD143, DD149, DD157-E17 were untreated or treated with ICOS-L (0.5, 5, 50pg/ml). Data shows mean +/- SD (n=3). (A) The data are shown as the percentage of TCRap+ cells in CD45+ CD4+ CD8+ population. (B) The data are shown as the percentage of TCRap+ cells in CD45+ CD4- CD8+ population. Data were analysed using paired t test *p<0.05.

Figure 4 shows a schematic view of an example of a six-stage method for generating T cells from iPSCs.

Detailed Description

This invention relates to the use of Inducible Co-stimulator ligand (ICOS-L) to increase the expression of ab T cell receptor. The presence of ICOS-L during the differentiation of haematopoietic progenitor cells into progenitor T cells and/or the maturation of progenitor T cells into double positive CD4+ CD8+ T cells is shown herein to increase the proportion of T cells in the population that express ab T cell receptor. The double positive CD4+ CD8+ TCR ab+ T cells may then be activated and expanded to produce single positive CD8+ or CD4+ TCR ab+ T cells which may be useful, for example, in immunotherapy.

Populations of TCR ab+ T cells are produced as described herein by (i) differentiating a population of haematopoietic progenitor cells into progenitor T cells and (ii) maturing the progenitor T cells to produce a population of CD4+ CD8+ T cells, wherein one or both of (i) and (ii) are performed in the presence of Inducible Co-stimulator ligand (ICOS-L). The presence of ICOS-L in either or both steps increases the proportion of cells that differentiate and/or mature into TCR ab+ T cells. The TCR ab+ cells may be CD4+ CD8+ T cells, which may then be activated and expanded into single positive CD8+ or single positive CD4+

T cells. The presence of ICOS-L during the differentiation of haematopoietic progenitor cells (HPCs) into T cell progenitors may increase the proportion of TCR ab+ cells in the population of progenitor T cells relative to the absence of ICOS-L during the differentiation. For example, the presence of ICOS-L may increase the proportion of TCR ab+ cells in the population of CD3+ cells. The presence of ICOS-L during the maturation of T cell progenitor cells into TCR ab+ T cells may increase the proportion of progenitor T cells that mature into TCR ab+ T cells relative to the absence of ICOS-L during the maturation. For example, the proportion of TCR ab+ CD8+ single positive (SP) T cells and/or the proportion of TCR ab+ CD4+ CD8+ double positive (DP) T cells may be increased by ICOS-L.

A method described herein may further comprise providing a population of haematopoietic progenitor cells (HPCs) for use in a method of producing TCR ab+ T cells as described herein.

HPCs (also called hematopoietic stem and progenitor cells or HSPCs) are multipotent stem cells that are committed to a hematopoietic lineage and are capable of further hematopoietic differentiation into all blood cell types including myeloid and lymphoid lineages, including monocytes, B cells, NK cells, NKT cells, TILs and T cells. HPCs may express CD34. HPCs may co-express CD45. HPCs may also co-express CD117, CD133, CD45 and FLK1 (also known as KDR or VEGFR2). HPCs may be negative for expression of CD38 and other lineage specific markers. For example, HPCs may display one or more, preferably all of CD34+ CD133+ CD45+ FLK1+ CD38-.

In some preferred embodiments, HPCs are produced in vitro trom induced pluripotent stem cells (iPSCs).

For example, a population of TCR ab+ T cells may be produced by a method comprising;

(i) differentiating a population of iPSCs into HPCs,

(ii) differentiating the HPCs into T cell progenitors; and

(ii) maturing the population of progenitor T cells to produce a population of TCR c^+ T cells, wherein one or both of step (ii) and step (iii) is performed in presence of ICOS-L. iPSCs may be differentiated into HPCs using a three-step process that includes mesoderm and haemogenic endothelium stages. For example, a method may comprise;

(i) differentiating the population of iPSCs into mesoderm cells,

(ii) differentiating the mesoderm cells into haemogenic endothelial cells, and

(iii) differentiating the haemogenic endothelial cells into a population of HPCs.

Induced pluripotent stem cells (iPSCs) are pluripotent cells which are derived from non-pluripotent, fully differentiated donor or antecedent cells. iPSCs are capable of self-renewal in vitro and exhibit an undifferentiated phenotype and are potentially capable of differentiating into any foetal or adult cell type of any of the three germ layers (endoderm, mesoderm and ectoderm). The population of iPSCs may be clonal i.e. genetically identical cells descended from a single common ancestor cell. iPSCs may express one or more of the following pluripotency associated markers: POU5f1 (Oct4), Sox2, Alkaline Phosphatase, SSEA-3, Nanog, SSEA-4, Tra-1-60, KLF4 and c-myc, preferably one or more of POU5f1 , NANOG and SOX2. An iPSC may lack markers associated with specific differentiative fates, such as Bra, Sox17, FoxA2, aFP, Sox1 , NCAM, GATA6, GATA4, Handl and CDX2. In particular, an iPSC may lack markers associated with endodermal fates.

Preferably, the iPSCs are human IPSCs (hiPSCs).

In some embodiments, iPSCs may be gene edited, for example to inactivate or delete HLA genes or other genes associated with immunogenicity or GVHD, or may be gene edited to include nucleic acid encoding an exogenous antigen receptor such as for example a TCR, CAR or NKCR.

IPSCs may be derived or reprogramed from donor cells, which may be somatic cells or other antecedent cells obtained from a source, such as a donor individual. The donor cells may be mammalian, preferably human cells. Suitable donor cells include adult fibroblasts and blood cells, for example peripheral blood cells, such as HPCs or mononuclear cells.

Suitable donor cells for reprogramming into iPSCs as described herein may be obtained from a donor individual. In some embodiments, the donor individual may be the same person as the recipient individual to whom the T cells will be administered following production as described herein (autologous treatment). In other embodiments, the donor individual may be a different person to the recipient individual to whom the T cells will be administered following production as described herein (allogeneic treatment). For example, the donor individual may be a healthy individual who is human leukocyte antigen (HLA) matched (either before or after donation) with a recipient individual suffering from cancer. In other embodiments, the donor individual may not be HLA matched with the recipient individual. Preferably, the donor individual may be a neonate (new-born), for example the donor cells may be obtained from a sample of umbilical cord blood.

Suitable donor individuals are preferably free of communicable viral (e.g. HIV, HPV, CMV) and adventitious agents (e.g. bacteria, mycoplasma), and free of known genetic abnormalities.

In some embodiments, a population of peripheral blood cells, such as HPCs, for reprogramming may be isolated from a blood sample, preferably an umbilical cord sample, obtained from the donor individual. Suitable methods for the isolation of HPCs and other peripheral blood cells, are well-known in the art and include, for example magnetic activated cell sorting (see, for example, Gaudernack et al 1986 J Immunol Methods 90 179), fluorescent activated cell sorting (FACS: see for example, Rheinherz et al (1979) PNAS 76 4061), and cell panning (see for example, Lum et al (1982) Cell Immunol 72 122). HPCs may be identified in a sample of blood cells by expression of CD34. In other embodiments, a population of fibroblasts for reprogramming may be isolated from a skin biopsy following dispersal using collagenase or trypsin and outgrowth in appropriate cell culture conditions. In some embodiments, IPSCs may be derived from antigen-specific T cells. For example, the T cells may comprise nucleic acid encoding ab TCRs that bind to an antigen, such as a tumor antigen, displayed in complex with a class 1 MHC. Antigen-specific T cells for use in the generation of iPSCs may be obtained by screening a diverse population of T cells with peptide epitopes from the target antigen displayed on a class I or II MHC molecule on the surface of an antigen presenting cell, such as a dendritic cell, or by isolating from a tumour sample from a cancer patient.

Donor cells are typically reprogrammed into iPSCs by the introduction of reprogramming factors, such as Oct4, Sox2 and Klf4 into the cell. The reprogramming factors may be proteins or encoding nucleic acids and may be introduced into the differentiated cells by any suitable technique, including plasmid, transposon or more preferably, viral transfection or direct protein delivery. Other reprogramming factors, for example Klf genes, such as Klf-1 , -2, -4 and -5; Myc genes such as C-myc, L-myc and N-myc; Nanog; SV40 Large T antigen; Lin28; and short hairpins (shRNA) targeting genes such as p53, may also be introduced into the cell to increase induction efficiency. Following introduction of the reprogramming factors, the donor cells may be cultured. Cells expressing pluripotency markers may be isolated and/or purified to produce a population of iPSCs. Techniques for the production of iPSCs are well-known in the art (Yamanaka et al Nature 2007; 448:313-7; Yamanaka 62007 Jun 7; 1(1):39-49; Kim et al Nature. 2008 Jul 31 ; 454(7204):646-50;

Takahashi Cell. 2007 Nov 30; 131 (5):861 -72. Park et al Nature. 2008 Jan 10; 451 (7175):141-6; Kimet et al Cell Stem Cell. 2009 Jun 5;4(6):472-6; Vallier, L, et al. Stem Cells, 2009. 9999(999A): p. N/A; Baghbaderani et al 2016; Stem Cell Rev. 2016 Aug; 12(4):394-420; Baghbaderani et al. (2015) Stem Cell Reports, 5(4), 647-659).

Conventional techniques may be employed for the culture and maintenance of iPSCs (Vallier, L. et al Dev. Biol. 275, 403-421 (2004), Cowan, C.A. et al. N. Engl. J. Med. 350, 1353-1356 (2004), Joannides, A. et al. Stem Cells 24, 230-235 (2006) Klimanskaya, I. et al. Lancet 365, 1636-1641 (2005), Ludwig, T.E. et al. Nat. Biotechnol. 24, 185-187 (2006)). IPSCs for use in the present methods may be grown in defined conditions or on feeder cells. For example, iPSCs may be conventionally cultured in a culture dish on a layer of feeder cells, such as irradiated mouse embryonic fibroblasts (MEF), at an appropriate density (e.g. 10 5 to 10 6 cells/60mm dish), or on an appropriate substrate, in a feeder conditioned or defined iPSC maintenance medium. iPSCs for use in the present methods may be passaged by enzymatic or mechanical means. In some embodiments, iPSCs may be passaged on matrigel™ or an ECM protein, such as vibronectin, in an iPSC maintenance medium, such as mTeSR™1 orTeSR™2 (StemCell Technologies) or E8 flex (Life Thermo) culture medium.

Differentiation and maturation of the cell populations in the steps of the methods described herein is induced by culturing the cells in a culture medium supplemented with a set of differentiation factors. The set of differentiation factors that is listed for each culture medium is preferably exhaustive and medium may be devoid of other differentiation factors. In preferred embodiments, the culture media are chemically defined media. For example, a culture medium may consist of a chemically defined nutrient medium that is supplemented with an effective amount of one or more differentiation factors, as described below. A chemically defined nutrient medium may comprise a basal medium that is supplemented with one or more serum-free culture medium supplements.

Differentiation factors are factors which modulate, for example promote or inhibit, a signalling pathway which mediates differentiation in a mammalian cell. Differentiation factors may include growth factors, cytokines and small molecules which modulate one or more of the Activin/Nodal, FGF, Wnt or BMP or signalling pathways thereof. Examples of differentiation factors include Activin/Nodal, FGFs, BMPs, retinoic acid, vascular endothelial growth factor (VEGF), stem cell factor (SCF), TGFp ligands, GDFs, LIF, Interleukins, GSK-3 inhibitors and phosphatidylinositol 3-kinase (PI3K) inhibitors.

Differentiation factors which are used in one or more of the media described herein include TGFp ligands, such as activin, fibroblast growth factor (FGF), bone morphogenetic protein (BMP), stem cell factor (SCF), vascular endothelial growth factor (VEGF), GSK-3 inhibitors (such as CHIR-99021), interleukins, and hormones, such as IGF-1 and angiotensin II. A differentiation factor may be present in a medium described herein in an amount that is effective to modulate a signalling pathway in cells cultured in the medium.

In some embodiments, a differentiation factor listed above or below may be replaced in a culture medium by a factor that has the same effect (i.e. stimulation or inhibition) on the same signalling pathway. Suitable factors are known in the art and include proteins, nucleic acids, antibodies and small molecules.

The extent of differentiation of the cell population during each step may be determined by monitoring and/or detecting the expression of one or more cell markers in the population of differentiating cells. For example, an increase in the expression of markers characteristic of the more differentiated cell type or a decrease in the expression of markers characteristic of the less differentiated cell type may be determined. The expression of cell markers may be determined by any suitable technique, including immunocytochemistry, immunofluorescence, RT-PCR, immunoblotting, fluorescence activated cell sorting (FACS), and enzymatic analysis. In preferred embodiments, a cell may be said to express a marker if the marker is detectable on the cell surface. For example, a cell which is stated herein not to express a marker may display active transcription and intracellular expression of the marker gene but detectable levels of the marker may not be present on the surface of the cell.

A population of partially differentiated cells, for example mesoderm cells, haemogenic endothelium (HE; i.e. haemogenic endothelial cells or HECs), HPCs, or T cell progenitors, that is produced by a step in the methods described herein may be cultured, maintained or expanded before the next differentiation step. Partially differentiated cells may be expanded by any convenient technique.

After each step, the population of partially differentiated cells which is produced by that step may be free or substantially free from other cell types. For example, the population may contain 60% or more, 70% or more, 80% or more or 90% or more partially differentiated cells, following culture in the medium. Preferably, the population of cells is sufficiently free of other cell types that no purification is required. If required, the population of partially differentiated cells may be purified by any convenient technique, such as MACs or FACS.

Cells may be cultured in a monolayer, in the absence of feeder cells, on a surface or substrate coated with extracellular matrix protein, such as fibronectin, laminin or collagen. Suitable techniques for cell culture are well-known in the art (see, for example, Basic Cell Culture Protocols, C. Helgason, Humana Press Inc. U.S. (15 Oct 2004) ISBN: 1588295451 ; Human Cell Culture Protocols (Methods in Molecular Medicine S.) Humana Press Inc., U.S. (9 Dec 2004) ISBN: 1588292223; Culture of Animal Cells: A Manual of Basic Technique, R. Freshney, John Wiley & Sons Inc (2 Aug 2005) ISBN: 0471453293, Ho WY et al J Immunol Methods. (2006) 310:40-52, Handbook of Stem Cells (ed. R. Lanza) ISBN: 0124366430) Basic Cell Culture Protocols’ by J. Pollard and J. M. Walker (1997), ‘Mammalian Cell Culture: Essential Techniques’ by A.

Doyle and J. B. Griffiths (1997), ‘Human Embryonic Stem Cells’ by A. Chiu and M. Rao (2003), Stem Cells: From Bench to Bedside’ by A. Bongso (2005), Peterson & Loring (2012)Human Stem Cell Manual: A Laboratory Guide Academic Press and ‘Human Embryonic Stem Cell Protocols’ by K. Turksen (2006).

Media and ingredients thereof may be obtained from commercial sources (e.g. Gibco, Roche, Sigma, Europa bioproducts, R&D Systems). Standard mammalian cell culture conditions may be employed for the above culture steps, for example 37°C, 5% or 21% Oxygen, 5% Carbon Dioxide. Media is preferably changed every two days and cells allowed to settle by gravity.

Cells may be cultured in a culture vessel. Suitable cell culture vessels are well-known in the art and include culture plates, dishes, flasks, bioreactors, and multi-well plates, for example 6-well, 12-well or 96-well plates. The culture vessel are preferably treated for tissue culture, for example by coating one or more surfaces of the vessel with an extracellular matrix protein, such as fibronectin, laminin or collagen. Culture vessels may be treated for tissue culture using standard techniques, for example by incubating with a coating solution as described herein, or may be obtained from pre-treated from commercial suppliers.

In a first stage, iPSCs may be differentiated into mesoderm cells by culturing the population of iPSCs under suitable conditions to promote mesodermal differentiation. For example, the iPSCs cells may be cultured sequentially in first, second and third mesoderm induction media to induce differentiation into mesoderm cells.

A suitable first mesoderm induction medium may stimulate SMAD2 and SMAD3 and/or SMAD2 and SMAD3 mediated signalling pathways. For example, the first mesoderm induction medium may comprise activin.

A suitable second mesoderm induction medium may (i) stimulate SMAD1 , SMAD2, SMAD3, SMAD5 and SMAD9 and/or SMAD1 , SMAD2, SMAD3, SMAD5 and SMAD9 mediated signalling pathways and (ii) have fibroblast growth factor (FGF) activity. For example, the second mesoderm induction medium may comprise activin, preferably activin A, BMP, preferably BMP4 and FGF, preferably bFGF.

A suitable third mesoderm induction medium may (i) stimulate SMAD1 , SMAD2, SMAD3, SMAD5 and SMAD9 and/or SMAD1 , SMAD2, SMAD3, SMAD5 and SMAD9 mediated signalling pathways (ii) have fibroblast growth factor (FGF) activity and (iii) inhibit glycogen synthase kinase 3b. For example, the third mesoderm induction medium may comprise activin, preferably activin A, BMP, preferably BMP4, FGF, preferably bFGF, and a GSK3 inhibitor, preferably CHIR99021.

The first, second and third mesoderm induction media may be devoid of differentiation factors other than the differentiation factors set out above.

SMAD2 and SMAD3 and/or SMAD2 and SMAD3 mediated intracellular signalling pathways may be stimulated by the first, second and third mesoderm induction media through the presence in the media of a first TGFp ligand. The first TGFp ligand may be Activin. Activin (Activin A: NCBI Gene ID: 3624 nucleic acid reference sequence NM_002192.2 Gl: 62953137, amino acid reference sequence NP_002183.1 Gl: 4504699) is a dimeric polypeptide which exerts a range of cellular effects via stimulation of the Activin/Nodal pathway (Vallier et al., Cell Science 118:4495-4509 (2005)). Activin is readily available from commercial sources (e.g. Stemgent Inc. MA USA; Miltenyi Biotec Gmbh, DE). Conveniently, the concentration of Activin in a medium described herein may be from 1 to 100ng/ml, preferably about 5 to 50ng/ml.

The fibroblast growth factor (FGF) activity of the second and third mesoderm induction media may be provided by the presence of fibroblast growth factor (FGF) in the media. Fibroblast growth factor (FGF) is a protein factor which stimulates cellular growth, proliferation and cellular differentiation by binding to a fibroblast growth factor receptor (FGFR). Suitable fibroblast growth factors include any member of the FGF family, for example any one of FGF1 to FGF14 and FGF15 to FGF23. Preferably, the FGF is FGF2 (also known as bFGF, NCBI GenelD: 2247, nucleic acid sequence NM_002006.3 Gl: 41352694, amino acid sequence NP_001997.4 Gl: 41352695); FGF7 (also known as keratinocyte growth factor (or KGF), NCBI GenelD: 2247, nucleic acid sequence NM_002006.3 Gl: 41352694, amino acid sequence NP_001997.4 Gl: 41352695); or FGF10 (NCBI GenelD: 2247, nucleic acid sequence NM_002006.3 Gl: 41352694, amino acid sequence NP_001997.4 Gl: 41352695). Most preferably, the fibroblast growth factor is FGF2.

Conveniently, the concentration of FGF, such as FGF2 in a medium described herein may be from 0.5 to 50ng/ml, preferably about 5ng/ml. Fibroblast growth factors, such as FGF2, FGF7 and FGF10, may be produced using routine recombinant techniques or obtained from commercial suppliers (e.g. R&D Systems, Minneapolis, MN; Stemgent Inc, USA; Miltenyi Biotec Gmbh, DE).

SMAD1 , SMAD5 and SMAD9 and/or SMAD1 , SMAD5 and SMAD9 mediated intracellular signalling pathways may be stimulated by the second and third mesoderm induction media through the presence in the media of a second TGFp ligand.

The second TGFp ligand may be a Bone Morphogenic Protein (BMP). Bone Morphogenic Proteins (BMPs) bind to Bone Morphogenic Protein Receptors (BMPRs) and stimulate intracellular signalling through pathways mediated by SMAD1 , SMAD5 and SMAD9. Suitable Bone Morphogenic Proteins include any member of the BMP family, for example BMP2, BMP3, BMP4, BMP5, BMP6 or BMP7. Preferably the second TGFp ligand is BMP2 (NCBI GenelD: 650, nucleic acid sequence NM_001200.2 Gl: 80861484; amino acid sequence NP_001191.1 Gl: 4557369) or BMP4 (NCBI GenelD: 652, nucleic acid sequence NM_001202.3 Gl: 157276592; amino acid sequence NP_001193.2 Gl: 157276593). Suitable BMPs include BMP4. Conveniently, the concentration of a Bone Morphogenic Protein, such as BMP2 or BMP4 in a medium described herein may be from 1 to 500ng/ml, preferably about 10ng/ml. BMPs may be produced using routine recombinant techniques or obtained from commercial suppliers (e.g. R&D, Minneapolis, USA, Stemgent Inc, USA; Miltenyi Biotec Gmbh, DE).

The GSK3p inhibition activity of the third mesoderm induction medium may be provided by the presence of a GSK3p inhibitor in the medium. GSK3p inhibitors inhibit the activity of glycogen synthase kinase 3b (Gene ID 2932: EC2.7.11.26). Preferred inhibitors specifically inhibit the activity of glycogen synthase kinase 3b. Suitable inhibitors include CHIR99021 ( 6-((2-((4-(2,4-Dichlorophenyl)-5-(4-methyl-1H-imidazol-2-yl) pyrimidin - 2-yl)amino)ethyl)amino)nicotinonitrile\ Ring D. B. et al., Diabetes, 52:588-595 (2003)) alsterpaullone, kenpaullone, BIO(6-bromoindirubin-3'-oxime (Sato et al Nat Med. 2004 Jan;10(1):55-63), SB216763 ( 3-(2,4 - dichlorophenyl)-4-(1 -methyl-1 H-indol-3-yl)-1 H-pyrrole-2,5-dione), Lithium and SB415286 ( 3-[(3-chloro-4 - hydroxyphenyl)amino]-4-(2-nitrophenyl)-1 H-pyrrole-2,5-dione; Coghlan et al Chem Biol. 2000 Oct;7(10):793- 803). In some preferred embodiments, the ObK3b inhibitor is CHIR99021.Suitable glycogen synthase kinase 3b inhibitors may be obtained from commercial suppliers (e.g. Stemgent Inc. MA USA; Cayman Chemical Co. Ml USA; Selleckchem, MA USA). For example, the third mesoderm induction medium may contain 0.1 to 100pM of a 0eK3b inhibitor, such as CHIR99021 , preferably about 10pM.

In preferred embodiments, the first, second and third mesoderm induction media are chemically defined media. For example, the first mesoderm induction medium may consist of a chemically defined nutrient medium supplemented with an effective amount of activin, preferably activin A, for example 50ng/ml activin A; the second mesoderm induction medium may consist of a chemically defined nutrient medium supplemented with an effective amount of activin preferably activin A, for example 5ng/ml activin A, BMP, preferably BMP4, for example 10ng/ml BMP4; and FGF, preferably bFGF (FGF2), for example 5ng/ml bFGF; and the third mesoderm induction medium may consist of a chemically defined nutrient medium supplemented with an effective amount of activin preferably activin A, for example 5ng/ml activin A, BMP, preferably BMP4, for example 10ng/ml BMP4; FGF, preferably bFGF (FGF2), for example 5ng/ml bFGF; and GSK3 inhibitor, preferably CHIR-99021 , for example 10pM CHIR-99021.

A chemically defined medium (CDM) is a nutritive solution for culturing cells which contains only specified components, preferably components of known chemical structure. A CDM is devoid of undefined components or constituents which include undefined components, such as feeder cells, stromal cells, serum, serum albumin and complex extracellular matrices, such as matrigel™ For example, a CDM does not contain stromal cells, such as OP9 cells, expressing Notch ligands, such as DLL1 or DLL4.

The CDM or chemically defined nutrient medium may comprise a chemically defined basal medium. Suitable chemically defined basal media include Iscove’s Modified Dulbecco’s Medium (IMDM), Ham’s F12,

Advanced Dulbecco’s modified eagle medium (DMEM) (Price et al Focus (2003), 25 3-6), Williams E (Williams, G.M. et al Exp. Cell Research, 89, 139-142 (1974)), RPMI-1640 (Moore, G.E. and Woods L.K., (1976) Tissue Culture Association Manual. 3, 503-508) and StemPro™-34 (ThermoFisher Scientific).

The basal medium may be supplemented by serum-free culture medium supplements and/or additional components in the medium. Suitable supplements and additional components are described above and may include L-glutamine or substitutes, such as GlutaMAX-1™, ascorbic acid, monothiolglycerol (MTG), antibiotics such as penicillin and streptomycin, human serum albumin, for example recombinant human serum albumin, such as Cellastim™ (Merck/Sigma) and Recombumin™ (albumedix.com), insulin, transferrin and 2-mercaptoethanol. A basal medium may be supplemented with a serum substitute, such as Knockout Serum Replacement (KOSR; Invitrogen).

The iPSCs may be cultured in the first mesoderm induction medium for 1 to 12 hours, for example any of 1 ,

2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 hours, preferably about 4 hours; then cultured in the second mesoderm induction medium for 30 to 54 hours, for example any of 35, 36, 37, 38, 39, 40, 41 , 42, 43, 44, 45, 46, 47, 48, 49 or 50 hours, preferably about 44 hours; and then cultured in the third mesoderm induction medium for 36 to 60 hours, for example any of 37, 38, 39, 40, 41 , 42, 43, 44, 45, 46, 47, 48, 49, 50, 51 , 52 or 53 hours, preferably about 48 hours to produce a population of mesodermal cells.

Mesoderm cells are partially differentiated progenitor cells that are committed to mesodermal lineages and are capable of differentiation under appropriate conditions into all cell types in the mesenchyme (fibroblast), muscle, bone, adipose, vascular and haematopoietic systems. Mesoderm cells may express one or more mesodermal markers. For example, the mesoderm cells may express any one, two, three, four, five, six or all seven of Brachyury, Goosecoid, MixM , KDR, FoxA2, GATA6 and PDGFaR.

In a second stage, mesoderm cells may be differentiated into haemogenic endothelial (HE) cells by culturing the population of mesoderm cells under suitable conditions to promote HE differentiation. For example, the mesoderm cells cells may be cultured in an HE induction medium.

A suitable HE induction medium may (i) stimulate cKIT receptor (CD117; KIT receptor tyrosine kinase) and/or cKIT receptor (CD117; KIT receptor tyrosine kinase) mediated signalling pathways and (ii) stimulate VEGFR and/or VEGFR mediated signalling pathways. For example, the HE induction medium may comprise SCF and VEGF.

Vascular endothelial growth factor (VEGF) is a protein factor of the PDGF family which binds to VEGFR tyrosine kinase receptors and stimulates vasculogenesis and angiogenesis. Suitable VEGFs include any member of the VEGF family, for example any one of VEGF-A to VEGF-D and PIGF. Preferably, the VEGF is VEGF-A (also known as VEGF, NCBI Gene ID: 7422, nucleic acid sequence NM_001025366.2, amino acid sequence NP_001020537.2). Preferably, the VEGFR and/or VEGFR mediated signalling pathways are VEGFR2 (KDR/Flk-1) and/or VEGFR2 (KDR/Flk-1) mediated signalling pathways. VEGF is readily available from commercial sources (e.g. R&D Systems, USA). Conveniently, the concentration of VEGF in an HE induction medium described herein may be from 1 to 10Ong/ml, for example any of about 5, 7, 10, 12, 15, 17, 20, 25, 30, 35, 40, 45 or 50 ng/ml, preferably about 15 ng/ml.

In some examples of HE induction media, VEGF may be replaced by a VEGF activator or agonist that stimulates VEGFR (or VEGFR2 (KDR/Flk-1)) and/or VEGFR (or VEGFR2 (KDR/Flk-1)) mediated signalling pathways. Suitable VEGF activators are known in the art and include proteins, such as gremlin (Mitola et al (2010) Blood 116(18) 3677-3680) nucleic acids, such as shRNA (e.g. Turunen et al Circ Res. 2009 Sep 11 ; 105(6):604-9), CRISPR-based plasmids (e.g. VEGF CRISPR activation plasmid; Santa Cruz Biotech, USA), antibodies and small molecules.

Stem cell factor (SCF) is a cytokine that binds to the KIT receptor (KIT proto-oncogene, receptor tyrosine kinase) (CD117; SCFR) and is involved in haematopoiesis. SCF (also called KITLG, NCBI GenelD: 4254) may have the reference nucleic acid sequence NM_000899.5 or NM_03994.5 and the reference amino acid sequence NP_000890.1 or NP_003985.5. SCF is readily available from commercial sources (e.g. R&D Systems, USA). Conveniently, the concentration of SCF in an HE induction medium described herein may be from 1 to 1000ng/ml, for example any of about 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900 ng/ml, preferably about 100 ng/ml.

In preferred embodiments, the HE induction medium is a chemically defined medium. For example, the HE induction medium may consist of a chemically defined nutrient medium supplemented with effective amounts of VEGF, for example 15ng/ml VEGF; and SCF, for example 100ng/ml SCF. Preferably, mesoderm cells are cultured in an HE induction medium consisting of a chemically defined nutrient medium and two differentiation factors, wherein the two differentiation factors being SCF and VEGF.

Suitable chemically defined nutrient media are described above and include StemPro™-34 (ThermoFisher Scientific) or a basal medium such as IMDM supplemented with albumin, insulin, selenium transferrin, and lipids as described below.

The mesoderm cells may be cultured in the HE induction medium for 2 to 6 days or 3 to 5 days, preferably about 4 days, to produce a population of HE cells.

Haemogenic endothelial (HE) are partially differentiated endothelial progenitor cells that have hematopoietic potential and are capable of differentiation under appropriate conditions into haematopoietic lineages. HE cells may express CD34 and, in some embodiments, may not express CD73 or CXCR4 (CD184). In some embodiments, the HE cells may have the phenotype D34+ CD73-, or the phenotype CD34+ CD73- CXCR4-.

In a third stage, haemogenic endothelial (HE) cells may be differentiated into haematopoietic progenitor cells (HPCs) by culturing the population of HE cells under suitable conditions to promote haematopoietic differentiation. For example, the HE cells may be cultured in a haematopoietic induction medium. A suitable haematopoietic induction medium may stimulate the following (i) cKIT receptor (CD 117; KIT receptor tyrosine kinase) and/or cKIT receptor (CD117; KIT receptor tyrosine kinase) mediated signalling pathways, (ii) VEGFR and/or VEGFR mediated signalling pathways, preferably VEGFR2 and/or VEGFR2 mediated signalling pathways, (iii) MPL (CD110) and/or MPL (CD110) mediated signalling pathways (iv)

FLT3 and/or FLT3 mediated signalling pathways (v) IGF1R and/or IGF1R mediated signalling pathways (vi) SMAD1 , 5 and 9 and/or SMAD1 , 5 and 9 mediated signalling pathways (vii) Hedgehog and/or Hedgehog signalling pathways (viii) EpoR and/or EpoR mediated signalling pathway and (ix) AGTR2 and/or AGTR2 mediated signalling pathways. A suitable haematopoietic induction medium may also inhibit AGTR1 (angiotensin II type 1 receptor (ATi)) and/or the AGTR1 (angiotensin II type 1 receptor (ATi)) mediated signaling pathway. A suitable haematopoietic induction medium may also have interleukin (IL) activity and FGF activity.

For example, a haematopoietic induction medium may comprise the differentiation factors: VEGF, SCF, Thrombopoietin (TPO), Flt3 ligand (Flt3L), IL-3, IL-6, IL-7, IL-11 , IGF-1 , BMP, FGF, Sonic hedgehog (SHH), erythropoietin (EPO), angiotensin II, and an angiotensin II type 1 receptor (ATi) antagonist. An example of a suitable haematopoietic induction medium is the Stage 3 medium shown in Table 1 below.

Thrombopoietin (TPO) is a glycoprotein hormone that regulates platelet production. TPO (also called THPO, NCBI Gene ID: 7066) may have the reference nucleic acid sequence NM_000460.4 and the reference amino acid sequence NP_000451.1. TPO is readily available from commercial sources (e.g. R&D Systems, USA; Miltenyi Biotec Gmbh, DE). Conveniently, the concentration of TPO in a haematopoietic induction medium described herein may be from 3 to 300ng/ml, for example any of about 0.1 , 0.25, 0.5, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90 or 100, 110, 120, 130, 140, 150, 160, 170,

180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280 or 290 ng/ml, preferably about 30 ng/ml.

Flt3 ligand (Fms- related tyrosine kinase 3 ligand or FLT3L) is a cytokine with haematopoietic activity which binds to the FLT3 receptor and stimulates the proliferation and differentiation of progenitor cells. Flt3 ligand (also called FLT3LG, NCBI GenelD: 2323) may have the reference nucleic acid sequence NM_001204502.2 and the reference amino acid sequence NP_001191431.1. Flt3 is readily available from commercial sources (e.g. R&D Systems, USA; Miltenyi Biotec Gmbh, DE). Conveniently, the concentration of Flt3 ligand in a haematopoietic induction medium described herein may be from 0.25 to 250ng/ml, for example any of about 0.1 , 0.25, 0.5, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90 or 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230 or 240 ng/ml, preferably about 25 ng/ml.

Interleukins (ILs) are cytokines that play major roles in immune development and function. ILs in a haematopoietic induction medium may include IL-3, IL-6, IL-7, and IL-11.

IL-3 (also called IL3 or MCGF, NCBI GenelD: 3562) may have the reference nucleic acid sequence NM_000588.4 and the reference amino acid sequence NP_000579.2. IL-3 is readily available from commercial sources (e.g. R&D Systems, USA; Miltenyi Biotec Gmbh, DE). Conveniently, the concentration of IL-3 in a haematopoietic induction medium described herein may be from 0.25 to 250ng/ml, for example any of about 0.1 , 0.25, 0.5, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90 or 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230 or240 ng/ml, preferably about 25 ng/ml.

IL-6 (also called IL6 or HGF, NCBI GenelD: 3569) may have the reference nucleic acid sequence NM_000600.5 and the reference amino acid sequence NP_000591.5. IL-6 is readily available from commercial sources (e.g. R&D Systems, USA; Miltenyi Biotec Gmbh, DE). Conveniently, the concentration of IL-6 in a haematopoietic induction medium described herein may be from 0.1 to 100ng/ml, for example any of about 0.1 , 0.25, 0.5, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90 or 95 ng/ml, preferably about 10 ng/ml.

IL-7 (also called IL7, NCBI GenelD: 3574) may have the reference nucleic acid sequence NM_000880.4 and the reference amino acid sequence NP_000871.1. IL-7 is readily available from commercial sources (e.g. R&D Systems, USA; Miltenyi Biotec Gmbh, DE). Conveniently, the concentration of IL-7 in a haematopoietic induction medium described herein may be from 0.1 to 100ng/ml, for example any of about 0.1 , 0.25, 0.5, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90 or 95 ng/ml, preferably about 10 ng/ml.

IL-11 (also called AGIF, NCBI GenelD: 3589) may have the reference nucleic acid sequence NM_000641.4 and the reference amino acid sequence NP_000632.1. IL-11 is readily available from commercial sources (e.g. R&D Systems, USA; Miltenyi Biotec Gmbh, DE). Conveniently, the concentration of IL-11 ligand in a haematopoietic induction medium described herein may be from 0.5 to 100ng/ml, for example any of about 0.1 , 0.25, 0.5, 0.75, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90 or 95 ng/ml, preferably about 5 ng/ml.

Insulin-like growth factor 1 (IGF-1) is a hormone that binds to the tyrosine kinases IGF-1 receptor (IGF1R) and insulin receptor and activates the multiple signalling pathways. IGF-1 (also called IGF or MGF, NCBI GenelD: 3479) may have the reference nucleic acid sequence NM_000618.5 and the reference amino acid sequence NP_000609.1. IGF-1 is readily available from commercial sources (e.g. R&D Systems, USA). Conveniently, the concentration of IGF-1 in a haematopoietic induction medium described herein may be from 0.25 to 250ng/ml, for example any of about 0.1 , 0.25, 0.5, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 20, 23, 25, 27, 30, 35, 40, 45, 50, 60, 70, 80, 90 or 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230 or 240 ng/ml, preferably about 25 ng/ml.

Sonic hedgehog (SHH) is a ligand of the hedgehog signalling pathway that regulates vertebrate organogenesis. SHH (also called TPT or HHG1 , NCBI GenelD: 6469) may have the reference nucleic acid sequence NM_000193.4 and the reference amino acid sequence NP_000184.1. SHH is readily available from commercial sources (e.g. R&D Systems, USA; Miltenyi Biotec Gmbh, DE). Conveniently, the concentration of SHH in a haematopoietic induction medium described herein may be from 0.25 to 250ng/ml, for example any of about 0.1 , 0.25, 0.5, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 20, 23, 25, 27, 30, 35, 40, 45, 50, 60, 70, 80, 90 or 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230 or 240 ng/ml, preferably about 25 ng/ml.

Erythropoietin (EPO) is a glycoprotein cytokine that binds to the erythropoietin receptor (EpoR) and stimulates erythropoiesis. EPO (also called DBAL, NCBI GenelD: 2056) may have the reference nucleic acid sequence NM_000799.4 and the reference amino acid sequence NP_000790.2. EPO is readily available from commercial sources (e.g. R&D Systems, USA; PreproTech, USA). Conveniently, the concentration of EPO in haematopoietic induction medium described herein may be from 0.02 to 20 U/ml, for example any of about 0.01 , 0.025, 0.05, 0.075, 0.1 , 0.5, 0.75, 1.0, 1 .5, 2.0, 2.5, 3, 4, 5, 6, 7, 8, 9, 10 13, 15, 17, or 19 U/ml, preferably about 2U/ml.

Angiotensin II is a heptapeptide hormone that is formed by the action of angiotensin converting enzyme (ACE) on angiotensin I. Angiotensin II stimulates vasoconstriction. Angiotensin I and II are formed by the cleavage of angiotensinogen (also called AGT, NCBI GenelD: 183), which may have the reference nucleic acid sequence NM_000029.4 and the reference amino acid sequence NP_000020.1 . Angiotensin II is readily available from commercial sources (e.g. R&D Systems, USA; Tocris, USA). Conveniently, the concentration of Angiotensin II in a haematopoietic induction medium described herein may be from 0.05 to 50ng/ml, for example any of about 0.01 , 0.025, 0.05, 0.075, 0.1 , 0.5, 0.75, 1.0, 1.5, 2.0, 2.5, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40 or 50 ng/ml, preferably about 5 ng/ml.

Angiotensin II type 1 receptor (ATi) antagonists (ARBs) are compounds that selectively block the activation of ATi receptor (AGTR1 ; Gene ID 185). Suitable ATi antagonists include losartan (2-Butyl-4-chloro-1-{[2'- (1 H-tetrazol-5-yl)-4-biphenylyl]methyl}-1 H-imidazol-5-yl)methanol), valsartan ((2S)-3-Methyl-2-(pentanoyl{[2'- (1 H-tetrazol-5-yl)biphenyl-4-yl]methyl}amino)butanoic acid), and telmisartan (4'[(1 ,4'-Dimethyl-2'-propyl[2,6'- bi-1 H-benzimidazol]-1 '-yl)methyl][1 ,1 '-biphenyl]-2-carboxylic acid. In some preferred embodiments, the ATi antagonist is losartan. Suitable ATi antagonists may be obtained from commercial suppliers (e.g. Tocris, USA; Cayman Chemical Co. Ml USA). Conveniently, the concentration of angiotensin II type 1 receptor (ATi) antagonist in a haematopoietic induction medium described herein may be from 1 to 1000 pM, for example any of about 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900 pM, preferably about 100 pM.

In preferred embodiments, the haematopoietic induction medium is a chemically defined medium. For example, the haematopoietic induction medium may consist of a chemically defined nutrient medium supplemented with effective amounts of VEGF, for example 15ng/ml; SCF, for example 100ng/ml; thrombopoietin (TPO), for example 30ng/ml; Flt3 ligand (FLT3L), for example 25ng./ml; IL-3, for example 25ng/ml; IL-6, for example 10ng/ml; IL-7, for example 10 ng/ml; IL-11 , for example 5 ng/ml; IGF-1 , for example 25 ng/ml; BMP, for example BMP4 at 10ng/ml; FGF, for example bFGF at 5ng/ml; Sonic hedgehog (SHH), for example 25ng/ml; erythropoietin (EPO), for example 2 u/ml; angiotensin II, for example 10gg/ml, and an angiotensin II type 1 receptor (ATi) antagonist, for example losartan, at 100pM. Suitable chemically defined nutrient media are described above and include StemPro™-34 PLUS (ThermoFisher Scientific) or a basal medium such as IMDM supplemented with albumin, insulin, selenium transferrin, and lipids as described below.

The HE cells may be cultured in the haematopoietic induction medium for 8-21 days, for example any of about 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19 or 20 days, preferably about 16 days, to produce the population of HPCs.

Following the generation of HPCs from HE cells, a population of HPCs expressing one or more cell surface markers, such as CD34, may be purified, for example by magnetic activated cell sorting (MACS), before being subjected to further differentiation. For example, a population of CD34+ HPCs may be purified. The CD34+ HPCs may be purified after 8 days, for example 8, 9 or10 days, culture in the HE induction medium. The CD34+ HPCs may be purified after 16 days of differentiation, for example on day 16, 17 or18 of the differentiation method.

In a fourth stage, haematopoietic progenitor cells (HPCs) may be differentiated into progenitor T cells by culturing the population of HPCs under suitable conditions to promote lymphoid differentiation. For example, the haematopoietic progenitor cells may be cultured in a lymphoid expansion medium.

Haematopoietic progenitor cells (HPCs) may be differentiated into progenitor T cells and DP (double positive) T cells in the absence of stromal cells, such as OP9-DI4 stromal cells, feeder cells or serum.

A lymphoid expansion medium is a cell culture medium that promotes the lymphoid differentiation of HPCs into progenitor T cells.

A suitable lymphoid expansion medium may (i) stimulate cKIT receptor (CD117; KIT receptor tyrosine kinase) and/or cKIT receptor (CD117; KIT receptor tyrosine kinase) mediated signalling pathways, (ii) stimulate MPL (CD110) and/or mediated signalling pathways (iii) FLT3 and/or FLT3 mediated signalling pathways and (iv) have interleukin (IL) activity. For example, a lymphoid expansion medium may comprise the differentiation factors SCF, FLT3L, TPO and IL7.

In preferred embodiments, the lymphoid expansion medium is a chemically defined medium. For example, the lymphoid expansion medium may consist of a chemically defined nutrient medium supplemented with effective amounts of the above differentiation factors. Suitable lymphoid expansion media are well-known in the art and include Stemspan™ SFEM II (Cat # 9605; StemCell Technologies Inc, CA).with Stemspan™ lymphoid expansion supplement (Cat # 9915; StemCell Technologies Inc, CA).

The HPCs may be cultured on a surface during differentiation into progenitor T cells. For example, the HPCs may be cultured on a surface of a culture vessel, bead or other biomaterial or polymer. Preferably, the surface may be coated with a factor that stimulates Notch signalling, for example a Notch ligand, such as Delta-like 1 (DLL1) or Delta-like 4 (DLL4). Suitable Notch ligands are well-known in the art and available from commercial suppliers.

The surface may also be coated with an extracellular matrix protein, such as fibronectin, vitronectin, laminin or collagen and/or one or more cell surface adhesion proteins, such as VCAM1 . In some embodiments, the surface for HPC culture may have a coating that comprises a factor that stimulates Notch signalling, for example a Notch ligand, such as DLL4, without the extracellular matrix protein or cell surface adhesion protein.

In some embodiments, the surface for HPC culture may have a coating that comprises a factor that stimulates Notch signalling, for example a Notch ligand, such as DLL4, an extracellular matrix protein, such as vitronectin, and a cell surface adhesion protein, such as VCAM1 . The surface may be coated with an extracellular matrix protein, factor that stimulates Notch signalling and cell surface adhesion protein by contacting the surface with a coating solution. For example, the coating solution may be incubated on the surface under suitable conditions to coat the surface. Conditions may, for example, include about 2 hours at room temperature. Coating solutions comprising an extracellular matrix protein and a factor that stimulates Notch signalling are available from commercial suppliers (StemSpan™ Lymphoid Differentiation Coating Material; Cat # 9925; Stem Cell Technologies Inc, CA) and may be supplemented with ICOS-L for use as described herein.

The HPCs may be cultured in the lymphoid expansion medium on the substrate for a time sufficient for the HPCs to differentiate into progenitor T cells. For example, the HPCs may be cultured for 2-6 weeks, 2 to 5 weeks or 2-4 weeks, preferably 3 weeks.

Progenitor T cells are multi-potent lymphopoietic progenitor cells that are capable of giving rise to ab T cells, gd T cells, tissue resident T cells and NK T cells. Progenitor T cells may commit to the ab T cell lineage after pre-TCR selection in the thymus. Progenitor T cells may be capable of in vivo thymus colonization and may be capable of committing to the ab T cell lineage after pre-TCR selection in the thymus. Progenitor T cells may also be capable of maturation into cytokine-producing CD3 + T-cells.

Progenitor T cells may express CD5 and CD7 i.e. the progenitor T cells may have a CD5+CD7+ phenotype. Progenitor T cells may also co-express CD44, CD25 and CD2. For example, progenitor T cells may have a CD5+, CD7+ CD44+, CD25+ CD2+ phenotype. Progenitor T cells may also co-express CD45. Progenitor T cells may lack expression, for example cell surface expression, of CD3, CD4 and CD8.

In a fifth stage, progenitor T cells may be matured into TCR ab+ T cells by culturing the population of progenitor T cells under suitable conditions to promote T cell maturation. For example, the progenitor T cells may be cultured in a T cell maturation medium. A T cell maturation medium is a cell culture medium that promotes the maturation of progenitor T cells into mature T cells. A suitable T cell maturation medium may (i) stimulate cKIT receptor (CD117; KIT receptor tyrosine kinase) and/or cKIT receptor (CD117; KIT receptor tyrosine kinase) mediated signalling pathways (ii) FLT3 and/or FLT3 mediated signalling pathways and (iii) have interleukin (IL) activity. For example, a T cell maturation medium may comprise the differentiation factors SCF, FLT3L, and IL7.

In preferred embodiments, the T cell maturation medium is a chemically defined medium. For example, the T cell maturation medium may consist of a chemically defined nutrient medium supplemented with effective amounts of the above differentiation factors. Suitable T cell maturation media are well-known in the art and include Stemspan™ SFEM II (Cat # 9605; StemCell Technologies Inc, CA) with Stemspan™ T cell maturation supplement (Cat # 9930; StemCell Technologies Inc, CA) and other media suitable for expansion of PBMCs and CD3+ cells, such as ExCellerate Human T cell expansion medium (R& D Systems, USA). Other suitable T cell maturation media may include a basal medium such as IMDM, supplemented with ITS, albumin and lipids, as described elsewhere herein and further supplemented with effective amounts of the above differentiation factors.

The progenitor T cells may be cultured on a surface. For example, the progenitor T cells may be cultured on a surface of a culture vessel, bead or other biomaterial or polymer.

Preferably, the surface may be coated with a factor that stimulates Notch signalling, for example a Notch ligand, such as Delta-like 1 (DLL1) or Delta-like 4 (DLL4). Suitable Notch ligands are well-known in the art and available from commercial suppliers. The surface may also be coated with an extracellular matrix protein, such as fibronectin, vitronectin, laminin or collagen and/or one or more cell surface adhesion proteins, such as VCAM1. Suitable coatings are well-known in the art and described elsewhere herein.

The progenitor T cells may be cultured in the T cell maturation medium on the substrate for a time sufficient for the progenitor T cells to mature into TCR ab+ T cells. For example, the progenitor T cells may be cultured for 1-4 weeks, preferably 2 or 3 weeks.

In some embodiments, the TCR ab+ T cells produced by maturation of progenitor T cells may be double positive CD4+CD8+ T cells.

In the methods of the invention, the HPCs are differentiated into progenitor T cells and/or the progenitor T cells matured into TCR ab+ T cells, in the presence of Inducible Co-stimulator ligand (ICOS-L). For example, the HPCs and/or progenitor T cells may be cultured in a culture medium comprising ICOS-L or on a surface coated with ICOS-L.

ICOS-L is a protein of the immunoglobulin superfamily that comprises an Ig-like C2-type (immunoglobulinlike) domain and an Ig-like V-type (immunoglobulin-like) domain. ICOS-L is a costimulatory signal forT-cell proliferation and cytokine secretion and induces B-cell proliferation and differentiation into plasma cells. ICOS-L is widely expressed in lymph nodes, leukocytes and spleen, and may also be detected on activated monocytes and dendritic cells. ICOS-L (Gene ID 23308; also called ICOSLG; B7-H2 or CD275) is preferably human ICOS-L and may have the amino acid sequence of database entry 075144, NP_001269979.1 , NP_001269980.1 , NP_001269981.1 , NP_001352688.1 , and NP_056074.1 ICOS-L may have the amino acid sequence of SEQ ID NO: 1. In some preferred embodiments, ICOS-L as described herein may comprise the extracellular domain of ICOS-L, for example residues 19 to 256 of SEQ ID NO: 1.

ICOS-L may be produced synthetically or recombinantly and is available from commercial suppliers (for example, Creative Biomart, Novoprotein, R&D Systems, Abnova; Stratech Scientific Limited; Sino Biological).

The surface may be coated with ICOS-L using a coating solution. For example the coating solution may be incubated on the surface under suitable conditions to coat the surface. Conditions may for example include about 2 hours at room temperature. The coating solution may for example comprise 0.5pg/ml to 50pg/ml, for example any of about 0.5, 1 , 2, 3, 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, 30, 35, 40, 45, or 50 pg/ml ICOS-L, or 1 pg/ml to 25 pg/ml ICOS-L, for example about 5pg/ml ICOS-L. Coating solutions suitable for supplementation with ICOS-L are described elsewhere herein.

The presence of ICOS-L is shown herein to increase the expression of T cell receptor (TCR) by CD4+ CD8+ T cells. TCRs are disulphide-linked membrane anchored heterodimeric proteins that comprise highly variable alpha (a) and beta (b) chains expressed as a complex with invariant CD3 chain molecules. T cells expressing this type of TCRs (ab TCRs) may be referred to as ab (or a:b) T cells.

TCRs bind specifically to major histocompatibility complexes (MHC) on the surface of cells that display a peptide fragment of a target antigen. For example, TCRs may bind specifically to a major histocompatibility complex (MHC) on the surface of cancer cells that displays a peptide fragment of a tumour antigen. Alternatively, TCRs may recognise specific antigen or peptide thereof independent of presentation by MHC.

T cells comprising such TCRs may be produced according to the methods of the present invention. An MHC is a set of cell-surface proteins which allow the acquired immune system to recognise ‘foreign’ molecules. Proteins are intracellularly degraded and presented on the surface of cells by the MHC. MHCs displaying ‘foreign’ peptides, such a viral or cancer associated peptides, are recognised by T cells with the appropriate TCRs, prompting cell destruction pathways. MHCs on the surface of cancer cells may display peptide fragments of tumour antigen i.e. an antigen which is present on a cancer cell but not the corresponding non- cancerous cell. T cells which recognise these peptide fragments may exert a CD4+ CD8+ effect on the cancer cell.

Progenitor T cells may be matured into TCR ab+ T cells by the methods described above. T cells (also called T lymphocytes) are white blood cells that play a central role in cell-mediated immunity. T cells can be distinguished from other lymphocytes by the presence of a T cell receptor (TCR) on the cell surface. There are several types of T cells, each type having a distinct function. T helper cells (TH cells) are known as CD4 + T cells because they express the CD4 surface glycoprotein.

CD4 + T cells play an important role in the adaptive immune system and help the activity of other immune cells by releasing T cell cytokines and helping to suppress or regulate immune responses. They are essential for the activation and growth of CD4+ CD8+ T cells. CD4+ CD8+ T cells (Tc cells, CTLs, killer T cells, CD4+ CD8+ T cells) are known as CD8 + T cells because they express the CD8 surface glycoprotein. CD8 + T cells act to destroy virus-infected cells and tumour cells. Most CD8 + T cells express TCRs that can recognise a specific antigen displayed on the surface of infected or damaged cells by a class I MHC molecule. Specific binding of the TCR and CD8 glycoprotein to the antigen and MHC molecule leads to T cell-mediated destruction of the infected or damaged cells.

TCR ab+ T cells produced as described herein may be double positive CD4+CD8+ T cells or single positive CD4+ or CD8+ T cells.

TCR ab+ T cells produced as described herein may be mature CD3+ T cells. For example, the cells may have a c^TCR+ CD3+ CD45+ CD28+ phenotype.

A population of T cells produced using ICOS-L as described herein may have an increased proportion of cells that express c^TCR relative to populations produced in the absence of ICOS-L. For example, the proportion of c^TCR+ T cells in a population produced using ICOS-L may be at least 10%, at least 20% or at least 30% higher than the proportion of c^TCR+ T cells in a population produced without ICOS-L.

Following maturation of progenitor T cells (stage 5), the population of T cells may be predominantly double positive CD4+CD8+ T cells.

In a sixth stage, the population of TCR ab+ T cells may be activated and/or expanded to produce or increase the proportion of single positive CD4+ T cells, or more preferably single positive CD8+ T cells. Suitable methods for activating and expanding T cells are well-known in the art. For example, T cells may be exposed to a T cell receptor (TCR) agonist under appropriate culture conditions. Suitable TCR agonists include ligands, such as peptides displayed on a class I or II MHC molecules (MHC-peptide complexes) on the surface of a bead or an antigen presenting cell, such as a dendritic cell, and soluble factors, such as anti- TCR antibodies, for example anti-CD28 antibodies, and multimeric MHC-peptide complexes, such as MHC- peptide tetramers, pentamers or dextramers.

Activation refers to the state of a T cell that has been sufficiently stimulated to induce detectable cellular proliferation. Activation can also be associated with induced cytokine production, and detectable effector functions. The term "activated T cells" refers to, among other things, T cells that are undergoing cell division.

An anti-TCR antibody may specifically bind to a component of the TCR, such as sCD3, aCD3 or aCD28. Anti-TCR antibodies suitable for TCR stimulation are well-known in the art (e.g. OKT3) and available from commercial suppliers (e.g. eBioscience CO USA). In some embodiments, T cells may be activated by exposure to anti-aCD3 antibodies and IL2, IL-7 or IL15. More preferably, T cells are activated by exposure to anti-aCD3 antibodies and anti-aCD28 antibodies. The activation may occur in the presence or absence of CD14 + monocytes. The T cells may be activated with anti-CD3 and anti-CD28 antibody coated beads. For example, PBMCs or T cell subsets including CD4 + and/or CD8 + cells may be activated, without feeder cells (antigen presenting cells) or antigen, using antibody coated beads, for example magnetic beads coated with anti-CD3 and anti-CD28 antibodies, such as Dynabeads® Human T-Activator CD3/CD28 (ThermoFisher Scientific). In other embodiments, soluble tetrameric antibody complexes that bind CD3, CD28 and CD2 cell surface ligands, such as ImmunoCult™ Human CD3/CD28/CD2 T Cell Activator or Human CD3/CD28 T Cell Activator, may be used to activate the T cells. . In other embodiments, T cells may be activated with an MHC-peptide complex, preferably a multimeric MHC-peptide complex, optionally in combination with an anti- CD28 antibody.

In some embodiments, TCR ab+ T cells, for example double positive CD4+CD8+ T cells, may be cultured in a T cell maturation medium as described herein supplemented with IL-15. The medium may be further supplemented with a T cell receptor (TCR) agonist, for example one or more anti-TCR antibodies, such as anti-aCD3 antibodies, and anti-aCD28 antibodies, as described above.

The TCR ab+ T cells may be cultured using any convenient technique to produce the expanded population. Suitable culture systems include stirred tank fermenters, airlift fermenters, roller bottles, culture bags or dishes, and other bioreactors, in particular hollow fibre bioreactors. The use of such systems is well-known in the art

TCR ab+ T cells produced as described herein may express an ab TCR that binds a target antigen. For example, the ab TCR may bind specifically to cancer cells that express a tumor antigen. The T cells may be useful for example in immunotherapy, as described below.

In some embodiments, the ab TCR expressed by the T cells may be naturally expressed (i.e. an endogenous TCR). For example, the T cells may be produced as described herein from iPSCs that are derived from Tumour Infiltrating Lymphocytes (TILs). TILs, for example tumour resident CD3+ CD8+ cells, may be obtained from an individual with a cancer condition using standard techniques. Alternatively, the T cells may be produced as described herein from iPSCs that are derived from T cells that bind to a peptide fragment of the target antigen displayed on a class I or II MHC molecule on the surface of an antigen presenting cell, such as a dendritic cell; or a population of T cells produced as described herein may be screened for binding to a peptide fragment of the target antigen displayed on a class I or II MHC molecule, and T cells that bind to the displayed peptide fragment identified.

In other embodiments, the ab TCR is not naturally expressed by the cells (i.e. the TCR is exogenous or heterologous). Suitable heterologous ab TCR may bind specifically to class I or II MHC molecules displaying peptide fragments of a target antigen. For example, the T cells may be modified to express a heterologous ab TCR that binds specifically to class I or II MHC molecules displaying peptide fragments of a tumour antigen expressed by the cancer cells in a cancer patient. In some embodiments the TCR may recognise a target antigen or a peptide fragment of a target antigen on the cancer cell independently of MHC presentation. Tumour antigens expressed by cancer cells in the cancer patient may identified using standard techniques. Preferred tumour antigens include NY-ES01 , PRAME, alpha-fetoprotein (AFP), MAGE A4, MAGE A1 , MAGE A10 and MAGE B2, most preferably NY-ESO-1 , MAGE-A4 and MAGE-A10.

A heterologous TCR may be a synthetic or artificial TCR i.e. a TCR that does not exist in nature. For example, a heterologous TCR may be engineered to increase its affinity or avidity for a tumour antigen (i.e. an affinity enhanced TCR). The affinity enhanced TCR may comprise one or more mutations relative to a naturally occurring TCR, for example, one or more mutations in the hypervariable complementarity determining regions (CDRs) of the variable regions of the TCR a and b chains. These mutations increase the affinity of the TCR for MHCs that display a peptide fragment of a tumour antigen expressed by cancer cells. Suitable methods of generated affinity enhanced TCRs include screening libraries of TCR mutants using phage or yeast display and are well known in the art (see for example Robbins et al J Immunol (2008)

180(9):6116; San Miguel et al (2015) Cancer Cell 28 (3) 281-283; Schmitt et al (2013) Blood 122348-256; Jiang et al (2015) Cancer Discovery 5 901). Preferred affinity enhanced TCRs may bind to cancer cells expressing one or more of the tumour antigens NY-ES01 , PRAME, alpha-fetoprotein (AFP), MAGE A4, MAGE A1 , MAGE A10 and MAGE B2.

Expression of a heterologous ab TCR may alter the immunogenic specificity of the T cells produced as described herein so that they recognise or display improved recognition for one or more target antigens, e.g. tumour antigens that are present on the surface of the cancer cells of an individual with cancer. In some embodiments, the T cells produced as described herein may display reduced binding or no binding to cancer cells in the absence of the heterologous ab TCR. For example, expression of the heterologous ab TCR may increase the affinity and/or specificity of the cancer cell binding of a T cell relative to T cells that do not express the ab TCR.

The term "heterologous" refers to a polypeptide or nucleic acid that is foreign to a particular biological system, such as a host cell, and is not naturally present in that system. A heterologous polypeptide or nucleic acid may be introduced to a biological system by artificial means, for example using recombinant techniques. For example, heterologous nucleic acid encoding a polypeptide may be inserted into a suitable expression construct which is in turn used to transform a host cell to produce the polypeptide. A heterologous polypeptide or nucleic acid may be synthetic or artificial or may exist in a different biological system, such as a different species or cell type. An endogenous polypeptide or nucleic acid is native to a particular biological system, such as a host cell, and is naturally present in that system. A recombinant polypeptide is expressed from heterologous nucleic acid that has been introduced into a cell by artificial means, for example using recombinant techniques. A recombinant polypeptide may be identical to a polypeptide that is naturally present in the cell or may be different from the polypeptides that are naturally present in that cell.

T cells may be modified to express the heterologous ab TCR by the introduction of heterologous encoding nucleic acid into cells at any stage in the method described herein. For example, heterologous encoding nucleic acid may be introduced into iPSCs, HPCs or progenitor T cells. In some preferred embodiments, cells may be transduced with heterologous nucleic acid encoding an ab TCR after culture in lymphoid expansion medium, for example after 2 weeks culture in lymphoid expansion medium (stage 4) as described herein. Heterologous nucleic acid encoding a TCR may encode all the sub-units of the receptor. For example, nucleic acid encoding a TCR may comprise a nucleotide sequence encoding a TCR a chain and a nucleotide sequence encoding a TCR b chain.

Nucleic acid may be introduced into the cells by any convenient technique. When introducing or incorporating a heterologous nucleic acid into an iPSC, HPC or progenitor T cell, certain considerations must be taken into account, well-known to those skilled in the art. The nucleic acid to be inserted should be assembled within a construct or vector which contains effective regulatory elements which will drive transcription in the T cell. Many known techniques and protocols for manipulation and transformation of nucleic acid, for example in preparation of nucleic acid constructs, introduction of DNA into cells and gene expression are described in detail in Protocols in Molecular Biology, Second Edition, Ausubel et al. eds. John Wiley & Sons, 1992. In some embodiments, nucleic acid may be introduced into the cells by gene editing.

For example, a DNA double strand break (DSB) at a target site may be induced by a CRISPR/Cas9 system and the repair of the DSB may introduce the heterologous nucleic acid into the cell genome at the target site or the nucleic acid may be introducing using an rAAV vector (AAV mediated gene editing; Hirsch et al 2014 Methods Mol Biol 1114291-307).

Suitable techniques for introducing the expression vector into the iPSCs, HPCs or progenitor T cells are well known in the art and include calcium phosphate transfection, DEAE-Dextran, electroporation, liposome- mediated transfection, gene editing and transduction using retrovirus or other virus, e.g. vaccinia or lentivirus. Preferably, nucleic acid encoding the heterologous ab TCR may be contained in a viral vector, most preferably a gamma retroviral vector or a lentiviral vector, such as a VSVg-pseudotyped lentiviral vector. A method described herein may comprise transducing a population of cells, for example iPSCs,

HPCs or progenitor T cells, with a viral vector to produce a transduced population of genetically modified cells. The cells may be transduced by contact with a viral particle comprising the nucleic acid. Viral particles for transduction may be produced according to known methods. For example, HEK293T cells may be transfected with plasmids encoding viral packaging and envelope elements as well as a lentiviral vector comprising the coding nucleic acid. A VSVg-pseudotyped viral vector may be produced in combination with the viral envelope glycoprotein G of the Vesicular stomatitis virus (VSVg) to produce a pseudotyped virus particle. For example, solid-phase transduction may be performed without selection by culture on retronectin- coated, retroviral vector-preloaded tissue culture plates.

Following production, the population of TCR ab+ T cells, for example DP CD4+CD8+ cells, SP CD4+ cells or SP CD8+ cells, may be isolated and/or purified. Any convenient technique may be used, including fluorescence-activated cell sorting (FACS) or magnetic-activated cell sorting using antibody coated magnetic particles (MACS). The population of TCR ap+ T cells, for example DP CD4+CD8+ cells, SP CD4+ cells or SP CD8+ cells, may be expanded and/or concentrated. Optionally, the population of TCR ab+ T cells produced as described herein may be stored, for example by cryopreservation, before use.

A population of TCR ab+ T cells may be admixed with other reagents, such as buffers, carriers, diluents, preservatives and/or pharmaceutically acceptable excipients. Suitable reagents are described in more detail below. A method described herein may comprise admixing the population of TCR ab+ T cells with a pharmaceutically acceptable excipient.

Pharmaceutical compositions suitable for administration (e.g. by infusion), include aqueous and non- aqueous isotonic, pyrogen-free, sterile injection solutions which may contain anti-oxidants, buffers, preservatives, stabilisers, bacteriostats, and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. Examples of suitable isotonic vehicles for use in such formulations include Sodium Chloride Injection, Ringer’s Solution, or Lactated Ringer’s Injection. Suitable vehicles can be found in standard pharmaceutical texts, for example, Remington’s Pharmaceutical Sciences, 18th edition, Mack Publishing Company, Easton, Pa., 1990.

In some preferred embodiments, the TCR ab+ T cells, which may be DP CD4+CD8+ T cells, SP CD4+ T cells or preferably SP CD8+ T cells, may be formulated into a pharmaceutical composition suitable for intravenous infusion into an individual.

The term “pharmaceutically acceptable” as used herein pertains to compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgement, suitable for use in contact with the tissues of a subject (e.g., human) without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. Each carrier, excipient, etc. must also be “acceptable” in the sense of being compatible with the other ingredients of the formulation.

An aspect of the invention provides a population of TCR ab+ T cells, which may be for example DP CD4+CD8+ T cells, SP CD4+ T cells or SP CD8+ T cells, produced by a method described above.

The population of TCR ab+ T cells may be for use as a medicament. For example, a population of mature TCR ab+ T cells as described herein may be used in cancer immunotherapy therapy, for example adoptive T cell therapy.

Adoptive cellular therapy or adoptive immunotherapy refers to the adoptive transfer of human T lymphocytes that express TCRs that are specific for antigens or peptides thereof expressed on target cells and/or TCRs that are specific for peptide MHC complexes expressed on target cells.

This can be used to treat a range of diseases depending upon the target chosen, e.g., tumour specific antigens to treat cancer. Adoptive cellular therapy involves removing a portion of a donor’s or the patient’s cells, for example, white blood cells. The cells are then used to generate iPSCs in vitro and these iPSCs are used to efficiently generate T cells that are specific for antigens or peptides thereof expressed on target cells and/or specific for peptide MHC complexes on target cells as described herein. The T cells may be expanded, washed, concentrated, and/or then frozen to allow time for testing, shipping and storage until a patient is ready to receive the infusion of cells.

Other aspects of the invention provide the use of a population of TCR ab+ T cells as described herein for the manufacture of a medicament for the treatment of cancer, a population of TCR ab+ T cells as described herein for the treatment of cancer, and a method of treatment of cancer comprising administering a population of TCR ab+ T cells as described herein to an individual in need thereof.

The population of TCR ab+ T cells may be autologous i.e. the TCR ab+ T cells were originally obtained from the same individual to whom they are subsequently administered (i.e. the donor and recipient individual are the same).

The population of TCR ab+T cells may be allogeneic i.e. the TCR ab+ T cells may be originally obtained from a different individual to the individual to whom they are subsequently administered (i.e. the donor and recipient individual are different). Allogeneic refers to a graft derived from a different animal of the same species.

The donor and recipient individuals may be HLA matched to avoid GVHD and other undesirable immune effects, such as rejection. Alternatively, the donor and recipient individuals may not be HLA matched, or HLA genes in the cells from the donor individual may be modified, for example by gene editing, to remove any HLA mismatch with the recipient.

A suitable population of TCR ab+ T cells for administration to a recipient individual may be produced by a method comprising providing an initial population of cells, preferably T cells, obtained from a donor individual, reprogramming the cells into iPSCs and differentiating the iPSCs into T cells that express an ab TCR which binds specifically to cancer cells and/or an antigen or peptide thereof presented by cancer cells optionally in complex with MHC, in the recipient individual.

Following administration of the TCR ab+ T cells, the recipient individual may exhibit a T cell mediated immune response against cancer cells in the recipient individual. This may have a beneficial effect on the cancer condition in the individual.

As used herein, the terms "cancer," "neoplasm," and "tumour" are used interchangeably and, in either the singular or plural form, refer to cells that have undergone a malignant transformation that makes them pathological to the host organism.

Primary cancer cells can be readily distinguished from non-cancerous cells by well-established techniques, particularly histological examination. The definition of a cancer cell, as used herein, includes not only a primary cancer cell, but any cell derived from a cancer cell ancestor. This includes metastasized cancer cells, and in vitro cultures and cell lines derived from cancer cells. When referring to a type of cancer that normally manifests as a solid tumour, a "clinically detectable" tumour is one that is detectable on the basis of tumour mass; e.g., by procedures such as computed tomography (CT) scan, magnetic resonance imaging (MRI), X-ray, ultrasound or palpation on physical examination, and/or which is detectable because of the expression of one or more cancer-specific antigens in a sample obtainable from a patient.

Cancer conditions may be characterised by the abnormal proliferation of malignant cancer cells and may include leukaemias, such as AML, CML, ALL and CLL, lymphomas, such as Hodgkin lymphoma, non- Hodgkin lymphoma and multiple myeloma, and solid cancers such as sarcomas, skin cancer, melanoma, bladder cancer, brain cancer, breast cancer, uterus cancer, ovary cancer, prostate cancer, lung cancer, colorectal cancer, cervical cancer, liver cancer, head and neck cancer, oesophageal cancer, pancreas cancer, renal cancer, adrenal cancer, stomach cancer, testicular cancer, cancer of the gall bladder and biliary tracts, thyroid cancer, thymus cancer, cancer of bone, and cerebral cancer, as well as cancer of unknown primary (CUP).

Cancer cells within an individual may be immunologically distinct from normal somatic cells in the individual (i.e. the cancerous tumour may be immunogenic). For example, the cancer cells may be capable of eliciting a systemic immune response in the individual against one or more antigens expressed by the cancer cells. The tumour antigens that elicit the immune response may be specific to cancer cells or may be shared by one or more normal cells in the individual.

The cancer cells of an individual suitable for treatment as described herein may express the antigen and/or may be of correct HLA type to bind the ab TCR expressed by the T cells.

An individual suitable for treatment as described above may be a mammal. In preferred embodiments, the individual is a human. In other preferred embodiments, non-human mammals, especially mammals that are conventionally used as models for demonstrating therapeutic efficacy in humans (e.g. murine, primate, porcine, canine, or rabbit animals) may be employed.

In some embodiments, the individual may have minimal residual disease (MRD) after an initial cancer treatment.

An individual with cancer may display at least one identifiable sign, symptom, or laboratory finding that is sufficient to make a diagnosis of cancer in accordance with clinical standards known in the art. Examples of such clinical standards can be found in textbooks of medicine such as Harrison’s Principles of Internal Medicine, 15th Ed., Fauci AS et al., eds., McGraw-Hill, New York, 2001 . In some instances, a diagnosis of a cancer in an individual may include identification of a particular cell type (e.g. a cancer cell) in a sample of a body fluid or tissue obtained from the individual. An anti-tumour effect is a biological effect which can be manifested by a reduction in the rate of tumour growth, decrease in tumour volume, a decrease in the number of tumour cells, a decrease in the number of metastases, an increase in life expectancy, or amelioration of various physiological symptoms associated with the cancerous condition. An "anti-tumour effect" can also be manifested by the ability of the peptides, polynucleotides, cells, particularly T cells produced according to the methods of the present invention, and antibodies described herein in prevention of the occurrence of tumour in the first place

Treatment may be any treatment and/or therapy, whether of a human or an animal (e.g. in veterinary applications), in which some desired therapeutic effect is achieved, for example, the inhibition or delay of the progress of the condition, and includes a reduction in the rate of progress, a halt in the rate of progress, amelioration of the condition, cure or remission (whether partial or total) of the condition, preventing, delaying, abating or arresting one or more symptoms and/or signs of the condition or prolonging survival of a subject or patient beyond that expected in the absence of treatment.

Treatment may also be prophylactic (i.e. prophylaxis). For example, an individual susceptible to or at risk of the occurrence or re-occurrence of cancer may be treated as described herein. Such treatment may prevent or delay the occurrence or re-occurrence of cancer in the individual.

In particular, treatment may include inhibiting cancer growth, including complete cancer remission, and/or inhibiting cancer metastasis. Cancer growth generally refers to any one of a number of indices that indicate change within the cancer to a more developed form. Thus, indices for measuring an inhibition of cancer growth include a decrease in cancer cell survival, a decrease in tumour volume or morphology (for example, as determined using computed tomographic (CT), sonography, or other imaging method), a delayed tumour growth, a destruction of tumour vasculature, improved performance in delayed hypersensitivity skin test, an increase in the activity of T cells, and a decrease in levels of tumour-specific antigens. Administration of T cells modified as described herein may improve the capacity of the individual to resist cancer growth, in particular growth of a cancer already present the subject and/or decrease the propensity for cancer growth in the individual.

The TCR ab+ T cells or the pharmaceutical composition comprising the TCR ab+ T cells may be administered to a subject by any convenient route of administration, whether systemically/ peripherally or at the site of desired action, including but not limited to; parenteral, for example, by infusion. Infusion involves the administration of the T cells in a suitable composition through a needle or catheter. Typically, T cells are infused intravenously or subcutaneously, although the T cells may be infused via other non-oral routes, such as intramuscular injections and epidural routes. Suitable infusion techniques are known in the art and commonly used in therapy (see, e.g., Rosenberg et al., New Eng. J. of Med., 319:1676, 1988).

Typically, the number of cells administered is from about 10 5 to about 10 10 per Kg body weight, for example any of about 1 , 2, 3, 4, 5, 6, 7, 8, or 9, x 10 5 , x 10 6 , x 10 7 , x 10 8 , x 10 9 , or x 10 10 cells per individual, typically 2x10 8 to 2x10 10 cells per individual, typically over the course of 30 minutes, with treatment repeated as necessary, for example at intervals of days to weeks. It will be appreciated that appropriate dosages of the TCR ab+ T cells, and compositions comprising the TCR ab+ T cells, can vary from patient to patient. Determining the optimal dosage will generally involve the balancing of the level of therapeutic benefit against any risk or deleterious side effects of the treatments of the present invention. The selected dosage level will depend on a variety of factors including, but not limited to, the activity of the particular cells, cytokine release syndrome (CRS), the route of administration, the time of administration, the rate of loss or inactivation of the cells, the duration of the treatment, other drugs, compounds, and/or materials used in combination, and the age, sex, weight, condition, general health, and prior medical history of the patient. The amount of cells and the route of administration will ultimately be at the discretion of the physician, although generally the dosage will be to achieve local concentrations at the site of action which achieve the desired effect without causing substantial harmful or deleterious side-effects.

While the TCR ab+ T cells may be administered alone, in some circumstances the TCR ab+ T cells may be administered cells in combination with the target antigen, APCs displaying the target antigen, CD3/CD28 beads, IL-2, IL7 and/or IL15 to promote expansion in vivo of the population of TCR ab+ T cells.

Administration in combination may be by separate, simultaneous or sequential administration of the combined components.

The population of TCR ab+ T cells may be administered in combination with one or more other therapies, such as cytokines e.g. IL-2, CD4+ CD8+ chemotherapy, radiation and immuno-oncology agents, including checkpoint inhibitors, such as anti-B7-H3, anti-B7-H4, anti-TIM3, anti-KIR, anti-LAG3, anti-PD-1 , anti-PD-L1 , and anti-CTLA4 antibodies. Administration in combination may be by separate, simultaneous or sequential administration of the combined components.

The one or more other therapies may be administered by any convenient means, preferably at a site which is separate from the site of administration of the TCR ab+ T cells.

Administration of TCR ab+ T cells can be effected in one dose, continuously or intermittently (e.g., in divided doses at appropriate intervals) throughout the course of treatment. Methods of determining the most effective means and dosage of administration are well known to those of skill in the art and will vary with the formulation used for therapy, the purpose of the therapy, the target cell being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician. Preferably, the TCR ab+ T cells are administered in a single transfusion, for example of any of 500 million, 1 billion, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15 billion T cells for example at least 1 x 10 9 T cells.

Other aspects of the invention provide a coating solution for treating the surface of a T cell culture vessel, the solution comprising ICOS-L

The coating solution may further comprise a factor that stimulates Notch signalling, for example a Notch ligand, such as Delta-like 1 (DLL1) or Delta-like 4 (DLL4). The coating solution may further comprise an extracellular matrix protein, such as fibronectin, vitronectin, laminin or collagen and/or a cell surface adhesion protein, such as VCAM1 .

Another aspect of the invention provides a culture vessel for T cell culture comprising a surface coated with ICOS-L

In addition to ICOS-L, the coating of the surface of the culture vessel may further comprise a factor that stimulates Notch signalling, for example a Notch ligand, such as Delta-like 1 (DLL1) or Delta-like 4 (DLL4). The coating may further comprise an extracellular matrix protein, such as fibronectin, vitronectin laminin or collagen and/or a cell surface adhesion protein, such as VCAM1 .

Suitable cell culture vessels are well known in the art and include culture plates, dishes, flasks, bioreactors, and multiwell plates, for example 6-well, 12-well or 96-well plates.

The culture vessel may further comprise a culture medium, for example a lymphoid expansion medium or a T cell maturation medium, as described above. In some embodiments, immune cells, such as haematopoietic progenitor cells, progenitor T cells or T cells may be present in the culture medium on the surface of the vessel.

Suitable coating solutions and culture vessels may be used in a method of producing T cells described above.

Other aspects and embodiments of the invention provide the aspects and embodiments described above with the term “comprising” replaced by the term “consisting of and the aspects and embodiments described above with the term “comprising” replaced by the term ’’consisting essentially of.

It is to be understood that the application discloses all combinations of any of the above aspects and embodiments described above with each other, unless the context demands otherwise. Similarly, the application discloses all combinations of the preferred and/or optional features either singly or together with any of the other aspects, unless the context demands otherwise.

Modifications of the above embodiments, further embodiments and modifications thereof will be apparent to the skilled person on reading this disclosure, and as such, these are within the scope of the present invention.

Unless defined otherwise, all 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. Although any compositions and methods similar or equivalent to those described herein can be used in the practice or testing of the methods of the disclosure, exemplary compositions and methods are described herein. Any of the aspects and embodiments of the disclosure described herein may also be combined. For example, the subject matter of any dependent or independent claim disclosed herein may be multiply combined ( e.g ., one or more recitations from each dependent claim may be combined into a single claim based on the independent claim on which they depend).

Ranges provided herein include all values within a particular range described and values about an endpoint for a particular range. The figures and tables of the disclosure also describe ranges, and discrete values, which may constitute an element of any of the methods disclosed herein. Concentrations described herein are determined at ambient temperature and pressure. This may be, for example, the temperature and pressure at room temperature or in within a particular portion of a process stream. Preferably, concentrations are determined at a standard state of 25 °C and 1 bar of pressure. The term “about” means a value within two standard deviations of the mean for any particular measured value.

As used herein and in the claims, the singular forms “a,” “and,” and “the” include plural reference unless the context clearly dictates otherwise. Thus, for example, reference to “a peptide chain” is a reference to one or more peptide chains and includes equivalents thereof known to those skilled in the art.

All documents and sequence database entries mentioned in this specification are incorporated herein by reference in their entirety for all purposes.

“and/or” where used herein is to be taken as specific disclosure of each of the two specified features or components with or without the other. For example “A and/or B” is to be taken as specific disclosure of each of (i) A, (ii) B and (iii) A and B, just as if each is set out individually herein.

Experimental

Methods hiPSC Culture iPSCs were routinely cultured in mTeSRI (SCT) on Matrigel (BD Corning) using tissue culture plasticware, in 5% CO2, 5% C>2at 37C. hiPSCs were harvested manually using an EasyPassage tool (Invitrogen) and cells seeded at 1 :6 or 1 :12 ratios, in media with Y27632 (R&D Systems) at 10uM for the first 48h of culture. For differentiation, hiPSCs were passaged onto either matrigel or vitronectin at low density cultures using 1 :48 or 1 :98 split ratio. Seeding density was about 1 colony per field of view, when viewed under x4 magnification on the microscope, at 24h post seeding. hiPSCs were cultured in mTeSR2 or E8 flex (SCT), depending on the cell culture matrix used, for approx. 4-5 days until colonies are compacted and distinct cells are no longer visible.

HiPSC maintenance medium (mTeSR2 or E8 flex) was removed the cells washed twice with DMEM/F12.

2 mL of StemPro34 PLUS (StemPro34 from Invitrogen; StemPro34 basal media, with supplement added and Penicillin Streptomycin (1% v/v: Invitrogen) and Glutamine (2mM: Invitrogen) , Ascorbic Acid (5C^g/ml:

Sigma Aldrich) and monothioglycerol (100 mM: Sigma Aldrich), further supplemented with 50 ng/mL of Activin was added and incubated for 4 hours. Volumes are dependent of culture flask size, typically at least 2mls/ 9cm 2 , and 20mls /150cm 2 . After 4 hours, the medium was removed and the cells washed twice with DMEM/F12 to remove residual high concentration Activin A. The medium was replaced with 2 ml_ of StemPro34 PLUS supplemented with 5 ng/mL of Activin A, 10 ng/ml of BMP4 and 5 ng/ml of bFGF and incubated for 44 hours (Stage 1 media). The medium was then replaced with fresh Stage 1 media and supplemented with 10 pM CHIR-99021 and further cultured for 48 hours.

On Day 4, the medium was removed and the cells washed twice with DMEM/F12 to remove residual stage 1 cytokines. The medium was then replaced with StemPro34 PLUS supplemented with 100 ng/mL of SCF and 15 ng/ml of VEGF and incubated for 48 hours (Stage 2 media). The medium was then replenished with fresh Stage 2 media and the cells cultured for a further 48 hours.

The medium was then replaced by the Stage 3 medium shown in Table 1 and the cells cultured for between 16-18 days, with 1 :1 (v/v) feeding every 48h. Typically this involved harvest of media and collection of cells in suspension by centrifugation (at 300g, 10 min), and returning suspension cells to culture with fresh media (i.e. 20mls for a T150 flask).

On approx d 16-18 depending on hiPSC line used, (confirmed separately by flow cytometry prior to day of harvest) we isolate CD34+ cells from resulting monolayers for onward culture. Here we routinely include hiPSCs lines designated ChiPSC31 (Takara), NIH2 (WT: a sub clone of MR1.1 from Lonza)) and sub-clones of NIH2: c3F3 and c1A12. CD34+ cells were harvested by sequential incubation with Accutase (SCT: for 30 mins at 37C) and then Collagenase II (Invitrogen: 2mg/ml) for 30 mins at 37C. Cell suspensions were collected and washed (x2 centrifugation at 300g for 12 min in DMEM/F12), prior to CD34+ cell isolation via Magnetic activated beads (MACS) isolation (Miltenyi: according to manufacturer’s instructions).

Following MACS isolation of CD34+ cells, these were then cryopreserved at 2x10 5 cells / vial in CS10 (SCT), firstly at -80C at slow rate freezing, then in liquid nitrogen for long term storage.

For continued lymphoid proliferation and differentiation, Stem Cell Technologies proprietary 2 stage (Lymphoid Proliferation U cell Maturation) media was employed (according to manufacturer’s instructions) as outlined below, together with iCOS ligand.

ICOS-L stimulation at Staae 4 hiPSC derived CD34+ haematopoietic progenitors were isolated after a 16 day differentiation protocol, described in methods. CD34+ haematopoietic progenitors were isolated by MACS separation and stored in liquid nitrogen in CS10 (Stem Cell Technologies). Cryo preserved hiPSC derived CD34+ cells were then thawed, washed in basal media, and cultured in StemCell technologies (SCT) T-cell generation kit for 21 days in Lymphoid Proliferation media (LP media). Cells were seeded at 10c10 L 4 cells per well in 1 ml of LP media either on the standard SCT’s coating or the SCT’S coating + ICOS-L (5pg/ml) into 24 well plates (DD166-A16 from NIH2 WT, C3F3 and C1A12 clones). Or cells were seeded at 5x10 L 4 cells per well in 1 ml of LP media either on the standard SCT’s coating or the SCT’S coating + ICOS-L (0.5, 5, 50pg/ml) into 24 well plates (DD166-C16 and DD166-D16 from NIH2 WT, C3F3 and C1A12 clones).

ICOS-L was added to the coating matrix for two hours prior to adding the cells. Cells were fed with fresh media every 3-4 days. After 2 weeks, cells were harvested, counted and seeded at 0.5x10 L 6 cells per well in 1 ml of LP media either on the standard SCT’s coating or the SCT’S coating + ICOS-L (5pg/ml) into 24 well plate (DD166-A16). Or cells were harvested, counted and seeded at 1x10 L 5 cells per well in 0.2ml of LP media either on the standard SCT’s coating or the SCT’S coating + ICOS-L (0.5, 5, 50pg/ml) into 96 well plate (DD166-C16 and DD166-D16). Cells were cultured for a further week in LP media with feeding every 3- 4 days and phenotyped after one week.

ICOS-L stimulation at Stape 5 hiPSC derived CD34 cells (from ChiPSC31 DD143, DD149 and DD157-E17) were defrosted as before, and cultured in StemCell technologies (SCT) T-cell generation kit for 21 to 25 days in LP media.

The cells were then subsequently seeded at 5x10 L 6 cells or 1x10 L 6 cells per well in 1ml of T Cell Maturation media (TM media) either on the standard SCT’s coating or the SCT’S coating + ICOS-L (0.5, 5, 50pg/ml) into 24 well plate. ICOS-L was added to the coating matrix for two hours prior to adding the cells. Cells were cultured for a further 2 weeks in TM media with feeding every 3-4 days and phenotyped after two weeks.

Cell phenotypinp

On the day of the assay, the cells were counted using a Cellometer (Nexcelon Biosciences, LLC) and automated analysis, washed twice in FACS Buffer and phenotyped by flow cytometry (BD Fortessa) after staining using relevant T-cell specific surface markers as identified in the figures. The list of antibody clones and fluorochrome used with their supplier, can be found in Table 2.

Results

The addition of ICOS-L at 0.5ug/ml to the SCT coating matrix during stage 4 does not significantly enhance the proportion of TCRap+ cells within CD45+ CD3+, however the addition of ICOS-L at 5 and 50ug/ml to the coating matrix during stage 4 significantly increases the proportion of TCRap+ cells within CD45+ CD3+.

This is observed across 3 separate cell lines: WT, c3F3 and c1A12 (Figure 1) and also shown as mean data (Figure 2 where n=3) where ** is P value of 0.01 and *** is a P value 0.001.

The addition of ICOSL at 5ug/ml to the coating matrix during stage 5 increases the proportion of TCRap+ cells within CD45+ CD8+ CD4+ and significantly increases the proportion of TCRap+ cells within CD45+ CD8+ CD4- T cells (Figure 3). Sequences

MRLGSPGLLF LLFSSLRADT QEKEVRAMVG SDVELSCACP EGSRFDLNDV YVYWQTSESK TWTYHIPQN SSLENVDSRY RNRALMSPAG MLRGDFSLRL FNVTPQDEQK FHCLVLSQSL GFQEVLSVEV TLHVAANFSV PVVSAPHSPS QDELTFTCTS INGYPRPNVY WINKTDNSLL DQALQNDTVF LNMRGLYDW SVLRIARTPS VNIGCCIENV LLQQNLTVGS QTGNDIGERD KITENPVSTG EKNAATWSIL AVLCLLVWA VAIGWVCRDR CLQHSYAGAW AVSPETELTG HV SEQ ID NO: 1 (ICOS-L; signal sequence dotted underline; transmembrane domain underline)

Table 1

Table 2: Antibodies used to phenotype T cells.