Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
OPTIMIZED LENTIVIRAL VECTOR COMPROMISING MINIMAL ENHANCER ELEMENTS FOR STEM CELL GENE THERAPY OF HEMOGLOBINOPATHIES
Document Type and Number:
WIPO Patent Application WO/2020/168004
Kind Code:
A9
Abstract:
In certain embodiments a lentiviral vector having optimized reduced size LCR with improved enhancer activity is provided. In certain embodiments direct treatment of a subject by direct introduction of the vector(s) described herein is contemplated. The lentiviral compositions may be formulated for delivery by any available route including, but not limited to parenteral (e.g., intravenous), intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), transmucosal, rectal, and vaginal.

Inventors:
KOHN DONALD B (US)
HOLLIS ROGER PAUL (US)
MORGAN RICHARD A (US)
COOPER AARON ROSS (US)
Application Number:
PCT/US2020/017998
Publication Date:
July 15, 2021
Filing Date:
February 12, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV CALIFORNIA (US)
International Classes:
C07K14/805; C12N7/01; C12N15/09; C12N15/11; C12N15/67; C12N15/86
Attorney, Agent or Firm:
HUNTER, Tom et al. (US)
Download PDF:
Claims:
CLAIMS

What Is claimed Is:

1. A recombinant lentiviral vector (LV) comprising: an expression cassette comprising a nucleic acid construct comprising: a human β-globin locus control region comprising a plurality of reduced length hypersensitive site (HS) sequences where the nucleic acid sequence of said reduced length hypersensitive site (HS) sequences consist of one or more sequences independently selected from the group consisting of vector A HS1.1, vector B HS1.1, vector C HS1.1, vector D HS1.1, vector E HS1.1, vector A HS1.2, vector B HS1.2, vector C HS1.2, vector D HS1.2, vector A HS1.3, vector B HS1.3, vector A HS1.4, vector B HS1.4, vector A HS2.1, vector B HS2.1, vector C HS2.1, vector D HS2.1, vector E HS2.1, vector C HS2.2, vector A HS3.1, vector B HS3.1, vector C HS3.1, vector D HS3.1, vector E HS3.1, vector A HS3.2, vector B HS3.2, vector C HS3.2, vector A HS4.1, vector B HS4.1, vector C HS4.1, vector D HS4.1, vector E HS4.1, vector A HS5.1, vector B HS5.1, vector C HS5.1, vector D HS5.1, and vector E HS5.1, or where the reduced length hypersensitive site sequences having at least 90%, or at least 95%, or at least 98%, or at least 99% sequence identify with the nucleotide sequence of the foregoing reduced length hypersensitive site sequences; and a heterologous gene to be expressed by said construct operably linked to said human β-globin locus control region; and where said LV is a TAT-independent and self-inactivating (SIN) lentiviral vector.

2. The vector of claim 1, wherein the nucleic acid sequence of said reduced length hypersensitive site (HS) sequences consist of sequences independently selected from the group consisting of vector A HS1.1, vector B HS1.1, vector C HS1.1, vector D HS1.1, vector E HS1.1, vector A HS2.1, vector B HS2.1, vector C HS2.1, vector D HS2.1, vector E HS2.1, vector A HS3.1, vector B HS3.1, vector C HS3.1, vector D HS3.1, vector E HS3.1, vector A HS4.1, vector B HS4.1, vector C HS4.1, vector D HS4.1, vector E HS4.1, vector A HS5.1, vector B HS5.1, vector C HS5.1, vector D HS5.1, and vector E HS5.1.

3. The vector according to any one of claims 1-2, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequences: consisting of or comprising the sequence of one of vector A HS1.1, vector B HS1.1, vector C HS1.1, vector D HS1.1, or vector E HS1.1; consisting of or comprising the sequence of one of vector a HS 2.1, vector B HS2.1, vector C HS2.1, vector D HS2.1, or vector E HS2.1; consisting of or comprising the sequence of one of vector A HS3.1, vector B HS3.1, vector C HS3.1, vector D HS3.1, or vector E HS3.1; consisting of or comprising the sequence of one of vector A HS41., vector B HS4.1, vector C HS4.1, vector D HS4.1, or vector E HS4.1; and consisting of or comprising the sequence of one of vector A HS5.1, vector B HS5.1, vector C HS5.1, vector D HS5.1, or vector E HS5.1.

4. The vector according to any one of claims 1-3, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.1 o f vector A.

5. The vector according to any one of claims 1-3, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.1 of vector B.

6. The vector according to any one of claims 1-3, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.1 of vector C.

7. The vector according to any one of claims 1-3, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.1 of vector D.

8. The vector according to any one of claims 1-3, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.1 of vector E.

9. The vector according to any one of claims 1-8, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS2.1 of vector A.

10. The vector according to any one of claims 1-8, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS2.1 of vector B.

11. The vector according to any one of claims 1-8, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS2.1 of vector C.

12. The vector according to any one of claims 1-8, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS2.1 of vector D.

13. The vector according to any one of claims 1-8, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS2.1 of vector E.

14. The vector according to any one of claims 1-13, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS3.1 of vector A.

15. The vector according to any one of claims 1-13, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS3.1 of vector B.

16. The vector according to any one of claims 1-13, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS3.1 of vector C.

17. The vector according to any one of claims 1-13, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS3.1 of vector D.

18. The vector according to any one of claims 1-13, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS3.1 of vector E.

19. The vector according to any one of claims 1-18, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS4.1 of vector A.

20. The vector according to any one of claims 1-18, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS4.1 of vector B.

21. The vector according to any one of claims 1-18, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS4.1 of vector C.

22. The vector according to any one of claims 1-18, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS4.1 of vector D.

23. The vector according to any one of claims 1-18, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS4.1 of vector E.

24. The vector according to any one of claims 1-23, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS5.1 of vector A.

25. The vector according to any one of claims 1-23, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS5.1 of vector B.

26. The vector according to any one of claims 1-23, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS5.1 of vector C.

27. The vector according to any one of claims 1-23, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS5.1 of vector D.

28. The vector according to any one of claims 1-23, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS5.1 of vector E.

29. The vector according to any one of claims 1-28, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.2 of vector A.

30. The vector according to any one of claims 1-28, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.2 of vector B.

31. The vector according to any one of claims 1-28, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.2 of vector C.

32. The vector according to any one of claims 1-28, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.2 of vector D.

33. The vector according to any one of claims 1-32, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.3 of vector A.

34. The vector according to any one of claims 1-32, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.3 of vector B.

35. The vector according to any one of claims 1-34, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.4 of vector A.

36. The vector according to any one of claims 1-34, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.4 of vector B.

37. The vector according to any one of claims 1-36, wherein said wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS2.2 of vector C.

38. The vector according to any one of claims 1-37, wherein said wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS3.2 of vector A.

39. The vector according to any one of claims 1-37, wherein said wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS3.2 of vector B.

40. The vector according to any one of claims 1-39, wherein said wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS3.3 of vector C.

41. The vector of claim 1, wherein said β-globin locus control region comprises or consists of reduced length hypersensitive site (HS) sequences: vector A HS1.1, vector A HS1.2, vector A HS1.3, vector A HS1.4, vector A HS2.1, vector A HS3.1, vector A HS3.2, vector A HS4.1, and vector A HS5.1.

42. The vector of claim 1, wherein said β-globin locus control region comprises or consists of reduced length hypersensitive site (HS) sequences: vector B HS1.1, vector B HS1.2, vector B HS1.3, vector B HS1.4, vector B HS2.1, vector B HS3.1, vector B HS3.2, vector B HS4.1, and vector B HS5.1.

43. The vector of claim 1, wherein said β-globin locus control region comprises or consists of reduced length hypersensitive site (HS) sequences: vector C HS1.1, vector C HS1.2, vector C HS2.1, vector C HS2.2, vector C HS3.1, vector C HS3.2, vector C HS4.1, and vector C HS5.1.

44. The vector of claim 1, wherein said β-globin locus control region comprises or consists of reduced length hypersensitive site (HS) sequences: vector D HS1.1, vector D HS1.2, vector D HS2.1, vector D HS3.1, vector D HS4.1, and vector D HS51.

45. The vector of claim 1, wherein said β-globin locus control region comprises or consists of reduced length hypersensitive site (HS) sequences: vector E HS1.1, vector E HS2.1, vector E HS3.1, vector E HS 4.1, and vector E HS 51.

46. The vector according to any one of claims 1-45, wherein the reduced length hypersensitive site (HS) sequences are concatenated in order of increasing HS number:

HS1.1 (when present) - HS 1.2 (when present) - HS1.3 (when present)

- HS1.4 (when present - HS2 (when present) - HS2.1 (when present) - HS 2.2 (when present) - HS 3.1(when present) - HS3.2 (when present) - HS4.1 (when present) - HS5.1 (when present).

47. The vector of claim 1, wherein said β-globin locus control region comprises or consists of the LCR sequence of vector A.

48. The vector of claim 1, wherein said β-globin locus control region comprises or consists of the LCR sequence of vector B.

49. The vector of claim 1, wherein said β-globin locus control region comprises or consists of the LCR sequence of vector C.

50. The vector of claim 1, wherein said β-globin locus control region comprises or consists of the LCR sequence of vector D.

51. The vector of claim 1 , wherein said β-globin locus control region comprises or consists of the LCR sequence of vector E.

52. The vector according to any one of claims 1-51, wherein said heterologous gene comprises a recombinant human beta globin gene encoding a beta globin polypeptide.

53. The vector of claim 52, wherein said human beta globin gene comprises a wild-type beta globin gene.

54. The vector of claim 52, wherein said human beta globin gene comprises an anti-sickling human beta globin gene encoding an anti-sickling beta globin polypeptide.

55. The vector of claim 54, wherein said anti-sickling human beta globin gene encoding an anti-sickling-beta globin polypeptide comprise one or more mutations selected from the group consisting of Glyl6Asp, Glu22Ala and Thr87Gln.

56. The vector of claim 55, wherein said beta globin gene comprises the mutation Glyl6Asp.

57. The vector according to any one of claims 55-56, wherein said beta globin gene comprises the mutation Glu22Ala.

58. The vector according to any one of claims 55-57, wherein said beta globin gene comprises the mutation Thr87Gln.

59. The vector of claim 55, wherein said anti-sickling human β-globin gene comprises about 2.3 kb of recombinant human β-globin gene including exons and introns under the control of said human β-globin locus control region.

60. The vector according to any one of claims 1-59, wherein said β-globin gene comprises β-globin intron 2 with a 375 bp Rsal deletion from IVS2.

61. The vector according to any one of claims 1-60, wherein said β-globin gene comprises an Sspl (S) to Rsal (R) deletion (~220bp).

62. The vector according to any one of claims 1-61, wherein said vector comprises a human Ankyrin insulator element.

63. The vector according to any one of claims 1-62, further comprising an insulator in the 3’ LTR.

64. The vector of claim 63, wherein said insulator comprises FB (FII/BEAD-A), a 77 bp insulator element, which contains the minimal CTCF binding site enhancer-blocking components of the chicken β-globin 5’ Dnasel-hypersensitive site 4 (5’ HS4).

65. The vector according to any one of claims 1-64, wherein said vector comprises a ψ region vector genome packaging signal.

66. The vector according to any one of claims 1-65, wherein the 5’ LTR comprises a CMV enhancer/promoter.

67. The vector according to any one of claims 1-66, wherein said vector comprises a Rev Responsive Element (RRE).

68. The vector according to any one of claims 1-67, wherein said vector comprises a central polypurine tract.

69. The vector according to any one of claims 1-68, wherein said vector comprises a post-translational regulatory element.

70. The vector of claim 69, wherein the posttranscriptional regulatory element is modified Woodchuck Post-transcriptional Regulatory Element (WPRE).

71. The vector of claim 1, wherein said vector comprises the nucleic acid sequence of vector A.

72. The vector of claim 1, wherein said vector comprises the nucleic acid sequence of vector B.

73. The vector of claim 1, wherein said vector comprises the nucleic acid sequence of vector C.

74. The vector of claim 1, wherein said vector comprises the nucleic acid sequence of vector D.

75. The vector of claim 1, wherein said vector comprises the nucleic acid sequence of vector E.

76. The vector according to any one of claims 1-75, wherein said vector is incapable of reconstituting a wild-type lentivims through recombination.

77. A host cell transduced with a vector according to any one of claims 1-

76.

78. The host cell of claim 77, wherein the cell is a stem cell.

79. The host cell of claim 78, wherein said cell is a stem cell derived from bone marrow, and/or from umbilical cord blood, and/or from peripheral blood.

80. The host cell of claim 77, wherein the cell is a 293T cell.

81. The host cell of claim 77, wherein, wherein the cell is a human hematopoietic progenitor cell.

82. The host cell of claim 81 , wherein the human hematopoietic progenitor cell is a CD34+ cell.

83. A method of treating a hemoglobinopathy, in a subject, said method comprising: transducing a stem cell and/or progenitor cell from said subject with a vector according to any one of claims 1-76; and transplanting said transduced cell or cells derived therefrom into said subject where said cells or derivatives therefrom express said anti-sickling human beta globin gene.

84. The method of claim 83, wherein the cell is a stem cell.

85. The host cell of claim 83, wherein said cell is a stem cell derived from bone marrow.

86. The method of claim 83, wherein, wherein the cell is a human hematopoietic progenitor cell.

87. The method of claim 86, wherein the human hematopoietic progenitor cell is a CD34+ cell.

88. The method according to any one of claims 83-87, wherein said hemoglobinopathy is sickle cell disease.

89. The method according to any one of claims 83-87, wherein said hemoglobinopathy is β-thalassemia.

Description:
OPTIMIZED LENTIVIRAL VECTOR COMPRISING MINIMAL

ENHANCER ELEMENTS FOR STEM CELL GENE THERAPY OF

HEMOGLOBINOPATHIES

CROSS-REFERENCE TO RELATED APPLICATIONS [0001] This application claims benefit of and priority to USSN 62/805,817, filed on February 14, 2019, which is incorporated herein by reference inits entirety for all purposes.

STATEMENT OF GOVERNMENTAL SUPPORT

[ Not Applicable ]

INCORPORATION BY REFERENCE OF SEQUENCE LISTING PROVIDED AS A

TEXT FILE

[0002] A Sequence Listing is provided herewith as a text file, “UCLA-

P207P_ST25.txt” created on February 13, 2019 and having a size of 95,997 bytes. The contents of the text file are incorporated by reference herein in their entirety.

BACKGROUND [0003] Sickle cell disease (SCD) is one of the most common monogenic disorders worldwide and is a major cause of morbidity and early mortality (Hoffman et al. (2009) Hematology: Basic Principles and Practice. 5th ed. London, United Kingdom, Churchill Livingstone). SCD affects approximately 80,000 Americans, and causes significant neurologic, pulmonary, and renal injury, as well as severe acute and chronic pain that adversely impacts quality of life. It is estimated that approximately 240,000 children are bom annually in Africa with SCD and 80% die by their second birthday. The average lifespan of subjects with SCD in the United States is approximately 40 years and this has remained unchanged over the last 3-4 decades.

[0004] SCD is caused by a single amino acid change in β-globin (Glu 6 to Val 6) which leads to hemoglobin polymerization and red blood cell (rbc) sickling. SCD typically results in continual low-grade ischemia and episodic exacerbations or “crises” resulting in tissue ischemia, organ damage, and premature death.

[0005] Although SCD is well characterized, there is still no ideal long-term treatment.

Current therapies are based on induction of fetal hemoglobin (HbF) to inhibit polymerization of sickle hemoglobin (HbS) (Voskaridou et al. (2010) Blood, 115(12): 2354-2363) and cell dehydration (Eaton and Hofrichter (1987) Blood, 70(5): 1245-1266) or reduction of the percentage of HbS by transfusions (Stamatoyannopoulos et al, eds. (2001) Molecular Basis of Blood Diseases. 3rd ed. Philadelphia, Pennsylvania, USA: WB Saunders). Allogeneic human stem cell transplantation (HSCT) from bone marrow (BM) or umbilical cord blood (UCB) or mobilized peripheral blood stem cells (mPBSC) is a potentially curative therapy, although only a small percentage of patients have undergone this procedure, mostly children with severe symptoms who had HLA-matched sibling donors (Bolanos-Meade and Brodsky (2009) Curr. Opin. Oncol. 21(2): 158-161; Rees et al. (2010) Lancet, 376(9757): 2018-2031; Shenoy (2011) Hematology Am Soc Hematol Educ Program. 2011: 273-279).

[0006] Transplantation of allogeneic cells carries the risk of graft- versus host disease

(GvHD), which can be a cause of extensive morbidity. HSCT using UCB from matched unrelated donors holds reduced risk of acute or chronic GvHD compared with using BM; however, there is a higher probability of engraftment failure using UCB as a result of its lower cell dose and immunologic immaturity (Kamani et al. (2012) Biol. Blood Marrow Transplant. 18(8): 1265-1272; Locatelli and Pagliara (2012) Pediatr. Blood Cancer. 59(2): 372-376).

[0007] Gene therapy with autologous human stem cells (HSCs) is an alternative to allogeneic HSCT, since it avoids the limitations of finding a matched donor and the risks of GvHD and graft rejection. For gene therapy application in SCD patients, one source for autologous HSC would be BM, due to the complications previously described when G-CSF was used to collect autologous peripheral blood stem cells (PBSCs) in SCD patients (Abboud et al. (1998) Lancet351(9107): 959; Adler et al. (2001) Blood, 97(10): 3313-3314; Fitzhugh et al. (2009) Cytotherapy, 11(4): 464-471). However, more recently, plerixafor, an immunostimulat, can be used to mobilize hematopoietic stem cells into the blood stream.

The stem cells can then be extracted from the blood and use. Although general anesthesia imposes a risk for SCD patients as well, current best medical practices can minimize these (Neumayr et al. (1998 ) Am. J. Hematol. 57(2): 101-108).

[0008] The development of integrating vectors for β-globin gene transfer has been challenging due to the complex regulatory elements needed for high-level, erythroid-specific expression (Lisowski a & Sadelain (2008) Br. J. Haematol. 141(3): 335-345). γ-Retroviral vectors were unable to transfer these β-globin expression cassettes intact (Gelinas et al.

(1989) Adv. Exp. Med. Biol. 271: 135-148; Gelinas et al. (1989) Prog. Clin. Biol. Res. 316B: 235-249). In contrast, lentiviral vectors (LV) can transfer β-globin cassettes intact with relatively high efficiency, although the titers of these vectors are reduced compared with those of vectors bearing simpler cassettes (see, e.g., May et al. (2000) Nature 406(6791): 82- 86; Pawliuk et al. (2001) Science, 294(5550): 2368-2371). In the last decade, many groups have developed different β-globin LV for targeting β-hemoglobinopathies, with successful therapeutic results following transplantation of ex vivo- modified HSC in mouse models (May et al. (2000) Nature406(6791): 82-86; Pawliuk et al. (2001) Science, 294(5550): 2368-2371; Levasseur et al. (2003 ) Blood, 102(13):4312-4319; Hanawa et al. (2004) Blood, 104(8): 2281-2290; Puthenveetil et al. (2004) Blood, 104(12): 3445-3453; Miccio et al. (2008) Proc. Natl. Acad. Sci. USA, 105(30):10547-10552; Pestina et al. (2008) Mol. Ther. 17(2): 245-252). Recently, Bluebird Bio, updated trial results with longer follow-up from lentiviral vector treated patients with beta-thalassemia and sickle cell disease. Across three small studies testing the gene therapy in the two blood diseases, patients given LentiGlobin saw their levels of the crucial oxygen-carrying protein hemoglobin rise to approach normal, eliminating the need for blood transfusions in most over the studied period.

[0009] Sickle patients with hereditary persistence of fetal hemoglobin (HbF) (HPFH) have improved survival and amelioration of clinical symptoms, with maximal clinical benefits observed when the HbF is elevated above threshold values (e.g., 8%-15% of the total cellular Hb) (Voskaridou et al. (2010) Blood, 115(12): 2354-2363; Platt et al. (1994) N. Engl. J. Med. 330(23): 1639-1644). Therefore, some gene therapy strategies have employed viral vectors carrying the human γ-globin gene ( HBG1/2 ). However, these constructs expressed HbF poorly in adult erythroid cells, since fetal- specific transcription factors are required for high-level expression of the γ-globin gene (Chakalova et al. (2005) Blood105(5): 2154-2160; Russell (2007) Eur. J. Haematol. 79(6): 516-525). These limitations have been overcome by embedding the exons encoding human γ-globin within the human β-globin gene 5' promoter and 3' enhancer elements (Hanawa et al. (2004) Blood, 104(8): 2281-2290; Persons et al. (2002) Blood, 101(6): 2175-2183; Perumbeti et al. (2009) Blood, 114(6): 1174-1185). Breda et al. (2012) PLoS One, 7(3): e32345 used an LV vector encoding the human hemoglobin (HBB) gene to increase the expression of normal HbA in CD34 + -derived erythroid cells from SCD patients, however, the expression level needed when the HBB gene is used would be higher than would be required for HBG1/2 gene expression to achieve therapeutic benefits in SCD patients.

[0010] Another approach is to modify β-globin genes to confer antisickling activity by substituting key amino acids from γ-globin. The modified β-globin cassette should yield the necessary high-level, erythroid- specific expression in adult erythroid cells. Pawliuk et al. (2001) Science, 294(5550): 2368-2371 designed an LV carrying a human β-globin gene with the amino acid modification T87Q. The glutamine at position 87 of γ-globin has been implicated in the anti-sickling activity of HbF (Nagel et al. (1979) Proc. Natl. Acad Sci., USA, 76(2): 670-672). This anti-sickling construct corrected SCD in 2 murine models of the disease, and a similar LV has been used in a clinical trial for β-thalassemia and SCD in France (Cavazzana-Calvo et al. (2010) Nature, 467(7313): 318-322).

[0011] Townes and colleagues have taken a similar approach, developing a recombinant human anti-sickling β-globin gene (HBBAS3) encoding a β-globin protein (HbAS3) that has 3 amino substitutions compared with the original (HbA): T87Q for blocking the lateral contact with the canonical Val 6 of HbS, E22A to disrupt axial contacts (McCune et al. (1994) Proc. Natl. Acad. Sci. USA, 91(21): 9852-9856) and G16D, which confers a competitive advantage over sickle-β-globin chains for interaction with the α-globin polypeptide. Functional analysis of the purified HbAS3 protein demonstrated that this recombinant protein had potent activity to inhibit HbS tetramer polymerization (Levasseur et al. (2004) J. Biol. Chem. 279(26): 27518-27524.). Levasseur et al. (2003) Blood, 102(13): 4312-4319, showed efficient transduction of BM stem cells from a murine model of SCD with a self-inactivating (SIN) LV carrying the HBBAS3 transgene that resulted in normalized rbc physiology and prevented the pathological manifestations of SCD.

[0012] Unfortunately, current β-globin expression vectors, suffer from low vector titer and sub-optimal gene transfer to hematopoietic stem cells, representing a major barrier toward the effective implementation of this gene therapy strategy to the clinic.

SUMMARY

[0013] Various embodiments contemplated herein may include, but need not be limited to, one or more of the following: [0014] Embodiment 1: A recombinant lentiviral vector (LV) comprising: an expression cassette comprising a nucleic acid construct comprising:

[0015] a human β-globin locus control region comprising a plurality of reduced length hypersensitive site (HS) sequences where the nucleic acid sequence of said reduced length hypersensitive site (HS) sequences consist of one or more sequences independently selected from the group consisting of vector A HS1.1, vector B HS1.1, vector C HS1.1, vector D HS1.1, vector E HS1.1, vector A HS1.2, vector B HS1.2, vector C HS1.2, vector D HS1.2, vector A HS1.3, vector B HS1.3, vector A HS1.4, vector B HS1.4, vector A HS2.1, vector B HS2.1, vector C HS2.1, vector D HS2.1, vector E HS2.1, vector C HS2.2, vector A HS3.1, vector B HS3.1, vector C HS3.1, vector D HS3.1, vector E HS3.1, vector A HS3.2, vector B HS3.2, vector C HS3.2, vector A HS4.1, vector B HS4.1, vector C HS4.1, vector D HS4.1, vector E HS4.1, vector A HS5.1, vector B HS5.1, vector C HS5.1, vector D HS5.1, and vector E HS5.1, or where the reduced length hypersensitive site sequences having at least 90%, or at least 95%, or at least 98%, or at least 99% sequence identify with the nucleotide sequence of the foregoing reduced length hypersensitive site sequences; and

[0016] a heterologous gene to be expressed by said construct operably linked to said human β-globin locus control region; and where said LV is a TAT- independent and self-inactivating (SIN) lentiviral vector. [0017] Embodiment 2: The vector of embodiment 1, wherein the nucleic acid sequence of said reduced length hypersensitive site (HS) sequences consist of sequences independently selected from the group consisting of vector A HS1.1, vector B HS1.1, vector C HS1.1, vector D HS1.1, vector E HS1.1, vector A HS2.1, vector B HS2.1, vector C HS2.1, vector D HS2.1, vector E HS2.1, vector A HS3.1, vector B HS3.1, vector C HS3.1, vector D HS3.1, vector E HS3.1, vector A HS4.1, vector B HS4.1, vector C HS4.1, vector D HS4.1, vector E HS4.1, vector A HS5.1, vector B HS5.1, vector C HS5.1, vector D HS5.1, and vector E HS5.1.

[0018] Embodiment 3: The vector according to any one of embodiments 1-2, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequences:

[0019] consisting of or comprising the sequence of one of vector A HS1.1, vector B HS1.1, vector C HS1.1, vector D HS1.1, or vector E HS1.1;

[0020] consisting of or comprising the sequence of one of vector a HS 2.1, vector B HS2.1, vector C HS2.1, vector D HS2.1, or vector E HS2.1; [0021] consisting of or comprising the sequence of one of vector A HS3.1, vector B HS3.1, vector C HS3.1, vector D HS3.1, or vector E HS3.1;

[0022] consisting of or comprising the sequence of one of vector A HS41., vector B HS4.1, vector C HS4.1, vector D HS4.1, or vector E HS4.1; and [0023] consisting of or comprising the sequence of one of vector A HS5.1, vector B HS5.1, vector C HS5.1, vector D HS5.1, or vector E HS5.1.

[0024] Embodiment 4: The vector according to any one of embodiments 1-3, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.1 o f vector A. [0025] Embodiment 5: The vector according to any one of embodiments 1-3, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS 1.1 of vector B.

[0026] Embodiment 6: The vector according to any one of embodiments 1-3, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS 1.1 of vector C.

[0027] Embodiment 7: The vector according to any one of embodiments 1-3, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS 1.1 of vector D. [0028] Embodiment 8: The vector according to any one of embodiments 1-3, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS 1.1 of vector E.

[0029] Embodiment 9: The vector according to any one of embodiments 1-8, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS2.1 of vector A.

[0030] Embodiment 10: The vector according to any one of embodiments 1-8, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS2.1 of vector B.

[0031] Embodiment 11 : The vector according to any one of embodiments 1-8, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS2.1 of vector C.

[0032] Embodiment 12: The vector according to any one of embodiments 1-8, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS2.1 of vector D. [0033] Embodiment 13: The vector according to any one of embodiments 1-8, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS2.1 of vector E.

[0034] Embodiment 14: The vector according to any one of embodiments 1-13, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS3.1 of vector A. [0035] Embodiment 15: The vector according to any one of embodiments 1-13, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS3.1 of vector B.

[0036] Embodiment 16: The vector according to any one of embodiments 1-13, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS3.1 of vector C.

[0037] Embodiment 17: The vector according to any one of embodiments 1-13, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS3.1 of vector D. [0038] Embodiment 18: The vector according to any one of embodiments 1-13, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS3.1 of vector E.

[0039] Embodiment 19: The vector according to any one of embodiments 1-18, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS4.1 of vector A.

[0040] Embodiment 20: The vector according to any one of embodiments 1-18, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS4.1 of vector B.

[0041] Embodiment 21: The vector according to any one of embodiments 1-18, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS4.1 of vector C.

[0042] Embodiment 22: The vector according to any one of embodiments 1-18, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS4.1 of vector D. [0043] Embodiment 23: The vector according to any one of embodiments 1-18, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS4.1 of vector E.

[0044] Embodiment 24: The vector according to any one of embodiments 1-23, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS5.1 of vector A. [0045] Embodiment 25: The vector according to any one of embodiments 1-23, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS5.1 of vector B.

[0046] Embodiment 26: The vector according to any one of embodiments 1-23, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS5.1 of vector C.

[0047] Embodiment 27: The vector according to any one of embodiments 1-23, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS5.1 of vector D. [0048] Embodiment 28: The vector according to any one of embodiments 1-23, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS5.1 of vector E.

[0049] Embodiment 29: The vector according to any one of embodiments 1-28, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.2 of vector A.

[0050] Embodiment 30: The vector according to any one of embodiments 1-28, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.2 of vector B.

[0051] Embodiment 31: The vector according to any one of embodiments 1-28, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.2 of vector C.

[0052] Embodiment 32: The vector according to any one of embodiments 1-28, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.2 of vector D. [0053] Embodiment 33: The vector according to any one of embodiments 1-32, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.3 of vector A.

[0054] Embodiment 34: The vector according to any one of embodiments 1-32, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS 1.3 of vector B . [0055] Embodiment 35: The vector according to any one of embodiments 1-34, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.4 of vector A.

[0056] Embodiment 36: The vector according to any one of embodiments 1-34, wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.4 of vector B.

[0057] Embodiment 37: The vector according to any one of embodiments 1-36, wherein said wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS2.2 of vector C.

[0058] Embodiment 38: The vector according to any one of embodiments 1-37, wherein said wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS3.2 of vector A. [0059] Embodiment 39: The vector according to any one of embodiments 1-37, wherein said wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS3.2 of vector B.

[0060] Embodiment 40: The vector according to any one of embodiments 1-39, wherein said wherein said β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS3.3 of vector C.

[0061] Embodiment 41: The vector of embodiment 1, wherein said β-globin locus control region comprises or consists of reduced length hypersensitive site (HS) sequences: vector A HS1.1, vector A HS1.2, vector A HS1.3, vector A HS1.4, vector A HS2.1, vector A HS3.1, vector A HS3.2, vector A HS4.1, and vector A HS5.1.

[0062] Embodiment 42: The vector of embodiment 1, wherein said β-globin locus control region comprises or consists of reduced length hypersensitive site (HS) sequences: vector B HS1.1, vector B HS1.2, vector B HS1.3, vector B HS1.4, vector B HS2.1, vector B HS3.1, vector B HS3.2, vector B HS4.1, and vector B HS5.1.

[0063] Embodiment 43: The vector of embodiment 1, wherein said β-globin locus control region comprises or consists of reduced length hypersensitive site (HS) sequences: vector C HS1.1, vector C HS1.2, vector C HS2.1, vector C HS2.2, vector C HS3.1, vector C HS3.2, vector C HS4.1, and vector C HS5.1.

[0064] Embodiment 44: The vector of embodiment 1, wherein said β-globin locus control region comprises or consists of reduced length hypersensitive site (HS) sequences: vector D HS1.1, vector D HS1.2, vector D HS2.1, vector D HS3.1, vector D HS4.1, and vector D HS51.

[0065] Embodiment 45: The vector of embodiment 1, wherein said β-globin locus control region comprises or consists of reduced length hypersensitive site (HS) sequences: vector E HS1.1, vector E HS2.1, vector E HS3.1, vector E HS 4.1, and vector E HS 51. [0066] Embodiment 46: The vector according to any one of embodiments 1-45, wherein the reduced length hypersensitive site (HS) sequences are concatenated in order of increasing HS number HS1.1 (when present) - HS 1.2 (when present) - HS1.3 (when present) - HS1.4 (when present - HS2 (when present) - HS2.1 (when present) - HS 2.2 (when present) - HS 3.1(when present) - HS3.2 (when present) - HS4.1 (when present) - HS5.1 (when present).

[0067] Embodiment 47: The vector of embodiment 1, wherein said β-globin locus control region comprises or consists of the LCR sequence of vector A.

[0068] Embodiment 48: The vector of embodiment 1, wherein said β-globin locus control region comprises or consists of the LCR sequence of vector B. [0069] Embodiment 49: The vector of embodiment 1, wherein said β-globin locus control region comprises or consists of the LCR sequence of vector C.

[0070] Embodiment 50: The vector of embodiment 1, wherein said β-globin locus control region comprises or consists of the LCR sequence of vector D.

[0071] Embodiment 51 : The vector of embodiment 1 , wherein said β-globin locus control region comprises or consists of the LCR sequence of vector E.

[0072] Embodiment 52: The vector according to any one of embodiments 1-51, wherein said heterologous gene comprises a recombinant human beta globin gene encoding a beta globin polypeptide.

[0073] Embodiment 53: The vector of embodiment 52, wherein said human beta globin gene comprises a wild-type beta globin gene. [0074] Embodiment 54: The vector of embodiment 52, wherein said human beta globin gene comprises an anti-sickling human beta globin gene encoding an anti-sickling beta globin polypeptide.

[0075] Embodiment 55: The vector of embodiment 54, wherein said anti-sickling human beta globin gene encoding an anti-sickling-beta globin polypeptide comprise one or more mutations selected from the group consisting of Glyl6Asp, Glu22Ala and Thr87Gln.

[0076] Embodiment 56: The vector of embodiment 55, wherein said beta globin gene comprises the mutation Glyl6Asp.

[0077] Embodiment 57: The vector according to any one of embodiments 55-56, wherein said beta globin gene comprises the mutation Glu22Ala.

[0078] Embodiment 58: The vector according to any one of embodiments 55-57, wherein said beta globin gene comprises the mutation Thr87Gln.

[0079] Embodiment 59: The vector of embodiment 55, wherein said anti-sickling human β-globin gene comprises about 2.3 kb of recombinant human β-globin gene including exons and introns under the control of said human β-globin locus control region.

[0080] Embodiment 60: The vector according to any one of embodiments 1-59, wherein said β-globin gene comprises β-globin intron 2 with a 375 bp Rsal deletion from IVS2.

[0081] Embodiment 61: The vector according to any one of embodiments 1-60, wherein said β-globin gene comprises an Sspl (S) to Rsal (R) deletion (~220bp).

[0082] Embodiment 62: The vector according to any one of embodiments 1-61, wherein said vector comprises a human Ankyrin insulator element.

[0083] Embodiment 63: The vector according to any one of embodiments 1-62, further comprising an insulator in the 3’ LTR. [0084] Embodiment 64: The vector of embodiment 63, wherein said insulator comprises FB (FII/BEAD-A), a 77 bp insulator element, which contains the minimal CTCF binding site enhancer-blocking components of the chicken β-globin 5’ Dnasel-hypersensitive site 4 (5' HS4).

[0085] Embodiment 65: The vector according to any one of embodiments 1-64, wherein said vector comprises a ψ region vector genome packaging signal. [0086] Embodiment 66: The vector according to any one of embodiments 1-65, wherein the 5’ LTR comprises a CMV enhancer/promoter.

[0087] Embodiment 67: The vector according to any one of embodiments 1-66, wherein said vector comprises a Rev Responsive Element (RRE). [0088] Embodiment 68: The vector according to any one of embodiments 1-67, wherein said vector comprises a central polypurine tract

[0089] Embodiment 69: The vector according to any one of embodiments 1-68, wherein said vector comprises a post-translational regulatory element.

[0090] Embodiment 70: The vector of embodiment 69, wherein the posttranscriptional regulatory element is modified Woodchuck Post-transcriptional Regulatory Element (WPRE).

[0091] Embodiment 71: The vector of embodiment 1, wherein said vector comprises the nucleic acid sequence of vector A.

[0092] Embodiment 72: The vector of embodiment 1, wherein said vector comprises the nucleic acid sequence of vector B.

[0093] Embodiment 73: The vector of embodiment 1, wherein said vector comprises the nucleic acid sequence of vector C.

[0094] Embodiment 74: The vector of embodiment 1, wherein said vector comprises the nucleic acid sequence of vector D. [0095] Embodiment 75: The vector of embodiment 1, wherein said vector comprises the nucleic acid sequence of vector E.

[0096] Embodiment 76: The vector according to any one of embodiments 1-75, wherein said vector is incapable of reconstituting a wild-type lentivims through recombination. [0097] Embodiment 77: A host cell transduced with a vector according to any one of embodiments 1-76.

[0098] Embodiment 78: The host cell of embodiment 77, wherein the cell is a stem cell.

[0099] Embodiment 79: The host cell of embodiment 78, wherein said cell is a stem cell derived from bone marrow, and/or from umbilical cord blood, and/or from peripheral blood. [0100] Embodiment 80: The host cell of embodiment 77, wherein the cell is a 293T cell.

[0101] Embodiment 81: The host cell of embodiment 77, wherein, wherein the cell is a human hematopoietic progenitor cell. [0102] Embodiment 82: The host cell of embodiment 81, wherein the human hematopoietic progenitor cell is a CD34+ cell.

[0103] Embodiment 83: A method of treating a hemoglobinopathy, in a subject, said method comprising: transducing a stem cell and/or progenitor cell from said subject with a vector according to any one of embodiments 1-76; and transplanting said transduced cell or cells derived therefrom into said subject where said cells or derivatives therefrom express said anti-sickling human beta globin gene.

[0104] Embodiment 84: The method of embodiment 83, wherein the cell is a stem cell.

[0105] Embodiment 85: The host cell of embodiment 83, wherein said cell is a stem cell derived from bone marrow.

[0106] Embodiment 86: The method of embodiment 83, wherein, wherein the cell is a human hematopoietic progenitor cell.

[0107] Embodiment 87: The method of embodiment 86, wherein the human hematopoietic progenitor cell is a CD34 + cell. [0108] Embodiment 88: The method according to any one of embodiments 83-87, wherein said hemoglobinopathy is sickle cell disease.

[0109] Embodiment 89: The method according to any one of embodiments 83-87, wherein said hemoglobinopathy is β-thalassemia.

Definitions. [0110] A "reduced length hypersensitive site (HS) sequence" refers to an HS sequence that is shorter in length than the corresponding wild type HS sequence, e.g., HS2, HS3, and HS4 as previously defined (e.g., HS2 (~1.20kb), HS3 (~1.28kb), and HS4 (~l.lkb)) (see, e.g., Forrester et al. (1986) Proc. Natl. Acad. Sci. USA, 83: 1359-1363). In certain embodiments the reduced length HS sequence expressly excludes one or more of the HS core sequence(s) as described in PCT Publication No: WO 2013/071309 (PCT/US2012/064878) which is incorporated herein by reference for the core HS sequences described therein (e.g., core HS2 (~420 bp), core HS3 (~340bp), and/or core HS4 (~410 bp)).

[0111] "Recombinant" is used consistently with its usage in the art to refer to a nucleic acid sequence that comprises portions that do not naturally occur together as part of a single sequence or that have been rearranged relative to a naturally occurring sequence. A recombinant nucleic acid is created by a process that involves the hand of man and/or is generated from a nucleic acid that was created by hand of man (e.g. , by one or more cycles of replication, amplification, transcription, etc.). A recombinant virus is one that comprises a recombinant nucleic acid. A recombinant cell is one that comprises a recombinant nucleic acid.

[0112] As used herein, the term "recombinant lentiviral vector" or "recombinant LV) refers to an artificially created polynucleotide vector assembled from an LV and a plurality of additional segments as a result of human intervention and manipulation.

[0113] By "globin nucleic acid molecule" is meant a nucleic acid molecule that encodes a globin polypeptide. In various embodiments the globin nucleic acid molecule may include regulatory sequences upstream and/or downstream of the coding sequence.

[0114] By "globin polypeptide" is meant a protein having at least 85%, or at least 90%, or at least 95%, or at least 98% amino acid sequence identity to a human alpha, beta or gamma globin. [0115] The term "therapeutic functional globin gene" refers to a nucleotide sequence the expression of which leads to a globin that does not produce a hemoglobinopathy phenotype, and which is effective to provide therapeutic benefits to an individual with a defective globin gene. The functional globin gene may encode a wild-type globin appropriate for a mammalian individual to be treated, or it may be a mutant form of globin, preferably one which provides for superior properties, for example superior oxygen transport properties or anti-sickling properties. The functional globin gene includes both exons and introns, as well as globin promoters and splice donors/acceptors.

[0116] By "an effective amount" is meant the amount of a required agent or composition comprising the agent to ameliorate or eliminate symptoms of a disease relative to an untreated patient. The effective amount of composition(s) used to practice the methods described herein for therapeutic treatment of a disease varies depending upon the manner of administration, the age, body weight, and general health of the subject. Ultimately, the attending physician or veterinarian will decide the appropriate amount and dosage regimen. Such amount is referred to as an "effective" amount.

[0117] The term "sequence identity" refers to the extent to which two optimally aligned polynucleotide or polypeptide sequences are invariant throughout a window of alignment of components, e.g. , nucleotides or amino acids. "Identity" can be readily calculated by known methods including, but not limited to, those described in: Computational Molecular Biology (Lesk, A. M., ed.) Oxford University Press, New York (1988); Biocomputing: Informatics and Genome Projects (Smith, D. W., ed.) Academic Press, New York (1993); Computer Analysis of Sequence Data, Part I (Griffin, A. M., and Griffin, H. G., eds.) Humana Press, New Jersey (1994); Sequence Analysis in Molecular Biology (von

Heinje, G., ed.) Academic Press (1987); and Sequence Analysis Primer (Gribskov, M. and Devereux, J., eds.) Stockton Press, New York (1991); and the like.

[0118] An "identity fraction" for aligned segments of a test sequence and a reference sequence is the number of identical components which are shared by the two aligned sequences divided by the total number of components in the reference sequence segment, i.e. , the entire reference sequence or a smaller defined part of the reference sequence. Percent sequence identity is typically represented as the identity fraction multiplied by 100. As used herein, the term "percent sequence identity" or "percent identity" refers to the percentage of identical nucleotides in a linear polynucleotide sequence of a reference ("query") polynucleotide molecule (or its complementary strand) as compared to a test ("subject") polynucleotide molecule (or its complementary strand) when the two sequences are optimally aligned (with appropriate nucleotide insertions, deletions, or gaps totaling less than 20 percent of the reference sequence over the window of comparison).

[0119] Optimal alignment of sequences for aligning a comparison window is well known to those skilled in the art and may be conducted by tools such as the local homology algorithm of Smith and Waterman, the homology alignment algorithm of Needleman and Wunsch, the search for similarity method of Pearson and Lipman, and optionally by computerized implementations of these algorithms such as GAP, BESTFIT, FASTA, and TFASTA available as part of the GCG® Wisconsin Package® (Accelrys Inc., Burlington, Mass.). The comparison of one or more polynucleotide sequences may be to a full-length polynucleotide sequence or a portion thereof, or to a longer polynucleotide sequence. In various embodiments "percent identity" may also be determined using BLASTX version 2.0 for translated nucleotide sequences and BLASTN version 2.0 for polynucleotide sequences. [0120] The percent of sequence identity can be determined using the "Best Fit" or "Gap" program of the Sequence Analysis Software Package.TM. (Version 10; Genetics Computer Group, Inc., Madison, Wis.). "Gap" utilizes the algorithm of Needleman and Wunsch (1970) J. Mol. Biol. 48: 443-453, to find the alignment of two sequences that maximizes the number of matches and minimizes the number of gaps. "BestFit" performs an optimal alignment of the best segment of similarity between two sequences and inserts gaps to maximize the number of matches using the local homology algorithm of Smith and Waterman (1981) Adv. Appl. Math., 2: 482-489, Smith et al. (1983) Nucleic Acids Res. 11: 2205-2220. Useful methods for determining sequence identity are also disclosed in Guide to Huge Computers (Martin J. Bishop, ed., Academic Press, San Diego (1994)), and Carillo et al. (1988) Appl. Math, 48: 1073). Certain illustrative computer programs for determining sequence identity include, but are not limited to, the Basic Local Alignment Search Tool (BLAST) programs, that are publicly available from National Center Biotechnology Information (NCBI) at the National Library of Medicine, National Institute of Health, Bethesda, Md. 20894; see BLAST Manual, Altschul et al., NCBI, NLM, NIH; (Altschul et al., J. Mol. Biol. 215:403-410 (1990)); version 2.0 or higher of BLAST programs allows the introduction of gaps (deletions and insertions) into alignments; for peptide sequence, BLASTX can be used to determine sequence identity; and for polynucleotide sequence, BLASTN can be used to determine sequence identity.

BRIEF DESCRIPTION OF THE DRAWINGS

[0121] Figure 1, panels A-C, illustrate the library and oligonucleotide design: Panel A) Each sequence is composed of lOObp and each subsequent sequence is offset by four nucleotides. The entire library is duplicated three times by assigning three unique barcodes to each sequence. An antisense library is then designed in similar fashion. Panel B) Each oligonucleotide includes 55bp of “backend” sequence needed for downstream cloning into a lentiviral reporter vector. Cloning results in the placement of the enhancer fragment upstream of the promoter and barcode upstream of the polyadenylation signal. Panel C) Sliding lOObp window that moves 4bp at a time across large DNA regions in the context of a lentiviral vector to assay for enhancer activity. Added benefit of selecting for sequences that are successfully packaged and transferred to target cell.

[0122] Figure 2. After microarray oligonucleotide synthesis the plasmid library is assembled, packaged into lentiviral vector particles and transferred to Human Umbilical Derived Cord Blood 2 (HUDEP2:HD2) cells. Cells are harvested for RNA and DNA after culture under erythroid conditions to induce globin expression. gDNA and RNA are extracted, RNA is converted to cDNA, barcodes amplified from plasmid, cDNA and gDNA, and PCR products sequenced. cDNA barcodes are normalized to plasmid or gDNA barcodes to create a normalized map of enhancer activity. [0123] Figure 3, panels A & B, shows maps of intrinsic enhancer regions across the endogenous glbin LCR. Panel A) Map of sequence intrinsic enhancer regions across the endogenous globin LCR overlaid with other ChhiP_Seq data sets. Panel B) Schema showing different cutoff levels and the lengths of concatenated mini-LCRs produced with alignment of sequence to dendogenous globin LCR. [0124] Figure 4 illustrates the CCL-BAS3-FB lentiviral vector along with 4 illustrative reduced length lentiviral vectors (vectors A (80) B (90), C (95), D (97.5), and E ((98.5)).

[0125] Figure 5 shows the raw titer produced by vectors B, C, D, and E, compared to the CCL-BAS3-FB. [0126] Figure 6 shows the average %BAS3/total/VCN in HUDEP cells produced by vectors B, C, D, and ED, compared to the CCL-BAS3-FB.

[0127] Figure 7 shows the gene transfer efficiency at various multiplicities of infection (MOI) to CD34+ bone marrow derived HSPCs for vectors A, B, C, and D, compared to the CCL-BAS3-FB. [0128] Figures 8, panel A, shows the average %BAS3/total/VCN vs vector length for vectors B, C, D, and E, compared to the CCL-BAS3-FB when CD34+ HSPCs were transduced at various MOI and cultured under erythroid culture conditions. Panel A demonstrates that enhancer length correlates with %BAS3/total/VCN. Panel B shows the VCNs that were achieved at the 1.0 x 10 7 (TU/mL) transduction condition for the experiment shown in panel A. Panel C shows the average %BAS3/Total expression avhieved at the 1.0 x 10 7 (TU/mL) transduction condition for the experiment shown in panel A. Panel D shows the %BAS3/total/VCN at the 1.0 x 10 7 (TU/mL) transduction condition for the experiment shown in panel A.

[0129] Figure 9 shows week 4 in vivo data produce after transplanting Lin(-) bone marrow cells derived from SCD mouse model into lethally irradiated recipient mice. Lin(-) bone marrow cells were transduced using vector C (97.5) or CCL-BAS3-FB. Panel A shows %Engraftment of mononuclear cells in peripheral blood, Panel B shows %engraftment of the red blood cell compartment by HPLC, Panel C shows VCNs seen in peripheral blood, Panel D shows %Hb BAS3/Total Hb expression seen in perihpheral blood by HPLC, Pandel E shows %HB BAS3/Total Hb normalized to VCN.

DETAILED DESCRIPTION [0130] It is believed that autologous stem cell gene therapy for sickle cell disease (SCD) or other hemoglobinopathies (e.g., β-thallasemia, etc.) has the potential to treat these illnesses without the need for immune suppression of current allogeneic hematopoietic stem cell transplantation (HSCT) approaches. In particular, it is believed that autologous stem cell gene therapy that introduces, for example, anti-sickling human beta globin into hematopoietic cells (or progenitors thereof) can provide effective therapy for SCD (including, for example, normalized red blood cell (RBC) physiology and prevention of the manifestations of SCD) or certain other hemoglobinopathies.

[0131] Current β-globin expression vectors, however, suffer from low vector titer and sub-optimal gene transfer to hematopoietic stem cells, representing a major barrier toward the effective implementation of this gene therapy strategy to the clinic. Without being bound to a particular theory, it is believed that the predominant factor most likely affecting vector performance is overall vector length.

[0132] One solution to reducing LCR size has been to use protein binding and histone marking data to redefine boundaries of HS fragments and reduce size. This problem suffers from two defects: 1) This approach may fail to identify enhancer sequences due for example to transient protein binding or histone marking; and 2) This approach may over or under estimate enhancer boundaries as the identified boundaries reflect the footprint of protein bound to DNA during immunoprecipitation.

[0133] Our solution has been to identify “sequence intrinsic” enhancers (actual DNA sequences that provide enhancer activity) of LCR in the context of a lentiviral vector. As illustrated in Figure 1, panels A-C, a sliding 100bp window that moves 4bp at a time across large DNA regions in the context of a lentiviral vector was used to assay for enhancer activity. This provides the added benefit of selecting for sequences that are successfully packaged and transferred to target cell. [0134] More specifically, we have harnessed the power of massively parallel automated DNA synthesis and next-generation sequencing to map enhancer activity across large DNA regions (~16kb) in cell types of choice. Our DNA enhancer mapping method can be used to generate novel synthetic enhancers of minimal size to drive tissue specific expression of transgenes. A current limitation to developing tissue-specific enhancers of minimal length is a lack of knowledge regarding the exact boundaries of sequence intrinsic enhancers (the actual DNA sequence that provides enhancer function) in a given cell type. Current technologies such as ChIP-sequencing (ChIP-seq) and its variants, provide vague boundaries of enhancer location based on protein binding and often fail to identify sequence intrinsic enhancers when proteins transiently bind, modify local chromatin structure, and dissociate before they can be fixed in place by DNA-protein crosslinking (a key step in implementing the above-mentioned technologies). [0135] By generating targeted enhancer maps spanning large DNA regions, we can construct streamlined enhancers by concatenating those regions that are (for example) in the 95th, 90th, or 85th percentile of relative enhancer strength. Moreover, this approach is superior to current methods used to identify tissue-specific enhancers as the exact boundaries (+/- 4bp) of enhancer sequences are provided. [0136] We have used this targeted enhancer mapping approach to map sequence intrinsic enhancer function of the endogenous human globin locus region (LCR) in HUDEP2 cells (an erythrocyte-like cell line). The map revealed regions of the LCR unknown to possess enhancer activity and assisted in defining the intrinsic boundaries of the LCR’s known enhancer regions. We then set a 95% cutoff based on enhancer strength and concatenated the strongest enhancer sequences to produce a synthetic “mini- LCR” of minimal length (1.6kb) that possesses sequences from various combinations of the LCR’s HS1, HS2, HS3, HS4, HS5, and intervening sequences.

[0137] To generate a targeted enhancer map, we started by designing an oligonucleotide that contains base pair positions 1-100 derived from the larger (~16kb) DNA sequence being interrogated. The subsequent oligonucleotide contained positions 4-104, the next 8-108, and so on until complete coverage of the larger DNA sequence was achieved (total of ~4e3 oligos). The library was duplicated a total of three times by assigning three unique ~13bp barcodes to each sequence (total of ~1.2e4 oligos) and an antisense library was made doubling the total size of the library (total of ~2.4e4 oligos). Each oligo included ~55bp of “backend” sequence required for downstream cloning into a lentiviral reporter vector (Figure 1, panels A-C). Our library construction strategy allowed for placement of the lOObp “interrogation sequence” upstream of the promoter, and placement of the barcode between the transgene and polyadenylation signal (allowing for expression of the barcode in mRNA).

[0138] After microarray oligonucleotide synthesis, the library was assembled and packaged into lentiviral vector particles and transferred to a cell-line of choice. In our example, we used the erythrocyte-like cell line, HUDEP2. Cells were harvested for RNA and DNA after they are partially differentiated down the erythroid lineage. The RNA was then used to make CDNA (using primers specific to the barcode containing reporter gene transcript), barcodes were then amplified from cDNA and gDNA, and PCR products were submitted for next generation sequencing to facilitate the computational quantification and analysis of barcode reads (Figure 2). The cDNA barcode reads were then normalized to the

DNA barcodes reads to generate a high-resolution map of enhancer activity across the large DNA region being interrogated. A 95% cutoff was then set and those regions displaying the strongest enhancer activity were then concatenated to produce a streamlined synthetic enhancer element (Figure 3). [0139] Our method for generating targeted enhancer maps in a given cell type has facilitated the creation of a synthetic enhancers capable of driving lineage specific expression of a therapeutic transgene. Lentiviral vectors comprising these synthetic enhancers are schematically illustrated in Figure 4, which illustrates four novel constructs (designated vectors A-D) compared to the CCL-BAS3-FB construct. [0140] Figure 5 illustrates vector titer produced by each of these constructs and it is noted that vectors A-D all produce a higher titer than CCL-BAS3-FB. As shown in Figure 6, the average %BAS3/Total/VCN decreased with decreasing enhancer length, indicating that (as expected) as you increase the cut-off of functional DNA sequences to reduce concatenated sequence length you begin to remove some of the sequences required for maximal enhancer function. Figure 7 illustrates the results of primary cell studies. As shown therein, vector B produced higher vector copy number (VCN) and as a result higher %BAS3/Total than the CCL-AS3-FB construct. However the %BAS3/Total/VCN was lower for vector B than CCL-AS3-FB, which is expected as this vector B does not have all of the strong enhancer sequences present in its concantenated LCR sequence. Similarly, Figure 8 shows that gene transfer of vector B is linear, resulting in a linear increase of total expression, and that the expression per VCN is predictable unlike the CCL-AS3-FB contract.

[0141] Figure 9 illustrates the results of an in vivo study using transduced Lin(-) bone marrow cells from an SCD mouse model. Cells were transfected with vector C (97.5) and CCL-BAS3-FB at equal vector doses demonstrating that the findings from in vitro generated data in primary human cells (figure 8) can also be recapitulated in a mouse model of SCD. Vector C displayed superior gene transfer and as a result superior total AS3 expression, however the expression per VCN is reduced as this vector does not contain all of the enhancer sequences that are present in CCL-AS3-FB.

[0142] In view of these and othter data, we have created reduced size synthetic enhancers capable of driving lineage specific (hematopoietic cell specific) expression of a therapeutic transgene (e.g., a transgene for the treatment of hemoglobinopathies). However, in certain emdobiments, the synthetic enhancers can also be redefined sequences, as well as sequences not included or larger than those initially identified.

[0143] Accordingly, in certain embodiments, a recombinant lentiviral vector (LV) is provided where the vector comprises a human β-globin locus control region comprising a plurality of reduced length hypersensitive site (HS) sequences where the nucleic acid sequence of the reduced length hypersensitive site (HS) sequences consists of (or compriss) one or more sequences independently selected from the group consisting of vector A HS1.1, vector B HS1.1, vector C HS1.1, vector D HS1.1, vector E HS1.1, vector A HS1.2, vector B HS1.2, vector C HS1.2, vector A HSU, vector B HSU, vector A HS1.4, vector B HS1.4, vector A HS2.1, vector B HS2.1, vector C HS2.1, vector D HS2.1, vector E HS2.1, vector C HS2.2, vector A HS3.1, vector B HS3.1, vector C HS3.1, vector D HS3.1, vector E HS3.1, vector A HS3.2, vector B HS3.2, vector C HS3.2, vector A HS4.1, vector B HS4.1, vector C HS4.1, vector D HS4.1, vector E HS4.1, vector A HS5.1, vector B HS5.1, vector C HS5.1, vector D HS5.1, and vector E HS5.1 (see, e.g., Figure 4, and Sequence Listing provided herein), or where the reduced length hypersensitive site sequences having at least 90%, or at least 95%, or at least 98%, ro at least 99% sequence identify with the nucleotide sequence of the foregoing reduced length hypersensitive site sequences. In certain embodiments the nucleic acid sequence of said reduced length hypersensitive site (HS) sequences consists of sequences independently selected from the group consisting of vector A HS1.1, vector B HS1.1, vector C HS1.1, vector D HS1.1, vector E HS1.1, vector A HS2.1, vector B HS2.1, vector C HS2.1, vector D HS2.1, vector E HS2.1, vector A HS3.1, vector B HS3.1, vector C HS3.1, vector D HS3.1, vector E HS3.1, vector A HS4.1, vector B HS4.1, vector C HS4.1, vector D HS4.1, vector E HS4.1, vector A HS5.1, vector B HS5.1, vector C HS5.1, vector D HS5.1, and vector E HS5.1. [0144] In certain embodiments the β-globin locus control region comprises (or consists of) reduced length hypersensitive site (HS) sequences:

[0145] consisting of or comprising the sequence of one of vector A HS1.1, vector B HS1.1, vector C HS1.1, vector D HS1.1, or vector E HS1.1; [0146] consisting of or comprising the sequence of one of vector a HS 2.1, vector B HS2.1, vector C HS2.1, vector D HS2.1, or vector E HS2.1;

[0147] consisting of or comprising the sequence of one of vector A HS3.1, vector B HS3.1, vector C HS3.1, vector D HS3.1, or vector E HS3.1;

[0148] consisting of or comprising the sequence of one of vector A HS41., vector B HS4.1, vector C HS4.1, vector D HS4.1, or vector E HS4.1; and

[0149] consisting of or comprising the sequence of one of vector A HS5.1, vector B HS5.1, vector C HS5.1, vector D HS5.1, or vector E HS5.1 (see, e.g., Sequence Listing provided herein).

[0150] In certain embodiments the β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS 1.1 o f vector A. In certain embodiments the β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.1 of vector B. In certain embodiments the β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.1 of vector C. In certain embodiments the β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.1 of vector D. In certain embodiments the β-globin locus control region comprises reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.1 of vector E. [0151] In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS2.1 of vector A. In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS2.1 of vector B. In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS2.1 of vector C. In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS2.1 of vector D. In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS2.1 of vector E.

[0152] In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS3.1 of vector A. In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS3.1 of vector B. In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS3.1 of vector C. In certain embodiments the the β-globin locus control region comprises

(or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS3.1 of vector D. In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS3.1 of vector E. [0153] In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS4.1 of vector A. In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS4.1 of vector B. In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS4.1 of vector C. In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS4.1 of vector D. In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS4.1 of vector E.

[0154] In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS5.1 of vector A. In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS5.1 of vector B. In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS5.1 of vector C. In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS5.1 of vector D. In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS5.1 of vector E.

[0155] In certain embodiments the the β-globin locus control region comprises (or further comprises) a In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.2 of vector A. In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS 1.2 of vector B. In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.2 of vector C. [0156] In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HSU of vector A. In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HSU of vector B. [0157] In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.4 of vector A. In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS1.4 of vector B. [0158] In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS2.2 of vector C.

[0159] In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS3.2 of vector A. In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS3.2 of vector B. [0160] In certain embodiments the the β-globin locus control region comprises (or further comprises) a reduced length hypersensitive site (HS) sequence consisting of or comprising the sequence of HS3.3 of vector C.

[0161] In certain embodiments the β-globin locus control region comprises or consists of reduced length hypersensitive site (HS) sequences: vector A HS1.1, vector A HS1.2, vector A HS1.3, vector A HS1.4, vector A HS2.1, vector A HS3.1, vector A HS3.2, vector A HS4.1, and vector A HS5.1.

[0162] In certain embodiments the β-globin locus control region comprises or consists of reduced length hypersensitive site (HS) sequences: vector B HS1.1, vector B HS1.2, vector B HSU, vector B HS1.4, vector B HS2.1, vector B HS3.1, vector B HS3.2, vector B HS4.1, and vector B HS5.1.

[0163] In certain embodiments the β-globin locus control region comprises or consists of reduced length hypersensitive site (HS) sequences: vector C HS1.1, vector C HS1.2, vector C HS2.1, vector C HS2.2, vector C HS3.1, vector C HS3.2, vector C HS4.1, and vector C HS5.1.

[0164] In certain embodiments the β-globin locus control region comprises or consists of reduced length hypersensitive site (HS) sequences: vector D HS1.1, vector D HS2.1, vector D HS3.1, vector D HS4.1, and vector D HS51.

[0165] In certain embodiments the β-globin locus control region comprises or consists of reduced length hypersensitive site (HS) sequences: vector E HS1.1, vector E HS2.1, vector E HS3.1, vector E HS 4.1, and vector E HS 51.

[0166] In certain embodiments the reduced length hypersensitive site (HS) sequences are concatenated in order of increasing HS number: HS1.1 (when present) - HS 1.2 (when present) - HS1.3 (when present) - HS1.4 (when present - HS2 (when present) - HS2.1 (when present) - HS 2.2 (when present) - HS 3.1 (when present) - HS3.2 (when present) - HS4.1 (when present) - HS5.1 (when present).

[0167] In certain embodiments the β-globin locus control region comprises or consists of the LCR sequence of vector A. In certain embodiments the β-globin locus control region comprises or consists of the LCR sequence of vector B. In certain embodiments the β-globin locus control region comprises or consists of the LCR sequence of vector C. In certain embodiments the β-globin locus control region comprises or consists of the LCR sequence of vector D. In certain embodiments the β-globin locus control region comprises or consists of the LCR sequence of vector E.

[0168] In certain embodiments the vector comprises or consists of the nucleic acid sequence of vector A (recognizing that the transgene can be altered, e.g. , can be a wild type globin gene, etc.). In certain embodiments the vector comprises or consists of the nucleic acid sequence of vector B (recognizing that the transgene can be altered, e.g., can be a wild type globin gene, etc.). In certain embodiments the vector comprises or consists of the nucleic acid sequence of vector C (recognizing that the transgene can be altered, e.g., can be a wild type globin gene, etc.). In certain embodiments the vector comprises or consists of the nucleic acid sequence of vector D (recognizing that the transgene can be altered, e.g. , can be a wild type globin gene, etc.). In certain embodiments the vector comprises or consists of the nucleic acid sequence of vector E (recognizing that the transgene can be altered, e.g., can be a wild type globin gene, etc.).

[0169] In view of the foregoing, an improved LV is provided for the introduction of a normal wild-type or an anti-sickling beta globin into stem and progenitor cells {e.g. , hematopoietic stem and progenitor cells) that can then be transplanted into a subject in need thereof {e.g., a subject that has the sickle cell mutation, a subject with β-thalassemia, etc.).

[0170] In various embodiments the improved vectors described herein are capable of driving lineage-restricted expression of an anti-sickling β-globin like gene (βΑ83), a wild- type β-globin gene, or any other heterologous gene it is desired to express. Optimization of the LCR, as described above, with the primary goal of reducing length provides smaller effective vectors with improved ehancer activity.

[0171] Additionally, in certain embodiments, elements can be added to the optimized vectors such as the murine GATA1. In certain embodiments a human Ankyrin insulator (~150 bp) element can be included. These vectors, rationally-designed for reduced sizes of the LCR fragments and added transcriptional enhancing elements are believed to be produced at higher titers than the original β-globin lenti viral vector and have improved gene transfer to human HSC while retaining strong erythroid-specific gene expression. Such improved lentiviral vectors can be effective for gene therapy of hemoglobinopathies such as sickle cell disease and β-thalassemia.

[0172] In certain embodiments, one or more of the reduced HS sequences described herein can be used in combination with a full-length {e.g., wildtype) HS sequence. Thus, for example, in certain embodiments, a reduced length HS2.1 may be used in combination with a reduced length HS3.1 and/or HS3.2, or with a wildtype HS3, or with a reduced length HS4.1 or a wildtype HS4, or with a reduced length HS3.1 and/or HS3.2 and a wildtype HS4, or with a wildtype HS3 and a reduced length HS4.1. In certain embodiments, a reduced length HS3.1 and/or HS3.2 may be used in combination with a reduced length HS2.1 and/or HS2.2, or with a wildtype HS2, or with a reduced length HS4.1 or a wildtype HS4, or with a reduced length HS2.1 and/or HS2.2 and a wildtype HS4, or with a wildtype HS2 and a reduced length HS4.1. Thus, for example, in certain embodiments, a reduced length HS4.1 may be used in combination with a reduced length HS2.1 and/or HS2.2, or with a wildtype HS2, or with a reduced length HS3.1 and/or HS3.2 or a wildtype HS3, or with a reduced length HS2.1 nad/or HS2.2 and a wildtype HS3, or with a wildtype HS2 and a reduced length HS3. In certain embodiments, particularly where a mGATAlq-HS2 and/or human ankyrin insulator element is present, the hypersensitive sites can comprise or consist of a wildtype HS3 and a wildtype HS2. The foregoing combinations of reduce enhancer regions are illustrative and non-limiting. Thus, for example, any of the fragments described herein can either be reduced or redefined (i.e., of a similar size to the original butconsisting of a non contigious concatenated sequence). Using the teaching sprovided herein, numerous other reduced LCR regions are available to one of skill in the art.

[0173] In certain embodiments the human β -globin gene in the vectors contemplated herein comprises an anti-sickling human β -globin gene encoding an anti-sickling β -globin polypeptide. In certain embodiments the anti-sickling version of a human β -globin gene used in the vector comprises one, two, or three mutations selected from the group consisting of GlylbAsp, Glu22Ala and Thr87Gln (see, e.g., Levasseur (2004) J. Biol. Chem. 279(26): 27518-27524). Without being bound to a particular theory, it is believed the Glu22Ala mutation increases affinity to a-chain, the Thr87Gln mutation blocks lateral contact with Val6 of β8 protein, and the Glyl6Asp mutation decreases axial contact between globin chains.

[0174] In certain embodiments the vectors described herein comprise a human

Ankyrin insulator element

[0175] In certain embodiments the vectors described herein comprise a murine GATA1-HS2.

[0176] In various embodiments, the LVs described herein can have additional safety features that can include, for example, the presence of an insulator (e.g., an FB insulator in the 3’LTR). Additionally, or alternatively, in certain embodiments, the HIV LTR has been substituted with an alternative promoter (e.g. , a CMV) to yield a higher titer vector without the inclusion of the HIV TAT protein during packaging. Other strong promoters (e.g., RSV, and the like can also be used).

[0177] In certain embodiments the vectors contemplated herein include the various elements shown in the vectors (vectors A, B, C, D, or E) illustrated in Figure 4.

[0178] As shown above, the vectors described herein are effective to transduce cells at high titer and to also provide high levels of expression.

[0179] In view of these results, it is believed that LVs described herein, e.g., recombinant TAT-independent, SIN LVs that express a human beta-globin gene can be used to effectively treat hemoglobinopathies in subjects (e.g. , human and non-human mammals). Such hemoglobinopathies include, but are not limited to sickle cell disease (SCD) and β- thalassemia. It is believed these vectors can be used for the modification of stem cells (e.g., hematopoietic stem and progenitor cells) that can be introduced into a subject in need thereof for the treatment of, e.g., SCD or β-thalassemia. Moreover, it appears that the resulting cells will produce enough of the transgenic β-globin protein to demonstrate significant improvement in subject health. It is also believed the vectors can be directly administered to a subject to achieve in vivo transduction of the target (e.g., hematopoietic stem or progenitor cells) and thereby also effect a treatment of subjects in need thereof.

[0180] As noted above, in various embodiments the LVs described herein can comprise various safety features. For example, the HIV LTR has been substituted with a CMV promoter to yield higher titer vector without the inclusion of the HIV TAT protein during packaging. In certain embodiments an insulator (e.g., the FB insulator) is introduced into the 3’LTR for safety. The LVs are also constructed to provide efficient transduction and high titer. [0181] It will be appreciated that the foregoing elements are illustrative and need not be limiting. In view of the teachings provided herein, suitable substitutions for these elements will be recognized by one of skill in the art and are contemplated within the scope of the teachings provided herein.

Antl-slckllng β-glnbin gene and expression cassette, [0182] As indicated above, in various embodiments the LV described herein comprise an expression cassette encoding a wild-type β-globin gene, or an anti-sickling human β- globin gene. On illustrative, but non-limiting cassette is βΑ83 which comprises an ~2.3 kb recombinant human β-globin gene (exons and introns) with three amino acid substitutions (Thr87Gln; Glyl6Asp; and Glu22Ala) under the control of transcriptional control elements (e.g., the human β-globin gene 5’ promoter (e.g., ~266 bp), the human β-globin 3’ enhancer (e.g., ~260 bp ), β-globin intron 2 with a ~375 bp Rsal deletion from IVS2, and a ~3.4 kb composite human β-globin locus control region (e.g., HS2 -1203 bp; HS3 -1213 bp; HS4 -954 bp). One embodiment of a βΑ83 cassette is described by Levasseur (2003) Blood 102: 4312-4319.

[0183] In certain embodiments the β-globin gene comprises a Sspl (S) to Rsal (R) deletion (~220bp), e.g., as described by Antoniou et al. 1998) Nucl. Acids Res., 26(3): 721- 729.

[0184] The βΑ83 cassette, however, is illustrative and need not be limiting. Using the teaching provided herein, numerous variations will be available to one of skill in the art. Such variations include, for example, use of a gene encoding a wild-type β-globin, use of a gene comprising one or two mutations selected from the group consisting of Thr87Gln, Glyl6Asp, and Glu22Ala, and/or further or alternative mutations to the β-globin to further enhance non-sickling properties, alterations in the transcriptional control elements (e.g., promoter and/or enhancer), variations on the intron size/structure, and the like.

TAT -Indeoendent and Self inactivating lentiviral vectors.

[0185] To further improve safety, in various embodiments, the lentiviral vectors described herein comprise a TAT-independent, self-inactivating (SIN) configuration. Thus, in various embodiments it is desirable to employ in the LVs described herein an LTR region that has reduced promoter activity relative to wild-type LTR. Such constructs can be provided that are effectively "self-inactivating" (SIN) which provides a biosafety feature.

SIN vectors are ones in which the production of full-length vector RNA in transduced cells is greatly reduced or abolished altogether. This feature minimizes the risk that replication- competent recombinants (RCRs) will emerge. Furthermore, it reduces the risk that that cellular coding sequences located adjacent to the vector integration site will be aberrantly expressed.

[0186] Furthermore, a SIN design reduces the possibility of interference between the LTR and the promoter that is driving the expression of the transgene. SIN LVs can often permit full activity of the internal promoter. [0187] The SIN design increases the biosafety of the LVs. The majority of the HIV LTR is comprised of the U3 sequences. The U3 region contains the enhancer and promoter elements that modulate basal and induced expression of the HIV genome in infected cells and in response to cell activation. Several of these promoter elements are essential for viral replication. Some of the enhancer elements are highly conserved among viral isolates and have been implicated as critical virulence factors in viral pathogenesis. The enhancer elements may act to influence replication rates in the different cellular target of the virus

[0188] As viral transcription starts at the 3’ end of the U3 region of the 5’ LTR, those sequences are not part of the viral mRNA and a copy thereof from the 3’ LTR acts as template for the generation of both LTR’s in the integrated provirus. If the 3’ copy of the U3 region is altered in a retroviral vector construct, the vector RNA is still produced from the intact 5’

LTR in producer cells, but cannot be regenerated in target cells. Transduction of such a vector results in the inactivation of both LTR’s in the progeny virus. Thus, the retrovirus is self-inactivating (SIN) and those vectors are known as SIN transfer vectors. [0189] In certain embodiments self-inactivation is achieved through the introduction of a deletion in the U3 region of the 3’ LTR of the vector DNA, i.e. , the DNA used to produce the vector RNA. During RT, this deletion is transferred to the 5’ LTR of the proviral DNA. Typically, it is desirable to eliminate enough of the U3 sequence to greatly diminish or abolish altogether the transcriptional activity of the LTR, thereby greatly diminishing or abolishing the production of full-length vector RNA in transduced cells. However, it is generally desirable to retain those elements of the LTR that are involved in polyadenylation of the viral RNA, a function typically spread out over U3, R and U5. Accordingly, in certain embodiments, it is desirable to eliminate as many of the transcriptionally important motifs from the LTR as possible while sparing the polyadenylation determinants. [0190] The SIN design is described in detail in Zufferey et al. (1998) J Virol. 72(12): 9873-9880, and in U.S. Patent No: 5,994,136. As described therein, there are, however, limits to the extent of the deletion at the 3’ LTR. First, the 5’ end of the U3 region serves another essential function in vector transfer, being required for integration (terminal dinucleotide+att sequence). Thus, the terminal dinucleotide and the att sequence may represent the 5’ boundary of the U3 sequences which can be deleted. In addition, some loosely defined regions may influence the activity of the downstream polyadenylation site in the R region. Excessive deletion of U3 sequence from the 3’LTR may decrease polyadenylation of vector transcripts with adverse consequences both on the titer of the vector in producer cells and the transgene expression in target cells.

[0191] Additional SIN designs are described in U.S. Patent Publication No: 2003/0039636. As described therein, in certain embodiments, the lentiviral sequences removed from the LTRs are replaced with comparable sequences from a non-lenti viral retrovirus, thereby forming hybrid LTRs. In particular, the lentiviral R region within the LTR can be replaced in whole or in part by the R region from a non-lentiviral retrovirus. In certain embodiments, the lentiviral TAR sequence, a sequence which interacts with TAT protein to enhance viral replication, is removed, preferably in whole, from the R region. The TAR sequence is then replaced with a comparable portion of the R region from a non- lentiviral retrovirus, thereby forming a hybrid R region. The LTRs can be further modified to remove and/or replace with non-lentiviral sequences all or a portion of the lentiviral U3 and U5 regions.

[0192] Accordingly, in certain embodiments, the SIN configuration provides a retroviral LTR comprising a hybrid lentiviral R region that lacks all or a portion of its TAR sequence, thereby eliminating any possible activation by TAT, wherein the TAR sequence or portion thereof is replaced by a comparable portion of the R region from a non-lentiviral retrovirus, thereby forming a hybrid R region. In a particular embodiment, the retroviral LTR comprises a hybrid R region, wherein the hybrid R region comprises a portion of the HIV R region (e.g. , a portion comprising or consisting of the nucleotide sequence shown in SEQ ID

NO: 10 in US 2003/0039636) lacking the TAR sequence, and a portion of the MoMSV R region (e.g., a portion comprising or consisting of the nucleotide sequence shown in SEQ ID NO: 9 in 2003/0039636) comparable to the TAR sequence lacking from the HIV R region.

In another particular embodiment, the entire hybrid R region comprises or consists of the nucleotide sequence shown in SEQ ID NO: 11 in 2003/0039636.

[0193] Suitable lentiviruses from which the R region can be derived include, for example, HIV (HIV-1 and HIV-2), EIV, SIV and FIV. Suitable retro viruses from which non- lentiviral sequences can be derived include, for example, MoMSV, MoMLV, Friend, MSCV, RSV and Spumavimses. In one illustrative embodiment, the lentivirus is HIV and the non- lentiviral retrovirus is MoMSV.

[0194] In another embodiment described in US 2003/0039636, the LTR comprising a hybrid R region is a left (5’) LTR and further comprises a promoter sequence upstream from the hybrid R region. Preferred promoters are non-lentiviral in origin and include, for example, the U3 region from a non-lentiviral retrovirus (e.g., the MoMSV U3 region). In one particular embodiment, the U3 region comprises the nucleotide sequence shown in SEQ ID NO: 12 in US 2003/0039636. In another embodiment, the left (5’) LTR further comprises a lentiviral U5 region downstream from the hybrid R region. In one embodiment, the U5 region is the HIV U5 region including the HIV att site necessary for genomic integration. In another embodiment, the U5 region comprises the nucleotide sequence shown in SEQ ID NO: 13 in US 2003/0039636. In yet another embodiment, the entire left (5’) hybrid LTR comprises the nucleotide sequence shown in SEQ ID NO: 1 in US 2003/0039636.

[0195] In another illustrative embodiment, the LTR comprising a hybrid R region is a right (3’) LTR and further comprises a modified (e.g. , truncated) lentiviral U3 region upstream from the hybrid R region. The modified lentiviral U3 region can include the att sequence, but lack any sequences having promoter activity, thereby causing the vector to be SIN in that viral transcription cannot go beyond the first round of replication following chromosomal integration. In a particular embodiment, the modified lentiviral U3 region upstream from the hybrid R region consists of the 3’ end of a lentiviral (e.g. , HIV) U3 region up to and including the lentiviral U3 att site. In one embodiment, the U3 region comprises the nucleotide sequence shown in SEQ ID NO: 15 in US 2003/0039636. In another embodiment, the right (3’) LTR further comprises a polyadenylation sequence downstream from the hybrid R region. In another embodiment, the polyadenylation sequence comprises the nucleotide sequence shown in SEQ ID NO: 16 in US 2003/0039636. In yet another embodiment, the entire right (5’) LTR comprises the nucleotide sequence shown in SEQ ID NO: 2 or 17 of US 2003/0039636.

[0196] Thus, in the case of HIV based LV, it has been discovered that such vectors tolerate significant U3 deletions, including the removal of the LTR TATA box (e.g., deletions from -418 to -18), without significant reductions in vector titers. These deletions render the LTR region substantially transcriptionally inactive in that the transcriptional ability of the LTR in reduced to about 90% or lower.

[0197] It has also been demonstrated that the trans-acting function of Tat becomes dispensable if part of the upstream LTR in the transfer vector construct is replaced by constitutively active promoter sequences (see, e.g., Dull et al. (1998) J Virol. 72(11): 8463- 8471. Furthermore, we show that the expression of rev in trans allows the production of high-titer HIV-derived vector stocks from a packaging construct which contains only gag and pol. This design makes the expression of the packaging functions conditional on complementation available only in producer cells. The resulting gene delivery system, conserves only three of the nine genes of HIV-1 and relies on four separate transcriptional units for the production of transducing particles.

[0198] In one embodiments illustrated in Example 1, the cassette expressing an anti- sickling β-globin (e.g., βΑ83) is placed in the pCCL LV backbone, which is a SIN vector with the CMV enhancer/promoter substituted in the 5' LTR.

[0199] It will be recognized that the CMV promoter typically provides a high level of non-tissue specific expression. Other promoters with similar constitutive activity include, but are not limited to the RSV promoter, and the SV40 promoter. Mammalian promoters such as the beta-actin promoter, ubiquitin C promoter, elongation factor 1 apromoter, tubulin promoter, etc. , may also be used.

[0200] The foregoing SIN configurations are illustrative and non-limiting. Numerous SIN configurations are known to those of skill in the art. As indicated above, in certain embodiments, the LTR transcription is reduced by about 95% to about 99%. In certain embodiments LTR may be rendered at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95% at least about 96%, at least about 97%, at least about 98%, or at least about 99% transcriptionally inactive.

Insulator element

[0201] In certain embodiments, to further enhance biosafety, insulators are inserted into the lentiviral vectors described herein. Insulators are DNA sequence elements present throughout the genome. They bind proteins that modify chromatin and alter regional gene expression. The placement of insulators in the vectors described herein offer various potential benefits including, inter alia : 1) Shielding of the vector from positional effect variegation of expression by flanking chromosomes (i.e., barrier activity); and 2) Shielding flanking chromosomes from insertional trans- activation of gene expression by the vector

(enhancer blocking). Thus, insulators can help to preserve the independent function of genes or transcription units embedded in a genome or genetic context in which their expression may otherwise be influenced by regulatory signals within the genome or genetic context (see, e.g. , Burgess-Beusse el al. (2002) Proc. Natl. Acad. Sci. USA, 99: 16433; and Zhan el al. (2001) Hum. Genet., 109: 471). In the present context insulators may contribute to protecting lenti virus -expressed sequences from integration site effects, which may be mediated by cis- acting elements present in genomic DNA and lead to deregulated expression of transferred sequences. In various embodiments LVs are provided in which an insulator sequence is inserted into one or both LTRs or elsewhere in the region of the vector that integrates into the cellular genome.

[0202] The first and best characterized vertebrate chromatin insulator is located within the chicken β-globin locus control region. This element, which contains a DNase-I hypersensitive site-4 (cHS4), appears to constitute the 5' boundary of the chicken β-globin locus (Rrioleau et al. (1999) EMBO J. 18: 4035-4048). A 1.2-kb fragment containing the cHS4 element displays classic insulator activities, including the ability to block the interaction of globin gene promoters and enhancers in cell lines (Chung et al. (1993) Cell, 74: 505-514), and the ability to protect expression cassettes in Drosophila (Id.), transformed cell lines (Rikaart et al. (1998) Genes Dev. 12: 2852-2862), and transgenic mammals (Wang et al. (1997) Nat. Biotechnol, 15: 239-243; Taboit-Dameron et al. (1999) Transgenic Res., 8: 223- 235) from position effects. Much of this activity is contained in a 250-bp fragment. Within this stretch is a 49-bp cHS4 core (Chung et al. (1997) Proc. Natl. Acad. Sci., USA, 94: 575- 580) that interacts with the zinc finger DNA binding protein CTCF implicated in enhancer- blocking assays (Bell et al. (1999) Cell, 98: 387-396).

[0203] One illustrative and suitable insulator is FB (FII/BEAD-A), a 77 bp insulator element, that contains the minimal CTCF binding site enhancer-blocking components of the chicken β-globin 5’ HS4 insulators and a homologous region from the human T-cell receptor alpha/delta blocking element alpha/delta I (BEAD-I) insulator described by Ramezani et al. (2008) Stem Cell 26: 3257-3266. The FB “synthetic” insulator has full enhancer blocking activity. This insulator is illustrative and non-limiting. Other suitable insulators may be used including, for example, the full-length chicken beta-globin HS4 or insulator sub-fragments thereof, the ankyrin gene insulator, and other synthetic insulator elements. [0204] In various embodiments the vectors described herein further comprise a packaging signal. A "packaging signal," "packaging sequence," or "psi sequence" is any nucleic acid sequence sufficient to direct packaging of a nucleic acid whose sequence comprises the packaging signal into a retroviral particle. The term includes naturally occurring packaging sequences and also engineered variants thereof. Packaging signals of a number of different retroviruses, including lentiviruses, are known in the art. Rev Responsive Element (RRE).

[0205] In certain embodiments the lentiviral vectors described herein comprise a Rev response element (RRE) to enhance nuclear export of unspliced RNA. RREs are well known to those of skill in the art. Illustrative RREs include, but are not limited to RREs such as that located at positions 7622-8459 in the HIV NL4-3 genome (Genbank accession number

AF003887) as well as RREs from other strains of HIV or other retroviruses. Such sequences are readily available from Genbank or from the database with URL hiv- web.lanl.gov/content/index.

Central PolvPurine Tract (cPPT). [0206] In various embodiments the lentiviral vectors described herein further include a central polypurine tract. Insertion of a fragment containing the central polypurine tract (cPPT) in lentiviral (e.g., HIV-1) vector constructs is known to enhance transduction efficiency drastically, reportedly by facilitating the nuclear import of viral cDNA through a central DNA flap. Expression-Stimulating Posttranscriptional Regulatory Element (PRE)

[0207] In certain embodiments the lentiviral vectors (LVs) described herein may comprise any of a variety of posttranscriptional regulatory elements (PREs) whose presence within a transcript increases expression of the heterologous nucleic acid (e.g., βΑ83) at the protein level. PREs may be particularly useful in certain embodiments, especially those that involve lentiviral constructs with modest promoters.

[0208] One type of PRE is an intron positioned within the expression cassette, which can stimulate gene expression. However, introns can be spliced out during the life cycle events of a lentivirus. Hence, if introns are used as PRE’s they are typically placed in an opposite orientation to the vector genomic transcript. [0209] Posttranscriptional regulatory elements that do not rely on splicing events offer the advantage of not being removed during the viral life cycle. Some examples are the posttranscriptional processing element of herpes simplex vims, the posttranscriptional regulatory element of the hepatitis B vims (HPRE) and the woodchuck hepatitis virus (WPRE). Of these the WPRE is typically preferred as it contains an additional cis-acting element not found in the HPRE. This regulatory element is typically positioned within the vector so as to be included in the RNA transcript of the transgene, but outside of stop codon of the transgene translational unit. [0210] The WPRE is characterized and described in U.S. Pat. No: 6,136,597. As described therein, the WPRE is an RNA export element that mediates efficient transport of RNA from the nucleus to the cytoplasm. It enhances the expression of transgenes by insertion of a ris-acting nucleic acid sequence, such that the element and the transgene are contained within a single transcript. Presence of the WPRE in the sense orientation was shown to increase transgene expression by up to 7- to 10-fold. Retroviral vectors transfer sequences in the form of cDNAs instead of complete intron-containing genes as introns are generally spliced out during the sequence of events leading to the formation of the retroviral particle. Introns mediate the interaction of primary transcripts with the splicing machinery. Because the processing of RNAs by the splicing machinery facilitates their cytoplasmic export, due to a coupling between the splicing and transport machineries, cDNAs are often inefficiently expressed. Thus, the inclusion of the WPRE in a vector results in enhanced expression of transgenes.

Transduced Host Cells and Methods of cell transduction. [0211] The recombinant lentiviral vectors (LV) and resulting virus described herein are capable of transferring a heterologous nucleic acid (e.g., a nucleic acid encoding an anti- sickling β-globin) sequence into a mammalian cell. In various embodiments, for delivery to cells, vectors described herein are preferably used in conjunction with a suitable packaging cell line or co-transfected into cells in vitro along with other vector plasmids containing the necessary retroviral genes (e.g. , gag and pol) to form replication incompetent virions capable of packaging the vectors of the present invention and infecting cells.

[0212] The recombinant LVs and resulting virus described herein are capable of transferring a nucleic acid (e.g., a nucleic acid encoding an anti-sickling β-globin or other sequence) into a mammalian cell. For delivery to cells, various vectors described herein are preferably used in conjunction with a suitable packaging cell line or co-transfected into cells in vitro along with other vector plasmids containing the necessary retroviral genes (e.g. , gag and pol) to form replication incompetent virions capable of packaging the vectors of the present invention and infecting cells.

[0213] In certain embodiments the vectors are introduced via transfection into the packaging cell line. The packaging cell line produces viral particles that contain the vector genome. Methods for transfection are well known by those of skill in the art. After cotransfection of the packaging vectors and the transfer vector to the packaging cell line, the recombinant virus is recovered from the culture media and titered by standard methods used by those of skill in the art. Thus, the packaging constructs can be introduced into human cell lines by calcium phosphate transfection, lipofection or electroporation, generally together with or without a dominant selectable marker, such as neomycin, DHFR, Glutamine synthetase, followed by selection in the presence of the appropriate drug and isolation of clones. In certain embodiments the selectable marker gene can be linked physically to the packaging genes in the construct.

[0214] Stable cell lines wherein the packaging functions are configured to be expressed by a suitable packaging cell are known (see, e.g., U.S. Patent No. 5,686,279, which describes packaging cells). In general, for the production of virus particles, one may employ any cell that is compatible with the expression of lentiviral Gag and Pol genes, or any cell that can be engineered to support such expression. For example, producer cells such as 293T cells and HT1080 cells may be used.

[0215] The packaging cells with a lentiviral vector incorporated therein form producer cells. Producer cells are thus cells or cell-lines that can produce or release packaged infectious viral particles carrying the therapeutic gene of interest (e.g. , modified β-globin). These cells can further be anchorage dependent which means that these cells will grow, survive, or maintain function optimally when attached to a surface such as glass or plastic. Some examples of anchorage dependent cell lines used as lentiviral vector packaging cell lines when the vector is replication competent are HeLa or 293 cells and PERC.6 cells. [0216] Accordingly, in certain embodiments, methods are provided of delivering a gene to a cell which is then integrated into the genome of the cell, comprising contacting the cell with a virion containing a lentiviral vector described herein. The cell (e.g. , in the form of tissue or an organ) can be contacted (e.g. , infected) with the virion ex vivo and then delivered to a subject (e.g. , a mammal, animal or human) in which the gene (e.g., anti-sickling β- globin) will be expressed. In various embodiments the cell can be autologous to the subject (i.e., from the subject) or it can be non-autologous (i.e., allogeneic or xenogenic) to the subject. Moreover, because the vectors described herein are capable of being delivered to both dividing and non-dividing cells, the cells can be from a wide variety including, for example, bone marrow cells, mesenchymal stem cells (e.g., obtained from adipose tissue), and other primary cells derived from human and animal sources. Alternatively, the virion can be directly administered in vivo to a subject or a localized area of a subject (e.g., bone marrow). [0217] Of course, as noted above, the lentivectors described herein will be particularly useful in the transduction of human hematopoietic progenitor cells or a hematopoietic stem cells, obtained either from the bone marrow, the peripheral blood or the umbilical cord blood, as well as in the transduction of a CD4 + T cell, a peripheral blood B or T lymphocyte cell, and the like. In certain embodiments particularly preferred targets are CD34 + hematopoetic stem and progenitor cells.

Gene therapy.

[0218] In still other embodiments, methods are provide for transducing a human hematopoietic stem cell. In certain embodiments the methods involve contacting a population of human cells that include hematopoietic stem cells with one of the foregoing lentivectors under conditions to effect the transduction of a human hematopoietic progenitor cell in said population by the vector. The stem cells may be transduced in vivo or in vitro, depending on the ultimate application. Even in the context of human gene therapy, such as gene therapy of human stem cells, one may transduce the stem cell in vivo or, alternatively, transduce in vitro followed by infusion of the transduced stem cell into a human subject. In one aspect of this embodiment, the human stem cell can be removed from a human, e.g., a human patient, using methods well known to those of skill in the art and transduced as noted above. The transduced stem cells are then reintroduced into the same or a different human.

Stem cell/progenitor cell gate therapy. [0219] In various embodiments the lentivectors described herein are particularly useful for the transduction of human hematopoietic progenitor cells or haematopoietic stem cells (HSCs), obtained either from the bone marrow, the peripheral blood or the umbilical cord blood, as well as in the transduction of a CD4 + T cell, a peripheral blood B or T lymphocyte cell, and the like. In certain embodiments particularly preferred targets are CD34 + hematopoietic stem and progenitor cells.

[0220] When cells, for instance CD34 + cells, dendritic cells, peripheral blood cells or tumor cells are transduced ex vivo, the vector particles are incubated with the cells using a dose generally in the order of between 1 to 50 multiplicities of infection (MOI) which also corresponds to 1 x 10 5 to 50 x 10 5 transducing units of the viral vector per 10 5 cells. This can include amounts of vector corresponding to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, and 50 MOI. Typically, the amount of vector may be expressed in terms of HT-29 transducing units (TU). [0221] In certain embodiments cell-based therapies involve providing stem cells and/or hematopoietic precursors, transduce the cells with the lentivirus encoding, e.g., an anti-sickling human β-globin, and then introduce the transformed cells into a subject in need thereof (e.g., a subject with the sickle cell mutation). [0222] In certain embodiments the methods involve isolating population of cells, e.g., stem cells from a subject, optionally expand the cells in tissue culture, and administer the lentiviral vector whose presence within a cell results in production of an anti-sickling β- globin in the cells in vitro. The cells are then returned to the subject, where, for example, they may provide a population of red blood cells that produce the anti-sickling β globin. [0223] In some illustrative, but non-limiting, embodiments, a population of cells, which may be cells from a cell line or from an individual other than the subject, can be used. Methods of isolating stem cells, immune system cells, etc., from a subject and returning them to the subject are well known in the art. Such methods are used, e.g. , for bone marrow transplant, peripheral blood stem cell transplant, etc. , in patients undergoing chemotherapy. [0224] Where stem cells are to be used, it will be recognized that such cells can be derived from a number of sources including bone marrow (BM), cord blood (CB), mobilized peripheral blood stem cells (mPBSC), and the like. In certain embodiments the use of induced pluripotent stem cells (IPSCs) is contemplated. Methods of isolating hematopoietic stem cells (HSCs), transducing such cells and introducing them into a mammalian subject are well known to those of skill in the art.

[0225] In certain embodiments a lentiviral vector described herein (see, e.g., Figure 4) is used in stem cell gene therapy for SCD by introducing the βΑ83 anti-sickling β-globin gene into the bone marrow stem cells of patients with sickle cell disease followed by autologous transplantation.

Direct introduction of vector.

[0226] In certain embodiments direct treatment of a subject by direct introduction of the vector(s) described herein is contemplated. The lentiviral compositions may be formulated for delivery by any available route including, but not limited to parenteral (e.g., intravenous), intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), transmucosal, rectal, and vaginal. Commonly used routes of delivery include inhalation, parenteral, and transmucosal. [0227] In various embodiments pharmaceutical compositions can include an LV in combination with a pharmaceutically acceptable carrier. As used herein the language "pharmaceutically acceptable carrier" includes solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. Supplementary active compounds can also be incorporated into the compositions.

[0228] In some embodiments, active agents, i.e., a lentiviral described herein and/or other agents to be administered together the vector, are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such compositions will be apparent to those skilled in the art. Suitable materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc. Liposomes can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No.

4,522,811. In some embodiments the composition is targeted to particular cell types or to cells that are infected by a virus. For example, compositions can be targeted using monoclonal antibodies to cell surface markers, e.g., endogenous markers or viral antigens expressed on the surface of infected cells.

[0229] It is advantageous to formulate compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit comprising a predetermined quantity of a LV calculated to produce the desired therapeutic effect in association with a pharmaceutical carrier.

[0230] A unit dose need not be administered as a single injection but may comprise continuous infusion over a set period of time. Unit dose of the LV described herein may conveniently be described in terms of transducing units (T.U.) of lentivector, as defined by titering the vector on a cell line such as HeLa or 293. In certain embodiments unit doses can range from 10 3 , 10 4 , 10 5 , 10 6 , 10 7 , 10 8 , 10 9 , 10 10 , 10 11 , 10 12 , 10 13 T.U. and higher.

[0231] Pharmaceutical compositions can be administered at various intervals and over different periods of time as required, e.g., one time per week for between about 1 to about 10 weeks; between about 2 to about 8 weeks; between about 3 to about 7 weeks; about 4 weeks; about 5 weeks; about 6 weeks, etc. It may be necessary to administer the therapeutic composition on an indefinite basis. The skilled artisan will appreciate that certain factors can influence the dosage and timing required to effectively treat a subject, including but not limited to the severity of the disease or disorder, previous treatments, the general health and/or age of the subject, and other diseases present. Treatment of a subject with a LV can include a single treatment or, in many cases, can include a series of treatments.

[0232] Illustrative, but non-limiting, doses for administration of gene therapy vectors and methods for determining suitable doses are known in the art. It is furthermore understood that appropriate doses of a LV may depend upon the particular recipient and the mode of administration. The appropriate dose level for any particular subject may depend upon a variety of factors including the age, body weight, general health, gender, and diet of the subject, the time of administration, the route of administration, the rate: of excretion, other administered therapeutic agents, and the like.

[0233] In certain embodiments lentiviral gene therapy vectors described herein can be delivered to a subject by, for example, intravenous injection, local administration, or by stereotactic injection (see, e.g., Chen et al. (1994) Proc. Natl. Acad. Sci. USA, 91: 3054). In certain embodiments vectors may be delivered orally or inhalationally and may be encapsulated or otherwise manipulated to protect them from degradation, enhance uptake into tissues or cells, etc. Pharmaceutical preparations can include a LV in an acceptable diluent, or can comprise a slow release matrix in which a LV is imbedded. Alternatively or additionally, where a vector can be produced intact from recombinant cells, as is the case for retroviral or lentiviral vectors as described herein, a pharmaceutical preparation can include one or more cells which produce vectors. Pharmaceutical compositions comprising a LV described herein can be included in a container, pack, or dispenser, optionally together with instructions for administration.

[0234] The foregoing compositions, methods and uses are intended to be illustrative and not limiting. Using the teachings provided herein other variations on the compositions, methods and uses will be readily available to one of skill in the art.

[0235] The approach to generate reduced length enhance regions is superior to previous strategies for generating tissue-specific enhancers for, among other reasons: 1) The cost of goods is decreased due to a low number of outputs required to be tested, 2) Strength of synthetic enhancers may be superior to those produced with current methods, or they may be less active but more suitable for LV-mediated delivery, and 3). Enhancers can be of minimal length.

[0236] Additionally, without being bound to a particular theory, it is believed the enhancer mapping strategy described herein can be modified to generate genome-wide enhancer maps using a similar cloning strategy and sonicated human genomic DNA and that the mapping strategies can be used to generate synthetic enhancers responsive to an array of distinct cellular perturbations.

[0237] It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims. All publications, patents, and patent applications cited herein are hereby incorporated by reference in their entirety for all purposes.