Viral Vectors for Prophylaxis and Therapy of Hemoglobinopathies

Rivella; Stefano ;   et al.

Patent Application Summary

U.S. patent application number 17/694841 was filed with the patent office on 2022-06-30 for viral vectors for prophylaxis and therapy of hemoglobinopathies. The applicant listed for this patent is The Children's Hospital of Philadelphia, CORNELL UNIVERSITY. Invention is credited to Gerd Blobel, Laura Breda, Wulan Deng, Alisa Dong, Stefano Rivella.

Application Number20220202958 17/694841
Document ID /
Family ID1000006207789
Filed Date2022-06-30

United States Patent Application 20220202958
Kind Code A1
Rivella; Stefano ;   et al. June 30, 2022

Viral Vectors for Prophylaxis and Therapy of Hemoglobinopathies

Abstract

Provided are compositions and methods for inducing expression of human beta-globin in erythrocytes for use in prophylaxis and/or therapy of a hemoglobinopathy in an individual. The method generally entails introducing into CD34+ cells a polynucleotide encoding: i) a 5' long terminal repeat (LTR) and a self-inactivating 3' LTR; ii) at least one polyadenylation signal; iii) at least one promoter; iv) a globin gene locus control region (LCR); v) an ankyrin insulator element (Ank); vi) a Woodchuck Post-Regulatory Element (WPRE) configured such that the WPRE does not integrate into a target genome; and vii) a sequence that is a reverse complement of a sequence encoding human beta-globin, and can include beta-globin that has a .beta.T87Q mutation. Intron 2 of the beta globin gene can be a complete intron. Modified erythrocyte progenitor cells, recombinant vectors and virions comprising recombinant polynucleotides, and methods of making the vectors and virions are included.


Inventors: Rivella; Stefano; (New York, NY) ; Breda; Laura; (New York, NY) ; Dong; Alisa; (New York, NY) ; Blobel; Gerd; (Bala Cynwyd, PA) ; Deng; Wulan; (Ashburn, VA)
Applicant:
Name City State Country Type

CORNELL UNIVERSITY
The Children's Hospital of Philadelphia

Ithaca
Philadelphia

NY
PA

US
US
Family ID: 1000006207789
Appl. No.: 17/694841
Filed: March 15, 2022

Related U.S. Patent Documents

Application Number Filing Date Patent Number
15545159 Jul 20, 2017 11311632
PCT/US16/14269 Jan 21, 2016
17694841
62105829 Jan 21, 2015

Current U.S. Class: 1/1
Current CPC Class: C12N 2830/50 20130101; C12N 15/867 20130101; C12N 2740/15043 20130101; A61K 48/005 20130101; C12N 2830/40 20130101; C12N 2830/52 20130101; C12N 2830/48 20130101; C12N 15/86 20130101
International Class: A61K 48/00 20060101 A61K048/00; C12N 15/86 20060101 C12N015/86; C12N 15/867 20060101 C12N015/867

Goverment Interests



STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH

[0002] This invention was made with government support under contract no. 1R01HL102449 awarded by the National Institutes of Health. The government has certain rights in the invention.
Claims



1. A method for inducing expression of human beta-globin in erythrocytes for use in prophylaxis and/or therapy of a hemoglobinopathy in an individual comprising introducing into erythrocyte progenitor cells a polynucleotide encoding: i) a 5' long terminal repeat (LTR) and a self-inactivating 3' LTR; ii) at least one polyadenylation signal; iii) at least one promoter; iv) a globin gene locus control region (LCR); v) an ankyrin insulator element (Ank); vi) a Woodchuck Post-Regulatory Element (WPRE) configured such that the WPRE does not integrate into a target genome; and vii) a sequence that is a reverse complement of a sequence encoding modified human beta-globin comprising a (3T87Q mutation (B-globinM), wherein the sequence encoding the B-globinM comprises a first intron (intron 1) between exon 1 and exon 2, and a second intron (intron 2) between exon 2 and exon 3 of said B-globinM sequence, wherein intron 2 comprises more than 476 nucleotides of human B-globinM intron 2 sequence; wherein, subsequent to the introducing, the erythrocyte progenitor cells differentiate into erythrocytes, wherein the erythrocytes are present in the individual and the erythrocytes produce more human beta-globin than a control.

2. The method of claim 1, wherein the control comprises a human beta-globin value obtained from control cells, wherein the control cells comprise erythrocytes from an individual who has the hemoglobinopathy, wherein the erythrocytes are progeny of cells into which a control viral vector is introduced, the control viral vector comprising the 5' LTR, the 3' LTR, the at least one polyadenylation signal, the at least one promoter, the LCR, the Ank, a WPRE, and the sequence encoding the B-globinM, but wherein the sequence encoding the B-globinM in the control viral vector comprises 476 or fewer nucleotides of the human B-globinM intron 2 sequence.

3. The method of claim 1, wherein the lentiviral vector further comprise a sequence encoding a fusion of an Ldb1 transcription factor and a zinc finger (ZF) domain.

4. The method of claim 1, wherein the lentiviral vector further comprises a sequence encoding an RNA polynucleotide that is a reverse complement of mRNA encoding transferrin receptor 1, and wherein the RNA polynucleotide comprises a microRNA or an shRNA sequence and is capable of decreasing transferrin receptor 1 mRNA in an RNAi-mediated process.

5. The method of claim 1, wherein the erythrocyte progenitor cells comprise CD34+ cells, wherein the CD34+ cells are separated from the individual before the introducing the lentiviral vector to the cells, and wherein the CD34+ cells are introduced into the individual subsequent to the introducing of the lentiviral vector.

6. The method of claim 1, wherein the intron 2 comprises 851 nucleotides of the adult human B-globinM intron 2 sequence.

7. The method of claim 1, wherein the increased human beta-globin comprises adult hemoglobin, fetal hemoglobin, B-globinM, or a combination thereof.

8. The method of claim 7, wherein the hemoglobinopathy comprises sickle-cell anemia (SCA) or beta-thalassemia.

9. A lentiviral vector for inducing expression of human beta-globin in erythrocytes and/or erythrocyte progenitor cells for use in prophylaxis and/or therapy of a hemoglobinopathy in an individual, the lentiviral vector comprising: i) a 5' long terminal repeat (LTR) and a self-inactivating 3' LTR; ii) at least one polyadenylation signal; iii) at least one promoter; iv) a globin gene locus control region (LCR); v) an ankyrin insulator element (Ank); vi) a Woodchuck Post-Regulatory Element (WPRE) configured such that the WPRE does not integrate into a target genome; vii) a sequence that is a reverse complement of a sequence encoding modified adult human beta-globin comprising a .beta.T87Q mutation (B-globinM), wherein the sequence encoding the B-globinM comprises a first intron (intron 1) between exon 1 and exon 2, and a second intron (intron 2) between exon 2 and exon 3 of said B-globinM sequence, wherein intron 2 comprises more than 476 nucleotides of adult human B-globinM intron 2 sequence.

10. The lentiviral vector of claim 9, wherein the lentiviral vector further comprise a sequence encoding a fusion of an Ldb1 transcription factor and a zinc finger (ZF) domain.

11. The lentiviral vector of claim 9, wherein the lentiviral vector further comprises a sequence encoding a RNA polynucleotide that a reverse complement of mRNA encoding transferrin receptor 1, and wherein the RNA polynucleotide comprises a microRNA or a shRNA sequence and is capable of decreasing transferrin receptor 1 mRNA in a RNAi-mediated process.

12. The lentiviral vector claim 9, wherein the lentiviral vector is present in CD34+ cells, wherein the CD34+ cells have been separated from an individual who has a hemoglobinopathy.

13. The lentiviral vector of claim 12, wherein the lentiviral vector is present in a virion.

14. A pharmaceutical composition comprising viral particles, wherein the viral particles each comprise a RNA strand which comprises a lentiviral vector of claim 9.

15. A method of making a viral particle preparation for use in prophylaxis and/or therapy for one or more hemoglobinopathies comprising introducing a plasmid encoding a lentiviral vector of claim 1 into packaging cells which comprise a DNA packaging plasmid which encodes at least one virion protein, and a DNA envelope plasmid which encodes an envelope protein, and allowing expression of the virion protein and the envelope protein such that viral particles form, and separating the viral particles from the packaging cells.
Description



CROSS REFERENCE TO RELATED APPLICATIONS

[0001] This application is a divisional application of U.S. patent application Ser. No. 15/545,159, filed Jul. 20, 2017, which is a .sctn. 371 application of PCT/US2016/014269, filed Jan. 21, 2016, which claims priority to U.S. Provisional application No. 62/105,829, filed on Jan. 21, 2015. The foregoing applications are incorporated by reference herein.

FIELD

[0003] The present disclosure related to compositions and methods useful for prophylaxis and/or therapy of hemoglobinopathies.

BACKGROUND

[0004] There are a variety of hemoglobinopathies that affect large portions of the human population. For example, sickle-cell anemia (SCA) is a blood related disorder that affects the structure of the hemoglobin molecules. In SCA the hemoglobin molecule is defective, causing the entire blood cell to change shape (Steinberg, M. H., Forget, B. G., Higgs, D. R. & Nagel, R. L. Disorders of hemoglobin: Genetics, Pathophysiology and Clinical Management, (Cambridge University Press, Cambridge, UK, 2001)). After these abnormal hemoglobin molecules release oxygen in circulation, they may cluster together and form long, rod-like structures, which become rigid and assume sickle shape. Unlike healthy red blood cells, which are usually smooth and donut-shaped, sickled red blood cells cannot squeeze through small blood vessels. Instead, they stack up and cause blockages that deprive organs and tissues of oxygen-carrying blood. This produces periodic episodes of pain and ultimately can damage tissues and vital organs and lead to other serious medical problems. Normal red blood cells live about 120 days in the bloodstream, but sickled red cells die after about 10 to 20 days. Because they cannot be replaced fast enough, the blood is chronically short of red blood cells, leading to anemia.

[0005] SCA affects millions throughout the world. It is particularly common among people whose ancestors come from Sub-Saharan Africa, South America, Cuba, Central America, Saudi Arabia, India, and Mediterranean countries such as Turkey, Greece, and Italy. In the Unites States, it affects around 72,000 people, most of whose ancestors come from Africa. The disease occurs in about 1 in every 500 African-American births and 1 in every 1000 to 1400 Hispanic-American births. About 2 million Americans, or 1 in 12 African Americans, carry the sickle cell allele.

[0006] Beta-thalassemia is one of the two the most common congenital anemias and is due to partial or complete lack of synthesis of beta-globin chains and hemoglobin. Patients affected by beta-thalassemia have mutations in the beta-globin gene. The World Health Organization (WHO) estimates that 50,000 to 100,000 children are born with symptomatic features of beta-thalassemia every year. Cooley's anemia, also known as beta-thalassemia major, the most severe form of this disease, is characterized by ineffective erythropoiesis (IE) and extra medullary hematopoiesis (EMH) requiring regular blood transfusions to sustain life.

[0007] In beta-thalassemia intermedia, where a greater number of beta-globin chains are synthesized, the clinical picture is milder and patients do not require frequent transfusions (Musallam, K. M., et al. Non-transfusion-dependent thalassemias. Haematologica 98, 833-844 (2013); Rivella, S. The role of ineffective erythropoiesis in non-transfusion-dependent thalassemia. Blood reviews 26 Suppl 1, S12-15 (2012); Ginzburg, Y. & Rivella, S. beta-thalassemia: a model for elucidating the dynamic regulation of ineffective erythropoiesis and iron metabolism. Blood 118, 4321-4330 (2011)). However, hemoglobin levels often decrease over time, splenomegaly appears, and patients suffer from progressive iron overload due to increased gastrointestinal iron absorption. Current disease management includes prenatal diagnosis, transfusion therapy, iron chelation and allogeneic bone marrow transplantation (BMT), which is limited by finding a compatible bone marrow donor and presents many risks and complications. Beta-thalassemia or Cooley's anemia has a serious impact on the life of those afflicted, as well as on society in general. Therefore, potential gene therapy approaches that provide therapy to these patients would be highly relevant. In this regard, mutations leading to Cooley's anemia can be classified as beta0, such as beta0-39, in which a single point mutation creates a stop codon and no beta-globin protein is produced, or beta+, such as beta+-IVS1-110, where a mutation in the first intron results in alternative splicing and insufficient beta-globin chain synthesis (Musallam, K. M., et al. Non-transfusion-dependent thalassemias. Haematologica 98, 833-844 (2013); Rivella, S. The role of ineffective erythropoiesis in non-transfusion-dependent thalassemia. Blood reviews 26 Suppl 1, S12-15 (2012); Ginzburg, Y. & Rivella, S. beta-thalassemia: a model for elucidating the dynamic regulation of ineffective erythropoiesis and iron metabolism. Blood 118, 4321-4330 (2011)). Previous research has shown that it is possible to rescue beta-thalassemia in mouse models by lentiviral-mediated beta-globin gene transfer (May, C., et al. Successful treatment of murine beta-thalassemia intermedia by transfer of the human beta-globin gene. Blood 99, 1902-1908. (2002); May, C., et al. Therapeutic hemoglobin synthesis in beta-thalassaemic mice expressing lentivirus-encoded human beta-globin. Nature 406, 82-86 (2000); Rivella, S., et al. A novel murine model of Cooley anemia and its rescue by lentiviral-mediated human beta-globin gene transfer. Blood 101, 2932-2939 (2003)). However, these animals are characterized by a complete deletion of the mouse beta-globin gene. Additional hemoglobinopathies are characterized by mutations in the beta-globin gene that lead to additional aberrant beta-globin chains and patho-physiological sequelae similar to sickle cell anemia and/or beta-thalassemia. Thus, there is an ongoing and unmet need for improved compositions and methods for treating hemoglobinopathies. The present disclosure is pertinent to this need.

SUMMARY

[0008] The present disclosure provides compositions and methods for prophylaxis and/or therapy of hemoglobinopathies. In general, hemoglobinopathies comprise disorders that are characterized by reduced synthesis or abnormal structure of the hemoglobin molecule. Thalassemias are considered a type of hemoglobinopathy, with the understanding that certain forms of hemoglobinopathies are typically inherited single-gene disorders that result in abnormal structure of one of the globin chains of the hemoglobin molecule, whereas thalassemias are considered other forms of hemoglobinopathies associated with underproduction of normal globin proteins, such as those cases that arise due to mutations in regulatory genes. Thus, the disclosure is pertinent to individuals in need of treatment for disorders that include but are not necessarily limited to those characterized by altered hemoglobin structure, such as in hemoglobin C disease, hemoglobin S-C disease, sickle cell anemia, as well as in various types of thalassemia that are well known in the art, including but not necessarily limited to beta-thalassemia. This is also pertinent to patients that have mixed hemoglobinopathy features, such as hemoglobin S/thalassemia, for instance.

[0009] In one aspect the disclosure provides a method for inducing expression of human beta-globin in erythrocytes for use in prophylaxis and/or therapy of a hemoglobinopathy in an individual. The method generally comprises introducing into erythrocyte progenitor cells, typically CD34+ cells, a polynucleotide encoding: i) a 5' long terminal repeat (LTR) and a self-inactivating 3' LTR (self-inactivating meaning the 3' LTR comprises a deletion relative to its native sequence, and thus results is replication incompetent); ii) at least one polyadenylation signal; iii) at least one promoter; iv) a globin gene locus control region (LCR); v) an ankyrin insulator element (Ank); vi) a Woodchuck Post-Regulatory Element (WPRE) configured such that the WPRE does not integrate into a target genome; and vii) a sequence that is a reverse complement to a sequence encoding modified human beta-globin comprising a .beta.T87Q mutation (B-globinM). The sequence of human B-globinM is included with this disclosure. The B-globinM is expressed from integrated DNA that is derived from the viral genome. Thus, the B-globinM sequence in the RNA genome of the modified lentiviral vectors of this disclosure is configured so that the B-globinM is expressed in the context of the double stranded DNA that is from integration of the modified lentiviral genome. The expression can in certain embodiments be exclusive to certain cell types, such as erythrocytes. It will be recognized in certain instances that DNA sequences are provided, but those skilled in the art can readily envision the RNA equivalent of the DNA sequence, such as when a DNA sequence is used to illustrate features of a lentiviral genome.

[0010] The sequence encoding the B-globinM comprises a first intron (intron 1) between exon 1 and exon 2, and a second intron (intron 2) between exon 2 and exon 3. In certain embodiments intron 2 comprises more than 476 nucleotides of the human B-globinM intron 2 sequence, and may comprise up to the full intron 2 sequence. After introducing the lentiviral construct into erythrocyte progenitor cells the lentiviral construct integrates into one or more chromosomes in the progenitor cells, and the progenitor cells differentiate into erythrocytes. The erythrocytes derived from the erythrocyte progenitor cells produce the beta-globin protein.

[0011] The disclosure provides for modified erythrocytes that produce more human beta-globin than a suitable control. In one embodiment the control comprises a human beta-globin value obtained from control cells. In one non-limiting approach the control cells comprise erythrocytes from an individual who has the hemoglobinopathy, wherein the erythrocytes are progeny of progenitor cells into which a control viral vector was introduced. The control viral vector can comprise, for example, the 5' LTR, the 3' LTR, the at least one polyadenylation signal, the at least one promoter, the LCR, the Ank, a WPRE, and the sequence encoding the B-globinM, but the sequence encoding the B-globinM in the control viral vector comprises 476 or fewer nucleotides of the human B-globinM intron 2 sequence, and thus comprises an intron 2 that contains a deletion. In one embodiment, the modified erythrocytes produce increased adult hemoglobin, fetal hemoglobin, B-globinM, or a combination thereof, relative to a control.

[0012] In one embodiment the lentiviral vectors can further comprise a sequence encoding a fusion of an Ldb1 transcription factor and a zinc finger (ZF) domain. In certain embodiments the lentiviral vector can comprise a sequence encoding an RNA polynucleotide that is at has reverse complementarity to mRNA encoding transferrin receptor 1. The RNA polynucleotide is capable of decreasing transferrin receptor 1 mRNA by, for example, an RNAi-mediated process.

[0013] In certain approaches the disclosure includes modifying erythrocyte progenitor cells that are CD34+ cells. The CD34+ cells can be separated from the individual before the lentiviral vector is introduced into them. The CD34+ cells can be introduced into the individual subsequent to the introduction of the lentiviral vector. The lentiviral vector can be introduced using any suitable approach. In one embodiment the erythrocyte progenitor cells are infected with viral particles comprising a lentiviral vector, such as a recombinant+strand viral polynucleotide. In certain embodiments, approximately 50 infection particles (IP) per cell are used. In certain embodiments the cells into which the vector is introduced can be enriched for a certain cell type, such as CD34+ cells. In certain approaches the cells are held for a period of from 2-3 days after infection prior to introducing into an individual in need thereof In embodiments the cells can be frozen and introduced into the patients after they are thawed. The lentiviral vectors can be introduced into a patient using any suitable method. In one approach they are administered by intravenous infusion, which may be performed subsequent to any other technique, such as after depletion of bone marrow cells, i.e., myeloablation. Those skilled in the art will recognize that a cells characterized by a suitable vector copy number (VCN) can be used. In one embodiment the VCN is approximately 1 VCN per cell.

[0014] The disclosure includes the lentiviral vectors themselves, the components of which are as described above for use in a method of the disclosure. Lentiviral vectors comprise modified, recombinant polynucleotides, and can comprise RNA or DNA polynucleotides. In certain embodiments a lentiviral vector of this disclosure comprises an isolated polynucleotide, or an isolated preparation of virions that comprise the lentiviral vector. In an embodiment a recombinant lentiviral vector of the disclosure is present in CD34+ cells, wherein the CD34+ cells have been separated from an individual.

[0015] Also provided is a method of making a viral particle preparation for use in prophylaxis and/or therapy for one or more hemoglobinopathies. This approach comprises introducing a plasmid encoding a lentiviral vector as described above into packaging cells, wherein the packaging cells comprise a DNA packaging plasmid which encodes at least one virion protein, and wherein the packaging cells comprises a DNA envelope plasmid which encodes an envelope protein. The packaging and envelope plasmids express their respective proteins, which facilitate formation of virions which comprise an RNA lentiviral vector of this disclosure.

DESCRIPTION OF THE FIGURES

[0016] FIGS. 1A-1C. The level of HbF/GFP expressing cells increases proportionally to the amount of molecules of GG1-SA vector integrated. Lentiviral construct carrying the bicistronic cassette that expresses the Ldb1 SA domain under the ankyrin promoter (Ank Pr.) and the green fluorescence protein (GFP) through an internal ribosomal entry site (IRES). The Woodchuck hepatitis virus Posttrascriptional Regulatory Element (WPRE) increases RNA stability and protein yield. FIG. 1A: Percentage of F, S and A.sub.2 hemoglobins measured by HPLC in differentiated untreated erythroid cells (left) or with up to 0.7 copies of ZF-Ldb1-viral molecules integrated on average (middle and right). Increase in HbF (FIG. 1B) and GFP (FIG. 1C) expressing cells measured by flow cytometry in cells untreated or with 0.26 and 0.67 copies/cell of ZF-Ldb1-viral molecules integrated.

[0017] FIGS. 2A-2C. pCL-ZF-Ldb1 supports high levels of .gamma.-globin/hemoglobin F induction and concurrently reduces sickle globin levels in sickle cell disease (SCD) erythroblasts. FIG. 2A: .beta.-like globin mRNA content (.gamma., left, and .beta.s, middle) was measured by Q-PCR and normalized by endogenous GAPDH expression. .beta.-like globin expression changes were further normalized by .alpha.-globin expression (which should remain unchanged and is a measure of differentiation across samples). Right: transgenic ZF-Ldb1 mRNA expression in treated samples is plotted against integration of pCL-ZF-Ldb1. FIG. 2B: HbF (left), HbS (middle) and HbA2 (right) changes in all SCD samples treated with pCL-ZF-Ldb1 LV. FIG. 2C: Net HbF % increase and HbS %-A2% decrease.

[0018] FIGS. 3A-3B. Quantification of tetrameric Hbs and single globin chains in SCD cells untreated and treated with pCL-ZF-Ldb1. FIG. 3A: Content of HbF increase (left) and all Hbs (right) in erythroid cells without or with pCL-ZF-Ldb1. FIG. 3B: Single globin quantification by liquid chromatography in denaturing conditions. On left, the quantity of single .beta.- and .gamma.-globin chains (.mu.g) is calculated over the quantity of single .alpha.-globin chain. On right, means of .gamma.A+G chains/all .beta.-like chains area indicated. .mu.g of Hbs or single globin chains in hemolysates were extrapolated from calibration curves obtained with standard samples with known Hb concentration.

[0019] FIGS. 4A-4D. pCL-ZF-Ldb1-transduced SCD cells expressing HbF have a reduced expression of HbS compared to untransduced cells. FIG. 4A: The number of GFP expressing SCD erythroblasts after pCL-ZF-Ldb1 integration (right) is compared to untransduced erythroblast (left). FIG. 4B: Percentage of HbF producing cells in permeabilized erythroblasts (left, no anti-HbF Ab), untransduced (center) and pCL-ZF-Ldb1 transduced erythroblasts (right). FIG. 4C: .beta.-globin expressing cells within the HbF positive populations from B (center and right) are compared to permeabilized cells (left, no anti-HbB Ab). FIG. 4D: Quantification of HbF of SCD samples (from B) analyzed by HPLC.

[0020] FIGS. 5A-5C. Expression of y-globin repressor genes in healthy cells, sickle cells untreated or treated with pCL-ZF-Ldb1 LV. Bcl11A and C-Myb, (FIG. 5A) and SOX6 and KLF1 (FIG. 5B) genes' expression is expressed in function of the level of cell differentiation marker glycophorin A (GPA) and normalized by GAPDH expression. The expression of KEL (FIG. 5C, left), the gene that encodes Kell, a blood group antigen, was chosen as internal control. Expression of transgenic Ldb1 in transduced samples (FIG. 5C, right) was confirmed in the same samples analyzed for the other messenger RNAs.

[0021] FIGS. 6A-6B. Hemoglobin F increase/Sickle hemoglobin decrease in CD34+-derived SCD erythroid cells treated with pCL-ZF-Ldb1 LV and with HbF inducers in vitro. FIG. 6A: (Top) Net increase of HbF % and (bottom) net decrease of HbS % in SCD erythroblasts treated with the HbF inducers decitabine, tranylcypromine, hydroxyurea, pomalidomide, butyrate or with the LV carrying ZF-Ldb1. FIG. 6B: Erythroid cell count (hemoglobinized cells, or benzidine+stained cells) for each treatment has been normalized over the untreated sample. In A-B n=11, except for Hu and pCL-ZF-Ldb1 in which n=10, while for But n=9.

[0022] FIGS. 7A-7C. Differential single globin chain synthesis in cells treated with pCL-ZF-Ldb1. FIG. 7A: Chromatographic profile of representative sickle cells hemolysate untreated (UT) or treated with pCL-ZF-Ldb1. FIG. 7B: Calibration curve obtained using known hemoglobin concentration of blood from SCD transgenic mouse. FIG. 7C: Areas under peak of single globin chains extrapolated from the calibration curve (from B) in samples untreated or after treatment with pCL-ZF-Ldb1 (n=5).

[0023] FIG. 8. Trend of moderately elevated HbF levels in CD34+-derived SCD erythroid cells at steady state. Comparative HPLC assessment of HbF% between healthy and SCD erythroid samples in which .gamma.-globin repressors BCl11a, c-Myb, KLF1 and SOX6 were quantified

[0024] FIG. 9. Cytotoxicity effect and dose/response calibration. Erythroid cells count measurements (benzidine plus count) of cells (N=2) at different doses of drugs in comparison with integration of .about.1 copy/cell of GG1-SA. The arrows in black indicate the dose of each drug chosen for the bulk of the experiments.

[0025] FIG. 10. Variation of HbF % SCD erythroid cells treated with pCL-ZF-Ldb1 vector and with HbF inducers in vitro at different times. Net increase of HbF % in SCD erythroblasts treated with the HbF inducers 5-aza-cytidine or with the LV carrying ZF-Ldb1, according to FIG. 16(L) and FIG. 17(E). Kruskal-Wallis with Dunn's multiple comparison test.

[0026] FIG. 11. Graphical maps of vectors. Topmost vector map is pCL-ZF-Ldb1, also referred to as pCL20cAnkyrinGG1DDiGFP. Second from vector map is ALS-10. Third from top vector map is CT9Ank. Bottom vector map is ALS-10T. Each vector comprises a 5' and a 3' self-inactivating long terminal repeat (5'LTR and 3' SinLTR, respectively). Also shown is "B-globinM" which is a mutant beta-globin is known as the "(3T87Q" form. The B-globinM is configured in the vector such that it is expressed from an integrated DNA that is derived from the RNA genome via a well-known process. "Ank" is an Ankyrin insulator. "IRES" is an internal ribosomal entry site. "P" designates a promoter. "LCR" is a Locus control region. "GFP" is enhanced green fluorescence protein. "pA" is a polyadenylation signal. "WPRE" is the Woodchuck Post-Regulatory Element. "fI1" is a full beta-globin gene intron 1. "fI2" is a full beta-globin gene intron 2. "I1S" is a modified beta-globin gene intron 1 comprising a microRNA targeting the transferrin receptor. "SV40 oriR-pA" is an origin of replication and polyadenylation signal. Beta-globin gene exons 1, 2 and 3 are labeled accordingly.

[0027] FIG. 12 provides a graphical summary of results comparing adult hemoglobin values (HbA) obtained from .beta.0/+ or .beta.0/0 erythroblasts patient CD34+ cells into which the AnkT9W and ALS10 vectors were introduced, as labeled.

[0028] FIG. 13 provides an annotated polynucleotide and encoded protein sequence of the ZF-Ldb1 vector and the DNA equivalent of the RNA. The nucleotide sequence in FIG. 13 is SEQ ID NO:1. The ZF-LDB1 AA sequence (shown as GG1=ZF and DDi=LDB1, with the HA terminal amino acid sequence) is SEQ ID NO:2. Vector features labeled in boxes are shown above the pertinent sequences.

[0029] FIG. 14 provides an annotated polynucleotide sequence of the ALS-10 vector as the DNA equivalent of the RNA and encoded protein sequence. The nucleotide sequence shown in FIG. 14 is provided as SEQ ID NO:3. Vector features in boxes are shown above the pertinent sequences. The location of the initiating .beta.T87Q beta globin methionine codon in its antiparallel configuration is the "CAT" triplet to the immediate left of the beta globin 5' UTR, reading in the 5' to 3' direction.

[0030] FIG. 15 provides an annotated sequence of the non-mutated beta-globin cDNA. The cDNA sequence is SEQ ID NO:4. Also shown is the beta-globin amino acid sequence which is provided in SEQ ID NO:5. The .beta.T87Q mutation occurs at the Threonine which is shown in the sequence at position 88. The mutation is referred to as .beta.T87Q according to convention wherein the first Methionine at position 1 is not included in the amino acid numbering. In the .beta.T87Q mRNA, the codon for the Threonine at position 88 is replaced by a codon encoding Glutamine.

[0031] FIG. 16. (Top) Experimental flow for expansion, differentiation and treatment of human SCD CD-34+ cells with pCL-ZF-Ldb1 and/or HbF pharmacological inducers. (Bottom) Type of HbF inducer, biological activity and dose used in the study.

[0032] FIG. 17. Experimental flow for expansion, differentiation and treatment of human SCD CD-34+ cells with pCL-ZF-Ldb1 or HbF pharmacological inducers.

DETAILED DESCRIPTION

[0033] The present disclosure provides compositions and methods for prophylaxis and/or therapy for hemoglobinopathies. In this regard, and as is well known in the art, a few months after birth children start producing red cells containing adult hemoglobin, which is the oxygen carrier molecule made of alpha- and beta-globin chains, encoded, respectively, by the alpha- and beta-globin genes. In many hemoglobinopathies, mutations in the gene encoding the beta-globin chain impair the synthesis of adult hemoglobin or lead to the production of abnormal adult hemoglobin. This leads to limited production of red cells or synthesis of abnormal red cells. For these reasons, patients require blood transfusion for survival. Before birth and during the first months of life, children express fetal hemoglobin, which comprises gamma- and alpha-chains. Generally, after birth the gene encoding the gamma-chain is silenced, while the beta-globin gene is activated, switching the production of hemoglobin from fetal to adult. In rare cases, individuals that do not silence the gamma-globin gene and also carry mutations in the beta-globin gene are spared from the disease associated with reduced or abnormal production of adult hemoglobin. Therefore, reactivation of fetal hemoglobin might be therapeutic in ameliorating diseases associated with mutations in the beta-globin gene. Activation and silencing of the gamma-globin gene depends on the proximity of a genomic region called locus control region (LCR), which maps .about.40 to 60 kilobases upstream of the gamma- and beta-globin genes and is associated with many factors that activate transcription. This region is required to "loop" and bind the promoter of the gene that needs to be activated. When the gamma-globin gene is expressed, the LCR loops and binds on the gamma-globin promoter. After birth, the LCR moves away from the gamma-globin promoter and loops and binds the beta-globin promoter leading to silencing of the gamma gene and activation of the globin-gene. The transcription co-factor Ldb1 is involved in the LCR looping to the promoters of the gamma- and beta-globin genes. Ldb1 alone does not promote the binding of the LCR to the promoter, but requires additional factors. Artificial zinc-finger (ZF) proteins have the ability of binding specific sequences on the DNA. Ldb1 has been fused to a specific ZF protein that binds the gamma-globin promoter. In transgenic mice, this protein promotes the looping and binding of the LCR to the promoter of the gamma globin gene, activating its expression (Deng, W., et al. Controlling long-range genomic interactions at a native locus by targeted tethering of a looping factor. Cell 149, 1233-1244 (2012)).

[0034] An alternative way to approach therapy of hemoglobinopathies associated with mutation in the beta-globin gene is by inserting in the bone marrow cells (and in particular in the hematopoietic stem cells or HSCs) a functional beta-globin gene. Therefore, gene transfer of the non-mutated form of the beta-globin gene by lentiviral vectors could potentially restore production of the human beta-globin protein in patients and be utilized for gene therapy trials. Lentiviral vectors are well characterized for their ability to infect and insert the human beta-globin gene into HSCs. In this regard, the present disclosure provides novel viral vectors that are designed for prophylaxis and/or therapy of hemoglobinopathies. The viral vectors include but are not necessarily limited to lentiviral vectors. In various embodiments an isolated mammalian hematopoietic progenitor cell or an isolated mammalian stem cell comprising a recombinant lentiviral vector is provided.

[0035] Various embodiments of lentiviral vectors that are subjects of this disclosure are shown in FIG. 11. Features of these vectors include but are not necessarily limited to the capability to, when introduced to an appropriate cell: 1) reactivate expression of fetal hemoglobin, and/or 2) express a novel transgene adult hemoglobin and/or, concurrently, 3) inactivate the expression of mutant hemoglobin. Certain features of the vectors are known in the art, and FIGS. 13 and 14 provide representative sequences of them. In particular, any suitable IRES sequence can be used, and those skilled in the art will recognize, given the benefit of this disclosure, which promoter sequences can be adapted for use in embodiments of the invention. Likewise, the LCR sequence is known in the art, as are suitable polyadenylation signals that can function, for example, in erythrocytes. The representative sequences of this disclosure, such as those shown in FIGS. 13 and 14, can be altered according to well-known parameters, so long as they impart to erythrocytes the ability to produced therapeutically effective amounts of elevated globin. In certain cases polynucleotide sequences can be identical to those presented herein, or they can have least 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99% identity across a contiguous segment of the sequences.

[0036] It is expected that lentiviral vectors of this disclosure will result in the expression of therapeutic levels of hemoglobins where previous vectors have failed to do so. In embodiments, these vectors can be specifically adapted to optimize production of beta-globin plus gamma-globin mRNA, and protein levels in presence of different mutations associated with beta-thalassemia, sickle cell anemia and other hemoglobinopathies. Certain aspects of this disclosure involve combinations of genomic elements, specific embodiments of which are depicted schematically in FIG. 11. The disclosure includes each and every polynucleotide sequence disclosed herein, the RNA equivalent of every DNA polynucleotide (i.e., where uracil replaces thymine) and every DNA equivalent of every RNA, and the complementary sequence and the reverse complement of every polynucleotide sequence. The disclosure includes every amino acid sequence, and all polynucleotide sequences encoding the amino acid sequences. Contiguous segments of polynucleotides and polypeptides sequences are also included. In non-limiting examples, sequences of this disclosure can comprise or consist of any one or any combination of the contiguous segments of each vector construct disclosed herein, including but not limited to the segments and sequences depicted in FIGS. 11, 13, 14 and 15, the RNA equivalents thereof, and the DNA and RNA reverse complements thereof. In aspects of the disclosure integration of a lentiviral vector of the disclosure results in an integrated functional globin gene, the expression of which produces a globin molecule that is effective to a participate in providing a therapeutic benefit to an individual with a globinopathy. The gene may exhibit conditional expression, such as being expressed in erythrocytes, but not being expressed in for example, hematopoietic stem cells comprising an integrated DNA segment derived from the lentiviral construct, wherein the stem cells differentiate into the erythrocytes that express the globin molecule. The disclosure also includes cells, which comprise the recombinant polynucleotides.

[0037] The present disclosure includes a demonstration of using the ALS10 lentiviral vector depicted in FIG. 11 to elevate HbA production in cells from hemoglobinopathy patients. In particular, the disclosure includes a demonstration that the ALS10 vector is superior to previously available approaches for improving HbA production in the most severe thalassemic specimens, i.e., those individuals with .beta.0/0 phenotype. In this regard, the disclosure provides a demonstration of using ALS10 to modify CD34+ cells from .beta.0/0 phenotype patients such that HbA production is improved relative to a suitable control, and those skilled in the art will recognize suitable controls given the benefit of this disclosure. In embodiments, the control can be a single value or a range of values. For example, a control can be a standardized curve or an area on a graph. In one embodiment the control comprises the increase in HbA produced using a construct described in Breda, L., et al., Therapeutic hemoglobin levels after gene transfer in beta-thalassemia mice and in hematopoietic cells of beta-thalassemia and sickle cells disease patients. PLoS One, 2012. 7(3): p. e32345, which is known in the art as "AnkT9W."

[0038] In more detail, and without intending to be constrained by any particular theory, the approach of the current disclosure was taken in part in an effort to increase adult hemoglobin in thalassemic and SCD cells in a tissue specific manner, and proportionally to the number of viral molecules inserted. To attempt to reduce genome toxicity believed to be caused by random integration of viral vectors, we strived to maintain viral integration within an average of 2 copies/cell.

[0039] It has been demonstrated in thalassemic specimens with moderate to medium range of HbA reduction (.beta.+/+ and .beta.+/0, respectively) 0.6 copies of AnkT9W were sufficient to generate HbA levels comparable to those detected in healthy or carrier cells, which is about 80-90% (Breda et al, Plos One, 2012). However, in specimens with the most severe phenotype, .beta.0/0, in which no adult HbA is observed, 0.6 copies of AnkT9W could not meet the level of adult hemoglobin observed in healthy or carrier cells, which was a therapeutic threshold that we strived to achieve in the presently provided approach. In connection with this we modified AnkT9W to generate ALS10. In ALS10, the Woodchuck Post-Regulatory Element (WPRE) was eliminated from the integrating sequence to increase the safety of the vector. The WPRE was part of the integrated portion of AnkT9W since it was placed between the LCR and the 3' LTR. The original purpose of the WPRE was to increase the titer of the lentivirus as it had been previously shown to have that effect, but we have now determined that it is dispensable in the current position. WPRE is a viral sequence and is not required for the expression of the beta-globin gene. It is considered safer to limit as much as possible the amount of viral sequences that are integrated in the genome of patient cells. For this reason, we moved this sequence in the vector to the non-integrating region. This preserves the WPRE's ability to produce high titers of viral particles but excludes the WPRE from the genome of patient cells. In order to preserve the ability of WPRE to increase viral titers without having it in the integrating sequence, we removed the WPRE from the integrating portion (before the 3'LTR) and added it back after the 3'LTR so that it is positioned outside the sequence that is integrated in the chromosome(s) of target cells. We also added a strong bovine growth hormone polyA tail after the WPRE region (outlined in FIG. 11). Our data indicate that the modifications do not decrease viral titer during production of compositions of this disclosure. However, we also modified the segment of the construct that results in expression of HbA in erythrocytes that are derived from CD34+ cells into which the lentiviral vector is introduced. In particular, the portion of the beta-globin gene intron 2 that is deleted in the AnkT9W construct is annotated in FIG. 14. As can be seen from that annotation, the beta-globin gene intron 2 encompasses nucleotides 4772-5621, inclusive, and is thus 851 nucleotides in length. However, the intron 2 in AnkT9W comprises a deletion of the 375 nucleotides spanning nucleotides 5,164 through 5,537, inclusive (as designated in FIG. 14). As a consequence of that deletion, the AnkT9W beta-globin gene intron 2 is 476 nucleotides in length. In contrast, and while not intending to be bound by any particular theory, it is considered that including a longer segment of intron 2 in the context of the ALS10 construct is at least partially responsible for the unexpected and statistically significant increase in HbA in the .beta.0/0 patient cells. In this regard, when compared to AnkT9W, ALS10 showed significant improvement and in particular leads to much higher level of HbA in the most severe thalassemic patient specimens, namely, the .beta.0/0 phenotype. It is therefore reasonable expect that the present approach will benefit patients with hemoglobinopathies that are not necessarily due to a .beta.0/0 phenotype, such as SCD. A summary of the results demonstrating this advantage of ALS10 is presented in FIG. 12. The disclosure thus includes lentiviral vectors and cells comprising them, and the integrated construct, wherein the beta-globin gene comprises an intron 2 of more than 476 nucleotides in length, and up to 851 nucleotides in length. The intron is accordingly between 477-875 nucleotides, inclusive, and including all integers and ranges of integers there between. The intron 2 can be thus comprise or consist of 477, 478, 479, 480, 481, 482, 483, 484, 485, 486, 487, 488, 489, 490, 491, 492, 493, 494, 495, 496, 497, 498, 499, 500, 501, 502, 503, 504, 505, 506, 507, 508, 509, 510, 511, 512, 513, 514, 515, 516, 517, 518, 519, 520, 521, 522, 523, 524, 525, 526, 527, 528, 529, 530, 531, 532, 533, 534, 535, 536, 537, 538, 539, 540, 541, 542, 543, 544, 545, 546, 547, 548, 549, 550, 551, 552, 553, 554, 555, 556, 557, 558, 559, 560, 561, 562, 563, 564, 565, 566, 567, 568, 569, 570, 571, 572, 573, 574, 575, 576, 577, 578, 579, 580, 581, 582, 583, 584, 585, 586, 587, 588, 589, 590, 591, 592, 593, 594, 595, 596, 597, 598, 599, 600, 601, 602, 603, 604, 605, 606, 607, 608, 609, 610, 611, 612, 613, 614, 615, 616, 617, 618, 619, 620, 621, 622, 623, 624, 625, 626, 627, 628, 629, 630, 631, 632, 633, 634, 635, 636, 637, 638, 639, 640, 641, 642, 643, 644, 645, 646, 647, 648, 649, 650, 651, 652, 653, 654, 655, 656, 657, 658, 659, 660, 661, 662, 663, 664, 665, 666, 667, 668, 669, 670, 671, 672, 673, 674, 675, 676, 677, 678, 679, 680, 681, 682, 683, 684, 685, 686, 687, 688, 689, 690, 691, 692, 693, 694, 695, 696, 697, 698, 699, 700, 701, 702, 703, 704, 705, 706, 707, 708, 709, 710, 711, 712, 713, 714, 715, 716, 717, 718, 719, 720, 721, 722, 723, 724, 725, 726, 727, 728, 729, 730, 731, 732, 733, 734, 735, 736, 737, 738, 739, 740, 741, 742, 743, 744, 745, 746, 747, 748, 749, 750, 751, 752, 753, 754, 755, 756, 757, 758, 759, 760, 761, 762, 763, 764, 765, 766, 767, 768, 769, 770, 771, 772, 773, 774, 775, 776, 777, 778, 779, 780, 781, 782, 783, 784, 785, 786, 787, 788, 789, 790, 791, 792, 793, 794, 795, 796, 797, 798, 799, 800, 801, 802, 803, 804, 805, 806, 807, 808, 809, 810, 811, 812, 813, 814, 815, 816, 817, 818, 819, 820, 821, 822, 823, 824, 825, 826, 827, 828, 829, 830, 831, 832, 833, 834, 835, 836, 837, 838, 839, 840, 841, 842, 843, 844, 845, 846, 847, 848, 849, 850, 851 nucleotides. The polynucleotide sequence of the human beta globin gene is depicted in FIG. 13. It includes the 3' enhancer element, the 3' untranslated region (UTR), the polyA signal, exon 3, intron 2, exon 2, intron 1, exon 1, the 5' UTR, and the beta globin promoter segment. It is shown in the 3'-5' orientation because, as is well known in the art, it is anti-parallel to the depicted remainder of the construct and is expressed only after integration and in the context of a double stranded DNA region wherein one strand of the DNA comprises the beta globin gene sequence presented in FIG. 14

[0040] In ALS10 the sequence of the beta-globin gene is also modified to increase its ability to prevent the sickling of the sickle-beta-globin chains (referred to as "B-globinM" in FIG. 11). This mutant beta-globin is known as the .beta.T87Q form, due to its amino acid substitution at the 87th position. .beta.T87Q has been used to improve hematological parameters of the SAD and BERK mouse models of SCA (Pawliuk, R., et al. Correction of sickle cell disease in transgenic mouse models by gene therapy. Science 294, 2368-2371. (2001)). This form was also used in the first successful clinical trial to correct a patient with .beta.0/.beta.E thalassemia (Cavazzana-Calvo, M., et al. Transfusion independence and HMGA2 activation after gene therapy of human beta-thalassaemia. Nature 467, 318-322 (2010)).

[0041] The ankyrin insulator that is well known in the art and was present in AnkT9W is also present in ALS10. ALS10 has the ankyrin insulator in the 3'LTR, while the vector AnkT9W has the ankyrin insulator between the promoter and the LCR.

[0042] In one aspect of this disclosure, an intron of the beta-globin gene, or other locations such as the 3'UTR, can be modified to include a polynucleotide that targets and decreases the synthesis of the transferrin receptor 1 (TR1). The rational of this approach is based on our observations that decreased erythroid iron intake can be beneficial in beta-thalassemia and, potentially other hemoglobinopathies, because this decreases formation of heme molecules. Heme is normally included in hemoglobin molecules but in many hemoglobinopathies, due to the reduction in synthesis of beta-globin chains, there is an excess of heme not bound to hemoglobin, and these molecules are toxic to red cells. An excess of heme is responsible for apoptosis of erythroblasts and for altering the intracellular content, production of reactive oxygen species (ROS), and for reducing membrane stability and plasticity of erythrocytes, leading to their reduced lifespan, hemolysis and/or sickling. A combination vector that expressed gamma-globin and concurrently knocked-down sickle beta-globin via small hairpin RNA has been described (Samakoglu, S., et al. A genetic strategy to treat sickle cell anemia by coregulating globin transgene expression and RNA interference. Nature biotechnology 24, 89-94 (2006)) and this approach can be incorporated in embodiments of the present disclosure (i.e., FIG. 11, in ALS-10T, signified by "I1S"). With respect to polynucleotides that target TR1 and that can be included in vectors of this disclosure, shRNA is one non-limiting example of an RNAi-mediated approach. But RNAi-based inhibition can be achieved using any suitable RNA polynucleotide that is targeted to TR1 mRNA. With respect to shRNAs, they are known in the art to adopt a typical hairpin secondary structure that contains a paired sense and antisense portion, and a short loop sequence between the paired sense and antisense portions. shRNA is delivered to the cytoplasm where it is processed by DICER into siRNAs. siRNA is recognized by RNA-induced silencing complex (RISC), and once incorporated into RISC, siRNAs facilitate cleavage and degradation of targeted mRNA. In embodiments, an shRNA polynucleotide segment or DNA segment encoding it included for use in suppressing TR1 expression can comprise or consist of between 45-100 nucleotides, inclusive, and including all integers between 45 and 100. The portion of the shRNA that has reverse complementarity to the TR1 mRNA mRNA can be from 21-29 nucleotides, inclusive, and including all integers between 21 and 29. In another approach, a ribozyme that can specifically cleave TR1 mRNA can be included. In another embodiment, microRNA (.mu.RNA) targeted to the TR1 mRNA can be used.

[0043] The present disclosure provides an illustration that vector encoding a fusion of an Ldb1 transcription factor and a zinc finger (ZF) domain (ZF-Ldb1) is useful for approaching certain hemoglobinopathies. A ZF-Lbd1 gene can be incorporated into vectors of this disclosure in several configurations. In one non-limiting approach using ALS-10 in FIG. 11 as a representative example, the ZF-Ldb1 gene can be positioned such that the vector comprises consecutively (with the same stranded-ness as the beta-globin coding sequence) the 5'LTR, a polyadenylation signal, the ZF-Ldb1 region and a either a promoter that can drive expression of a separate mRNA encoding the ZF-Ldb1 protein, or an IRES so that the mRNA encoding the ZF-Ldb1 protein can be made as a distinct protein, but from the same mRNA that encodes the beta-globin protein. The rational for including ZF-Ldb1 into ALS is twofold: 1) the ZF-Ldb1 will move the LCR from the promoter of the mutant beta-globin gene to that of the gamma-globin gene. In this way the production of mutant RNA will be reduced or shut down, while expression of the functional gamma-globin gene will be activated: 2) The expression of transgenic beta-globin gene (considered to be a potentially curative gene carried by the lentiviral vector) will not be affected in the presence of ZF-Ldb1. Without intending to be constrained by theory this is expected to lead to an additive or synergistic effect by the production of both hemoglobins: fetal hemoglobin (HbF, .alpha.2.gamma.2, from the endogenous locus), and adult hemoglobin (HbA or .alpha.2.beta.2, from the therapeutic vector). As hemoglobinopathies in humans are characterized by more than 300 mutations, it is reasonable to predict that the many or all hemoglobinopathies could be improved by a vector of this disclosure. In this regard, we demonstrated that a lentiviral vector encoding the ZF-Ldb1 cassette (pCL-ZF-Ldb1) increases synthesis of fetal hemoglobin (HbF, .alpha.2.gamma.2) in CD34-derived erythroid cells from normal individuals and SCA patients. As HbF increased, the synthesis of adult hemoglobin (HbA) or sickle hemoglobin (HbS or .alpha.2.beta.s2) diminished, respectively in cells derived from normal individuals or SCA patients. We also investigated the number of vectors integrated per cell, on average. This number is indicated as number of integration per cell or vector copy number (VCN). We observed that the increase in VCN was associated with an increase of HbF both in normal and SCA cells. The number of HbF positive cells measured by flow cytometry also increased proportionally to the VCN. .beta., .gamma.- and .alpha.-globin mRNA levels measured by quantitative PCR also showed that the relative ratio .gamma./.alpha. increased while the .beta./.alpha. or .beta.S/.alpha. were reduced in the samples treated with the ZF-Ldb1 vector. Taken together these data suggest that the ZF-Ldb1 can redirect the LCR enhancing effect from the .beta.-globin promoter to the .gamma.-globin promoter. In SCA this effect is particularly beneficial given the toxicity caused by the accumulation of HbS. Examples 1-6 further confirm potential usefulness of including ZF-Ldb1 in ALS10. In particular, these Examples demonstrate infection of hematopoietic stem cells isolated from blood of SCD patients with a lentivirus expressing the ZF-Ldb1 transgene and differentiation of them into mature erythroid cells in vitro. HbF synthesis induced by ZF-Ldb1 was compared to that obtained in specimens treated with hydroxyurea and various additional HbF inducers. ZF-Ldb1 increased HbF synthesis and simultaneously reduced sickle Hb (HbS), establishing a balanced synthesis between .alpha.- and functional .beta.-like globins. The induction of HbF in cells treated with ZF-Ldb1 was roughly three times higher (+34%), than that observed using decitabine and pomalidomide; tranylcypromine had an intermediate effect, while butyrate and hydroxyurea showed marginal HbF induction. Notably, erythroid cell differentiation and viability remained unaltered in ZF-Ldb1 expressing cells. Thus, lentiviral-mediated ZF-Ldb1 gene transfer appears superior to existing drug regimens for affecting SCD erythroid cells and it is reasonable to expect that combining ZF- to an ALS10 vector will provide an effective approach to prophylaxis and/or therapy of a variety of hemoglobinopathies. The Examples provide also provide a demonstration of the effectiveness of ALS10 in .beta.0/0 patient cells.

[0044] Compositions comprising recombinant lentiviral vectors are provided. In certain approaches pharmaceutical compositions are provided and can be prepared by mixing, for example, virions comprising a lentiviral vector of this disclosure and any suitable pharmaceutically acceptable carriers, excipients and/or stabilizers. Some examples of compositions suitable for mixing with IL-8 can be found in: Remington: The Science and Practice of Pharmacy (2005) 21st Edition, Philadelphia, Pa. Lippincott Williams & Wilkins. In certain approaches.

[0045] The disclosure includes methods of making a virion preparations for use in prophylaxis and/or therapy of hemoglobinopathies. In one embodiment this method comprises introducing a plasmid encoding a lentiviral vector of this disclosure into packaging cells. The packaging cells comprise a DNA packaging plasmid, which encodes at least one virion protein, and a DNA envelope plasmid, which encodes a suitable viral envelope protein. The packaging and envelope plasmids express the respective proteins, which facilitate formation of virions which comprise a recombinant RNA lentiviral vector of this disclosure. Suitable packaging systems that can be adapted to produce virions of this disclosure are commercially available, such as from Addgene (Australia).

[0046] The following Examples are intended to illustrate various aspects of the present disclosure but are not meant to be limiting in any way.

EXAMPLE 1

[0047] This Example demonstrates that lentivirally expressed ZF-Ldb1 raises HbF levels in CD34+-derived sickle erythroblasts. To obtain the data presented here, we used the lentiviral vector pCL-ZF-Ldb1, (FIG. 11, top vector map), which carries a zinc-finger protein that specifically binds the .gamma.-globin promoters, fused to the self-association (SA) domain of Ldb1, as well as a green fluorescent protein under the control of the erythroid specific ankyrin promoter. The effects of ZF-Ldb1 expression on chromatin contacts within the .beta.-globin locus have been shown using 3C experiments in healthy adult erythroblasts. ZF-Ldb1 expression promotes the juxtaposition of the .gamma.-globin promoters with the LCR leading to transcription [Deng, W., et al., Reactivation of developmentally silenced globin genes by forced chromatin looping. Cell, 2014. 158(4): p. 849-60]. This is accompanied by a concomitant reduction in the expression of adult globin genes, compatible with a mechanism in which the fetal and adult promoters compete for LCR enhancer activity. This example describes an attempt to improve the amount of functional hemoglobin in CD34+ cells isolated from sickle cell patients. Sickle CD34+ cells produce predominantly HbS (.alpha.2.beta.s2) once differentiated into erythroblasts in vitro. Other hemoglobins, as HbF (.alpha.2.gamma.2) and HbA2 (.alpha.2.beta.2) are also produced to a lower degree (FIG. 1A, left). Following infection with pCL-ZF-Ldb1 vector, sickle cells dramatically increased HbF synthesis in a manner proportional to the copy number of viral integrants. In a representative experiment the same SCD derived erythroid cells (shown in FIG. 1A, left) transduced with 0.26 or 0.67 viral molecules on average per cell (VCN or vector copy number) produce 22% or 44% (FIG. 1A, middle and right, respectively) more HbF than the control sample. Concomitantly, with progressively higher viral integration the number of HbF (FIG. 1B) and GFP (FIG. 1C) expressing erythroblasts increased, as determined by flow cytometry measurements. The gain in the fraction of GFP+ cells is somewhat lower, likely the result of reduced translation efficiency that is often observed downstream of the internal ribosomal entry site (IRES) between the ZF-Ldb1 cassette and the GFP gene.

EXAMPLE 2

[0048] This Example demonstrates that transgenic ZF-Ldb1 supports high levels of fetal globin induction and concurrently reduces sickle globin levels in SCD erythroblasts. In particular, we analyzed the mRNA and protein content of erythroblasts derived from 10 SCD subjects. CD34+ cells isolated from peripheral blood mononuclear cells (PBMCs) were frozen and used for independent replicate experiments. Infection with pCL-ZF-Ldb1 was performed on pro-erythroblast within the first 10 days of the expansion phase. At this time cells still express high levels of CD1117 and CD44 markers and do not express glycophorin A (data not shown) (Breda, L., et al., Therapeutic hemoglobin levels after gene transfer in beta-thalassemia mice and in hematopoietic cells of beta-thalassemia and sickle cells disease patients. PLoS One, 2012. 7(3): p. e32345).

[0049] To assess the ability of ZF-Ldb1 to reactivate HbF we measured the amount of .gamma.-globin mRNA expressed in cells untreated or after transduction. These values were normalized to GAPDH and to .alpha.-globin gene expression whose expression is directly proportional to the erythroid differentiation stage in the cells. Cells with 1.1 copies of pCL-ZF-Ldb1 produced on average a 3-fold increase of the .gamma./.alpha. globin ratio (from 0.2.+-.0.11 to 0.6.+-.0.33), compared to untreated cells and, simultaneously, a reduction of the .beta./.alpha. ratio (from 0.38.+-.0.08 to 0.29.+-.0.16) (FIG. 2A, left and center), confirming the ability of the ZF-Ldb1 construct to partially redirect the LCR from the .beta.- to the .gamma.-globin promoter. These changes were observed only in cells expressing transgenic Ldb1, whose expression was proportional to the level of ZF-Ldb1 integration (FIG. 2A, right).

[0050] On average, cells treated with pCL-ZF-Ldb1 produced nearly 40% more HbF (63.10%.+-.14.01) compared to untreated cells (27.27%.+-.16.29) (FIG. 2B, left) and lowered production of HbS (-35.65%) and HbA2 (-5.18%) (FIG. 2B, center and right). Reduction of HbA2 is likely the result of reduced contact frequencies between .quadrature.-globin and the LCR in the presence of ZF-Ldb1. A summary of net HbA increase and HbS and HbA2 decrease is provided in FIG. 3C.

EXAMPLE 3

[0051] To establish the effect of ZF-Ldb1 treatment on the total amount of cellular Hb we measured absolute Hb content per differentiated cell. Importantly, in spite of significant shifts in the ration of fetal to adult globin, total Hb synthesis changes (HbF+HbS+HbA2) remained essentially unaltered (FIG. 3A, right). To corroborate this evidence is the fact that .beta.S chains are diminished while both .gamma.A+.gamma.G chains are increased in specimens treated with ZF-Ldb1, analyzed by reversed-phase liquid chromatography (FIG. 3B and S1) which allows for the quantification of single globin chains rather than to tetrameric hemoglobin abundance. The reduced amount of HbS was confirmed by flow cytometry (FIG. 4). ZF-Ldb1-expressing SCD erythroblasts, which can be tracked by GFP expression (FIG. 4A), have a greater fraction of HbF positive cells (FIG. 4B), and within the HbF positive population, a lower fraction of HbS compared to untransduced SCD erythroblasts (FIG. 4C). Untreated baseline HbF positive erythroblasts (FIG. 4B, center) were less frequent and contained less than half the HbF/erythroid cell when compared to the ZF-Ldb1 expressing cells (FIG. 4D).

EXAMPLE 4

[0052] This Example demonstrates that y-globin gene repressors SOX6 and KLF1 are down regulated in sickle erythroblasts. In particular, BCL11A, SOX6, C-MYb and KLF1 have emerged as salient repressors of .gamma.-globin during erythroid differentiation. We investigated the impact of pCL-ZF-Ldb1 on transcription of these repressors in both wild type and sickle cell disease derived erythroid cells. RT-qPCR analyses indicates that differentiated erythroblasts obtained from SCD patients present a different pattern of expression for certain negative regulators of .gamma.-globin gene expression, compared to erythroblasts obtained from healthy individuals. BCL11A and C-MYB messenger RNAs present comparable level of expression (FIG. 5A), whereas both KLF1 and SOX6 show significant down regulation in SCD specimens (0.17.+-.0.05 and 4.49.+-.0.76, respectively) compared to healthy ones (0.55.+-.0.43 and 8.29.+-.3.14, respectively) (FIG. 5B). All samples analyzed present similar expression of KEL, an internal control mRNA that increases with level of differentiation. These trends are independent from the level of transgene expression (FIGS. 5 and 5C, right). These data could indicate a more permissive chromatin state of the .gamma.-globin gene in SCD cells in culture, as suggested by a trend of higher content of HbF at steady state in these cells compared to healthy ones (FIG. 7).

EXAMPLE 5

[0053] This Example demonstrates that HbF induction in SCD erythroblasts ZF-Ldb1-mediated is greater than induction mediated by pharmacological inducers. A scheme of the experimental procedures is illustrated in FIG. 16. Briefly, erythroid progenitor SCD cells were infected with pCL-ZF-Ldb1 within days 9-11 in expansion phase, or treated at days 1 and 3 of differentiation phase with pharmacological inducers 5-aza-2'-deoxy-cytidine (0.5 .mu.M), tranyl-cypromine (1.5 .mu.M), hydroxyurea (150 .mu.M), pomalidomide (30 .mu.M), and butyrate (100 .mu.M), These concentrations were determined through evaluation of efficacy (net increase of HbF) versus toxicity (cell death); the original scaling dosage was extrapolated from the recent literature (FIG. 9) [Watanapokasin, Y., et al., In vivo and in vitro studies of fetal hemoglobin induction by hydroxyurea in beta-thalassemia/hemoglobin E patients. Exp Hematol, 2005. 33(12): p. 1486-92; Moutouh-de Parseval, L. A., et al., Pomalidomide and lenalidomide regulate erythropoiesis and fetal hemoglobin production in human CD34+cells. J Clin Invest, 2008. 118(1): p. 248-58; Shi, L., et al., Lysine-specific demethylase 1 is a therapeutic target for fetal hemoglobin induction. Nat Med, 2013. 19(3): p. 291-4]. Studies were performed on day 8 of differentiation phase, at the orthochromatophilic stage when high amounts of hemoglobin were accumulated. As measured by HPLC, pCL-ZF-Ldb1 had the most robust increase of HbF and decrease in HbS. Specifically, on average the net HbF increase in SCD erythroblast treated with the lentivirus was 34.2%.+-.12.53, versus 15.19%.+-.12.77 with 5-aza-cytidine (p<0.01), 8.08%.+-.6.28 with tranyl-cypromine (p<0.001), 2.39%.+-.2.13 with hydroxyurea (p<0.001), 11.84%.+-.9.02 with pomalidomide (p<0.01), and 4.84%.+-.5.03 with butyrate (p<0.001). ZF-Ldb1 expressing cells presented HbF significantly higher than untreated cells (p<0.001) (FIG. 6A, top). Conversely, on average the net HbS decrease in SCD erythroblast treated with the lentivirus was -31.36%.+-.11.61, versus -17.30%.+-.15.04 with 5-aza-cytidine, -6.93%.+-.6.64 with tranyl-cypromine, -3.48%.+-.4.68 with hydroxyurea, 11.61-%.+-.9.27 with pomalidomide, and -4.90%.+-.7.94 with butyrate (FIG. 6A, bottom).

[0054] Along with recovery of greater levels of functional Hb, cells expressing ZF-Ldb1 did not show significant changes in viability compared to untreated samples, whereas cells treated with pomalidomide, butyrate and hydroxyurea showed reduced viability (FIG. 6B). These differences were confirmed in a subset of samples treated with pharmacological inducers at earlier time points (FIG. 17) to exclude biases due to a delay to drug response (FIG. 10). Taken together, pCL-ZF-Ldb1 was superior to all tested compounds in augmenting HbF and F-Cell levels and importantly was associated with minimal toxicity.

[0055] It will be apparent from the foregoing that a lentiviral vector carrying the SA Ldb1 domain linked to a ZF protein, which selectively binds the .gamma.-globin promoters, significantly increased HbF synthesis and exceeds previously described pharmacological inducers. It is therefore reasonable that adding an SA Ldb1 domain linked to a ZF protein to an ALS10 vector of this disclosure will likely enhance beneficial properties of the combined vectors.

EXAMPLE 6

[0056] This Example provides a description of the materials and methods used to obtain the results described in Examples 1-5.

[0057] Human and Animal Ethics

[0058] Peripheral blood samples from SCD patients were obtained during automated red cell exchange as part of their routine clinical care at Montefiore Medical Center. Since the samples were unlinked and de-identified medical waste, the Montefiore Medical Center Institutional Review Board deemed them to be IRB exempt.

[0059] Construct

[0060] The ZFs targeting HS2 of the human .gamma.-globin promoters are known in the art. The SA domain containing amino acids 1-200 of Ldb1 was inserted C-terminal to the ZF. The SA domain was attached in C-terminal to GG1 tagged with HA.

[0061] Vector Production and Titration

[0062] Viral stocks were generated by co-transfection of the gene transfer plasmid (pCL-ZF-Ldb1) together with the envelope plasmid (VSV-G), the packaging plasmid (pMDLg/p RRE), and the pRSV-REV plasmid into 293T cells. An aliquot (5.times.10.sup.6) of 293T cells was seeded into cell culture dishes (10 cm) 24 hours prior to transfection in Iscove's modification of Eagle's medium (DMEM, Cellgro, Manassas, Va.) with 10% fetal bovine serum, 100 U/ml penicillin, and 100 mg/ml streptomycin, at 37.degree. C. under 5% CO2. The culture medium was changed 2 hours prior to transfection. The precipitate was formed by adding the plasmids to 450 uL of 0.1.times.TE (0.1.times.TE is 10 mM Tris plus 1 mM EDTA) and 50 .mu.L of 2 M CaCl2, then adding 500 .mu.L of 2.times. HEPES-buffered saline (281 mM NaCl, 100 mM HEPES, 1.5 mM Na2HPO4) drop wise after which the precipitate was vortexed and immediately added to the cultures. The medium (10 ml) was replaced after 16 hours. Viral supernatants were collected at 24 and 48 hours, cleared by low speed centrifugation, and filtered through cellulose acetate (0.2 .mu.m). Following ultracentrifugation, serial dilutions of concentrated virus (5; 0.5 and 0.05 .mu.L, respectively) were used to infect 1.times.10.sup.5 NIH 3T3 cells (ATCC, Manassas, Va.) in 1 mL of transfection buffer complemented with polybrene (Millipore, Billerica, Ma.) at a final concentration of 8 .mu.g/mL. Genomic DNA was extracted after 3 days (Qiagen kit, Valencia, Calif.). The multiplicity of infection (MOI) was calculated using the following formula: number of cells (1.times.10.sup.5).times.dilution factor (1 mL/.mu.L viral preparation).times.VCN (measured via real-time PCR, using oligos for WPRE element and ID gene, see PCR and Real Time PCR).

[0063] Real Time (RT)-PCR

[0064] Retrotranscription of total mRNA was done using the SuperScript.TM. II First Strand Kit (Invitrogen, Carlsbad, Calif.). Q-PCR reactions were performed using the ABI Prism 7700 Sequence Detection System (Applied Biosystems, Foster City, Calif.), with either TaqMan (TaqMan PCR 2.times. Master mix; Applied Biosystems) or SYBR Green (iTaq.TM. SYBR.RTM. Green Supermix, Bio-Rad Laboratories, Hercules, Calif.) chemistry. Quantitative real-time PCR assays of globin and GAPDH transcripts were carried out using gene-specific double fluorescently labeled probes. The following primer and probe sequences were used (forward, reverse and probe, when used, of each gene, respectively): .beta.: Fw: 5'-CAAGAAAGTGCTCGGTGCCT-3' (SEQ ID NO:6); Rev: 5'-GCAAAGGTGCCCTTGAGGT-3' (SEQ ID NO:7); 5'-FAM-TAGTGATGGCCTGGCTCACCTGGAC-TAMRA-3' (SEQ ID NO:8); .alpha.: Fw: 5'-TCCCCACCACCAAGACCTAC-3' (SEQ ID NO:9); Rev: 5'-CCTTAACCTGGGCAGAGCC-3' (SEQ ID NO:10); 5'-FAM-TCCCGCACTTCGACCTGAGCCA-TAMRA-3' (SEQ ID NO:11); .gamma.: Fw: 5'-TGGCAAGAAGGTGCTGACTTC-3' (SEQ ID NO:12); Rev: 5'-TCACTCAGCTGGGCAAAGG (SEQ ID NO:13); 5'-FAM-TGG GAGATGCCATAAAGCACCTGG-TAMRA-3 (SEQ ID NO:14); BCL11A: Fw: 5'-TGATGTGTGTCCATTGGTGTGAGC-3' (SEQ ID NO:15); Rev: 5'-TGCGAACTTGAACGTCAGGAGTCT, SOX-6 (SEQ ID NO:16): Fw: 5'-AGCTGCTTTCGGCTTTCTCCCTTA-3' (SEQ ID NO:17); Rev: 5'-CCTTTGCATTTGCAGCAGTTCAGC-3' (SEQ ID NO:18); C-MYB: Fw: 5'-TCAACCGATCATCCCTCACACTCT-3' (SEQ ID NO:19); Rev: 5'-AATCAGCAGCGCTTCCATTCAAGG-3' (SEQ ID NO:20), KLF-1: Fw: 5'-GCTGCCTCCACCCAAGTG-3' (SEQ ID NO:21); Rev: 5'-ACCAACTCTGGGCAGTCACAT-3' (SEQ ID NO:22), Kell: Fw: 5'-AGCAACCACCCATGCCTGCC-3' (SEQ ID NO:23); Rev: 5'-CTCGGGCCAAAGGCCTCACG-3' SEQ ID NO:24). For real-time PCR of the reference genes, we used as an endogenous control the human glyceraldehyde-3-phosphate dehydrogenase (GAPDH) kit, in which the probe is fluorescently labeled with VIC (Applied Biosystems). The number of integrations (VCN) was quantified by Q-PCR using oligos (Fw: 5'-CGGCTGTTGGGCACTGA-3' SEQ ID NO:25); Rev: 5'-GGAAGGTCCGCTGGATTGA-3' SEQ ID NO:26)) and a probe (5'-FAM-ATGGCTGCTCGCCTGTGTTGCC-TAMRA-3' SEQ ID NO:27) for a specific sequence present in the vector (WPRE) and compared it to an endogenous control present in two copies within the genome (ID-1 Fw: 5'-AAGGTGAGCAAGGTGGAGATTC-3 SEQ ID NO:28); Rev: 5'-TTCCGAGTTCAGCTCCAACTG-3' SEQ ID NO:29).

[0065] Two-phase liquid cultures, benzidine staining and transduction

[0066] CD34+ cells selection from blood samples was performed by immunomagnetic separation, using the CD34 microbeads kit (Miltenyi Biotec Inc., Auburn, Calif.). These cells were then expanded following a modified version of the protocol described by Leberbauer and colleagues. Cells were seeded in 5 mL of serum-free StemSpan with 50 .mu.L of StemSpan CC-100 cytokine cocktail (both from Stemcell Technologies, Vancouver, BA, Canada), 2 U/mL Erytrhopoietin (Amgen, Thousand Oaks, Calif.), 10-6 M dexamethasone (Sigma) and 1% penicillin streptomycin. CD34+ cultures were kept undifferentiated by refreshing the medium twice a week and density gradient centrifugation was used to remove both dead and spontaneously differentiating cells. At this stage cells were either frozen (with 50% characterized Hyclone FBS, 10% DMSO, Sigma, and 40% Iscove's Modified DMEM, Cellgro), or used for experiments. After 10 days in phase I, cells were transferred into phase II media containing a-modified essential medium supplemented with 30% fetal calf serum and 10-5 M .beta.-mercaptoethanol. Erythropoietin was added (5 U/mL) to stimulate erythroid differentiation. Cells were infected with serial dilutions of the virus. Cells were collected on day 7-10 of phase II for all analyses. The level of differentiation was assessed by benzidine staining. At this stage, cells were analyzed by flow cytometry for GFP, HbF and HbB expression.

[0067] Treatment with HbF Inducers

[0068] We treated peripheral blood derived CD34+ cells (see procedure) with HbF inducer drugs: Hydroxyurea (HU), 5-azacytidine, pomalidomide, sodium butyrate, Tranyl-cypromine (TCP). Tranyl-cypromine (TCP), 5-aza-2'-deoxy-cytidine, pomalidomide and sodium butyrate. We first titrated the drugs to find the most efficient and less toxic concentration according to published data. Hydroxyurea (HU; Sigma-Aldrich), dissolved in water, was added to the culture medium at final concentrations of 150 .mu.M. Tranylcypromine (Sigma-Aldrich) was dissolved in water and added to the culture medium at final concentrations 1.5 .mu.M. 5-azacytidine (DAC; Sigma-Aldrich) was dissolved in water and added to the culture medium at final concentrations of 0.5 .mu.M. Pomalidomide (Sigma-Aldrich) was dissolved in DMSO and was added to the culture medium at final concentrations of 30 .mu.M. Sodium butyrate (Sigma-Aldrich) was dissolved in water and added to the culture medium at final concentration of 100 .mu.M. The treatment timeline is shown in FIG. 16.

[0069] Tetrameric and single chain analysis by high performance liquid chromatography (HPLC)

[0070] Red cell pellets were lysed with HPLC-grade water, and the resulting membrane-free hemolysates loaded into a System Gold 126 Solvent Module instrument (Beckman Coulter, Fullerton, Calif.). Hemoglobins were separated on a weak cation-exchange PolyCAT A column (PolyLC, Inc, Columbia, Md.), and detected at a wavelength of 415 nm. The Hbs were bound to the column with mobile phase A (20 mmol/L Bis-Tris, 2 mmol/L KCN, pH 6.96) and eluted with mobile phase B (20 mmol/L Bis-Tris, 2 mmol/L KCN, 200 mmol/L NaCl, pH 6.55). Single chain quantification was assessed via reversed-phase HPLC. Hb samples in this case were injected on a Hitachi D-7000 HSM Series apparatus (Hitachi Instruments, San Jose, Calif.) using a Zorbax 5 .mu.m 300SB-C8 300 .ANG., LC 150.times.2.1 mm column (Agilent Technologies, Santa Clara, Calif.) and a gradient from 20% to 60% acetonitrile in 0.1% trifluoroacetic acid in 25 minutes, with UV detection at 215 nm. Serial dilutions of a solution with known concentrations of HbA and HbF (Analytical Control System, Inc, Fishers, Ind.) were used to generate a calibration curve, where the peak areas were plotted against the concentration values. Types and relative quantity of Hbs in samples were assessed by comparison to standard hemoglobin controls.

EXAMPLE 7

[0071] This Example demonstrates that introducing the lentiviral vector ALS10 into CD34+ cells from .beta.0/0 phenotype samples, and thus the most severe thalassemic specimens, results in statistically significantly elevated levels of HbA produced by erythrocytes derived from the modified CD34+ cells. The elevation in HbA is relative to a previously describe construct, which is used in this Example as a comparison control (AnkT9W, from Breda et al, Plos One, 2012), which did not include a complete intron 2. Thus, when compared to the previously described construct, ALS10 showed significant and unexpected improvement, as demonstrated by the results depicted in FIG. 12. To obtain the results presented in FIG. 12, the following materials and methods were used.

[0072] Vector Production and Titration

[0073] Viral stocks were generated by co-transfection of the gene transfer plasmid (GG1-SA) together with the envelope plasmid (VSV-G), the packaging plasmid (pMDLg/p RRE), and the pRSV-REV vector into 293T cells. An aliquot (5.times.10.sup.6) of 293T cells was seeded into cell culture dishes (10 cm) 24 hours prior to transfection in Iscove's modification of Eagle's medium (DMEM, Cellgro, Manassas, Va.) with 10% fetal bovine serum, 100 U/ml penicillin, and 100 mg/ml streptomycin, at 37.degree. C. under 5% CO2. The culture medium was changed 2 hours prior to transfection. The precipitate was formed by adding the plasmids to 450 uL of 0.1.times.TE (0.1.times.TE is 10 mM Tris plus 1 mM EDTA) and 50 .mu.L of 2 M CaCl2, then adding 500 .mu.L of 2.times. HEPES-buffered saline (281 mM NaCl, 100 mM HEPES, 1.5 mM Na2HPO4) dropwise after which the precipitate was vortexed and immediately added to the cultures. The medium (10 ml) was replaced after 16 hours. Viral supernatants were collected at 24 and 48 hours, cleared by low speed centrifugation, and filtered through cellulose acetate (0.2 .mu.m). Following concentration by ultracentrifugation, serial dilutions of concentrated virus (5; 0.5 and 0.05 .mu.L, respectively) were used to infect 1.times.10.sup.5 NIH 3T3 cells (ATCC, Manassas, Va.) in 1 mL of transfection buffer complemented with polybrene (Millipore, Billerica, Mass.) at a final concentration of 8 .mu.g/mL. Genomic DNA was extracted after 3 days (Qiagen kit, Valencia, Calif.). The multiplicity of infection (MOI) was calculated using the following formula: number of cells (1.times.10.sup.5).times.dilution factor (1 mL/.mu.L viral preparation)'VCN (measured via real-time PCR, using oligos for Psi element and ID gene, by Real Time PCR).

[0074] Two-Phase Liquid Cultures, Benzidine Staining and Transduction

[0075] Consented patients with .beta.0/0 and healthy individuals donated between 20 and 30 mL of peripheral blood; alternatively 30 to 60 mL of peripheral blood from .beta.0/0 patients were obtained from the discarded blood from the red cell exchange therapeutic procedure. We selected CD34+ cells by immunomagnetic separation, using the CD34 microbeads kit (Miltenyi Biotec Inc., Auburn, Calif.) and then expanded these cells following a modified version of the protocol described by Leberbauer and colleagues. Cells were seeded in 5 mL of serum-free StemSpan with 50 .mu.L of StemSpan CC-100 cytokine cocktail (both from Stemcell Technologies, Vancouver, BA, Canada), 2 U/mL Erytrhopoietin (Amgen, Thousand Oaks, Calif.), 10-6 M dexamethasone (Sigma) and 1% penicillin streptomycin. CD34+ cultures were kept undifferentiated by refreshing the medium twice a week and density gradient centrifugation was used to remove both dead and spontaneously differentiating cells. At this stage cells were either frozen (with 50% characterized Hyclone FBS, 10% DMSO, Sigma, and 40% Iscove's Modified DMEM, Cellgro), or used for experiments. After 10 days in phase I, cells were transferred into phase II media containing .alpha.-modified essential medium supplemented with 30% fetal calf serum and 10-5 M .beta.-mercaptoethanol. Erythropoietin was added (5 U/mL) to stimulate erythroid differentiation. Cells were infected with serial dilutions of the virus. Cells were collected on day 7-10 of phase II for all analyses. The level of differentiation was assessed by benzidine staining.

[0076] High Performance Liquid Chromatography (HPLC)

[0077] Cell pellets were lysed with HPLC-grade water and loaded into a System Gold 126 Solvent Module instrument (Beckman Coulter, Fullerton, Calif.). Hemoglobins were separated on a PolyCAT A column (PolyLC, Inc, Columbia, Md.), which is packed with silica-based material with a bonded coating of polyaspartic acid, and detected at a wavelength of 415 nm. The Hbs were bound to the column with mobile phase A (20 mmol/L Bis-Tris, 2 mmol/L KCN, pH 6.96) and eluted with mobile phase B (20 mmol/L Bis-Tris, 2 mmol/L KCN, 200 mmol/L NaCl, pH 6.55). Serial dilutions of a solution with known concentrations of HbA and HbF (Analytical Control System, Inc, Fishers, Ind.) were used to generate a calibration curve, where the absorbance detected at 415 nm was plotted against the concentration values. Types and quantity of hemoglobins in samples were assessed by comparison to standard hemoglobin controls.

[0078] While the invention has been described through specific embodiments, routine modifications will be apparent to those skilled in the art and such modifications are intended to be within the scope of the present invention.

Sequence CWU 1

1

2912875DNAartificial sequenceZF-Ldb1 vector as DNA equivalent of RNA lentivirus 1cgcgttcgaa ggggcaacca ggggtccgcg cgccgaggcc tggggagcgg ggcctcctgg 60ggttggggga ggaggtgctc ttgtaatctg cggtggtccc caggcgggcg ccacccctcc 120gcccgcccgt gccgggagcg cccggcccga cagcaagcgc ctctggggcc gataaggccc 180tcgggggcct ggcccgcacg tcacaggccc cgcagaggct gcggtgagtc cgccagcccc 240agctgctcct cctcaagccc ccaaggccct tcggcggcaa ttcccaccgg tcgccaccat 300ggcccaggcg gccttggagc ccggagagaa accctacaaa tgcccggagt gtggaaagtc 360cttcagcgac tgtagggacc tggcacgcca ccagcgaacc cacacaggcg agaagcccta 420taaatgtcca gagtgtggaa agagcttctc tagaaacgat gcactgactg agcaccaacg 480cacccatacc ggggaaaaac cttataaatg tcctgagtgt ggtaaaagtt tttctcaatt 540ggctcatctc cgcgctcacc aacgcacgca tactggtgag aagccctata agtgccccga 600atgcggcaag agtttttctc aaagcgggga ccttagaaga caccaaagaa cccataccgg 660cgaaaaacct tacaagtgtc ccgagtgcgg aaaatctttt tcacgcaaag acaatttgaa 720gaaccaccag cggacacaca ccggtgaaaa gccttacaaa tgtcccgaat gtggcaagtc 780attctcagac cccggagccc tggtgcgcca tcagcgcact cacactggca agaagactag 840cggccaggcc ggccaggcta gcccgaaaaa gaaacgcaaa gttgggcgcg ccctggatcg 900ggatgtgggc ccaactccca tgtacccacc tacatacctg gagcctggga tcgggaggca 960cacaccatat ggtaaccaaa ccgactatag aatatttgag cttaacaaac ggctacagaa 1020ctggacagag gagtgtgaca atctctggtg ggatgctttc acaactgagt tctttgaaga 1080tgacgccatg ctgaccatca ctttctgctt ggaggatgga ccaaagagat ataccattgg 1140ccggaccctg ataccacgct acttccgaag catttttgag gggggtgcca cagagctgta 1200ctacgtgctc aagcacccca aggaggcatt ccacagcaac ttcgtgtccc tcgactgtga 1260ccagggcagc atggtgaccc agcacggcaa acccatgttt acccaggtgt gtgtggaagg 1320ccggttgtac ctggagttca tgtttgacga catgatgcgg ataaagacgt ggcacttcag 1380catccggcaa cacagagagc tcatccccag aagtatcctg gccatgcacg cccaggaccc 1440ccagatgctg gatcagctgt ccaaaaacat tacccggtgt gggctgtcct taattaacta 1500cccgtacgac gttccggact acgcttcttg aatcggtagg aattaattct gcagcggccg 1560cggatccgcc cctctccctc ccccccccct aacgttactg gccgaagccg cttggaataa 1620ggccggtgtg cgtttgtcta tatgttattt tccaccatat tgccgtcttt tggcaatgtg 1680agggcccgga aacctggccc tgtcttcttg acgagcattc ctaggggtct ttcccctctc 1740gccaaaggaa tgcaaggtct gttgaatgtc gtgaaggaag cagttcctct ggaagcttct 1800tgaagacaaa caacgtctgt agcgaccctt tgcaggcagc ggaacccccc acctggcgac 1860aggtgcctct gcggccaaaa gccacgtgta taagatacac ctgcaaaggc ggcacaaccc 1920cagtgccacg ttgtgagttg gatagttgtg gaaagagtca aatggctctc ctcaagcgta 1980ttcaacaagg ggctgaagga tgcccagaag gtaccccatt gtatgggatc tgatctgggg 2040cctcggtgca catgctttac atgtgtttag tcgaggttaa aaaaacgtct aggccccccg 2100aaccacgggg acgtggtttt cctttgaaaa acacgatgat aatatggcca caaccatggt 2160gagcaagggc gaggagctgt tcaccggggt ggtgcccatc ctggtcgagc tggacggcga 2220cgtaaacggc cacaagttca gcgtgtccgg cgagggcgag ggcgatgcca cctacggcaa 2280gctgaccctg aagttcatct gcaccaccgg caagctgccc gtgccctggc ccaccctcgt 2340gaccaccctg acctacggcg tgcagtgctt cagccgctac cccgaccaca tgaagcagca 2400cgacttcttc aagtccgcca tgcccgaagg ctacgtccag gagcgcacca tcttcttcaa 2460ggacgacggc aactacaaga cccgcgccga ggtgaagttc gagggcgaca ccctggtgaa 2520ccgcatcgag ctgaagggca tcgacttcaa ggaggacggc aacatcctgg ggcacaagct 2580ggagtacaac tacaacagcc acaacgtcta tatcatggcc gacaagcaga agaacggcat 2640caaggtgaac ttcaagatcc gccacaacat cgaggacggc agcgtgcagc tcgccgacca 2700ctaccagcag aacaccccca tcggcgacgg ccccgtgctg ctgcccgaca accactacct 2760gagcacccag tccgccctga gcaaagaccc caacgagaag cgcgatcaca tggtcctgct 2820ggagttcgtg accgccgccg ggatcactct cggcatggac gagctgtaca agtaa 28752410PRTartificial sequenceZF-LDB1 fusion 2Met Ala Gln Ala Ala Leu Glu Pro Gly Glu Lys Pro Tyr Lys Cys Pro1 5 10 15Glu Cys Gly Lys Ser Phe Ser Asp Cys Arg Asp Leu Ala Arg His Gln 20 25 30Arg Thr His Thr Gly Glu Lys Pro Tyr Lys Cys Pro Glu Cys Gly Lys 35 40 45Ser Phe Ser Arg Asn Asp Ala Leu Thr Glu His Gln Arg Thr His Thr 50 55 60Gly Glu Lys Pro Tyr Lys Cys Pro Glu Cys Gly Lys Ser Phe Ser Gln65 70 75 80Leu Ala His Leu Arg Ala His Gln Arg Thr His Thr Gly Glu Lys Pro 85 90 95Tyr Lys Cys Pro Glu Cys Gly Lys Ser Phe Ser Gln Ser Gly Asp Leu 100 105 110Arg Arg His Gln Arg Thr His Thr Gly Glu Lys Pro Tyr Lys Cys Pro 115 120 125Glu Cys Gly Lys Ser Phe Ser Arg Lys Asp Asn Leu Lys Asn His Gln 130 135 140Arg Thr His Thr Gly Glu Lys Pro Tyr Lys Cys Pro Glu Cys Gly Lys145 150 155 160Ser Phe Ser Asp Pro Gly Ala Leu Val Arg His Gln Arg Thr His Thr 165 170 175Gly Lys Lys Thr Ser Gly Gln Ala Gly Gln Ala Ser Pro Lys Lys Lys 180 185 190Arg Lys Val Gly Arg Ala Leu Asp Arg Asp Val Gly Pro Thr Pro Met 195 200 205Tyr Pro Pro Thr Tyr Leu Glu Pro Gly Ile Gly Arg His Thr Pro Tyr 210 215 220Gly Asn Gln Thr Asp Tyr Arg Ile Phe Glu Leu Asn Lys Arg Leu Gln225 230 235 240Asn Trp Thr Glu Glu Cys Asp Asn Leu Trp Trp Asp Ala Phe Thr Thr 245 250 255Glu Phe Phe Glu Asp Asp Ala Met Leu Thr Ile Thr Phe Cys Leu Glu 260 265 270Asp Gly Pro Lys Arg Tyr Thr Ile Gly Arg Thr Leu Ile Pro Arg Tyr 275 280 285Phe Arg Ser Ile Phe Glu Gly Gly Ala Thr Glu Leu Tyr Tyr Val Leu 290 295 300Lys His Pro Lys Glu Ala Phe His Ser Asn Phe Val Ser Leu Asp Cys305 310 315 320Asp Gln Gly Ser Met Val Thr Gln His Gly Lys Pro Met Phe Thr Gln 325 330 335Val Cys Val Glu Gly Arg Leu Tyr Leu Glu Phe Met Phe Asp Asp Met 340 345 350Met Arg Ile Lys Thr Trp His Phe Ser Ile Arg Gln His Arg Glu Leu 355 360 365Ile Pro Arg Ser Ile Leu Ala Met His Ala Gln Asp Pro Gln Met Leu 370 375 380Asp Gln Leu Ser Lys Asn Ile Thr Arg Cys Gly Leu Ser Leu Ile Asn385 390 395 400Tyr Pro Tyr Asp Val Pro Asp Tyr Ala Ser 405 410313550DNAartificial sequenceALS10 as DNA equilvalent of RNA lentiviral vector 3ctgtcagacc aagtttactc atatatactt tagattgatt taaaacttca tttttaattt 60aaaaggatct aggtgaagat cctttttgat aatctcatga ccaaaatccc ttaacgtgag 120ttttcgttcc actgagcgtc agaccccgta gaaaagatca aaggatcttc ttgagatcct 180ttttttctgc gcgtaatctg ctgcttgcaa acaaaaaaac caccgctacc agcggtggtt 240tgtttgccgg atcaagagct accaactctt tttccgaagg taactggctt cagcagagcg 300cagataccaa atactgtcct tctagtgtag ccgtagttag gccaccactt caagaactct 360gtagcaccgc ctacatacct cgctctgcta atcctgttac cagtggctgc tgccagtggc 420gataagtcgt gtcttaccgg gttggactca agacgatagt taccggataa ggcgcagcgg 480tcgggctgaa cggggggttc gtgcacacag cccagcttgg agcgaacgac ctacaccgaa 540ctgagatacc tacagcgtga gctatgagaa agcgccacgc ttcccgaagg gagaaaggcg 600gacaggtatc cggtaagcgg cagggtcgga acaggagagc gcacgaggga gcttccaggg 660ggaaacgcct ggtatcttta tagtcctgtc gggtttcgcc acctctgact tgagcgtcga 720tttttgtgat gctcgtcagg ggggcggagc ctatggaaaa acgccagcaa cgcggccttt 780ttacggttcc tggccttttg ctggcctttt gctcacatgt tctttcctgc gttatcccct 840gattctgtgg ataaccgtat taccgccttt gagtgagctg ataccgctcg ccgcagccga 900acgaccgagc gcagcgagtc agtgagcgag gaagcggaag agcgcccaat acgcaaaccg 960cctctccccg cgcgttggcc gattcattaa tgcagctggc acgacaggtt tcccgactgg 1020aaagcgggca gtgagcgcaa cgcaattaat gtgagttagc tcactcatta ggcaccccag 1080gctttacact ttatgcttcc ggctcgtatg ttgtgtggaa ttgtgagcgg ataacaattt 1140cacacaggaa acagctatga ccatgattac gccaagcgcg caattaaccc tcactaaagg 1200gaacaaaagc tggagctgca agcttggcca ttgcatacgt tgtatccata tcataatatg 1260tacatttata ttggctcatg tccaacatta ccgccatgtt gacattgatt attgactagt 1320tattaatagt aatcaattac ggggtcatta gttcatagcc catatatgga gttccgcgtt 1380acataactta cggtaaatgg cccgcctggc tgaccgccca acgacccccg cccattgacg 1440tcaataatga cgtatgttcc catagtaacg ccaataggga ctttccattg acgtcaatgg 1500gtggagtatt tacggtaaac tgcccacttg gcagtacatc aagtgtatca tatgccaagt 1560acgcccccta ttgacgtcaa tgacggtaaa tggcccgcct ggcattatgc ccagtacatg 1620accttatggg actttcctac ttggcagtac atctacgtat tagtcatcgc tattaccatg 1680gtgatgcggt tttggcagta catcaatggg cgtggatagc ggtttgactc acggggattt 1740ccaagtctcc accccattga cgtcaatggg agtttgtttt ggcaccaaaa tcaacgggac 1800tttccaaaat gtcgtaacaa ctccgcccca ttgacgcaaa tgggcggtag gcgtgtacgg 1860tgggaggtct atataagcag agctcgttta gtgaaccggg gtctctctgg ttagaccaga 1920tctgagcctg ggagctctct ggctaactag ggaacccact gcttaagcct caataaagct 1980tgccttgagt gcttcaagta gtgtgtgccc gtctgttgtg tgactctggt aactagagat 2040ccctcagacc cttttagtca gtgtggaaaa tctctagcag tggcgcccga acagggacct 2100gaaagcgaaa gggaaaccag agctctctcg acgcaggact cggcttgctg aagcgcgcac 2160ggcaagaggc gaggggcggc gactggtgag tacgccaaaa attttgacta gcggaggcta 2220gaaggagaga gatgggtgcg agagcgtcag tattaagcgg gggagaatta gatcgcgatg 2280ggaaaaaatt cggttaaggc cagggggaaa gaaaaaatat aaattaaaac atatagtatg 2340ggcaagcagg gagctagaac gattcgcagt taatcctggc ctgttagaaa catcagaagg 2400ctgtagacaa atactgggac agctacaacc atcccttcag acaggatcag aagaacttag 2460atcattatat aatacagtag caaccctcta ttgtgtgcat caaaggatag agataaaaga 2520caccaaggaa gctttagaca agatagagga agagcaaaac aaaagtaaga ccaccgcaca 2580gcaagcggcc gctgatcttc agacctggag gaggagatat gagggacaat tggagaagtg 2640aattatataa atataaagta gtaaaaattg aaccattagg agtagcaccc accaaggcaa 2700agagaagagt ggtgcagaga gaaaaaagag cagtgggaat aggagctttg ttccttgggt 2760tcttgggagc agcaggaagc actatgggcg cagcctcaat gacgctgacg gtacaggcca 2820gacaattatt gtctggtata gtgcagcagc agaacaattt gctgagggct attgaggcgc 2880aacagcatct gttgcaactc acagtctggg gcatcaagca gctccaggca agaatcctgg 2940ctgtggaaag atacctaaag gatcaacagc tcctggggat ttggggttgc tctggaaaac 3000tcatttgcac cactgctgtg ccttggaatg ctagttggag taataaatct ctggaacaga 3060tttggaatca cacgacctgg atggagtggg acagagaaat taacaattac acaagcttaa 3120tacactcctt aattgaagaa tcgcaaaacc agcaagaaaa gaatgaacaa gaattattgg 3180aattagataa atgggcaagt ttgtggaatt ggtttaacat aacaaattgg ctgtggtata 3240taaaattatt cataatgata gtaggaggct tggtaggttt aagaatagtt tttgctgtac 3300tttctatagt gaatagagtt aggcagggat attcaccatt atcgtttcag acccacctcc 3360caaccccgag gggacccgac aggcccgaag gaatagaaga agaaggtgga gagagagaca 3420gagacagatc cattcgatta gtgaacggat ctcgacggta tcggttaact tttaaaagaa 3480aaggggggat tggggggtac agtgcagggg aaagaatagt agacataata gcaacagaca 3540tacaaactaa agaattacaa aaacaaatta caaaaattca aaattttatc gataagcttg 3600ggagttccgc gtgttggggg tggaccatcc tctaggtatt gaataagaaa aatgaagtta 3660aggtggttga tggtaacact atgctaataa ctgcagagcc agaagcacca taagggacat 3720gataagggag ccagcagacc tctgatctct tcctgaatgc taatcttaaa catcctgagg 3780aagaatggga cttccatttg gggtgggcct atgatagggt aataagacag tagtgaatat 3840caagctacaa aaagccccct ttcaaattct tctcagtcct aacttttcat actaagccca 3900gtccttccaa agcagactgt gaaagagtga tagttccggg agactagcac tgcagattcc 3960gggtcactgt gagtggggga ggcagggaag aagggctcac aggacagtca aaccatgccc 4020cctgtttttc cttcttcaag tagacctcta taagacaaca gagacaacta aggctgagtg 4080gccaggcgag gagaaaccat ctcgccgtaa aacatggaag gaacacttca ggggaaaggt 4140ggtatctcta agcaagagaa ctgagtggag tcaaggctga gagatgcagg ataagcaaat 4200gggtagtgaa aagacattca tgaggacagc taaaacaata agtaatgtaa aatacagcat 4260agcaaaactt taacctccaa atcaagcctc tacttgaatc cttttctgag ggatgaataa 4320ggcataggca tcaggggctg ttgccaatgt gcattagctg tttgcagcct caccttcttt 4380catggagttt aagatatagt gtattttccc aaggtttgaa ctagctcttc atttctttat 4440gttttaaatg cactgacctc ccacattccc tttttagtaa aatattcaga aataatttaa 4500atacatcatt gcaatgaaaa taaatgtttt ttattaggca gaatccagat gctcaaggcc 4560cttcataata tcccccagtt tagtagttgg acttagggaa caaaggaacc tttaatagaa 4620attggacagc aagaaagcga gcttagtgat acttgtgggc cagggcatta gccacaccag 4680ccaccacttt ctgataggca gcctgcactg gtggggtgaa ttctttgcca aagtgatggg 4740ccagcacaca gaccagcacg ttgcccagga gctgtgggag gaagataaga ggtatgaaca 4800tgattagcaa aagggcctag cttggactca gaataatcca gccttatccc aaccataaaa 4860taaaagcaga atggtagctg gattgtagct gctattagca atatgaaacc tcttacatca 4920gttacaattt atatgcagaa atatttatat gcagaaatat tgctattgcc ttaacccaga 4980aattatcact gttattcttt agaatggtgc aaagaggcat gatacattgt atcattattg 5040ccctgaaaga aagagattag ggaaagtatt agaaataaga taaacaaaaa agtatattaa 5100aagaagaaag cattttttaa aattacaaat gcaaaattac cctgatttgg tcaatatgtg 5160tacacatatt aaaacattac actttaaccc ataaatatgt ataatgatta tgtatcaatt 5220aaaaataaaa gaaaataaag tagggagatt atgaatatgc aaataagcac acatatattc 5280caaatagtaa tgtactaggc agactgtgta aagttttttt ttaagttact taatgtatct 5340cagagatatt tccttttgtt atacacaatg ttaaggcatt aagtataata gtaaaaattg 5400cggagaagaa aaaaaaagaa agcaagaatt aaacaaaaga aaacaattgt tatgaacagc 5460aaataaaaga aactaaaacg atcctgagac ttccacactg atgcaatcat tcgtctgttt 5520cccattctaa actgtaccct gttacttctc cccttcctat gacatgaact taaccataga 5580aaagaagggg aaagaaaaca tcaagggtcc catagactca ccctgaagtt ctcaggatcc 5640acgtgcagct tgtcacagtg cagctcactc agtgtggcaa aggtgccctt gaggttgtcc 5700aggtgagcca ggccatcact aaaggcaccg agcactttct tgccatgagc cttcacctta 5760gggttgccca taacagcatc aggagtggac agatccccaa aggactcaaa gaacctctgg 5820gtccaagggt agaccaccag cagcctaagg gtgggaaaat agaccaatag gcagagagag 5880tcagtgccta tcagaaaccc aagagtcttc tctgtctcca catgcccagt ttctattggt 5940ctccttaaac ctgtcttgta accttgatac caacctgccc agggcctcac caccaacttc 6000atccacgttc accttgcccc acagggcagt aacggcagac ttctcctcag gagtcaggtg 6060caccatggtg tctgtttgag gttgctagtg aacacagttg tgtcagaagc aaatgtaagc 6120aatagatggc tctgccctga cttttatgcc cagccctggc tcctgccctc cctgctcctg 6180ggagtagatt ggccaaccct agggtgtggc tccacagggt gaggtctaag tgatgacagc 6240cgtacctgtc cttggctctt ctggcactgg cttaggagtt ggacttcaaa ccctcagccc 6300tccctctaag atatatctct tggccccata ccatcagtac aaattgctac taaaaacatc 6360ctcctttgca agtgtattta cgtaatattt ggaatcacag cttggtaagc atattgaaga 6420tcgttttccc aattttctta ttacacaaat aagaaattga tgcactaaaa gtggaagagt 6480tttgtctacc ataattcagc tttgggatat gtagatggat ctcttcctgc gtctccagaa 6540tatgcaaaat acttacagga cagaatggat gaaaactcta cctcagttct aagcatatct 6600tctccttatt tggattaaaa ccttctggta agaaaagaaa aaaaatatat atatatatgt 6660gtatatatac acacatacat atacatatat atgcattcat ttgttgttgt ttttcttaat 6720ttgctcatgg tatatgtgta tatatatata tatatattca ggaaataata tattctagaa 6780tatgtcacat tctgtctcag gcatccattt tctttatgat gccgtttgag gtggagtttt 6840agtcaggtgg tcagcttctc cttttttttg ccatctgccc tgtaagcatc ctgctgggga 6900cccagatagg agtcatcact ctaggctgag aacatctggg cacacaccct aagcctcagc 6960atgactcatc atgactcagc attgctgtgc ttgagccaga aggtttgctt agaaggttac 7020acagaaccag aaggcggggg tggggcactg accccgacag gggcctggcc agaactgctc 7080atgcttggac tatgggaggt cactaatgga gacacacaga aatgtaacag gaactaagga 7140aaaactgaag cttatttaat cagagatgag atgctggaag ggatagaggg agctgagctt 7200gtaaaaagta tagtaatcat tcagcaaatg gttttgaagc acctgctgga tgctaaacac 7260tattttcagt gcttgaatca taaataagaa taaaacatgt atcttattcc ccacaagagt 7320ccaagtaaaa aataacagtt aattataatg tgctctgtcc cccaggctgg agtgcagtgg 7380cacgatctca gctcactgca acctccgcct cccgggttca agcaattctc ctgcctcagc 7440caccctaata gctgggatta caggtgcaca ccaccatgcc aggctaattt ttgtactttt 7500tgtagaggca gggtatcacc atgttgtcca agatggtctt gaactcctga gctccaagca 7560gtccacccac ctcagcctcc caaagtgcta tctgcggccg cctatctgta ccactagtct 7620cgagaagctt tcattaaaaa aagtctaacc agctgcattc gactttgact gcagcagctg 7680gttagaaggt tctactggag gagggtccca gcccattgct aaattaacat caggctctga 7740gactggcagt atatctctaa cagtggttga tgctatcttc tggaacttgc ctgctacatt 7800gagaccactg acccatacat aggaagccca tagctctgtc ctgaactgtt aggccactgg 7860tccagagagt gtgcatctcc tttgatcctc ataataaccc tatgagatag acacaattat 7920tactcttact ttatagatga tgatcctgaa aacataggag tcaaggcact tgcccctagc 7980tgggggtata ggggagcagt cccatgtagt agtagaatga aaaatgctgc tatgctgtgc 8040ctcccccacc tttcccatgt ctgccctcta ctcatggtct atctctcctg gctcctggga 8100gtcatggact ccacccagca ccaccaacct gacctaacca cctatctgag cctgccagcc 8160tataacccat ctgggccctg atagctggtg gccagccctg accccacccc accctccctg 8220gaacctctga tagacacatc tggcacacca gctcgcaaag tcaccgtgag ggtcttgtgt 8280ttgctgagtc aaaattcctt gaaatccaag tccttagaga ctcctgctcc caaatttaca 8340gtcatagact tcttcatggc tgtctccttt atccacagaa tgattccttt gcttcattgc 8400cccatccatc tgatcctcct catcagtgca gcacagggcc catgagcagt agctgcagag 8460tctcacatag gtctggcact gcctctgaca tgtccgacct taggcaaatg cttgactctt 8520ctgagctcag tcttgtcatg gcaaaataaa gataataata gtgttttttt atggagttag 8580cgtgaggatg gaaaacaata gcaaaattga ttagactata aaaggtctca acaaatagta 8640gtagatttta tcatccatta atccttccct ctcctctctt actcatccca tcacgtatgc 8700ctcttaattt tcccttacct ataataagag ttattcctct tattatattc ttcttatagt 8760gattctggat attaaagtgg gaatgagggg caggccacta acgaagaaga tgtttctcaa 8820agaagccatt ctccccacat agatcatctc agcagggttc aggaagataa aggaggatca 8880aggtcgaagg taggaactaa ggaagaacac tgggcaagtg gatcctgagc cccttttcct 8940ctaactgaaa gaaggaaaaa aaaaatggaa cccaaaatat tctacatagt ttccatgtca 9000cagccagggc tgggcagtct cctgttattt cttttaaaat aaatatatca tttaaatgca 9060taaataagca aaccctgctc gggaatggga gggagagtct ctggagtcca ccccttctcg 9120gccctggctc tgcagatagt gctatcaaag ccctgacaga gccctgccca ttgctgggcc 9180ttggagtgag tcagcctagt agagaggcag ggcaagccat ctcatagctg ctgagtggga 9240gagagaaaag ggctcattgt ctataaactc aggtcatggc tattcttatt ctcacactaa 9300gaaaaagaat gagatgtcta catataccct gcgtcccctc ttgtgtactg gggcccccaa 9360gagctctcta aaagtgatgg caaagtcatt gcgctagatg ccatcccatc tattataaac 9420ctgcatttgt ctccacacac cagtcatgga caataaccct cctcccaggt ccacgtgctt 9480gtctttgtat aatactcaag taatttcgga aaatgtattc

tttcaatctt gttctgttat 9540tcctgtttca atggcttagt agaaaaagta catacttgtt ttcccataaa ttgacaatag 9600acaatttcac atcaatgtct atatgggtcg ttgtgtttgc tgtgtttgca aaaactcaca 9660ataactttat attgttacta ctctaagaaa gttacaacat ggtgaataca agagaaagct 9720attacaagtc cagaaaataa aagttatcat cttgaggcct cagctttcta ggaataatat 9780caatattaca aaattaatct aacaattatg aacagcaatg agataatatg tacaaagtac 9840ccagacctat gtggtagagc atcaaggaag cgcattgcgg agcagttttt tgtttgtttg 9900tttttgtatt ctgtttcgtg aggcaaggtt tcactctgct gtccaggctg gagtgcagtg 9960gcaagatcat gtctcactgc agccttgaca ctacacgtgc tttaagacca atgacttaca 10020aggcagctgt agatcttagc cactttttaa aagaaaaggg gggactggaa gggctaattc 10080actcccaacg aagacaagat ctgctttttg ctagcgcggc cgctctagac tagtggggcc 10140cgtgcaattg aagccggctg gcgccaagct tctctgcagg atgtgcgggc caggcccccg 10200agggccttat cggccccaga ggcgcttgct gtcgggccgg gcgctcccgg cacgggcggg 10260cggaggggtg gcgcccgcct ggggaccgca gattacaaga gcacctcctc ccccaacccc 10320aggaggcccc gctccccagg cctcggccgg cgcggacccc tggttgcccc ggatgtacag 10380gcatgcgtcg acctcgaggg ggggcccggt accgctagca ctgggtctct ctggttagac 10440cagatctgag cctgggagct ctctggctaa ctagggaacc cactgcttaa gcctcaataa 10500agcttgcctt gagtgcttca agtagtgtgt gcccgtctgt tgtgtgactc tggtaactag 10560agatccctca gaccctttta gtcagtgtgg aaaatctcta gcaggtcgac aatcaacctc 10620tggattacaa aatttgtgaa agattgactg gtattcttaa ctatgttgct ccttttacgc 10680tatgtggata cgctgcttta atgcctttgt atcatgctat tgcttcccgt atggctttca 10740ttttctcctc cttgtataaa tcctggttgc tgtctcttta tgaggagttg tggcccgttg 10800tcaggcaacg tggcgtggtg tgcactgtgt ttgctgacgc aacccccact ggttggggca 10860ttgccaccac ctgtcagctc ctttccggga ctttcgcttt ccccctccct attgccacgg 10920cggaactcat cgccgcctgc cttgcccgct gctggacagg ggctcggctg ttgggcactg 10980acaattccgt ggtgttgtcg gggaagctga cgtcctttcc atggctgctc gcctgtgttg 11040ccacctggat tctgcgcggg acgtccttct gctacgtccc ttcggccctc aatccagcgg 11100accttccttc ccgcggcctg ctgccggctc tgcggcctct tccgcgtctt cgccttcgcc 11160ctcagacgag tcggatctcc ctttgggccg cctccccgcc tggaattcga gctcggtacc 11220tgatcagcct cgactgtgcc ttctagttgc cagccatctg ttgtttgccc ctcccccgtg 11280ccttccttga ccctggaagg tgccactccc actgtccttt cctaataaaa tgaggaaatt 11340gcatcgcatt gtctgagtag gtgtcattct attctggggg gtggggtggg gcaggacagc 11400aagggggagg attgggaaga caatagcagg catgctgggg atgcggtggg ctctatggct 11460tctgaggaaa gaaccagctg gggctcgaga tccactagtt ctagcctcga ggctagagcg 11520gccgccaccg cggtagtagt tcatgtcatc ttattattca gtatttataa cttgcaaaga 11580aatgaatatc agagagtgag aggaacttgt ttattgcagc ttataatggt tacaaataaa 11640gcaatagcat cacaaatttc acaaataaag catttttttc actgcattct agttgtggtt 11700tgtccaaact catcaatgta tcttatcatg tctggctcta gctatcccgc ccctaactcc 11760gcccagttcc gcccattctc cgccccatgg ctgactaatt ttttttattt atgcagaggc 11820cgaggccgcc tcggcctctg agctattcca gaagtagtga ggaggctttt ttggaggcct 11880aggcttttgc gtcgagacgt acccaattcg ccctatagtg agtcgtatta cgcgcgctca 11940ctggccgtcg ttttacaacg tcgtgactgg gaaaaccctg gcgttaccca acttaatcgc 12000cttgcagcac atcccccttt cgccagctgg cgtaatagcg aagaggcccg caccgatcgc 12060ccttcccaac agttgcgcag cctgaatggc gaatggcgcg acgcgccctg tagcggcgca 12120ttaagcgcgg cgggtgtggt ggttacgcgc agcgtgaccg ctacacttgc cagcgcccta 12180gcgcccgctc ctttcgcttt cttcccttcc tttctcgcca cgttcgccgg ctttccccgt 12240caagctctaa atcgggggct ccctttaggg ttccgattta gtgctttacg gcacctcgac 12300cccaaaaaac ttgattaggg tgatggttca cgtagtgggc catcgccctg atagacggtt 12360tttcgccctt tgacgttgga gtccacgttc tttaatagtg gactcttgtt ccaaactgga 12420acaacactca accctatctc ggtctattct tttgatttat aagggatttt gccgatttcg 12480gcctattggt taaaaaatga gctgatttaa caaaaattta acgcgaattt taacaaaata 12540ttaacgttta caatttccca ggtggcactt ttcggggaaa tgtgcgcgga acccctattt 12600gtttattttt ctaaatacat tcaaatatgt atccgctcat gagacaataa ccctgataaa 12660tgcttcaata atattgaaaa aggaagagta tgagtattca acatttccgt gtcgccctta 12720ttcccttttt tgcggcattt tgccttcctg tttttgctca cccagaaacg ctggtgaaag 12780taaaagatgc tgaagatcag ttgggtgcac gagtgggtta catcgaactg gatctcaaca 12840gcggtaagat ccttgagagt tttcgccccg aagaacgttt tccaatgatg agcactttta 12900aagttctgct atgtggcgcg gtattatccc gtattgacgc cgggcaagag caactcggtc 12960gccgcataca ctattctcag aatgacttgg ttgagtactc accagtcaca gaaaagcatc 13020ttacggatgg catgacagta agagaattat gcagtgctgc cataaccatg agtgataaca 13080ctgcggccaa cttacttctg acaacgatcg gaggaccgaa ggagctaacc gcttttttgc 13140acaacatggg ggatcatgta actcgccttg atcgttggga accggagctg aatgaagcca 13200taccaaacga cgagcgtgac accacgatgc ctgtagcaat ggcaacaacg ttgcgcaaac 13260tattaactgg cgaactactt actctagctt cccggcaaca attaatagac tggatggagg 13320cggataaagt tgcaggacca cttctgcgct cggcccttcc ggctggctgg tttattgctg 13380ataaatctgg agccggtgag cgtgggtctc gcggtatcat tgcagcactg gggccagatg 13440gtaagccctc ccgtatcgta gttatctaca cgacggggag tcaggcaact atggatgaac 13500gaaatagaca gatcgctgag ataggtgcct cactgattaa gcattggtaa 135504754DNAhuman 4cggctgtcat cacttagacc tcaccctgtg gagccacacc ctagggttgg ccaatctact 60cccaggagca gggagggcag gagccagggc tgggcataaa agtcagggca gagccatcta 120ttgcttacat ttgcttctga cacaactgtg ttcactagca acctcaaaca gacaccatgg 180tgcacctgac tcctgaggag aagtctgccg ttactgccct gtggggcaag gtgaacgtgg 240atgaagttgg tggtgaggcc ctgggcaggc tgctggtggt ctacccttgg acccagaggt 300tctttgagtc ctttggggat ctgtccactc ctgatgctgt tatgggcaac cctaaggtga 360aggctcatgg caagaaagtg ctcggtgcct ttagtgatgg cctggctcac ctggacaacc 420tcaagggcac ctttgccaca ctgagtgagc tgcactgtga caagctgcac gtggatcctg 480agaacttcag gctcctgggc aacgtgctgg tctgtgtgct ggcccatcac tttggcaaag 540aattcacccc accagtgcag gctgcctatc agaaagtggt ggctggtgtg gctaatgccc 600tggcccacaa gtatcactaa gctcgctttc ttgctgtcca atttctatta aaggttcctt 660tgttccctaa gtccaactac taaactgggg gatattatga agggccttga gcatctggat 720tctgcctaat aaaaaacatt tattttcatt gcaa 7545147PRThuman 5Met Val His Leu Thr Pro Glu Glu Lys Ser Ala Val Thr Ala Leu Trp1 5 10 15Gly Lys Val Asn Val Asp Glu Val Gly Gly Glu Ala Leu Gly Arg Leu 20 25 30Leu Val Val Tyr Pro Trp Thr Gln Arg Phe Phe Glu Ser Phe Gly Asp 35 40 45Leu Ser Thr Pro Asp Ala Val Met Gly Asn Pro Lys Val Lys Ala His 50 55 60Gly Lys Lys Val Leu Gly Ala Phe Ser Asp Gly Leu Ala His Leu Asp65 70 75 80Asn Leu Lys Gly Thr Phe Ala Thr Leu Ser Glu Leu His Cys Asp Lys 85 90 95Leu His Val Asp Pro Glu Asn Phe Arg Leu Leu Gly Asn Val Leu Val 100 105 110Cys Val Leu Ala His His Phe Gly Lys Glu Phe Thr Pro Pro Val Gln 115 120 125Ala Ala Tyr Gln Lys Val Val Ala Gly Val Ala Asn Ala Leu Ala His 130 135 140Lys Tyr His145620DNAartificial sequencePrimer 6caagaaagtg ctcggtgcct 20719DNAartificial sequencePrimer 7gcaaaggtgc ccttgaggt 19825DNAartificial sequencePrimer 8tagtgatggc ctggctcacc tggac 25920DNAartificial sequencePrimer 9tccccaccac caagacctac 201019DNAartificial sequencePrimer 10ccttaacctg ggcagagcc 191122DNAartificial sequencePrimer 11tcccgcactt cgacctgagc ca 221221DNAartificial sequencePrimer 12tggcaagaag gtgctgactt c 211319DNAartificial sequencePrimer 13tcactcagct gggcaaagg 191424DNAartificial sequencePrimer 14tgggagatgc cataaagcac ctgg 241524DNAartificial sequencePrimer 15tgatgtgtgt ccattggtgt gagc 241624DNAartificial sequencePrimer 16tgcgaacttg aacgtcagga gtct 241724DNAartificial sequencePrimer 17agctgctttc ggctttctcc ctta 241824DNAartificial sequencePrimer 18cctttgcatt tgcagcagtt cagc 241924DNAartificial sequencePrimer 19tcaaccgatc atccctcaca ctct 242024DNAartificial sequencePrimer 20aatcagcagc gcttccattc aagg 242118DNAartificial sequencePrimer 21gctgcctcca cccaagtg 182221DNAartificial sequencePrimer 22accaactctg ggcagtcaca t 212320DNAartificial sequencePrimer 23agcaaccacc catgcctgcc 202420DNAartificial sequencePrimer 24ctcgggccaa aggcctcacg 202517DNAartificial sequencePrimer 25cggctgttgg gcactga 172619DNAartificial sequencePrimer 26ggaaggtccg ctggattga 192722DNAartificial sequencePrimer 27atggctgctc gcctgtgttg cc 222822DNAartificial sequencePrimer 28aaggtgagca aggtggagat tc 222921DNAartificial sequencePrimer 29ttccgagttc agctccaact g 21

* * * * *


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed