WO2018195297A1 - Optimized lentiviral vector for xla gene therapy - Google Patents

Optimized lentiviral vector for xla gene therapy Download PDF

Info

Publication number
WO2018195297A1
WO2018195297A1 PCT/US2018/028331 US2018028331W WO2018195297A1 WO 2018195297 A1 WO2018195297 A1 WO 2018195297A1 US 2018028331 W US2018028331 W US 2018028331W WO 2018195297 A1 WO2018195297 A1 WO 2018195297A1
Authority
WO
WIPO (PCT)
Prior art keywords
btk
cell
cells
seq
vector
Prior art date
Application number
PCT/US2018/028331
Other languages
English (en)
French (fr)
Inventor
David J. Rawlings
Karen SOMMER
Swati SINGH
Original Assignee
Seattle Children's Hospital (dba Seattle Children's Research Institute)
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Seattle Children's Hospital (dba Seattle Children's Research Institute) filed Critical Seattle Children's Hospital (dba Seattle Children's Research Institute)
Priority to JP2019557373A priority Critical patent/JP7471821B2/ja
Priority to US16/605,740 priority patent/US20200325458A1/en
Priority to CN201880043435.0A priority patent/CN110809627A/zh
Priority to EP18788504.1A priority patent/EP3612238A4/en
Priority to AU2018256412A priority patent/AU2018256412A1/en
Publication of WO2018195297A1 publication Critical patent/WO2018195297A1/en
Priority to JP2024063151A priority patent/JP2024084849A/ja

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4612B-cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1205Phosphotransferases with an alcohol group as acceptor (2.7.1), e.g. protein kinases
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/10Protein-tyrosine kinases (2.7.10)
    • C12Y207/10002Non-specific protein-tyrosine kinase (2.7.10.2), i.e. spleen tyrosine kinase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • A61K48/0058Nucleic acids adapted for tissue specific expression, e.g. having tissue specific promoters as part of a contruct
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector
    • C12N2740/15043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16041Use of virus, viral particle or viral elements as a vector
    • C12N2740/16043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/22Vectors comprising a coding region that has been codon optimised for expression in a respective host
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/008Vector systems having a special element relevant for transcription cell type or tissue specific enhancer/promoter combination
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/46Vector systems having a special element relevant for transcription elements influencing chromatin structure, e.g. scaffold/matrix attachment region, methylation free island

Definitions

  • aspects of the present invention concern compositions and methods for treating, inhibiting or ameliorating X linked agammaglobulinemia (XLA) in subjects that have been identified or selected as being ones that would benefit from a therapy to treat, inhibit, or ameliorate XLA.
  • XLA X linked agammaglobulinemia
  • Exemplary aspects include constructs and methods for gene therapy, which restore or increase Bruton's tyrosine kinase (BTK) expression.
  • BTK Bruton's tyrosine kinase
  • X linked agammaglobulinemia is a rare X-linked genetic disorder resulting from mutations in the Bruton's tyrosine kinase (BTK) gene. These mutations contribute to the failure of afflicted individuals to generate mature B cells and the inability of these B cells to respond to B cell antigen receptor engagement, as well as, other cellular signals. Affected males are unable to generate protective antibody responses to pathogen challenge and eventually succumb to viral or bacterial infection. Current therapy has not changed for over 5 decades and consists of immunoglobulin replacement and targeted antimicrobial agents. Despite this therapy, XLA subjects continue to suffer from chronic infections and are at an increased risk for a range of morbid or life-threatening complications. In rare settings, XLA subjects have been treated with stem cell transplantation without conditioning or using reduced intensity conditioning with variable outcomes. The need for more therapies to inhibit, treat, or ameliorate XLA is manifest.
  • BTK Bruton's tyrosine kinase
  • the constructs set forth herein utilize a truncated ubiquitous chromatin opening element (UCOE) element, a conserved enhancer element derived from intronic regions within the human BTK locus in association with the human BTK proximal promoter to drive expression of a human codon-optimized BTK cDNA.
  • UCOE ubiquitous chromatin opening element
  • LV vectors using this construct in mouse gene therapy experiments mediate sustained BTK expression in B and myeloid cells in primary and secondary transplant recipients and rescue B cell development and function without evidence of viral toxicity.
  • this construct represents a unique LV vector for gene therapy treatment, inhibition, or amelioration of human XLA.
  • a polynucleotide for sustained Bruton's tyrosine kinase (BTK) expression comprising: a first sequence encoding an ubiquitous chromatin opening element (UCOE), a second sequence encoding a promoter and a third sequence encoding BTK.
  • the UCOE is 2kb, 1.5kb, lkb, 0.75kb, 0.5kb or 0.25 kb or any number of kilobases in between a range defined by any two afore mentioned values.
  • the first sequence comprises the nucleic acid sequence set forth in SEQ ID NO: 1, and/or SEQ ID NO: 2.
  • the promoter is a BTK promoter.
  • the BTK promoter comprises the nucleic acid sequence set forth in SEQ ID NO: 5.
  • the third sequence is codon optimized for expression in humans.
  • the third sequence comprises the sequence set forth in SEQ ID NO: 6 or SEQ ID NO: 7.
  • the promoter is a B cell specific promoter.
  • the B cell specific promoter comprises the B cell specific promoter B29.
  • the B29 promoter sequence comprises a sequence set forth in SEQ ID NO: 46.
  • the B cell specific promoter is an endogenous promoter.
  • the polypeptide further comprises one or more enhancer elements.
  • the one or more enhancer elements comprises at least one DNase Hypersensitive Site (DHS).
  • the DNase Hypersensitive Site is DNase Hypersensitive Site 1 (DHS1), DNase Hypersensitive Site 2, (DHS2) DNase Hypersensitive Site 3 (DHS3), DNase Hypersensitive Site 4 (DHS4) and/or DNase Hypersensitive Site 5 (DHS5).
  • the DNase Hypersensitive Site comprises a sequence set forth in SEQ ID NO: 3.
  • the one or more enhancer elements comprise at least one intronic region.
  • the at least one intronic region is from a human BTK locus that is in association with a human BTK proximal promoter.
  • the at least one intronic region is intron 4, intron 5 and/or intron 13 of the human BTK locus that is in association with the human BTK proximal promoter.
  • the intronic region comprises the sequence set forth in SEQ ID NO: 9 (intron 4), SEQ ID NO: 10 (intron 5) and/or SEQ ID NO: 11 (intron 13).
  • the one or more enhancer elements comprise the sequence set forth in SEQ ID NO: 4, SEQ ID NO: 14 or SEQ ID NO: 15.
  • the UCOE is in a reverse orientation or forward orientation. In some alternatives, the UCOE is in a forward orientation.
  • the one or more enhancer elements comprises the sequences set forth in SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19 and/or SEQ ID NO: 20.
  • the polynucleotide further comprises a gene upstream of a BTK promoter.
  • the gene upstream of a BTK promoter is a BTK enhancer.
  • the BTK enhancer comprises the sequence set forth in SEQ ID NO: 21 or SEQ ID NO: 22.
  • 0.7UCOE comprises a sequence set forth in SEQ ID NO: 2.
  • the enhancer comprises a sequence set forth in SEQ ID NO: 3 or 4.
  • a vector for sustaining BTK expression in cells comprising: a first sequence encoding an ubiquitous chromatin opening element (UCOE), a second sequence encoding a promoter, and a third sequence encoding BTK.
  • the first sequence comprises the nucleic acid sequence set forth in SEQ ID NO: 1, and/or SEQ ID NO: 2.
  • the promoter is a BTK promoter.
  • the promoter comprises the nucleic acid sequence set forth in SEQ ID NO: 5.
  • the third sequence is codon optimized for expression in humans.
  • the third sequence comprises the sequence set forth in SEQ ID NO: 6 or SEQ ID NO: 7.
  • the vector further comprises a B cell specific promoter.
  • the B cell specific promoter comprises the B cell specific promoter B29.
  • the B29 promoter sequence comprises a sequence set forth in SEQ ID NO: 46.
  • the B cell specific promoter is an endogenous promoter.
  • the vector further comprises one or more enhancer elements.
  • the one or more enhancer elements comprise at least one intronic region.
  • the one or more enhancer elements comprises a DNase Hypersensitive Site (DHS).
  • DHS DNase Hypersensitive Site
  • the DNase Hypersensitive Site is DNase Hypersensitive Site 1 (DHS1), DNase Hypersensitive Site 2, (DHS2) DNase Hypersensitive Site 3 (DHS3), DNase Hypersensitive Site 4 (DHS4) and/or DNase Hypersensitive Site 5 (DHS5).
  • the DNase Hypersensitive Site comprises a sequence set forth in SEQ ID NO: 3.
  • the at least one intronic region is from a human BTK locus that is in association with a human BTK proximal promoter.
  • the at least one intronic region is intron 4 intron 5 and/or intron 13 of the human BTK locus that is in association with the human BTK proximal promoter.
  • the intronic region comprises the sequence set forth in SEQ ID NO: 9 (intron 4), SEQ ID NO: 10 (intron 5) and/or SEQ ID NO: 11 (intron 13).
  • the one or more enhancer elements comprise the sequence set forth in SEQ ID NO: 4, SEQ ID NO: 14 or SEQ ID NO: 15.
  • the UCOE is in a reverse orientation or forward orientation. In some alternatives, the UCOE is in a forward orientation.
  • the vector is a lentiviral-based vector of a B lineage specific lentiviral vector.
  • the cells are B cells.
  • the cells are myeloid cells.
  • the cells are hematopoietic stem cells.
  • the cells are CD34+ hematopoietic stem cells.
  • the one or more enhancer elements comprises the sequences set forth in SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19 and/or SEQ ID NO: 20.
  • the polynucleotide further comprises a gene upstream of a BTK promoter.
  • the gene upstream of a BTK promoter is a BTK enhancer.
  • the BTK enhancer comprises the sequence set forth in SEQ ID NO: 21 or SEQ ID NO: 22.
  • 0.7UCOE comprises a sequence set forth in SEQ ID NO: 2.
  • the enhancer comprises a sequence set forth in SEQ ID NO: 3 or 4.
  • a cell for expression of BTK comprising a polynucleotide, which comprises a first sequence encoding a UCOE, a second sequence encoding a promoter and a third sequence encoding BTK.
  • the polynucleotide is in a vector.
  • the vector is a lentiviral vector.
  • the cell is a B cell.
  • the cells are a myeloid cell.
  • the cell is a hematopoietic stem cell.
  • the cell is a CD34+ hematopoietic stem cell.
  • 0.7UCOE comprises a sequence set forth in SEQ ID NO: 2.
  • the enhancer comprises a sequence set forth in SEQ ID NO: 3 or 4.
  • methods of promoting or increasing B cell survival, proliferation and/or differentiation in a subject in need thereof comprises administering the cells of any one of the alternatives herein to the subject or cells comprising the polynucleotide of any one of the alternatives herein or the vector of any one of the alternatives herein to the subject in need and, optionally identifying or selecting the subject as one that would benefit from receiving a therapy that would promote B cell survival, proliferation and/or differentiation in advance of administering the cells and/or, optionally, measuring B cell survival, proliferation and/or differentiation in said subject or in a biological sample obtained from said subject after receiving the administration of the cells.
  • the cells are from the subject and, wherein the cells are genetically modified by introducing the polynucleotide of any one of the alternatives herein or the vector of any one of the alternatives herein into the cells.
  • the administering is performed by adoptive cell transfer.
  • the cells are B cells.
  • the cells are myeloid cells.
  • the cells are hematopoietic stem cells.
  • the cells are CD34+ hematopoietic stem cells.
  • the subject is male.
  • the subject is suffering from XLA.
  • the subject is selected to receive immunoglobulin replacement therapy.
  • the subject is selected to receive targeted anti-microbial agents.
  • 0.7UCOE comprises a sequence set forth in SEQ ID NO: 2.
  • the enhancer comprises a sequence set forth in SEQ ID NO: 3 or 4.
  • X linked agammaglobulinemia X linked agammaglobulinemia
  • the methods comprise administering the cell of any one of the alternatives herein to the subject or a cell comprising the polynucleotide of any one of the alternatives herein or the vector of any one of the alternatives herein to the subject in need and, optionally identifying or selecting the subject as one that would benefit from receiving a therapy for XLA or disease symptoms associated with XLA and/or, optionally, measuring an improvement in the progression of XLA or an improvement in a disease symptom associated with XLA in said subject.
  • XLA X linked agammaglobulinemia
  • the cell is from the subject, wherein the cell is genetically modified by introducing the polynucleotide of any one of the alternatives herein or the vector of any one of the alternatives herein into the cell.
  • the administering is performed by adoptive cell transfer.
  • the cell is a B cell.
  • the cells are myeloid cells.
  • the cells are hematopoietic stem cells.
  • the cells are CD34+ hematopoietic stem cells.
  • the subject is male.
  • the subject is selected to receive immunoglobulin replacement therapy.
  • the subject is selected to receive targeted anti-microbial agents.
  • 0.7UCOE comprises a sequence set forth in SEQ ID NO: 2.
  • the enhancer comprises a sequence set forth in SEQ ID NO: 3 or 4.
  • Figure 1 shows the optimized gene delivery platform for XLA.
  • Figure 2 shows methods for optimizing lentiviral vectors for gene therapy for XLA.
  • Figure 3 shows the methods for the preclinical models for the therapy plan.
  • FIG. 4 shows BTK expression in lymphocyte subsets: BTK expression in bone marrow (BM), spleen (SP) and peritoneal fluid (PF) B cells, monocytes and neutrophils from XLA mice transplanted with lentiviral vector 0.7UCOE or with 0.7UCOE- 14,5 expressing human BTK, as measured by flow cytometry and expressed as percent of the control BTK + population.
  • BM bone marrow
  • SP spleen
  • PF peritoneal fluid
  • FIG. 5 shows rescue of B cell development and function. Analysis of BTK expression in B subsets from primary recipients. Splenic B cell subsets (right panel) are represented in order from least to more mature subsets: early B cell development (pre B cell, pro B cell, immature and mature B cell) to late B cell development (transitional 1 (Tl), transitional 2 (T2), marginal zone precursor (MZP), marginal zone (MZ) and follicular mature (FM).
  • pre B cell early B cell development
  • pro B cell pro B cell
  • immature and mature B cell late B cell development
  • Tl transitional 1
  • T2 transitional 2
  • MZP marginal zone precursor
  • MZ marginal zone
  • FM follicular mature
  • Figure 6 shows both vectors 0.7 UCOE.57Xp. BTK and 0.7UCOE 1-4,5 BTKp. BTK rescue B cell development and function.
  • A Absolute counts for splenic B cells (Right panel).
  • B B cell proliferation in response to IgM stimulation. Naive B cells were stimulated with soluble IgM. The percentage of BTK+ B cells proliferating at 72 hours is shown as normalized to WT Mock.
  • FIG. 7 shows responses to immunization with T-dependent antigens and autoantibody production.
  • A Cumulative data for the levels of NP-specific IgG in serum from treated mice immunized with NP-CGG in Alum was detected by ELISA and measured relative to an IgG standard. ELISAs were used to detect levels of high-affinity NP-IgG before (-) and after primary (10) immunization, as well as following re-challenge or (20) secondary immunization.
  • B Levels of anti-dsDNA IgG and IgG2c in serum from treated mice were measured by ELISA at the end point of the experiment depicted as absorbance readings (OD450). Serum from autoimmune WAS-/- chimeric mice are used as positive control (black triangles).
  • Figure 8 shows viral integration number per cell.
  • A Average viral integration number per cell in total BM and in splenic CD43- (B cell) vs. CD43+ (non B) cells in primary transplant mice as measured by qPCR.
  • FIG. 9 shows expression profiles of four LV constructs and rescue of B cell development and function in primary recipient mice.
  • (9A) Schematic of the lentiviral constructs with RRL backbone used to express human BTK in murine cells with the BTK promoter (BTKp), a 1.5kb ubiquitous chromatin opening element (UCOE) or ⁇ enhancer, and codon-optimized human BTK cDNA (co).
  • n 14 (WT Mock), 12 (KO Mock), 5 (BTKp), 24 (1 5kb.UCOE.BTKp), 18 (1.5kb.UCOE.BTKp.co), 21 ( ⁇ . ⁇ ).
  • B cells (B220+) in the bone marrow, spleen, and peritoneum were stained for surface markers indicative of B cell subsets and analyzed for counts or percentage of live lymphocytes: (9C) Early B cell development: Pro-B (CD43+, IgM-), Pre-B (CD43-, IgM-, IgD-), Immature (CD43-, IgM+ IgD-), and Mature (CD43-, IgM+, IgD+); (9D) late B cell development: transitional Tl (CD24hi, CD21-), transitional T2 (CD24hi, CD21int), marginal zone/precursor MZ/MZP (CD24hi, CD21hi), and follicular FM (CD24int, CD21int); (9E) peritoneum B cells: B2 (B220hi), Bib (B2201o, CD5-), and B la (B2201o, CD5+).
  • BM,SP,PE 11,13, 14 (WT Mock); 10, 11,10 (KO Mock); 5,5,5 (BTKp); 20,21,22 (UCOE.BTKP); 14, 14,17 (UCOE.BTKp.co); 18,19,20 ( ⁇ . ⁇ )
  • Total splenocytes from treated mice were stimulated with media alone, anti- murine IgM, LPS or PMA/Ionomycin and proliferation was measured by incorporation of 3H-thymidine, depicted as the counts per minute (cpm) averaged from 3 replicate wells.
  • n 10 (WT Mock), 7 (KO Mock), 5 (BTKp), 13 (1.5kbUCOE.BTKp), 9 (1.5 kbUCOE.BTKp.co), 10 ( ⁇ . ⁇ ).
  • 9G-9H Total serum IgG (9G) and IgM (9H) in serum from treated mice was measured by ELISA.
  • FIG 10 shows ⁇ . ⁇ gene therapy treated mice develop broad specificity IgG autoreactive antibodies.
  • FIG 11 shows 1.5kbUCOE.BTKp and 1.5kbUCOE.BTKp.co lead to sustained BTK expression with lower copy numbers in primary and secondary recipients.
  • Splenic granulocytes (CDl lb+, GRlhi) from secondary transplant mice were evaluated for %BTK+ cells by flow cytometry,
  • (a) Graphical analysis of %BTK+ granulocytes in secondary transplant recipients. Data represent mean ⁇ SEM from 3 independent experiments with n 3 (WT Mock), 2 (KO Mock), 3 (BTKp), 8 (1.5.UCOE.BTKp), 7 ( ⁇ . ⁇ ).
  • Figure 12 shows restoration of BTK expression in affected hematopoietic lineages and B cell development in gene therapy mice treated with 0.7kb UCOE.BTKp.co LV.
  • the proportion of BTK+ cells and numerical reconstitution of B cell subsets was assessed at 16-25 weeks post-transplant,
  • UCOE ubiquitous chromatin opening element
  • b-c Percent of BTK+ cells in bone marrow (b) and splenic (c) lymphocyte subsets (Neutrophils, Monocytes, B cells and T cells) from gene therapy treated groups, determined by flow cytometry.
  • Figure 13 shows reconstitution of B cell function following 0.7.UCOE.Bkp.co LV gene therapy in primary recipients.
  • Assays of B cell function after 16- 25 weeks post bone marrow transplantation. Bars show the mean ⁇ SD (a) The percentage of B cells (CD43- splenocyte) that underwent > 1 cell division 72 hours after incubation with anti-mouse IgM antibodies, LPS or P/I (as measured by CFSE dilution and read out by flow cytometry). Representative data from one independent experiment; n 4 (WT Mock), 3 (KO Mock), and 5 (0.7.UCOE.BTKp.BTK.co).
  • Serum from 2 autoimmune WAS chimera with high serum anti-DNA antibodies was run as a positive control (e).
  • VCN Viral copy number
  • n for primary recipients 2( BTKp.BTK), 5 (1.5kb UCOE.BTKp.BTK), 6 ( Eu.BTKp.BTK ), 6 (0.7kb.UCOE.BTKp.co).
  • n for secondary recipients 4 (1.5kb.UCOE.BTKp.BTK) and 6 (0.7 UCOE.BTKp.co).
  • Figure 15 shows 0.7UCOE.BTKp.co leads to sustained BTK expression and lower copy numbers in XLA CD34 cells.
  • XLA and control CD34 cells were transduced with 0.7UCOE.BTKp.co at various multiplicities of infection (MOI) and cultured in vitro for 15 days
  • MOI multiplicities of infection
  • FIG. 15 shows 0.7UCOE.BTKp.co leads to sustained BTK expression and lower copy numbers in XLA CD34 cells.
  • XLA and control CD34 cells were transduced with 0.7UCOE.BTKp.co at various multiplicities of infection (MOI) and cultured in vitro for 15 days
  • MOI multiplicities of infection
  • FIG 16 shows the BTK promoter mimics BTK's endogenous expression pattern in mice.
  • (a-b) Bone marrow, spleen, peritoneal wash, peripheral blood and thymus were collected from a wildtype mouse and cells were stained for B, myeloid, T and NK cell markers, intracellular BTK and read on a flow cytometer. Samples from the various tissues are designated by the shape of the symbol, as outlined in the legend. The percent of cells that are BTK+ is shown for each subset (a), as well as the median fluorescence intensity of BTK+ cells (b).
  • Figure 17 shows (a) Schematic of lentiviral constructs under control of the BTK promoter with ⁇ enhancer and human BTK cDNA, either wild type of codon- optimized, with a T2A-linked GFP.
  • (b) Chicken BTK-/- DT40 cells were transduced with BTK-GFP or coBTK-GFP constructs; histograms show GFP and BTK expression,
  • Figure 18 shows (a) Representative plots of BTK expression by flow cytometry in peripheral blood B cells and myeloid cells from gene therapy-treated KO mice, (b) BTK marking in bone marrow, spleen, peritoneal B and myeloid cells from gene therapy treated mice was determined by flow cytometry and the median fluorescence intensity of the BTK+ cells is shown.
  • Figure 19 shows (a) Survival curve of ⁇ . ⁇ primary transplant mice with high and low anti-dsDNA IgG titers, compared to controls, (b) Correlation of BTK MFI of BTK+ cells and Anti-dsDNA IgG titers as measured by flow cytometry and ELISA, respectively, in ⁇ . ⁇ primary transplant mice.
  • Figure 20 shows sera from WT Mock (4), 1.5kb. UCOE.BTKp (4) and ⁇ . ⁇ (8) mice were analyzed by autoantigen array for levels of IgM and IgG reactive to 88 murine antigens. Data from each row was subject to z-transformation and Z-scores are displayed on a colorimetric scale from lowest reactivity (red) to highest (blue).
  • Figure 21 shows the development of B cells and the pathway in which the enzyme BTK is involved.
  • Figure 22 shows development of the optimal BTK LV gene therapy vector.
  • Figure 23 shows LV vectors containing the BTK endogenous promoters. As shown, the 2 kb CBX3-HNRPA2B1 element- UCOE 1.5- eliminates the A2 promoter but retains the CBX3 in reverse orientation.
  • Figure 24 shows the development of LV vectors containing the BTK endogenous promoter. As shown, ⁇ associated with higher level Btk MFI in B cells.
  • Figure 25 shows the results from a FACs assay of LV vectors containing the Btk endogenous promoter.
  • Figure 26 shows the expression of BTK in myeloid cells that were obtained from splenocytes of treated mice.
  • Figure 27 shows the results of BTK expression in B cells in which the LV expressing the BTK comprises a B cell specific promoter ⁇ . ⁇ 29.
  • Figure 28 shows BTK expression in B cells and monocytes from the lentiviral vector comprising the promoter region BTKpro.BTK
  • Figure 29 shows BTK expression in B cells and monocytes from the lentiviral vector comprising the promoter region BTKpro.BTK and the ubiquitous chromatin opening element (UCOE).
  • BTKpro.BTK ubiquitous chromatin opening element
  • Figure 30 shows BTK expression in B cells and monocytes from the lentiviral vector comprising the promoter region ⁇ . ⁇ . ⁇ (BTK promoter) and huBTK.
  • Figure 31 shows the expression profile of BTK of several BTK promoters in B cells and myeloid cells of the bone marrow, spleen and peritoneum.
  • Figure 32 shows the expression profile of BTK of several BTK promoters in B cells and myeloid cells of the bone marrow, spleen and peritoneum.
  • the vectors used were: WT mock, DKO mock, BTKp, UCOE BTKp, UCOE.57Xp.co and ⁇ .
  • Figure 33 shows B Cell Development Restoration of Mature B Cell Subsets. The black colored regions show that more mature peripheral B cell populations.
  • Figure 34 shows B Cell Development Numerical Reconstitution of B Cell Populations (bone marrow, spleen and Peritoneum). As shown, the data summarize findings from up to 40 recipient mice per vector and similar numbers of controls.
  • Figure 35 shows B cell proliferation in cells expressing four different types of lentiviral vectors (mock wt, DKO mock, lentiviral vector with UCOE.57Xp and lentiviral vector ⁇ , ⁇ ).
  • the vital dye dilution vs Btk stain showed that UCOE led to very similar results to the WT mice in the experiments.
  • Figure 36 shows B cell proliferation in cells with the following vectors: WT, WT mock, DKO, DKO mock, UCOE.57Xp and ⁇ .57 ⁇ .
  • Figure 37 shows the results of IgM and IgG production in cells with the following vectors: WT, WT mock, DKO, DKO mock, BTKp, UCOE.BTKp and ⁇ , ⁇ .
  • the bottom panel shows T independent immune responses in cells with the following vectors: WT, WT mock, DKO, DKO mock, UCOE.57Xp, WT-0 and KO-0. It is noted that the ⁇ IgG levels actually slightly above WT mock recipients.
  • Figure 38 shows T-dependent immune responses in cells with the following vectors: WT, WT mock, DKO, DKO mock, BTKp, UCOE.BTKp, UCOE.57Xp.co, WT-0, ⁇ , ⁇ and KO-0.
  • FIG 39 shows T-dependent immunization: affinity maturation. Affinity maturation for individual UCOE animals and controls led to very similar results.
  • Figure 40 shows evidence for auto-antibody production.
  • Figure 41 shows evidence for auto-antibody production using a heat map of cells that have the vectors WT mock, UCOE.57Xp and ⁇ , ⁇ . This was performed for testing the production of IgM and IgG.
  • Figure 42 shows association of antibody levels and percent mouse survival with BTK expression.
  • Figure 43 shows long-term stem cell marking in neutrophils with BTK expression in secondary recipient mice.
  • Figure 44 shows evidence for long-term stem cell marking in VCN in primary and secondary recipients that have the vectors: mock, BTKp, XJCOE.BTKp, UCOE.BTKp.co, and ⁇ . ⁇ .
  • Figure 45 shows the impact of LV therapy on long-term survival.
  • Figure 46 shows the summary of testing of alternative enhancer elements within BTK promoter LV.
  • Figure 47 shows that 0.7UCOE.BTKp and 0.7UCOE.IE.BTKp rescue BTK expression and splenic B cell counts.
  • Figure 48 shows that 0.7UCOE.BTKp and 0.7UCOE.IE.BTKp rescue B cell function.
  • Figure 49 shows that 0.7UCOE.IE exhibits an improved safety profile over 0.7UCOE.
  • Figure 50 shows new BTK constructs with DNase Hypersensitive Sites.
  • Figure 51 shows the identification of DNase Hypersensitive sites.
  • Figure 52 shows a schematic of the constructs that were evaluated for BTK expression in cells.
  • Figure 53 shows in vivo comparison of DHS constructs to 0.7UCOE and 0.7UCOE.IE.
  • Figure 54 shows peripheral blood lymphocyte distribution in B cells, T cells, monocytes and neutrophils 15 weeks post-transplant. BTK reconstitution was also shown in lymphocyte subsets in cells with the following vectors: KO unirradiated, KO Mock, 0.7 UCOE, 0.7 UCOE.IE, 0.7 UCOEDHS4, 0.7 UCOE.DHS12, 0.7 UCOE.DHS124, 0.7 UCOEl-5, WT mock and WT unirradiated.
  • Figure 55 shows BTK experimental summary in cells with the vectors: KO Mock, 0.7 UCOE, 0.7 UCOE.IE, 0.7 UCOEDHS4, 0.7 UCOE.DHS12, 0.7 UCOE.DHS124, 0.7 UCOEl-5 and WT mock.
  • Figure 56 shows BTK expression in cells with vectors with either the UCOE element or DHS4 element.
  • Figure 57 shows BTK expression experiments with cells that have the following vectors: O.IVCOE. BTK ⁇ .coBTK, KO Mock, 0.7 UCOE, DHS4, DHSl-5 and WT Mock.
  • Figure 58 shows peripheral blood lymphocyte distribution— 12 weeks post transplantation with cells with the following vectors: KO Mock, WT Mock, 0.7 UCOE, DHS4 and DHSl-5.
  • Figure 59 shows 6 week and 12 week peripheral blood lymphocyte distribution in B cells, monocytes and neutrophils that have the following vectors: KO unirradiated, KO Mock, 0.7 UCOE, DHS4, DHSl-5, WT Mock, and WT unirradiated.
  • Figure 60 shows 6 week and 12 week BTK expression in B cells, monocytes and neutrophils that have the following vectors: KO unirradiated, KO Mock, 0.7 UCOE, DHS4, DHS1-5, WT Mock, and WT unirradiated.
  • Figure 61 shows that BTK is expressed across cell subsets after a 12 week bleed following cell transplantation.
  • Figure 62 shows the experimental set up for the in vitro comparison of original coBTK (codon optimized BTK) vs new coBTK.
  • Figure 63 shows in vitro Comparison— TBK Lineage Negative Cells, Volume Matched Virus (day 7 BTK stain).
  • Figure 64 shows the percent BTK expression in cells that received different concentrations of the lentivirus vectors for BTK expression.
  • the vectors used were the following: KO Mock, 0.7 UCOE, DHS4, 0.7 UCOE.newcoBTK, 0.7 UCOE.newcoBTK, and WT spleen.
  • Figure 65 shows in vivo comparison of original vs new coBTK.
  • Figure 66 shows additional techniques for optimizing the UCOE elements to increase or enhance BTK expression.
  • Figure 67 shows additional techniques for optimizing the UCOE elements to increase or enhance BTK expression for the 1.5kb UCOE.
  • Figure 68 shows 0.7UCOE.BTKp vs. 0.7UCOEfwd.BTKp: in vitro test with matched volume virus.
  • Figure 69 shows an outline for finalizing the clinical BTK LV construct.
  • Figure 70 shows an outline for finalizing the clinical BTK LV construct and planned final in vivo testing for a codon optimized BTK.
  • Figure 71 is a table showing approved apheresis of XLA patient's stem cells.
  • Figure 72 shows a human chimera in NSG.
  • Figure 73 shows human lymphocyte reconstitution in B cells of the bone marrow and the spleen.
  • Figure 74 shows phenotype of engrafted XLA stem cell (spleen) such as the expression of IgD and IgM.
  • Figure 75 shows a phenotype of engrafted XLA stem cell (spleen) such as the expression of CD24 and CD38.
  • Figure 76 shows B cell developmental block (BM) indicating that there is an equivalent number of % ProB cells between the XLA group and the healthy group.
  • Figure 77 shows B cell developmental block (BM) indicating that patients with XLA have a significantly higher % pre-B cells compared to a healthy control.
  • Figure 78 shows B cell developmental block (BM) indicating that patients with XLA have cells that are blocked at the Pre B cell stage. These Pre B cells are able to migrate into the spleen.
  • CD179a/CD179b surrogate light chain is disulfide-linked to membrane-bound Ig mu heavy chain in association with a signal transducer CD79a/CD79b heterodimer to form a B cell receptor-like structure, so-called preB cell receptor (preBCR).
  • Figure 79 shows a B cell developmental block (BM). Development of XLA B cells are blocked at Pre B cell stage. Pre B cells are able to migrate to the spleen.
  • Figure 80 shows that that B cells function by Ca 2+ flux. However, the XLA B cells are unable to flux with IgM as compared to the control cells.
  • Figure 81 shows the block of the pathway in B cell development, in which XLA causes the B cell development to be blocked at the Pre B cell stage.
  • Figure 82 shows an outline of the results and conclusions from the experiments.
  • Figure 83 shows a diagram outlining the human lentiviral transduction of stem cells.
  • Figure 84 shows a diagram outlining preclinical modeling- human HSC.
  • Figure 85 shows results from the Human lentiviral transduction of Stem cells. As shown there is 70% viability with 0.7UCOE.BTKp.BTK compare to DHS4.
  • Figure 86 shows BTK expression at D15 with XLA P2. As shown, in a non-selective environment 0.7 UCOE.BTKpBTK leads to higher expression of BTK compare to DHS4.
  • Figure 87 shows an outline of the conclusion from the experiments and follow up experiments.
  • Figure 88 shows preclinical modeling for Human HSC. For these experiments, CD34+ cells are transduced with either mock vector or the vector: UCOE.57Xjp.co.
  • Figure 89 shows preclinical modeling- Human HSC.
  • Cells were transduced with the vector UCOE.5rXpro.co-op.SrX (UCOE.5rXp.co) GFP.
  • Figure 90 shows a diagram of the experimental methods for transduction of the NSG recipient mice.
  • Figure 91 shows the results from the transformed recipient mice: % GFP expression, viral copy expression in the cells of the bone marrow and the spleen, as well as, the expression in B cells and the myeloid cells.
  • Figure 92 shows the analysis to identify conserved non-coding sequences by comparing mouse and human BTK gene sequences and all the vectors that are tested for expression of BTK. The regions identified and located were within the BTK promoter, upstream from the BTK promoter and proximal to the neighboring gene (BTK enhancer or BTKe), within part of intron 1, introns 4, 5, and 13.
  • BTK enhancer or BTKe the neighboring gene
  • Figure 93 shows results from truncation of 1.5kb UCOE to 0.7kb and identification of potential DNA enhancer elements may improve titer and BTK expression in Lentiviral constructs, (a) Truncation of 1.5kb Ubiquitous Chromatin Opening Element (UCOE) to 0.7kb.
  • the UCOE element spans a large, CpG-rich region across the divergently transcribed promoter regions for the housekeeping genes CBX3 and HNRPA2B1, and has traditionally been truncated to a 1.5-2.2kb region by various groups for use in protein expression constructs.
  • the 1.5kb UCOE used here starts at Exon 1 of CBX3 and spans past CBX3 Alternate Exon 1.
  • DHS DNasel Hypersensitive sites
  • FIGS. 1-10 Shown here are diagrams of the lentiviral constructs with RRL backbone expressing either version of codon optimized human BTK (coBTK or co2BTK) with a 0.7 kb ubiquitous chromatin opening element (0.7UCOE), with or without addition of DHS4 downstream of the 0.7UCOE element.
  • 0.7UCOE ubiquitous chromatin opening element
  • Figure 94 shows restoration of BTK expression in affected hematopoietic lineages and B cell development in gene therapy mice treated with 0.7UCOE vectors. The proportion of BTK+ cells and numerical reconstitution of B and myeloid cell subsets was assessed at 19-23 weeks post-transplant.
  • Figure 94 A Representative flow plots showing intracellular BTK staining in splenic B cells at endpoint analysis,
  • b-d Percent of BTK+ cells in bone marrow ( Figure 94B), spleen ( Figure 94C), and peritoneal (Figure 94D) lymphocyte subsets from gene therapy treated groups, determined by flow cytometry.
  • Cell subsets are defined as Neutrophils (CDl lb+ GR1+), Monocytes (CDl lb+), and B cells (B220+).
  • Figure 94E-F Stacked bars show the average counts of B cell subsets in bone marrow ( Figure 94E), spleen ( Figure 94F), and peritoneum ( Figure 94G).
  • Late B cell development (spleen): transitional Tl (CD24hi, CD21-), transitional T2 (CD24hi, CD21int), marginal zone/precursor MZ/MZP (CD24hi, CD21hi), and follicular FM (CD24int, CD21int).
  • Peritoneal B cell subsets Bl (IgM+ CD43+), and B2 (CD43-).
  • Figure 94H Rescue of BTK expression in B cell subsets, measured by flow cytometry. ( Figure 941) BTK+ MFI across B cell development, normalized to WT Mock in each individual experiment.
  • FIG. 95D-F At endpoint analysis, B cells were isolated from splenocytes by CD43- magnetic separation, labeled with Cell Trace Violet and stimulated in vitro with IgM, LPS, or a media control.
  • Figure 95D The percentage of BTK+ B cells that underwent > 1 cell division 72 hours after incubation with anti-mouse IgM antibodies, LPS or media only (read out by flow cytometry).
  • Figure 95E BTK+ MFI of cells after each division (D0-D4), normalized to WT Mock.
  • Figure 95F Representative flow plots showing BTK staining and Cell Trace dilution in B cells 72 hours post-IgM stimulation, gated on live and B220+ BTK+ cells.
  • Figure 96 shows vector safety considerations.
  • Figure 96A-B Levels of anti-dsDNA IgG (Figure 96A) and anti-dsDNA IgG2c (Figure 96B) in serum from treated mice were measured by ELISA and are depicted as absorbance readings (OD450). Serum from the known autoimmune-prone WAS chimeric mouse model and mice treated with Eu.BTKp were included as positive controls.
  • Figure 96C Genomic DNA was isolated from total bone marrow and spleen at endpoint analysis and the number of viral integrations per cell (VCN) was quantified by qPCR.
  • Figure 97 shows a schematic of a transfection protocol.
  • Figure 98 shows equivalent bone marrow engraftment of human hematopoietic cells between treatment groups. Human stem cells from XLA patient, either with or without gene therapy treatments, engrafted into the bone marrow equivalently as those from a healthy donor. 98A. Representative flow plots showing various markers of human immune cells including human and mouse CD45 (a marker for hematopoietic cells) CD33 (myeloid cells) and CD19 (B cells), CD4 and CD 8 (T cells).
  • Human hematopoietic cells are hCD45+ and mCD45- from total live BM cells; CD33 and CD19 markers were analyzed from hCD45+gate; CD4 and CD 8 were analyzed from the CD33-CD19- gate.
  • 98B and 98C. % human CD45 and total number of hCD45 cells engrafted in BM; n 4 for each cohort (XL A3, XLA3+LV 0.7UCOE.BTKp.BTKco2, XL A3 + LV 0.7UCOE.DHS4.BTKp.BTKco2, and healthy donor).
  • Figure 99 shows Pre-clinical modeling XLA3 : Spleen analysis 12 weeks post-transplant.
  • Figure 100 shows Pre-clinical modeling XLA3 : Spleen analysis 12 weeks post-transplant and results of mouse recipients of XLA3 HSC treated with gene therapy using 0.7UCOE.BTKp BTKco2 or 0.7UCOE.DHS4.BTKp.BTKco2.
  • Figure 101 shows representative flow plot for B cell development subsets in spleen.
  • Figure 102 shows that recipients of LV transduced XLA3 HSC (using 0.7UCOE.BTKp.BTKco2 or 0.7UCOE.DHS4 BTKp.BTKco2 ) have an increased proportion of splenic immature B cells (including CD19+CD24+CD38+ B cells and CD19+CD24+CD38+IgM+ cells) compared to XLA controls.
  • Graphs summarize the results from flow cytometry shown in Figure 4, looking at specific subpopulations of immature B cells.
  • A % of Immature B cells (CD24+ CD38+ ) in the spleen
  • B % of immature B cells that are IgM+
  • C- D Total number of immature B cells (CD24+ CD38+) and CD24+hCD38+ IgM+ cells in the spleen
  • E overlay of CD 10 histogram , showing the mean fluorescence intensity MFI shift in CD 10 compared to healthy donor.
  • Figure 103 shows that recipients of LV transduced XLA HSC (0.7UCOE.BTKp.BTKco2 or 0.7UCOE.DHS4 BTKp.BTKco2 ) have increased proportions of mature B cells (CD19 + CD24 low CD38 low IgM + and IgD + ) compared to XLA controls.
  • Figure 104 shows a representative flow plot for B cell development subsets in the bone marrow.
  • Figure 105 shows that recipients of LV transduced XLA HSC (using 0.7UCOE BTKp BTKco2 or DHS4 BTKpBTK.co2 ) exhibit an increased proportion of CD 19 + CD24 + CD38 + IgM + immature B cells compared to XLA controls.
  • Figure 106 shows that recipients of LV transduced XLA HSC (using LV 0.7UCOE BTKp BTKco2 or LV 0.7UCOE.DHS4 BTKp.BTK.co2) exhibit gene marking of 0.2-2 viral copy number (VCN)/cell in vivo.
  • VCN viral copy number
  • Figure 107 shows that recipients of XLA3 HSC that received gene therapy with LV 0.7UCOE.BTKp.BTKco2 or 0.7UCOE.DHS4.BTK.pBTKco2 produce B cells that secrete IgM in vivo.
  • Figure 108 shows that recipients of XL A3 HSC transduced with LV 0.7UCOE.BTKp.BTKco2 or 0.7UCOE.DHS4.BTKp.BTKco2 exhibit a restored capacity for B cells to flux calcium in response to B cell receptor (BCR) engagement.
  • Figure 109 shows the B cell differentiation protocol.
  • Figure 110 shows a representative flow plot for in vitro B cell class switching.
  • Figure 111 shows that recipients of LV transduced XLA HSC (using 0.7UCOE BTKp BTKco2 or DHS4 BTKpBTK.co2) generate B cells that are capable of responding to cytokine and T-cell dependent signals leading to antibody secretion.
  • Polynucleotide refers to “nucleic acid” or “nucleic acid molecule,” such as deoxyribonucleic acid (DNA) or ribonucleic acid (RNA), oligonucleotides, fragments generated by the polymerase chain reaction (PCR), and fragments generated by any of ligation, scission, endonuclease action, and exonuclease action.
  • DNA deoxyribonucleic acid
  • RNA ribonucleic acid
  • PCR polymerase chain reaction
  • Nucleic acid molecules can be composed of monomers that are naturally-occurring nucleotides (such as DNA and RNA), or analogs of naturally-occurring nucleotides (e.g, enantiomeric forms of naturally-occurring nucleotides), or a combination of both.
  • Modified nucleotides can have alterations in sugar moieties and/or in pyrimidine or purine base moieties.
  • Sugar modifications include, for example, replacement of one or more hydroxyl groups with halogens, alkyl groups, amines, and azido groups, or sugars can be functionalized as ethers or esters.
  • the entire sugar moiety can be replaced with sterically and electronically similar structures, such as aza-sugars and carbocyclic sugar analogs.
  • modifications in a base moiety include alkylated purines and pyrimidines, acylated purines or pyrimidines, or other well-known heterocyclic substitutes.
  • Nucleic acid monomers can be linked by phosphodiester bonds or analogs of such linkages. Analogs of phosphodiester linkages include phosphorothioate, phosphorodithioate, phosphoroselenoate, phosphorodiselenoate, phosphoroanilothioate, phosphoranilidate, phosphoramidate, and the like.
  • nucleic acid molecule also includes so-called “peptide nucleic acids,” which comprise naturally-occurring or modified nucleic acid bases attached to a polyamide backbone. Nucleic acids can be either single stranded or double stranded. In some alternatives, a nucleic acid sequence encoding a fusion protein is provided. In some alternatives, the nucleic acid is RNA or DNA.
  • Coding for" or “encoding” have their plain and ordinary meaning when read in light of the specification, and may include but is not limited to, for example, the property of specific sequences of nucleotides in a polynucleotide, such as a gene, a cDNA, or an mRNA, to serve as templates for synthesis of other macromolecules such as a defined sequence of amino acids.
  • a gene codes for a protein if transcription and translation of mRNA corresponding to that gene produces the protein in a cell or other biological system.
  • BTK Bruton's tyrosine kinase
  • BTK has its plain and ordinary meaning when read in light of the specification, and may include but is not limited to, for example, an enzyme that in humans is encoded by the BTK gene.
  • BTK is a kinase that plays a crucial role in B-cell development.
  • BTK plays a crucial role in B cell maturation as well as mast cell activation through the high-affinity IgE receptor.
  • Mutations in the BTK gene are implicated in the primary immunodeficiency disease X-linked agammaglobulinemia (Bruton's agammaglobulinemia); sometimes abbreviated to XLA. Patients with XLA have normal pre-B cell populations in their bone marrow but these cells fail to mature and enter the circulation.
  • X-linked agammaglobulinemia as described herein is a genetic disorder that affects the body's ability to fight infection. As the form of agammaglobulinemia that is X-linked, it is much more common in males. In people with XLA, the white blood cell formation process does not generate mature B cells, which manifests as a complete or near- complete lack of proteins called gamma globulins, including antibodies, in their bloodstream.
  • X-linked agammaglobulinemia (XLA) is characterized by recurrent bacterial infections in affected males in the first two years of life. Recurrent otitis is the most common infection prior to diagnosis.
  • a "promoter” is a region of DNA that initiates transcription of a specific gene.
  • the promoters can be located near the transcription start site of a gene, on the same strand and upstream on the DNA (the 5 'region of the sense strand).
  • the promoter can be a conditional, inducible or a constitutive promoter.
  • the promoter can be specific for bacterial, mammalian or insect cell protein expression.
  • the nucleic acid further comprises a promoter sequence.
  • the promoter is specific for mammalian protein expression.
  • the promoter is a conditional, inducible or a constitutive promoter.
  • Ubiquitous chromatin opening elements are regulatory elements that are derived from promoter-containing CpG islands of ubiquitously expressed housekeeping genes. It was proposed that regulatory elements from such promoters possess a chromatin-remodeling function allowing the maintenance of chromatin in a permissive configuration resulting in high and consistent expression of genes in their proximity. Although originally relatively large (up to 16 kb), new, smaller, synthetic UCOEs can lead to high expression of the transgene. Ubiquitous chromatin elements and their functions is described in Figure 1. In some alternatives, 0.7UCOE comprises a sequence set forth in SEQ ID NO: 2. In some alternatives, the enhancer comprises a sequence set forth in SEQ ID NO: 3 or 4.
  • Codon optimization has its plain and ordinary meaning when read in light of the specification, and may include but is not limited to, for example, the design process of altering codons to codons known to increase maximum protein expression efficiency.
  • codon optimization for expression in human is preferred, wherein codon optimization can be performed by using algorithms that are known to those skilled in the art so as to create synthetic genetic transcripts optimized for high mRNA and protein yield in humans.
  • Programs containing algorithms for codon optimization in humans are readily available. Such programs can include, for example, OptimumGeneTM or GeneGPS® algorithms.
  • human codon optimized sequences can be obtained commercially, for example, from Integrated DNA Technologies.
  • optimization can also be performed to reduce the occurrence of secondary structure in a polynucleotide.
  • optimization of the sequences in the vector can also be performed to reduce the total GC/AT ratio.
  • Strict codon optimization can lead to unwanted secondary structure or an undesirably high GC content that leads to secondary structure.
  • the secondary structures affect transcriptional efficiency.
  • Programs such as GeneOptimizer can be used after codon usage optimization, for secondary structure avoidance and GC content optimization. These additional programs can be used for further optimization and troubleshooting after an initial codon optimization to limit secondary structures that can occur after the first round of optimization.
  • Alternative programs for optimization are readily available.
  • the vector comprises sequences that are optimized for secondary structure avoidance and/or the sequences are optimized to reduce the total GC/AT ratio and/or the sequences are optimized for expression in humans.
  • the gene encoding BTK is codon optimized.
  • the codon optimized BTK comprises a sequence set forth in SEQ ID NO: 6 or SEQ ID NO: 7.
  • Enhancers are short regions of DNA that can be bound by proteins (activators) to increase the likelihood that transcription of a particular gene will occur.
  • the activators can also be referred to as transcription factors.
  • Enhancers can be either cis-acting, or Trans-acting (acting away from the gene) and can be located up to 1 Mbp (1,000,000 bp) away from the gene and can be upstream or downstream from the start site, and either in the forward or backward direction.
  • the size of an enhancer can be of a size 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400 or 1500 bp or any number of base pairs in between a range defined by any two aforementioned values.
  • Dnase I hypersensitive site is a region of chromatin that is sensitive to cleavage by the DNase I enzyme. In these specific regions of the genome, chromatin has lost its condensed structure, exposing the DNA and making it accessible. This raises the availability of DNA to degradation by enzymes, such as DNase I. These accessible chromatin zones are functionally related to transcriptional activity, since this remodeled state is necessary for the binding of proteins such as transcription factors. As described in the alternatives herein is "Dnase I hypersensitive site 4" (DHS4).
  • DHS4 Dnase I hypersensitive site 4
  • DHS4 is an enhancer element that is located at -18 kb from a ⁇ -globin promoter and can include binding sites for both erythroid specific and ubiquitous proteins and plays an important role as a regulatory element.
  • the vector for expression of BTK comprises at least one DNase Hypersensitive Site.
  • the DNase Hypersensitive Site is DNase Hypersensitive Site 1 (DHSl), DNase Hypersensitive Site 2, (DHS2) DNase Hypersensitive Site 3 (DHS3), DNase Hypersensitive Site 4 (DHS4) and/or DNase Hypersensitive Site 5 (DHS5).
  • Intratron as described herein, is any nucleotide sequence within a gene that is removed by RNA splicing during maturation of the final RNA product.
  • the vector comprises at least one intronic region.
  • the at least one intronic region is from a human BTK locus that is in association with a human BTK proximal promoter.
  • Vector is a nucleic acid used to introduce heterologous nucleic acids into a cell that has regulatory elements to provide expression of the heterologous nucleic acids in the cell.
  • Vectors include but are not limited to plasmid, minicircles, yeast, and viral genomes.
  • the vector is a viral vector.
  • the viral vector is a lentiviral vector.
  • B cells as described herein are a type of white blood cell of the lymphocyte subtype. They are also known as B lymphocytes. B cells can function in the humoral immunity component of the adaptive immune system by secreting antibodies. Additionally, B cells present antigen (they are also classified as professional antigen- presenting cells (APCs) and secrete cytokines. In some alternatives of the cells provided herein, the cells are B cells.
  • APCs professional antigen- presenting cells
  • Myeloid cells has its plain and ordinary meaning when read in light of the specification, and may include but is not limited to, for example, a granulocyte or monocyte precursor cell in bone marrow or spinal cord, or a resemblance to those found in the bone marrow or spinal cord.
  • the myeloid cell lineage includes circulating monocytic cells in the peripheral blood and the cell populations that they become following maturation, differentiation, and/or activation. These populations include non-terminally differentiated myeloid cells, myeloid derived suppressor cells, and differentiated macrophages. Differentiated macrophages include non-polarized and polarized macrophages, resting and activated macrophages.
  • the myeloid lineage can also include granulocytic precursors, polymorphonuclear derived suppressor cells, differentiated polymorphonuclear white blood cells, neutrophils, granulocytes, basophils, eosinophils, monocytes, macrophages, microglia, myeloid derived suppressor cells, dendritic cells and erythrocytes.
  • microglia can differentiate from myeloid progenitor cells.
  • the cells are myeloid cells.
  • Hematopoietic stem cells are precursor cells that can give rise to myeloid cells such as, for example, macrophages, monocytes, macrophages, neutrophils, basophils, eosinophils, erythrocytes, megakaryocytes/platelets, dendritic cells and lymphoid lineages (such as, for example, T-cells, B-cells, K-cells).
  • HSCs have a heterogeneous population in which three classes of stem cells exist, which are distinguished by their ratio of lymphoid to myeloid progeny in the blood (L/M).
  • the cells are hematopoietic stem cells.
  • Subjects or patients have their plain and ordinary meaning when read in light of the specification, and may include but is not limited to, for example, any organism upon which the alternatives described herein may be used or administered, e.g, for experimental, diagnostic, prophylactic, and/or therapeutic purposes.
  • Subjects or patients include, for example, animals.
  • the subject is mice, rats, rabbits, non-human primates, and humans.
  • the subject is a cow, sheep, pig, horse, dog, cat, primate or a human.
  • the subject is a human male.
  • adoptive cellular therapy or “adoptive cell transfer,” as described herein refers to the transfer of cells, most commonly immune-derived cells, back into the same patient or into a new recipient host with the goal of transferring the immunologic functionality and characteristics into the new host.
  • adoptive cellular therapy or adoptive cell transfer comprises administering cells for expression of BTK to a subject in need.
  • BTK is expressed in both B cells and myeloid cells where it also contributes to normal functional responses in both lineages. Failure to express BTK leads to XLA. Conversely, overexpression of activated or wild type BTK can lead to cell transformation and/or developmental blockade ("Early arrest in B cell development in transgenic mice that express the E41K Bruton's tyrosine kinase mutant under the control of the CD19 promoter region" J Immunol. 1999 Jun l; 162(l l):6526-33; "Correction of B-cell development in Btk-deficient mice using lentiviral vectors with codon-optimized human BTK.” Leukemia.
  • the unique constructs of the alternatives herein utilize a truncated UCOE element, a conserved enhancer element derived from intronic regions within the human BTK locus in association with the human BTK proximal promoter to drive expression of a human codon-optimized BTK cDNA.
  • the optimal LV vectors of the one of the exemplary alternatives is referred to as 0.7 UCOE.DHS4.BTKpro.coBTK.
  • X-linked agammaglobulinemia is a hereditary X-linked immunodeficiency disorder caused by a mutation in the BTK gene (Bruton's Tyrosine Kinase). This disease affects approximately 1 in 100,000 males.
  • the clinical manifestations can include: lack mature B cells and serum immunoglobulins, susceptibility to lung, sinus, and skin infections with encapsulated bacteria, risks for sudden death due to bacteria sepsis, chronic and systemic infections with enteroviruses, chronic inflammatory bowel disease and increased risk for malignancy such as colon and other types of cancer.
  • the current treatment options for XLA can include lifelong pooled human immunoglobulin (IVIg or SCIg) every 3-4 weeks.
  • IVIg or SCIg lifelong pooled human immunoglobulin
  • the candidates for gene therapy can include those with single gene hematopoietic disorder.
  • Bruton's Tyrosine Kinase is a cytoplasmic non-receptor protein tyrosine kinase whose main role is in B cell receptor signaling pathway. BTK promotes B cell survival, proliferation and differentiation. BTK can maintain sustained calcium signal following BCR (B cell receptor) engagement and promotes FKB activation. Furthermore, BTK also plays role in cytokine, growth factor, and TLR signaling pathways. A role for BTK is shown in Figure 21, where BTK leads to the development of the B cells. As shown, XLA leads to a block at the Pre-B cell stage. BTK is expressed in B and myeloid lineages, and not expressed in T cells. The expression profile of endogenous BTK is shown in Figure 16.
  • the optimized lentiviral vectors will be used in gene therapy for subjects with X-linked agammaglobulinemia (XL A) designed to lead to long-term curative therapy for this disease.
  • XL A X-linked agammaglobulinemia
  • Shown in Figure 22 is an exemplary alternative in the development of the optimal BTK lentiviral gene therapy vector. This alternative would lead to a safe viral delivery platform and the expression of desired lineages with optimal levels of protein expression.
  • the optimized BTK lentiviral vector comprises a ubiquitous chromatin opening element (UCOE).
  • UCOE ubiquitous chromatin opening element
  • the UCOE can provide stable expression of transgenes regardless of integration site and can also confer resistance to silencing in an adjacent promoter (see Figure 1).
  • the optimized LV vectors for BTK expression were also shown to increase BTK expression in cells.
  • a few exemplary vectors that were used in several alternatives are shown in Figure 23.
  • the lentiviral vectors comprising ⁇ 57 ⁇ promoters also had an increase of BTK in myeloid cells.
  • this LV vector will lead to curative therapy, in particular XLA gene therapy.
  • the gene delivery platform was specialized for XLA therapy. This would allow restoration of endogenous BTK expression in B cells and myeloid cells, and rescue of immunological responses in those suffering from XLA.
  • the vector safety profile was further evaluated.
  • Previously investigated lentiviral vectors for XLA included those with promoter and transcription elements such as the BTK minimal promoter, the Ig heavy chain ⁇ intronic enhancer, as well as the 1.5 kb ubiquitous chromatin opening element.
  • promoter and transcription elements such as the BTK minimal promoter, the Ig heavy chain ⁇ intronic enhancer, as well as the 1.5 kb ubiquitous chromatin opening element.
  • the lentiviral vectors tested in the pre-clinical models in the alternatives described herein include 0.7 UCOE.BTKp.BTK and 0.7 UCOE.I-
  • the cells were then administered to the XLA mice, which were analyzed at 20-25 weeks post cell transfer.
  • the BTK expression was analyzed by flow cytometry. A secondary cellular transfer was also performed for long term repopulation of the stem cells (See Figure
  • the LV vector that included conserved BTK regulatory elements (derived from BTK introns 4 and 5) in association with the endogenous BTK improves BTK expression per viral integration. Also 0.7UCOE.I4,5.BTKpBTK LV comprises an efficient candidate for XLA gene therapy.
  • FIG. 9 Shown in Figure 9 are the expression profiles of four LV constructs and rescue of B cell development and function in primary recipient mice.
  • the vectors used were BTKpro.BTK, 1 5UCOE.BTKpro.BTK, 1.5UCOE.BTKpro.coBTK (human codon optimized BTK) and ⁇ . BTKpro.BTK.
  • the vector, the ⁇ . ⁇ . ⁇ was shown to increase BTK expression in all cells as shown in Figure 9.
  • the vector, the ⁇ . ⁇ . ⁇ was also shown to increase IgG expression in all cells as shown in Figure 10.
  • Increased BTK expression was also seen for the vector ⁇ . ⁇ . ⁇ in granulocytes, bone marrow and spleen B cells as seen in Figure 11.
  • the 1.5kbUCOE.BTKp and 1.5kbUCOE.BTKp.co vectors were shown to lead to sustained BTK expression with lower copy numbers in primary and secondary recipients (Figure 11).
  • the vector 0.7UCOE.BTKp.co was shown to lead to sustained BTK expression and lower copy numbers in XLA CD34 cells (Figure 15).
  • XLA and control CD34 cells were transduced with 0.7UCOE.BTKp.co at various multiplicities of infection (MOI) and cultured in vitro for 15 days. As shown, the XLA transduced cells had similar viability as the healthy control cells.
  • the BTK promoter in the lentiviral vectors were also used to evaluate BTK expression in wild type mice. As shown, the BTK promoter mimics BTK's endogenous expression pattern in mice ( Figure 16).
  • the ⁇ promoter was then tested for expression enhancement in the lentiviral vectors. Two vectors were tested, which had the ⁇ promoter as shown in Figure 17 panel a ( ⁇ enhancer and human BTK cDNA, either wild type or human codon- optimized, with a T2A-linked GFP).
  • Chicken BTK-/- DT40 cells were transduced with BTK- GFP or coBTK-GFP constructs; histograms show GFP and BTK expression ( Figure 17 panel b).
  • BTK-GFP and coBTK-GFP transduced cells were stained with an Indo-1 Ester AM fluorescent dye and stimulated with anti-IgM; calcium mobilization was monitored via flow cytometry. As shown, lentiviral constructs under control of the BTK promoter with ⁇ enhancer and human BTK cDNA led to controlled expression of the BTK-GFP in the chicken cells.
  • FIG. 18 Representative plots of BTK expression after flow cytometry of peripheral blood B cells and myeloid cells from gene therapy-treated KO mice was also performed ( Figure 18).
  • the vectors tested were: WT Mock, KO Mock, BTKp, 1 5kb.UCOE.BTKp, 1.5kb.UCOE.BTKp.co, and ⁇ . ⁇ .
  • Shown in Figure 18 panel c are the representative flow cytometry plots from bone marrow of gene therapy treated mice, stained with markers for early B cell development.
  • ⁇ . ⁇ primary transplant mice also had increased survival as compared to the control XLA mice treated with cells that were transduced with the mock vectors.
  • B cell specific promoters were also examined for their influence on BTK production.
  • a lentivector was manufactured, which comprised the ⁇ enhancer element and a B cell specific promoter, B29, fused to a gene encoding human BTK (huBTK) (Figure 27).
  • huBTK human BTK
  • the BTK promoter region was also examined in another alternative, to evaluate the influence of the BTK promoter in a lentiviral vector for BTK expression.
  • a lentiviral vector was manufactured, which comprised the BTK promoter (BTKpro) fused to the gene encoding human BTK (huBTK).
  • BTKpro BTK promoter
  • huBTK human BTK
  • BTK expression was not comparable to the expression in wild type cells.
  • the lentiviral vector comprising the BTK promoter region was unable to lead to increased expression of BTK and the expression of BTK was comparable to the knock out cells.
  • a lentiviral vector was manufactured, which comprised a ubiquitous chromatin opening element and a BTK promoter (BTKpro) fused to a gene encoding human BTK (Figure 29).
  • BTKpro BTK promoter
  • Figure 29 the enhancer element along with the promoter led to increase of BTK expression in both B cells, as well as, monocytes.
  • the expression was not comparable to the expression in the wild type cells.
  • a lentiviral vector was manufactured, which comprised a ⁇ enhancer and a BTK promoter (BTKpro) fused to a gene encoding human BTK (Figure 30).
  • BTKpro BTK promoter
  • Figure 30 the enhancer element along with the promoter led to increase of BTK expression in both B cells and monocytes.
  • the results show that the expression BTK in the transduced cells exceeded the amounts of BTK in the wild type cells.
  • the data shows that the ⁇ enhancer led to the most BTK expression of the vectors that were tested.
  • these constructs represent various iterations of 3 different elements (0.7UCOE, Enhancer, or BTK coding sequence, all having the same BTK promoter):
  • Human codon optimization can be performed by using algorithms that are known to those skilled in the art so as to create synthetic genetic transcripts optimized for high mRNA and protein yield in humans.
  • two constructs were tested for their ability to lead to increased BTK expression in cells.
  • the lentiviral constructs comprised the ⁇ element and the BTK promoter region.
  • the genes for expression were BTK and a human codon optimized BTK, both fused to GFP gene transcripts.
  • human codon optimized BTK led to an increase in GFP (MFI 2288), as well as, an increase in GFP.
  • BTK promoter BTK promoter
  • UCOE.5r p.co ubiquitous chromatin opening element
  • BTK promoter plus human codon optimized BTK BTK promoter
  • ⁇ .57 ⁇ ⁇ element plus BTK promoter
  • BTK promoter led to the highest expression of BTK in bone marrow, spleen and peritoneum from the B cells and the myeloid cells.
  • XLA ubiquitous chromatin opening element plus BTK promoter
  • UCOE.5r p.co ubiquitous chromatin opening element, BTK promoter plus codon optimized BTK
  • ⁇ , ⁇ ⁇ element plus BTK promoter
  • BTK promoter for BTK expression led to cells that were of the mature peripheral B cell population, indicating that the levels of BTK produced from the transduced cell would lead to restoration of mature B cell subsets in the mice ( Figure 33).
  • BTK deficient mice that were administered cells expressing BTK.
  • the cells were transduced with the following vectors: WT mock, DKO mock, BTKp (BTK promoter), UCOE.
  • BTKp BTK promoter
  • ubiquitous chromatin opening element plus BTK promoter ubiquitous chromatin opening element plus BTK promoter
  • UCOE.5r p.co ubiquitous chromatin opening element, BTK promoter plus human codon optimized BTK
  • FIG. 34 Shown in Figure 34 are the data that summarize the finding from 40 recipient mice that received the transduced cells for BTK expression. Mice that received the cells that were transduced with the BTK expression vector comprising UCOE.5r p.co , ⁇ .57 ⁇ or UCOE. BTKp were able to develop mature B cells in the bone marrow, spleen and peritoneum. Accordingly, aspects of the invention concern a lentiviral vector that comprises ubiquitous chromatin opening element, BTK promoter, and human codon optimized BTK, which can be used in methods to promote mature B cell development in subjects suffering from XL A.
  • B cell proliferation was also shown in mice that were treated with cells transduced with the lentiviral vectors that comprised the ubiquitous chromatin opening element and BTK promoter (XJCOE.BTKp) and a lentiviral vector that comprise the ⁇ element fused to the BTK promoter region ( ⁇ , ⁇ ) ( Figure 35).
  • Controls include the WT mock as well as the DKO mock cells.
  • Fluorescence activated cell sorting was performed on the cells using anti-IgM antibodies.
  • the cells with expressing BTK from the two lentiviral vectors UCOE.57Xp and ⁇ .57 ⁇ had expression of IgM, which is a basic antibody produced by B cells.
  • the cells were also treated with PMA and lonomycin.
  • PMA is used for activating PKC, while lonomycin, is used to trigger calcium release, which is needed for FAT signaling ( Figure 35).
  • the transduced cells were administered to mice and further tested for T cell dependent immune responses.
  • the cells were transduced with the following lentiviral vectors: WT mock, DKO, DKO mock, BTKp, UCOE.BTKp, and UCOE.BTKp.co (human codon optimized BTK), ⁇ . ⁇ . ⁇ (human codon optimized BTK), and ⁇ .57 ⁇ . T cell immune responses were then evaluated.
  • the cells transduced with ⁇ .57 ⁇ lentiviral vectors led to increased IgG and IgM expression ( Figure 38 -41).
  • the antibody levels were also associated with BTK expression, as well as, survival of the mice.
  • mice that were administered cells transduced with ⁇ .57 ⁇ lentiviral vectors led to increased mouse survival, as compared to the mice that were administered the DKO mock cells.
  • BTK expression was also evaluated in neutrophils and secondary recipient mice. As shown in Figure 43, administered cells transduced with ⁇ .57 ⁇ lentiviral vectors led to increased BTK expression in secondary recipient mice. However, there was increased viral copy number in secondary recipients in mice that were administered the viral vector BTKp ( Figure 44). Evaluation of the lentiviral therapy on long term survival provided evidence that the vectors UCOE.5riCp.co, UCOE.57Xp and ⁇ , ⁇ led to increased mouse survival.
  • BTK promoters in the lentiviral vectors exhibit significant BTK expression in B and myeloid cells.
  • UCOE.BTKpro and ⁇ . ⁇ rescue B cell development, absolute B cell numbers, B cell proliferation, and immune responses. Myeloid expression is also rescued with UCOE.BTKpro and ⁇ . ⁇ .
  • ⁇ -containing vectors lead to high-titer autoantibody production, thus, making them potentially unsafe for clinical use.
  • UCOE-Btkp- Btk vectors exhibit functional rescue at much lower viral copy number compared with non- UCOE vectors.
  • UCOE-Btkp-Btk vectors exhibit sustained marking in both murine and human HSC.
  • the UCOE.BTKp-coBTK lentiviral vector represents an improved and unique clinical vector platform for additional modification.
  • the 1.5 kb UCOE was truncated to 0.7UCOE to create the lentiviral vector 0.7UCOE.i?7Xp ( Figure 2).
  • Potential human BTK enhancer elements were also added to create 0 OCOE.JE.BTKp as shown in Figure 2.
  • the intronic regions included intron 4 and 5 from a human BTK locus that is in association with a human BTK proximal promoter.
  • 0.7UCOE.IE The cells transduced with 0.7UCOE.IE exhibited an improved safety profile over 0.7UCOE, as well (see Figure 49). As shown, 0.7UCOE and 0.7UCOE.IE both produce low autoantibody titers compared to autoimmune controls (and previous non-Btk promoter vectors, while 0.7UCOE.IE exhibits similar efficacy with few viral integrations per cell.
  • Hypersensitive sites (Figure 50).
  • the vectors were the following: O ⁇ UCOEJJTXp.coBTK
  • 0.7UCOE.ffi4,5,.57Xp.coBTK 0.7UCOE element, intron 4 and 5 of the human BTK locus that is in association with the human BTK proximal promoter, BTK promoter and the human codon optimized BTK.
  • constructs with candidate introns and DNAse hypersensitive sites were constructed ( Figure 52). Constructs were the following: 0.7UCOE.BTKp.coBTK, 0.7UCOE.IE.BTKp.coBTK, 0.7UCOE.DHS4.BTKp.coBTK, 0.7UCOE.DHSl,2.BTKp.coBTK, 0.7UCOE.DHSl,2,4.BTKp.coBTK, and 0.7UCOE.DHS1- 5 BTKp.coBTK.
  • BTK expression in vivo was examined using the following vectors: 0.7UCOE.BTKp.coBTK, 0.7UCOE.DHS4.BTKp.coBTK and 0.7UCOE.DHSl-5.BTKp.coBTK.
  • Lin-cells were harvested from TBK donor mice.
  • transduction a matched volume was achieved bylOul virus/million cells (target: matched input VCN of ⁇ 3, as predicted by in vitro test) followed by a 16 hour transduction, 4xl0e 6 cells/ml in SCGM transduction media (mSCF, mTPO) + polybrene.
  • the 6 week peripheral blood lymphocyte distribution is shown in Figure 59.
  • the cells expressing BTK from 0.7UCOE, 0.7UCOE.DHS4 and 0.7UCOE.DHS1-4 had an increase in B cell production (Figure 59).
  • BTK expression was shown in the B cells, monocytes and neutrophils at the evaluation at 6 weeks and 12 weeks in mice that were administered the cells comprising the 0.7UCOE, 0.7UCOE.DHS4 and 0.7UCOE.DHS1-4 lentiviral vectors ( Figure 60).
  • BTK was shown to be expressed across all the subsets ( Figure 61).
  • the objective of the following experiments was to compare the BTK expression/staining of two different versions of human codon optimized BTK.
  • the set-up included: Isolated Lin- cells from TBK mice, Transduction media: complete SCGM + mSCF + mTPO + polybrene, addition of 5, 10, or 20ul virus to 1x106 cells (4x106 cells/ml), 7-day in vitro culture, then BTK stain and VCN at Day 7.
  • the lentiviral constructs were: 0.7UCOE.BTKp.coBTK (Titer: 1.17E +09 ), 0.7UCOE.DHS4.BTKp.coBTK (Titer: 1.09E +09 ), 0.7UCOE.BTKp.newcoBTK (Titer: 1.81E +08 ), and 0.7UCOE.DHS4.BTKp.newcoBTK (Titer: 1.13E +08 ) ( Figure 62).
  • the experimental groups include: 0.7UCOE.BTKp.coBTK (Titer: 1.17E +09 ) (5 mice), 0.7UCOE.DHS4.BTKp.coBTK (Titer: 1.09E +09 ) (5 mice), 0.7UCOE.BTKp.newcoBTK (Titer: 1.81E+ +08 ) (5 mice), 0.7UCOE.DHS4.BTKp.newcoBTK (Titer: 1.13E +08 ) (5 mice), KO Mock (3 mice), WT Mock (5 mice) and Unirradiated controls (1 mouse).
  • Transduction set-up includes: Volume match with lOul/million cells (for consistency with current in vivo experiments) ( Figure71).
  • lentiviral vectors were tested: KO mock, 0.7UCOE.BTKp (7.52E +07 ), 0.7UCOEfwd.BTKp col4 (6.79E +08 ), 0.7UCOEfwd.BTKp col6 (1.79E +09 ), and WT mock.
  • the levels of BTK between the lentiviral vectors 0.7UCOE.BTKp (7.52E +07 ), 0.7UCOEfwd.BTKp col4 (6.79E +08 ), 0.7UCOEfwd.BTKp col6 (1.79E +09 ) were similar, however VCN was increased at increased volume of virus.
  • the BTK promoter is a robust promoter.
  • 0.7kb UCOE effectively prevents silencing of BTK expression and leads to increased vector titer. Furthermore, placing the UCOE in the forward orientation increases titer significantly, but does not alter BTK expression. This indicates that the reverse UCOE orientation performs equivalent to high titer constructs.
  • enhancer elements it was shown that 0.7UCOE.IE.Btkp exhibits rescue of BTK expression function with fewer viral integrations compared to 0.7UCOE.Btkp and the addition of smaller enhancer elements (DHS sites) has not increased titer compared to larger IE construct.
  • UCOE in the forward orientation increases IE titer by >1 log indicating that the forward orientation of the UCOE will increase titer in all constructs.
  • 0.7UCOE.DHS4.Btkp exhibits increased BTK expression in B and myeloid cells as compared to 0.7UCOE.IE.Btkpro indicating that 0.7UCOE.DHS4.Btkp vector is a particularly robust vector.
  • the new construct leads to a significant increase in BTK expression as compared to the previous co-Btk construct.
  • the XLA B cell phenotype in the periphery include a markedly reduced percentage of B cells, higher percentage of transition/ immature B cells (CD38+ CD24+ CD 10 high), and a lack of mature B cells (CD38- CD24- CD10 low ).
  • FIG. 76 The phenotype of the engrafted XLA stem cell (spleen) is shown in Figure 76 for patients XLA P2, P3 and P4. As shown, the patients exhibited equivalent %Pro B cell between the XLA and healthy group.
  • the B cell developmental block was examined for all XLA patients. As shown in Figure 77, BTK allows development of Pro B cell to a mature B cell. The cells were analyzed for hCD19+, CD22+ and CD179a+. As shown, the XLA patients had a significantly higher percentage of pre-B cells as compared to the healthy control. These XLA B cells are thus blocked at the Pre B cell stage in which they are then able to migrate to the spleen ( Figure 78 and 85). [0216] The B cell function by Ca 2+ was also examined in the XLA cells ( Figure 80. As shown, the XLA B cells were not able to flux with IgM as compared to the control.
  • XLA patients have a lower number and percentage of B cells in spleen. Additionally, the B cell development is arrested at the pre-B cell stage in the bone marrow. The next step was to recapitulate the XLA patient's B cell phenotype in NSG mice to examine the effects of the BTK expressing lentiviral vector system.
  • BTK expression was also seen in the cells derived from patient XLA P2 transduced with the 0.7UCOE.BTK.pBTK lentiviral vector. As shown in a non-selective environment, 0.7UCOE.BTKpBK leads to a higher expression of BTK, as compared to the lentiviral vector comprising the DNAse hypersensitivity site 4 region ( Figure 86).
  • test construct comprises the
  • UCOE element a BTK promoter, a human codon optimized BTK gene, as well as, the T2A-
  • Intron 4 and 5 contig were tested in vivo in mouse gene therapy model, calling it intronic enhancer 4, 5 or IE4-5. These studies used the enhancer/promoter elements to drive expression of a codon-optimized human BTK coding sequence, and showed that inclusion of IE4-5 improved expression of BTK per viral copy number.
  • polypeptides that encode the sequences for the BTK for expression are set forth in SEQ ID NO's 33-35, and 41-45.
  • DHS1, 2, and 3 DNase hypersensitive sites in B and myeloid cells identified within intron 1 (DHS1, 2, and 3) that were also for testing, due to some evidence that intron 1 may improve MFI in vitro.
  • DHS 3 and 4 were identified as having properties of transcriptional enhancers in B cells by the ENCODE segmentation analysis; DHS5 was identified as a CTCF binding site/candidate insulator.
  • Truncation of 1.5kb UCOE to 0.7kb and identification of potential DNA enhancer elements may improve titer and BTK expression in Lentiviral constructs.
  • the UCOE element spans a large, CpG-rich region across the divergently transcribed promoter regions for the housekeeping genes CBX3 and HNRPA2B 1, and has traditionally been truncated to a 1.5-2.2kb region by various groups for use in protein expression constructs.
  • the 1.5kb UCOE used here starts at Exon 1 of CBX3 and spans past CBX3 Alternate Exon 1. Truncation of this region to 0.7kb eliminates the region downstream of Alt. Ex. 1.
  • DHS DNasel Hypersensitive sites
  • DHS3, DHS4, and DHS5 blue boxes.
  • ENCODE Genome Segmentation identified a predicted Enhancer element around DHS4 (yellow box). Exons are shown as black boxes.
  • Figure 94 are the results from restoration of BTK expression in affected hematopoietic lineages and B cell development in gene therapy mice treated with 0.7UCOE vectors. The proportion of BTK+ cells and numerical reconstitution of B and myeloid cell subsets was assessed at 19-23 weeks post-transplant.
  • Figure 94A Representative flow plots showing intracellular BTK staining in splenic B cells at endpoint analysis.
  • Figure 94B-D Percent of BTK+ cells in bone marrow ( Figure 94B), spleen ( Figure 94C), and peritoneal (Figure 94D) lymphocyte subsets from gene therapy treated groups, determined by flow cytometry.
  • Cell subsets are defined as Neutrophils (CDl lb+ GR1+), Monocytes (CDl lb+), and B cells (B220+).
  • Figure 94E-F Stacked bars show the average counts of B cell subsets in bone marrow ( Figure 94E), spleen ( Figure 94F), and peritoneum ( Figure 94G.
  • Late B cell development (spleen): transitional Tl (CD24hi, CD21-), transitional T2 (CD24hi, CD21int), marginal zone/precursor MZ/MZP (CD24hi, CD21hi), and follicular FM (CD24int, CD21int).
  • Peritoneal B cell subsets Bl (IgM+ CD43+), and B2 (CD43-).
  • Figure 94H Rescue of BTK expression in B cell subsets, measured by flow cytometry. ( Figure 941) BTK+ MFI across B cell development, normalized to WT Mock in each individual experiment.
  • Figure 95 shows the restoration of B cell function in vivo and in vitro.
  • Figure 95A Mice were immunized with NP-CGG in Alum at 12 weeks post-transplant. Levels of NP-specific IgG in serum from immunized mice was measured by ELISA and expressed relative to an IgG standard. High-affinity NP-IgG was measured from serum prior to (-) and 10 days following primary immunization (1°). One month following primary challenge, mice were re-challenged with P-CGG in PBS and serum was collected 10 days later (2°).
  • FIG. 95B-C Total serum IgG (Figure 95B) and IgM (Figure 95C) in serum from treated mice was measured by ELISA and endpoint analysis (21-23 weeks post- transplant).
  • Figure 95D-F At endpoint analysis, B cells were isolated from splenocytes by CD43- magnetic separation, labeled with Cell Trace Violet and stimulated in vitro with IgM, LPS, or a media control.
  • Figure 95D The percentage of BTK+ B cells that underwent > 1 cell division 72 hours after incubation with anti-mouse IgM antibodies, LPS or media only (read out by flow cytometry).
  • Figure 95E BTK+ MFI of cells after each division (D0-D4), normalized to WT Mock.
  • Figure 96 shows some vector safety considerations. Levels of anti-dsDNA IgG (Figure 96A) and anti-dsDNA IgG2c (Figure 96B) in serum from treated mice were measured by ELISA and are depicted as absorbance readings (OD450). Serum from the known autoimmune-prone WAS chimeric mouse model and mice treated with Eu.BTKp were included as positive controls. ( Figure 96C) Genomic DNA was isolated from total bone marrow and spleen at endpoint analysis and the number of viral integrations per cell (VCN) was quantified by qPCR.
  • VCN viral integrations per cell
  • CD34 cell source XLA Patient 3 (missense mutation) and healthy donor #15.
  • the transduction protocol comprises one hit of LV after a 48 hour prestimulation in SCGM (TPO, FLT3 and SCF at lOOng/ml) .
  • the MOI used 5.
  • the lentivirus used was 0.7 UCOE.
  • BTKp.BTK .co2 (titer: 7 X 10 8 ) and DHS4.co2 (titer: 1 X 10 9 )
  • mice Healthy donor #15 (HD)
  • mice XLA P3 mock treated (XLAP3)
  • mice 0.7 UCOE.BTKp.BTK.co2 (0.7UCOE)
  • mice 0.7 UCOE.DHS4.BTKp.BTK.co2 (DHS4)
  • FIG 98 Shown in Figure 98 are the results from an equivalent bone marrow engraftment of human hematopoietic cells between treatment groups.
  • Human stem cells from XLA patient either with or without gene therapy treatments, engrafted into the bone marrow equivalently as those from a healthy donor.
  • Representative flow plots showing various markers of human immune cells including human and mouse CD45 (a marker for hematopoietic cells) CD33 (myeloid cells) and CD19 (B cells), CD4 and CD 8 (T cells).
  • Human hematopoietic cells are hCD45+ and mCD45- from total live BM cells; CD33 and CD 19 markers were analyzed from hCD45+gate; CD4 and CD8 were analyzed from the CD33-CD19- gate.
  • Figures 98B and 98C. % human CD45 and total number of hCD45 cells engrafted in BM; n 4 for each cohort (XL A3, XLA3+LV 0.7UCOE.BTKp.BTKco2, XL A3 + LV 0.7UCOE.DHS4.BTKp.BTKco2, and healthy donor).
  • FIG. 99 Shown in Figure 99, are the results of mouse recipients of XLA3 HSC given gene therapy using 0.7UCOE.BTKp BTKco2 or 0.7UCOE.DHS4.BTKp.BTKco2 showing significantly increased numbers of splenic human hematopoietic cells.
  • Figure 99A Representative flow plots showing markers of human immune cells including human and mouse CD45 (a marker for hematopoietic cells) CD33 (myeloid cells) and CD19 (B cells), CD4 and CD 8 (T cells).
  • Human hematopoietic cells are both hCD45+ and mCD45-; CD33 and CD 19 markers were analyzed from the hCD45+gate; CD4 and CD8 were analyzed from the CD33-CD19- gate.
  • FIG. 101 Shown in Figure 101, is a representative flow plot for B cell development subsets in spleen.
  • Human CD24 and hCD38 cells were gated from human CD19+cells. Immature B cells (hCD24+hCD38+) are IgM+ IgD - CDlOhigh; mature B cells (CD241ow CD381ow) are IgM+IgD+ CD 10 low.
  • FIG. 102 Shown in Figure 102 are the results of recipients of LV transduced XLA3 HSC (using 0.7UCOE.BTKp.BTKco2 or 0.7UCOE.DHS4 BTKp.BTKco2). As shown, the recipients have an increased proportion of splenic immature B cells (including CD19+CD24+CD38+ B cells and CD19+CD24+CD38+IgM+ cells) compared to XLA controls.
  • Graphs summarize the results from flow cytometry shown in Figure 4, looking at specific subpopulations of immature B cells.
  • A % of Immature B cells (CD24+ CD38+ ) in the spleen
  • B % of immature B cells that are IgM+
  • C- D Total number of immature B cells (CD24+ CD38+ ) and CD24+hCD38+ IgM+ cells in the spleen
  • E overlay of CD 10 histogram , showing the mean fluorescence intensity MFI shift in CD 10 compared to healthy donor.
  • recipients of LV transduced XLA HSC (0.7UCOE.BTKp.BTKco2 or 0.7UCOE.DHS4 BTKp.BTKco2 ) have increased proportions of mature B cells (CD19 + CD24 low CD38 low IgM + and IgD + ) compared to XLA controls.
  • Figures 104A-C % of mature B cells (hCD24 low hCD38 low ) and the percent of mature B cells that are IgM + and IgM + IgD + in the spleen, respectively;
  • Figures 103D-F Total number of splenic mature B cells and mature B cells that are IgM + and IgM + IgD + , respectively;
  • Figure 103G histogram overlay of CD 10, showing similar MFI of CD 10 as that of a healthy donor.
  • Shown in Figure 104 is a representative flow plot for B cell development subsets in the bone marrow.
  • Blue color represents individual mice treated with LV 0.7.UCOE.BTKp.BTKco2, and orange color individual mice treated with LV 0,7UCOE.DHS4.UCOE.BTKp.BTKco2.
  • recipients of LV transduced XLA HSC (using LV 0.7UCOE BTKp BTKco2 or LV 0.7UCOE.DHS4 BTKp.BTK.co2) exhibit gene marking of 0.2-2 viral copy number (VCN)/cell in vivo.
  • VCN viral copy number
  • the number of viral integrations per cell were calculated in BM (A) and Spleen (B), as well as the original CD34 cells prior to transplantation (C) as determined by quantitative PCR. No significant difference between 0.7UCOE.Co2.BTKp.BTK and DHS4.BTKp.BTKCo2.
  • recipients of XLA3 HSC that received gene therapy with LV 0.7UCOE.BTKp.BTKco2 or 0.7UCOE.DHS4.BTK.pBTKco2 produce B cells that secrete IgM in vivo.
  • FIG. 108 recipients of XL A3 HSC transduced with LV 0.7UCOE.BTKp.BTKco2 or 0.7UCOE.DHS4.BTKp.BTKco2 exhibit a restored capacity for B cells to flux calcium in response to B cell receptor (BCR) engagement.
  • BCR B cell receptor
  • Figure 108A provides a representative flow plots showing the gating strategy for human CD 19+ cells evaluated for the calcium flux.
  • Figure 108B provides a kinetic analysis of intracellular calcium levels by flow cytometry after BCR engagement with human IgM antibodies.
  • FIG. 110 Shown in Figure 110, is a representative flow plot for in vitro B cell class switching. Splenic cells from recipient mice were cultured in B cell differentiation protocol (previous slide) then stained for markers allowing the identification of plasma B cells. Media supernatant was also collected for determining human IgM and IgG levels by ELISA.
  • recipients of LV transduced XLA HSC (using 0.7UCOE BTKp BTKco2 or DHS4 BTKpBTK.co2) generate B cells that are capable of responding to cytokine and T-cell dependent signals leading to antibody secretion.
  • 0.5x 10 6 total splenic cells (pooled from mice in each cohort) were cultured in IMDM + 10% FBS + 2-mercaptoethanol media.
  • Phase I cells were cultured for 7 days with MegaCD40L (lOOng/ml) + CpG ODN 2006 ( ⁇ g/ml) + IL-2 (50ng/ml) + IL-10 (50ng/ml) + IL-15 (lOng/ml).
  • Phase I cells were washed with PBS 2x and cultured for 3 days in Phasell media (IMDM+ 10% FBS + BME supplemented with IL-2 (50ng/ml) + IL-6 (50ng/ml) + IL-10 (50ng/ml) + IL-15 (lOng/ml), then switched to Phase III media (EVIDM+ 10% FBS + BME supplemented with IL-6 (50ng/ml) + IL-15 (lOng/ml) + IFN-a 2B (lOOU/ml) for 4 days.
  • Phase III media EIDM+ 10% FBS + BME supplemented with IL-6 (50ng/ml) + IL-15 (lOng/ml) + IFN-a 2B (lOOU/ml) for 4 days.
  • media supernatants were collected and the levels of human IgG (A) and IgM (B) were determined by ELISA using a human IgM and IgG standard.
  • a polynucleotide for sustained Bruton's tyrosine kinase (BTK) expression is provided.
  • the polynucleotide can comprise a first sequence encoding a ubiquitous chromatin opening element (UCOE), a second sequence encoding a promoter and a third sequence encoding BTK.
  • UCOE ubiquitous chromatin opening element
  • the UCOE is 2kb, 1.5kb, lkb, 0.75kb, 0.5kb or 0.25 kb or any number of kilobases in between a range defined by any two afore mentioned values.
  • the first sequence comprises the nucleic acid sequence set forth in SEQ ID NO: 1, and/or SEQ ID NO: 2.
  • the promoter is a BTK promoter.
  • the BTK promoter comprises the nucleic acid sequence set forth in SEQ ID NO: 5.
  • the third sequence is codon optimized for expression in humans.
  • the third sequence comprises the sequence set forth in SEQ ID NO: 6 or SEQ ID NO: 7.
  • the promoter is a B cell specific promoter.
  • the B cell specific promoter comprises the B cell specific promoter, B29.
  • the B29 promoter sequence comprises a sequence set forth in SEQ ID NO: 46.
  • the B cell specific promoter is an endogenous promoter.
  • the polypeptide further comprises one or more enhancer elements.
  • the one or more enhancer elements comprise at least one DNase Hypersensitive Site (DHS).
  • the DNase Hypersensitive Site is DNase Hypersensitive Site 1 (DHS1), DNase Hypersensitive Site 2, (DHS2) DNase Hypersensitive Site 3 (DHS3), DNase Hypersensitive Site 4 (DHS4) and/or DNase Hypersensitive Site 5 (DHS5).
  • the DNase Hypersensitive Site comprises a sequence set forth in SEQ ID NO: 3.
  • the one or more enhancer elements comprise at least one intronic region.
  • the at least one intronic region is from a human BTK locus that is in association with a human BTK proximal promoter.
  • the at least one intronic region is intron 4, intron 5 and/or intron 13 of the human BTK locus that is in association with the human BTK proximal promoter.
  • the intronic region comprises the sequence set forth in SEQ ID NO: 9 (intron 4), SEQ ID NO: 10 (intron 5) and/or SEQ ID NO: 11 (intron 13).
  • the one or more enhancer elements comprise the sequence set forth in SEQ ID NO: 4, SEQ ID NO: 14 or SEQ ID NO: 15.
  • the UCOE is in a reverse orientation or forward orientation. In some alternatives, the UCOE is in a forward orientation.
  • the one or more enhancer elements comprises the sequences set forth in SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19 and/or SEQ ID NO: 20.
  • the polynucleotide further comprises a gene upstream of a BTK promoter.
  • the gene upstream of a BTK promoter is a BTK enhancer.
  • the BTK enhancer comprises the sequence set forth in SEQ ID NO: 21 or SEQ ID NO: 22.
  • the polypeptide comprises a sequence set forth in SEQ ID NO's: 33, 34, 35, 41, 42, 43, 44 or 45.
  • 0.7UCOE comprises a sequence set forth in SEQ ID NO: 1.
  • 0.7UCOE comprises a sequence set forth in SEQ ID NO: 2.
  • the enhancer comprises a sequence set forth in SEQ ID NO: 3 or 4.
  • a vector for sustaining BTK expression in cells is also provided.
  • the vector can comprise a first sequence encoding an ubiquitous chromatin opening element (UCOE), a second sequence encoding a promoter, and a third sequence encoding BTK.
  • the first sequence comprises the nucleic acid sequence set forth in SEQ ID NO: 1, and/or SEQ ID NO: 2.
  • the promoter is a BTK promoter.
  • the promoter comprises the nucleic acid sequence set forth in SEQ ID NO: 5.
  • the third sequence is codon optimized for expression in humans.
  • the third sequence comprises the sequence set forth in SEQ ID NO: 6 or SEQ ID NO: 7.
  • the vector further comprises a B cell specific promoter.
  • the B cell specific promoter comprises the B cell specific promoter, B29.
  • the B29 promoter sequence comprises a sequence set forth in SEQ ID NO: 46.
  • the B cell specific promoter is an endogenous promoter.
  • the vector further comprises one or more enhancer elements.
  • the one or more enhancer elements comprise at least one intronic region.
  • the one or more enhancer elements comprise a DNase Hypersensitive Site (DHS).
  • the DNase Hypersensitive Site is DNase Hypersensitive Site 1 (DHS1), DNase Hypersensitive Site 2, (DHS2) DNase Hypersensitive Site 3 (DHS3), DNase Hypersensitive Site 4 (DHS4) and/or DNase Hypersensitive Site 5 (DHS5).
  • the DNase Hypersensitive Site comprises a sequence set forth in SEQ ID NO: 3.
  • the at least one intronic region is from a human BTK locus that is in association with a human BTK proximal promoter.
  • the at least one intronic region is intron 4 intron 5 and/or intron 13of the human BTK locus that is in association with the human BTK proximal promoter.
  • the intronic region comprises the sequence set forth in SEQ ID NO: 9 (intron 4), SEQ ID NO: 10 (intron 5) and/or SEQ ID NO: 11 (intron 13).
  • the one or more enhancer elements comprise the sequence set forth in SEQ ID NO: 4, SEQ ID NO: 14 or SEQ ID NO: 15.
  • the UCOE is in a reverse orientation or forward orientation. In some alternatives, the UCOE is in a forward orientation.
  • the vector is a lentiviral-based vector of a B lineage specific lentiviral vector.
  • the cells are B cells. In some alternatives, the cells are myeloid cells. In some alternatives, the cells are hematopoietic stem cells. In some alternatives, the cells are CD34+ hematopoietic stem cells.
  • the one or more enhancer elements comprise the sequences set forth in SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19 and/or SEQ ID NO: 20.
  • the polynucleotide further comprises a gene upstream of a BTK promoter.
  • the gene upstream of a BTK promoter is a BTK enhancer.
  • the BTK enhancer comprises the sequence set forth in SEQ ID NO: 21 or SEQ ID NO: 22.
  • the polypeptide or vector comprises a sequence set forth in SEQ ID NO's: 33, 34, 35, 41, 42, 43, 44 or 45.
  • 0.7UCOE comprises a sequence set forth in SEQ ID NO: 2.
  • the enhancer comprises a sequence set forth in SEQ ID NO: 3 or 4.
  • a cell for expression of BTK comprising: a polynucleotide, which comprises a first sequence encoding an ubiquitous chromatin opening element (UCOE), a second sequence encoding a promoter and a third sequence encoding BTK.
  • the polynucleotide is in a vector.
  • the vector can comprise a first sequence encoding a ubiquitous chromatin opening element (UCOE), a second sequence encoding a promoter, and a third sequence encoding BTK.
  • the first sequence comprises the nucleic acid sequence set forth in SEQ ID NO: 1, and/or SEQ ID NO: 2.
  • the promoter is a BTK promoter.
  • the promoter comprises the nucleic acid sequence set forth in SEQ ID NO: 5
  • the third sequence is codon optimized for expression in humans.
  • the third sequence comprises the sequence set forth in SEQ ID NO: 6 or SEQ ID NO: 7.
  • the vector further comprises a B cell specific promoter.
  • the B cell specific promoter comprises the B cell specific promoter, B29.
  • the B29 promoter sequence comprises a sequence set forth in SEQ ID NO: 46.
  • the B cell specific promoter is an endogenous promoter.
  • the vector further comprises one or more enhancer elements. In some alternatives, the one or more enhancer elements comprise at least one intronic region.
  • the one or more enhancer elements comprises a DNase Hypersensitive Site (DHS).
  • DHS DNase Hypersensitive Site
  • the DNase Hypersensitive Site is DNase Hypersensitive Site 1 (DHS1), DNase Hypersensitive Site 2, (DHS2) DNase Hypersensitive Site 3 (DHS3), DNase Hypersensitive Site 4 (DHS4) and/or DNase Hypersensitive Site 5 (DHS5).
  • DNase Hypersensitive Site comprises a sequence set forth in SEQ ID NO: 3.
  • the at least one intronic region is from a human BTK locus that is in association with a human BTK proximal promoter.
  • the at least one intronic region is intron 4 intron 5 and/or intron 13 of the human BTK locus that is in association with the human BTK proximal promoter.
  • the intronic region comprises the sequence set forth in SEQ ID NO: 9 (intron 4), SEQ ID NO: 10 (intron 5) and/or SEQ ID NO: 11 (intron 13).
  • the one or more enhancer elements comprise the sequence set forth in SEQ ID NO: 4 (SEQ ID NO: 4: intron 4-5), SEQ ID NO: 14 or SEQ ID NO: 15.
  • the UCOE is in a reverse orientation or forward orientation. In some alternatives, the UCOE is in a forward orientation.
  • the vector is a lentiviral-based vector of a B lineage specific lentiviral vector.
  • the cells are B cells.
  • the cells are myeloid cells.
  • the cells are hematopoietic stem cells.
  • the cells are CD34+ hematopoietic stem cells.
  • the one or more enhancer elements comprises the sequences set forth in SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19 and/or SEQ ID NO: 20.
  • the polynucleotide further comprises a gene upstream of a BTK promoter.
  • the gene upstream of a BTK promoter is a BTK enhancer.
  • the BTK enhancer comprises the sequence set forth in SEQ ID NO: 21 or SEQ ID NO: 22.
  • the vector is a lentiviral vector.
  • the cell is a B cell.
  • the cells are myeloid cells.
  • the cell is a hematopoietic stem cell.
  • the cell is a CD34+ hematopoietic stem cell.
  • 0.7UCOE comprises a sequence set forth in SEQ ID NO: 2.
  • the enhancer comprises a sequence set forth in SEQ ID NO: 3 or 4.
  • a method of promoting B cell survival, proliferation and/or differentiation in a subject in need thereof comprising administering the cell of any one of the alternatives herein to the subject or a cell comprising the polynucleotide of any one of the alternatives herein or the vector of any one of the alternatives herein to the subject in need and, optionally identifying the subject as one that would benefit from receiving a therapy that would promote B cell survival, proliferation and/or differentiation in advance of administering the cell and/or, optionally, measuring B cell survival, proliferation and/or differentiation in said subject or in a biological sample obtained from said subject.
  • the vector can comprise a first sequence encoding a ubiquitous chromatin opening element (UCOE), a second sequence encoding a promoter, and a third sequence encoding BTK.
  • the first sequence comprises the nucleic acid sequence set forth in SEQ ID NO: 1, and/or SEQ ID NO: 2.
  • the promoter is a BTK promoter.
  • the promoter comprises the nucleic acid sequence set forth in SEQ ID NO: 5.
  • the third sequence is codon optimized for expression in humans.
  • the third sequence comprises the sequence set forth in SEQ ID NO: 6 or SEQ ID NO: 7.
  • the vector further comprises a B cell specific promoter.
  • the B cell specific promoter comprises the B cell specific promoter, B29.
  • the B29 promoter sequence comprises a sequence set forth in SEQ ID NO: 46.
  • the B cell specific promoter is an endogenous promoter.
  • the vector further comprises one or more enhancer elements.
  • the one or more enhancer elements comprise at least one intronic region.
  • the one or more enhancer elements comprise a DNase Hypersensitive Site (DHS).
  • the DNase Hypersensitive Site is DNase Hypersensitive Site 1 (DHS1), DNase Hypersensitive Site 2, (DHS2) DNase Hypersensitive Site 3 (DHS3), DNase Hypersensitive Site 4 (DHS4) and/or DNase Hypersensitive Site 5 (DHS5).
  • the DNase Hypersensitive Site comprises a sequence set forth in SEQ ID NO: 3.
  • the at least one intronic region is from a human BTK locus that is in association with a human BTK proximal promoter.
  • the at least one intronic region is intron 4 intron 5 and/or intron 13of the human BTK locus that is in association with the human BTK proximal promoter.
  • the intronic region comprises the sequence set forth in SEQ ID NO: 9 (intron 4), SEQ ID NO: 10 (intron 5) and/or SEQ ID NO: 11 (intron 13).
  • the one or more enhancer elements comprise the sequence set forth in SEQ ID NO: 4 (SEQ ID NO: 4: intron 4-5), SEQ ID NO: 14 or SEQ ID NO: 15.
  • the UCOE is in a reverse orientation or forward orientation. In some alternatives, the UCOE is in a forward orientation.
  • the vector is a lentiviral-based vector of a B lineage specific lentiviral vector.
  • the cells are B cells. In some alternatives, the cells are myeloid cells. In some alternatives, the cells are hematopoietic stem cells. In some alternatives, the cells are CD34+ hematopoietic stem cells.
  • the one or more enhancer elements comprises the sequences set forth in SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19 and/or SEQ ID NO: 20.
  • the polynucleotide further comprises a gene upstream of a BTK promoter.
  • the gene upstream of a BTK promoter is a BTK enhancer.
  • the BTK enhancer comprises the sequence set forth in SEQ ID NO: 21 or SEQ ID NO: 22.
  • the cell is from the subject and, wherein the cell is genetically modified by introducing the polynucleotide or the vector of any one of alternatives described above into the cell.
  • the administering is performed by adoptive cell transfer.
  • the cell is a B cell. In some alternatives, the cells are myeloid cells. In some alternatives, the cell is a hematopoietic stem cell. In some alternatives, the cell is a CD34+ hematopoietic stem cell. In some alternatives, the subject is male. In some alternatives, the subject is suffering from X linked agammaglobulinemia (XLA). In some alternatives, the subject is selected to receive immunoglobulin replacement therapy. In some alternatives, the subject is selected to receive targeted anti-microbial agents. In some alternatives, the polypeptide or vector comprises a sequence set forth in SEQ ID NO's: 33, 34, 35, 41, 42, 43, 44 or 45.
  • the polypeptide or vector comprises a sequence set forth in SEQ ID NO's: 33, 34, 35, 41, 42, 43, 44 or 45.
  • 0.7UCOE comprises a sequence set forth in SEQ ID NO: 2.
  • the enhancer comprises a sequence set forth in SEQ ID NO: 3 or 4.
  • a method of treating, inhibiting, or ameliorating X linked agammaglobulinemia (XLA) or disease symptoms associated with XLA in a subject in need thereof comprising: administering the cell of any one of the alternatives herein to the subject or a cell comprising the polynucleotide of any one of the alternatives herein or the vector of any one of the alternatives herein to the subject in need and, optionally identifying the subject as one that would benefit from receiving a therapy for XLA or disease symptoms associated with XLA and/or, optionally, measuring an improvement in the progression of XLA or an improvement in a disease symptom associated with XLA in said subject.
  • XLA X linked agammaglobulinemia
  • the vector can comprise a first sequence encoding a ubiquitous chromatin opening element (UCOE), a second sequence encoding a promoter, and a third sequence encoding BTK.
  • the first sequence comprises the nucleic acid sequence set forth in SEQ ID NO: 1 and/or SEQ ID NO: 2.
  • the promoter is a BTK promoter.
  • the promoter comprises the nucleic acid sequence set forth in SEQ ID NO: 5.
  • the third sequence is codon optimized for expression in humans.
  • the third sequence comprises the sequence set forth in SEQ ID NO: 6 or SEQ ID NO: 7.
  • the vector further comprises a B cell specific promoter.
  • the B cell specific promoter comprises the B cell specific promoter, B29.
  • the B29 promoter sequence comprises a sequence set forth in SEQ ID NO: 46.
  • the B cell specific promoter is an endogenous promoter.
  • the vector further comprises one or more enhancer elements.
  • the one or more enhancer elements comprise at least one intronic region.
  • the one or more enhancer elements comprise a DNase Hypersensitive Site (DHS).
  • the DNase Hypersensitive Site is DNase Hypersensitive Site 1 (DHS1), DNase Hypersensitive Site 2, (DHS2) DNase Hypersensitive Site 3 (DHS3), DNase Hypersensitive Site 4 (DHS4) and/or DNase Hypersensitive Site 5 (DHS5).
  • the DNase Hypersensitive Site comprises a sequence set forth in SEQ ID NO: 3.
  • the at least one intronic region is from a human BTK locus that is in association with a human BTK proximal promoter.
  • the at least one intronic region is intron 4 intron 5 and/or intron 13of the human BTK locus that is in association with the human BTK proximal promoter.
  • the intronic region comprises the sequence set forth in SEQ ID NO: 9 (intron 4), SEQ ID NO: 10 (intron 5) and/or SEQ ID NO: 11 (intron 13).
  • the one or more enhancer elements comprise the sequence set forth in SEQ ID NO: 4 (SEQ ID NO: 4: intron 4-5), SEQ ID NO: 14 or SEQ ID NO: 15.
  • the UCOE is in a reverse orientation or forward orientation. In some alternatives, the UCOE is in a forward orientation.
  • the vector is a lentiviral-based vector of a B lineage specific lentiviral vector.
  • the cells are B cells. In some alternatives, the cells are myeloid cells. In some alternatives, the cells are hematopoietic stem cells. In some alternatives, the cells are CD34+ hematopoietic stem cells.
  • the one or more enhancer elements comprises the sequences set forth in SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19 and/or SEQ ID NO: 20.
  • the polynucleotide further comprises a gene upstream of a BTK promoter.
  • the gene upstream of a BTK promoter is a BTK enhancer.
  • the BTK enhancer comprises the sequence set forth in SEQ ID NO: 21 or SEQ ID NO: 22.
  • the vector is a lentiviral-based vector of a B lineage specific lentiviral vector.
  • the cells are B cells. In some alternatives, the cells are myeloid cells.
  • the cells are hematopoietic stem cells. In some alternatives, the cells are CD34+ hematopoietic stem cells. In some alternatives, the cell is from the subject, wherein the cell is genetically modified by introducing the polynucleotide of any one of the alternatives herein or the vector of any one of the alternatives herein into the cell. In some alternatives, the administering is performed by adoptive cell transfer. In some alternatives, the cell is a B cell. In some alternatives, the cells are myeloid cells. In some alternatives, the cell is a hematopoietic stem cell. In some alternatives, the cell is a CD34+ hematopoietic stem cell. In some alternatives, the subject is male.
  • the subject is selected to receive immunoglobulin replacement therapy. In some alternatives, the subject is selected to receive targeted anti-microbial agents.
  • the polypeptide or vector comprises a sequence set forth in SEQ ID NO's: 33, 34, 35, 41, 42, 43, 44 or 45. In some alternatives, 0.7UCOE comprises a sequence set forth in SEQ ID NO: 2. In some alternatives, the enhancer comprises a sequence set forth in SEQ ID NO: 3 or 4.
  • nucleic acid comprising the promoter with GFP sequence is below

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Immunology (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Medicinal Chemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Biochemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Virology (AREA)
  • Molecular Biology (AREA)
  • Epidemiology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Mycology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Hematology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
PCT/US2018/028331 2017-04-21 2018-04-19 Optimized lentiviral vector for xla gene therapy WO2018195297A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
JP2019557373A JP7471821B2 (ja) 2017-04-21 2018-04-19 Xla遺伝子療法用の最適化されたレンチウイルスベクター
US16/605,740 US20200325458A1 (en) 2017-04-21 2018-04-19 Optimized lentiviral vector for xla gene therapy
CN201880043435.0A CN110809627A (zh) 2017-04-21 2018-04-19 用于xla基因疗法的优化的慢病毒载体
EP18788504.1A EP3612238A4 (en) 2017-04-21 2018-04-19 OPTIMIZED LENTIVIRAL VECTOR FOR XLA GENE THERAPY
AU2018256412A AU2018256412A1 (en) 2017-04-21 2018-04-19 Optimized lentiviral vector for XLA gene therapy
JP2024063151A JP2024084849A (ja) 2017-04-21 2024-04-10 Xla遺伝子療法用の最適化されたレンチウイルスベクター

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762488523P 2017-04-21 2017-04-21
US62/488,523 2017-04-21

Publications (1)

Publication Number Publication Date
WO2018195297A1 true WO2018195297A1 (en) 2018-10-25

Family

ID=63856377

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/028331 WO2018195297A1 (en) 2017-04-21 2018-04-19 Optimized lentiviral vector for xla gene therapy

Country Status (6)

Country Link
US (1) US20200325458A1 (enIt.pdf)
EP (1) EP3612238A4 (enIt.pdf)
JP (2) JP7471821B2 (enIt.pdf)
CN (1) CN110809627A (enIt.pdf)
AU (1) AU2018256412A1 (enIt.pdf)
WO (1) WO2018195297A1 (enIt.pdf)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020041647A1 (en) 2018-08-24 2020-02-27 Calimmune, Inc. Vector production in serum free media
WO2022123559A1 (en) * 2020-12-07 2022-06-16 Noga Therapeutics Ltd. Lentiviral vectors for therapeutic expression of btk in hematopoietic cells

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6974667B2 (en) * 2000-06-14 2005-12-13 Gene Logic, Inc. Gene expression profiles in liver cancer
US20160004814A1 (en) * 2012-09-05 2016-01-07 University Of Washington Through Its Center For Commercialization Methods and compositions related to regulation of nucleic acids

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
KERNS, H. ET AL.: "B Cell -Specific Lentiviral Gene Therapy Leads To Sustained B- Cell Functional 1 Recovery In A Murine Model Of X-Linked Agammaglobulinemia", BLOOD, vol. 115, 21 January 2010 (2010-01-21), pages 2146 - 2155, XP055541268 *
KERNS, H. ET AL.: "Lentiviral Vectors Using A Ubiquitously Acting Chromatin Opening Element 1 And Endogenous Btk Promoter Restore B And Myeloid Cell Defects In Murine X-Linked : Agammaglobulinemia", MOLECULAR THERAPY, vol. 19, no. 1, May 2011 (2011-05-01), pages S136, XP055541263 *
See also references of EP3612238A4 *
SINGH, S. ET AL.: "BTK-Promoter LV Vectors Utilizing Conserved Intron Element Mediate 1 Functional Rescue In Murine XLA", MOLECULAR THERAPY, vol. 23, no. 1, May 2015 (2015-05-01), pages S93, XP055541252 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020041647A1 (en) 2018-08-24 2020-02-27 Calimmune, Inc. Vector production in serum free media
WO2022123559A1 (en) * 2020-12-07 2022-06-16 Noga Therapeutics Ltd. Lentiviral vectors for therapeutic expression of btk in hematopoietic cells
US11464872B2 (en) 2020-12-07 2022-10-11 Noga Therapeutics Ltd. Lentiviral vectors for therapeutic expression of BTK in haematopoietic cells

Also Published As

Publication number Publication date
CN110809627A (zh) 2020-02-18
EP3612238A1 (en) 2020-02-26
US20200325458A1 (en) 2020-10-15
JP2024084849A (ja) 2024-06-25
AU2018256412A1 (en) 2019-11-07
JP2020517268A (ja) 2020-06-18
JP7471821B2 (ja) 2024-04-22
EP3612238A4 (en) 2020-12-30

Similar Documents

Publication Publication Date Title
Lee et al. Transgenic expression of the activating natural killer receptor Ly49H confers resistance to cytomegalovirus in genetically susceptible mice
Banks et al. Lymphotoxin-alpha-deficient mice. Effects on secondary lymphoid organ development and humoral immune responsiveness.
KR20200097760A (ko) 유전자 편집을 위한 cpf1-관련 방법 및 조성물
JPH07503848A (ja) 遺伝子標的現象による同型遺伝子接合
JP2024084849A (ja) Xla遺伝子療法用の最適化されたレンチウイルスベクター
WO2015183885A1 (en) Methods and compositions for perturbing gene expression in hematopoietic stem cell lineages in vivo
KR20220038362A (ko) 재조합 ad35 벡터 및 관련 유전자 요법 개선
CA3128216A1 (en) Gene-regulating compositions and methods for improved immunotherapy
CN113429472A (zh) Cd94和nkg2a基因人源化的非人动物及其制备方法和应用
KR20210120019A (ko) 세포-특이적 전사 조절 서열 및 이의 용도
TWI817994B (zh) 基因修飾細胞及其製備方法
JP7404237B2 (ja) ヒトインターロイキン15を発現する免疫不全マウス
US20130189781A1 (en) Loss of de novo dna methyltransferases promotes expansion of normal hematopoietic stem cells
CN108070614B (zh) 人源化基因改造动物模型的制备方法及应用
CA2451608C (en) Process for constructing a mutant utilizing the igh 3' enhancer
CN117795065A (zh) 使用细胞穿透crispr-cas系统在原代免疫细胞中进行基因编辑
CN109280674B (zh) 一种筛选抗体的非人模式动物的构建方法及其应用
WO2023041035A1 (en) Genetically modified non-human animal with human or chimeric genes
CN113817770B (zh) Cd73基因人源化的非人动物的构建方法及应用
US20240009236A1 (en) Modified nk cells with reduced ccr5 expression and methods of their use
EP4132966A1 (en) A method of generating a library of populations of cells comprising a transgene integrated into the genome at a target locus
Rieß The role of Roquin proteins in B cell physiology and pathology
JP2023525969A (ja) インビトロおよびインビボでのcd3+細胞における遺伝子治療産物の発現のためのプロモータ配列
Tsai The molecular programs controlling follicular helper T cell differentiation and function
Wilhelm The genomic organisation and transcriptional regulation of natural killer receptor genes

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 18788504

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2019557373

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2018256412

Country of ref document: AU

Date of ref document: 20180419

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2018788504

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2018788504

Country of ref document: EP

Effective date: 20191121