WO2014085593A1 - Targeting bcl11a distal regulatory elements for fetal hemoglobin reinduction - Google Patents

Targeting bcl11a distal regulatory elements for fetal hemoglobin reinduction Download PDF

Info

Publication number
WO2014085593A1
WO2014085593A1 PCT/US2013/072236 US2013072236W WO2014085593A1 WO 2014085593 A1 WO2014085593 A1 WO 2014085593A1 US 2013072236 W US2013072236 W US 2013072236W WO 2014085593 A1 WO2014085593 A1 WO 2014085593A1
Authority
WO
WIPO (PCT)
Prior art keywords
cell
cells
dna
mammal
expression
Prior art date
Application number
PCT/US2013/072236
Other languages
French (fr)
Inventor
Stuart H. Orkin
Daniel E. BAUER
Jian Xu
Original Assignee
Children's Medical Center Corporation
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
Priority to AU2013352156A priority Critical patent/AU2013352156B2/en
Priority to KR1020157016749A priority patent/KR102240555B1/en
Application filed by Children's Medical Center Corporation filed Critical Children's Medical Center Corporation
Priority to US14/647,547 priority patent/US9822355B2/en
Priority to ES13858006T priority patent/ES2708948T3/en
Priority to BR112015011995-6A priority patent/BR112015011995B1/en
Priority to SG11201504038XA priority patent/SG11201504038XA/en
Priority to CN201810979764.8A priority patent/CN109554350B/en
Priority to CN201380071452.2A priority patent/CN104955943B/en
Priority to DK13858006.3T priority patent/DK2925864T3/en
Priority to MX2015006528A priority patent/MX361412B/en
Priority to CA2892860A priority patent/CA2892860C/en
Priority to EP13858006.3A priority patent/EP2925864B1/en
Priority to JP2015544205A priority patent/JP6542124B2/en
Priority to EP18203436.3A priority patent/EP3502240B1/en
Publication of WO2014085593A1 publication Critical patent/WO2014085593A1/en
Priority to IL238949A priority patent/IL238949B/en
Priority to SA515360489A priority patent/SA515360489B1/en
Priority to ZA2015/04004A priority patent/ZA201504004B/en
Priority to HK16103646.4A priority patent/HK1215720A1/en
Priority to US15/816,083 priority patent/US10472619B2/en
Priority to AU2018278850A priority patent/AU2018278850B2/en
Priority to US16/573,389 priority patent/US11542493B2/en
Priority to AU2021218012A priority patent/AU2021218012B2/en
Priority to US18/074,107 priority patent/US20230348887A1/en

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
    • 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
    • C12N15/01Preparation of mutants without inserting foreign genetic material therein; Screening processes therefor
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/465Hydrolases (3) acting on ester bonds (3.1), e.g. lipases, ribonucleases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/795Porphyrin- or corrin-ring-containing peptides
    • C07K14/805Haemoglobins; Myoglobins
    • 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
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/102Mutagenizing nucleic acids
    • 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
    • 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
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases RNAses, DNAses

Abstract

Provided herein are methods and compositions for increasing fetal hemoglobin levels in a cell by disrupting BCL11A expression at the genomic level. Also provided herein are methods and compositions relating to the treatment of hemoglobinopathies by reinduction of fetal hemoglobin levels.

Description

TARGETING BCL11A DISTAL REGULATORY ELEMENTS FOR FETAL HEMOGLOBIN
REINDUCTION
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This application claims benefit under 35 U.S.C. § 119(e) of U.S. Provisional Application No. 61/730,323 filed November 27, 2012, of U.S. Provisional Application No. 61/730,369 filed
November 27, 2012, of U.S. Provisional Application No. 61/776,144 filed March 11, 2013, and of U.S. Provisional Application No. 61/889,174 filed October 10, 2013, the contents of each of which are incorporated herein by reference in their entireties.
GOVERNMENT SUPPORT
[0002] This invention was made with Government support under Grant No. 5RO1HL032259 awarded by the National Institutes of Health. The Government has certain rights in the invention.
BACKGROUND
[0003] Normal adult hemoglobin comprises four globin proteins, two of which are alpha (a) proteins and two of which are beta (β) proteins. During mammalian fetal development, particularly in humans, the fetus produces fetal hemoglobin, which comprises two gamma (y)-globin proteins instead of the two β-globin proteins. During the neonatal period, a globin switch occurs, referred to as the "fetal switch", at which point, erythroid precursors switch from making predominantly γ-globin to making predominantly β -globin. The developmental switch from production of predominantly fetal hemoglobin or HbF (α2γ2) to production of adult hemoglobin or HbA (α2β2) begins at about 28 to 34 weeks of gestation and continues shortly after birth until HbA becomes predominant. This switch results primarily from decreased transcription of the gamma-globin genes and increased transcription of beta-globin genes. On average, the blood of a normal adult contains less than 1% HbF, though residual HbF levels have a variance of over 20 fold in healthy adults and are genetically controlled.
[0004] Hemoglobinopathies encompass a number of anemias of genetic origin in which there is a decreased production and/or increased destruction (hemolysis) of red blood cells (RBCs). These also include genetic defects that result in the production of abnormal hemoglobins with a concomitant impaired ability to maintain oxygen concentration. Some such disorders involve the failure to produce normal β -globin in sufficient amounts, while others involve the failure to produce normal β -globin entirely. These disorders associated with the β -globin protein are referred to generally as β- hemoglobinopathies. For example, β -thalassemias result from a partial or complete defect in the expression of the β -globin gene, leading to deficient or absent HbA. Sickle cell anemia results from a point mutation in the β -globin structural gene, leading to the production of an abnormal (sickle) hemoglobin (HbS). HbS is prone to polymerization, particularly under deoxygenated conditions. HbS RBCs are more fragile than normal RBCs and undergo hemolysis more readily, leading eventually to anemia.
[0005] Recently, the search for treatment aimed at reduction of globin chain imbalance or predisposition to hemoglobin polymerization in patients with β -hemoglobinopathies has focused on the pharmacologic manipulation of fetal hemoglobin (α2γ2; HbF). The therapeutic potential of such approaches is suggested by observations of the mild phenotype of individuals with co-inheritance of both homozygous β-thalassemia and hereditary persistence of fetal hemoglobin (HPFH), as well as by those patients with homozygous β-thalassemia who synthesize no adult hemoglobin, but in whom a reduced requirement for transfusions is observed in the presence of increased concentrations of fetal hemoglobin. Furthermore, it has been observed that certain populations of adult patients with β chain abnormalities have higher than normal levels of fetal hemoglobin (HbF), and have been observed to have a milder clinical course of disease than patients with normal adult levels of HbF. For example, a group of Saudi Arabian sickle-cell anemia patients who express 20-30% HbF have only mild clinical manifestations of the disease. It is now accepted that hemoglobin disorders, such as sickle cell anemia and the β- thalassemias, are ameliorated by increased HbF production.
[0006] As mentioned earlier, the switch from fetal hemoglobin to adult hemoglobin (α2γ2; HbA) usually proceeds within six months after parturition. However, in the majority of patients with β- hemoglobinopathies, the upstream γ globin genes are intact and fully functional, so that if these genes become reactivated, functional hemoglobin synthesis could be maintained during adulthood, and thus ameliorate disease severity. Unfortunately, the in vivo molecular mechanisms underlying the globin switch are not well understood.
[0007] Evidence supporting the feasibility of reactivation of fetal hemoglobin production comes from experiments in which it was shown that peripheral blood, containing clonogenic cells, when given the appropriate combination of growth factors, produce erythroid colonies and bursts in semisolid culture. Individual cells in such colonies can accumulate fetal hemoglobin (HbF), adult hemoglobin (HbA) or a combination of both. In cultures from adult blood, nucleated red cells accumulate either HbA (F-A+) only, or a combination of HbF and HbA (F+A+). Importantly, individual colonies contain both F+ and F- cells, indicating that both types are progeny from the same circulating stem cells. Thus, during the early stages of development in culture, cells execute an option, through currently unknown mechanisms, whether or not to express HbF. The proportion of adult F+ cells developing in culture does not appear to be preprogrammed in vivo, but appears to depend on culture conditions: A shift into the combined HbF and HbA expression pathway can, for example, be achieved in vitro by high serum concentrations, due to the activity of an unidentified compound that can be absorbed on activated charcoal.
[0008] Overall, identification of molecules that play a role in the globin switch is important for the development of novel therapeutic strategies that interfere with adult hemoglobin and induce fetal hemoglobin synthesis. Such molecules would provide new targets for the development of therapeutic interventions for a variety of hemoglobinopathies in which reactivation of fetal hemoglobin synthesis would significantly ameliorate disease severity and morbidity.
SUMMARY
[0009] Provided herein are methods and compositions for increasing fetal β-globin levels in a cell by disrupting BCL11A expression at the genomic level. Also provided herein are methods and compositions relating to the treatment of hemoglobinopathies by reinduction of fetal β-globin levels.
[0010] One aspect described herein relates to a method for producing a progenitor cell having decreased BCL11A mRNA or protein expression, the method comprising contacting an isolated progenitor cell with an agent that binds the genomic DNA of the cell on chromosome 2 location
60,716,189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly), thereby reducing the mRNA or protein expression of BCL11A.
[0011] Another aspect described herein relates to a method for producing a genetic engineered human cell having at least one genetic modification comprising contacting an isolated cell with an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA-targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location 60,716, 189-60,728,612 causing at least one genetic modification therein.
[0012] Also provided herein in another aspect is an isolated genetic engineered human cell having at least one genetic modification on chromosome 2 location 60,716, 189-60,728,612. In one embodiment, the at least one genetic modification is a deletion in the genomic DNA at the specified location. In one embodiment, the isolated genetic engineered human cell has reduced or decreased mRNA or protein expression of BCL11A compared to a control cell that has no one genetic modification on chromosome 2 location 60,716, 189-60,728,612.
[0013] Another aspect described herein relates to a use of an isolated genetic engineered human cell having at least one genetic modification on chromosome 2 location 60,716,189-60,728,612 described herein for the purpose of increasing the fetal hemoglobin levels in a mammal.
[0014] Another aspect described herein relates to a use of an isolated genetic engineered human cell having at least one genetic modification on chromosome 2 location 60,716,189-60,728,612 described herein for the treatment a hemoglobinopathy in a mammal.
[0015] Another aspect described herein relates to a use of an isolated genetic engineered human cell having at least one genetic modification on chromosome 2 location 60,716,189-60,728,612 described herein for the manufacturer of medicament for the treatment a hemoglobinopathy in a mammal whereby the fetal hemoglobin levels in a mammal is increased. [0016] Another aspect described herein is a composition comprising isolated genetic engineered human cells having at least one genetic modification on chromosome 2 location 60,716,189-60,728,612. In one embodiment, the composition further comprises a pharmaceutically acceptable carrier.
[0017] Another aspect described herein relates to a use of a composition comprising isolated genetic engineered human cells having at least one genetic modification on chromosome 2 location 60,716,189-60,728,612 for the purpose of increasing the fetal hemoglobin levels in a mammal.
[0018] Another aspect described herein relates to a use of a composition comprising isolated genetic engineered human cells having at least one genetic modification on chromosome 2 location 60,716,189-60,728,612 for the treatment a hemoglobinopathy in a mammal.
[0019] Another aspect described herein relates to a use of a composition comprising isolated genetic engineered human cells having at least one genetic modification on chromosome 2 location 60,716,189-60,728,612 for the manufacturer of medicament for the treatment a hemoglobinopathy in a mammal whereby the fetal hemoglobin levels in a mammal is increased.
[0020] Another aspect described herein is a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA-targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of a human cell on chromosome 2 location 60,716,189-60,728,612 causing at least one genetic modification therein. In one embodiment, the composition further comprises a pharmaceutically acceptable carrier.
[0021] Another aspect described herein relates to a use of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA-targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of a human cell on chromosome 2 location 60,716, 189-60,728,612 causing at least one genetic modification therein for the purpose of increasing the fetal hemoglobin levels in a mammal.
[0022] Another aspect described herein relates to a use of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA-targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of a human cell on chromosome 2 location 60,716, 189-60,728,612 causing at least one genetic modification therein for the treatment a hemoglobinopathy in a mammal.
[0023] Another aspect described herein relates to a use of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA-targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of a human cell on chromosome 2 location 60,716, 189-60,728,612 causing at least one genetic modification therein for the manufacturer of medicament for the treatment a hemoglobinopathy in a mammal whereby the fetal hemoglobin levels in a mammal is increased. [0024] In one embodiment, provided herein is a use of an agent that binds the genomic DNA of the cell on chromosome 2 location 60,716, 189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly) for increasing the fetal hemoglobin in a mammal or for the treatment of a hemoglobinopathy in the mammal or for reducing the mRNA or expression of BCL1 lA, wherein the mRNA or protein expression of BCL11A is reduced.
[0025] In one embodiment, provided herein is a use of an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA- targeting endonuclease for increasing the fetal hemoglobin in a mammal or for the treatment of a hemoglobinopathy in the mammal or for reducing the mRNA or expression of BCL1 lA, wherein the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location
60,716,189-60,728,612 causing at least one genetic modification therein.
[0026] In one embodiment, provided herein is a use of an effective amount of a composition comprising at least a DNA-targeting enzyme or a vector carrying the coding sequence of a DNA- targeting enzyme for increasing the fetal hemoglobin in a mammal or for the treatment of a
hemoglobinopathy in the mammal or for reducing the mRNA or expression of BCL1 lA, wherein the DNA-targeting enzyme produces at least one epigenetic modification in the genomic DNA of the cell on chromosome 2, thereby affecting the mRNA or expression of BCL11A. In one embodiment, the at least one epigenetic modification is at location 60,716,189-60,728,612. In another embodiment, the effect of the one epigenetic modification is reducing the mRNA or protein expression of BCL11A. In one embodiment, the at least one epigenetic modification in the genomic DNA of the cell on chromosome 2 indirectly or directly affects the location 60,716,189-60,728,612 of chromosome 2.
[0027] In one embodiment, provided herein is a use of any isolated cells described herein for increasing the fetal hemoglobin in a mammal or for the treatment of a hemoglobinopathy in the mammal.
[0028] In one embodiment, provided herein is a use of a composition comprising isolated genetic engineered human cells for increasing the fetal hemoglobin in a mammal or for the treatment of a hemoglobinopathy in the mammal, wherein the cells have at least one genetic modification on chromosome 2 location 60,716, 189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly) made by the process of contacting the cells with an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA- targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location 60,716,189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly) causing at least one genetic modification therein.
[0029] In one embodiment, provided herein is a use of a composition comprising isolated genetic engineered human cells for increasing the fetal hemoglobin in a mammal or for the treatment of a hemoglobinopathy in the mammal, wherein the cells have at least one epigenetic modification on chromosome 2. In one embodiment, the at least one epigenetic modification on chromosome 2 is at location 60,716, 189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly). In another embodiment, at least one epigenetic modification on chromosome 2 is made by the process of contacting the cells with an effective amount of a composition comprising at least a DNA-targeting enzyme or a vector carrying the coding sequence of a DNA-targeting enzyme whereby the DNA- targeting enzyme produces at least one epigenetic modification in the genomic DNA of the cell on chromosome 2 which affects the location 60,716,189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly) causing therein.
[0030] In one embodiment, provided herein is a use of any isolated cells described herein or any one of the compositions described herein for the manufacture of a medicament for increasing the fetal hemoglobin in a mammal in need thereof or for the treatment of a hemoglobinopathy in a mammal.
[0031] Another aspect described herein is a method of increasing fetal hemoglobin levels in a cell, the method comprising the steps of: contacting an isolated cell with an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA- targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location 60,716,189-60,728,612 causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in said cell, or its progeny, relative to the cell prior to the contacting.
[0032] Another aspect described herein is a method for increasing fetal hemoglobin levels in a mammal in need thereof, the method comprising the steps of contacting an isolated hematopoietic progenitor cell in said mammal with an effective amount of a composition comprising at least a DNA- targeting endonuclease or a vector carrying the coding sequence of a DNA-targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location 60,716, 189-60,728,612 causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in said mammal, relative to expression prior to said contacting.
[0033] Another aspect described herein is a method for increasing fetal hemoglobin levels in a mammal in need thereof, the method comprising transplanting an isolated genetic engineered human cell having at least one genetic modification on chromosome 2 location 60,716, 189-60,728,612 into the mammal.
[0034] In one embodiment, this disclosure provides a method for increasing fetal hemoglobin levels in a mammal in need thereof, the method comprising the steps of providing an isolated population of hematopoietic progenitor cells or hematopoietic stem cells from the mammal in ex vivo, and contacting the population of hematopoietic progenitor or stem cells with an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA- targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location 60,716,189-60,728,612 causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in the mammal, relative to expression prior to the contacting.
[0035] In one embodiment, this disclosure provides a method for increasing fetal hemoglobin levels in a mammal in need thereof, the method comprising the steps of isolating a population of hematopoietic progenitor cells or hematopoietic stem cells from the mammal, and contacting in ex vivo the population of hematopoietic progenitor or stem cells with an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA- targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location 60,716,189-60,728,612 causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in the mammal, relative to expression prior to the contacting.
[0036] In one embodiment, this disclosure provides a method for increasing fetal hemoglobin levels in a mammal in need thereof, the method comprising the steps of providing isolating a population of hematopoietic progenitor cells or hematopoietic stem cells from the mammal and deleting the genomic DNA of the cells on chromosome 2 location 60,716, 189-60,728,612 causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in the mammal, relative to expression prior to the contacting.
[0037] In one embodiment, this disclosure provides a method for increasing fetal hemoglobin levels in a mammal in need thereof, the method comprising the steps of isolating a population of hematopoietic progenitor cells or hematopoietic stem cells from the mammal and ex vivo deleting the genomic DNA of the cells on chromosome 2 location 60,716, 189-60,728,612 causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in the mammal, relative to expression prior to the contacting.
[0038] In one embodiment, this disclosure provides a method of treatment of a hemoglobinopathy in a mammal comprising the steps of:(a) providing hematopoietic progenitor cells or hematopoietic stem cells or iPSCs; (b) contacting the cells ex vivo or in vitro with an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA- targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location 60,716,189-60,728,612 causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in the mammal, relative to expression prior to the contacting; and (c) administering the of step (b) into the mammal.
[0039] In one embodiment, this disclosure provides a method of treatment of a hemoglobinopathy in a mammal comprising the steps of:(a) isolating hematopoietic progenitor cells or hematopoietic stem cells from the mammal; (b) contacting the cells ex vivo or in vitro with an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA-targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location 60,716,189-60,728,612 causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in the mammal, relative to expression prior to the contacting; and (c) administering the of step (b) into the mammal.
[0040] In one embodiment, this disclosure provides a method of treatment of a hemoglobinopathy in a mammal comprising the steps of: (a) providing hematopoietic progenitor cells or hematopoietic stem cells or iPSCs; (b) ex vivo deleting the genomic DNA of the cells on chromosome 2 location 60,716, 189- 60,728,612 causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in the mammal, relative to expression prior to the contacting; and (c) administering the cells of step (b) into the mammal.
[0041] In one embodiment, this disclosure provides a method of treatment of a hemoglobinopathy in a mammal comprising the steps of:(a) isolating hematopoietic progenitor cells or hematopoietic stem cells from the mammal; (b) ex vivo deleting the genomic DNA of the cells on chromosome 2 location 60,716,189-60,728,612 causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in the mammal, relative to expression prior to the contacting; and (c) administering the of step (b) into the mammal.
[0042] In one embodiment, this disclosure provides a method of treatment of a hemoglobinopathy in a mammal (e.g. a human) comprising introducing a composition described herein comprising isolated genetic engineered cells having at least one genetic modification on chromosome 2 location 60,716,189- 60,728,612 whereby fetal hemoglobin expression is increased in the mammal.
[0043] In one embodiment, this disclosure provides a method of treatment of a hemoglobinopathy in a mammal (e.g. a human) comprising increasing fetal hemoglobin expression in the mammal by method described herein.
[0044] In one embodiment of this aspect and all other aspects described herein, the isolated cell or isolated population of cells is/are human cell(s).
[0045] In one embodiment of this aspect and all other aspects described herein, the isolated cell or isolated population of cells is/are progenitor cell(s).
[0046] In one embodiment of this aspect and all other aspects described herein, the human cell is a hematopoietic progenitor cell.
[0047] In one embodiment of this aspect and all other aspects described herein, the human cell is an induced pluripotent stem cell.
[0048] In one embodiment of this aspect and all other aspects described herein, the induced pluripotent stem cell is hematopoietic progenitor cell.
[0049] In one embodiment of this aspect and all other aspects described herein, the hematopoietic progenitor is a cell of the erythroid lineage. [0050] In one embodiment of this aspect and all other aspects described herein, the hematopoietic progenitor cell or isolated is contacted ex vivo or in vitro or in vivo.
[0051] In one embodiment of this aspect and all other aspects described herein, the at least one genetic modification is a deletion.
[0052] In one embodiment of this aspect and all other aspects described herein, the deletion removes the entire region between chromosome 2 location 60,716,189-60,728,612 or removes a portion of the region resulting in disruption of one of more DNAse 1 -hypersensitive sites (DHS).
[0053] In another embodiment of this aspect and all other aspects described herein, the deletion comprises one or more of the DNAse 1 -hypersensitive sites (DHS) +62, +58, and +55 as described herein in the Examples section.
[0054] In another embodiment of this aspect and all other aspects described herein, the deletion comprises one or more of the SNP markers described in Table 2.
[0055] In another embodiment of this aspect and all other aspects described herein, the deletion comprises one or more of the fragments listed in Table 7.
[0056] In another embodiment of this aspect and all other aspects described herein, the deletion removes the entire region between chromosome 2 location 60,716,189-60,728,612 or removes a portion of the region resulting in disruption of one of more DNAse 1 -hypersensitive sites (DHS). In one embodiment, as used herein, the term "portion" in the context of genomic deletion is at least 20%-80% of the specified region.
[0057] In further embodiment of any treatment method, the method comprises chemotherapy and/or radiation therapy to remove or reduced the endogenous hematopoietic progenitor or stem cells in the mammal.
[0058] In one embodiment of any method, the contacted cells having at least one genetic modification can be cryopreserved and stored until the cells are needed for administration into a mammal.
[0059] In one embodiment of any described method, the hematopoietic progenitor or stem cells or isolated cells can be substituted with an iPSCs described herein.
[0060] In one embodiment of any described method, the hematopoietic progenitor or stem cells or iPSCs or isolated cells are autologous to the mammal, meaning the cells are derived from the same mammal. In another of the embodiments of the described method, the hematopoietic progenitor or stem cells or iPSCs or isolated cells are non-autologous to the mammal, meaning the cells are not derived from the same mammal, but another mammal of the same species. For example, the mammal is a human.
[0061] In one embodiment of any treatment method, the method further comprises selecting a mammal in need of increased fetal hemoglobin expression. [0062] In one embodiment of any treatment method, the method further comprises selecting a mammal in need of treatment of a hemoglobinopathy.
[0063] In any embodiment of any treatment method described, the hemoglobinopathy is a β- hemoglobinopathy.
[0064] In any embodiment of any treatment method described, the hemoglobinopathy is β- thalassemia.
[0065] In any embodiment of any treatment method described, the hemoglobinopathy is sickle cell anemia.
BRIEF DESCRIPTION OF DRAWINGS
[0066] FIG. 1A shows the distribution of 636 SNPs previously published to be associated with erythroid traits at P < 5 x 10"8 with respect to promoter, exonic, intronic, 3'UTR, and intergenic sequences. For comparison, genomic distribution of these regions is displayed.
[0067] FIG. IB is a graph plotting the cumulative distribution of the distances of the 636 erythroid trait-associated SNPs with respect to nearest erythroid enhancer. Erythroid enhancers were defined by sequences more than 2kb from the TSSs of Refseq-annotated genes with DNase I hypersensitivity, presence of H3K4mel, either H3K27ac or H3K9ac, and absence of H3K4me3 and H3K27me3. The distance of mean ± SD of 50 sets of 636 randomly permuted non-erythroid trait-associated SNPs (from GWAS database), SNPs from the Affy 6.0 genotyping array, or random SNPs were also plotted.
[0068] FIG. 2A shows ChlP followed by massively parallel sequencing performed from CD34+- cell-derived erythroid precursors with antibodies to H3K27me3, H3K4me3, H3K4mel, H3K27ac, GATA1, TALI, and PolII. Nuclei isolated from erythroid precursors, fetal brain, and B- and T- lymphocytes subject to DNase I treatment with sites of cleavage determined by massively parallel sequencing. HbF-associated SNPs includes those associated with HbF level or F-cell number at P < 5 x 10~8 and sentinel SNPs those with highest association to HbF or F-cell number in a given GWAS. Three adjacent erythroid DHS are labeled as +62, +58, and +55 based on distance in kb from BCLllA TSS.
[0069] FIG. 2B shows ChlP-qPCR of primary human erythroid precursors at BCLllA intron-2, normalized to 1% input chromatin. DHS +62, +58, and +55 from Figure 2A shaded. Enrichment at negative control (GAPDH, OCT4) and positive control (β-globin LCR HS3 and ct-globin HS-40) loci displayed for comparison.
[0070] FIG. 2C shows chromosome conformation capture performed in primary human erythroid precursors across BCLllA locus using BCLllA promoter as anchor. Interaction frequency is normalized to LCR-HBB interaction.
[0071] FIG. 3A shows Healthy anonymous donors from whom hematopoietic stem/progenitor cells were available were genotyped at rsl427407 to identify heterozygous individuals. Five donors were identified. The hematopoietic stem/progenitor cells were subject to erythroid differentiation culture. Chromatin was isolated from erythroblasts, and immunoprecipitated by GATA1 or TALI. ChIP DNA or input DNA was subject to a pyrosequencing reaction to quantify the relative abundance of the rs 1427407 G-allele.
[0072] FIG. 3B shows data from Healthy anonymous donors from whom hematopoietic stem/progenitor cells were available were genotyped at rsl427407, rs7606173, and rs7569946 to identify individuals heterozygous for the rsl427407-rs7606173 haplotype as well as rs7569946. Three donors were identified. Haplotyping revealed that the rs7569946 G-allele was on the same chromosome as the rsl427407 G-allele and rs7606173 C-allele in each. The hematopoietic stem/progenitor cells were subject to erythroid differentiation culture. RNA and genomic DNA were isolated, and cDNA was produced by reverse transcription. Paired gDNA and cDNA samples were subject to a pyrosequencing reaction to quantify the relative abundance of the 7569946 G-allele.
[0073] FIG.3C shows mean HbF for rsl427407-rs7606173 haplotypes in the CSSCD cohort. The mean HbF level was 4.05% (SD 3.10) in 213 rsl427407-rs7606173 G-C individuals, 7.08% (SD 4.50) in 254 rsl427407-rs7606173 T-G/G-T heterozygotes, and 11.21% (SD 4.37) in 60 rsl427407-rs7606173 T-G individuals. The P-values correspond to one-tailed student t-tests. The haplotype frequencies in CSSCD are: TG : 24.5%, TC 0.085%, GC 42.3%, GG 33.1%.
[0074] FIGs. 4A-4C show data from a 12.4-kb fragment of BCL11A intron-2 encompassing the DHSs +62, +58 and +55 (encompassing +52.0-64.4 kb from TSS) cloned upstream of an Hsp68 minimal promoter and lacZ reporter gene flanked by H19 insulator elements. Transient transgenic murine embryos generated by nuclear injection at the one-cell stage.
[0075] FIG. 4A shows E12.5 transient transgenic embryo stained with X-gal.
[0076] FIG. 4B shows cell suspensions isolated from the peripheral blood and fetal liver of stable transgenic E12.5 embryos. Cytospins were stained with X-gal and counterstained with Nuclear Red.
[0077] FIG. 4C shows data from bone marrow erythroblasts (CD71+/Terl 19+) and splenic lymphocytes (CD 19+ for B-lymphocytes and CD3+ for T-lymphocytes) that were isolated and sorted from young adult stable transgenics. Cells were subject to X-gal staining or RNA isolation followed by RT-qPCR. Gene expression normalized to GAPDH, and expressed relative to T-lymphocytes, which express neither BCL11A nor lacZ.
[0078] FIGs. 5A-5C show mouse erythroleukemia (MEL) cells and pro-B lymphoid cells transfected with two pairs of TALENs each designed to generate a DSB on either end of the orthologous 10 kb BCL11A intron-2 erythroid enhancer from +50.4-+60.4 kb. Clones (called Δ50.4-60.4) were isolated with biallelic deletion of the 10-kb segment.
[0079] FIG. 5 A shows RT-qPCR performed of Bell la with primer pairs recognizing sequences upstream, spanning, and downstream of intron-2. [0080] FIG. 5B shows an immunoblot of Δ50.4-60.4 with anti-BCL 11 A.
[0081] FIG. 5C shows globin gene expression in Δ50.4-60.4 MEL clones. A common primer pair recognizes the adult β-globins β2 and β ΐ, while independent primers recognize the embryonic β-globins sy and βΗΙ.
[0082] FIG. 6 shows mouse zygote pronuclei injected with lacZ reporter construct. Transgenic embryos were isolated at E12.5. Embryos were genotyped by lacZ PCR. The fraction of transgenic embryos with X-gal staining of fetal liver is reported.
[0083] FIG. 7 shows one to two kb sequence fragments cloned into enhancer construct with TK minimal promoter and GFP. Enhancer reporter constructs were delivered by lentiviral vectors to primary human erythroid precursors. Transfected cells were selected by puromycin resistance. Mean GFP fluorescence intensity was measured.
[0084] FIG. 8 shows data relating to chromatin profiling of mouse erythroid cells that reveals an orthologous enhancer signature at Bell la intron-2. Mouse tracks obtained from previously published global mouse erythroid chromatin profiling, with histone modifications and DNase-I cleavage from and GATA1 and TALI ChlP-seq from. Dotted rectangle bounds orthologous enhancer signature defining Δ50.4-60.4 element targeted for TALEN-mediated deletion.
[0085] FIG. 9A is a schematic of a TALEN-mediated genome engineering strategy used herein. TALENs are sequence-specific nucleases. Two pairs of TALENs were engineered to generate double strand breaks, one at Bell la +50.4 and the other at +60.4. Clones were isolated that had repaired the two DSBs by NHEJ with excision of the intervening 10-kb segment. Clones were screened by PCR with primers 5', 3', internal and spanning the 10-kb deletion.
[0086] FIG. 9B shows Southern blotting of Hindlll digested genomic DNA from Δ50.4-60.4 clones corroborated that these clones had expected excision allele and lacked a non-excised allele.
[0087] FIG. 10 shows Sanger sequencing of PCR products from Δ50.4-60.4 clones. 5' (+50.4) and 3' (+60.4) left and right TALEN recognition sequences with intervening spacers is shown. Some alleles showed evidence of end-joining directly from each digested spacer sequence whereas other alleles showed loss of hundreds of additional nucleotides. Only one allele each was isolated from MEL clone #1 and pro-B clone #2.
[0088] FIG. l 1A shows the genotype data obtained in 1,178 individuals from CSSCD for 38 variants within BCL11A +62, +58 or +55 DHSs. Most highly significant associations to HbF level among common (MAF > 1%) SNPs (n = 10) prior to (rsl427407) or following (rs7606173) conditional analysis on rsl427407. SNP coordinates chromosome 2, buildingl9.
[0089] FIG. 1 IB the HbF association analyses at BCL11A. Genotype data obtained in 1,178 individuals from CSSCD for 38 variants within BCL11A +62, +58 or +55 DHSs. Sentinel SNPs are those with the highest association to HbF level or F-cell number in prior GWAS (7-12). These SNPs are shown with respect to BCL11A intron-2 with the 3 DHSs +62, +58 and +55 indicated.
DETAILED DESCRIPTION
[0090] The methods and compositions described herein relate, in part, to the discovery of a distal regulatory region upstream of the BCL11A gene that can regulate expression of the BCL11A protein. The BCL11A protein acts as a stage specific regulator of fetal hemoglobin expression by repressing γ- globin induction. Accordingly, the methods and compositions provided herein are novel methods for the regulation of γ-globin expression in eythroid cells. More specifically, these activities can be harnessed in methods for the treatment of β -hemoglobinopathies by induction of γ-globin via inhibition of the BCL11A gene product.
[0091] In one embodiment, provided herein is a method for producing an isolated progenitor cell having decreased BCL11A mRNA or protein expression, the method comprising contacting an isolated progenitor cell with an agent that binds the genomic DNA of the cell on chromosome 2 location 60,716,189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly), thereby reducing the mRNA or protein expression of BCL11A.
[0092] In one embodiment, provided herein is a method for producing an isolated progenitor cell having decreased BCL11A mRNA or protein expression, the method comprising providing an isolated progenitor cell and contacting the isolated progenitor cell with an agent that binds the genomic DNA of the cell on chromosome 2 location 60,716, 189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly), thereby reducing the mRNA or protein expression of BCL11A.
[0093] In one embodiment, provided herein is a method for producing an isolated progenitor cell having decreased BCL11A mRNA or protein expression, the method comprising contacting an isolated progenitor cell with an agent that binds produces an epigenetic modification in the genomic DNA of the cell on chromosome 2 thereby reducing the mRNA or protein expression of BCL11A. In one embodiment, the epigenetic modification in the genomic DNA is at chromosome 21ocation 60,716,189- 60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly).
[0094] In one embodiment, provided herein is a method for producing an isolated progenitor cell having decreased BCL11A mRNA or protein expression, the method comprising providing an isolated progenitor cell and contacting the isolated progenitor cell with an agent that produces an epigenetic modification in the genomic DNA of the cell on chromosome 2 thereby reducing the mRNA or protein expression of BCL11A. In one embodiment, the epigenetic modification in the genomic DNA is at chromosome 21ocation 60,716,189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly).
[0095] The disclosure described herein, in a preferred embodiment, does not concern a process for cloning human beings, processes for modifying the germ line genetic identity of human beings, uses of human embryos for industrial or commercial purposes or processes for modifying the genetic identity of animals which are likely to cause them suffering without any substantial medical benefit to man or animal, and also animals resulting from such processes.
[0096] One aspect described herein relates to a method for producing an isolated genetic engineered human cell having at least one genetic modification comprising contacting the cell with an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA-targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location 60,716, 189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly) causing at least one genetic modification therein.
[0097] Another aspect provided herein relates to a method of increasing fetal hemoglobin levels in an isolated cell, the method comprising decreasing the BCL11A mRNA or protein expression in the cell. In one aspect, the decrease of BCL11A mRNA or protein expression is achieved by causing at least one genetic modification at the genomic DNA of the cell on chromosome 2 location 60,716,189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly). In another aspect, the decrease of BCL11A mRNA or protein expression is achieved by causing at least one genetic modification at the genomic DNA of the cell on chromosome 2 location 60,716, 189-60,728,612 that results in epigenetic modification of the genetic function at chromosome 2 location 60,716, 189-60,728,612. In this aspect, the BCL11A enhancer activity located within this chromosome 2 location 60,716,189-60,728,612 is reduce. By decrease in this aspect, the enhancer activity in enhancing BCL11A mRNA or protein expression in the cell is at least 5% lower is at least 10% lower, at least 20% lower, at least 30% lower, at least 40% lower, at least 50% lower, at least 60% lower, at least 70% lower, at least 80% lower, at least 90% lower, at least 1-fold lower, at least 2-fold lower, at least 5 -fold lower, at least 10 fold lower, at least 100 fold lower, at least 1000-fold lower, or more compared to a control cell that is not treated in any method disclosed herein. By decrease of the BCL11A mRNA or protein expression in the cell means that protein expression is at least 5% lower is at least 10% lower, at least 20% lower, at least 30% lower, at least 40% lower, at least 50% lower, at least 60% lower, at least 70% lower, at least 80% lower, at least 90% lower, at least 1-fold lower, at least 2-fold lower, at least 5 -fold lower, at least 10 fold lower, at least 100 fold lower, at least 1000-fold lower, or more compared to a control cell that is not treated in any method disclosed herein.
[0098] Another aspect provided herein relates to a method of increasing fetal hemoglobin levels in an isolated cell, the method comprising providing an isolated human cell or progenitor cell and decreasing the BCL11A mRNA or protein expression in the cell.
[0099] Another aspect provided herein relates to a method of increasing fetal hemoglobin levels in a cell, the method comprising the steps of: contacting an isolated human cell with an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA-targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location 60,716, 189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly) causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in the cell, or its progeny, relative to the cell prior to the contacting.
[0100] Another aspect provided herein relates to a method of increasing fetal hemoglobin levels in a cell, the method comprising the steps of providing an isolated human cell or progenitor cell, contacting an isolated human cell or progenitor cell with an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA-targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location 60,716, 189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly) causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in the cell, or its progeny, relative to the cell prior to the contacting.
[0101] Another aspect described herein relates to a method for increasing fetal hemoglobin levels in a mammal in need thereof, the method comprising decreasing the BCL11A mRNA or protein expression in a hematopoietic progenitor cell in the mammal. In one aspect, the decrease of BCL11A mRNA or protein expression is achieved by causing at least one genetic modification at the genomic DNA of the cell on chromosome 2 location 60,716,189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly). In another aspect, the decrease of BCL11A mRNA or protein expression is achieved by causing at least one epigenetic modification at the genomic DNA of the cell on chromosome 2. In another aspect, the decrease of BCL11A mRNA or protein expression is achieved by causing at least one epigenetic modification at the genomic DNA of the cell on chromosome 2 location 60,716,189-60,728,612.
[0102] Another aspect described herein relates to a method for increasing fetal hemoglobin levels in a mammal in need thereof, the method comprising providing an isolated human cell or progenitor cell from a mammal and decreasing the BCL11A mRNA or protein expression in the cell. In one aspect, the method further comprises selecting a mammal in need of increasing fetal hemoglobin levels therein.
[0103] Another aspect described herein relates to a method for increasing fetal hemoglobin levels in a mammal in need thereof, the method comprising the steps of contacting a hematopoietic progenitor cell in the mammal with an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA-targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location
60,716,189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly) causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in the mammal, relative to expression prior to the contacting.
[0104] Another aspect described herein relates to a method for increasing fetal hemoglobin levels in a mammal in need thereof, the method comprising the steps of providing an isolated human cell or progenitor cell or an isolated population of hematopoietic progenitor cells from a mammal contacting the human cell or progenitor cell or hematopoietic progenitor cell with an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA- targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location 60,716,189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly) causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in said mammal, relative to expression prior to the contacting.
[0105] Another aspect provided herein relates to a method for increasing fetal hemoglobin levels in a mammal in need thereof, the method comprising transplanting a genetic engineered human cell as described herein into the mammal.
[0106] In one embodiment of this aspect and all other aspects described herein, the method further comprises providing an isolated cell or an isolated progenitor cell or an isolated population of cells which can be progenitor cell or hematopoietic progenitor cell.
[0107] In one embodiment of this aspect and all other aspects described herein, the isolated cell is an isolated progenitor cell.
[0108] In one embodiment of this aspect and all other aspects described herein, the isolated progenitor cell is an isolated human cell.
[0109] In one embodiment of this aspect and all other aspects described herein, the isolated human cell is a hematopoietic progenitor cell.
[0110] In another embodiment of this aspect and all other aspects described herein, the
hematopoietic cell is a cell of the erythroid lineage. Methods of isolating hematopoietic progenitor cell are well known in the art, e.g., by flow cytometric purification of CD34+ or CD133+ cells, microbeads conjugated with antibodies against CD34 or CD133, markers of hematopoietic progenitor cell.
Commercial kits are also available, e.g., MACS® Technology CD34 MicroBead Kit, human, and CD34 MultiSort Kit, human, and STEMCELL™ Technology EasySep™ Mouse Hematopoietic Progenitor Cell Enrichment Kit.
[0111] In another embodiment of this aspect and all other aspects described herein, the human cell is an induced pluripotent stem cell (iPSC).
[0112] In another embodiment of this aspect and all other aspects described herein, the contacting of any cell described herein can be ex vivo or in vitro or in vivo.
[0113] In another embodiment of this aspect and all other aspects described herein, the contacting of any cell described herein comprises contacting with an agent that binds the genomic DNA of the cell on chromosome 2 and produces an epigenetic modification in the genome of the cell on chromosome 2, thereby reducing the mRNA or protein expression of BCL11A. In one embodiment, the epigenetic modification is on chromosome 2 location 60,716, 189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly).
[0114] In one embodiment of this aspect and all other aspects described herein, the at least one epigenetic modification in the genomic DNA of the cell on chromosome 2 indirectly or directly affects the location 60,716,189-60,728,612 of chromosome 2.
[0115] As used herein, "indirectly affecting the location 60,716, 189-60,728,612 of chromosome 2" refers to long distance effects of epigenetic modification in the genomic DNA of the cell on chromosome 2 the location 60,716, 189-60,728,612 of chromosome 2.
[0116] In another embodiment of this aspect and all other aspects described herein, the contacting of any cell described herein comprises contact with an agent that binds the genomic DNA of the cell on chromosome 2 location 60,716, 189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly), and produces an epigenetic modification on chromosome 2, thereby reducing the mRNA or protein expression of BCL11A.
[0117] In another embodiment of this aspect and all other aspects described herein, the contacting of any cell described herein comprises contact with an effective amount of a composition comprising at least a DNA-targeting enzyme or a vector carrying the coding sequence of a DNA-targeting enzyme whereby the DNA-targeting enzyme produces an epigenetic modification on chromosome 2, thereby reducing the mRNA or protein expression of BCL11A.
[0118] In another embodiment of this aspect and all other aspects described herein, the contacting of any cell described herein comprises contact with an effective amount of a composition comprising at least a DNA-targeting enzyme or a vector carrying the coding sequence of a DNA-targeting enzyme whereby the DNA-targeting enzyme produces an epigenetic modification on chromosome 2 location 60,716,189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly) thereby reducing the mRNA or protein expression of BCL11A. In one aspect, fetal hemoglobin expression is increased in the mammal, relative to expression prior to the contacting.
[0119] In another embodiment of this aspect and all other aspects described herein, the hematopoietic progenitor cell, the isolated human cell, or isolated cell is contacted ex vivo or in vitro.
[0120] In another embodiment of this aspect and all other aspects described herein, the at least one genetic modification is a deletion. In another embodiment of this aspect and all other aspects described herein, the at least one epigenetic modification.
[0121] In another embodiment of this aspect and all other aspects described herein, the deletion comprises one or more of the DNAse 1 -hypersensitive sites (DHS) +62, +58, and +55 as described herein in the Examples section. In another embodiment of this aspect and all other aspects described herein, the deletion consists essentially of one or more of the DNAse 1 -hypersensitive sites (DHS) +62, +58, and +55 as described herein in the Examples section. In another embodiment, the deletion consists of one or more of the DNAse 1 -hypersensitive sites (DHS) +62, +58, and +55 as described herein in the Examples section.
[0122] In another embodiment of this aspect and all other aspects described herein, the epigenetic modification comprises or affects one or more of the DNAse 1 -hypersensitive sites (DHS) +62, +58, and +55 as described herein in the Examples section. As used herein, the phrase "affects one or more of the DNAse 1 -hypersensitive sites" means natural function of these DNAse 1 -hypersensitive sites (DHS) +62, +58, and +55 are reduce, for example, access to transcription factors or DNA degradation enzymes such as DNase I. In general, DNase I hypersensitive sites (DHSs) are regions of chromatin which are 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, like 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. Accordingly, the epigenetic modification contemplated herein results in reduced access to DNA degradation enzymes that is at least 5% lower is at least 10% lower, at least 20% lower, at least 30% lower, at least 40% lower, at least 50% lower, at least 60% lower, at least 70% lower, at least 80% lower, at least 90% lower, at least 1-fold lower, at least 2-fold lower, at least 5-fold lower, at least 10 fold lower, at least 100 fold lower, at least 1000-fold lower, or more compared to a control cell that is not treated in any method disclosed herein.
[0123] In another embodiment of this aspect and all other aspects described herein, the deletion comprises one or more of the SNP markers described in Table 2. In another embodiment of this aspect and all other aspects described herein, the deletion consists essentially of one or more of the SNP markers described in Table 2. In another embodiment of this aspect and all other aspects described herein, the deletion consists of one or more of the SNP markers described in Table 2.
[0124] In another embodiment of this aspect and all other aspects described herein, the epigenetic modification comprises or affects one or more of the SNP markers described in Table 2. As used herein, the phrase "affects one or more of the SNP markers" means natural function(s) of these SNPs are reduce, for example, access to transcription factors. For example, methylation of these SNPs would reduce the binding of transcription factors, leading to reduced mRNA or protein expression of BCL11A.
[0125] In another embodiment of this aspect and all other aspects described herein, the deletion comprises one or more of the fragments listed in Table 7. In another embodiment of this aspect and all other aspects described herein, the deletion consists essentially of one or more of the fragments listed in Table 7. In another embodiment of this aspect and all other aspects described herein, the deletion consists of one or more of the fragments listed in Table 7. In another embodiment of this aspect and all other aspects described herein, the deletion is from 60,716,189 to 60,728,612, from 60,716,189 to 60,723,870, from 60,722,992 to 60,728,612, from 60,717,236 to 60,719,036, from 60,722,006 to 60,723,058, from 60,724,917 to 60,726,282, from 60,616,396 to 60,618,032, from 60,623,536 to 60,624,989, from 60,626,565 to 60,628, 177, from 60,717,236 to 60,719,036, from 60,721,212 to 60,722,958, from
60,724,780 to 60,726,471, from 60,739,075 to 60,740,154, from 60,748,003 to 60,749,009, from
60,826,438 to 60,827,601, or from 60,831,589 to 60,833,556.
[0126] In another embodiment of this aspect and all other aspects described herein, the epigenetic modification comprises or affects one or more of the fragments listed in Table 7. As used herein, the phrase "affects one or more of the fragments listed in Table 7" means natural function(s) of these fragments are reduce, for example, access to transcription factors. For example, methylation of these fragments would reduce the binding of transcription factors, leading to reduced mRNA or protein expression of BCL11A.
[0127] In another embodiment of this aspect and all other aspects described herein, the epigenetic modification is from 60,716,189 to 60,728,612, from 60,716,189 to 60,723,870, from 60,722,992 to 60,728,612, from 60,717,236 to 60,719,036, from 60,722,006 to 60,723,058, from 60,724,917 to 60,726,282, from 60,616,396 to 60,618,032, from 60,623,536 to 60,624,989, from 60,626,565 to 60,628,177, from 60,717,236 to 60,719,036, from 60,721,212 to 60,722,958, from 60,724,780 to 60,726,471, from 60,739,075 to 60,740, 154, from 60,748,003 to 60,749,009, from 60,826,438 to 60,827,601, or from 60,831,589 to 60,833,556.
[0128] In another embodiment of this aspect and all other aspects described herein, the deletion removes the entire region between chromosome 2 location 60,716,189-60,728,612 or removes a portion of the region resulting in disruption of one of more DNAse 1 -hypersensitive sites (DHS). As used herein, the term "disruption" refers to a decrease in erythroid transcription of BCL11A in a cell comprising a disruption of one or more DNAse -1 hypersensitive sites by at least 10% (e.g., at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 99% or even 100% (i.e., no detectable erythroid transcription)) compared to a cell not having such a disruption. In one embodiment, the disruption comprises an inability of a modified DNAse- lhypersensitive site to bind to its native transcription factors (e.g., GATA1 and TALI).
[0129] In another embodiment of this aspect and all other aspects described herein, the epigenetic modification that interferes with the establishment and/or maintenance of the epigenetic signature at the enhancer region on chromosome 2 location 60,716, 189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly) thereby leading to reduced mRNA or protein expression of BCL11A, and increasing fetal hemoglobin expression in the mammal.
[0130] In one embodiment of this aspect and all other aspects described herein, the epigenetic modification that interferes with the establishment and/or maintenance of the epigenetic signature at the enhancer region on chromosome 2 location 60,716, 189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly) includes but is not limited to epigenetic modifications that affects DNase I sensitivity, epigenetic modifications that affects histone modifications, epigenetic modifications that affects GATA1/TAL1 binding, and epigenetic modifications that affects long-range promoter interaction of the promoter of BCL11A.
[0131] For example, an epigenetic modification that interferes with the establishment and/or maintenance of the epigenetic signature at the enhancer region on chromosome 2 location 60,716, 189- 60,728,612 include but is not limited to at least one deletion within chromosome 2 location 60,716,189- 60,728,612 such that the overall function of this region is affected whereby the mRNA and expression of BCL11A is reduced or decreased. For example, the deletion is at the DNasel sensitivity regions chromosome 2 location 60,716, 189-60,728,612, e.g., +62, +58, and +55. The deletion could be at +62 or +58 or +55 or combination thereof. For examples, at +62 and +58, +58 and +55, +62 and +55, or at all three +62, +58, and +55.
[0132] As another example, an epigenetic modification that interferes with the establishment and/or maintenance of the epigenetic signature at the enhancer region on chromosome 2 location 60,716, 189- 60,728,612 include but is not limited to changes in the histone modifications on chromosome 2 that is not at location 60,716,189-60,728,612, or changes in the histone modifications on chromosome 2 at location 60,716,189-60,728,612, or both histone modifications on chromosome 2 not at location 60,716,189- 60,728,612 as well as at at location 60,716,189-60,728,612 such that the overall function of this region is affected whereby the mRNA and expression of BCL11A is reduced or decreased.
[0133] In another embodiment, an epigenetic modification that interferes with the establishment and/or maintenance of the epigenetic signature at the enhancer region on chromosome 2 location 60,716,189-60,728,612 include but is not limited to an insertion of at least one engineered specific - repressor sequence that change the epigenetic features of noncoding elements at chromosome 2 location 60,716,189-60,728,612 and thus result in repression of target gene expression. The first is specifically focused on epigenetically repressing individual enhancers. In other words, insertion of engineered specific-repressor sequences into chromosome 2 would prospectively interfering with epigenetic modification at the BCL11A erythroid enhancer which eventually leads to reduced BCL11A gene expression.
[0134] Any methods known in the art can be used to produce the epigenetic modification contemplated. For example, as described in Mendenhall E. M. et al., Nat. Biotechnol. 08 September 2013, and Maeder ML et al., Nat Biotechnol. 09 October 2013 2013.
[0135] In one embodiment of this aspect and all other aspects described herein, the insertion of at least one engineered specific-repressor sequence on any location chromosome 2 results in but is not limited to reduced DNasel sensitivity regions at chromosome 2 location 60,716, 189-60,728,612, e.g., +62, +58, and +55; increased histone modifications on chromosome 2 location 60,716, 189-60,728,612; reduced transcription factors binding to the GATA1/TAL1 of the enhancer region on chromosome 2 location 60,716, 189-60,728,612; and reduced or weakened interaction between the chromosome 2 location 60,716, 189-60,728,612 with the BCL11A promoter. [0136] In one embodiment of this aspect and all other aspects described herein, the overall effects of the insertion of at least one engineered specific -repressor sequence on any location chromosome 2 is reduced or decreased mRNA and expression of BCL11A.
[0137] In some embodiments, as used in the context of mRNA and expression of BCL11A, interaction between the chromosome 2 location 60,716, 189-60,728,612 or BCL11A enhancer with the BCL11A promoter, and transcription factors binding to the GATA1/TAL1 of the enhancer region, the term "reduced" or "decreased" refers to at least 5% lower is at least 10% lower, at least 20% lower, at least 30% lower, at least 40% lower, at least 50% lower, at least 60% lower, at least 70% lower, at least 80% lower, at least 90% lower, at least 1-fold lower, at least 2-fold lower, at least 5-fold lower, at least 10 fold lower, at least 100 fold lower, at least 1000-fold lower, or more compared to the control situation that is in the absence of the epigenetic modification or insertion of engineered sequences disclosed herein. By decrease of the BCL11A mRNA or protein expression in the cell means that protein expression is at least 5% lower is at least 10% lower, at least 20% lower, at least 30% lower, at least 40% lower, at least 50% lower, at least 60% lower, at least 70% lower, at least 80% lower, at least 90% lower, at least 1-fold lower, at least 2-fold lower, at least 5 -fold lower, at least 10 fold lower, at least 100 fold lower, at least 1000-fold lower, or more compared to a control cell that does not have the epigenetic modification or insertion of engineered sequences disclosed herein.
[0138] In one embodiment of this aspect and all other aspects described herein, the insertion of at least one engineered specific-repressor sequence occurs within the DNasel sensitivity regions of chromosome 2 location 60,716, 189-60,728,612, e.g., +62, +58, and +55. The insertion could be at the 5'end of +62 or +58 or +55 or at the 3'end of +62 or +58 or +55, or between +62 and +58, or between +58 and +55, or between +55 and +62.
[0139] In one embodiment of this aspect and all other aspects described herein, the insertion of at least one engineered specific-repressor sequence changes the DNasel sensitivity regions of chromosome 2 location 60,716,189-60,728,612.
[0140] In one embodiment of this aspect and all other aspects described herein, the epigenetic modifications changes the DNasel sensitivity regions of chromosome 2 location 60,716, 189-60,728,612.
[0141] In one embodiment of this aspect and all other aspects described herein, the epigenetic modifications changes the histone modifications on chromosome 2 location 60,716, 189-60,728,612.
[0142] In one embodiment of this aspect and all other aspects described herein, the insertion of at least one engineered specific-repressor sequence changes the histone modifications on chromosome 2 location 60,716, 189-60,728,612.
[0143] In one embodiment of this aspect and all other aspects described herein, the epigenetic modifications changes the GATAl/TALl binding of the enhancer region on chromosome 2 location 60,716,189-60,728,612 such that the overall function of this region is affected whereby the mRNA and expression of BCL11A is reduced or decreased. For example, the binding of transcription factors to the GATA1/TAL1.
[0144] In one embodiment of this aspect and all other aspects described herein, the insertion of at least one engineered specific-repressor sequence occurs within the GATA1/TAL1 as described herein. The insertion can be at the 5' end or 3 'end of GATA1 or TALI. The insertion can be between GATA1 and TALI. The insertion changes the GATAl/TALl binding of the enhancer region on chromosome 2 location 60,716, 189-60,728,612 such that the overall function of this region is affected whereby the mRNA and expression of BCL11A is reduced or decreased. For example, the binding of transcription factors to the GATAl/TALl.
[0145] In one embodiment of this aspect and all other aspects described herein, the epigenetic modification changes the interaction between the BCL11A enhancer and the BCL11A promoter. In one embodiment, the interaction is reduced or weakened such that the overall function of this region is affected whereby the mRNA and expression of BCL11A is reduced or decreased.
[0146] In one embodiment of this aspect and all other aspects described herein, the epigenetic modifications changes the interaction between the chromosome 2 location 60,716,189-60,728,612 with the BCL11A promoter. In one embodiment, the interaction is reduced or weakened such that the overall function of this region is affected whereby the mRNA and expression of BCL11A is reduced or decreased.
[0147] Also provided herein in another aspect is an isolated genetic engineered human cell having at least one genetic modification on chromosome 2 location 60,716, 189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly) made by the process of contacting the cell with an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA-targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location 60,716, 189-60,728,612 causing at least one genetic modification therein.
[0148] In one embodiment of this aspect and all other aspects described herein, the isolated genetic engineered human cell having at least one epigenetic modification at the genomic DNA of the cell on chromosome 2. In another of this aspect and all other aspects described herein, the isolated genetic engineered human cell having at least one epigenetic modification at the genomic DNA of the cell on chromosome 2 location 60,716, 189-60,728,612.
[0149] In some aspects of any of these isolated genetic engineered human cells having at least one epigenetic modification, the cells are transplanted into a mammal for use in increasing the fetal hemoglobin in the mammal.
[0150] In one embodiment of this aspect and all other aspects described herein, the isolated genetic engineered human cell having at least one genetic modification at the genomic DNA of the cell on chromosome 2 location 60,716, 189-60,728,612 is transplanted into a mammal for use in increasing the fetal hemoglobin in the mammal.
[0151] In one embodiment of this aspect and all other aspects described herein, the isolated genetic engineered human cell having at least one genetic modification at the genomic DNA of the cell on chromosome 2 location 60,716, 189-60,728,612 is stored for later use by cryopreservation.
[0152] In some aspects of any of those isolated genetic engineered human cells having at least one epigenetic modification, the cells are stored for later use by cryopreservation.
[0153] In one embodiment of this aspect and all other aspects described herein, the isolated genetic engineered human cell having at least one genetic modification at the genomic DNA of the cell on chromosome 2 location 60,716, 189-60,728,612 is cryopreserved, thawed and transplanted into mammal for use in increasing the fetal hemoglobin in the mammal.
[0154] In some aspects of any of those isolated genetic engineered human cells having at least one epigenetic modification, cryopreserved, thawed and transplanted into mammal for use in increasing the fetal hemoglobin in the mammal.
[0155] Another aspect provided herein relates to a composition comprising isolated genetic engineered human cells, wherein the cells have at least one genetic modification on chromosome 2 location 60,716, 189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly) made by the process of contacting the cells with an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA-targeting
endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location 60,716, 189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly) causing at least one genetic modification therein.
[0156] Another aspect provided herein relates to a composition comprising isolated genetic engineered human cells, wherein the cells have at least one epigenetic modification on chromosome 2. In one embodiment, the at least one epigenetic modification on chromosome 2 is at location 60,716, 189- 60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly). In another embodiment, at least one epigenetic modification on chromosome 2 is made by the process of contacting the cells with an effective amount of a composition comprising at least a DNA-targeting enzyme or a vector carrying the coding sequence of a DNA-targeting enzyme whereby the DNA-targeting enzyme produces at least one epigenetic modification in the genomic DNA of the cell on chromosome 2 which affects the location 60,716,189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly) causing therein.
[0157] In one embodiment of this aspect and all other aspects described herein, the composition causes an increase in fetal hemoglobin mRNA or protein expression in the contact cell. [0158] In one embodiment of this aspect and all other aspects described herein, the cells of any compositions described are autologous, to the mammal who is the recipient of the cells in a
transplantation procedure, ie., the cells of the composition are derived or harvested from the mammal prior to any described modification.
[0159] In one embodiment of this aspect and all other aspects described herein, the cells of any compositions described are non-autologous to the mammal who is the recipient of the cells in a transplantation procedure, ie., the cells of the composition are not derived or harvested from the mammal prior to any described modification.
[0160] In one embodiment of this aspect and all other aspects described herein, the cells of any compositions described are at the minimum HLA type matched with to the mammal who is the recipient of the cells in a transplantation procedure.
[0161] In one embodiment of this aspect and all other aspects described herein, the cells of any compositions described are isolated progenitor cells prior to any described modification.
[0162] In one embodiment of this aspect and all other aspects described herein, the cells of any compositions described are isolated hematopoietic progenitor cells prior to any described modification.
[0163] In one embodiment of this aspect and all other aspects described herein, the cells of any compositions described are isolated induced pluripotent stem cells prior to any described modification.
[0164] In another embodiment of this aspect and all other aspects described herein, the deletion comprises one or more of the DNAse 1 -hypersensitive sites (DHS) +62, +58, and +55 as described herein in the Examples section. In another embodiment of this aspect and all other aspects described herein, the deletion consists essentially of one or more of the DNAse 1 -hypersensitive sites (DHS) +62, +58, and +55 as described herein in the Examples section. In another embodiment, the deletion consists of one or more of the DNAse 1 -hypersensitive sites (DHS) +62, +58, and +55 as described herein in the Examples section. In one embodiment, as used herein, the term "portion" in the context of genomic deletion is at least 10% to about 100% of the specified region. In other embodiments, the portion deleted is at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 99% or even 100% of the specified region.
[0165] In another embodiment of this aspect and all other aspects described herein, the deletion comprises one or more of the SNP markers described in Table 2. In another embodiment of this aspect and all other aspects described herein, the deletion consists essentially of one or more of the SNP markers described in Table 2. In another embodiment of this aspect and all other aspects described herein, the deletion consists of one or more of the SNP markers described in Table 2.
[0166] In another embodiment of this aspect and all other aspects described herein, the deletion comprises one or more of the fragments listed in Table 7. In another embodiment of this aspect and all other aspects described herein, the deletion consists essentially of one or more of the fragments listed in Table 7. In another embodiment of this aspect and all other aspects described herein, the deletion consists of one or more of the fragments listed in Table 7. In another embodiment of this aspect and all other aspects described herein, the deletion is from 60,716,189 to 60,728,612, from 60,716,189 to 60,723,870, from 60,722,992 to 60,728,612, from 60,717,236 to 60,719,036, from 60,722,006 to 60,723,058, from 60,724,917 to 60,726,282, from 60,616,396 to 60,618,032, from 60,623,536 to 60,624,989, from
60,626,565 to 60,628, 177, from 60,717,236 to 60,719,036, from 60,721,212 to 60,722,958, from
60,724,780 to 60,726,471, from 60,739,075 to 60,740,154, from 60,748,003 to 60,749,009, from
60,826,438 to 60,827,601, or from 60,831,589 to 60,833,556.
[0167] In another embodiment of this aspect and all other aspects described herein, the deletion removes the entire region between chromosome 2 location 60,716,189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly) or removes a portion of the region resulting in disruption of one of more DNAse 1 -hypersensitive sites (DHS).
[0168] In one embodiment of this aspect and all other aspects described herein, the method further comprises selecting a mammal in need of increasing fetal hemoglobin.
[0169] In one embodiment of this aspect and all other aspects described herein, the mammal has been diagnosed with a hemoglobinopathy.
[0170] In one embodiment of this aspect and all other aspects described herein, the mammal in need of increasing fetal hemoglobin has been diagnosed with a hemoglobinopathy.
[0171] In one embodiment of this aspect and all other aspects described herein, the
hemoglobinopathy is a β -hemoglobinopathy.
[0172] In one embodiment of this aspect and all other aspects described herein, the
hemoglobinopathy is sickle cell disease.
[0173] In one embodiment of this aspect and all other aspects described herein, the
hemoglobinopathy is β -thalassemia.
[0174] In one embodiment of this aspect and all other aspects described herein, the contacted cell, human cell, hematopoietic progenitor cell or its progeny is administered to the mammal.
[0175] In one embodiment, this disclosure provides a method for increasing fetal hemoglobin levels in a mammal in need thereof, the method comprising the steps of providing an isolated population of hematopoietic progenitor cells or hematopoietic stem cells from the mammal in ex vivo, and contacting the population of hematopoietic progenitor or stem cells with an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA- targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location 60,716,189-60,728,612 causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in the mammal, relative to expression prior to the contacting. In further embodiment of this method, the contacted population of hematopoietic progenitor or stem cells having increased fetal hemoglobin expression is cryopreserved and stored or reintroduced into the mammal. In another embodiment, the cryopreserved population of hematopoietic progenitor or stem cells having increased fetal hemoglobin expression is thawed and then reintroduced into the mammal. In further embodiment of this method, the method comprises chemotherapy and/or radiation therapy to remove or reduced the endogenous hematopoietic progenitor or stem cells in the mammal. In any of the embodiment of the described method, the hematopoietic progenitor or stem cells can be substituted with an iPSCs described herein.
[0176] In one embodiment, this disclosure provides a method for increasing fetal hemoglobin levels in a mammal in need thereof, the method comprising the steps of isolating a population of hematopoietic progenitor cells or hematopoietic stem cells from the mammal, and contacting in ex vivo the population of hematopoietic progenitor or stem cells with an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA- targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location 60,716,189-60,728,612 causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in the mammal, relative to expression prior to the contacting. In further embodiment of this method, the ex vivo contacted population of hematopoietic progenitor or stem cells having increased fetal hemoglobin expression is cryopreserved and stored or reintroduced into the mammal. In another embodiment, the cryopreserved population of hematopoietic progenitor or stem cells having increased fetal hemoglobin expression is thawed and then reintroduced into the mammal. In further embodiment of this method, the method comprises chemotherapy and/or radiation therapy to remove or reduced the endogenous hematopoietic progenitor or stem cells in the mammal. In any of the embodiment of the described method, the hematopoietic progenitor or stem cells can be substituted with an iPSCs derived from the mammal. In any embodiment of the method, the method further comprises selecting a mammal in need of increased fetal hemoglobin expression.
[0177] In one embodiment, this disclosure provides a method for increasing fetal hemoglobin levels in a mammal in need thereof, the method comprising the steps of providing isolating a population of hematopoietic progenitor cells or hematopoietic stem cells from the mammal and deleting the genomic DNA of the cells on chromosome 2 location 60,716, 189-60,728,612 causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in said mammal, relative to expression prior to said contacting. In further embodiment of this method, the population of
hematopoietic progenitor or stem cells with deleted genomic DNA and having increased fetal hemoglobin expression is cryopreserved and stored or reintroduced into the mammal. In another embodiment, the population of hematopoietic progenitor or stem cells with deleted genomic DNA and having increased fetal hemoglobin expression is thawed and then reintroduced into the mammal. In further embodiment of this method, the method comprises chemotherapy and/or radiation therapy to remove or reduced the endogenous hematopoietic progenitor or stem cells in the mammal. In any of the embodiment of the described method, the hematopoietic progenitor or stem cells can be substituted with an iPSCs described herein. In any of the embodiment of the described method, the hematopoietic progenitor or stem cells or iPSCs are analogous to the mammal, meaning the cells are derived from the same mammal. In another of the embodiment of the described method, the hematopoietic progenitor or stem cells or iPSCs are non-analogous to the mammal, meaning the cells are not derived from the same mammal, but another mammal of the same species. For example, the mammal is a human. In any embodiment of the method, the method further comprises selecting a mammal in need of increased fetal hemoglobin expression.
[0178] In one embodiment, this disclosure provides a method for increasing fetal hemoglobin levels in a mammal in need thereof, the method comprising the steps of isolating a population of hematopoietic progenitor cells or hematopoietic stem cells from the mammal and ex vivo deleting the genomic DNA of the cells on chromosome 2 location 60,716, 189-60,728,612 causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in said mammal, relative to expression prior to said contacting. In further embodiment of this method, the population of
hematopoietic progenitor or stem cells with deleted genomic DNA and having increased fetal hemoglobin expression is cryopreserved and stored or reintroduced into the mammal. In another embodiment, the cryopreserved population of hematopoietic progenitor or stem cells having increased fetal hemoglobin expression is thawed and then reintroduced into the mammal. In further embodiment of this method, the method comprises chemotherapy and/or radiation therapy to remove or reduced the endogenous hematopoietic progenitor or stem cells in the mammal. In any of the embodiment of the described method, the hematopoietic progenitor or stem cells can be substituted with an iPSCs derived from the mammal. In any embodiment of the method, the method further comprises selecting a mammal in need of increased fetal hemoglobin expression.
[0179] In one embodiment of any method described, the method further comprises selecting a mammal in need of increased fetal hemoglobin expression. Exemplary mammal in need of increased fetal hemoglobin expression is one that has been diagnosed with a hemoglobinopathy.
[0180] In one embodiment, this disclosure provides a method of treatment of a hemoglobinopathy in a mammal comprising the steps of:(a) providing hematopoietic progenitor cells or hematopoietic stem cells or iPSCs; (b) contacting the cells ex vivo or in vitro with an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA- targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location 60,716,189-60,728,612 causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in said mammal, relative to expression prior to said contacting; and (c) administering the of step (b) into the mammal.
[0181] In one embodiment of any method, the cells after step (b) can be cryopreserved till they are needed for administration into the mammal. In further embodiment of this method, the method comprises chemotherapy and/or radiation therapy to remove or reduced the endogenous hematopoietic progenitor or stem cells in the mammal. In any of the embodiment of the described method, the hematopoietic progenitor or stem cells or iPSCs are autologous to the mammal, meaning the cells are derived from the same mammal. In another of the embodiment of the described method, the hematopoietic progenitor or stem cells or iPSCs are non-autologous to the mammal, meaning the cells are not derived from the same mammal, but another mammal of the same species. For example, the mammal is a human.
[0182] In one embodiment of any method described, the method further comprises selecting a mammal in need of treatment of a hemoglobinopathy.
[0183] In one embodiment, this disclosure provides a method of treatment of a hemoglobinopathy in a mammal comprising the steps of:(a) isolating hematopoietic progenitor cells or hematopoietic stem cells from the mammal;(b) contacting the cells ex vivo or in vitro with an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA-targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location 60,716,189-60,728,612 causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in the mammal, relative to expression prior to said contacting; and (c) administering the of step (b) into the mammal.
[0184] In one embodiment, the cells after step (b) can be cryopreserved till they are needed for administration into the mammal. In any embodiment of the method, the method further comprises selecting a mammal in need of treatment of a hemoglobinopathy.
[0185] In one embodiment, this disclosure provides a method of treatment of a hemoglobinopathy in a mammal comprising the steps of: (a) providing hematopoietic progenitor cells or hematopoietic stem cells or iPSCs; (b) ex vivo deleting the genomic DNA of the cells on chromosome 2 location 60,716, 189- 60,728,612 causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in said mammal, relative to expression prior to said contacting; and (c) administering the cells of step (b) into the mammal.
[0186] In one embodiment, the cells after step (b) can be cryopreserved till they are needed for administration into the mammal. In further embodiment of this method, the method comprises chemotherapy and/or radiation therapy to remove or reduced the endogenous hematopoietic progenitor or stem cells in the mammal. In any of the embodiment of the described method, the hematopoietic progenitor or stem cells or iPSCs are analogous to the mammal, meaning the cells are derived from the same mammal. In another of the embodiments of the described method, the hematopoietic progenitor or stem cells or iPSCs are non-analogous to the mammal, meaning the cells are not derived from the same mammal, but another mammal of the same species. For example, the mammal is a human. In any embodiment of the method, the method further comprises selecting a mammal in need of treatment of a hemoglobinopathy. [0187] In one embodiment, this disclosure provides a method of treatment of a hemoglobinopathy in a mammal comprising the steps of: (a) isolating hematopoietic progenitor cells or hematopoietic stem cells from the mammal; (b) ex vivo deleting the genomic DNA of the cells on chromosome 2 location 60,716,189-60,728,612 causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in the mammal, relative to expression prior to said contacting; and (c) administering the of step (b) into the mammal.
[0188] In one embodiment, the cells after step (b) can be cryopreserved till they are needed for administration into the mammal. In further embodiment of this method, the method comprises chemotherapy and/or radiation therapy to remove or reduced the endogenous hematopoietic progenitor or stem cells in the mammal. In any embodiment of the method, the method further comprises selecting a mammal in need of treatment of a hemoglobinopathy.
[0189] In one embodiment, this disclosure provides a method of treatment of a hemoglobinopathy in a mammal (e.g. a human) comprising introducing a composition described herein comprising isolated genetic engineered cells having at least one genetic modification on chromosome 2 location 60,716,189- 60,728,612 whereby fetal hemoglobin expression is increased in the mammal. In further embodiment of this method, the method comprises chemotherapy and/or radiation therapy to remove or reduced the endogenous hematopoietic progenitor or stem cells in the mammal. In any embodiment of the method, the method further comprises selecting a mammal in need of treatment of a hemoglobinopathy.
[0190] In one embodiment, this disclosure provides a method of treatment of a hemoglobinopathy in a mammal (e.g. a human) comprising increasing fetal hemoglobin expression in the mammal by method described herein.
[0191] In any embodiment of any treatment method described, the hemoglobinopathy is a β- hemoglobinopathy.
[0192] In any embodiment of any treatment method described, the hemoglobinopathy is β- thalassemia.
[0193] In any embodiment of any treatment method described, the hemoglobinopathy is sickle cell anemia.
[0194] In one of embodiment of any described method, the hematopoietic progenitor or stem cells or iPSCs are autologous to the mammal, meaning the cells are derived from the same mammal. In another of the embodiment of any described method, the hematopoietic progenitor or stem cells or iPSCs are non-autologous to the mammal, meaning the cells are not derived from the same mammal, but another mammal of the same species. For example, the mammal is a human.
[0195] In one of embodiment of any described method, the contacting of any cell described herein can be ex vivo or in vitro or in vivo. [0196] In another embodiment of any described method, the contacting of any cell described herein comprises contact with an agent that binds the genomic DNA of the cell on chromosome 2 and produces an epigenetic modification in the genome of the cell on chromosome 2, thereby reducing the mRNA or protein expression of BCL11A. In one embodiment, the epigenetic modification is on chromosome 2 location 60,716, 189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly).
[0197] In another embodiment of any described method, the contacting of any cell described herein comprises contact with an agent that binds the genomic DNA of the cell on chromosome 2 location 60,716,189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly), and produces an epigenetic modification on chromosome 2, thereby reducing the mRNA or protein expression of BCL11A.
[0198] In another embodiment of any described method, the contacting of any cell described herein comprises contact with an effective amount of a composition comprising at least a DNA-targeting enzyme or a vector carrying the coding sequence of a DNA-targeting enzyme whereby the DNA- targeting enzyme produces an epigenetic modification on chromosome 2, thereby reducing the mRNA or protein expression of BCL11A.
[0199] In another embodiment of any described method, the contacting of any cell described herein comprises contact with an effective amount of a composition comprising at least a DNA-targeting enzyme or a vector carrying the coding sequence of a DNA-targeting enzyme whereby the DNA- targeting enzyme produces an epigenetic modification on chromosome 2 location 60,716, 189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly) thereby reducing the mRNA or protein expression of BCL11A. In one aspect, fetal hemoglobin expression is increased in the mammal, relative to expression prior to the contacting.
[0200] In another embodiment of any described method, the hematopoietic progenitor cell, the isolated human cell, or isolated cell is contacted ex vivo or in vitro.
[0201] In another embodiment of any described method, the at least one genetic modification is a deletion. In another embodiment of this aspect and all other aspects described herein, the at least one epigenetic modification.
[0202] In one embodiment, provided herein is a use of an agent that binds the genomic DNA of the cell on chromosome 2 location 60,716, 189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly) for increasing the fetal hemoglobin in a mammal or for the treatment of a hemoglobinopathy in the mammal or for reducing the mRNA or expression of BCL1 lA, wherein the mRNA or protein expression of BCL11A is reduced.
[0203] In one embodiment, provided herein is a use of an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA- targeting endonuclease for increasing the fetal hemoglobin in a mammal or for the treatment of a hemoglobinopathy in the mammal or for reducing the mRNA or expression of BCL1 lA, wherein the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location
60,716,189-60,728,612 causing at least one genetic modification therein.
[0204] In one embodiment, provided herein is a use of an effective amount of a composition comprising at least a DNA-targeting enzyme or a vector carrying the coding sequence of a DNA- targeting enzyme for increasing the fetal hemoglobin in a mammal or for the treatment of a
hemoglobinopathy in the mammal or for reducing the mRNA or expression of BCL1 lA, wherein the DNA-targeting enzyme produces at least one epigenetic modification in the genomic DNA of the cell on chromosome 2, thereby affecting the mRNA or expression of BCL11A. In one embodiment, the at least one epigenetic modification is at location 60,716,189-60,728,612. In another embodiment, the effect of the one epigenetic modification is reducing the mRNA or protein expression of BCL11A.
[0205] In one embodiment, provided herein is a use of any isolated cells described herein for increasing the fetal hemoglobin in a mammal or for the treatment of a hemoglobinopathy in the mammal.
[0206] In one embodiment, provided herein is a use of a composition comprising isolated genetic engineered human cells for increasing the fetal hemoglobin in a mammal or for the treatment of a hemoglobinopathy in the mammal, wherein the cells have at least one genetic modification on chromosome 2 location 60,716, 189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly) made by the process of contacting the cells with an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA- targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location 60,716,189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly) causing at least one genetic modification therein.
[0207] In one embodiment, provided herein is a use of a composition comprising isolated genetic engineered human cells for increasing the fetal hemoglobin in a mammal or for the treatment of a hemoglobinopathy in the mammal, wherein the cells have at least one epigenetic modification on chromosome 2. In one embodiment, the at least one epigenetic modification on chromosome 2 is at location 60,716, 189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly). In another embodiment, at least one epigenetic modification on chromosome 2 is made by the process of contacting the cells with an effective amount of a composition comprising at least a DNA-targeting enzyme or a vector carrying the coding sequence of a DNA-targeting enzyme whereby the DNA- targeting enzyme produces at least one epigenetic modification in the genomic DNA of the cell on chromosome 2 which affects the location 60,716,189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly) causing therein.
[0208] In one embodiment, provided herein is a use of any isolated cells described herein or any one of the compositions described herein for the manufacture of a medicament for increasing the fetal hemoglobin in a mammal or for the treatment of a hemoglobinopathy in the mammal. [0209] In one embodiment of use of the composition described herein, the composition causes an increase in fetal hemoglobin mRNA or protein expression in the contact cell.
[0210] In one embodiment of use of the composition described herein, the cells of any
compositions described are autologous, to the mammal who is the recipient of the cells in a
transplantation procedure, ie., the cells of the composition are derived or harvested from the mammal prior to any described modification.
[0211] In one embodiment of use of the composition described herein, the cells of any
compositions described are non-autologous to the mammal who is the recipient of the cells in a transplantation procedure, ie., the cells of the composition are not derived or harvested from the mammal prior to any described modification.
[0212] In one embodiment of use of the composition described herein, the cells of any
compositions described are at the minimum HLA type matched with to the mammal who is the recipient of the cells in a transplantation procedure.
[0213] In one embodiment of use of the composition described herein, the cells of any
compositions described are isolated progenitor cells prior to any described modification.
[0214] In one embodiment of use of the composition described herein, the cells of any
compositions described are isolated hematopoietic progenitor cells prior to any described modification.
[0215] In one embodiment of use of the composition described herein, the cells of any
compositions described are isolated induced pluripotent stem cells prior to any described modification.
[0216] In one embodiment of use of the composition described herein, the cells of any
compositions described are cryopreserved prior to use.
[0217] It is known that there are HbF-associated variations at BCL11A. Six GWAS of HbF level (or the highly correlated trait F-cell number) have been conducted in individuals of European, African and Asian descent, each identifying trait-associated variants within BCL11A (7-12). The same variants are associated with the clinical severity of SCD and β-thalassemia (9, 10, 50), consistent with HbF as a major modifier of these disorders. Variation at BCL11A is estimated to explain -15% of the trait variance in HbF level (7, 12, 43). Four different SNPs have been identified as most highly associated with the trait (rsl427407 (7), rsl 1886868 (8), rs4671393 (9) and rs766432 (10-12)); these sentinel SNPs cluster within 3 kb of each other in BCL11A intron-2 (Figs. 2A and 1 IB). Haplotypes including the sentinel SNPs appear to better explain the HbF association than any individual SNP (12, 43). Fifty SNPs at the BCL11A locus and twenty-seven SNPs within intron-2 have been associated with HbF level with genome-wide significance (P < 5 x 10-8). Despite large-scale resequencing efforts, coding variants of BCL11A have not been described (43). [0218] Previously, the inventors used the CSSCD to fine-map the association signal with HbF at the BCLl 1A locus and reported a strong association with rs4671393 (43). In that study, rsl427407 was imputed. Two additional SNPs, rs766432 and rsl 1886868 have also been identified in prior studies as sentinel SNPs most highly trait-associated (8, 10, 11, 51). In a subset of individuals (n = 728) for which genotypes at all four sentinel SNPs were available, the association result was not significant at rs4671393, rs766432 or rsl 1886868 following conditioning on genotypes at rsl427407; conversely, the association remained highly significant for rsl427407 upon conditioning on rs4671393, rs766432 or rsl 1886868 (Table 4). Therefore, rs 1427407 is the SNP most strongly associated with HbF level within the erythroid DHSs and better accounts for the trait association than other previously described sentinel SNPs.
[0219] Conditional analysis demonstrated associations that remained significant after conditioning on rsl427407. The most significant residual association was for rs7606173 in DHS +55 (P = 9.66 x 10- 11); rs7599488 in DHS +62, which we had previously reported (43), was only slightly less significant (P = 2.43 x 10-10) (Table 1). Analysis of rare DNA sequence variants within the three DHSs did not yield additional independent HbF-associated signals (Table 5).
[0220] The inventors have found that allele-specific transcription factor (TF) binding are involved with BCLl 1A expression. Allele-specific biochemical studies were performed using informative heterozygotes to control for trans-acting differences between samples and to ensure equal abundance of both alleles, substantiated by equal representation of alleles in paired gDNA (Figs. 2B and 2C).
rs 1427407 is found directly at the center of a GATA1 and TALI binding peak at DHS +62 (Fig. 2B). In the ChlP assays performed, chromatin was sonicated to approximately 500-bp fragments. The five primary human erythroid precursor samples heterozygous for rs 1427407 used for ChlP-qPCR were Sanger sequenced at the erythroid DHSs. The only other heterozygous SNP within 500-bp of rs 1427407 in any of these samples was rs7599488 (304-bp 3' of rsl427407) which was heterozygous in just two of the five samples. This SNP does not fall within GATA1 or TALI binding motifs. It therefore appears unlikely that another SNP within DHS +62 could account for the observed allele-specific TF binding.
[0221] In addition, the inventors have found that there is an association between BCL 11 A expression and HbF level. The inventors' studies provide an estimate of the change in BCLl 1A expression that may result in a clinically meaningful increase in HbF level. Among a limited set of human lymphoblastoid cell lines were previously reported correlation of the high HbF-associated A- allele of rs4671393 with reduced BCLl 1A expression (13). Extension of these experiments to a larger collection of genotyped lines failed to confirm this observation. Hence, The inventors have found that the HbF-associated rsl427407-rs7606173 haplotype influence BCLl 1A expression in an erythroid-specific context, a possibility consistent with the DNase I sensitivity findings. BCLl 1A mRNA expression in primary erythroid precursors differed by 1.7-fold between the high-HbF rsl427407-rs7606173 T-G and low-HbF G-C haplotypes (Fig. 3B); correspondingly, median HbF levels were 10.6% and 3.1% in T-G and G-C homozygotes, respectively (Fig. 3C). Of note, the results demonstrating allele-specific expression of BCLl 1A in primary human erythroid cells were observed in cells heterozygous for the rsl427407-rs7606173 haplotype, and thus the modest effects on BCLl 1A expression reflect the combined effects of all functional SNPs within the haplotype. While inheritance of a protective BCLl 1A haplotype is clinically beneficial on a population basis (9, 10, 50), the average level of HbF in T-G homozygotes remains below that required to prevent morbidity from SCD. The sensitivity of HbF level to BCLl 1A expression, however, predicts that relief of disease severity might require only a modest further reduction in BCLl 1A expression.
[0222] The inventors further investigated the developmental regulation of globin genes and BCLl 1 A. During human development, yolk sac-derived ε-globin is superseded in the first trimester by fetal liver-derived γ-globin. Following birth, as erythropoiesis shifts from the liver to the bone marrow, γ- globin is gradually silenced and β -globin predominates. Only a single switch in globin gene expression occurs in mouse ontogeny. During this transition, which occurs at mid-gestation, the circulating yolk sac- derived primitive erythrocytes express embryonic-stage globins sy and βΗΙ, whereas the fetal liver definitive erythroblasts express adult-stage globins β 1 and β2. Concordant with this developmental switch, BCLl 1A is expressed in the definitive but not primitive-stage erythroid lineage and required for the change in globin gene expression (16, 52).
[0223] In the stable transgenic BCLl 1A +52.0-64.4 reporter lines at 10.5 dpc, lacZ expression was observed only in the fetal liver primordium and not in the circulating blood within the embryo, placenta or yolk sac (Fig. 6A). These results, coupled with the finding of lacZ expression in the 12.5 dpc definitive fetal liver erythroblasts but not yolk sac-derived primitive circulating erythrocytes (Fig. 4B), demonstrate that the BCLl 1A composite enhancer sequences drive expression in a developmentally-specific pattern concordant with endogenous globin gene switching.
[0224] A series of deletion mutants was generated to refine the minimal elements required for erythroid enhancer activity. Sequences containing the central +58 DHS were sufficient for erythroid enhancer activity. Those sequences containing only the flanking +62 or +55 elements were unable to direct erythroid gene expression (Fig. 6B). To test the ability of the DHSs to enhance gene expression in primary human erythroid precursors, we used lentiviral delivery of a GFP reporter system as previously described (39). Similarly, the +58 DHS enhanced gene expression in this reporter assay (Fig. 7).
[0225] The inventors decided to generate cell lines with a Bell la enhancer deletion to investigate the requirement of the enhancer for BCLl 1A expression. Stable erythroid cells with disruption of the enhancer were generated. Since there are no suitable adult-stage human erythroid cell lines, and as proof of principle, the inventors turned to the murine system. Mouse erythroleukemia (MEL) cells depend on BCLl 1A for an adult-stage pattern of globin gene expression (14). The inventors identified an orthologous erythroid composite enhancer at mouse Bell la intron-2. Like the human GWAS-marked intron-2 BCLl 1A enhancer, these sequences possessed a series of erythroid-specific DHSs. In addition, these sequences were decorated by H3K4mel and H3K27ac, lacked H3K4me3 and H3K27me3, and occupied by both GATA1 and TALI in mouse erythroid chromatin (Fig. 8). Composite regulatory elements including a series of adjacent DHSs have been shown to be critical for gene expression at numerous loci, including among others the β-globin locus control region, a-globin multispecies conserved sequences, and IgH regulatory region (53-55). We observed species-specific unique features of the composite enhancer. For example, The inventors identified the conserved mouse sequences to each of the three human DHSs +62, +58 and +55, and found erythroid DNase I hypersensitivity at the +62 and +55 conserved sequences, however the +58 conserved sequences lacked DNase I hypersensitivity.
[0226] PCR and Southern blotting verified excision of the +50.4-60.4 kb intronic segment of Bell la in three unique MEL clones and two unique pre-B lymphocyte clones (Fig. 9). Sanger-sequenced breakpoints were characteristic of TALEN-mediated cleavage with subsequent NHEJ repair (Fig. 10). Upon deletion of the intronic segment, we observed dramatic reduction in BCLl 1A transcript in the MEL cell clones by RT-qPCR, using primer pairs detecting exon junctions upstream, spanning or downstream of the deletion (Fig. 5A).
Definitions
[0227] For convenience, certain terms employed in the entire application (including the specification, examples, and appended claims) are collected here. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
[0228] As used herein, the phrase "agent that binds the genomic DNA of the cell on chromosome 2 location 60,716, 189-60,728,612" refers to small molecules, nucleic acids, proteins, peptides or oligonucleotides that can bind to the location within the genomic DNA {e.g., chromosome 2 location 60,716,189-60,728,612) and represses mRNA or protein expression of BCLl 1A in a cell by at least 20% compared to the mRNA or protein level of BCLl 1A in a cell not treated with such an agent. In one embodiment, the agent "interferes with BCLl 1A interactions with BCLl 1A binding partners," as that phrase is used herein.
[0229] As used herein, the term "small molecule" refers to a chemical agent including, but not limited to, peptides, peptidomimetics, amino acids, amino acid analogs, polynucleotides, polynucleotide analogs, aptamers, nucleotides, nucleotide analogs, organic or inorganic compounds (i.e., including heteroorganic and organometallic compounds) having a molecular weight less than about 10,000 grams per mole, organic or inorganic compounds having a molecular weight less than about 5,000 grams per mole, organic or inorganic compounds having a molecular weight less than about 1,000 grams per mole, organic or inorganic compounds having a molecular weight less than about 500 grams per mole, and salts, esters, and other pharmaceutically acceptable forms of such compounds.
[0230] A "nucleic acid", as described herein, can be RNA or DNA, and can be single or double stranded, and can be selected, for example, from a group including: nucleic acid encoding a protein of interest, oligonucleotides, nucleic acid analogues, for example peptide- nucleic acid (PNA), pseudo- complementary PNA (pc-PNA), locked nucleic acid (LNA) etc. Such nucleic acid sequences include, for example, but are not limited to, nucleic acid sequence encoding proteins, for example that act as transcriptional repressors, antisense molecules, ribozymes, small inhibitory nucleic acid sequences, for example but are not limited to RNAi, shRNAi, siRNA, micro RNAi (mRNAi), antisense oligonucleotides etc.
[0231] By "interferes with BCL 11 A interactions with BCL 11 A binding partners" is meant that the amount of interaction of BCLl 1A with the BCLl 1A binding partner is at least 5% lower in populations treated with a BCLl 1A inhibitor, than a comparable, control population, wherein no BCLl 1A inhibitor is present. It is preferred that the amount of interaction of BCLl 1A with the BCLl 1A binding partner in a BCLl lA-inhibitor treated population is at least 10% lower, at least 20% lower, at least 30% lower, at least 40% lower, at least 50% lower, at least 60% lower, at least 70% lower, at least 80% lower, at least 90% lower, at least 1-fold lower, at least 2-fold lower, at least 5 -fold lower, at least 10 fold lower, at least 100 fold lower, at least 1000-fold lower, or more than a comparable control treated population in which no BCLl 1A inhibitor is added. At a minimum, BCLl 1A interaction can be assayed by determining the amount of BCLl 1A binding to the BCLl 1A binding partner using techniques standard in the art, including, but not limited to, mass spectrometry, immunoprecipitation, or gel filtration assays.
Alternatively, or in addition, BCLl 1A activity can be assayed by measuring fetal hemoglobin expression at the mRNA or protein level following treatment with a candidate BCLl 1A inhibitor.
[0232] In one embodiment, BCL 11 A activity is the interaction of BCL 11 A with its binding partners: GATA-1, FOG-1, components of the NuRD complex, matrin-3, MTA2 and RBBP7.
Accordingly, any antibody or fragment thereof, small molecule, chemical or compound that can block this interaction is considered an inhibitor of BCLl 1A activity.
[0233] As used herein, the term "genetic engineered cell" refers to a cell that comprises at least one genetic modification, as that term is used herein.
[0234] As used herein, the term "genetic modification" refers to a disruption at the genomic level resulting in a decrease in BCLl 1A expression or activity in a cell. Exemplary genetic modifications can include deletions, frame shift mutations, point mutations, exon removal, removal of one or more DNAse 1 -hypersensitive sites (DHS) (e.g., 2, 3, 4 or more DHS regions), etc.
[0235] By "inhibits BCLl 1A expression" is meant that the amount of expression of BCLl 1A is at least 5% lower in a cell or cell population treated with a DNA-targeting endonuc lease, than a comparable, control cell or cell population, wherein no DNA-targeting endonuclease is present. It is preferred that the percentage of BCLl 1A expression in a treated population is at least 10% lower, at least 20% lower, at least 30% lower, at least 40% lower, at least 50% lower, at least 60% lower, at least 70% lower, at least 80% lower, at least 90% lower, at least 1-fold lower, at least 2-fold lower, at least 5-fold lower, at least 10 fold lower, at least 100 fold lower, at least 1000-fold lower, or more than a comparable control treated population in which no DNA-targeting endonuclease is added.
[0236] By "inhibits BCL11A activity" is meant that the amount of functional activity of BCL11A is at least 5% lower in a cell or cell population treated with the methods described herein, than a comparable, control cell or population, wherein no DNA-targeting endonuclease is present. It is preferred that the percentage of BCL11A activity in a BCL1 lA-inhibitor treated population is at least 10% lower, at least 20% lower, at least 30% lower, at least 40% lower, at least 50% lower, at least 60% lower, at least 70% lower, at least 80% lower, at least 90% lower, at least 1-fold lower, at least 2-fold lower, at least 5 -fold lower, at least 10 fold lower, at least 100 fold lower, at least 1000-fold lower, or more than a comparable control treated population in which no DNA-targeting endonuclease is added. At a minimum, BCL11A activity can be assayed by determining the amount of BCL11A expression at the protein or mRNA levels, using techniques standard in the art. Alternatively, or in addition, BCL11A activity can be determined using a reporter construct, wherein the reporter construct is sensitive to BCL11A activity. The γ-globin locus sequence is recognizable by the nucleic acid-binding motif of the BCL11A construct.
[0237] In one embodiment, as used herein, the term "DNA targeting endonuclease" refers to an endonuclease that generates a double-stranded break at a desired position in the genome (e.g., chromosome 2 location 60,716, 189-60,728,612) without producing undesired off-target double-stranded breaks. The DNA targeting endonuclease can be a naturally occurring endonuclease (e.g., a bacterial meganuclease) or it can be artificially generated (e.g., engineered meganucleases, TALENs, or ZFNs, among others).
[0238] In another embodiment, as used herein, the term "DNA targeting endonuclease" refers to an endonuclease that generates a single-stranded break or a "nick" or break on one strand of the DNA phosphate sugar backbone at a desired position in the genome (e.g., chromosome 2 location 60,716, 189- 60,728,612) without producing undesired off-target DNA stranded breaks.
[0239] As used herein, the term "vector" refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a "plasmid", which refers to a circular double stranded DNA loop into which additional nucleic acid segments can be ligated. Another type of vector is a viral vector, wherein additional nucleic acid segments can be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian vectors) are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome.
Moreover, certain vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as "recombinant expression vectors", or more simply "expression vectors." In general, expression vectors of utility in recombinant DNA techniques are often in the form of plasmids. In the present specification, "plasmid" and "vector" can be used interchangeably as the plasmid is the most commonly used form of vector. However, the methods and compositions described herein can include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, lentiviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions.
[0240] Within an expression vector, "operably linked" is intended to mean that the nucleotide sequence of interest is linked to the regulatory sequence(s) in a manner which allows for expression of the nucleotide sequence (e.g., in an in vitro transcription/translation system or in a target cell when the vector is introduced into the target cell). The term "regulatory sequence" is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Such regulatory sequences are described, for example, in Goeddel; Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, CA (1990). Regulatory sequences include those which direct constitutive expression of a nucleotide sequence in many types of host cell and those which direct expression of the nucleotide sequence only in certain host cells (e.g., tissue-specific regulatory sequences). Furthermore, the DNA-targeting endonuclease can be delivered by way of a vector comprising a regulatory sequence to direct synthesis of the DNA-targeting endonuclease at specific intervals, or over a specific time period. It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the choice of the target cell, the level of expression desired, and the like.
[0241] As used herein the term "cleaves" generally refers to the generation of a double-stranded break in the DNA genome at a desired location.
[0242] As used herein, the term "effective amount of a composition comprising at least a DNA- targeting endonuclease" refers to an amount of a DNA-targeting endonuclease that yields sufficient endonuclease activity to generate a double-stranded break in the desired location of the genome. In one embodiment, the effective amount of a DNA-targeting endonuclease generates a double-stranded break at the desired genetic locus in at least 20% of the cells in a population contacted with the composition (e.g., at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 99%, or even 100% of the cells in the population comprise a genetic modification produced by the DNA-targeting endonuclease composition).
[0243] As used herein the term "increasing the fetal hemoglobin levels" in a cell indicates that fetal hemoglobin is at least 5% higher in populations treated with an agent that disrupts BCL11A mRNA or protein expression (e.g., a DNA-targeting endonuclease) by binding to genomic DNA at chromosome 2 location 60,716, 189-60,728,612, than in a comparable, control population, wherein no agent is present. It is preferred that the percentage of fetal hemoglobin expression in a population treated with such an agent that binds the genomic DNA at chromosome 2 location 60,716,189-60,728,612 is at least 10% higher, at least 20% higher, at least 30% higher, at least 40% higher, at least 50% higher, at least 60% higher, at least 70% higher, at least 80% higher, at least 90% higher, at least 1-fold higher, at least 2-fold higher, at least 5 -fold higher, at least 10 fold higher, at least 100 fold higher, at least 1000-fold higher, or more than a control treated population of comparable size and culture conditions. The term "control treated population" is used herein to describe a population of cells that has been treated with identical media, viral induction, nucleic acid sequences, temperature, confluency, flask size, pH, etc., with the exception of the addition of the agent that binds genomic DNA at chromosome 2 location 60,716, 189-60,728,612. In one embodiment, any method known in the art can be used to measure an increase in fetal hemoglobin expression, e. g. Western Blot analysis of fetal γ-globin protein and quantifying mRNA of fetal γ-globin.
[0244] The term "isolated cell" as used herein refers to a cell that has been removed from an organism in which it was originally found, or a descendant of such a cell. Optionally the cell has been cultured in vitro, e.g., in the presence of other cells. Optionally the cell is later introduced into a second organism or re-introduced into the organism from which it (or the cell from which it is descended) was isolated.
[0245] The term "isolated population" with respect to an isolated population of cells as used herein refers to a population of cells that has been removed and separated from a mixed or heterogeneous population of cells. In some embodiments, an isolated population is a substantially pure population of cells as compared to the heterogeneous population from which the cells were isolated or enriched. In some embodiments, the isolated population is an isolated population of human hematopoietic progenitor cells, e.g., a substantially pure population of human hematopoietic progenitor cells as compared to a heterogeneous population of cells comprising human hematopoietic progenitor cells and cells from which the human hematopoietic progenitor cells were derived.
[0246] The term "substantially pure," with respect to a particular cell population, refers to a population of cells that is at least about 75%, preferably at least about 85%, more preferably at least about 90%, and most preferably at least about 95% pure, with respect to the cells making up a total cell population. That is, the terms "substantially pure" or "essentially purified," with regard to a population of hematopoietic progenitor cells, refers to a population of cells that contain fewer than about 20%, more preferably fewer than about 15%, 10%, 8%, 7%, most preferably fewer than about 5%, 4%, 3%, 2%, 1%, or less than 1%, of cells that are not hematopoietic progenitor cells as defined by the terms herein.
[0247] As used herein, the term "treating" includes reducing or alleviating at least one adverse effect or symptom of a condition, disease or disorder. For example, the term "treating" and "treatment" refers to administering to a subject an effective amount of a composition, e.g., an effective amount of a composition comprising a population of hematopoietic progenitor cells so that the subject has a reduction in at least one symptom of the disease or an improvement in the disease, for example, beneficial or desired clinical results. For purposes of this disclosure, beneficial or desired clinical results include, but are not limited to, alleviation of one or more symptoms, diminishment of extent of disease, disease stabilization (e.g., not worsening), delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total) , whether detectable or undetectable. In some embodiments, treating can refer to prolonging survival as compared to expected survival if not receiving treatment. Thus, one of skill in the art realizes that a treatment can improve the disease condition, but may not be a complete cure for the disease. In some embodiments, treatment can include prophylaxis. However, in alternative embodiments, treatment does not include prophylaxis.
[0248] The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
[0249] As used herein, the terms "pharmaceutically acceptable", "physiologically tolerable" and grammatical variations thereof, as they refer to compositions, carriers, diluents and reagents, are used interchangeably and represent that the materials are capable of administration to or upon a mammal without the production of undesirable physiological effects such as nausea, dizziness, gastric upset and the like. A pharmaceutically acceptable carrier will not promote the raising of an immune response to an agent with which it is admixed, unless so desired. The preparation of a pharmacological composition that contains active ingredients dissolved or dispersed therein is well understood in the art and need not be limited based on formulation. Typically such compositions are prepared as injectable either as liquid solutions or suspensions, however, solid forms suitable for solution, or suspensions, in liquid prior to use can also be prepared. The preparation can also be emulsified or presented as a liposome composition. The active ingredient can be mixed with excipients which are pharmaceutically acceptable and compatible with the active ingredient and in amounts suitable for use in the therapeutic methods described herein. Suitable excipients are, for example, water, saline, dextrose, glycerol, ethanol or the like and
combinations thereof. In addition, if desired, the composition can contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents and the like which enhance the effectiveness of the active ingredient. The therapeutic composition of the present invention can include pharmaceutically acceptable salts of the components therein. Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the polypeptide) that are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, tartaric, mandelic and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine and the like. Physiologically tolerable carriers are well known in the art. Exemplary liquid carriers are sterile aqueous solutions that contain no materials in addition to the active ingredients and water, or contain a buffer such as sodium phosphate at physiological pH value, physiological saline or both, such as phosphate-buffered saline. Still further, aqueous carriers can contain more than one buffer salt, as well as salts such as sodium and potassium chlorides, dextrose, polyethylene glycol and other solutes. Liquid compositions can also contain liquid phases in addition to and to the exclusion of water. Exemplary of such additional liquid phases are glycerin, vegetable oils such as cottonseed oil, and water-oil emulsions. The amount of an active agent used with the methods described herein that will be effective in the treatment of a particular disorder or condition will depend on the nature of the disorder or condition, and can be determined by standard clinical techniques.
[0250] As used herein, "prevention" or "preventing," when used in reference to a disease, disorder or symptoms thereof, refers to a reduction in the likelihood that an individual will develop a disease or disorder, e.g., a hemoglobinopathy. The likelihood of developing a disease or disorder is reduced, for example, when an individual having one or more risk factors for a disease or disorder either fails to develop the disorder or develops such disease or disorder at a later time or with less severity, statistically speaking, relative to a population having the same risk factors and not receiving treatment as described herein. The failure to develop symptoms of a disease, or the development of reduced (e.g., by at least 10% on a clinically accepted scale for that disease or disorder) or delayed (e.g., by days, weeks, months or years) symptoms is considered effective prevention.
[0251] In connection with contacting a cell with a DNA-targeting endonuclease to decrease BCL11A expression, the phrase "increasing fetal hemoglobin levels in a cell" indicates that fetal hemoglobin in a cell or population of cells is at least 5% higher in the cell or population of cells treated with the DNA-targeting endonuclease, than a comparable, control population, wherein no DNA-targeting endonuclease is present. It is preferred that the fetal hemoglobin expression in a DNA-targeting endonuclease treated cell is at least 10% higher, at least 20% higher, at least 30% higher, at least 40% higher, at least 50% higher, at least 60% higher, at least 70% higher, at least 80% higher, at least 90% higher, at least 1-fold higher, at least 2-fold higher, at least 5 -fold higher, at least 10 fold higher, at least 100 fold higher, at least 1000-fold higher, or more than a comparable control treated population. The term "control treated population" is used herein to describe a population of cells that has been treated with identical media, viral induction, nucleic acid sequences, temperature, confluency, flask size, pH, etc., with the exception of the addition of the BCL11A inhibitor.
[0252] The term "mammal" is intended to encompass a singular "mammal" and plural "mammals," and includes, but is not limited to humans; primates such as apes, monkeys, orangutans, and
chimpanzees; canids such as dogs and wolves; felids such as cats, lions, and tigers; equids such as horses, donkeys, and zebras; food animals such as cows, pigs, and sheep; ungulates such as deer and giraffes; rodents such as mice, rats, hamsters and guinea pigs; and bears. In some preferred embodiments, a mammal is a human.
[0253] Accordingly, in one embodiment, the mammal has been diagnosed with a
hemoglobinopathy. In a further embodiment, the hemoglobinopathy is a β -hemoglobinopathy. In one preferred embodiment, the hemoglobinopathy is a sickle cell disease. As used herein, "sickle cell disease" can be sickle cell anemia, sickle-hemoglobin C disease (HbSC), sickle beta-plus-thalassaemia (HbS/p+), or sickle beta-zero-thalassaemia (HbS/βΟ). In another preferred embodiment, the
hemoglobinopathy is a β -thalassemia. [0254] As used herein, the term "hemoglobinopathy" means any defect in the structure or function of any hemoglobin of an individual, and includes defects in the primary, secondary, tertiary or quaternary structure of hemoglobin caused by any mutation, such as deletion mutations or substitution mutations in the coding regions of the β-globin gene, or mutations in, or deletions of, the promoters or enhancers of such genes that cause a reduction in the amount of hemoglobin produced as compared to a normal or standard condition. The term further includes any decrease in the amount or effectiveness of hemoglobin, whether normal or abnormal, caused by external factors such as disease, chemotherapy, toxins, poisons, or the like.
[0255] In one embodiment, the term "effective amount", as used herein, refers to the amount of a cell composition that is safe and sufficient to treat, lesson the likelihood of, or delay the development of a hemoglobinopathy. The amount can thus cure or result in amelioration of the symptoms of the hemoglobinopathy, slow the course of hemoglobinopathy disease progression, slow or inhibit a symptom of a hemoglobinopathy, slow or inhibit the establishment of secondary symptoms of a hemoglobinopathy or inhibit the development of a secondary symptom of a hemoglobinopathy. The effective amount for the treatment of the hemoglobinopathy depends on the type of hemoglobinopathy to be treated, the severity of the symptoms, the subject being treated, the age and general condition of the subject, the mode of administration and so forth. Thus, it is not possible or prudent to specify an exact "effective amount". However, for any given case, an appropriate "effective amount" can be determined by one of ordinary skill in the art using only routine experimentation.
[0256] As used herein the term "comprising" or "comprises" is used in reference to compositions, methods, and respective component(s) thereof, that are essential to the invention, yet open to the inclusion of unspecified elements, whether essential or not.
[0257] As used herein the term "consisting essentially of refers to those elements required for a given embodiment. The term permits the presence of additional elements that do not materially affect the basic and novel or functional characteristic(s) of that embodiment of the invention.
[0258] The term "consisting of refers to compositions, methods, and respective components thereof as described herein, which are exclusive of any element not recited in that description of the embodiment.
[0259] As used in this specification and the appended claims, the singular forms "a," "an," and "the" include plural references unless the context clearly dictates otherwise. Thus for example, references to "the method" includes one or more methods, and/or steps of the type described herein and/or which will become apparent to those persons skilled in the art upon reading this disclosure and so forth. It is understood that the foregoing detailed description and the following examples are illustrative only and are not to be taken as limitations upon the scope of the invention. Various changes and modifications to the disclosed embodiments, which will be apparent to those of skill in the art, may be made without departing from the spirit and scope of the present invention. Further, all patents, patent applications, and publications identified are expressly incorporated herein by reference for the purpose of describing and disclosing, for example, the methodologies described in such publications that might be used in connection with the present invention. These publications are provided solely for their disclosure prior to the filing date of the present application. Nothing in this regard should be construed as an admission that the inventors are not entitled to antedate such disclosure by virtue of prior invention or for any other reason. All statements as to the date or representation as to the contents of these documents are based on the information available to the applicants and do not constitute any admission as to the correctness of the dates or contents of these documents.
Hemoglobinopathies
[0260] Fetal hemoglobin (HbF) is a tetramer of two adult ct-globin polypeptides and two fetal β- like γ-globin polypeptides. During gestation, the duplicated γ-globin genes constitute the predominant genes transcribed from the β-globin locus. Following birth, γ-globin becomes progressively replaced by adult β-globin, a process referred to as the "fetal switch" (3). The molecular mechanisms underlying this switch have remained largely undefined and have been a subject of intense research. The developmental switch from production of predominantly fetal hemoglobin or HbF (α2γ2) to production of adult hemoglobin or HbA (α2β2) begins at about 28 to 34 weeks of gestation and continues shortly after birth at which point HbA becomes predominant. This switch results primarily from decreased transcription of the gamma-globin genes and increased transcription of beta-globin genes. On average, the blood of a normal adult contains only about 2% HbF, though residual HbF levels have a variance of over 20 fold in healthy adults (Atweh, Semin. Hematol. 38(4):367-73 (2001)).
[0261] Hemoglobinopathies encompass a number of anemias of genetic origin in which there is a decreased production and/or increased destruction (hemolysis) of red blood cells (RBCs). These disorders also include genetic defects that result in the production of abnormal hemoglobins with a concomitant impaired ability to maintain oxygen concentration. Some such disorders involve the failure to produce normal β-globin in sufficient amounts, while others involve the failure to produce normal β-globin entirely. These disorders specifically associated with the β-globin protein are referred to generally as β- hemoglobinopathies. For example, β -thalassemias result from a partial or complete defect in the expression of the β-globin gene, leading to deficient or absent HbA. Sickle cell anemia results from a point mutation in the β-globin structural gene, leading to the production of an abnormal (sickled) hemoglobin (HbS). HbS RBCs are more fragile than normal RBCs and undergo hemolysis more readily, leading eventually to anemia (Atweh, Semin. Hematol. 38(4):367-73 (2001)). Moreover, the presence of a BCLUA genetic variant, HBS IL-MYB variation, ameliorates the clinical severity in beta-thalassemia. This variant has been shown to be associated with HbF levels. It has been shown that there is an odds ratio of 5 for having a less severe form of beta-thalassemia with the high-HbF variant (Galanello S. et al., 2009, Blood, in press).
[0262] The search for treatment aimed at reduction of globin chain imbalance in patients with β- hemoglobinopathies has focused on the pharmacologic manipulation of fetal hemoglobin (α2γ2; HbF). The important therapeutic potential of such approaches is suggested by observations of the mild phenotype of individuals with co-inheritance of both homozygous β-thalassemia and hereditary persistence of fetal hemoglobin (HPFH), as well as by those patients with homozygous β-thalassemia who synthesize no adult hemoglobin, but in whom a reduced requirement for transfusions is observed in the presence of increased concentrations of fetal hemoglobin. Furthermore, it has been observed that certain populations of adult patients with β chain abnormalities have higher than normal levels of fetal hemoglobin (HbF), and have been observed to have a milder clinical course of disease than patients with normal adult levels of HbF. For example, a group of Saudi Arabian sickle-cell anemia patients who express 20-30% HbF have only mild clinical manifestations of the disease (Pembrey, et al., Br. J.
Haematol. 40: 415-429 (1978)). It is now accepted that β -hemoglobinopathies, such as sickle cell anemia and the β-thalassemias, are ameliorated by increased HbF production. (Reviewed in Jane and
Cunningham Br. J. Haematol. 102: 415-422 (1998) and Bunn, N. Engl. J. Med. 328: 129-131 (1993)).
[0263] While the molecular mechanisms controlling the in vivo developmental switch from γ- to β- globin gene expression are currently unknown, there is accumulating evidence that external factors can influence γ-globin gene expression. The first group of compounds discovered having HbF reactivation activity were cytotoxic drugs. The ability to cause de novo synthesis of HbF by pharmacological manipulation was first shown using 5-azacytidine in experimental animals (DeSimone, Proc Natl Acad Sci U S A. 79(14):4428-31 (1982)). Subsequent studies confirmed the ability of 5-azacytidine to increase HbF in patients with β-thalassemia and sickle cell disease (Ley, et al., N. Engl. J. Medicine, 307: 1469- 1475 (1982), and Ley, et al., Blood 62: 370-380 (1983)). Additional experiments demonstrated that baboons treated with cytotoxic doses of arabinosylcytosine (ara-C) responded with striking elevations of F-reticulocytes (Papayannopoulou et al., Science. 224(4649):617-9 (1984)), and that treatment with hydroxyurea led to induction of γ-globin in monkeys or baboons (Letvin et. al., N Engl J Med.
310(14):869-73 (1984)).
[0264] The second group of compounds investigated for the ability to cause HbF reactivation activity was short chain fatty acids. The initial observation in fetal cord blood progenitor cells led to the discovery that γ-aminobutyric acid can act as a fetal hemoglobin inducer (Perrine et al., Biochem Biophys Res Commun. l48(2):694-700 (1987)). Subsequent studies showed that butyrate stimulated globin production in adult baboons (Constant oulakis et al., Blood. Dec; 72(6): 1961-7 (1988)), and it induced γ-globin in erythroid progenitors in adult animals or patients with sickle cell anemia (Perrine et al., Blood. 74(l):454-9 (1989)). Derivatives of short chain fatty acids such as phenylbutyrate (Dover et al., Br J Haematol. 88(3):555-61 (1994)) and valproic acid (Liakopoulou et al., 1 : Blood. 186(8):3227-35 (1995)) also have been shown to induce HbF in vivo. Given the large number of short chain fatty acid analogs or derivatives of this family, there are a number of potential compounds of this family more potent than butyrate. Phenylacetic and phenylalkyl acids (Torkelson et al., Blood Cells Mol Dis.
22(2): 150-8. (1996)), which were discovered during subsequent studies, were considered potential HbF inducers as they belonged to this family of compounds. Presently, however, the use of butyrate or its analogs in sickle cell anemia and β-thalassemia remains experimental and cannot be recommended for treatment outside of clinical trials.
[0265] Clinical trials aimed at reactivation of fetal hemoglobin synthesis in sickle cell anemia and β -thalassemia have included short term and long term administration of such compounds as 5-azacytidine, hydroxyurea, recombinant human erythropoietin, and butyric acid analogs, as well as combinations of these agents. Following these studies, hydroxyurea was used for induction of HbF in humans and later became the first and only drug approved by the Food and Drug Administration (FDA) for the treatment of hemoglobinopathies. However, varying drawbacks have contraindicated the long term use of such agents or therapies, including unwanted side effects and variability in patient responses. For example, while hydroxyurea stimulates HbF production and has been shown to clinically reduce sickling crisis, it is potentially limited by myelotoxicity and the risk of carcinogenesis. Potential long term carcinogenicity would also exist in 5-azacytidine-based therapies. Erythropoietin-based therapies have not proved consistent among a range of patient populations. The short half-lives of butyric acid in vivo have been viewed as a potential obstacle in adapting these compounds for use in therapeutic interventions.
Furthermore, very high dosages of butyric acid are necessary for inducing γ-globin gene expression, requiring catheritization for continuous infusion of the compound. Moreover, these high dosages of butyric acid can be associated with neurotoxicity and multiorgan damage (Blau, et al., Blood 81: 529-537 (1993)). While even minimal increases in HbF levels are helpful in sickle cell disease, β-thalassemias require a much higher increase that is not reliably, or safely, achieved by any of the currently used agents (Olivieri, Seminars in Hematology 33: 24-42 (1996)).
[0266] Identifying natural regulators of HbF induction and production could provide a means to devise therapeutic interventions that overcome the various drawbacks of the compounds described above. Recent genome-wide association studies have yielded insights into the genetic basis of numerous complex diseases and traits (McCarthy et al., Nat Rev Genet 9, 356 (2008) and Manolio et. al. J Clin Invest 118, 1590 (2008)). However, in the vast majority of instances, the functional link between a genetic association and the underlying pathophysiology remains to be uncovered. The level of fetal hemoglobin (HbF) is inherited as a quantitative trait and clinically important, given its above-mentioned and well-characterized role in ameliorating the severity of the principal β -hemoglobinopathies, sickle cell disease and β -thalassemia (Nathan et. al., Nathan and Oski's hematology of infancy and childhood ed. 6th, pp. 2 v. (xiv, 1864, xli p.) 2003)). Two genome-wide association studies have identified three major loci containing a set of five common single nucleotide polymorphisms (SNPs) that account for -20% of the variation in HbF levels (Lettre et al., Proc Natl Acad Sci U S A (2008); Uda et al., Proc Natl Acad Sci U S A 105, 1620 (2008); Menzel et al., Nat Genet 39, 1197 (2007)). Moreover, several of these variants appear to predict the clinical severity of sickle cell disease (Lettre et al., Proc Natl Acad Sci U S A (2008)) and at least one of these SNPs may also affect clinical outcome in β-thalassemia (Uda et al., Proc Natl Acad Sci U S A 105, 1620 (2008)). The SNP with the largest effect size, explaining over 10% of the variation in HbF, is located in the second intron of a gene on chromosome 2, BCL11A. Whereas BCL11A, a C2H2-type zinc finger transcription factor, has been investigated for its role in lymphocyte development (Liu et al., Nat Immunol 4, 525 (2003) and Liu et al., Mol Cancer 5, 18 (2006)), its role in red blood cell production or globin gene regulation has not been previously assessed.
[0267] At the onset of the recombinant DNA era, studies of globin gene structure provided a strong molecular foundation for interrogating the fetal globin switch. Considerable effort has focused on delineating the cis-elements within the β -globin locus necessary for proper regulation of the genes within the β-like globin cluster. These studies relied on naturally occurring mutations and deletions that dramatically influence HbF levels in adults, and have been complemented by generation of transgenic mice harboring portions of the cluster (Nathan et. al., Nathan and Oski's hematology of infancy and childhood ed. 6th, pp. 2 v. (xiv, 1864, xli p.) 2003) and G. Stamatoyannopoulos, Exp Hematol 33, 259 (2005)). Although the precise cis-elements required for globin switching remain ill-defined, findings in transgenic mice have strongly indicated that the γ-globin genes are autonomously silenced in the adult stage, a finding that is most compatible with the absence of fetal-stage specific activators or the presence of a stage-specific repressor. The results of recent genetic association studies provide candidate genes to interrogate for their involvement in control of the γ-globin genes, such as BCL11A.
Hematopoietic progenitor cells
[0268] In one embodiment, the hematopoietic progenitor cell is contacted ex vivo or in vitro. In a specific embodiment, the cell being contacted is a cell of the erythroid lineage. In one embodiment, the cell composition comprises cells having decreased BCL11A expression.
[0269] "Hematopoietic progenitor cell" as the term is used herein, refers to cells of a stem cell lineage that give rise to all the blood cell types including the myeloid (monocytes and macrophages, neutrophils, basophils, eosinophils, erythrocytes, megakaryocytes/platelets, dendritic cells), and the lymphoid lineages (T-cells, B-cells, NK-cells). A "cell of the erythroid lineage" indicates that the cell being contacted is a cell that undergoes erythropoeisis such that upon final differentiation it forms an erythrocyte or red blood cell (RBC). Such cells belong to one of three lineages, erythroid, lymphoid, and myeloid, originating from bone marrow haematopoietic progenitor cells. Upon exposure to specific growth factors and other components of the haematopoietic microenvironment, haematopoietic progenitor cells can mature through a series of intermediate differentiation cellular types, all intermediates of the erythroid lineage, into RBCs. Thus, cells of the "erythroid lineage", as the term is used herein, comprise hematopoietic progenitor cells, rubriblasts, prorubricytes, erythroblasts, metarubricytes, reticulocytes, and erythrocytes.
[0270] In some embodiment, the hematopoietic progenitor cell has at least one of the cell surface marker characteristic of hematopoietic progenitor cells: CD34+, CD59+, Thyl/CD90+,CD381o/-, and C- kit/CDl 17+. Preferably, the hematopoietic progenitor cells have several of these markers. [0271] In some embodiments, the hematopoietic progenitor cells of the erythroid lineage have the cell surface marker characteristic of the erythroid lineage: CD71 and Terl 19.
[0272] Stem cells, such as hematopoietic progenitor cells, are capable of proliferation and giving rise to more progenitor cells having the ability to generate a large number of mother cells that can in turn give rise to differentiated or differentiable daughter cells. The daughter cells themselves can be induced to proliferate and produce progeny that subsequently differentiate into one or more mature cell types, while also retaining one or more cells with parental developmental potential. The term "stem cell" refers then, to a cell with the capacity or potential, under particular circumstances, to differentiate to a more specialized or differentiated phenotype, and which retains the capacity, under certain circumstances, to proliferate without substantially differentiating. In one embodiment, the term progenitor or stem cell refers to a generalized mother cell whose descendants (progeny) specialize, often in different directions, by differentiation, e.g., by acquiring completely individual characters, as occurs in progressive diversification of embryonic cells and tissues. Cellular differentiation is a complex process typically occurring through many cell divisions. A differentiated cell may derive from a multipotent cell which itself is derived from a multipotent cell, and so on. While each of these multipotent cells may be considered stem cells, the range of cell types each can give rise to may vary considerably. Some differentiated cells also have the capacity to give rise to cells of greater developmental potential. Such capacity may be natural or may be induced artificially upon treatment with various factors. In many biological instances, stem cells are also "multipotent" because they can produce progeny of more than one distinct cell type, but this is not required for "stem-ness." Self-renewal is the other classical part of the stem cell definition, and it is essential as used in this document. In theory, self-renewal can occur by either of two major mechanisms. Stem cells may divide asymmetrically, with one daughter retaining the stem state and the other daughter expressing some distinct other specific function and phenotype.
Alternatively, some of the stem cells in a population can divide symmetrically into two stems, thus maintaining some stem cells in the population as a whole, while other cells in the population give rise to differentiated progeny only. Generally, "progenitor cells" have a cellular phenotype that is more primitive (i.e., is at an earlier step along a developmental pathway or progression than is a fully differentiated cell). Often, progenitor cells also have significant or very high proliferative potential. Progenitor cells can give rise to multiple distinct differentiated cell types or to a single differentiated cell type, depending on the developmental pathway and on the environment in which the cells develop and differentiate.
[0273] In the context of cell ontogeny, the adjective "differentiated", or "differentiating" is a relative term. A "differentiated cell" is a cell that has progressed further down the developmental pathway than the cell it is being compared with. Thus, stem cells can differentiate to lineage-restricted precursor cells (such as a hematopoietic progenitor cell), which in turn can differentiate into other types of precursor cells further down the pathway (such as an erthyrocyte precursor), and then to an end-stage differentiated cell, such as an erthyrocyte, which plays a characteristic role in a certain tissue type, and may or may not retain the capacity to proliferate further.
Induced Pluripotent Stem Cells
[0274] In some embodiments, the genetic engineered human cells described herein are derived from isolated pluripotent stem cells. An advantage of using iPSCs is that the cells can be derived from the same subject to which the progenitor cells are to be administered. That is, a somatic cell can be obtained from a subject, reprogrammed to an induced pluripotent stem cell, and then re-differentiated into a hematopoietic progenitor cell to be administered to the subject {e.g., autologous cells). Since the progenitors are essentially derived from an autologous source, the risk of engraftment rejection or allergic responses is reduced compared to the use of cells from another subject or group of subjects. In some embodiments, the hematopoeitic progenitors are derived from non-autologous sources. In addition, the use of iPSCs negates the need for cells obtained from an embryonic source. Thus, in one embodiment, the stem cells used in the disclosed methods are not embryonic stem cells.
[0275] Although differentiation is generally irreversible under physiological contexts, several methods have been recently developed to reprogram somatic cells to induced pluripotent stem cells. Exemplary methods are known to those of skill in the art and are described briefly herein below.
[0276] As used herein, the term "reprogramming" refers to a process that alters or reverses the differentiation state of a differentiated cell {e.g., a somatic cell). Stated another way, reprogramming refers to a process of driving the differentiation of a cell backwards to a more undifferentiated or more primitive type of cell. It should be noted that placing many primary cells in culture can lead to some loss of fully differentiated characteristics. Thus, simply culturing such cells included in the term
differentiated cells does not render these cells non-differentiated cells {e.g., undifferentiated cells) or pluripotent cells. The transition of a differentiated cell to pluripotency requires a reprogramming stimulus beyond the stimuli that lead to partial loss of differentiated character in culture. Reprogrammed cells also have the characteristic of the capacity of extended passaging without loss of growth potential, relative to primary cell parents, which generally have capacity for only a limited number of divisions in culture.
[0277] The cell to be reprogrammed can be either partially or terminally differentiated prior to reprogramming. In some embodiments, reprogramming encompasses complete reversion of the differentiation state of a differentiated cell {e.g., a somatic cell) to a pluripotent state or a multipotent state. In some embodiments, reprogramming encompasses complete or partial reversion of the differentiation state of a differentiated cell {e.g., a somatic cell) to an undifferentiated cell {e.g., an embryonic-like cell). Reprogramming can result in expression of particular genes by the cells, the expression of which further contributes to reprogramming. In certain embodiments described herein, reprogramming of a differentiated cell {e.g., a somatic cell) causes the differentiated cell to assume an undifferentiated state (e.g., is an undifferentiated cell). The resulting cells are referred to as "reprogrammed cells," or "induced pluripotent stem cells (iPSCs or iPS cells)."
[0278] Reprogramming can involve alteration, e.g., reversal, of at least some of the heritable patterns of nucleic acid modification (e.g., methylation), chromatin condensation, epigenetic changes, genomic imprinting, etc., that occur during cellular differentiation. Reprogramming is distinct from simply maintaining the existing undifferentiated state of a cell that is already pluripotent or maintaining the existing less than fully differentiated state of a cell that is already a multipotent cell (e.g., a hematopoietic stem cell). Reprogramming is also distinct from promoting the self-renewal or proliferation of cells that are already pluripotent or multipotent, although the compositions and methods described herein can also be of use for such purposes, in some embodiments.
[0279] The specific approach or method used to generate pluripotent stem cells from somatic cells (broadly referred to as "reprogramming") is not critical to the claimed invention. Thus, any method that re-programs a somatic cell to the pluripotent phenotype would be appropriate for use in the methods described herein.
[0280] Reprogramming methodologies for generating pluripotent cells using defined combinations of transcription factors have been described induced pluripotent stem cells. Yamanaka and Takahashi converted mouse somatic cells to ES cell-like cells with expanded developmental potential by the direct transduction of Oct4, Sox2, Klf4, and c-Myc (Takahashi and Yamanaka, 2006). iPSCs resemble ES cells as they restore the pluripotency-associated transcriptional circuitry and muc of the epigenetic landscape. In addition, mouse iPSCs satisfy all the standard assays for pluripotency: specifically, in vitro differentiation into cell types of the three germ layers, teratoma formation, contribution to chimeras, germline transmission (Maherali and Hochedlinger, 2008), and tetraploid complementation (Woltjen et al., 2009).
[0281] Subsequent studies have shown that human iPS cells can be obtained using similar transduction methods (Lowry et al., 2008; Park et al., 2008; Takahashi et al., 2007; Yu et al., 2007b), and the transcription factor trio, OCT4, SOX2, and NANOG, has been established as the core set of transcription factors that govern pluripotency (Jaenisch and Young, 2008). The production of iPS cells can be achieved by the introduction of nucleic acid sequences encoding stem cell-associated genes into an adult, somatic cell, historically using viral vectors.
[0282] iPS cells can be generated or derived from terminally differentiated somatic cells, as well as from adult stem cells, or somatic stem cells. That is, a non-pluripotent progenitor cell can be rendered pluripotent or multipotent by reprogramming. In such instances, it may not be necessary to include as many reprogramming factors as required to reprogram a terminally differentiated cell. Further, reprogramming can be induced by the non- viral introduction of reprogramming factors, e.g., by introducing the proteins themselves, or by introducing nucleic acids that encode the reprogramming factors, or by introducing messenger RNAs that upon translation produce the reprogramming factors (see e.g., Warren et al., Cell Stem Cell, 2010 Nov 5;7(5):618-30). Reprogramming can be achieved by introducing a combination of nucleic acids encoding stem cell-associated genes including, for example Oct-4 (also known as Oct-3/4 or Pouf51), Soxl, Sox2, Sox3, Sox 15, Sox 18, NANOG, , Klfl, Klf2, Klf4, Klf5, NR5A2, c-Myc, 1-Myc, n-Myc, Rem2, Tert, and LIN28. In one embodiment, reprogramming using the methods and compositions described herein can further comprise introducing one or more of Oct-3/4, a member of the Sox family, a member of the Klf family, and a member of the Myc family to a somatic cell. In one embodiment, the methods and compositions described herein further comprise introducing one or more of each of Oct 4, Sox2, Nanog, c-MYC and Klf4 for reprogramming. As noted above, the exact method used for reprogramming is not necessarily critical to the methods and compositions described herein. However, where cells differentiated from the reprogrammed cells are to be used in, e.g., human therapy, in one embodiment the reprogramming is not effected by a method that alters the genome. Thus, in such embodiments, reprogramming is achieved, e.g., without the use of viral or plasmid vectors.
[0283] The efficiency of reprogramming (i.e., the number of reprogrammed cells) derived from a population of starting cells can be enhanced by the addition of various small molecules as shown by Shi, Y., et al (2008) Cell-Stem Cell 2:525-528, Huangfu, D., et al (2008) Nature Biotechnology 26(7):795- 797, and Marson, A., et al (2008) Cell-Stem Cell 3: 132-135. Thus, an agent or combination of agents that enhance the efficiency or rate of induced pluripotent stem cell production can be used in the production of patient-specific or disease-specific iPSCs. Some non-limiting examples of agents that enhance reprogramming efficiency include soluble Wnt, Wnt conditioned media, BIX-01294 (a G9a histone methyltransferase), PD0325901 (a MEK inhibitor), DNA methyltransferase inhibitors, histone deacetylase (HDAC) inhibitors, valproic acid, 5'-azacytidine, dexamethasone, suberoylanilide, hydroxamic acid (SAHA), vitamin C, and trichostatin (TSA), among others.
[0284] Other non-limiting examples of reprogramming enhancing agents include: Suberoylanilide Hydroxamic Acid (SAHA (e.g., MK0683, vorinostat) and other hydroxamic acids), BML-210,
Depudecin (e.g., (-)-Depudecin), HC Toxin, Nullscript (4-(l,3-Dioxo-lH,3H-benzo[de]isoquinolin-2-yl)- N-hydroxybutanamide), Phenylbutyrate (e.g., sodium phenylbutyrate) and Valproic Acid ((VP A) and other short chain fatty acids), Scriptaid, Suramin Sodium, Trichostatin A (TSA), APHA Compound 8, Apicidin, Sodium Butyrate, pivaloyloxymethyl butyrate (Pivanex, AN-9), Trapoxin B, Chlamydocin, Depsipeptide (also known as FR901228 or FK228), benzamides (e.g., CI-994 (e.g., N-acetyl dinaline) and MS-27-275), MGCD0103, NVP-LAQ-824, CBHA (m-carboxycinnaminic acid bishydroxamic acid), JNJ16241199, Tubacin, A-161906, proxamide, oxamflatin, 3-Cl-UCHA (e.g., 6-(3- chlorophenylureido)caproic hydroxamic acid), AOE (2-amino-8-oxo-9, 10-epoxydecanoic acid), CHAP31 and CHAP 50. Other reprogramming enhancing agents include, for example, dominant negative forms of the HDACs (e.g., catalytically inactive forms), siRNA inhibitors of the HDACs, and antibodies that specifically bind to the HDACs. Such inhibitors are available, e.g., from BIOMOL International, Fukasawa, Merck Biosciences, Novartis, Gloucester Pharmaceuticals, Aton Pharma, Titan Pharmaceuticals, Schering AG, Pharmion, MethylGene, and Sigma Aldrich.
[0285] To confirm the induction of pluripotent stem cells for use with the methods described herein, isolated clones can be tested for the expression of a stem cell marker. Such expression in a cell derived from a somatic cell identifies the cells as induced pluripotent stem cells. Stem cell markers can be selected from the non-limiting group including SSEA3, SSEA4, CD9, Nanog, Fbxl5, Ecatl, Esgl, Eras, Gdf3, Fgf4, Cripto, Daxl, Zpf296, Slc2a3, Rexl, Utfl, and Natl. In one embodiment, a cell that expresses Oct4 or Nanog is identified as pluripotent. Methods for detecting the expression of such markers can include, for example, RT-PCR and immunological methods that detect the presence of the encoded polypeptides, such as Western blots or flow cytometric analyses. In some embodiments, detection does not involve only RT-PCR, but also includes detection of protein markers. Intracellular markers may be best identified via RT-PCR, while cell surface markers are readily identified, e.g., by immunocytochemistry.
[0286] The pluripotent stem cell character of isolated cells can be confirmed by tests evaluating the ability of the iPSCs to differentiate to cells of each of the three germ layers. As one example, teratoma formation in nude mice can be used to evaluate the pluripotent character of the isolated clones. The cells are introduced to nude mice and histology and/or immunohistochemistry is performed on a tumor arising from the cells. The growth of a tumor comprising cells from all three germ layers, for example, further indicates that the cells are pluripotent stem cells.
Somatic Cells for reprogramming
[0287] Somatic cells, as that term is used herein, refer to any cells forming the body of an organism, excluding germline cells. Every cell type in the mammalian body— apart from the sperm and ova, the cells from which they are made (gametocytes) and undifferentiated stem cells— is a
differentiated somatic cell. For example, internal organs, skin, bones, blood, and connective tissue are all made up of differentiated somatic cells.
[0288] Additional somatic cell types for use with the compositions and methods described herein include: a fibroblast {e.g., a primary fibroblast), a muscle cell {e.g., a myocyte), a cumulus cell, a neural cell, a mammary cell, an hepatocyte and a pancreatic islet cell. In some embodiments, the somatic cell is a primary cell line or is the progeny of a primary or secondary cell line. In some embodiments, the somatic cell is obtained from a human sample, e.g., a hair follicle, a blood sample, a biopsy {e.g., a skin biopsy or an adipose biopsy), a swab sample {e.g., an oral swab sample), and is thus a human somatic cell.
[0289] Some non-limiting examples of differentiated somatic cells include, but are not limited to, epithelial, endothelial, neuronal, adipose, cardiac, skeletal muscle, immune cells, hepatic, splenic, lung, circulating blood cells, gastrointestinal, renal, bone marrow, and pancreatic cells. In some embodiments, a somatic cell can be a primary cell isolated from any somatic tissue including, but not limited to brain, liver, gut, stomach, intestine, fat, muscle, uterus, skin, spleen, endocrine organ, bone, etc. Further, the somatic cell can be from any mammalian species, with non-limiting examples including a murine, bovine, simian, porcine, equine, ovine, or human cell. In some embodiments, the somatic cell is a human somatic cell.
[0290] When reprogrammed cells are used for generation of hematopoietic progenitor cells to be used in the therapeutic treatment of disease, it is desirable, but not required, to use somatic cells isolated from the patient being treated. For example, somatic cells involved in diseases, and somatic cells participating in therapeutic treatment of diseases and the like can be used. In some embodiments, a method for selecting the reprogrammed cells from a heterogeneous population comprising reprogrammed cells and somatic cells they were derived or generated from can be performed by any known means. For example, a drug resistance gene or the like, such as a selectable marker gene can be used to isolate the reprogrammed cells using the selectable marker as an index.
[0291] Reprogrammed somatic cells as disclosed herein can express any number of pluripotent cell markers, including: alkaline phosphatase (AP); ABCG2; stage specific embryonic antigen-1 (SSEA-1); SSEA-3; SSEA-4; TRA-1-60; TRA-1-81; Tra-2-49/6E; ERas ECAT5, E-cadherin; β-ΙΙΙ-tubulin; ct- smooth muscle actin (ct-SMA); fibroblast growth factor 4 (Fgf4), Cripto, Daxl; zinc finger protein 296 (Zfp296); N-acetyltransferase-1 (Natl); (ES cell associated transcript 1 (ECAT1);
ESG1 DPPA5 ECAT2; ECAT3; ECAT6; ECAT7; ECAT8; ECAT9; ECAT10; ECAT15-1; ECAT15-2; Fthll7; Sall4; undifferentiated embryonic cell transcription factor (Utfl); Rexl; p53; G3PDH;
telomerase, including TERT; silent X chromosome genes; Dnmt3a; Dnmt3b; TRIM28; F-box containing protein 15 (Fbxl5); Nanog ECAT4; Oct3/4; Sox2; Klf4; c-Myc; Esrrb; TDGF1; GABRB3; Zfp42, FoxD3; GDF3; CYP25A1; developmental pluripotency-associated 2 (DPPA2); T-cell lymphoma breakpoint 1 (Tell); DPP A3 /Stella; DPPA4; other general markers for pluripotency, etc. Other markers can include Dnmt3L; Soxl5; Stat3; Grb2; β-catenin, and Bmil. Such cells can also be characterized by the down-regulation of markers characteristic of the somatic cell from which the induced pluripotent stem cell is derived.
Genome editing and DNA-targeting endonucleases
[0292] As used herein, the term "genome editing" refers to a reverse genetics method using artificially engineered nucleases to cut and create specific double-stranded breaks at a desired location(s) in the genome, which are then repaired by cellular endogenous processes such as, homologous recombination (FfR), homology directed repair (FfDR) and non-homologous end-joining (NFfEJ). NFfEJ directly joins the DNA ends in a double-stranded break, while FfDR utilizes a homologous sequence as a template for regenerating the missing DNA sequence at the break point.
[0293] Genome editing cannot be performed using traditional restriction endonucleases since most restriction enzymes recognize a few base pairs on the DNA as their target and the probability is very high that the recognized base pair combination will be found in many locations across the genome resulting in multiple cuts (i.e., not limited to a desired location). To overcome this challenge and create site-specific double-stranded breaks, several distinct classes of nucleases have been discovered and bioengineered to date. These are the meganucleases, Zinc finger nucleases (ZFNs), Cas9/CRISPR system, and
transcription-activator like effector nucleases (TALENs).
[0294] Meganucleases are commonly grouped into four families: the LAGLIDADG family, the GIY-YIG family, the His-Cys box family and the HNH family. These families are characterized by structural motifs, which affect catalytic activity and recognition sequence. For instance, members of the LAGLIDADG family are characterized by having either one or two copies of the conserved
LAGLIDADG motif (see Chevalier et al. (2001), Nucleic Acids Res. 29(18): 3757-3774). The
LAGLIDADG meganucleases with a single copy of the LAGLIDADG motif form homodimers, whereas members with two copies of the LAGLIDADG are found as monomers. Similarly, the GIY-YIG family members have a GIY-YIG module, which is 70-100 residues long and includes four or five conserved sequence motifs with four invariant residues, two of which are required for activity (see Van Roey et al. (2002), Nature Struct. Biol. 9: 806-811). The His-Cys box meganucleases are characterized by a highly conserved series of histidines and cysteines over a region encompassing several hundred amino acid residues (see Chevalier et al. (2001), Nucleic Acids Res. 29(18): 3757-3774). In the case of the NHN family, the members are defined by motifs containing two pairs of conserved histidines surrounded by asparagine residues (see Chevalier et al. (2001), Nucleic Acids Res. 29(18): 3757-3774). The four families of meganucleases are widely separated from one another with respect to conserved structural elements and, consequently, DNA recognition sequence specificity and catalytic activity.
[0295] Meganucleases are found commonly in microbial species and have the unique property of having very long recognition sequences (>14bp) thus making them naturally very specific for cutting at a desired location. This can be exploited to make site-specific double-stranded breaks in genome editing. One of skill in the art can use these naturally occurring meganucleases, however the number of such naturally occurring meganucleases is limited. To overcome this challenge, mutagenesis and high throughput screening methods have been used to create meganuclease variants that recognize unique sequences. For example, various meganucleases have been fused to create hybrid enzymes that recognize a new sequence. Alternatively, DNA interacting amino acids of the meganuclease can be altered to design sequence specific meganucleases (see e.g., US Patent 8,021,867). Meganucleases can be designed using the methods described in e.g., Certo, MT et al. Nature Methods (2012) 9:073-975; U.S. Patent Nos. 8,304,222; 8,021,867; 8, 119,381; 8, 124,369; 8, 129,134; 8,133,697; 8,143,015; 8,143,016; 8, 148,098; or 8, 163,514, the contents of each are incorporated herein by reference in their entirety. Alternatively, meganucleases with site specific cutting characteristics can be obtained using commercially available technologies e.g., Precision Biosciences' Directed Nuclease Editor™ genome editing technology.
[0296] ZFNs and TALENs restriction endonuclease technology utilizes a non-specific DNA cutting enzyme which is linked to a specific DNA sequence recognizing peptide(s) such as zinc fingers and transcription activator-like effectors (TALEs). Typically an endonuclease whose DNA recognition site and cleaving site are separate from each other is selected and the its cleaving portion is separated and then linked to a sequence recognizing peptide, thereby yielding an endonuclease with very high specificity for a desired sequence. An exemplary restriction enzyme with such properties is Fokl.
Additionally Fokl has the advantage of requiring dimerization to have nuclease activity and this means the specificity increases dramatically as each nuclease partner recognizes a unique DNA sequence. To enhance this effect, Fokl nucleases have been engineered that can only function as heterodimers and have increased catalytic activity. The heterodimer functioning nucleases avoid the possibility of unwanted homodimer activity and thus increase specificity of the double-stranded break.
[0297] Although the nuclease portions of both ZFNs and TALENs have similar properties, the difference between these engineered nucleases is in their DNA recognition peptide. ZFNs rely on Cys2- His2 zinc fingers and TALENs on TALEs. Both of these DNA recognizing peptide domains have the characteristic that they are naturally found in combinations in their proteins. Cys2-His2 Zinc fingers typically happen in repeats that are 3 bp apart and are found in diverse combinations in a variety of nucleic acid interacting proteins such as transcription factors. TALEs on the other hand are found in repeats with a one-to-one recognition ratio between the amino acids and the recognized nucleotide pairs. Because both zinc fingers and TALEs happen in repeated patterns, different combinations can be tried to create a wide variety of sequence specificities. Approaches for making site-specific zinc finger endonucleases include, e.g., modular assembly (where Zinc fingers correlated with a triplet sequence are attached in a row to cover the required sequence), OPEN (low-stringency selection of peptide domains vs. triplet nucleotides followed by high-stringency selections of peptide combination vs. the final target in bacterial systems), and bacterial one-hybrid screening of zinc finger libraries, among others. ZFNs for use with the methods and compositions described herein can be obtained commercially from e.g., Sangamo Biosciences™ (Richmond, CA).
[0298] It is contemplated herein that the Cas9/CRISPR system of genome editing be employed with the methods and compositions described herein. Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated (Cas) systems is useful for RNA-programmable genome editing (see e.g., Jinek, M. et al. Science (2012) 337(6096):816-821).
[0299] Alternatively, genome editing can be performed using recombinant adeno-associated virus (rAAV) based genome engineering, which is a genome-editing platform centered around the use of rAAV vectors that enables insertion, deletion or substitution of DNA sequences into the genomes of live mammalian cells. The rAAV genome is a single-stranded deoxyribonucleic acid (ssDNA) molecule, either positive- or negative-sensed, which is about 4.7 kilobase long. These single-stranded DNA viral vectors have high transduction rates and have a unique property of stimulating endogenous homologous recombination in the absence of causing double strand DNA breaks in the genome. One of skill in the art can design a rAAV vector to target a desired genomic locus and perform both gross and/or subtle endogenous gene alterations in a cell, such as a deletion. rAAV genome editing has the advantage in that it targets a single allele and does not result in any off-target genomic alterations. rAAV genome editing technology is commercially available, for example, the rAAV GENESIS™ system from Horizon™ (Cambridge, UK).
Pharmaceutically Acceptable Carriers
[0300] The methods of administering human hematopoietic progenitors to a subject as described herein involve the use of therapeutic compositions comprising hematopoietic progenitor cells.
Therapeutic compositions contain a physiologically tolerable carrier together with the cell composition and optionally at least one additional bioactive agent as described herein, dissolved or dispersed therein as an active ingredient. In a preferred embodiment, the therapeutic composition is not substantially immunogenic when administered to a mammal or human patient for therapeutic purposes, unless so desired.
[0301] In general, the hematopoietic progenitor cells described herein are administered as a suspension with a pharmaceutically acceptable carrier. One of skill in the art will recognize that a pharmaceutically acceptable carrier to be used in a cell composition will not include buffers, compounds, cryopreservation agents, preservatives, or other agents in amounts that substantially interfere with the viability of the cells to be delivered to the subject. A formulation comprising cells can include e.g., osmotic buffers that permit cell membrane integrity to be maintained, and optionally, nutrients to maintain cell viability or enhance engraftment upon administration. Such formulations and suspensions are known to those of skill in the art and/or can be adapted for use with the hematopoietic progenitor cells as described herein using routine experimentation.
[0302] A cell composition can also be emulsified or presented as a liposome composition, provided that the emulsification procedure does not adversely affect cell viability. The cells and any other active ingredient can be mixed with excipients which are pharmaceutically acceptable and compatible with the active ingredient and in amounts suitable for use in the therapeutic methods described herein.
[0303] Additional agents included in a cell composition as described herein can include pharmaceutically acceptable salts of the components therein. Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the polypeptide) that are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, tartaric, mandelic and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine and the like. Physiologically tolerable carriers are well known in the art. Exemplary liquid carriers are sterile aqueous solutions that contain no materials in addition to the active ingredients and water, or contain a buffer such as sodium phosphate at physiological pH value, physiological saline or both, such as phosphate-buffered saline. Still further, aqueous carriers can contain more than one buffer salt, as well as salts such as sodium and potassium chlorides, dextrose, polyethylene glycol and other solutes. Liquid compositions can also contain liquid phases in addition to and to the exclusion of water. Exemplary of such additional liquid phases are glycerin, vegetable oils such as cottonseed oil, and water-oil emulsions. The amount of an active compound used in the cell compositions as described herein that is effective in the treatment of a particular disorder or condition will depend on the nature of the disorder or condition, and can be determined by standard clinical techniques.
Administration & Efficacy
[0304] As used herein, the terms "administering," "introducing" and "transplanting" are used interchangeably in the context of the placement of cells, e.g. hematopoietic progenitor cells, as described herein into a subject, by a method or route which results in at least partial localization of the introduced cells at a desired site, such as a site of injury or repair, such that a desired effect(s) is produced. The cells e.g. hematopoietic progenitor cells, or their differentiated progeny can be administered by any appropriate route which results in delivery to a desired location in the subject where at least a portion of the implanted cells or components of the cells remain viable. The period of viability of the cells after administration to a subject can be as short as a few hours, e.g., twenty-four hours, to a few days, to as long as several years, i.e., long-term engraftment. For example, in some embodiments of the aspects described herein, an effective amount of hematopoietic progenitor cells is administered via a systemic route of administration, such as an intraperitoneal or intravenous route.
[0305] When provided prophylactically, hematopoietic progenitor cells described herein can be administered to a subject in advance of any symptom of a hemoglobinopathy, e.g., prior to the switch from fetal γ-globin to predominantly β-globin. Accordingly, the prophylactic administration of a hematopoietic progenitor cell population serves to prevent a hemoglobinopathy, as disclosed herein.
[0306] When provided therapeutically, hematopoietic progenitor cells are provided at (or after) the onset of a symptom or indication of a hemoglobinopathy, e.g., upon the onset of sickle cell disease.
[0307] In some embodiments of the aspects described herein, the hematopoietic progenitor cell population being administered according to the methods described herein comprises allogeneic hematopoietic progenitor cells obtained from one or more donors. As used herein, "allogeneic" refers to a hematopoietic progenitor cell or biological samples comprising hematopoietic progenitor cells obtained from one or more different donors of the same species, where the genes at one or more loci are not identical. For example, a hematopoietic progenitor cell population being administered to a subject can be derived from umbilical cord blood obtained from one more unrelated donor subjects, or from one or more non-identical siblings. In some embodiments, syngeneic hematopoietic progenitor cell populations can be used, such as those obtained from genetically identical animals, or from identical twins. In other embodiments of this aspect, the hematopoietic progenitor cells are autologous cells; that is, the hematopoietic progenitor cells are obtained or isolated from a subject and administered to the same subject, i.e., the donor and recipient are the same.
[0308] For use in the various aspects described herein, an effective amount of hematopoietic progenitor cells, comprises at least 102 hematopoietic progenitor cells, at least 5 X 102 hematopoietic progenitor cells, at least 103 hematopoietic progenitor cells, at least 5 X 103 hematopoietic progenitor cells, at least 10 hematopoietic progenitor cells, at least 5 X 10 hematopoietic progenitor cells, at least 105 hematopoietic progenitor cells, at least 2 X 105 hematopoietic progenitor cells, at least 3 X 105 hematopoietic progenitor cells, at least 4 X 105 hematopoietic progenitor cells, at least 5 X 105 hematopoietic progenitor cells, at least 6 X 105 hematopoietic progenitor cells, at least 7 X 105 hematopoietic progenitor cells, at least 8 X 105 hematopoietic progenitor cells, at least 9 X 105 hematopoietic progenitor cells, at least 1 X 106 hematopoietic progenitor cells, at least 2 X 106 hematopoietic progenitor cells, at least 3 X 106 hematopoietic progenitor cells, at least 4 X 106 hematopoietic progenitor cells, at least 5 X 106 hematopoietic progenitor cells, at least 6 X 106 hematopoietic progenitor cells, at least 7 X 106 hematopoietic progenitor cells, at least 8 X 106 hematopoietic progenitor cells, at least 9 X 106 hematopoietic progenitor cells, or multiples thereof. The hematopoietic progenitor cells can be derived from one or more donors, or can be obtained from an autologous source. In some embodiments of the aspects described herein, the hematopoietic progenitor cells are expanded in culture prior to administration to a subject in need thereof.
[0309] In one embodiment, the term "effective amount" as used herein refers to the amount of a population of human hematopoietic progenitor cells or their progeny needed to alleviate at least one or more symptom of a hemoglobinopathy, and relates to a sufficient amount of a composition to provide the desired effect, e.g., treat a subject having a hemoglobinopathy. The term "therapeutically effective amount" therefore refers to an amount of hematopoietic progenitor cells or a composition comprising hematopoietic progenitor cells that is sufficient to promote a particular effect when administered to a typical subject, such as one who has or is at risk for a hemoglobinopathy. An effective amount as used herein would also include an amount sufficient to prevent or delay the development of a symptom of the disease, alter the course of a symptom disease (for example but not limited to, slow the progression of a symptom of the disease), or reverse a symptom of the disease. It is understood that for any given case, an appropriate "effective amount" can be determined by one of ordinary skill in the art using routine experimentation.
[0310] As used herein, "administered" refers to the delivery of a hematopoietic stem cell composition as described herein into a subject by a method or route which results in at least partial localization of the cell composition at a desired site. A cell composition can be administered by any appropriate route which results in effective treatment in the subject, i.e. administration results in delivery to a desired location in the subject where at least a portion of the composition delivered, i.e. at least 1 x 104 cells are delivered to the desired site for a period of time. Modes of administration include injection, infusion, instillation, or ingestion. "Injection" includes, without limitation, intravenous, intramuscular, intra-arterial, intrathecal, intraventricular, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, sub capsular, subarachnoid, intraspinal, intracerebro spinal, and intrasternal injection and infusion. For the delivery of cells, administration by injection or infusion is generally preferred. [0311] In one embodiment, the cells as described herein are administered systemically. The phrases "systemic administration," "administered systemically", "peripheral administration" and "administered peripherally" as used herein refer to the administration of a population of hematopoietic progenitor cells other than directly into a target site, tissue, or organ, such that it enters, instead, the subject's circulatory system and, thus, is subject to metabolism and other like processes.
[0312] The efficacy of a treatment comprising a composition as described herein for the treatment of a hemoglobinopathy can be determined by the skilled clinician. However, a treatment is considered "effective treatment," as the term is used herein, if any one or all of the signs or symptoms of, as but one example, levels of fetal β-globin are altered in a beneficial manner, other clinically accepted symptoms or markers of disease are improved or ameliorated, e.g., by at least 10% following treatment with an inhibitor. Efficacy can also be measured by failure of an individual to worsen as assessed by
hospitalization or need for medical interventions (e.g., progression of the disease is halted or at least slowed). Methods of measuring these indicators are known to those of skill in the art and/or described herein. Treatment includes any treatment of a disease in an individual or an animal (some non-limiting examples include a human, or a mammal) and includes: (1) inhibiting the disease, e.g., arresting, or slowing the progression of sepsis; or (2) relieving the disease, e.g., causing regression of symptoms; and (3) preventing or reducing the likelihood of the development of infection or sepsis.
[0313] The treatment according to the present invention ameliorates one or more symptoms associated with a β-globin disorder by increasing the amount of fetal hemoglobin in the individual. Symptoms typically associated with a hemoglobinopathy, include for example, anemia, tissue hypoxia, organ dysfunction, abnormal hematocrit values, ineffective erythropoiesis, abnormal reticulocyte (erythrocyte) count, abnormal iron load, the presence of ring sideroblasts, splenomegaly, hepatomegaly, impaired peripheral blood flow, dyspnea, increased hemolysis, jaundice, anemic pain crises, acute chest syndrome, splenic sequestration, priapism, stroke, hand-foot syndrome, and pain such as angina pectoris.
[0314] In one embodiment, the hematopoietic progenitor cell is contacted ex vivo or in vitro with a DNA targeting endonuclease, and the cell or its progeny is administered to the mammal (e.g., human). In a further embodiment, the hematopoietic progenitor cell is a cell of the erythroid lineage. In one embodiment, a composition comprising a hematopoietic progenitor cell that was previously contacted with a DNA-targeting endonuclease and a pharmaceutically acceptable carrier and is administered to a mammal.
[0315] In one embodiment, any method known in the art can be used to measure an increase in fetal hemoglobin expression, e.g., Western Blot analysis of fetal hemoglobin protein and quantifying mRNA of fetal γ-globin.
[0316] In one embodiment, the hematopoietic progenitor cell is contacted with a DNA-targeting endonuclease in vitro, or ex vivo. In one embodiment, the cell is of human origin (e.g., an autologous or heterologous cell). In one embodiment, the composition causes an increase in fetal hemoglobin expression.
[0317] The present invention can be defined in any of the following alphabetized paragraphs:
[A] A method for producing a progenitor cell having decreased BCL 11 A mRNA or protein expression, the method comprising contacting an isolated progenitor cell with an agent that binds the genomic DNA of the cell on chromosome 2 location 60,716,189-60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly), thereby reducing the mRNA or protein expression of BCL 11 A.
[B] A method for producing an isolated genetic engineered human cell having at least one genetic modification comprising contacting an isolated cell with an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA-targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location 60,716,189-60,728,612 causing at least one genetic modification therein.
[C] The method of paragraph [A] or [B], wherein the isolated progenitor cell or isolated cell is a hematopoietic progenitor cell.
[D] The method of paragraph [C], wherein the hematopoietic progenitor is a cell of the erythroid lineage.
[E] The method of paragraph [A] or [B], wherein the isolated progenitor cell or isolated cell is an induced pluripotent stem cell.
[F] The method of paragraph [C], wherein the hematopoietic progenitor cell is contacted ex vivo or in vitro.
[G] The method of any one of paragraphs [A]-[F], wherein the at least one genetic modification is a deletion.
[H] The method of paragraph [G], wherein the deletion removes the entire region between chromosome 2 location 60,716, 189-60,728,612 or removes a portion of the region resulting in disruption of one of more DNAse 1 -hypersensitive sites (DHS).
[I] An isolated genetic engineered human cell having at least one genetic modification on chromosome 2 location 60,716, 189-60,728,612 according to paragraphs [B]-[H].
[J] A composition comprising isolated genetic engineered human cells of paragraph [I].
[K] A method of increasing fetal hemoglobin levels in a cell, the method comprising the steps of: contacting an isolated cell with an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA-targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location 60,716, 189-60,728,612 causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in said cell, or its progeny, relative to said cell prior to said contacting.
[L] The method of paragraph [K], wherein the isolated cell is a hematopoietic progenitor cell.
[M] The method of paragraph [K] or [L], wherein the hematopoietic progenitor cell is a cell of the erythroid lineage.
[N] The method of paragraph [K], wherein the isolated cell is an induced pluripotent stem cell.
[O] The method of paragraph [L] or [M], wherein the hematopoietic progenitor cell is contacted ex vivo or in vitro.
[P] The method of any one of paragraphs [K]-[0], wherein the at least one genetic modification is a deletion.
[Q] The method of paragraph [P], wherein the deletion removes the entire region between chromosome 2 location 60,716, 189-60,728,612 or removes a portion of the region resulting in disruption of one of more DNAse 1 -hypersensitive sites (DHS).
[R] A method for increasing fetal hemoglobin levels in a mammal in need thereof, the method comprising the steps of contacting an isolated hematopoietic progenitor cell in said mammal with an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA-targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location 60,716,189-60,728,612 causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in said mammal, relative to expression prior to the contacting.
[0318] This invention is further illustrated by the following example which should not be construed as limiting. The contents of all references cited throughout this application, as well as the figures and table are incorporated herein by reference.
EXAMPLE
[0319] The inventors have discovered and characterized regulatory elements of the BCL 11 A gene that are critical for its expression in erythroid lineage cells. Common genetic variants within these sequences are associated with fetal hemoglobin level and beta-globin disorder severity. These sequences comprise distal regulatory elements with an enhancer chromatin signature, possessing accessible chromatin, active histone marks, and occupancy by erythroid transcription factors. These elements interact with the BCLl 1A promoter and promote gene expression in erythroid cells but not other lineages that express BCLl 1A such as B-lymphocytes. These regulatory elements can be targeted for therapeutic purposes to achieve BCLl 1A inhibition and fetal hemoglobin reinduction. This can be achieved by mechanisms not limited to genome editing, nucleic acid or protein binding, and epigenetic modification. Advantages of this method include: disruption of a physiologic regulator of fetal hemoglobin level resulting in increased gamma-globin production and reduced beta-globin production; minimal effect on overall globin output or on red blood cell production or function; limitation of impact on cells outside of the erythroid lineage thus reducing potential toxicity.
Materials and Methods
Cell culture
[0320] Human CD34+ cells from mobilized peripheral blood of healthy donors were obtained from Centers of Excellence in Molecular Hematology at Yale University, New Haven, Connecticut and Fred Hutchinson Cancer Research Center, Seattle, Washington. The cells were subject to ex vivo erythroid maturation with a two-phase serum-free liquid culture protocol as previously described (39). Peripheral blood mononuclear cells (PBMCs) were obtained from healthy donors from Boston Children's Hospital. Erythroid differentiation from PBMCs was performed as previously described (40). Mouse
erythroleukemia (MEL) cells and 293T cells were cultured as previously described (39). Stably v-Abl transformed pre-B lymphocyte murine cells (derived as described (41)) were cultured in RPMI plus 2% penicillin-streptomycin, 15% FCS, 2% HEPES, 1% non-essential amino acids, 1% sodium pyruvate, 1% L-glutamine and 100 μΜ β-mercaptoethanol.
CMP and DNase I sensitivity
[0321] Chromatin immunoprecipitation and massively parallel sequencing were performed as described (39). The following antibodies were used: H3K27me3 (Millipore, 07-449), H3K4me3 (Millipore, 04-745), H3K4mel (Abeam, ab8895), H3K27ac (Abeam, ab4729), RNA Polymerase II (PolII, Santa Cruz, sc-899), GATAl (Abeam, abl 1852) and TALI (Santa Cruz, sc-12984). DNase I cleavage density performed and analyzed as previously described (42). For ChlP-qPCR, relative enrichment was determined by comparing amplification of ChIP material to 1% input chromatin by the ACt method. Loci previously reported to be occupied and non-occupied by GATAl and TALI were used as positive and negative controls respectively (39).
Chromosome conformation capture (3C)
[0322] 3C assay was performed as previously described (39) except as below. Nuclei from formaldehyde cross-linked primary human erythroid precursors were digested with Hindlll prior to ligation and reversal of cross-links. Quantitative real-time PCR was performed using iQ SYBR Green Supermix (Bio-Rad, 170-8880). A fragment containing the BCLl 1A promoter was used as the anchor region. To correct for amplification efficiency of different primers, a control template was prepared by digesting and ligating an equimolar mixture of two bacterial artificial chromosomes (BACs) comprising the complete human BCL11A locus (RP11-606L8 and RP11-139C22) and one the human β-globin cluster (CTD-3055E11). An interaction between fragments in HS1 HS2 and HS3 of the human 13-globin locus control region (LCR) served as a positive control. Interaction frequency was expressed as amplification relative to the known LCR interaction, normalized to the BAC control template.
Fine-mapping BCL11 A locus
[0323] Markers (all coordinates hgl9) were selected from within the three BCL11A intron-2 DHSs +62 (chr2:60,717,492-60,718,860), +58 (chr2:60,721,411-60,722,674) and +55 (chr2:60,724,802- 60,726,084). 21 markers were identified from the 1000 Genomes Project database using the North European (CEU), Nigerian (YRI) and African- American (ASW) reference populations (Table S I). 38 additional variants were present in dbSNP135 (Table 2). The inventors sequenced by Sanger chemistry the three DHS intervals in the DNA of 52 and 36 sickle cell disease (SCD) patients from the CSSCD cohort with high (> 8%) and low (< 2%) HbF levels, respectively. From this sequencing effort, seven novel sequence variants were identified (Table 3). Because most markers cluster in small genomic intervals, it was not possible to design genotyping assays for some of them. Of 66 non-redundant variants identified in the three DHSs, genotyping assays for 40 markers were performed in 1,263 participants from the CSSCD, an African-American SCD cohort for which genomic DNA (gDNA) is available and HbF levels are known (21). Markers were genotyped using the Sequenom iPLEX platform. Individuals and DNA sequence variants with a genotyping success rate < 90% were excluded. Overall genotype concordance estimated from triplicates was 100%. SNPs passing quality control (QC; n = 38) are listed in Tables 2 and 3, and shown schematically in Figs. 11A and 1 IB below the three DHSs. A substantial fraction of the genotyped SNPs are rare in the reference populations so not surprisingly monomorphic in the CSSCD (n = 18). After QC, 1, 178 individuals and 20 polymorphic SNPs remained for the analysis. HbF levels were modeled as previously described (9, 43). Association and conditional analyses of single variants (MAF > 1%) were performed with PLINK (44) using linear regression under an additive genetic model. Analysis of common variants (MAF > 1%) revealed that rs 1427407 in DHS +62 had the strongest association to HbF level (P = 7.23 x 10-50; Figs. 11A and 3B). Conditional analysis demonstrated that after conditioning on rsl427407 and rs7606173, no more SNPs were significant (Fig. 3B). Adjusting for principal components (PCs) on 855 individuals for whom genome-wide genotyping data was available to account for admixture and other confounders yielded similar results.
[0324] For rare and low- frequency variants (MAF < 5%), the inventors performed set-based analyses using each of the three DHSs +62, +58 and +55 as the testing unit. For these analyses, we used the sequence kernel association test (SKAT-O) program (45) with default parameters. The inventors selected the 5% threshold for MAF in order to maximize statistical power given our limited sample size, but note that markers with a MAF between 1% and 5% were also analyzed in the single variant analyses presented above. This variant overlap is accounted for using conditional analyses with the common variants independently associated with HbF levels. Two sets were found to be statistically significant, namely DHS +62 and DHS +55, but after conditioning on rs 1427407 and rs7606173, results were no longer statistically significant, suggesting weak LD between the rare/low-frequency variants and the common SNPs (Table 5). The inventors did not find evidence that rare and low-frequency sequence variants within the BCL11A DHSs influence HbF levels in SCD subjects, despite Sanger re-sequencing these DHSs in 88 subjects with extreme HbF phenotype.
[0325] The rsl427407-rs7606173 haplotype frequencies in CSSCD are: T-G 24.5%, T- C 0.085%, G-C 42.3%, G-G 33.1%. The mean HbF level is 4.05% (SD 3.10) in 213 rsl427407-rs7606173 G-C individuals, 7.08% (SD 4.50) in 254 rsl427407-rs7606173 T-G/G-T heterozygotes and 11.21% (SD 4.37) in 60 rsl427407-rs7606173 T-G individuals (Fig. 3C). For comparisons of HbF levels between genotypes, the P-values were determined by one-tailed student t-tests.
Molecular haplotyping
[0326] For two heterozygous SNPs on the same chromosome, there are two possible phases: A- B/a-b (model 1) and A-b/a-B (model 2). For SNPs within the 12-kb BCL11A intron-2 fragment +52.0- 64.4 kb, phase was determined by cloning PCR products and determining co-distribution of SNP alleles. To determine phase of rs7569946 and rsl427407 alleles (separated by 30.1 kb on chromosome 2), emulsion fusion PCR was performed as previously described (24, 25) with minor modification. Fusion PCR brings two regions of interest, from separate parts of the same chromosome, together into a single product. By carrying out the reaction in emulsion with aqueous microdroplets surrounded by oil, the preponderance of amplicons are derived from a single template molecule. Genomic DNA from individuals known to be doubly heterozygous for rs7569946 and rsl427407 served as template in the following 100 i&l reaction (with final concentrations listed): KOD Hot Start DNA Polymerase (14 U, Novagen, 71086), KOD buffer (IX), MgS04 (1.5 mM), dNTPs (0.2 mM each), rs7569946-F and rsl427407-R primers (1 i&M each), rs7569946-R primer (30 nM), rs7569946-R-revcomp-rsl427407-F bridging inner primer (30 nM), gDNA (200 ng). The 100 i&l aqueous reaction was added dropwise with stirring to 200 i&l oil phase to create an emulsion. Two 125 i&l aliquots of emulsion were amplified under the following conditions: 95 degrees 2 minutes; 45 cycles of 95 degrees 20 seconds, 60 degrees 10 seconds, 70 degrees 30 seconds; 70 degrees 2 minutes. Hexane extracted fusion PCR product was subject to nested PCR in 25 i&l as follows: KOD Hot Start DNA Polymerase (0.5 U), KOD buffer (IX), MgS04 (1.5 mM), dNTPs (0.2 mM each), rs7569946-nested-F and rsl427407-nested-R primers (300 nM each), extracted fusion PCR product (75 nl); 95 degrees 2 minutes; 35 cycles of 95 degrees 20 seconds, 60 degrees 10 seconds, 70 degrees 30 seconds; 70 degrees 2 minutes. The nested product was confirmed by agarose gel electrophoresis to constitute a single band of expected size. The purified product was cloned with the Zero Blunt PCR Cloning kit (Life Technologies, K2700-20). The Sanger sequencing of fusion amplicons enumerated clones of 4 possible sequences: A-B, a-b, A-b and a-B. The likelihood of each phase was calculated based on a multinomial distribution assumption (Table 6). The likelihood ratio for the two configurations was calculated as a measure for the statistical significance of the data fitting haplotype model 1 (as compared to model 2). A ratio approaching infinity suggests model 1, a ratio of 1 suggests equipoise and a ratio approaching zero suggests model 2.
Pyrosequencing
[0327] Healthy CD34+ cell donors were screened to identify five donors heterozygous for rs 1427407. These CD34+ cells were subject to ex vivo erythroid differentiation. Chromatin was isolated and Chip performed with GATA1 and TAL I antibodies. Input chromatin as compared to GATA1 or TAL I precipitated material was subject to pyrosequencing to determine allelic balance of rs 1427407. Healthy CD34+ donors were screened to identify three donors heterozygous for the rs 1427407- rs7606173 G-C/T-G haplotype. These CD34+ cells were subject to ex vivo erythroid differentiation. Complementary DNA (cDNA) and gDNA were subject to pyrosequencing to determine allelic balance of rs7569946.
[0328] PCR conditions as follows: 2X HotStarTaq master mix (QIAGEN, 203443), MgCl2 (final concentration 3 mM), template DNA (0.1-1 ng) and SNP-specific forward and reverse-biotinylated primers (200 nM each). PCR cycling conditions were: 941/4C 15 min; 45 cycles of 94°C 30 s; 60°C 30 s; 72°C 30 s; 72°C 5 min. One primer of each pair was biotinylated. The PCR product strand containing the biotinylated primer was bound to streptavidin beads and combined with a specific sequencing primer. The primed single stranded DNA was sequenced and genotype analyzed using the Pyrosequencing PSQ96 HS System (QIAGEN Pyrosequencing) following the manufacturer's instructions.
Transgenic mice
[0329] The enhancer reporter construct pWHERE-Dest was obtained from Dr. William Pu.
Modified from pWHERE (Invitrogen, pwhere) as previously described (46), the construct has murine HI 9 insulators flanking a CpG-free lacZ variant driven by a minimal Hsp68 minimal promoter with a Gateway destination cassette at the upstream MCS. Enhancer fragments were amplified from mouse gDNA, recombined into pDONR221 vector (Invitrogen, 12536-017) by BP clonase (Invitrogen, 11789020) and recombined into pWHERE-Dest vector with LR clonase (Invitrogen, 11791020).
Plasmids were digested with Pad to remove vector backbone. The lacZ enhancer reporter fragments were purified by gel electroelution and then concentrated using Wizard DNA Clean-Up System (Promega, A7280). Transgenic mice were generated by pronuclear injection to FVB fertilized eggs. Approximately 10 ng/μΐ of DNA solution was used for series of injections. CD-I females were used as recipients for injected embryos. 10.5 to 14.5 dpc embryos were dissected from surrogate mothers with whole-mount and tissue X-gal staining performed as previously described (47). X-gal stained cytospins were counterstained with Nuclear Fast Red (Vector Laboratories, H-3403). Tails used for PCR genotyping. Animal procedures were approved by the Children's Hospital Instititutional Animal Care and Use Committee. Human erythroid precursor enhancer assay
[0330] Genomic DNA fragments containing putative enhancer elements were cloned into pLVX- Puro (Clontech, 632164) upstream of a minimal TK promoter and GFP reporter gene as described (39). 293T cells were transfected with FuGene 6 reagent (Promega, E2691) according to manufacturer's protocol. The media was changed after 24 hours to SFEM medium supplied with 2% penicillin- streptomycin, and after 36 hours, supernatant was collected and filtered. CD34+ cell-derived erythroid cultures were transduced with lentivirus on expansion days 4 and 5 by spin-infection as previously described (39). Cells were resuspended in erythroid differentiation media 24 hours after the second infection. Selection with puromycin 1 μg/ml commenced 48 hours after infection. Transduced cells were analyzed after five days in differentiation media by flow cytometry for GFP mean fluorescence intensity.
Flow cytometry
[0331] Live cells were gated by exclusion of 7-aminoactinomycin D (7-AAD, BD Pharmingen, 559925). Bone marrow (for erythroblast) and spleen (for lymphocyte) suspensions were isolated from young adult transgenic mice. Following hypotonic lysis of mature red blood cells, live cells (7-AAD-) sorted based on staining with CD71-biotin (BD, 557416), streptavidin-APC (BD, 554067), Ter-119-PE (BD, 553673), CD19-APC (BD, 550992) or CD3-PE (BD, 100308). CD71+Terl 19+, CD19+ and CD3+ sorted populations used for cytospin and RNA isolation.
TALEN -mediated chromosomal deletion
[0332] Transcription activator-like effector nucleases (TALENs) were designed to generate cleavages at mouse Bell la intron-2 at sites +50.4 kb (termed 5' site) and +60.4 kb (3' site) relative to the TSS. The TALENs recognize the following sequences: CTTAAGGCAAGAATCACT (5' left),
CCATGCCTTTCCCCCCCT (5' right), GAGTTAAAATCAGAAATCT (3' left),
CTGACTAATTGATCAT (3' right). TALENs were synthesized with Golden Gate cloning (48) using the NN RVD to recognize G. The synthesized DNA binding domains were cloned into pcDNA3.1
(Invitrogen, V790-20) with the Fokl nuclease domain, A152 N-terminal domain and +63 C-terminal domain previously described (49). 2.5 &g of each of the four TALEN plasmids with 0.5 &g pmaxGFP (Lonza) were delivered to 2 x 106 MEL or pre-B cells by electroporation per manufacturer's protocol (Lonza, VCA-1005). GFP -positive cells were sorted by flow cytometry after 48 hours. Cells seeded by limiting dilution in 96-well plates to isolate individual clones. Clones screened by PCR of gDNA to detect the amplification of a short product from upstream of the 5' site and downstream of the 3' site indicating deletion of the intervening segment. Monoallelic deleted clones were subject to a second round of TALEN-mediated deletion to obtain biallelic deleted clones. Clones with biallelic deletion were identified by detecting absence of amplification from within the deleted fragment. Deletion frequency was approximately one in 50 alleles. Deletion was validated with Southern blotting. Genomic DNA was digested with Bmtl; a 561-bp probe (amplified from gDNA upstream of the 5' site) hybridizes to a 3.6 kb fragment from the wild-type allele and a 8.9 kb fragment from the A50.4-60.4 deleted allele. RT-qPCR and immunoblotting
[0333] RNA isolation with RNeasy columns (Qiagen, 74106), reverse transcription with iScript cDNA synthesis kit (Bio-Rad, 170-8890), qPCR with iQ SYBR Green Supermix (Bio-Rad, 170-8880) and immunoblotting performed as described (39). For the mouse 13-globin cluster genes, a common primer pair recognizes the adult 13-globins 132 and 131 while independent primers recognize the embryonic 13-globins sy and 13H1. The following antibodies were used for immunoblotting: BCLl 1A (Abeam, abl9487), GAPDH (Santa Cruz, sc-25778).
Results
[0334] Genome-wide association studies (GWAS) have ascertained numerous common genetic variants associated with traits, frequently localized to regulatory DNA. The hypothesis that regulatory variation may account for substantial heritability has undergone scarce experimental evaluation. Here the inventors show that a common genetic variation at BCLllA associated with fetal hemoglobin (HbF) level indicates noncoding sequences decorated by an erythroid enhancer chromatin signature. Fine mapping within this putative regulatory DNA reveals a motif-disrupting common variant associated with reduced transcription factor binding, diminished BCLl 1A expression, and elevated HbF. The surrounding sequences function in vivo as a developmental stage-specific lineage-restricted enhancer. By genome engineering, it was shown that this enhancer is required for erythroid BCLl 1A expression yet dispensable outside the erythroid lineage. These results illustrate how GWAS can highlight functional variants of modest impact within causal elements essential for appropriate gene expression. The GWAS- marked BCLllA enhancer represents a favorable therapeutic target for the β-globin disorders.
[0335] GWAS have been tremendously successful in identifying many thousands of common single nucleotide polymorphisms (SNPs) associated with human traits and diseases. However advancing from genetic association to causal biologic process has often been difficult. Challenges include the large number of correlated variants that may be associated with individual traits (owing to linkage
disequilibrium (LD)), the modest effect size of many variant-trait associations, and the location of many associated variants in the noncoding genome. Recent genome-scale chromatin mapping studies have highlighted the enrichment of GWAS variants in regulatory DNA elements, suggesting many causal variants may affect gene regulation. Nonetheless, few examples confirming the significance of causal variants or elements have been experimentally demonstrated.
[0336] The GWAS of HbF level have been particularly striking. Genetic variation at just three loci (HBB, HBS1L-MYB, and BCLllA) accounts for up to 50% of the heritable variation in HbF level. The same variants are also associated with the clinical severity of the major β-globinopathies sickle cell disease and β -thalassemia, perhaps not surprisingly since HbF is a major modifier of these disorders. Work from the inventor's laboratory and others have validated that BCLl 1A is a direct regulator of HbF levels. BCLl 1A is a transcriptional repressor that resides within multiprotein complexes occupying the β -globin gene cluster. While constitutive BCLl 1A deficiency results in embryonic lethality and impaired lymphocyte development, erythroid-specific deficiency of BCLl 1A counteracts developmental silencing of embryonic and fetal globin genes and is sufficient to rescue the hematologic and pathologic features of sickle cell disease in mouse models. Therefore BCLl 1A has emerged as a novel therapeutic target for the β-globin disorders. Understanding the consequences of impaired BCLl 1A is imperative. Human coding variants of BCLl 1A have not been described despite large-scale resequencing efforts. The BCLl 1 A variants associated with HbF levels reside in noncoding regions of BCLl 1 A. To further understand how common genetic variation impacts BCLl 1 A, HbF level, and β-globin disorder severity, the role of HbF - associated variants was investigated in detail.
Trait-associated variants near erythroid enhancers
[0337] Numerous GWAS as well as follow-up fine-mapping studies have been performed of erythroid traits (including phenotypes such as erythrocyte number and volume, hemoglobin
concentration, and HbF level), identifying 636 trait-associated SNPs at genome-wide significance (p<5e~ 8). These SNPs are enriched in promoters and coding regions as compared to random SNPs (Figure 1). Still the majority of the SNPs reside elsewhere in the genome. A global chromatin profiling of primary human erythroid precursors was performed, which identified an extensive set of distal erythroid enhancers defined by characteristic histone modifications and DNase I hypersensitivity. A strong colocalization of erythroid GWAS SNPs with enhancers was observed as compared to non-erythroid trait-associated SNPs, SNPs found on a common genotyping array, or randomly permuted SNPs. 13.5% of the erythroid trait-associated SNPs fell directly into erythroid enhancers, an 11.4-fold enrichment over random permuted enhancers (P < 1 x 10"4), as compared to 1.4% of non-erythroid trait-associated SNPs, representing a 1.4-fold enrichment (P = 0.0013). Many of the erythroid trait-associated SNPs were found in close proximity to erythroid enhancers (Figure IB). The median distance to erythroid enhancer was 16.0 kilobases (kb) for erythroid trait-associated SNPs, as compared to 238.0, 392.6, and 482.4 kb respectively for non-erythroid trait-associated, genotyping array, and randomly permuted SNPs. These results indicate that a substantial fraction of common variants associated with erythroid traits reside at or near erythroid enhancers, and are consistent with the hypothesis that causal variants often influence context-dependent gene regulation.
HbF GWAS mark an erythroid enhancer signature
[0338] Six GWAS have been conducted of HbF level (or the highly correlated trait F-cell number), including populations of European, African, and Asian descent. Each has identified trait-associated variants within BCLl 1 A. Variation at BCLl 1 A is estimated to explain -15% of the trait variance. Four different SNPs have been identified as most highly associated with the trait (rs 1427407, rsl 1886868, rs4671393, and rs766432; these so-called "sentinel" SNPs cluster within 3 kb of each other in BCLl 1 A intron-2 (Figure 2A). Haplotypes including the sentinel SNPs appear to better explain the HbF association than any individual SNP. Fifty SNPs at the BCLllA locus and twenty-seven SNPs within intron-2 have been associated with HbF level with at least genome-wide significance (P < 5 x 10"8).
[0339] The distribution of the HbF-associated SNPs at BCLllA were compared with DNase I sensitivity, an indicator of chromatin state suggestive of regulatory potential. In human erythroid precursors, three peaks of DNase I hypersensitivity were observed in intron-2, adjacent to and overlying the HbF-associated variants (Figure 2A), termed DNase I hypersensitive sites (DHSs) +62, +58, and +55 based on distance in kb from the TSS of BCLllA. Brain and B-lymphocytes, two tissues that express high levels of BCL11A, and T-lymphocytes, which do not express appreciable BCL11A, show unique patterns of DNase I sensitivity at the BCLllA locus, with a paucity of DNase I hypersensitivity overlying the trait-associated SNPs (Figure 2A).
[0340] The chromatin signature around the BCLllA locus was further analyzed by chromatin immunoprecipitation with massively parallel sequencing (ChlP-seq). ChlP-seq from primary human erythroid precursors revealed histone modifications with an enhancer signature overlying the trait- associated SNPs at BCLllA intron-2, including the presence of H3K4mel and H3K27ac and absence of H3K4me3 and H3K27me3 marks (Fig.2A). The master erythroid transcription factors GATAl and TALI occupy this enhancer region (Fig. 2A). ChlP-qPCR experiments demonstrated three discrete peaks of GATAl and TALI binding within BCLllA intron-2, each falling within an erythroid DHS (Fig. 2B).
[0341] One common feature of distal regulatory elements is long-range interaction with the promoters whose expression they regulate. The interactions between the BCLllA promoter and fragments across 250 kb of the BCLllA locus were evaluated, including sequences upstream, downstream, and intragenic. The greatest promoter interaction was observed within the region of intron-2 containing the erythroid DHS and trait-associated SNPs (Fig. 2C). These results indicate that these sequences have regulatory potential.
Regulatory variants
[0342] It was hypothesized that the causal trait-associated SNPs could function by modulating critical cis-regulatory elements. Therefore extensive genotyping of SNPs was performed within the three erythroid DHSs +62, +58, and +55 in 1263 DNA samples from the Cooperative Study of Sickle Cell Disease (CSSCD), an African-American sickle cell disease cohort for which genomic DNA is available and HbF levels are known. 66 DNA sequence variants located in the three DHSs from dbSNP were identified from reference populations CEU, YRI, and ASW within the 1000 Genomes Project, and by Sanger sequencing 88 individuals from the CSSCD with extreme HbF phenotype. 26 markers failed genotyping assay design and 18 were monomorphic (Tables 1 and 2). After quality control, 1178 individuals and 20 polymorphic SNPs remained for association testing. Analysis of common variants (minor allele frequency (MAF) > 1%) revealed that rs 1427407 in DHS +62 had the strongest association to HbF level (P = 7.23 x 10"50; Table 1). [0343] Previously, the inventors had used the CSSCD to fine-map the association signal with HbF at the BCLllA locus and reported a strong association with rs4671393; in that study, rsl427407 was imputed. Two additional SNPs, rs766432 and rsl 1886868 have also been identified in prior studies as sentinel SNPs most highly associated with HbF level (or F-cell number). In a subset of individuals (N = 728) for which genotypes at all four sentinel SNPs were known, when conditioned on genotypes at rsl427407, the association result was not significant at rs4671393, rs766432, or rsl 1886868; conversely, the association remained highly significant for rsl427407 when conditioning on rs4671393, rs766432, or rsl 1886868 (Table 3). Therefore rs 1427407 is the SNP most associated with HbF within the erythroid DHS and better accounts for the trait association than other previously described sentinel SNPs.
[0344] When conditioned on rs 1427407, other associations to HbF level were found within the three DHSs that were not completely lost (Table 1). The most significant association remaining was for rs7606173 in DHS +55 (P = 5.11 x 10"10); rs7599488 in DHS +58, which were previously reported, was only slightly less significant (P = 1.71 x 10"9) in this conditional analysis. After conditioning on rs 1427407 and rs7606173, no more SNPs were significant (Table 1). Adjusting for principal components (PCs) on 855 individuals for whom genome-wide genotyping data was available to account for admixture and other confounders yielded similar results (data not shown). The rsl427407-rs7606173 T-G haplotype was defined as that most highly associated with HbF level.
[0345] Rare and low-frequency variants (MAF < 5%) were also analyzed for their association with HbF levels using each of the three DHSs +62, +58, and +55 as a testing set. Two sets were found to be significant, namely DHS +62 and DHS +55, but after conditioning on rsl427407 and rs7606173, results were no longer significant, indicating weak LD between the rare/low- frequency variants and the common SNPs (Table 4). Therefore, no evidence that rare and low-frequency sequence variants within the BCLllA DHSs influence HbF levels in SCD patients was found, despite Sanger resequencing 88 individuals from CSSCD with extreme HbF phenotype.
[0346] The SNP rs 1427407 falls within a peak of GATA 1 and TAL 1 binding as determined by
ChlP-seq and ChlP-qPCR (Figs. 2A and 2B). The minor T-allele disrupts the G-nucleotide of a sequence element highly resembling a half-E-box/GATA composite motif [CTG(« )GATA]. This motif has been found to be highly enriched by GATA1 and TALI complexes in erythroid cells by ChlP-seq
experiments. Primary erythroid samples were identified from individuals heterozygous for the major G- allele and minor T-allele at rs 1427407 and subjected these samples to ChlP followed by pyrosequencing. The inventors identified an even balance of alleles in the input DNA. However more frequent binding to the G-allele was observed compared to the T-allele of approximately 60:40 in both the GATA1 and TALI immunoprecipitated chromatin samples (Fig. 3A).
[0347] It was previously reported that the high-HbF associated A-allele of rs4671393 was associated with BCL11A expression in human lymphoblastoid cell lines. The inventors were unable to reproduce a significant association between BCLllA genotype and expression level analyzing a larger set of lymphoblastoid cell lines (data not shown). It was speculated that the high-HbF-associated rs 1427407- rs7606173 haplotype influences BCL11A expression in an erythroid-speciflc context. The common synonymous SNP rs7569946 lies within exon-4 of BCLl 1 A and may serve as a marker of allelic expression. Three primary human erythroid samples were identified that were doubly heterozygous for the rsl427407-rs7606173 haplotype and rs7569946. The samples were subjected to molecular haplotyping by emulsion fusion PCR. The haplotyping demonstrated that for each sample the major rs7569946 G-allele was in phase with the low-HbF-associated rsl427407-rs7606173 G-C haplotype. Genomic DNA and cDNA were assayed by pyrosequencing of rs7569946 to determine allelic balance. Whereas the alleles were balanced in the genomic DNA, significant imbalance in the cDNA favoring increased expression of the low-HbF linked G-allele of rs7569946 was observed (Fig. 3B). These results indicate that the high-HbF rs 1427407 T-allele, which disrupts the half-E-box/GATA motif, is associated with reduced binding of GATAl and TALI and reduced expression of BCLl 1A in erythroid precursors. The mean HbF level in 60 high-HbF rsl427407-rs7606173 T-G haplotype homozygous individuals was 11.21% as compared to 4.05% in 213 low-HbF rsl427407-rs7606173 G-C haplotype homozygous individuals (P = 2.5 x 10 19) (Fig. 3C). In sum, the following evidence indicates that rs 1427407 is a causal SNP for HbF level: it has the highest association to the phenotype of any known variant, it accounts for the associations observed with previously described sentinel SNPs, it impacts a motif required for GATAl and TALI binding, and it is associated with GATAl and TALI binding as well as with BCLl 1A expression. However, variation at this position in the setting of common haplotypes is associated with only modest perturbation of BCLl 1A expression.
Enhancer sufficiency for erythroid expression
[0348] To understand the context within which these apparent regulatory trait-associated SNPs play their role, the function of the harboring cis-regulatory elements was explored. The inventors cloned -12- kb (+52.0-64.4 kb from TSS) which contained the three erythroid DHSs, and assayed enhancer potential in a transgenic reporter assay. In this assay putative enhancer sequences are positioned upstream of a minimal promoter (Hsp68) and reporter gene (lacZ) bounded by insulator sequences. Constructs were introduced to murine zygotes with reporter gene expression monitored throughout development.
Endogenous mouse BCLl 1A showed abundant expression throughout the developing central nervous system with much lower expression observed in the fetal liver. In contrast, reporter gene expression in the transgenic embryos was observed to be largely confined to the fetal liver, the site of definitive erythropoiesis, with lesser expression noted in the central nervous system (Fig. 4A).
[0349] A characteristic feature of globin genes is their developmental regulation. During human development, yolk sac-derived ε-globin is superseded in the first trimester by fetal liver-derived γ-globin. Around birth, γ-globin is gradually silenced and β -globin becomes activated. There is only a single switch in gene expression during mouse ontogeny. During this transition, which occurs at mid-gestation, the circulating yolk sac-derived primitive erythrocytes express embryonic-stage globins sy and βΗΙ whereas the fetal liver definitive erythroblasts express adult-stage globins β ΐ and β2. Concordant with this developmental switch, BCL11A expression is expressed in the definitive but not primitive stage erythroid lineage. Stable transgenic lines were derived from the BCL11A +52.0-64.4 reporter mice. In these mice at E12.5 circulating erythrocytes do not stain for X-gal whereas liver erythroblasts robustly stain positive (Fig. 4B). At E10.5 lacZ expression is only observed in the fetal liver primordium and not in the circulating blood within the embryo, placenta, or yolk sac (Fig. 6A). These results indicate that the GWAS-marked BCL11A intron-2 regulatory sequences are sufficient to specify developmentally appropriate gene expression.
[0350] Within the hematopoietic compartment, BCL 11 A expression is found in erythroid precursors and B-lymphocytes. Erythroid precursors and B-lymphocytes were isolated from transgenic young adult animals and expression of the lacZ reporter gene was evaluated. Endogenous BCL11A was expressed at 10.4-fold higher levels in splenic B-lymphocytes as compared to bone marrow erythroid precursors. However, lacZ expression was restricted to erythroid precursors and was not observed in B- lymphocytes (Fig.4C). These results indicate erythroid-specificity of these regulatory sequences.
[0351] A series of deletion mutants was generated to refine the minimal elements required for erythroid enhancer activity. Sequences containing the central +58 DHS were sufficient for erythroid enhancer activity. Those sequences containing only the flanking +62 or +55 elements were not able to direct erythroid gene expression (Fig. 6B). These DHSs were also tested for their ability to enhance gene expression in primary human erythroid precursors. The inventors used lentiviral delivery of a GFP reporter system with a minimal TK promoter as previously described. Similarly, only the +58 but not the +55 or +62 DHSs were able to enhance gene expression in this reporter assay (Fig. 7).
Enhancer requirement for erythroid expression
[0352] Next the inventors chose to determine the requirement of these regulatory sequences for appropriate expression of BCL11A and globin genes. Inspection of the Bell la locus in previously published global chromatin profiling of mouse erythroid cells revealed that this region of intron-2 possesses an orthologous enhancer signature with presence of H3K4mel and H3K27ac, absence of H3K4me3 and H3K27me3, and occupancy by GATA1 and TALI (Fig. 8). Moreover, erythroid-specific DNase I hypersensitivity was observed at these sequences. At each of the human erythroid DHSs +62, +58, and +55, evidence of evolutionary sequence conservation was observed, particularly within +62 and +55. To determine the requirement of these orthologous regulatory sequences for BCL11A expression, the mouse erythroleukemia cell line (MEL) was used. These cells depend on BCL11A expression for appropriate adult-stage pattern globin gene expression. Sequence-specific nucleases can result in the production of small chromosomal deletions. TALENs were engineered to introduce double-strand breaks to flank the orthologous 10-kb Bell la intron-2 sequences carrying the erythroid enhancer chromatin signature. Clones were screened for NHEJ-mediated repair and three unique clones were isolated that had undergone biallelic excision. PCR and Southern blotting verified excision of the intronic segment within clones (Fig. 9). Sanger-sequenced breakpoints were characteristic of TALEN-mediated cleavage with subsequent NHEJ repair (Fig. 10).
[0353] Expression of BCL11A was analyzed in the MEL cells with biallelic 10-kb intronic deletion. A dramatic reduction of BCL11A expression to -3% of baseline levels was observed (Fig. 5A). Similar reductions were noted with primer pairs detecting exon junctions upstream, spanning, or downstream of the deletion. By Western blotting, BCL11A expression was not detectable in the 10-kb enhancer deleted clones (Fig. 5B). MEL cells typically express high levels of the adult globin genes β 1 and β2 and low levels of the embryonic globin genes sy and βΗΙ. In the absence of the 10-kb enhancer for BCL1 lA, expression of adult globin genes was decreased by -2-5 -fold, whereas embryonic globin genes were considerably derepressed. The ratio of embryonic sy to adult β 1/2 was increased by a mean of 364-fold in three clones lacking the orthologous BCL11A erythroid enhancer (Fig. 5C).
[0354] To determine if the +50.4-60.4 kb intronic sequences were universally required for BCL11A expression, their loss was evalutated in a non-erythroid context. The same strategy of introduction of two pairs of TALENs to obtain clones with the NHEJ-mediated Δ50.4-60.4 deletion was employed in a pro-B lymphocyte cell line. Two unique Δ50.4-60.4 clones were isolated, and verified by PCR, Southern blotting, and Sanger sequencing (Figs. 9 and 10). In contrast to the erythroid cells, BCL11A expression was retained in the A50.4-60.4kb enhancer deleted pro-B cell clones at both the RNA and protein levels (Figures 5A and 5B). These results indicate the orthologous erythroid enhancer sequences are not required for integrity of transcription from the Bell la locus but only essential for erythroid gene expression.
[0355] An enhancer chromatin signature was found at intron-2 of BCL11A, directly overlying the HbF-associated SNPs. This region had numerous biochemical features of an enhancer, including occupancy by the histone marks H3K4mel and H3K27ac in the absence of H3K4me3 and H3K27me3, binding of the erythroid TFs GATA1 and TALI, erythroid-specific DNase-I hypersensitive sites, and long-range promoter interaction by 3C. Moreover, the inventors were able to fine-map this locus, using the DHSs as a guide, to identify the SNP rs 1424707 as being most highly associated with the trait, and entirely accounting for the trait association of the previously described and highly linked sentinel SNPs. In addition, it is shown herein that rs 1427407 disrupts a half-E-box GATA motif occupied by the transcription factors GATA1 and TALI in erythroid precursors. However, even after conditioning on rs 1427407 an association remains with several other SNPs in adjacent DHSs indicating a haplotype effect. It was found that haplotypes which possess a combination of SNP genotypes in adjacent elements that each modulate regulatory function cooperate to give the ultimate phenotype of BCL11A expression. Using heterozygous donors, a modest impact of the high-HbF associated haplotype was demonstrated on both TF binding and BCL11A expression.
[0356] These studies help to estimate the change in BCL 11 A expression required to result in a clinically meaningful increase in HbF level in patients with β-globin disorders. The difference in BCL11A expression between the high-HbF rsl427407-rs7606173 T-G and low-HbF G-C haplotypes was 1.7-fold and the HbF levels of T-G and G-C homozygotes were 11.2% and 4.1% (Fig.s 3B and 3C). HbF levels of >20% have been predicted to prevent the adverse consequences of SCD. A reduction in BCL11A expression of several-fold would likely approach this HbF goal.
[0357] This study identifies regulatory variation at BCL11A that impacts an erythroid enhancer. Many trait-associated SNPs are noncoding and have relatively small effect size. Due to these features, these SNPs are sometimes considered to be of negligible clinical importance. This study illustrates that a small effect size engendered by an individual noncoding variant does not preclude a large effect size of the underlying regulatory element. For example, despite a relatively modest impact of functional SNPs on expression of the underlying gene target BCL11A, the causal regulatory elements are essential for expression of BCL11A and globin genes in adult-stage erythroid precursors. The same regulatory element is dispensable for BCL11A expression in a non-erythroid lineage. A goal of studying protective alleles is to understand their underlying molecular mechanisms in an effort to reproduce this biological effect in at-risk individuals. Thus, many trait-associated polymorphisms will reside context-specific critical regulatory elements whose function may further illuminate the underlying biology of the trait beyond merely identifying the regulated gene. For example loss of BCL11A while resulting in impaired hemoglobin switching also results in impaired neurogenesis and lymphopoeisis and results in embryonic lethality. Ultimately a better understanding of the causal regulatory elements and associated regulatory networks underlying traits could identify novel therapeutic targets. The erythroid enhancer of BCL11A could itself constitute a favorable target for therapeutic genome editing in that ablation could impede BCL11A expression in erythroid precursors with resultant HbF derepression while preserving BCL11A expression in non-erythroid lineages.
References
1. L. Fugger, G. McVean, J. I. Bell, N. Engl. J. Med. 367, 2370-2371 (2012).
2. J. Ernst et al., Nature. 473, 43-49 (2011).
3. D. S. Paul et al., PLoS Genet. 7, el002139 (2011).
4. M. T. Maurano et al., Science. 337, 1190-1195 (2012).
5. P. van der Harst et al., Nature. 492, 369-375 (2012).
6. ENCODE Project Consortium et al., Nature. 489, 57-74 (2012).
7. S. Menzel et al., Nat. Genet. 39, 1197-1199 (2007).
8. M. Uda et al., Proc. Natl. Acad. Sci. U. S. A. 105, 1620-1625 (2008).
9. G. Lettre et al., Proc. Natl. Acad. Sci. U. S. A. 105, 11869-11874 (2008).
10. M. Nuinoon et al., Hum. Genet. 127, 303-314 (2010). 11. N. Solovieff et al., Blood. 1 15, 1815- 1822 (2010).
12. P. Bhatnagar et al., J. Hum. Genet. 56, 316-323 (2011).
13. V. G. Sankaran et al., Science. 322, 1839-1842 (2008).
14. J. Xu et al., Genes Dev. 24, 783-798 (2010).
15. J. Xu et al., Proc. Natl. Acad. Sci. U. S. A. 110, 6518-6523 (2013).
16. V. G. Sankaran et al., Nature. 460, 1093- 1097 (2009).
17. J. Xu et al., Science. 334, 993-996 (2011).
18. F. Esteghamat et al., Blood. 121, 2553-2562 (2013).
19. P. Liu et al., Nat. Immunol. 4, 525-532 (2003).
20. Y. Yu et al., J. Exp. Med. 209, 2467-2483 (2012).
21. M. D. Farber, M. Koshy, T. R. Kinney, J. Chronic Dis. 38, 495-505 (1985).
22. E. Soler et al., Genes Dev. 24, 277-289 (2010).
23. M. T. Kassouf et al., Genome Res. 20, 1064-1083 (2010).
24. D. J. Turner, M. E. Hurles, Nat. Protoc. 4, 1771-1783 (2009).
25. J. Tyson, J. A. Armour, BMC Genomics. 13, 693-2164-13-693 (2012).
26. M. Leid et al., Gene Expr. Patterns. 4, 733-739 (2004).
27. M. S. Kowalczyk et al., Mol. Cell. 45, 447-458 (2012).
28. H. J. Lee, E. Kim, J. S. Kim, Genome Res. 20, 81-89 (2010).
29. D. E. Bauer, S. C. Kamran, S. H. Orkin, Blood. 120, 2945-2953 (2012).
30. A. John et al., Development. 139, 1831-1841 (2012).
31. R. P. Patwardhan et al., Nat. Biotechnol. 30, 265-270 (2012).
32. A. Melnikov et al., Nat. Biotechnol. 30, 271-277 (2012).
33. K. A. Frazer, S. S. Murray, N. J. Schork, E. J. Topol, Nat. Rev. Genet. 10, 241-251 (2009).
34. A. N. Koehler, Curr. Opin. Chem. Biol. 14, 331-340 (2010).
35. F. D. Urnov, E. J. Rebar, M. C. Holmes, H. S. Zhang, P. D. Gregory, Nat. Rev. Genet. 11, 636-646 (2010).
36. J. K. Joung, J. D. Sander, Nat. Rev. Mol. Cell Biol. 14, 49-55 (2013).
37. J. van der Oost, Science. 339, 768-770 (2013). 38. Thanks to A. Woo, A. Cantor, M. Kowalczyk, S. Burns, J. Wright, J. Snow, J. Trowbridge and members of the Orkin laboratory, particularly C. Peng, P. Das, G. Guo, M. Kerenyi, and E. Baena, for discussions. C. Guo and F. Alt provided the pre-B cell line, A. He and W. Pu the pWHERE lacZ reporter construct, C. Currie and M. Nguyen technical assistance, D. Bates and T. Kutyavin expertise with sequence analysis, R.
39. J. Xu et al., Dev. Cell. 23, 796-811 (2012).
40. E. van den Akker, T. J. Satchwell, S. Pellegrin, G. Daniels, A. M. Toye, Haematologica. 95, 1594-1598 (2010).
41. A. L. Bredemeyer et al., Nature. 442, 466-470 (2006).
42. R. E. Thurman et al., Nature. 489, 75-82 (2012).
43. G. Galarneau et al., Nat. Genet. 42, 1049-1051 (2010).
44. S. Purcell et al., Am. J. Hum. Genet. 81, 559-575 (2007).
45. M. C. Wu et al., Am. J. Hum. Genet. 89, 82-93 (2011).
46. A. He, S. W. Kong, Q. Ma, W. T. Pu, Proc. Natl. Acad. Sci. U. S. A. 108, 5632-5637 (2011).
47. M. A. McDevitt, Y. Fujiwara, R. A. Shivdasani, S. H. Orkin, Proc. Natl. Acad. Sci. U. S. A. 94, 7976-7981 (1997).
48. T. Cermak et al., Nucleic Acids Res. 39, e82 (2011).
49. J. C. Miller et al., Nat. Biotechnol. 29, 143-148 (2011).
50. R. Galanello et al., Blood. 114, 3935-3937 (2009).
51. H. T. Bae et al., Blood. 120, 1961-1962 (2012).
52. K. E. McGrath et al., Blood. 117, 4600-4608 (2011).
53. D. Noordermeer, W. de Laat, IUBMB Life. 60, 824-833 (2008).
54. D. R. Higgs, D. Vernimmen, B. Wood, Adv. Genet. 61, 143-173 (2008).
55. E. Pinaud et al., Adv. Immunol. 110, 27-70 (2011). Table 1 Association analysis of common SNPs in BCLllA DHSs +62, +58, or +55
Conditional on
Conditional on
rsl 427407 and rsl427407
rs7606173
DHS Marker MAF β P β p β P
+62 rsl 11575474 0.0153 -0.2624 0.09762 -0.0851 0.5584 0.0486 0.7368
+62 rsl l2105713 0.0115 -0.3285 0.07755 -0.2137 0.2097 0.0859 0.6107
+62 rs74958177 0.0646 -0.3614 2.79xl0-6 -0.1838 0.01041 0.0832 0.2518
+62 rsl427407 0.2460 0.6634 7.23xl0_5° - - - -
+62 rs7599488 0.3148 -0.0047 0.9116 0.2622 2.43xl0_1° 0.0915 0.3547
+62 rsl 896293 0.1089 -0.2623 2.52xl0-5 -0.1248 0.03098 0.0241 0.6952
+58 rs6738440 0.2734 -0.3820 1.25xl0-18 -0.1935 5.64xl0-6 0.0223 0.6887
+55 rsl47910897 0.0132 -0.3656 0.03294 -0.2586 0.09945 0.1575 0.3101
+55 rsl48529953 0.0140 -0.3521 0.04034 -0.1423 0.3668 0.0098 0.9501
+55 rs7606173 0.4238 -0.4691 2.86xl0-34 -0.2632 9.66xl0-11 - -
Association analysis of common (MAF > 1%) SNPs BCLllA DHSs +62, +58, or +55 from 1178 individuals from CSSCD available for analysis. DHS, DNase I hypersensitive site. MAF, minor allele frequency.
Table 2 SNPs within BCLllA DHSs +62, +58, or +55
Mai or Minor
Marker CHR POS DHS Genotvped MAF
Allele Allele
rsl49113684 2 60,717,544 C A +62 Monomorp ic 0.0000 rsl 11575474 2 60,717,559 C T +62 YES 0.0157 rsl48272134 2 60,717,643 c A +62 Failed Assay Design - rsl 82773253 2 60,717,676 A G +62 Monomorphic 0.0000 rsl 88706265 2 60,717,769 c T +62 Monomorphic 0.0000 rs74958177 2 60,717,776 A G +62 YES 0.0645 rsl427407 2 60,718,043 G T +62 YES 0.2460 rs35262352 2 60,718,076 A - +62 Failed Assay Design - rs79781583 2 60,718,077 A T +62 Failed Assay Design - rs201428515 2 60,718,088 G A +62 Monomorphic 0.0000 rsl l2105713 2 60,718,278 G A +62 YES 0.1145 rs7599488 2 60,718,347 C T +62 YES 0.3149 rsl 13636744 2 60,718,540 C T +62 YES 0.0042 rs35259900 2 60,718,555 C T +62 Failed Assay Design - rsl l l911554 2 60,718,569 A G +62 Failed Assay Design - rsl37943695 2 60,718,574 G A +62 Monomorphic 0.0000 rs45579333 2 60,718,599 G A +62 Monomorphic 0.0000 rs77876582 2 60,718,639 C T +62 Failed Assay Design - rsl 12634025 2 60,718,708 G A +62 Failed Assay Design - rs45439602 2 60,718,721 G A +62 Failed Assay Design - rsl 12387548 2 60,718,762 C T +62 Failed Assay Design - rsl91369155 2 60,718,781 G A +62 Failed Assay Design - rs6723022 2 60,718,807 A C +62 Monomorphic 0.0000 rsl 1422901 2 60,718,819 G A +62 Failed Assay Design - rs200632291 2 60,718,824 A G +62 Failed Assay Design - rsl 1387709 2 60,718,826 A - +62 Failed Assay Design - rsl 896293 2 60,718,848 G T +62 YES 0.1088 rs71526487 2 60,721,587 T c +58 Failed Assay Design - rsl 85151573 2 60,721,639 G c +58 Monomorphic 0.0000 rs6721788 2 60,721,846 T c +58 YES 0.0025 rs76033449 2 60,721,900 G A +58 YES 0.0004 rs6706648 2 60,722,040 T c +58 Failed Genotyping - rs62142615 2 60,722,120 T c +58 YES 0.0081 rs35923541 2 60,722,197 T - +58 Monomorphic 0.0000 rs35815093 2 60,722,208 G - +58 Failed Assay Design - rsl47659683 2 60,722,219 G A +58 Failed Assay Design - rs6738440 2 60,722,241 A G +58 YES 0.2732 rsl 89178945 2 60,722,449 G A +58 Monomorphic 0.0000 rsl40819321 2 60,722,465 G A +58 YES 0.0064 rsl 81895125 2 60,722,609 A G +58 Monomorphic 0.0000 rs144676401 2 60,722,634 C T +58 Monomorphic 0.0000 rsl47910897 2 60,724,818 T c +55 YES 0.0132 rs34322220 2 60,724,831 T - +55 Monomorphic 0.0000 rsl48529953 2 60,724,967 A G +55 YES 0.0140 rsl 88426060 2 60,724,989 T G +55 Failed Assay Design - rsl91734859 2 60,724,994 A G +55 Failed Assay Design - rs45442493 2 60,725,043 G C +55 Monomorphic 0.0000 rs59444712 2 60,725,047 T C +55 Failed Assay Design - rs35173197 2 60,725,052 G - +55 Failed Assay Design - rsl 88151753 2 60,725,071 G A +55 Failed Assay Design - rsl 81041409 2 60,725,143 C A +55 Failed Assay Design - rsl 42174420 2 60,725,169 C A +55 Monomorphic 0.0000 rsl 87333125 2 60,725,342 C G +55 Monomorphic 0.0000 rs45566439 2 60,725,384 C T +55 Failed Assay Design - rs7606173 2 60,725,451 G c +55 YES 0.4235 rsl90502487 2 60,725,499 C T +55 Failed Assay Design - rsl51187913 2 60,725,714 G T +55 Monomorp ic 0.0000 rsl 13798461 2 60,725,727 T c +55 Monomorp ic 0.0000 rsl 81699714 2 60,726,054 G A +55 Failed Genotyping -
SNPs falling within BCLllA DHSs +62, +58, or +55 and present in either dbSNP or the 1000 Genomes data for YRI, CEU and ASW reference populations. Genotyped SNPs are identified and MAF within the CSSCD listed. Genomic coordinates hgl9.
Table 3 | Additional markers found by Sanger re-sequencing
Mai or Minor
Marker CHR POS DHS Genotyped MAF
Allele Allele
SS711589103 2 60,717,561 T A +62 YES 0.00085
SS711589106 2 60,718,048 c G +62 Failed Assay Design
SS711589108 2 60,722,056 G A +58 YES 0.00424
SS711589109 2 60,722,355 C T +58 YES 0.00085
SS711589110 2 60,722,358 C T +58 Failed Assay Design
SS711589111 2 60,725,211 G T +55 YES 0.00509
SS711589113 2 60,725,564 C A +55 YES 0.00127
88 individuals from CSSCD with extreme HbF phenotype underwent Sanger re-sequencing of the three DHSs within BCLllA. Identified novel markers listed. Genotyped SNPs are identified and MAF within the CSSCD listed. Genomic coordinates hgl9.
Table 4. Conditional analyses of four sentinel SNPs
Figure imgf000080_0001
Conditional analyses of four common sentinel SNPs previously associated with HbF levels{ {44;20; 19;36;37;515} }. All four were genotyped in 728 individuals fro CSSCD. It was not possible to calculate P for rs766432 when conditioning on rs4671393 (and vice versa) because these two markers are so strongly correlated (r2 = 0.997). r = 0.848 between rsl427407 and rs766432; r = 0.709 between rsl427407 and rsl 1886868; r = 0.850 between rsl427407 and rs4671393; r = 0.761 betw rs766432 and rsl 1886868; r2 = 0.758 between rsl 1886868 and rs4671393.
Table 5 Rare and low-frequency variant analysis
Figure imgf000081_0001
all 14 0.000631176 0.1503518 0.6908852
Rare and low- frequency variant analysis results (MAF < 5%). The
analysis was performed using the set-based SKAT-O algorithm using
the individual DHSs +62, +58, and +55 as three different sets. The
bottom row "all" shows the results of the tests when the three regions
were collapsed together.
Table 6 | Emulsion fusion haplotyping PCR sequencing
Likelihood ratio
Donor no. G-G A-T G-T A-G G-G/A-T phase
1 19 22 4 2 1.63xl029
2 22 14 2 3 3.78x1026
3 25 23 9 10 4.69xlOn
Emulsion fusion PCR analysis of rs7569946-rsl427407 haplotype. Fusion PCR
conducted in emulsion from three individual donors doubly heterozygous for
rs7569946 and rsl427407, generating a fusion amplicon encompassing both
S Ps. The fusion amplicon was cloned, and individual clones were Sanger
sequenced. The number of clones of each genotype is listed. The likelihood ratio
for the G-G/A-T as compared to G-T/A-G phase was calculated.
Table 7 | Coordinates of fragments for reporter assays
hgl9. chr2 BCLllA TSS fkb)
Reporter Name Start End Start End Length (bp) LacZ 52.0-64.4 60,716,189 60,728,612 64,444 52,021 12,423
56.8-64.4 60,716,189 60,723,870 64,444 56,763 7,681
52.0-57.6 60,722,992 60,728,612 57,641 52,021 5,620
+62 60,717,236 60,719,036 63,397 61,597 1,800
+58 60,722,006 60,723,058 58,627 57,575 1,052
+55 60,724,917 60,726,282 55,716 54,351 1,365
GFP +164 60,616,396 60,618,032 164,237 162,601 1,636
+156 60,623,536 60,624,989 157,097 155,644 1,453
+153 60,626,565 60,628, 177 154,068 152,456 1,612
+62 60,717,236 60,719,036 63,397 61,597 1,800
+58 60,721,212 60,722,958 59,421 57,675 1,746
+55 60,724,780 60,726,471 55,853 54,162 1,691
+41 60,739,075 60,740, 154 41,558 40,479 1,079
+32 60,748,003 60,749,009 32,630 31,624 1,006
-46 60,826,438 60,827,601 -45,805 -46,968 1, 163
-52 60,831,589 60,833,556 -50,956 -52,923 1,967
Coordinates of the putative enhancer fragments cloned in the enhancer reporter assays.
Chromosome 2 coordinates listed in hgl9 as well as in reference to the BCLllA TSS. Table 8. Oligonucleotide sequences.
Name Sequence Assay
mBcll la-5'-F AAAGAGCTGTCCGAAGTCCA TALEN deletion PCR
mBcll la-5'-R GGGCACTTCCTAGTCCCTCT TALEN deletion PCR
mBcll la-del 1-F TTTGAGCAGGAGGGAATTTG TALEN deletion PCR
mBcll la-del 1-R ATGTTGTGGTCCCTGTGGTT TALEN deletion PCR
mBcll la-del2-F GCAAGGCAGGTACCAAACAT TALEN deletion PCR
mBcll la-del2-R TAGAGATTCCAGGCCCCTTT TALEN deletion PCR
mBcll la-3'-F AGCAAGGAAAGGTGAAGCAG TALEN deletion PCR
mBcll la-3'-R CCCAATGTCTTCCGAACTGT TALEN deletion PCR
mBcll la- AGGCTGGTCTTGGGATTTTT TALEN deletion PCR
upstreamTALEN-F
mBcll la- GCCTTTAACAAGGGTGTCCA TALEN deletion PCR
downstreamTALEN-R
mBcll la-5'probe-F CATAGACCTGGGTCCTGGAA 5'-probe for Southern blot mBcll la-5'probe-R TTGCAGAGTGACTCCTGTGG 5'-probe for Southern blot hBCLl lA-52.0-F CCAGCCATACCCAAAACAAA lacZ reporter cloning
hBCLHA-64.4-R CTTTCCCTCTTGCCACTCAG lacZ reporter cloning
hBCLHA-56.8-F GGCAGAGAAGGCACAGTGA lacZ reporter cloning
hBCLHA-57.6-R GGCTGTCCTGGCATGTAAGT lacZ reporter cloning
hBCLHA-63.4-F AACAGACCCATGTGCTAGGC lacZ/GFP reporter cloning hBCLHA-61.6-R TGTGTGGACTGCCTTTTCTG lacZ/GFP reporter cloning hBCLHA-58.6-F GGGAAAAGGGAGAGGAAAAA lacZ reporter cloning
hBCLHA-57.6-R CTCAGAAAAATGACAGCACCA lacZ reporter cloning
hBCLHA-55.7-F GGACTCAGTGGCCTCTTTTG lacZ reporter cloning
hBCLHA-54.4-R GAAGATAATGGCAGCCCAGA lacZ reporter cloning
hBCLHA-164.2-F TGTGTGGCCAACCTGTAAAA GFP reporter cloning
hBCLHA-162.6-R CTCGCTCTGTTTCCCAGTTC GFP reporter cloning
hBCLHA-157.1-F CTCTCCGACGACCTCTTTTG GFP reporter cloning
hBCLHA-155.6-R GTAGGGAAGGGGCTACTTGG GFP reporter cloning
hBCLHA-154.1-F AGAGCCAAACTCCGTCTCAA GFP reporter cloning
hBCLHA-152.5-R AAATACCACAGCCCAACAGC GFP reporter cloning
hBCLHA-59.4-F GAACAGAGACCACTACTGGCAAT GFP reporter cloning
hBCLHA-57.7-R GGGGAAGGGGTATTGAATTG GFP reporter cloning
hBCLHA-55.9-F CTTCCACTGGATGGCACTTT GFP reporter cloning
hBCLHA-54.2-R ACTTCAGCCTCCAGCACTGT GFP reporter cloning
hBCLHA-41.6-F CCTCCCAGCAATGTAGGTGT GFP reporter cloning
hBCLHA-40.5-R TGGTGTGGTCCACTGTGACT GFP reporter cloning
hBCLHA-32.6-F GCAAGCTTAGCCCCTTCTTT GFP reporter cloning
hBCLHA-31.6-R TGAGGCAGAGTCAGATGTGG GFP reporter cloning
hBCLHA-n45.8-F CCCCGCTCAGAGTAAGTGAG GFP reporter cloning
hBCLl lA-n47.0-R GGAAACTGCCTATCCCATGA GFP reporter cloning
hBCLHA-n51.0-F CAACACCCCGATTTCAGACT GFP reporter cloning
hBCLHA-n52.9-R GAATGGTCCCGATCTCTTGA GFP reporter cloning
mGapdh-RT-F TGGTGAAGGTC GGTGTGAAC RT-qPCR (Gapdh)
mGapdh-RT-R CCATGTAGTTGAGGTCAATGAAGG RT-qPCR (Gapdh) mBcll la-RT-ele2-F AACCCCAGCACTTAAGCAAA RT-qPCR (Bell la exon-1/2) mBcll la-RT-ele2-R ACAGGTGAGAAGGTCGTGGT RT-qPCR (Bell la exon-1/2) mBcll la-RT-e2e3-F GCCCCAAACAGGAACACATA RT-qPCR (Bell la exon-2/3) mBcll la-RT-e2e3-R GGGGCATATTCTGCACTCAT RT-qPCR (Bell la exon-2/3) mBcll la-RT-e4e4-F ATGCGAGCTGTGCAACTATG RT-qPCR (Bell la exon-4/4, XLisoform) mBcll la-RT-e4e4-R GTAAACGTCCTTCCCCACCT RT-qPCR (Bell la exon-4/4, XLisoform) mBcll la-RT-e4e5-F CAGCTCAAAAGAGGGCAGAC RT-qPCR (Bell la exon-4/5, Lisoform) mBcll la-RT-e4e5-R GAGCTTCCATCCGAAAACTG RT-qPCR (Bell la exon-4/5, Lisoform) mHbby-RT-F TGGCCTGTGGAGTAAGGTCAA RT-qPCR (eY)
mHbby-RT-R GAAGCAGAGGACAAGTTCCCA RT-qPCR (eY)
mHbb-bhl-RT-F TGGACAACCTCAAGGAGACC RT-qPCR (bHl)
mHbb-bhl-RT-R ACCTCTGGGGTGAATTCCTT RT-qPCR (bHl)
mHbb-bl-RT-F TTTAACGATGGCCTGAATCACTT RT-qPCR (bl b2)
mHbb-bl-RT-R CAGCACAATCACGATCATATTGC RT-qPCR (bl b2)
lacZ-RT-F GCCAACATTGAGACACATGG RT-qPCR (lacZ)
lacZ-RT-R TGTCTCTCTGCACCATCCTG RT-qPCR (lacZ)
lacZ-F TTCAATGCTGTCAGGTGCTC PCR genotyping (lacZ)
lacZ-R GCCATGTGTCTCAATGTTGG PCR genotyping (lacZ)
rs7569946-F GTCTGCCCTCTTTTGAGCTG liaplotyping fusion PCR
rs7569946-R GACTCCAGACAATCGCCTTT liaplotyping fusion PCR
rs7569946-R-rc- AAAGGCGATTGTCTGGAGTCAACCTT bridging primer, liaplotyping fusion rsl427407-F CTTAGCACCCACAAAC PCR
rsl427407-R CATGTTACTGCAACTTGCTTTTT liaplotyping fusion PCR
rs7569946-nested-F AGATCCCTCCGTCCAGCTC liaplotyping fusion PCR
rsl427407-nested-R TGAAAGTTCAAGTAGATATCAGAAGG liaplotyping fusion PCR
3C-nBCLl lA-150.6-F AGCAAACCACACAGACTGAAGA 3C
3C-nBCLl lA-140.9-F CCAGAGCCATTTACGTCACA 3C
3C-nBCLl lA-114.1-F CAGAAGGGAATAAGGTACTCTGGA 3C
3C-nBCLl lA-111.5-F GTTTGGGCCTCAAGGTCTTT 3C
3C-nBCLl lA-109.1-F GAGGTTGGGAGTAAGCATTCTG 3C
3C-nBCLl lA-100.7-F ACGCATCAGAATGCCCATAG 3C
3C-nBCLl lA-92.3-F TTTTGAAAGAAAACGCTGACA 3C
3C-nBCLl lA-80.2-F TTCCAGCTGGTTAAATTTAGGG 3C
3C-nBCLl lA-77.2-F AGAAGGGGCCAGAAGAACAG 3C
3C-nBCLl lA-72.5-F CCTTCTTTTTCTTTCTTGGTTGC 3C
3C-nBCLl lA-66.8-F CCCTGCGTGCCATTAAAATA 3C
3C-nBCLl lA-61.2-F AAAGGCCTTGGGAAGAAAGA 3C
3C-nBCLl lA-59.1-F GCAAGTCAGTTGGGAACACA 3C
3C-nBCLl lA-57.1-F GGACTCAGTGGCCTCTTTTG 3C
3C-nBCLl lA-52.2-F CTGTCTCTGTCTCCCCCAAG 3C
3C-nBCLl lA-47-F CCAATGCTCCTGTAACAAAGG 3C
3C-nBCLl lA-43.5-F AATGCAGTAGGCAAAGAAGCA 3C
3C-nBCLl lA-38.6-F GAAATTTGGAAGGCCACAGA 3C
3C-nBCLl lA-29.3-F GCTTGCAACAATTAAAAGATGG 3C
3C-nBCLl lA-27.1-F GGTGACAAGGGAGAACCACT 3C
3C-nBCLl lA-20.9-F TGATTTCCTTGCAGCCTTTT 3C
3C-nBCLl lA-8.6-F CACACCCACAGCAACAAATG 3C
3C-nBCLl lApromoter-R TGCAGAGATCCCCCAAAGTA 3C
3C-nBCLl lA-n8.3-F CTCAGGGAGCAAGGGAAATA 3C
3C-nBCLl lA-nl2.6-F CCCTCCCAACAGGGATTTAT 3C
3C-nBCLl lA-nl9.5-F CAAAATTGAACACCTATGGTCTGA 3C
3C-nBCLl lA-n29.8-F AGGAAGACTTTGGCCTCCAT 3C
3 C-hBCL l l A-n34.6-F TTCC AAAC AATTATAC ACC AAC AAA 3 C
3C-nBCLl lA-n54-F TTTCATGGGGAATAGCCAAC 3C
3C-nBCLl lA-n78.2-F CCCTACTTGTTATTTGCTTCTGC 3C
3C-nBCLl 1 A-nl04.4-F AGC TG AAGTTTC AGGGAC C A 3C
3C-LCR-HS 1 -F CCAC ACCTGCCTTCCTTAGA 3C
3C-LCR-HS3-F TGCATATGATGGGGTAGCAG 3C
ChlP-nBCLl 1A-68.7-F AAGAGAAGGGGGAATTTGGA ChlP-qPCR
ChlP-nBCLl 1A-68.7-R TGGTGATAAGGGCAGGAAAC ChlP-qPCR
ChlP-nBCLl 1A-65.5-F AGGAAGCTGCAGAAAGGTGA ChlP-qPCR
ChlP-nBCLl 1A-65.5-R TGCTTCCCCAGGTTTAGATG ChlP-qPCR
ChlP-nBCLl 1A-64.7-F CCACTGCTACCCAAAACGAT ChlP-qPCR
ChlP-nBCLl 1A-64.7-R CAAGAGCGAAACTCCACCTC ChlP-qPCR ChlP-hBCLl 1A-63.9-F ACTGTGTGCCAAGTGACCAG ChlP-qPCR ChlP-hBCL 11 A-63.9-R CAGCTTCCTTCAGGTGCTTC ChlP-qPCR ChlP-hBCL 11 A-63.1 -F C ATGCTGCCTTTGTCTTCTG ChlP-qPCR ChlP-hBCL 11 A-63.1 -R TGTGGAGCTCTGGAATGATG ChlP-qPCR ChlP-hBCL 11 A-63.0-F GAGCTCCACAATCCAACTCC ChlP-qPCR ChlP-hBCL 11 A-63.0-R CCAGGAAGGAAATGAGAACG ChlP-qPCR ChlP-hBCL 11A-62.5-F ACCCACAAACATTTCCCTTCT ChlP-qPCR ChlP-hBCL 11A-62.5-R TTTGCTCTTCTCCAGGGTGT ChlP-qPCR ChlP-hBCL 11A-62.4-F TTTAAACAGCCACCCCACAC ChlP-qPCR ChlP-hBCL 11 A-62.4-R ACCACGTAGTTGGGCTTCAC ChlP-qPCR ChlP-hBCL 11A-62.2-F TTTCAACCATGGTCATCTGC ChlP-qPCR ChlP-hBCL 11A-62.2-R CCCTCTGGCATCAAAATGAG ChlP-qPCR ChIP-hBCLl lA-61.8-F GAACCTGGGAGGCAGAAGAT ChlP-qPCR ChIP-hBCLl lA-61.8-R TTTTTGGTGAGACGGAGATTT ChlP-qPCR ChIP-hBCLl lA-61.7-F CCGGGCAACAAGAGTAAATC ChlP-qPCR ChlP-hBCL 11 A-61.7-R ATGCCTAGGGTGTTTTGACG ChlP-qPCR ChIP-hBCLl lA-61.5-F CTCCGTGTTGAGAGCCAAGT ChlP-qPCR ChIP -hBCL 11 A-61.5-R TGTGTGGACTGCCTTTTCTG ChlP-qPCR ChlP-hBCL 11 A-61.3 -F C AGAAAAGGC AGTCC AC AC A ChlP-qPCR ChlP-hBCL 11 A-61.3 -R CCTCTCC AGATTCCCTCTC A ChlP-qPCR ChIP -hBCL 11 A-61.0-F AGCGAGACCCTGTCTCAAAA ChlP-qPCR ChIP -hBCL 11 A-61.0-R TCCAGCAGGCTTCAAAAAGT ChlP-qPCR ChlP-hBCL 11A-60.8-F GGTGGATAACCCCATCTCAG ChlP-qPCR ChlP-hBCL 11A-60.8-R GGAAATGAGAATGCCCTTTG ChlP-qPCR ChlP-hBCL 11A-60.5-F CAGTCTAGAAAGCCCCCTCA ChlP-qPCR ChlP-hBCL 11A-60.5-R GTGGGGGTTCAGTGGTTAGA ChlP-qPCR ChlP-hBCL 11A-60.3-F TCCATGGTGTGGAGTGTGTT ChlP-qPCR ChlP-hBCL 11 A-60.3-R ACCCACATGGCAACCAATAG ChlP-qPCR ChlP-hBCL 11A-60.0-F CCATTCCCTGGAGAGTTCAA ChlP-qPCR ChlP-hBCL 11A-60.0-R GGGGTCTCTTCCCATCATTT ChlP-qPCR ChlP-hBCL 11A-59.9-F ATGGGAAGAGACCCCAAAAC ChlP-qPCR ChlP-hBCL 11A-59.9-R GGACTCCGAACACCACACTT ChlP-qPCR ChlP-hBCL 11A-59.5-F GGGATCAGAGGTGAACAGGA ChlP-qPCR ChlP-hBCL 11A-59.5-R TTTAATCAGCTTCCGCCACT ChlP-qPCR ChlP-hBCL 11A-59.0-F TGGGGAGAGAAGAGTGGAAA ChlP-qPCR ChlP-hBCL 11A-59.0-R TTGCCAATTGGAGATTAGGG ChlP-qPCR ChlP-hBCL 11A-58.7-F TGCTCCGAGCTTGTGAACTA ChlP-qPCR ChlP-hBCL 11A-58.7-R GGGAAAGGGCCTGATAACTT ChlP-qPCR ChlP-hBCL 11A-58.3-F GAGAGTGCAGACAGGGGAAG ChlP-qPCR ChlP-hBCL 11A-58.3-R CCTCTTTCGGAAGGCTCTCT ChlP-qPCR ChlP-hBCL 11A-58.0-F TGGACTTTGCACTGGAATCA ChlP-qPCR ChlP-hBCL 11A-58.0-R GATGGCTGAAAAGCGATACA ChlP-qPCR ChlP-hBCL 11A-57.3-F GGGGAGATGATTGAAAGCAA ChlP-qPCR ChlP-hBCL 11 A-57.3-R AGAACTTTCCCGGTTCTGGT ChlP-qPCR ChlP-hBCL 11A-57.0-F GCTCTGGACACACAGCAAAA ChlP-qPCR ChlP-hBCL 1 1A-57.0-R TCAAATCCTTGCCTTGAACC ChlP-qPCR ChlP-hBCL 1 1A-56.6-F CCTCAAATCTCCCTCACTGG ChlP-qPCR ChlP-hBCL 1 1A-56.6-R GGGAA ATGGGTCC TGC TTT A ChlP-qPCR ChlP-hBCL 1 1A-56.3-F AGGGAGTACACCGCAGACAC ChlP-qPCR ChlP-hBCL 1 1A-56.3-R AAGGAAGGCTGCAAGGAAAT ChlP-qPCR ChlP-hBCL 1 1A-55.9-F GACTTAAACTGCCGCTCCTG ChlP-qPCR ChlP-hBCL 1 1A-55.9-R TGACTGGTAAGAGCCGATTG ChlP-qPCR ChlP-hBCL 1 1A-55.3-F GCTGGGGTGAGTCAAAAGTC ChlP-qPCR ChlP-hBCL 1 1A-55.3-R GGTCACCTTAAGGAGCCACA ChlP-qPCR ChlP-hBCL 1 1A-54.8-F GCACCTGCATTTGTTTTTCA ChlP-qPCR ChlP-hBCL 1 1A-54.8-R GGGTCAGATCACCTCTGCTC ChlP-qPCR ChlP-hBCL 1 1A-54.4-F AGGCATCCAAAGGGAAGAAT ChlP-qPCR ChlP-hBCL 1 1A-54.4-R GAAGATAATGGCAGCCCAGA ChlP-qPCR ChlP-hBCL 1 1A-54.0-F TGGGAAAGGTTGCACATTCT ChlP-qPCR ChlP-hBCLl 1A-54.0-R GGGCCTCAGGCTCTTTATCT ChlP-hBCLl 1A-53.4-F CCACTGCCAGGCTGTTTACT ChlP-hBCLl 1A-53.4-R GACCGAAAGGAGGAGAGGAG ChlP-hBCLl 1A-53.1-F CAGTTCCCCCATTATGCACT ChlP-hBCLl 1A-53.1-R CCCTTCTCTGAAGGCACATC ChlP-hBCLl 1A-52.7-F TTCAAGCCTTGGTGGATAGG ChlP-hBCLl 1A-52.7-R GCCAGGAAATTGGTGGTAGA ChlP-hBCLl 1A-52.3-F TGCCCACATGAGACATCTTT ChlP-hBCLl 1A-52.3-R AAATTGGCTGCCATTGAATC ChlP-hBCLl 1A-51.3-F CCACCAGAAGTCCTGGAAAA ChlP-hBCLl 1A-51.3-R TTGGAGGGACCTGATCTCTG ChlP-hBCLl 1A-50.2-F CCAAGATGGAGAAGCCACAT ChlP-hBCLl 1A-50.2-R TCTGTCTTGGGTCTCCTGGT ChlP-hBCLl 1A-49.8-F GAGAAGCCCTCAGCAAACAC ChlP-hBCLl 1A-49.8-R GGTTGCATCTTGGCTCCTAA ChlP-hBCLl 1A-49.5-F GAAATGCAGGAAAGGAACGA ChlP-hBCLl 1A-49.5-R TCTAGCAGATGGGGTTTTGG ChIP-hOct4-prom-F AGTC TGGGC AAC AAAGTGAGA ChIP-hOct4-prom-R AGAAACTGAGGAGAAGGATG ChIP-hHS3-F ATAGACCATGAGTAGAGGGCAGAC ChIP-hHS3-R TGATCCTGAAAACATAGGAGTCAA ChIP-hHS-40-F CAGATAACTGGGCCAACCAT ChIP-hHS-40-R ATTCACCCCTTTCCCTTGTC ChlP-hGAPDH-F CGTAGCTCAGGCCTCAAGAC ChlP-hGAPDH-R CGAACAGGAGGAGCAGAGAG
Oligonucleotides used in indicated experiments.

Claims

CLAIMS What is claimed:
1. A method for producing a progenitor cell having decreased BCL 11 A mRNA or protein expression, the method comprising contacting an isolated progenitor cell with an agent that binds the genomic DNA of the cell on chromosome 2 location 60,716,189- 60,728,612 (according to UCSC Genome Browser hg 19 human genome assembly), thereby reducing the mRNA or protein expression of BCL11A.
2. A method for producing an isolated genetic engineered human cell having at least one genetic modification comprising contacting an isolated cell with an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA-targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location
60,716,189-60,728,612 causing at least one genetic modification therein.
3. The method of claim 1 or 2, wherein the isolated progenitor cell or isolated cell is a hematopoietic progenitor cell.
4. The method of claim 3, wherein the hematopoietic progenitor is a cell of the erythroid lineage.
5. The method of claim 1 or 2, wherein the isolated progenitor cell or isolated cell is an induced pluripotent stem cell.
6. The method of claim 3, wherein the hematopoietic progenitor cell is contacted ex vivo or in vitro.
7. The method of any one of claims 1-6, wherein the at least one genetic modification is a deletion.
8. The method of claim 7, wherein the deletion removes the entire region between
chromosome 2 location 60,716,189-60,728,612 or removes a portion of the region resulting in disruption of one of more DNAse 1 -hypersensitive sites (DHS).
9. An isolated genetic engineered human cell having at least one genetic modification on chromosome 2 location 60,716,189-60,728,612 according to claims 2-8.
10. A composition comprising isolated genetic engineered human cells of claim 9.
11. A method of increasing fetal hemoglobin levels in a cell, the method comprising the steps of: contacting an isolated cell with an effective amount of a composition comprising at least a DNA-targeting endonuclease or a vector carrying the coding sequence of a DNA-targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location 60,716,189-60,728,612 causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in said cell, or its progeny, relative to said cell prior to said contacting.
12. The method of claim 11, wherein the isolated cell is a hematopoietic progenitor cell.
13. The method of claim 11 or 12, wherein the hematopoietic progenitor cell is a cell of the erythroid lineage.
14. The method of claim 11, wherein the isolated cell is an induced pluripotent stem cell.
15. The method of claim 12 or 13, wherein the hematopoietic progenitor cell is contacted ex vivo or in vitro.
16. The method of any one of claims 11-15, wherein the at least one genetic modification is a deletion.
17. The method of claim 16, wherein the deletion removes the entire region between
chromosome 2 location 60,716,189-60,728,612 or removes a portion of the region resulting in disruption of one of more DNAse 1 -hypersensitive sites (DHS).
18. A method for increasing fetal hemoglobin levels in a mammal in need thereof, the
method comprising the steps of contacting an isolated hematopoietic progenitor cell in said mammal with an effective amount of a composition comprising at least a DNA- targeting endonuclease or a vector carrying the coding sequence of a DNA-targeting endonuclease whereby the DNA-targeting endonuclease cleaves the genomic DNA of the cell on chromosome 2 location 60,716,189-60,728,612 causing at least one genetic modification therein, whereby fetal hemoglobin expression is increased in said mammal, relative to expression prior to said contacting.
9. A method for increasing fetal hemoglobin levels in a mammal in need thereof, the method comprising transplanting an isolated genetic engineered human cell of claim 9 or a composition of claim 10 into the mammal.
PCT/US2013/072236 2012-11-27 2013-11-27 Targeting bcl11a distal regulatory elements for fetal hemoglobin reinduction WO2014085593A1 (en)

Priority Applications (23)

Application Number Priority Date Filing Date Title
JP2015544205A JP6542124B2 (en) 2012-11-27 2013-11-27 Targeting of the BCL11A distal regulatory element for reinduction of fetal hemoglobin
EP13858006.3A EP2925864B1 (en) 2012-11-27 2013-11-27 Targeting bcl11a distal regulatory elements for fetal hemoglobin reinduction
US14/647,547 US9822355B2 (en) 2012-11-27 2013-11-27 Targeting BCL11A distal regulatory elements for fetal hemoglobin reinduction
KR1020157016749A KR102240555B1 (en) 2012-11-27 2013-11-27 Targeting bcl11a distal regulatory elements for fetal hemoglobin reinduction
BR112015011995-6A BR112015011995B1 (en) 2012-11-27 2013-11-27 METHOD FOR PRODUCING A HEMATOPOIETIC PROGENITOR CELL HAVING DECREASE BCL11A OR FOR INCREASE ITS FETAL HEMOGLOBIN LEVELS, COMPOSITION AND USE THEREOF
SG11201504038XA SG11201504038XA (en) 2012-11-27 2013-11-27 Targeting bcl11a distal regulatory elements for fetal hemoglobin reinduction
EP18203436.3A EP3502240B1 (en) 2012-11-27 2013-11-27 Targeting bcl11a distal regulatory elements for fetal hemoglobin reinduction
CN201380071452.2A CN104955943B (en) 2012-11-27 2013-11-27 The distal ends the targeting BCL11A controlling element induced again for fetal hemoglobin
DK13858006.3T DK2925864T3 (en) 2012-11-27 2013-11-27 DIRECTIONAL TARGETING OF DISTANT BCL11A CONTROLS FOR FETAL HEMOGLOBIN REINUCTION
MX2015006528A MX361412B (en) 2012-11-27 2013-11-27 Targeting bcl11a distal regulatory elements for fetal hemoglobin reinduction.
CA2892860A CA2892860C (en) 2012-11-27 2013-11-27 Targeting bcl11a distal regulatory elements for fetal hemoglobin reinduction
AU2013352156A AU2013352156B2 (en) 2012-11-27 2013-11-27 Targeting BCL11A distal regulatory elements for fetal hemoglobin reinduction
ES13858006T ES2708948T3 (en) 2012-11-27 2013-11-27 Distal regulatory elements of bcl11a as targets for the reinduction of fetal hemoglobin
CN201810979764.8A CN109554350B (en) 2012-11-27 2013-11-27 Targeting BCL11A distal regulatory elements for fetal hemoglobin re-induction
IL238949A IL238949B (en) 2012-11-27 2015-05-21 Targeting bcl11a distal regulatory elements for fetal hemoglobin reinduction
SA515360489A SA515360489B1 (en) 2012-11-27 2015-05-27 Targeting bcl11a distal regulatory elements for fetal hemoglobin reinduction
ZA2015/04004A ZA201504004B (en) 2012-11-27 2015-06-04 Targeting bcl11a distal regulatory elements for fetal hemoglobin reinduction
HK16103646.4A HK1215720A1 (en) 2012-11-27 2016-03-30 Targeting bcl11a distal regulatory elements for fetal hemoglobin reinduction bcl11a
US15/816,083 US10472619B2 (en) 2012-11-27 2017-11-17 Targeting BCL11A distal regulatory elements for fetal hemoglobin reinduction
AU2018278850A AU2018278850B2 (en) 2012-11-27 2018-12-11 Targeting bcl11a distal regulatory elements for fetal hemoglobin reinduction
US16/573,389 US11542493B2 (en) 2012-11-27 2019-09-17 Targeting BCL11A distal regulatory elements for fetal hemoglobin reinduction
AU2021218012A AU2021218012B2 (en) 2012-11-27 2021-08-16 Targeting bcl11a distal regulatory elements for fetal hemoglobin reinduction
US18/074,107 US20230348887A1 (en) 2012-11-27 2022-12-02 Targeting bcl11a distal regulatory elements for fetal hemoglobin reinduction

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US201261730369P 2012-11-27 2012-11-27
US201261730323P 2012-11-27 2012-11-27
US61/730,323 2012-11-27
US61/730,369 2012-11-27
US201361776144P 2013-03-11 2013-03-11
US61/776,144 2013-03-11
US201361889174P 2013-10-10 2013-10-10
US61/889,174 2013-10-10

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/647,547 A-371-Of-International US9822355B2 (en) 2012-11-27 2013-11-27 Targeting BCL11A distal regulatory elements for fetal hemoglobin reinduction
US15/816,083 Division US10472619B2 (en) 2012-11-27 2017-11-17 Targeting BCL11A distal regulatory elements for fetal hemoglobin reinduction

Publications (1)

Publication Number Publication Date
WO2014085593A1 true WO2014085593A1 (en) 2014-06-05

Family

ID=50828461

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/072236 WO2014085593A1 (en) 2012-11-27 2013-11-27 Targeting bcl11a distal regulatory elements for fetal hemoglobin reinduction

Country Status (18)

Country Link
US (4) US9822355B2 (en)
EP (2) EP2925864B1 (en)
JP (2) JP6542124B2 (en)
KR (1) KR102240555B1 (en)
CN (2) CN109554350B (en)
AU (3) AU2013352156B2 (en)
BR (1) BR112015011995B1 (en)
CA (1) CA2892860C (en)
DK (2) DK3502240T3 (en)
ES (2) ES2708948T3 (en)
HK (1) HK1215720A1 (en)
IL (1) IL238949B (en)
MX (1) MX361412B (en)
PT (2) PT3502240T (en)
SA (1) SA515360489B1 (en)
SG (3) SG11201504038XA (en)
WO (1) WO2014085593A1 (en)
ZA (1) ZA201504004B (en)

Cited By (82)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015033343A1 (en) 2013-09-03 2015-03-12 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Compositions and methods for expressing recombinant polypeptides
US9068179B1 (en) 2013-12-12 2015-06-30 President And Fellows Of Harvard College Methods for correcting presenilin point mutations
US9163284B2 (en) 2013-08-09 2015-10-20 President And Fellows Of Harvard College Methods for identifying a target site of a Cas9 nuclease
US9228207B2 (en) 2013-09-06 2016-01-05 President And Fellows Of Harvard College Switchable gRNAs comprising aptamers
WO2016005985A2 (en) 2014-07-09 2016-01-14 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Method for reprogramming cells
WO2016038616A1 (en) 2014-09-14 2016-03-17 Yeda Research And Development Co. Ltd. Nmda receptor antagonists for treating gaucher disease
US9322006B2 (en) 2011-07-22 2016-04-26 President And Fellows Of Harvard College Evaluation and improvement of nuclease cleavage specificity
US9322037B2 (en) 2013-09-06 2016-04-26 President And Fellows Of Harvard College Cas9-FokI fusion proteins and uses thereof
WO2016135557A2 (en) 2015-02-23 2016-09-01 Crispr Therapeutics Ag Materials and methods for treatment of hemoglobinopathies
WO2016135559A2 (en) 2015-02-23 2016-09-01 Crispr Therapeutics Ag Materials and methods for treatment of human genetic diseases including hemoglobinopathies
JP2016529907A (en) * 2013-09-04 2016-09-29 シーエスアイアール Site-specific nuclease single-cell assay targeting gene regulatory elements that silence gene expression
WO2016182917A1 (en) * 2015-05-08 2016-11-17 Children's Medical Center Corporation Targeting bcl11a enhancer functional regions for fetal hemoglobin reinduction
US9526784B2 (en) 2013-09-06 2016-12-27 President And Fellows Of Harvard College Delivery system for functional nucleases
WO2017009842A2 (en) 2015-07-16 2017-01-19 Biokine Therapeutics Ltd. Compositions and methods for treating cancer
EP3068881A4 (en) * 2013-11-13 2017-04-19 Children's Medical Center Corporation Nuclease-mediated regulation of gene expression
WO2017077394A2 (en) 2015-11-04 2017-05-11 Crispr Therapeutics Ag Materials and methods for treatment of hemoglobinopathies
WO2017118985A1 (en) 2016-01-06 2017-07-13 Yeda Research And Development Co. Ltd. Compositions and methods for treating malignant, autoimmune and inflammatory diseases
WO2017125931A1 (en) 2016-01-21 2017-07-27 The State Of Israel, Ministry Of Agriculture & Rural Development, Agricultural Research Organization (Aro) (Volcani Center) Parthenocarpic plants and methods of producing same
WO2017130205A1 (en) 2016-01-31 2017-08-03 Hadasit Medical Research Services And Development Ltd. Autosomal-identical pluripotent stem cell populations having non-identical sex chromosomal composition and uses thereof
WO2017138008A2 (en) 2016-02-14 2017-08-17 Yeda Research And Development Co. Ltd. Methods of modulating protein exocytosis and uses of same in therapy
WO2017153982A1 (en) 2016-03-06 2017-09-14 Yeda Research And Development Co. Ltd. Method for modulating myelination
US9834791B2 (en) 2013-11-07 2017-12-05 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
WO2018033929A1 (en) 2016-08-18 2018-02-22 Yeda Research And Development Co. Ltd. Diagnostic and therapeutic uses of exosomes
WO2018096547A1 (en) 2016-11-28 2018-05-31 Yeda Research And Development Co. Ltd. Isolated polynucleotides and polypeptides and methods of using same for expressing an expression product of interest
JP2018516556A (en) * 2015-05-12 2018-06-28 サンガモ セラピューティクス, インコーポレイテッド Nuclease-mediated gene expression regulation
WO2018122771A1 (en) 2016-12-29 2018-07-05 Ukko Inc. Methods for identifying and de-epitoping allergenic polypeptides
WO2018142416A1 (en) 2017-02-06 2018-08-09 Yeda Research And Development Co. Ltd. Isolated cells genetically modified to express a disarm system having an anti-phage activity and methods of producing same
US10077453B2 (en) 2014-07-30 2018-09-18 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
US10113163B2 (en) 2016-08-03 2018-10-30 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
WO2018207178A1 (en) 2017-05-07 2018-11-15 Yeda Research And Development Co. Ltd. Methods of treating psychiatric stress disorders
US10167457B2 (en) 2015-10-23 2019-01-01 President And Fellows Of Harvard College Nucleobase editors and uses thereof
WO2019021284A1 (en) 2017-07-24 2019-01-31 Yeda Research And Development Co. Ltd. Combination therapy for the treatment of cancer
WO2019038771A1 (en) 2017-08-23 2019-02-28 Technion Research & Development Foundation Limited Compositions and methods for improving alcohol tolerance in yeast
US10287588B2 (en) 2014-04-25 2019-05-14 The Childrens's Medical Center Corporation Compositions and methods to treating hemoglobinopathies
WO2019097514A1 (en) 2017-11-14 2019-05-23 Yeda Research And Development Co. Ltd. Hematopoietic stem cells with improved properties
WO2019145964A1 (en) 2018-01-29 2019-08-01 Yeda Research And Development Co. Ltd. Combination of a mek inhibitor and a cdk4/6 inhibitor for the treatment of sarcoma
WO2020008412A1 (en) 2018-07-04 2020-01-09 Ukko Inc. Methods of de-epitoping wheat proteins and use of same for the treatment of celiac disease
WO2020089892A1 (en) 2018-10-28 2020-05-07 Yeda Research And Development Co. Ltd. Prevention of age related clonal hematopoiesis and diseases associated therewith
US20200155606A1 (en) * 2017-05-10 2020-05-21 Editas Medicine, Inc. Crispr/rna-guided nuclease systems and methods
WO2020112979A3 (en) * 2018-11-30 2020-07-02 Bauer Daniel E Therapeutic gene editing for elane-associated disease
US10745677B2 (en) 2016-12-23 2020-08-18 President And Fellows Of Harvard College Editing of CCR5 receptor gene to protect against HIV infection
US10767156B2 (en) 2013-10-24 2020-09-08 Yeda Research And Development Co., Ltd. Polynucleotides encoding BREX system polypeptides and methods of using same
WO2020178822A1 (en) 2019-03-05 2020-09-10 The State Of Israel, Ministry Of Agriculture & Rural Development, Agricultural Research Organization (Aro) (Volcani Center) Genome-edited birds
WO2021001784A1 (en) 2019-07-04 2021-01-07 Ukko Inc. De-epitoped alpha gliadin and use of same for the management of celiac disease and gluten sensitivity
WO2021009763A1 (en) 2019-07-16 2021-01-21 Yeda Research And Development Co. Ltd. Methods of treating pain
WO2021084540A1 (en) 2019-10-30 2021-05-06 Yeda Research And Development Co. Ltd. Inhibitors of mmej pathway for prevention and treatment of pre-myeloid and myeloid malignancies
WO2021130752A1 (en) 2019-12-22 2021-07-01 Yeda Research And Development Co. Ltd. Systems and methods for identifying cells that have undergone genome editing
WO2021152587A1 (en) 2020-01-30 2021-08-05 Yeda Research And Development Co. Ltd. Treating acute liver disease with tlr-mik inhibitors
JP2021191755A (en) * 2015-05-13 2021-12-16 セルジーン コーポレイション TREATMENT OF β-THALASSEMIA USING ActRII LIGAND TRAPS
US11261441B2 (en) 2017-03-29 2022-03-01 Bluebird Bio, Inc. Vectors and compositions for treating hemoglobinopathies
US11268077B2 (en) 2018-02-05 2022-03-08 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of hemoglobinopathies
US11268082B2 (en) 2017-03-23 2022-03-08 President And Fellows Of Harvard College Nucleobase editors comprising nucleic acid programmable DNA binding proteins
WO2022074646A1 (en) 2020-10-05 2022-04-14 Protalix Ltd. Dicer-like knock-out plant cells
US11306324B2 (en) 2016-10-14 2022-04-19 President And Fellows Of Harvard College AAV delivery of nucleobase editors
WO2022079719A1 (en) 2020-10-15 2022-04-21 Yeda Research And Development Co. Ltd. Method of treating myeloid malignancies
US11319532B2 (en) 2017-08-30 2022-05-03 President And Fellows Of Harvard College High efficiency base editors comprising Gam
WO2022115498A1 (en) 2020-11-26 2022-06-02 Ukko Inc. Modified high molecular weight glutenin subunit and uses thereof
WO2022130388A2 (en) 2020-12-18 2022-06-23 Yeda Research And Development Co. Ltd. Compositions for use in the treatment of chd2 haploinsufficiency and methods of identifying same
WO2022130384A1 (en) 2020-12-17 2022-06-23 Yeda Research And Development Co. Ltd. Controlling ubiquitination of mlkl for treatment of disease
US11390884B2 (en) 2015-05-11 2022-07-19 Editas Medicine, Inc. Optimized CRISPR/cas9 systems and methods for gene editing in stem cells
US11447770B1 (en) 2019-03-19 2022-09-20 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
US11466271B2 (en) 2017-02-06 2022-10-11 Novartis Ag Compositions and methods for the treatment of hemoglobinopathies
WO2022239001A1 (en) 2021-05-10 2022-11-17 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Pharmaceutical compositions for treating neurological conditions
WO2022264132A1 (en) 2021-06-13 2022-12-22 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Method for reprogramming human cells
US11542496B2 (en) 2017-03-10 2023-01-03 President And Fellows Of Harvard College Cytosine to guanine base editor
US11542509B2 (en) 2016-08-24 2023-01-03 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
US11542493B2 (en) 2012-11-27 2023-01-03 The Children's Medical Center Corporation Targeting BCL11A distal regulatory elements for fetal hemoglobin reinduction
US11560566B2 (en) 2017-05-12 2023-01-24 President And Fellows Of Harvard College Aptazyme-embedded guide RNAs for use with CRISPR-Cas9 in genome editing and transcriptional activation
EP4130028A1 (en) 2021-08-03 2023-02-08 Rhazes Therapeutics Ltd Engineered tcr complex and methods of using same
WO2023012584A2 (en) 2021-08-03 2023-02-09 Genicity Limited Engineered tcr complex and methods of using same
WO2023062636A1 (en) 2021-10-14 2023-04-20 Weedout Ltd. Methods of weed control
US11661590B2 (en) 2016-08-09 2023-05-30 President And Fellows Of Harvard College Programmable CAS9-recombinase fusion proteins and uses thereof
EP4218771A1 (en) 2015-03-27 2023-08-02 Yeda Research and Development Co. Ltd Methods of treating motor neuron diseases
US11732274B2 (en) 2017-07-28 2023-08-22 President And Fellows Of Harvard College Methods and compositions for evolving base editors using phage-assisted continuous evolution (PACE)
US11788087B2 (en) 2017-05-25 2023-10-17 The Children's Medical Center Corporation BCL11A guide delivery
US11795443B2 (en) 2017-10-16 2023-10-24 The Broad Institute, Inc. Uses of adenosine base editors
WO2023240282A1 (en) 2022-06-10 2023-12-14 Umoja Biopharma, Inc. Engineered stem cells and uses thereof
US11851690B2 (en) 2017-03-14 2023-12-26 Editas Medicine, Inc. Systems and methods for the treatment of hemoglobinopathies
US11866726B2 (en) 2017-07-14 2024-01-09 Editas Medicine, Inc. Systems and methods for targeted integration and genome editing and detection thereof using integrated priming sites
US11898179B2 (en) 2017-03-09 2024-02-13 President And Fellows Of Harvard College Suppression of pain by gene editing
US11911415B2 (en) 2015-06-09 2024-02-27 Editas Medicine, Inc. CRISPR/Cas-related methods and compositions for improving transplantation
US11912985B2 (en) 2020-05-08 2024-02-27 The Broad Institute, Inc. Methods and compositions for simultaneous editing of both strands of a target double-stranded nucleotide sequence

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3009727A1 (en) * 2015-12-28 2017-07-06 Novartis Ag Compositions and methods for the treatment of hemoglobinopathies
US11326183B2 (en) 2016-02-12 2022-05-10 Bluebird Bio, Inc. VCN enhancer compositions and methods of using the same
EP4151720A1 (en) 2016-02-12 2023-03-22 Bluebird Bio, Inc. Vcn enhancer compositions and methods of using the same
EP3430142A1 (en) * 2016-03-14 2019-01-23 Editas Medicine, Inc. Crispr/cas-related methods and compositions for treating beta hemoglobinopathies
US20200330609A1 (en) * 2016-04-18 2020-10-22 Crispr Therapeutics Ag Materials and methods for treatment of hemoglobinopathies
CN109722415B (en) * 2017-10-27 2021-01-26 博雅辑因(北京)生物科技有限公司 Hematopoietic stem cell culture composition, culture medium and hematopoietic stem cell culture method
US20210047632A1 (en) * 2018-01-26 2021-02-18 The Children's Medical Center Corporation Targeting bcl11a distal regulatory elements with a cas9-cas9 fusion for fetal hemoglobin reinduction
EP3749767A1 (en) 2018-02-05 2020-12-16 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of hemoglobinopathies
CN109486850B (en) * 2018-12-04 2021-05-25 厦门大学 Optogenetic tool for UV-B light-down regulation of chromatin long-distance interaction
US20220193142A1 (en) * 2019-04-30 2022-06-23 Edigene Inc. Method for predicting effectiveness of treatment of hemoglobinopathy
CN111939271A (en) * 2019-04-30 2020-11-17 博雅辑因(北京)生物科技有限公司 Method for predicting treatment effectiveness of hemoglobinopathy
CN111705044B (en) * 2020-05-23 2022-12-27 南京大学 Construction of novel controllable high-activity G quadruplex DNA enzyme
EP4337232A1 (en) * 2021-05-13 2024-03-20 The Children's Medical Center Corporation Methods for stratifying subjects for fetal hemoglobin reinduction

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110182867A1 (en) * 2008-09-15 2011-07-28 Children's Medical Center Corporation Modulation of bcl11a for treatment of hemoglobinopathies
US8021867B2 (en) 2005-10-18 2011-09-20 Duke University Rationally-designed meganucleases with altered sequence specificity and DNA-binding affinity

Family Cites Families (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5061620A (en) 1990-03-30 1991-10-29 Systemix, Inc. Human hematopoietic stem cell
US5635387A (en) 1990-04-23 1997-06-03 Cellpro, Inc. Methods and device for culturing human hematopoietic cells and their precursors
US5460964A (en) 1992-04-03 1995-10-24 Regents Of The University Of Minnesota Method for culturing hematopoietic cells
US5409813A (en) 1993-09-30 1995-04-25 Systemix, Inc. Method for mammalian cell separation from a mixture of cell populations
US5677136A (en) 1994-11-14 1997-10-14 Systemix, Inc. Methods of obtaining compositions enriched for hematopoietic stem cells, compositions derived therefrom and methods of use thereof
US5928638A (en) 1996-06-17 1999-07-27 Systemix, Inc. Methods for gene transfer
US5731156A (en) * 1996-10-21 1998-03-24 Applied Imaging, Inc. Use of anti-embryonic hemoglobin antibodies to identify fetal cells
US8101349B2 (en) 1997-12-23 2012-01-24 Novartis Vaccines And Diagnostics, Inc. Gene products differentially expressed in cancerous cells and their methods of use II
FR2777909B1 (en) 1998-04-24 2002-08-02 Pasteur Institut USE OF TRIPLEX-STRUCTURED DNA SEQUENCES FOR THE TRANSFER OF NUCLEOTID SEQUENCES IN CELLS, RECOMBINANT VECTORS CONTAINING THESE TRIPLEX SEQUENCES
TR200401292T3 (en) 2000-12-01 2004-07-21 Max@Planck@Gesellschaft�Zur�F�Rderung�Der�Wissenschaften the rnaágirişimineáyoláaçanáküçükárnaámolekül
JP2006507841A (en) 2002-11-14 2006-03-09 ダーマコン, インコーポレイテッド Functional and ultrafunctional siRNA
EP1581056B1 (en) 2002-12-13 2010-07-21 Genetix Pharmaceuticals Inc. Therapeutic retroviral vectors for gene therapy
CA2566286A1 (en) 2004-05-11 2005-12-08 Rnai Co., Ltd. Polynucleotide causing rna interfere and method of regulating gene expression with the use of the same
US20080051431A1 (en) 2006-05-26 2008-02-28 Dominique Verhelle Methods and compositions using immunomodulatory compounds in combination therapy
EP2172547B1 (en) 2007-06-11 2016-01-06 Takara Bio Inc. Method for expression of specific gene
GB0713183D0 (en) 2007-07-06 2007-08-15 King S College London Method
US20110294114A1 (en) 2009-12-04 2011-12-01 Cincinnati Children's Hospital Medical Center Optimization of determinants for successful genetic correction of diseases, mediated by hematopoietic stem cells
CN102802412A (en) 2009-12-08 2012-11-28 海玛奎斯特医药公司 Methods and low dose regimens for treating red blood cell disorders
JP2011135864A (en) * 2009-12-30 2011-07-14 Korea Univ Research & Business Foundation Composition for retrodifferentiating somatic cell to embryonic stem cell-like cell, by using oct4 in combination with bmi1 or upstream regulator thereof, and method for generating embryonic stem cell-like cell using the same
EP3502254A1 (en) 2010-04-23 2019-06-26 Cold Spring Harbor Laboratory Novel structurally designed shrnas
WO2012073047A2 (en) 2010-12-03 2012-06-07 Genome Research Limited Compositions and methods
WO2012079046A2 (en) * 2010-12-10 2012-06-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of klf-1 and bcl11a genes
US9789139B2 (en) 2011-06-10 2017-10-17 Bluebird Bio, Inc. Gene therapy vectors for adrenoleukodystrophy and adrenomyeloneuropathy
KR102011532B1 (en) 2011-09-30 2019-08-16 블루버드 바이오, 인코포레이티드. Compounds for improved viral transduction
DK2836226T3 (en) 2012-02-24 2017-09-18 Hutchinson Fred Cancer Res COMPOSITIONS AND PROCEDURES FOR TREATING HEMOGLOBINOPATHY
EP2850188A4 (en) 2012-05-16 2016-01-20 Rana Therapeutics Inc Compositions and methods for modulating hemoglobin gene family expression
EA038924B1 (en) 2012-05-25 2021-11-10 Те Риджентс Оф Те Юниверсити Оф Калифорния Methods and compositions for rna-directed target dna modification and for rna-directed modulation of transcription
SG11201504038XA (en) 2012-11-27 2015-06-29 Childrens Medical Center Targeting bcl11a distal regulatory elements for fetal hemoglobin reinduction
CN113528577A (en) 2012-12-12 2021-10-22 布罗德研究所有限公司 Engineering of systems, methods and optimized guide compositions for sequence manipulation
WO2014093965A1 (en) 2012-12-14 2014-06-19 Case Western Reserve University Genomic rna packaging enhancer element
CA2910489A1 (en) 2013-05-15 2014-11-20 Sangamo Biosciences, Inc. Methods and compositions for treatment of a genetic condition
WO2015065964A1 (en) 2013-10-28 2015-05-07 The Broad Institute Inc. Functional genomics using crispr-cas systems, compositions, methods, screens and applications thereof
SI3068881T1 (en) 2013-11-13 2019-05-31 Children's Medical Center Corporation Nuclease-mediated regulation of gene expression
CN104955119B (en) 2014-03-26 2019-11-26 南京中兴新软件有限责任公司 A kind of method for switching network based on flow, device and terminal
WO2015164739A1 (en) 2014-04-25 2015-10-29 Bluebird Bio, Inc. Kappa/lambda chimeric antigen receptors
CN106536549B (en) 2014-04-25 2020-01-17 蓝鸟生物公司 MND promoter chimeric antigen receptor
JP6514717B2 (en) 2014-04-25 2019-05-15 ザ チルドレンズ メディカル センター コーポレーション Compositions and methods for treating abnormal hemoglobinopathy
US10619156B2 (en) 2014-05-28 2020-04-14 The Regents Of The University Of California Hybrid tRNA/pre-miRNA molecules and methods of use
SI3628687T1 (en) 2014-12-12 2021-12-31 2Seventy Bio, Inc. Bcma chimeric antigen receptors
WO2016182893A1 (en) 2015-05-08 2016-11-17 Teh Broad Institute Inc. Functional genomics using crispr-cas systems for saturating mutagenesis of non-coding elements, compositions, methods, libraries and applications thereof
EP3294879A4 (en) 2015-05-14 2019-02-20 University of Southern California Optimized gene editing utilizing a recombinant endonuclease system
US11279769B2 (en) 2015-08-31 2022-03-22 Helixmith Co., Ltd Anti-Sialyl Tn chimeric antigen receptors
GB201522243D0 (en) 2015-12-16 2016-01-27 Ucl Business Plc Treatment
EP4151720A1 (en) 2016-02-12 2023-03-22 Bluebird Bio, Inc. Vcn enhancer compositions and methods of using the same
WO2018218135A1 (en) 2017-05-25 2018-11-29 The Children's Medical Center Corporation Bcl11a guide delivery

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8021867B2 (en) 2005-10-18 2011-09-20 Duke University Rationally-designed meganucleases with altered sequence specificity and DNA-binding affinity
US8119381B2 (en) 2005-10-18 2012-02-21 Duke University Rationally-designed meganucleases with altered sequence specificity and DNA-binding affinity
US8124369B2 (en) 2005-10-18 2012-02-28 Duke University Method of cleaving DNA with rationally-designed meganucleases
US8129134B2 (en) 2005-10-18 2012-03-06 Duke University Methods of cleaving DNA with rationally-designed meganucleases
US8133697B2 (en) 2005-10-18 2012-03-13 Duke University Methods of cleaving DNA with rationally-designed meganucleases
US8143016B2 (en) 2005-10-18 2012-03-27 Duke University Methods of cleaving DNA with rationally-designed meganucleases
US8143015B2 (en) 2005-10-18 2012-03-27 Duke University Methods of cleaving DNA with rationally-designed meganucleases
US8148098B2 (en) 2005-10-18 2012-04-03 Duke University Methods of cleaving DNA with rationally-designed meganucleases
US8163514B2 (en) 2005-10-18 2012-04-24 Duke University Methods of cleaving DNA with rationally-designed meganucleases
US8304222B1 (en) 2005-10-18 2012-11-06 Duke University Rationally-designed meganucleases with altered sequence specificity and heterodimer formation
US20110182867A1 (en) * 2008-09-15 2011-07-28 Children's Medical Center Corporation Modulation of bcl11a for treatment of hemoglobinopathies

Non-Patent Citations (96)

* Cited by examiner, † Cited by third party
Title
A. HE; S. W. KONG; Q. MA; W. T. PU, PROC. NATL. ACAD. SCI. U. S. A., vol. 108, 2011, pages 5632 - 5637
A. JOHN ET AL., DEVELOPMENT, vol. 139, 2012, pages 1831 - 1841
A. L. BREDEMEYER ET AL., NATURE, vol. 442, 2006, pages 466 - 470
A. MELNIKOV ET AL., NAT. BIOTECHNOL., vol. 30, 2012, pages 271 - 277
A. N. KOEHLER, CURR. OPIN. CHEM. BIOL., vol. 14, 2010, pages 331 - 340
ATWEH, SEMIN. HEMATOL., vol. 38, no. 4, 2001, pages 367 - 73
BAUER, DANIEL E. ET AL.: "Reawakening fetal hemoglobin: prospects for new therapies for the B-globin disorders", BLOOD, vol. 120, no. 15, 17 August 2012 (2012-08-17), pages 2945 - 2953, XP002721961 *
BLAU ET AL., BLOOD, vol. 81, 1993, pages 529 - 537
BUNN, N. ENGL. J. MED., vol. 328, 1993, pages 129 - 131
CAO, ANTONIO ET AL.: "Recent advances in B-thalassemias", PEDIATRIC REPORTS, vol. 3, no. 2, 2011, pages 65 - 74, XP055255428 *
CERTO, MT ET AL., NATURE METHODS, vol. 9, 2012, pages 073 - 975
CHEVALIER ET AL., NUCLEIC ACIDS RES., vol. 29, no. 18, 2001, pages 3757 - 3774
CONSTANTOULAKIS ET AL., BLOOD., vol. 72, no. 6, December 1988 (1988-12-01), pages 1961 - 7
D. E. BAUER; S. C. KAMRAN; S. H. ORKIN, BLOOD, vol. 120, 2012, pages 2945 - 2953
D. J. TURNER; M. E. HURLES, NAT. PROTOC., vol. 4, 2009, pages 1771 - 1783
D. NOORDERMEER; W. DE LAAT, IUBMB LIFE, vol. 60, 2008, pages 824 - 833
D. R. HIGGS; D. VERNIMMEN; B. WOOD, ADV. GENET., vol. 61, 2008, pages 143 - 173
D. S. PAUL ET AL., PLOS GENET., vol. 7, 2011, pages E1002139
DESIMONE, PROC NATL ACAD SCI USA., vol. 79, no. 14, 1982, pages 4428 - 31
DOVER ET AL., BR J HAEMATOL., vol. 88, no. 3, 1994, pages 555 - 61
E. PINAUD ET AL., ADV. IMMUNOL., vol. 110, 2011, pages 27 - 70
E. SOLER ET AL., GENES DEV., vol. 24, 2010, pages 277 - 289
E. VAN DEN AKKER; T. J. SATCHWELL; S. PELLEGRIN; G. DANIELS; A. M. TOYE, HAEMATOLOGICA, vol. 95, 2010, pages 1594 - 1598
F. D. URNOV; E. J. REBAR; M. C. HOLMES; H. S. ZHANG; P. D. GREGORY, NAT. REV. GENET., vol. 11, 2010, pages 636 - 646
F. ESTEGHAMAT ET AL., BLOOD, vol. 121, 2013, pages 2553 - 2562
G. GALARNEAU ET AL., NAT. GENET., vol. 42, 2010, pages 1049 - 1051
G. LETTRE ET AL., PROC. NATL. ACAD. SCI. U. S. A., vol. 105, 2008, pages 11869 - 11874
GOEDDEL: "Gene Expression Technology: Methods in Enzymology", vol. 185, 1990, ACADEMIC PRESS
H. J. LEE; E. KIM; J. S. KIM, GENOME RES., vol. 20, 2010, pages 81 - 89
H. T. BAE ET AL., BLOOD, vol. 120, 2012, pages 1961 - 1962
HUANGFU, D. ET AL., NATURE BIOTECHNOLOGY, vol. 26, no. 7, 2008, pages 795 - 797
J. C. MILLER ET AL., NAT. BIOTECHNOL., vol. 29, 2011, pages 143 - 148
J. ERNST ET AL., NATURE, vol. 473, 2011, pages 43 - 49
J. K. JOUNG; J. D. SANDER, NAT. REV. MOL. CELL BIOL., vol. 14, 2013, pages 49 - 55
J. TYSON; J. A. ARMOUR, BMC GENOMICS, 2012, pages 13,693 - 2164,13-693
J. VAN DER OOST, SCIENCE, vol. 339, 2013, pages 768 - 770
J. XU ET AL., DEV. CELL., vol. 23, 2012, pages 796 - 811
J. XU ET AL., GENES DEV., vol. 24, 2010, pages 783 - 798
J. XU ET AL., PROC. NATL. ACAD. SCI. U. S. A., vol. 110, 2013, pages 6518 - 6523
J. XU ET AL., SCIENCE, vol. 334, 2011, pages 993 - 996
JANE; CUNNINGHAM, BR. J. HAEMATOL., vol. 102, 1998, pages 415 - 422
JINEK, M. ET AL., SCIENCE, vol. 337, no. 6096, 2012, pages 816 - 821
K. A. FRAZER; S. S. MURRAY; N. J. SCHORK; E. J. TOPOL, NAT. REV. GENET., vol. 10, 2009, pages 241 - 251
K. E. MCGRATH ET AL., BLOOD, vol. 117, 2011, pages 4600 - 4608
L. FUGGER; G. MCVEAN; J. I. BELL, N. ENGL. J. MED., vol. 367, 2012, pages 2370 - 2371
LETTRE ET AL., PROC NATL ACAD SCI U S A, 2008
LETVIN, N ENGL J MED., vol. 310, no. 14, 1984, pages 869 - 73
LEY ET AL., BLOOD, vol. 62, 1983, pages 370 - 380
LEY ET AL., N. ENGL. J. MEDICINE, vol. 307, 1982, pages 1469 - 1475
LIAKOPOULOU ET AL., BLOOD, vol. 186, no. 8, 1995, pages 3227 - 35
LIU ET AL., MOL CANCER, vol. 5, 2006, pages 18
LIU ET AL., NAT IMMUNOL, vol. 4, 2003, pages 525
M. A. MCDEVITT; Y. FUJIWARA; R. A. SHIVDASANI; S. H. ORKIN, PROC. NATL. ACAD. SCI. U. S. A., vol. 94, 1997, pages 7976 - 7981
M. C. WU ET AL., AM. J. HUM. GENET., vol. 89, 2011, pages 82 - 93
M. D. FARBER; M. KOSHY; T. R. KINNEY, J. CHRONIC DIS., vol. 38, 1985, pages 495 - 505
M. LEID ET AL., GENE EXPR. PATTERNS., vol. 4, 2004, pages 733 - 739
M. NUINOON ET AL., HUM. GENET., vol. 127, 2010, pages 303 - 314
M. S. KOWALCZYK ET AL., MOL. CELL, vol. 45, 2012, pages 447 - 458
M. T. KASSOUF ET AL., GENOME RES., vol. 20, 2010, pages 1064 - 1083
M. T. MAURANO ET AL., SCIENCE, vol. 337, 2012, pages 1190 - 1195
M. UDA ET AL., PROC. NATL. ACAD. SCI. U. S. A., vol. 105, 2008, pages 1620 - 1625
MAEDER ML ET AL., NAT BIOTECHNOL., 9 October 2013 (2013-10-09)
MANOLIO, J CLIN INVEST, vol. 118, 2008, pages 1590
MARSON, A. ET AL., CELL-STEM CELL, vol. 3, 2008, pages 132 - 135
MCCARTHY ET AL., NAT REV GENET, vol. 9, 2008, pages 356
MENDENHALL E. M. ET AL., NAT. BIOTECHNOL., 8 September 2013 (2013-09-08)
MENZEL ET AL., NAT GENET, vol. 39, 2007, pages 1197
MENZEL, STEPHAN ET AL.: "A QTL influencing F cell production maps to a gene encoding a zinc-finger protein on chromosome 2p15`", NATURE GENETICS, vol. 39, no. 10, 2007, pages 1197 - 1199, XP002506824 *
N. SOLOVIEFF ET AL., BLOOD, vol. 115, 2010, pages 1815 - 1822
NATHAN: "Nathan and Oski's hematology of infancy and childhood", 2003, pages: 2
NATHAN: "Nathan and Oski's hematology", 2003, pages: 2
NATURE, vol. 489, 2012, pages 57 - 74
OLIVIERI, SEMINARS IN HEMATOLOGY, vol. 33, 1996, pages 24 - 42
P. BHATNAGAR ET AL., J. HUM. GENET., vol. 56, 2011, pages 316 - 323
P. LIU ET AL., NAT. IMMUNOL., vol. 4, 2003, pages 525 - 532
P. VAN DER HARST ET AL., NATURE, vol. 492, 2012, pages 369 - 375
PAPAYANNOPOULOU ET AL., SCIENCE, vol. 224, no. 4649, 1984, pages 617 - 9
PEMBREY ET AL., BR. J. HAEMATOL., vol. 40, 1978, pages 415 - 429
PERRINE ET AL., BIOCHEM BIOPHYS RES COMMUN., vol. 148, no. 2, 1987, pages 694 - 700
PERRINE ET AL., BLOOD, vol. 74, no. 1, 1989, pages 454 - 9
R. E. THURMAN ET AL., NATURE, vol. 489, 2012, pages 75 - 82
R. GALANELLO ET AL., BLOOD, vol. 114, 2009, pages 3935 - 3937
R. P. PATWARDHAN ET AL., NAT. BIOTECHNOL., vol. 30, 2012, pages 265 - 270
S. MENZEL ET AL., NAT., vol. 39, 2007, pages 1197 - 1199
S. PURCELL ET AL., AM. J. HUM. GENET., vol. 81, 2007, pages 559 - 575
SANKARAN, VIJAY G: "Targeted therapeutic strategies for fetal hemoglobin induction", AMERICAN SOCIETY OF HEMATOLOGY EDUCATION PROGRAM BOOK, vol. 2011, no. 1, 2011, pages 459 - 465, XP055108964 *
SHI, Y. ET AL., CELL-STEM CELL, vol. 2, 2008, pages 525 - 528
T. CERMAK ET AL., NUCLEIC ACIDS RES., vol. 39, 2011, pages E82
TORKELSON ET AL., BLOOD CELLS MOL DIS., vol. 22, no. 2, 1996, pages 150 - 8
UDA ET AL., PROC NATL ACAD SCI U S A, vol. 105, 2008, pages 1620
V. G. SANKARAN ET AL., NATURE, vol. 460, 2009, pages 1093 - 1097
V. G. SANKARAN ET AL., SCIENCE, vol. 322, 2008, pages 1839 - 1842
VAN ROEY ET AL., NATURE STRUCT. BIOL., vol. 9, 2002, pages 806 - 811
WARREN ET AL., CELL STEM CELL, vol. 7, no. 5, 5 November 2010 (2010-11-05), pages 618 - 30
XU, JIAN ET AL.: "Transcriptional silencing of y-globin by BCLIIA involves long-range interactions and cooperation with SOX6", GENES & DEVELOPMENT, vol. 24, no. 8, 2010, pages 783 - 798, XP055255421 *
Y. YU ET AL., J. EXP. MED., vol. 209, 2012, pages 2467 - 2483

Cited By (131)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10323236B2 (en) 2011-07-22 2019-06-18 President And Fellows Of Harvard College Evaluation and improvement of nuclease cleavage specificity
US9322006B2 (en) 2011-07-22 2016-04-26 President And Fellows Of Harvard College Evaluation and improvement of nuclease cleavage specificity
US11542493B2 (en) 2012-11-27 2023-01-03 The Children's Medical Center Corporation Targeting BCL11A distal regulatory elements for fetal hemoglobin reinduction
US10508298B2 (en) 2013-08-09 2019-12-17 President And Fellows Of Harvard College Methods for identifying a target site of a CAS9 nuclease
US9163284B2 (en) 2013-08-09 2015-10-20 President And Fellows Of Harvard College Methods for identifying a target site of a Cas9 nuclease
US10954548B2 (en) 2013-08-09 2021-03-23 President And Fellows Of Harvard College Nuclease profiling system
US11920181B2 (en) 2013-08-09 2024-03-05 President And Fellows Of Harvard College Nuclease profiling system
WO2015033343A1 (en) 2013-09-03 2015-03-12 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Compositions and methods for expressing recombinant polypeptides
JP2016529907A (en) * 2013-09-04 2016-09-29 シーエスアイアール Site-specific nuclease single-cell assay targeting gene regulatory elements that silence gene expression
US10858639B2 (en) 2013-09-06 2020-12-08 President And Fellows Of Harvard College CAS9 variants and uses thereof
US9999671B2 (en) 2013-09-06 2018-06-19 President And Fellows Of Harvard College Delivery of negatively charged proteins using cationic lipids
US10912833B2 (en) 2013-09-06 2021-02-09 President And Fellows Of Harvard College Delivery of negatively charged proteins using cationic lipids
US9322037B2 (en) 2013-09-06 2016-04-26 President And Fellows Of Harvard College Cas9-FokI fusion proteins and uses thereof
US9388430B2 (en) 2013-09-06 2016-07-12 President And Fellows Of Harvard College Cas9-recombinase fusion proteins and uses thereof
US11299755B2 (en) 2013-09-06 2022-04-12 President And Fellows Of Harvard College Switchable CAS9 nucleases and uses thereof
US9340799B2 (en) 2013-09-06 2016-05-17 President And Fellows Of Harvard College MRNA-sensing switchable gRNAs
US9526784B2 (en) 2013-09-06 2016-12-27 President And Fellows Of Harvard College Delivery system for functional nucleases
US9228207B2 (en) 2013-09-06 2016-01-05 President And Fellows Of Harvard College Switchable gRNAs comprising aptamers
US10682410B2 (en) 2013-09-06 2020-06-16 President And Fellows Of Harvard College Delivery system for functional nucleases
US9737604B2 (en) 2013-09-06 2017-08-22 President And Fellows Of Harvard College Use of cationic lipids to deliver CAS9
US9340800B2 (en) 2013-09-06 2016-05-17 President And Fellows Of Harvard College Extended DNA-sensing GRNAS
US10597679B2 (en) 2013-09-06 2020-03-24 President And Fellows Of Harvard College Switchable Cas9 nucleases and uses thereof
US10767156B2 (en) 2013-10-24 2020-09-08 Yeda Research And Development Co., Ltd. Polynucleotides encoding BREX system polypeptides and methods of using same
US10640788B2 (en) 2013-11-07 2020-05-05 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAs
US10190137B2 (en) 2013-11-07 2019-01-29 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
US9834791B2 (en) 2013-11-07 2017-12-05 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
US11390887B2 (en) 2013-11-07 2022-07-19 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
US11021696B2 (en) 2013-11-13 2021-06-01 Children's Medical Center Corporation Nuclease-mediated regulation of gene expression
EP3068881A4 (en) * 2013-11-13 2017-04-19 Children's Medical Center Corporation Nuclease-mediated regulation of gene expression
EP3960856A1 (en) * 2013-11-13 2022-03-02 Children's Medical Center Corporation Nuclease-mediated regulation of gene expression
EP3492593A1 (en) * 2013-11-13 2019-06-05 Children's Medical Center Corporation Nuclease-mediated regulation of gene expression
US9840699B2 (en) 2013-12-12 2017-12-12 President And Fellows Of Harvard College Methods for nucleic acid editing
US11124782B2 (en) 2013-12-12 2021-09-21 President And Fellows Of Harvard College Cas variants for gene editing
US9068179B1 (en) 2013-12-12 2015-06-30 President And Fellows Of Harvard College Methods for correcting presenilin point mutations
US10465176B2 (en) 2013-12-12 2019-11-05 President And Fellows Of Harvard College Cas variants for gene editing
US11053481B2 (en) 2013-12-12 2021-07-06 President And Fellows Of Harvard College Fusions of Cas9 domains and nucleic acid-editing domains
US11739329B2 (en) 2014-04-25 2023-08-29 The Children's Medical Center Corporation Compositions and methods to treating hemoglobinopathies
US10662429B2 (en) 2014-04-25 2020-05-26 The Children's Medical Center Corporation Compositions and methods to treating hemoglobinopathies
US11124794B2 (en) 2014-04-25 2021-09-21 The Children's Medical Center Corporation Compositions and methods to treating hemoglobinopathies
US10287588B2 (en) 2014-04-25 2019-05-14 The Childrens's Medical Center Corporation Compositions and methods to treating hemoglobinopathies
WO2016005985A2 (en) 2014-07-09 2016-01-14 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Method for reprogramming cells
US10077453B2 (en) 2014-07-30 2018-09-18 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
US11578343B2 (en) 2014-07-30 2023-02-14 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
US10704062B2 (en) 2014-07-30 2020-07-07 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
WO2016038616A1 (en) 2014-09-14 2016-03-17 Yeda Research And Development Co. Ltd. Nmda receptor antagonists for treating gaucher disease
WO2016135557A2 (en) 2015-02-23 2016-09-01 Crispr Therapeutics Ag Materials and methods for treatment of hemoglobinopathies
WO2016135559A2 (en) 2015-02-23 2016-09-01 Crispr Therapeutics Ag Materials and methods for treatment of human genetic diseases including hemoglobinopathies
WO2016135558A2 (en) 2015-02-23 2016-09-01 Crispr Therapeutics Ag Materials and methods for treatment of hemoglobinopathies
EP4218771A1 (en) 2015-03-27 2023-08-02 Yeda Research and Development Co. Ltd Methods of treating motor neuron diseases
JP7288302B2 (en) 2015-05-08 2023-06-07 ザ チルドレンズ メディカル センター コーポレーション Methods of targeting BCL11A enhancer functional regions for fetal hemoglobin reinduction
JP2018524018A (en) * 2015-05-08 2018-08-30 ザ チルドレンズ メディカル センター コーポレイション Method for targeting the BCL11A enhancer functional region for reinduction of fetal hemoglobin
WO2016182917A1 (en) * 2015-05-08 2016-11-17 Children's Medical Center Corporation Targeting bcl11a enhancer functional regions for fetal hemoglobin reinduction
JP2022008430A (en) * 2015-05-08 2022-01-13 ザ チルドレンズ メディカル センター コーポレーション Methods for targeting bcl11a enhancer functional regions for fetal hemoglobin reinduction
EP3294873B1 (en) 2015-05-08 2020-08-19 The Children's Medical Center Corporation Targeting bcl11a enhancer functional regions for fetal hemoglobin reinduction
US11572543B2 (en) 2015-05-08 2023-02-07 The Children's Medical Center. Corporation Targeting BCL11A enhancer functional regions for fetal hemoglobin reinduction
US11390884B2 (en) 2015-05-11 2022-07-19 Editas Medicine, Inc. Optimized CRISPR/cas9 systems and methods for gene editing in stem cells
US10808020B2 (en) 2015-05-12 2020-10-20 Sangamo Therapeutics, Inc. Nuclease-mediated regulation of gene expression
JP7365374B2 (en) 2015-05-12 2023-10-19 サンガモ セラピューティクス, インコーポレイテッド Nuclease-mediated gene expression regulation
JP2018516556A (en) * 2015-05-12 2018-06-28 サンガモ セラピューティクス, インコーポレイテッド Nuclease-mediated gene expression regulation
EP3294866A4 (en) * 2015-05-12 2018-12-05 Sangamo Therapeutics, Inc. Nuclease-mediated regulation of gene expression
JP2021191755A (en) * 2015-05-13 2021-12-16 セルジーン コーポレイション TREATMENT OF β-THALASSEMIA USING ActRII LIGAND TRAPS
US11911415B2 (en) 2015-06-09 2024-02-27 Editas Medicine, Inc. CRISPR/Cas-related methods and compositions for improving transplantation
WO2017009842A2 (en) 2015-07-16 2017-01-19 Biokine Therapeutics Ltd. Compositions and methods for treating cancer
EP3943098A2 (en) 2015-07-16 2022-01-26 Biokine Therapeutics Ltd. Compositions and methods for treating cancer
EP3744340A2 (en) 2015-07-16 2020-12-02 Biokine Therapeutics Ltd. Compositions and methods for treating cancer
US10167457B2 (en) 2015-10-23 2019-01-01 President And Fellows Of Harvard College Nucleobase editors and uses thereof
US11214780B2 (en) 2015-10-23 2022-01-04 President And Fellows Of Harvard College Nucleobase editors and uses thereof
WO2017077394A2 (en) 2015-11-04 2017-05-11 Crispr Therapeutics Ag Materials and methods for treatment of hemoglobinopathies
WO2017118985A1 (en) 2016-01-06 2017-07-13 Yeda Research And Development Co. Ltd. Compositions and methods for treating malignant, autoimmune and inflammatory diseases
WO2017125931A1 (en) 2016-01-21 2017-07-27 The State Of Israel, Ministry Of Agriculture & Rural Development, Agricultural Research Organization (Aro) (Volcani Center) Parthenocarpic plants and methods of producing same
WO2017130205A1 (en) 2016-01-31 2017-08-03 Hadasit Medical Research Services And Development Ltd. Autosomal-identical pluripotent stem cell populations having non-identical sex chromosomal composition and uses thereof
WO2017138008A2 (en) 2016-02-14 2017-08-17 Yeda Research And Development Co. Ltd. Methods of modulating protein exocytosis and uses of same in therapy
WO2017153982A1 (en) 2016-03-06 2017-09-14 Yeda Research And Development Co. Ltd. Method for modulating myelination
US10113163B2 (en) 2016-08-03 2018-10-30 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
US10947530B2 (en) 2016-08-03 2021-03-16 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
US11702651B2 (en) 2016-08-03 2023-07-18 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
US11661590B2 (en) 2016-08-09 2023-05-30 President And Fellows Of Harvard College Programmable CAS9-recombinase fusion proteins and uses thereof
WO2018033929A1 (en) 2016-08-18 2018-02-22 Yeda Research And Development Co. Ltd. Diagnostic and therapeutic uses of exosomes
US11542509B2 (en) 2016-08-24 2023-01-03 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
US11306324B2 (en) 2016-10-14 2022-04-19 President And Fellows Of Harvard College AAV delivery of nucleobase editors
WO2018096547A1 (en) 2016-11-28 2018-05-31 Yeda Research And Development Co. Ltd. Isolated polynucleotides and polypeptides and methods of using same for expressing an expression product of interest
US10745677B2 (en) 2016-12-23 2020-08-18 President And Fellows Of Harvard College Editing of CCR5 receptor gene to protect against HIV infection
US11820969B2 (en) 2016-12-23 2023-11-21 President And Fellows Of Harvard College Editing of CCR2 receptor gene to protect against HIV infection
WO2018122771A1 (en) 2016-12-29 2018-07-05 Ukko Inc. Methods for identifying and de-epitoping allergenic polypeptides
WO2018142416A1 (en) 2017-02-06 2018-08-09 Yeda Research And Development Co. Ltd. Isolated cells genetically modified to express a disarm system having an anti-phage activity and methods of producing same
US11466271B2 (en) 2017-02-06 2022-10-11 Novartis Ag Compositions and methods for the treatment of hemoglobinopathies
US11898179B2 (en) 2017-03-09 2024-02-13 President And Fellows Of Harvard College Suppression of pain by gene editing
US11542496B2 (en) 2017-03-10 2023-01-03 President And Fellows Of Harvard College Cytosine to guanine base editor
US11851690B2 (en) 2017-03-14 2023-12-26 Editas Medicine, Inc. Systems and methods for the treatment of hemoglobinopathies
US11268082B2 (en) 2017-03-23 2022-03-08 President And Fellows Of Harvard College Nucleobase editors comprising nucleic acid programmable DNA binding proteins
US11261441B2 (en) 2017-03-29 2022-03-01 Bluebird Bio, Inc. Vectors and compositions for treating hemoglobinopathies
WO2018207178A1 (en) 2017-05-07 2018-11-15 Yeda Research And Development Co. Ltd. Methods of treating psychiatric stress disorders
EP3821888A1 (en) 2017-05-07 2021-05-19 Yeda Research and Development Co. Ltd Lxr agonists for treating psychiatric stress disorders
US11963982B2 (en) 2017-05-10 2024-04-23 Editas Medicine, Inc. CRISPR/RNA-guided nuclease systems and methods
US20200155606A1 (en) * 2017-05-10 2020-05-21 Editas Medicine, Inc. Crispr/rna-guided nuclease systems and methods
US11560566B2 (en) 2017-05-12 2023-01-24 President And Fellows Of Harvard College Aptazyme-embedded guide RNAs for use with CRISPR-Cas9 in genome editing and transcriptional activation
US11788087B2 (en) 2017-05-25 2023-10-17 The Children's Medical Center Corporation BCL11A guide delivery
US11866726B2 (en) 2017-07-14 2024-01-09 Editas Medicine, Inc. Systems and methods for targeted integration and genome editing and detection thereof using integrated priming sites
WO2019021284A1 (en) 2017-07-24 2019-01-31 Yeda Research And Development Co. Ltd. Combination therapy for the treatment of cancer
US11732274B2 (en) 2017-07-28 2023-08-22 President And Fellows Of Harvard College Methods and compositions for evolving base editors using phage-assisted continuous evolution (PACE)
WO2019038771A1 (en) 2017-08-23 2019-02-28 Technion Research & Development Foundation Limited Compositions and methods for improving alcohol tolerance in yeast
US11319532B2 (en) 2017-08-30 2022-05-03 President And Fellows Of Harvard College High efficiency base editors comprising Gam
US11932884B2 (en) 2017-08-30 2024-03-19 President And Fellows Of Harvard College High efficiency base editors comprising Gam
US11795443B2 (en) 2017-10-16 2023-10-24 The Broad Institute, Inc. Uses of adenosine base editors
WO2019097514A1 (en) 2017-11-14 2019-05-23 Yeda Research And Development Co. Ltd. Hematopoietic stem cells with improved properties
WO2019145964A1 (en) 2018-01-29 2019-08-01 Yeda Research And Development Co. Ltd. Combination of a mek inhibitor and a cdk4/6 inhibitor for the treatment of sarcoma
US11268077B2 (en) 2018-02-05 2022-03-08 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of hemoglobinopathies
WO2020008412A1 (en) 2018-07-04 2020-01-09 Ukko Inc. Methods of de-epitoping wheat proteins and use of same for the treatment of celiac disease
WO2020089892A1 (en) 2018-10-28 2020-05-07 Yeda Research And Development Co. Ltd. Prevention of age related clonal hematopoiesis and diseases associated therewith
WO2020112979A3 (en) * 2018-11-30 2020-07-02 Bauer Daniel E Therapeutic gene editing for elane-associated disease
WO2020178822A1 (en) 2019-03-05 2020-09-10 The State Of Israel, Ministry Of Agriculture & Rural Development, Agricultural Research Organization (Aro) (Volcani Center) Genome-edited birds
US11795452B2 (en) 2019-03-19 2023-10-24 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
US11643652B2 (en) 2019-03-19 2023-05-09 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
US11447770B1 (en) 2019-03-19 2022-09-20 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
WO2021001784A1 (en) 2019-07-04 2021-01-07 Ukko Inc. De-epitoped alpha gliadin and use of same for the management of celiac disease and gluten sensitivity
WO2021009763A1 (en) 2019-07-16 2021-01-21 Yeda Research And Development Co. Ltd. Methods of treating pain
WO2021084540A1 (en) 2019-10-30 2021-05-06 Yeda Research And Development Co. Ltd. Inhibitors of mmej pathway for prevention and treatment of pre-myeloid and myeloid malignancies
WO2021130752A1 (en) 2019-12-22 2021-07-01 Yeda Research And Development Co. Ltd. Systems and methods for identifying cells that have undergone genome editing
WO2021152587A1 (en) 2020-01-30 2021-08-05 Yeda Research And Development Co. Ltd. Treating acute liver disease with tlr-mik inhibitors
US11912985B2 (en) 2020-05-08 2024-02-27 The Broad Institute, Inc. Methods and compositions for simultaneous editing of both strands of a target double-stranded nucleotide sequence
WO2022074646A1 (en) 2020-10-05 2022-04-14 Protalix Ltd. Dicer-like knock-out plant cells
WO2022079719A1 (en) 2020-10-15 2022-04-21 Yeda Research And Development Co. Ltd. Method of treating myeloid malignancies
WO2022115498A1 (en) 2020-11-26 2022-06-02 Ukko Inc. Modified high molecular weight glutenin subunit and uses thereof
WO2022130384A1 (en) 2020-12-17 2022-06-23 Yeda Research And Development Co. Ltd. Controlling ubiquitination of mlkl for treatment of disease
WO2022130388A2 (en) 2020-12-18 2022-06-23 Yeda Research And Development Co. Ltd. Compositions for use in the treatment of chd2 haploinsufficiency and methods of identifying same
WO2022239001A1 (en) 2021-05-10 2022-11-17 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Pharmaceutical compositions for treating neurological conditions
WO2022264132A1 (en) 2021-06-13 2022-12-22 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Method for reprogramming human cells
EP4130028A1 (en) 2021-08-03 2023-02-08 Rhazes Therapeutics Ltd Engineered tcr complex and methods of using same
WO2023012584A2 (en) 2021-08-03 2023-02-09 Genicity Limited Engineered tcr complex and methods of using same
WO2023062636A1 (en) 2021-10-14 2023-04-20 Weedout Ltd. Methods of weed control
WO2023240282A1 (en) 2022-06-10 2023-12-14 Umoja Biopharma, Inc. Engineered stem cells and uses thereof

Also Published As

Publication number Publication date
AU2018278850B2 (en) 2021-05-27
AU2021218012A1 (en) 2021-09-09
BR112015011995A2 (en) 2017-07-11
US20200087651A1 (en) 2020-03-19
SG10202110062SA (en) 2021-11-29
US10472619B2 (en) 2019-11-12
IL238949A0 (en) 2015-07-30
EP3502240B1 (en) 2021-05-05
BR112015011995B1 (en) 2023-02-07
SG11201504038XA (en) 2015-06-29
MX361412B (en) 2018-12-05
IL238949B (en) 2019-06-30
PT2925864T (en) 2019-02-06
KR20150107726A (en) 2015-09-23
CA2892860A1 (en) 2014-06-05
CN104955943A (en) 2015-09-30
SG10201704008RA (en) 2017-06-29
AU2021218012B2 (en) 2023-06-29
AU2013352156B2 (en) 2018-12-06
EP2925864A1 (en) 2015-10-07
US20150307867A1 (en) 2015-10-29
MX2015006528A (en) 2016-07-13
CN104955943B (en) 2018-09-25
AU2013352156A1 (en) 2015-06-11
JP6542124B2 (en) 2019-07-10
US20180119130A1 (en) 2018-05-03
PT3502240T (en) 2021-08-11
US20230348887A1 (en) 2023-11-02
EP3502240A1 (en) 2019-06-26
SA515360489B1 (en) 2016-12-25
KR102240555B1 (en) 2021-04-16
EP2925864A4 (en) 2016-07-13
US11542493B2 (en) 2023-01-03
EP2925864B1 (en) 2018-10-31
JP6633602B2 (en) 2020-01-22
ES2884925T3 (en) 2021-12-13
ZA201504004B (en) 2016-11-30
US9822355B2 (en) 2017-11-21
ES2708948T3 (en) 2019-04-12
HK1215720A1 (en) 2016-09-09
JP2015536657A (en) 2015-12-24
CA2892860C (en) 2023-01-03
AU2018278850A1 (en) 2019-01-17
DK3502240T3 (en) 2021-08-09
CN109554350B (en) 2022-09-23
CN109554350A (en) 2019-04-02
JP2018085987A (en) 2018-06-07
DK2925864T3 (en) 2019-02-11

Similar Documents

Publication Publication Date Title
AU2021218012B2 (en) Targeting bcl11a distal regulatory elements for fetal hemoglobin reinduction
US20230235285A1 (en) Targeting bcl11a enhancer functional regions for fetal hemoglobin reinduction
US20210047632A1 (en) Targeting bcl11a distal regulatory elements with a cas9-cas9 fusion for fetal hemoglobin reinduction

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: 13858006

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 238949

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: MX/A/2015/006528

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2015544205

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2892860

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 14647547

Country of ref document: US

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112015011995

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2013352156

Country of ref document: AU

Date of ref document: 20131127

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: IDP00201503738

Country of ref document: ID

ENP Entry into the national phase

Ref document number: 20157016749

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2013858006

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 112015011995

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20150525