Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPOSITIONS AND METHODS FOR PREVENTING, AMELIORATING, OR TREATING SICKLE CELL DISEASE AND COMPOSITIONS AND METHODS FOR DISRUPTING GENES AND GENE SEGMENTS
Document Type and Number:
WIPO Patent Application WO/2024/095213
Kind Code:
A1
Abstract:
The present disclosure provides nucleic acids, compositions and vectors containing and their use for effecting gene editing and/or gene expression alteration on sickle cell disease (SCD)-associated genes, e.g., in vivo. The present disclosure further provides compositions for and methods of effecting gene editing and/or gene expression alteration, such as on SCD-associated genes in vivo, and methods of preventing, ameliorating, or treating SCD.

Inventors:
LEAVITT BLAIR (CA)
HILL AUSTIN (CA)
WAGNER PAMELA (CA)
CARON NICHOLAS (CA)
Application Number:
PCT/IB2023/061087
Publication Date:
May 10, 2024
Filing Date:
November 02, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
INCISIVE GENETICS INC (CA)
UNIV BRITISH COLUMBIA (CA)
International Classes:
C12N15/113; A61K9/51; A61K31/7088; A61K31/7105; A61K38/46; A61P7/00; C07K14/47; C12N9/22; C12N15/10; C12N15/12; C12N15/85; C12N15/88
Download PDF:
Claims:
CLAIMS What is claimed is: 1. One or more isolated guide RNAs (gRNAs) for Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-mediated gene editing, optionally which is/are effective for use in effecting CRISPR-mediated gene editing of at least one Sickle cell disease (SCD)-associated gene or a variant thereof and/or a promoter or enhancer thereof in vivo in a subject in need thereof, wherein the gRNAs comprise at least one CRISPR RNA (crRNA) sequence comprising a target-complementary sequence comprising at least 17 nucleic acids, optionally comprising 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleic acids, the target-complementary sequence comprising: (1) (i) the polynucleotide sequence of SEQ ID NO: 71,277,279, 87,287, or 289; or (ii) a polynucleotide sequence comprising one or more (optionally one, two, three, four, or five) mutations (e.g., deletions at the 3'-end) relative to the polynucleotide sequence of SEQ ID NO: 71,277,279, 87,287, or 289, respectively; or (2) (i) the polynucleotide sequence of SEQ ID NO: 271,273,275,281,283,285, 85, 25, 45, 47, 49, 65,67,69, 75,or77;or (ii) a polynucleotide sequence comprising one or more (optionally one, two, three, four, or five) mutations ( e.g., deletions at the 5' -end) relative to the polynucleotide sequence of SEQ ID NO: 271,273,275,281,283,285, 85, 25, 45, 47, 49, 65, 67, 69, 75, or 77, said mutations optionally at any nucleic acid position(s) other than the 4th to the 7th nucleic acid positions from the 3'-end of the polynucleotide sequence of SEQ ID NO: 271,273,275,281,283,285, 85, 25, 45, 47, 49, 65, 67, 69, 75, or 77, respectively, optionally wherein: in (1), the crRNA sequence further comprises a repeat sequence optionally at the 5'-end of the target-complementary sequence, the repeat sequence comprising (i) an optional 5' -end sequence, optionally comprising UAAUU or AAUU, (ii) a first stem sequence, optionally comprising UCUAC, (iii) a loop sequence, optionally comprising UCUU, UAAGU, UGUU, UUUU, UAUU, UGUUU, UUCG, or UUU, (iv) a second stem sequence which is reverse complementary to the first stem sequence, optionally comprising GUAGA, and (v) an optional 3'-end sequence, optionally comprising U, optionally wherein the repeat sequence comprises any one of SEQ ID NOS: 201-206; or in (2), the gRNA is: (I) a single guide RNA (sgRNA) comprising (i) a crRNA sequence comprising the target- complementary sequence and a crRNA backbone sequence and (ii) a trans-activating CRISPR RNA (tracrRNA) sequence in a single strand, optionally wherein the crRNA sequence and the tracrRNA sequence are linked via a linker optionally comprising SEQ ID NO: 139, further optionally wherein the gRNA comprises the target-complementary sequence followed by a sgRNA backbone sequence of any of SEQ ID NOS: 141-144, optionally wherein the sgRNA backbone sequence is followed by one or more uracils, further optionally 1-10 uracils, or (II) a dual guide RNA (dgRNA) formed by hybridization between (i) a crRNA sequence comprising the target-complementary sequence and a crRNA backbone sequence and (ii) a tracrRNA, optionally wherein the crRNA backbone sequence and the tracrRNA comprise SEQ ID NOS: 145 and 146, respectively, or SEQ ID NOS: 147 and 148, respectively, and optionally wherein: (i) the one or more gRNAs are synthetic or recombinant; and/or (ii) the one or more gRNAs comprise a synthetic sgRNA and comprises at least one chemical modification, optionally (ii-1) 2'-O-methylation further optionally at first three and last three bases and/or (ii-2) one or more 3' phosphorothioate bonds, further optionally between first three and last two bases. 2. A composition comprising one or more isolated gRNAs according to claim 1. 3. A polynucleotide or polynucleotides encoding the one or more isolated gRNAs of claim 1. 4. A vector comprising the polynucleotide or polynucleotides of claim 3 operably linked to one or more regulatory sequences. 5. A ribonucleoprotein (RNP), which comprises: (a) one or more isolated gRNAs of claim 1; complexed with (b) a Cas endonuclease, optionally wherein: the Cas endonuclease is: (i) selected from the group consisting ofCas12a or Cpfl, Cas9, Cas3, Cas8a2, Cas8b, Cas8c, Casl0, Casl 1, Cas12, Cas12b, Cas12f, Cas12j, Cas13, Cas13a, Cas14, C2cl, C2c3, and C2c2; (ii) a class 2 Cas endonuclease, optionally a type II, type V, or type VI Cas nuclease; (iii) Cas12a of Acidaminococcus sp. (AsCas12a), Lachnospiraceae bacterium (LbCas12a), Francisella novicida (FnCas12a), Moraxella bovoculi (MbCas12a), Coprococcus eutactus (CeCas 12a), Butyrivibrio fibrisolvens (BdCas 12a); (iv) Cas12a or any engineered variants thereof, optionally comprising any one of SEQ ID NOS: 210-215, 220-221, 230,240,250, and 260; (v) Cas9 of Streptococcus pyogenes (SpCas9), Staphylococcus aureus (SaCas9), Streptococcus thermophilus (StCas9), Neisseria meningitidis (NmCas9), Francisella novicida (FnCas9), Campylobacter jejuni (CjCas9), Streptococcus canis (ScCas9), or Staphylococcus auricularis (SauriCas9), or any engineered variants thereof, including SaCas9-HF, SpCas9-HF1, KKHSaCas9, eSpCas9, HypaCas9, Fokl-Fused dCas9, xCas9, SpRY (variant ofSpCas9), and SpG (variant ofSpCas9); and/or (vi) Cas9, optionally comprising any one of SEQ ID NOS: 150-161, and further optionally wherein the RNP is formed by mixing at an approximately equimolar ratio (I) a solution comprising the one or more isolated gRNAs, further optionally wherein the pH of the solution is about 6 to 8, about 6.5 to 7.5, further optionally about 7, and (II) a solution comprising the Cas endonuclease, optionally wherein the pH of the solution is about 6 to 8, about 6.5 to 7.5, further optionally about 7, further optionally wherein the mixing is for about 5 minutes. 6. An isolated oligodeoxynucleotide (ODN), which comprises a nucleic acid sequence having at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to SEQ ID NO: 190 or a reverse complement thereof and is about 30-160 nt, 50-120 nt, about 60-100 nt, about 70-90 nt, about 75-85 nt, about 80 nt or about 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 81, 82, 83, 84, or 85 nt in length, optionally wherein the ODN is a single-stranded oligo DNA nucleotide molecule (ssODN) or a double-stranded oligo DNA nucleotide molecule (dsODN). 7. A pharmaceutical composition comprising at least one cargo encapsulated in a carrier, optionally a lipid-based, transfection competent vesicle (TCV), wherein the at least one cargo is capable of: (i) effecting gene editing of at least one Sickle cell disease (SCD)-associated gene and/or a promoter or enhancer thereof in vivo in a subject in need thereof; and/or (ii) altering the expression, function, and/or effect of at least one SCD-associated gene in vivo in a subject in need thereof, optionally wherein the subject has or has a risk of developing SCD, which is optionally sickle cell anemia (SCA), Sickle cell-hemoglobin C (HbSC), or HbS ~-thalassaemia, optionally wherein the composition is for: (I) direct injection into the bone marrow of the subject; and/or (II) intravenous injection into the subject, optionally wherein the subject is administered at least one agent that promotes stem cell mobilization. 8. The pharmaceutical composition of claim 7, wherein the carrier is a lipid-based TCV which comprises at least one ionizable cationic lipid, optionally wherein: (i) the at least one ionizable cationic lipid comprises, essentially consists of, or consists of a lipid selected from the group consisting ofN,N-dimethyl-2,3-dioleyloxy)propylamine (DODMA), 1,2-dioleoyl-3-dimethylammonium propane ("DODAP"), 1,2-Dilinoleoyl-3- dimethylaminopropane (DLinDAP), N,N-dimethyl-2,2-di-(9Z, l 2Z)-9 ,l 2-octadecadien-1-yl- 1,3-dioxolane-4-ethanamine (KC2), ( 6Z,9Z,28Z,3 l Z)-heptatriaconta-6,9,28,31-tetraen- l 9-yl 4-( dimethylamino )butanoate (MC3), 6-((2-hexyldecanoyl)oxy)-N-( 6-((2- hexyldecanoyl)oxy)hexyl)-N-( 4-hydroxybutyl)hexan-1-aminium (ALC-0315), 8-[(2- hydroxyethyl)[6-oxo-6-(undecyloxy)hexyl]amino ]-octanoic acid, 1-octylnonyl ester (SM- 102), N,N-dioleyl-N,N-dimethylammonium chloride (DODAC), N,N-distearyl-N,N- dimethylammonium bromide (DDAB), N-(1-(2,3-dioleoyloxy)propyl)-N,N,N- trimethylammonium chloride (DOT AP), N-( 1-(2,3-dioleyloxyl)propyl)-N,N,N- trimethylammonium chloride (DOTMA), 1,2-DiLinoleyloxy-N ,N-dimethylaminopropane (DLinDMA), 1,2-Dilinolenyloxy-N,N-dimethylaminopropane (DLenDMA), 1,2- Dilinoleylcarbamoyloxy-3-dimethylaminopropane (DLin-C-DAP), l ,2-Dilinoleyoxy-3- ( dimethylamino )acetoxypropane (DLin- DAC), 1,2-Dilinoleyoxy-3-morpholinopropane (DLin-MA), 1,2-Dilinoleylthio-3-dimethylaminopropane (DLin-S-DMA), 1-Linoleoyl-2- linoleyloxy-3-dimethylaminopropane (DLin-2-DMAP), 1,2-Dilinoleyloxy-3- trimethylaminopropane chloride salt (DLin-TMA.Cl), 1,2-Dilinoleoyl-3- trimethylaminopropane chloride salt (DLin-T AR.Cl), l ,2-Dilinoleyloxy-3-(N- methylpiperazino )propane (DLin-MPZ), or 3-(N,N-Dilinoleylamino )-1,2-propanediol (DLinAP), 3-(N,N-Dioleylamino)-1,2-propanedio (DOAP), 1,2-Dilinoleyloxo-3-(2-N,N- dimethylamino )ethoxypropane (DLin-EG-DMA), 1,2-Dilinolenyloxy-N,N- dimethylaminopropane (DLin-K-DMA), 2,2-Dilinoleyl-4-dimethylaminomethyl-[ 1,3]- dioxolane (DLin-K-DMA) or analogs thereof, (3aR,5s,6aS)-N,N-dimethyl-2,2-di((9Z,12Z)- octadeca-9, 12-dienyl)tetrahydro-3 aH-cyclopenta[ d][l ,3]dioxol-5-amine (ALNY-100), N- (2,3-dioleyloxyl)propyl-N,N-N-triethylammonium chloride ("DOTMA"); 1,2-Dioleyloxy-3- trimethylaminopropane chloride salt ("DOT AP.Cl"); 3.beta.-(N-(N',N'- dimethylaminoethane )-carbamoyl)cholesterol ("DC-Chol"), N-( 1-(2,3-dioleyloxyl )propyl)-N- 2-( sperminecarboxamido )ethyl)-N,N-dimethyl-ammonium trifluoracetate ("DOSP A"), dioctadecylamidoglycyl carboxyspermine ("DOGS"), and N-(l ,2-dimyristyloxyprop-3-yl)- N,N-dimethyl-N-hydroxyethyl ammonium bromide ("DMRIE"), and any combinations thereof; (ii) the TCV further comprises at least one helper lipid, optionally wherein the at least one helper lipid comprises, essentially consists of, or consists of a lipid selected from the group consisting of dioleoylphosphatidylethanolamine (DOPE), distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), palmitoyloleoylphosphatidylcholine (POPC), palmitoyloleoyl-phosphatidylethanolamine (POPE), dioleoyl-phosphatidylethanolamine 4-(N-maleimidomethyl)-cyclohexane- l- carboxylate (DOPE-ma!), dipalmitoyl phosphatidyl ethanolamine (DPPE), dimyristoylphosphoethanolamine (DMPE), distearoyl-phosphatidyl-ethanolamine (DSPE), 16-O-monomethyl PE, 16-O-dimethyl PE, 18-1-trans PE, l-stearoyl-2-oleoyl- phosphatidyethanolamine (SOPE), and any combinations thereof; (iii) the TCV further comprises at least one phospholipid, optionally wherein the at least one phospholipid comprises, essentially consists of, or consists of a lipid selected from the group consisting of distearoylphosphatidylcholine (DSPC), dioleoyl phosphatidylethanolamine (DOPE), dipalmitoylphosphatidylcholine (DPPC), phosphocholine (DOPC), dimyristoylphosphatidylcholine (DMPC), phosphatidylcholine (PLPC), 1,2-distearoyl-sn- glycero-3-phosphocholine (DAPC), phosphatidylethanolamine (PE), egg phosphatidylcholine (EPC), dilauryloylphosphatidylcholine (DLPC), dimyristoylphosphatidylcholine (DMPC), 1- myristoyl-2-palmitoyl phosphatidylcholine (MPPC), l-palmitoyl-2-myristoyl phosphatidylcholine (PMPC), 1-palmitoyl-2-stearoyl phosphatidylcholine (PSPC), 1,2- diarachidoyl-sn-glycero-3-phosphocholine (DBPC), 1-stearoyl-2-palmitoyl phosphatidylcholine (SPPC), 1,2-dieicosenoyl-sn-glycero-3-phosphocholine (DEPC), palmitoyloleoyl phosphatidylcholine (POPC), lysophosphatidyl choline, dilinoleoylphosphatidylcholine distearoylphophatidylethanolamine (DSPE), dimyristoyl phosphatidylethanolamine (DMPE), dipalmitoyl phosphatidylethanolamine (DPPE), palmitoyloleoyl phosphatidylethanolamine (POPE), lysophosphatidylethanolamine, and any combinations thereof; (iv) the TCV further comprises at least one cholesterol or cholesterol derivative, optionally wherein the at least one cholesterol or cholesterol derivative comprises, essentially consists of, or consists of a cholesterol or cholesterol derivative selected from the group consisting of cholesterol, N,N-dimethyl-N-ethylcarboxamidocholesterol (DC-Chol), 1,4-bis(3-N- oleylamino-propyl)piperazine, imidazole cholesterol ester (ICE), and any combinations thereof; (v) the TCV further comprises at least one PEG or PEG-lipid, optionally wherein the at least one PEG-lipid comprises, essentially consists of, or consists of a PEG-lipid selected from the group consisting of PEG-myristoyl diglyceride (PEG-DMG) (e.g., 1,2-dimyristoyl-rac- glycero-3-methoxypolyethylene glycol-2000 (Avanti® Polar Lipids (Birmingham, AL)), which is a mixture of 1,2-DMG PEG2000 and 1,3-DMG PEG2000 (e.g., in about 97:3 ratio)), PEG-phosphatidylethanolamine and phosphatidic acid, PEG-ceramide conjugates (e.g., PEG- CerC 14 or PEG-CerC20), PEG-modified dialkylamines, PEG-modified 1,2-diacyloxypropan- 3-amines, and any combinations thereof; and/or (vi) the TCV is substantially, essentially, or entirely free of organic solvents and/or detergents, or any combination of(i) to (v), optionally wherein the TCV is formed by: (a) generating a first solution by dissolving all components of the TCV, optionally at about 20-35 mM, in ethanol; (b) providing a second solution, which is aqueous and contains sodium acetate and/or sodium citrate, optionally at about 25 mM, optionally wherein the pH of the solution is about 4; ( c) combining the first and second solutions by gentle mixing ( optionally repeated manual reciprocation of the TCV-generating fluid in a pipette), micromixing optionally using a staggered herringbone micromixer (SHM) or T-junction or Y-junction mixing, or extrusion; and ( d) removing ethanol, optionally by dialysis or evaporation, further optionally the size of the TCV before encapsulation of the at least one cargo is in a range of about 9 nm to about 80 nm at pH of about 4. 9. The pharmaceutical composition of claim 7 or 8, wherein: (a) the amount of the at least one ionizable cationic lipid relative to the total lipid components of the TCV is: ( a-1) about 20 mo!% to about 30 mo!%, about 20, 21, 22, 23, 24, 2526, 27, 28, 29 or 30 +/- 0.5 mo!%, about 10 mol¾ to about 70 mo!%, about 10 mo!% to about 60 mo!%, about 10 mo!% to about 50 mol%, about 10 mo!% to about 40 mo!%, about 10 mo!% to about 30 mo!%, about 15 mo!% to about 25 mol¾, about 18 mo!% to about 22 mo!%, about 19 mo!% to about 21 mo!%, about 19.5 mo!% to about 20.5 mo!%, about 19.8 mo!% to about 20.2 mo!%, or about 20 mo!% or about 30 mo!%; or (a-2) about 10 mo!% to about 70 mo!%, about 20 mo!% to about 70 mo!%, about 30 mo!% to about 70 mo!%, about 40 mo!% to about 70 mo!%, about 40 mo!% to about 60 mo!%, about 45 mo!% to about 55 mo!%, about 48 mo!% to about 52 mo!%, about 49 mo!% to about 51 mo!%, about 49.5 mo!% to about 50.5 mo!%, about 49.8 mo!% to about 50.2 mo!%, or about 50 mo!%; (b) in (ii), the amount of the at least one helper lipid relative to the total lipid components of the TCV is about 10 mo!% to about 60 mo!%, about 10 mo!% to about 50 mo!%, about 10 mo!% to about 40 mo!%, about 20 mo!% to about 40 mo!%, about 25 mo!% to about 35 mo!%, about 28 mo!% to about 32 mo!%, about 29 mo!% to about 31 mo!%, about 28, 29, 30, or 31 +/- 0.5 mo!%, about 29.5 mo!% to about 30.5 mo!%, about 29.8 mo!% to about 30.2 mo!%, or about 30 mo!%; (c) in (iii), the amount of the at least one phospholipid relative to the total lipid components of the TCV is about 5 mo!% to about 65 mo!%, about 5 mo!% to about 55 mo!%, about 5 mo!% to about 45 mo!%, about 5 mo!% to about 35 mo!%, about 5 mo!% to about 25 mo!%, about 5 mo!% to about 15 mo!%, about 8 mo!% to about 12 mo!%, about 8, 9, 10, 11 or 12 mo!%+/- 0.5 mo!%, about 9 mo!% to about 11 mo!%, about 9.5 mo!% to about 10.5 mo!%, about 9.8 mo!% to about 10.2 mo!%, or about 10 mo!%; ( d) in (iv), the amount of the at least one cholesterol or cholesterol derivative relative to the total lipid components of the TCV is about 20 mo!% to about 60 mo!%, about 25 mo!% to about 55 mo!%, about 30 mo!% to about 50 mo!%, about 35 mo!% to about 45 mo!%, about 38 mo!% to about 42 mo!%, about 35, 36, 37, 38, 39, 40, 41, 42, 43, 44 or 45 mo!%+/- 0.5 mo!%, about 39 mo!% to about 41 mo!%, about 39.5 mo!% to about 40.5 mo!%, about 39.8 mo!% to about 40.2 mol%, or about 40 mo!%, or about 39%; and/or (e) in (v), the amount of the at least one PEG or PEG-lipid relative to the total lipid components of the TCV is about 0.1 mo!% to about 5 mo!%, 0.1 mo!% to about 4 mo!%, 0.1 mo!% to about 3 mo!%, 0.1 mo!% to about 2 mo!%, about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9 or 2.0 mo!%, 0.5 mo!% to about 1.5 mo!%, 0.8 mo!% to about 1.2 mo!%, 0.9 mo!% to about 1.1 mo!%, or about 1 mo!%. 10. The pharmaceutical composition of any one of claims 7-9, wherein: (I) the TCV comprises, essentially consists of, or consists of: (i) at least one ionizable cationic lipid, which is optionally DODMA, DLinDMA, DLin-KC2- DMA, DLin-MC3-DMA, SM-102, and/or ALC-0315; (ii) at least one helper lipid, which is optionally DOPE; (iii) at least one phospholipid, which is optionally DSPC; and (iv) at least one cholesterol or cholesterol derivative, optionally wherein the amounts of the at least one ionizable cationic lipid, the at least one helper lipid, the at least one phospholipid, and the at least one cholesterol or cholesterol derivative, relative to the total components of the TCV, is about 20-30 mo!%, about 20-30 mo!%, about 10 mo!%, and about 40 mo!%, respectively; or (II) the TCV comprises, essentially consists of, or consists of: (i) at least one ionizable cationic lipid, which is optionally DODMA, DLinDMA, DLin-KC2- DMA, DLin-MC3-DMA, SM-102, and/or ALC-0315; (ii) at least one helper lipid, which is optionally DOPE; (iii) at least one phospholipid, which is optionally DSPC; (iv) at least one cholesterol or cholesterol derivative; and (v) at least one PEG or PEG-lipid, which is optionally PEG-DMG, optionally wherein the amounts of the at least one ionizable cationic lipid, the at least one helper lipid, the at least one phospholipid, the at least one cholesterol or cholesterol derivative, and the at least one PEG or PEG-lipid, relative to the total components of the TCV, is about 20 mo!%, about 30 mo!%, about 10 mo!%, about 39 mo!%, and about I mo!%, respectively, and wherein the TCV is substantially, essentially, or entirely free of ethanol, methanol, isopropanol, tetrahydrofuran (THF), dimethyl sulfoxide (DMSO), dimethyl fonnamide (DMF), and acetonitrile (ACN), optionally wherein the TCV is substantially, essentially, or entirely free of organic solvents and detergents, further optionally wherein the TCV is substantially, essentially, or entirely free of destabilizing agents and/or is stable for prolonged periods of time at about 1 to about 40 °C, about 5 to about 35 °C, about 10 to about 30 °C, or about 15 to about 25 °C, and further optionally wherein the TCV or the pharmaceutical composition further comprises and/or is stored in the presence of at least one cryoprotectant, optionally wherein: (a) the cryoprotectant comprises a sugar-based molecule, which is optionally sucrose, trehalose, or a combination thereof; (b) the concentration of the cryoprotectant is about 1 % to about 40 %, about 3% to about 30%, about 5% to about 30%, about 10% to about 20%, or about 15%; ( c) the TCV is stable at a freezing temperature, optionally at about -20°C or about -80°C, optionally for at least about one week, at least about two weeks, at least about three weeks, at least about a month, at least about two months, at least about four months, at least about five months, at least about six months, at least about nine months, at least about a year, or at least about two years, or longer, further optionally for about one week to about two year, about two weeks to about a year, about three weeks to about nine month, about one to about six months, about one to five months, about one to four months, about one to three months, or about one to two months; or (d) any combination of(a)-(c). 11. The pharmaceutical composition of any one of claims 7-10, wherein the at least one SCD- associated gene: (i) is BCLJ IA or a variant thereof, optionally encoding the amino acid sequence of SEQ ID NO: 6, and/or a promoter or enhancer region of BCLJ IA, preferably the erythroid-enhancer region (EER) ofBCLJJA; (ii) is HBGJ or a variant thereof, optionally encoding the amino acid sequence of SEQ ID NO: 8, and/or a promoter or enhancer region of HBGJ; (iii) is HBG2 or a variant thereof, optionally encoding the amino acid sequence of SEQ ID NO: 9, and/or a promoter or enhancer region of HBG2; (iv) is HEB or a variant thereof (such as the sickle cell hemoglobin (HbS) variant of HBB, also known as the ~S allele, or the hemoglobin C (HbC) variant of HEB), optionally comprising the polynucleotide sequence of SEQ ID NO: 11, 21 or 31 and/or encoding the amino acid sequence of SEQ ID NO: 1, 2 or 3, and/or a promoter or enhancer region of HEB; (v) is KLFJ or a variant thereof, optionally encoding the amino acid sequence of SEQ ID NO: 7, and/or a promoter or enhancer region of KLFJ; and/or (vi) comprises one or more genes selected from the group consisting of BCLJ JA, HBGJ, HBG2, and HEB (the sickle cell hemoglobin (HbS) variant, also known as the ~S allele), KLFJ, SOX6, GATAJ, NF-E4 (or NFE4), COUP-TF, NR2Cl (also known as TR2), NR2C2 (also known as TR4), genes encoding members of the MBD2 protein complex, IKZFJ (also known as Jkaros), genes encoding other members of PYR complex (CHD4, HDAC2, RBBP7, SMARCBJ, SMARCCJ, SMARCC2, SMARCDJ, and SMARCEJ), BRGJ and variants of any of the foregoing, and genes that directly or indirectly modulate the expression variants of any of the foregoing . 12. The pharmaceutical composition of any one of claims 7-11, wherein: (I) the gene editing is mediated by a protease, nuclease, endonuclease, meganuclease, zinc finger nuclease (ZFN), transcription activator-like nuclease (T ALEN), or clustered regularly interspaced short palindromic repeats (CRISPR)-associated (Cas) nuclease, optionally resulting in at least one nucleic acid insertion, deletion, or replacement (e.g., resulting in a nonsense, missense, or silent mutation) in the at least one SCD-associated gene; and/or (II) the at least one cargo is capable of effecting gene editing comprises, essentially consists of, or consists of: (a) a Cas nuclease, a RNA encoding a Cas nuclease, or a nucleic acid such as a DNA or RNA encoding a Cas nuclease, optionally wherein the Cas nuclease is: (i) selected from the group consisting ofCas12a or Cpfl, Cas 9, Cas3, Cas8a2, Cas8b, Cas8c, Casi 0, Casl 1, Cas12, Casl2b, Cas12f, Cas12j, Cas13, Casl3a, C2cl, C2c3, and C2c2, (ii) a class 2 Cas nuclease, optionally a type II, type V, or type VI Cas nuclease, (iii) Casl2a of Acidaminococcus sp. (AsCasl2a), Lachnospiraceae bacterium (LbCasl2a), Francisella novicida (FnCasl2a), Moraxella bovoculi (MbCasl2a), Coprococcus eutactus (CeCas 12a), Butyrivibrio jibrisolvens (BdCas 12a); (iv) Cas12a or any engineered variants thereof, optionally comprising any one of SEQ ID NOS: 210-215, 220-221, 230,240,250, and 260; (v) Cas 9, optionally Cas9 of Streptococcus pyogenes (SpCas9), Staphylococcus aureus (SaCas9), Streptococcus thermophilus (StCas9), Neisseria meningitidis (NmCas9), Francisella novicida (FnCas9), Campylobacter jejuni (CjCas9), Streptococcus canis (ScCas9), or Staphylococcus auricularis (SauriCas9), or any engineered variants thereof, including SaCas9-HF, SpCas9-HF1, KKHSaCas9, eSpCas9, HypaCas9, Fokl-Fused dCas9, xCas9, SpRY (variant ofSpCas9), and SpG (variant ofSpCas9); and/or (vi) Cas9 comprising any one of SEQ ID NOS: 150-161; and (b) a guide RNA (gRNA) comprising a target-complementary sequence which is complementary to a target sequence within the at least one SCD-associated gene and/or a promoter or enhancer thereof, or a nucleic acid encoding said gRNA, optionally wherein: ( 1) the gRNA comprises a repeat sequence optionally at the 5 '-end of the target- complementary sequence, the repeat sequence comprising (i) an optional 5 '-end sequence, optionally comprising UAAUU or AAUU, (ii) a first stem sequence, optionally comprising UCUAC, (iii) a loop sequence, optionally comprising UCUU, UAAGU, UGUU, UUUU, UAUU, UGUUU, UUCG, or UUU, (iv) a second stem sequence which is reverse complementary to the first stem sequence, optionally comprising GUAGA, and (v) an optional 3'-end sequence, optionally comprising U, optionally wherein the repeat sequence comprises any one of SEQ ID NOS: 201-206; or (2) (i) the gRNA is a single guide RNA (sgRNA) comprising (i-1) a crRNA sequence comprising the target-complementary sequence and a crRNA backbone sequence and (i-2) a trans-activating CRISPR RNA (tracrRNA) sequence in a single strand, optionally wherein the crRNA sequence and the tracrRNA sequence are linked via a linker optionally comprising SEQ ID NO: 139, further optionally wherein the gRNA comprises the target-complementary sequence followed by a sgRNA backbone sequence of any of SEQ ID NOS: 141-144, optionally wherein the sgRNA backbone sequence is followed by one or more uracils, further optionally 1- 10 uracils, or (ii) the gRNA is a dual guide RNA ( dgRNA) formed by hybridization between (ii-1) a crRNA sequence comprising the target-complementary sequence and a crRNA backbone sequence and (ii-2) a tracrRNA, optionally wherein the crRNA backbone sequence and the tracrRNA comprise SEQ ID NOS: 145 and 146, respectively, or SEQ ID NOS: 147 and 148, respectively, optionally wherein the at least one cargo comprises, essentially consists of, or consists of a ribonucleoprotein (RNP), which is a complex of the gRNA and the Cas nuclease, optionally an RNP according to claim 5, further optionally wherein the RNP is formed by mixing Cas9 and gRNA at an approximately equimolar ratio, optionally for about 5 minutes, further optionally wherein the pharmaceutical composition or the at least one cargo further comprises a DNA repair template, yet further optionally wherein the at least one cargo (which comprises the RNP or the RNP and the DNA repair template) encapsulated in the TCV is obtained by: (i) providing an aqueous solution comprising the TCV, optionally wherein the pH of the aqueous solution is about 3 to about 8, further optionally about 4 to about 7.5; and (ii) mixing the at least one cargo with the aqueous solution, wherein mixing is effected under conditions suitable for the at least one cargo to be encapsulate within the TCV, wherein the mixing comprises gentle mixing ( optionally repeated manual reciprocation of the TCV-generating fluid in a pipette), micromixing optionally using a staggered herringbone micromixer (SHM) or T-junction or Y-junction mixing, or extrusion, optionally wherein the mixing time is about 0.1 second to about 20 minutes; wherein the aqueous solution is substantially, essentially, or entirely free of ethanol, methanol, isopropanol, tetrahydrofuran (THF), dimethyl sulfoxide (DMSO), dimethyl formamide (DMF), and acetonitrile (ACN), optionally substantially, essentially, or entirely free of organic solvents and detergents, further optionally substantially, essentially, or entirely free of destabilizing agents, wherein the mixing is performed substantially, essentially, or entirely free of ethanol, methanol, isopropanol, tetrahydrofuran (THF), dimethyl sulfoxide (DMSO), dimethyl formamide (DMF), and acetonitrile (ACN), optionally substantially, essentially, or entirely free of organic solvents and detergents, further optionally substantially, essentially, or entirely free of destabilizing agents, and optionally wherein the final ethanol concentration after encapsulation is 5% (v/v) or below, preferably 0.5% (v/v) or below, further optionally the size of the TCV after encapsulation of the at least one cargo is in a range of about 80 nm to about 1000 nm and/or in arrange of about 100 nm to about 250 nm, at pH of about 7.5, and optionally wherein the at least one cargo (which comprises the RNP or the RNP and the DNA repair template) encapsulated in the TCV is comprised in a matrix vesicle, which is optionally for gradual release of the TCV. 13. The pharmaceutical composition of claim 12, wherein in (II): (A) the at least one SCD-associated gene comprises or consists of BCLJ JA or a variant thereof and/or the erythroid-enhancer region (EER) of BCLJ JA and/or a promoter or enhancer region ofBCLJJA, optionally wherein the gRNA directs the Cas protein to and hybridize to a target sequence, which is located between nucleotide positions 60450520 to 60553654 of Chromosome 2 (according to Gene Assembly GRCh38.pl 3, positive or negative strand) and/or a promoter or enhancer region of BCLJ IA, optionally within or overlapping with the polynucleotide sequence of SEQ ID NO: 4 or 5; and/or (B) the at least one SCD-associated gene comprises or consists of HBGI or a variant thereof and/or a promoter or enhancer region of HBGI, optionally wherein the gRNA directs the Cas protein to and hybridize to a target sequence, which is located between nucleotide positions 5248269 to 5249857 of Chromosome 11 (according to Gene Assembly GRCh38.p14, positive or negative strand) and/or a promoter or enhancer region of HBGI, optionally within or overlapping with the polynucleotide sequence of SEQ ID NO: 95 or 97, preferably within or overlapping with the BCLl lA-binding site thereof; (C) the at least one SCD-associated gene comprises or consists of HBG2 or a variant thereof and/or a promoter or enhancer region of HBG2, optionally wherein the gRNA directs the Cas protein to and hybridize to a target sequence, which is located between nucleotide positions 5253188 to 5254781 of Chromosome 11 (according to Gene Assembly GRCh38.p14, positive or negative strand) and/or a promoter or enhancer region of HBG2, optionally within or overlapping with the polynucleotide sequence of SEQ ID NO: 95 or 97, preferably within or overlapping with the BCLl lA-binding site thereof; (D) the at least one SCD-associated gene comprises or consists of HEB or a variant thereof (such as the sickle cell hemoglobin (HbS) variant of HEB, also known as the ~S allele, or the hemoglobin C (HbC) variant of HEB) and/or a promoter or enhancer region of HEB, optionally wherein the gRNA directs the Cas protein to and hybridize to a target sequence, which is located between nucleotide positions 5225464 to 5227071 of Chromosome 11 (according to Gene Assembly GRCh38.pl 3, positive or negative strand) and which is optionally within the polynucleotide sequence of SEQ ID NO: 11, 21, or 31 or the sequence complementary thereto, further optionally within or overlapping with exon 1 of HEB, further optionally wherein the pharmaceutical composition or the at least one cargo further comprises a DNA repair template which allows for a knock-in of or correction to the wildtype HEB gene sequence (SEQ ID NO: 11) or the polynucleotide sequence encoding the wildtype beta-globin amino acid sequence (SEQ ID NO: 1); and/or (E) the at least one SCD-associated gene comprises or consists of KLFl or a variant thereof and/or a promoter or enhancer region of KLF 1, optionally wherein the gRNA directs the Cas protein to and hybridize to a target sequence, which is located between nucleotide positions 12884422 to 12887201 of Chromosome 19 (according to Gene Assembly GRCh38.pl 3, positive or negative strand) and/or a promoter or enhancer region of KLF 1, optionally wherein, in any one or more of (A)-(C) and (E), the pharmaceutical composition or the at least one cargo further comprises an isolated ODN, which optionally comprises: (I) a ssODN comprising a nucleic acid sequence having less than 90%, less than 85%, less than 80%, less than 75%, less than 70%, less than 65%, less than 60%, less than 55%, less than 50%, less than 45%, less than 40%, less than 35%, less than 30%, less than 30%, less than 25%, less than 20%, less than 15%, or less than 10% complementarity to the sequence of a segment within the SCD-associated gene, wherein the segment comprises the target sequence and/or the sequence complementary thereto and is about 20-300 nt, about 25-200 nt, about 30-160 nt, about 40-140 nt, about 50-120 nt, about 60-100 nt, about 70-90 nt, about 75-85 nt, about 20 nt, about 25 nt, about 30 nt, about 35 nt, about 40 nt, about 45 nt, about 50 nt, about 55 nt, about 60 nt, about 65 nt, about 70 nt, about 75 nt, about 80 nt, about 85 nt, about 90 nt, about 95 nt, about 100 nt, about 105 nt, about 120 nt, about 125 nt, about 130 nt, about 135 nt, about 140 nt, about 145 nt, about 150 nt, about 155 nt, about 160 nt, about 175 nt, about 180 nt, about 185 nt, about 190 nt, about 195 nt, about 200 nt, about 205 nt, about 220 nt, about 225 nt, about 230 nt, about 235 nt, about 240 nt, about 245 nt, about 250 nt, about 255 nt, about 260 nt, about 275 nt, about 280 nt, about 285 nt, about 290 nt, about 295 nt, or about 300 nt in length; or (II) a dsODN, which comprises a first strand comprising a ssODN sequence of (I) and a second strand complementary to the first strand, further optionally wherein: (i) the isolated ODN is about 30-160 nt, 50-120 nt, about 60-100 nt, about 70-90 nt, about 75-85 nt, about 80 nt in length; and/or (ii) the nucleic acid sequence of the ODN has at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to SEQ ID NO: 190 or a reverse complement thereof. 14. The pharmaceutical composition of claim 13, wherein: in (A), (i) the target sequence is or comprises SEQ ID NO: 70 or at least the first 17, 18, or 19 nucleotides from the 3' end of SEQ ID NO: 70, and/or the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 71, or at least the first 17, 18, or 19 nucleotides thereof from the 5' end of SEQ ID NO: 71; (ii) the target sequence is or comprises SEQ ID NO: 276 or at least the first 17, 18, or 19 nucleotides from the 3' end of SEQ ID NO: 276, and/or the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 277, or at least the first 17, 18, or 19 nucleotides thereof from the 5' end of SEQ ID NO: 277; (iii) the target sequence is or comprises SEQ ID NO: 278 or at least the first 17, 18, or 19 nucleotides from the 3' end of SEQ ID NO: 278, and/or the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 279, or at least the first 17, 18, or 19 nucleotides thereof from the 5' end of SEQ ID NO: 279; (iv) the target sequence is or comprises SEQ ID NO: 270 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 270, and/or the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 271, or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 271; (v) the target sequence is or comprises SEQ ID NO: 272 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 272, and/or the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 273, or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 273; (vi) the target sequence is or comprises SEQ ID NO: 274 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 274, and/or the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 275, or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 275; ( vi) the target sequence is or comprises SEQ ID NO: 64 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 64, and/or the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 65, or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 65; ( vii) the target sequence is or comprises SEQ ID NO: 66 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 66, and/or the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 67 or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 67; and/or (viii) the target sequence is or comprises SEQ ID NO: 68 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 68, and/or the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 69 or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 69; optionally wherein the pharmaceutical composition or the at least one cargo further comprises an isolated ODN, which optionally comprises: (I) a ssODN comprising or consisting of a nucleic acid sequence having at least 80%, at least 85%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% complementarity to the sequence of a segment within BCLl 1 A or a variant thereof and/or the erythroid-enhancer region (EER) of BCLI JA and/or a promoter or enhancer region of BCLI IA, wherein the segment comprises the target sequence and/or the sequence complementary thereto and is about 20-300 nt, about 25-200 nt, about 30-160 nt, about 40-140 nt, about 50-120 nt, about 60-100 nt, about 70-90 nt, about 75-85 nt, about 20 nt, about 25 nt, about 30 nt, about 35 nt, about 40 nt, about 45 nt, about 50 nt, about 55 nt, about 60 nt, about 65 nt, about 70 nt, about 75 nt, about 80 nt, about 85 nt, about 90 nt, about 95 nt, about 100 nt, about 105 nt, about 120 nt, about 125 nt, about 130 nt, about 135 nt, about 140 nt, about 145 nt, about 150 nt, about 155 nt, about 160 nt, about 175 nt, about 180 nt, about 185 nt, about 190 nt, about 195 nt, about 200 nt, about 205 nt, about 220 nt, about 225 nt, about 230 nt, about 235 nt, about 240 nt, about 245 nt, about 250 nt, about 255 nt, about 260 nt, about 275 nt, about 280 nt, about 285 nt, about 290 nt, about 295 nt, or about 300 nt in length, optionally wherein: (i) the isolated ODN is about 30-160 nt, 50-120 nt, about 60-100 nt, about 70-90 nt, about 75-85 nt, about 80 nt or about 75, 76, 77, 78, 79, 81, 82, 83, 84, or 85 nt in length; and/or (ii) the nucleic acid sequence of the ssODN has at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to SEQ ID NO: 305,306,303,304, 301, or 302; or (II) a dsODN, which comprises a first strand comprising a ssODN sequence of (I) and a second strand complementary to the first strand; in (B) and/or (C) , (i) the target sequence is or comprises SEQ ID NO: 86 or at least the first 17, 18, or 19 nucleotides from the 3' end of SEQ ID NO: 86, and/or the target- complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 87 or at least the first 17, 18, or 19 nucleotides thereof from the 5' end of SEQ ID NO: 87; (ii) the target sequence is or comprises SEQ ID NO: 286 or at least the first 17, 18, or 19 nucleotides from the 3' end of SEQ ID NO: 286, and/or the target- complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 287 or at least the first 17, 18, or 19 nucleotides thereof from the 5' end of SEQ ID NO: 287; (iii) the target sequence is or comprises SEQ ID NO: 288 or at least the first 17, 18, or 19 nucleotides from the 3' end of SEQ ID NO: 288, and/or the target- complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 289 or at least the first 17, 18, or 19 nucleotides thereof from the 5' end of SEQ ID NO: 289; (iv) the target sequence is or comprises SEQ ID NO: 280 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 280, and/or the target- complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 281 or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 281; (v) the target sequence is or comprises SEQ ID NO: 282 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 282, and/or the target- complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 283 or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 283; (vi) the target sequence is or comprises SEQ ID NO: 284 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 284, and/or the target- complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 285 or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 285; (vii) the target sequence is or comprises SEQ ID NO: 84 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 84, and/or the target- complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 85 or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 85, optionally wherein the pharmaceutical composition or the at least one cargo further comprises an isolated ODN, which optionally comprises: (I) a ssODN comprising or consisting of a 5' homology arm, an optional central region, and a 3' homology arm, wherein: (a) (i) the 5' homology arm comprises or consists of the sequence corresponding to: (i-1) a sequence within SEQ ID NO: 195, which: contains at least 5, least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50 nucleotides, at least 55, at least 60, at least 65, at least 70, at least 75, or at least 80 nucleotides, about 5-80 nucleotides, about 10- 70 nucleotides, about 20-60 nucleotides, about 30-50 nucleotides, about 35-45 nucleotides, or about 40 nucleotides; and ends at the first, second, third, fourth, fifth, sixth, seventh, eighth, nineth, 10111, 1 \111, 1211\ 1311\ 1411\ 15111, 1611\ 17111, 1811\ 1911\ or 20111 nucleotide counting from the 3 '-end of SEQ ID NO: 195 or a more upstream nucleotide within SEQ ID NO: 195; (i-2) a sequence within SEQ ID NO: 195, which: contains about 35-40 nucleotides or about 40 nucleotides; and ends at the first nucleotide counting from the 3 '-end of SEQ ID NO: 195; (i-3) or a sequence comprising at least one (such as one, two, three, four, five, six, seven, eight, nine, or ten) mutation(s) relative to the sequence of (i-1) or (i-2), (ii) the optional central region: ( ii-1) is absent; or (ii-2) comprises a sequence of one, two, three, five, six, seven, eight, nine, ten, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more nucleotides optionally not comprising the five consecutive nucleotides of the BCLl lA-binding motif of 5' -TGACC-3', further optionally comprising or consisting of the sequence of 5'-TGAC-3', and (iii) the 3' homology arm comprises or consists of the sequence corresponding to: (iii-1) a sequence within SEQ ID NO: 196, which: contains at least 5, least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50 nucleotides, at least 55, at least 60, at least 65, at least 70, at least 75, or at least 80 nucleotides, about 5-80 nucleotides, about 10- 70 nucleotides, about 20-60 nucleotides, about 30-50 nucleotides, about 35-45 nucleotides, or about 40 nucleotides; and starts at the first, second, third, fourth, fifth, sixth, seventh, eighth, nineth, 10th, 111", 12th, 13th, 141", 151\ 16th, 171\ 18th, 191\201\21°1,221\231\24th, 25°1, 26th, 27th, 281\ 291", 301", 31th, 32°1, 33t11, 34°1, 35t11, 36th, 37th, 33th, 39°1, or 40th nucleotide counting from the 5 '-end of SEQ ID NO: 196 or a more upstream nucleotide within SEQ ID NO: 196; (iii-2) a sequence within SEQ ID NO: 196, which: contains about 35-40 nucleotides or about 40 or 41 nucleotides; and starts at the 13th nucleotide counting from the 5'-end of SEQ ID NO: 196; (iii-3) or a sequence comprising at least one (such as one, two, three, four, five, six, seven, eight, nine, or ten) mutation(s) relative to the sequence of(iii-1) or (iii-2), optionally wherein the ssODN comprises or consists of the. sequence of SEQ ID NO: 191; or (b) the sequence of the ssODN is fully complementary to any of the ssODNs of (a), optionally wherein the sequence of the ssODN comprises or consists of the sequence of SEQ ID NO: 192; or (II) a dsODN, which comprises a first strand comprising any of the ssODN sequences of (I) and a second strand complementary to the first strand; in (D), (i) the target sequence is or comprises SEQ ID NO: 24 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 24, and/or the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 25, or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 25; (ii) the target sequence is or comprises SEQ ID NO: 44 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 44 such as SEQ ID NO: 46, and/or the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 45, or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 45 such as SEQ ID NO: 47; and/or (iii) the target sequence is or comprises SEQ ID NO: 48 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 48, and/or the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 49, or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 49, optionally wherein the pharmaceutical composition or the at least one cargo further comprises a DNA repair template, which optionally comprise: (I) a single-stranded oligo DNA nucleotide molecule (ssODN) comprising or consisting of a 5' homology arm , a central region, and a 3' homology arm, wherein: ( a) (i) the 5' homology arm comprises or consists of (i-1) the sequence of SEQ ID NO: 112, (i-2) the sequence corresponding to the first nucleotide to at least the 20th nucleotide ( e.g., at least the 3ot11, such as the 39t11, at least the 4ot11, such as the 49t1,, or at least the 501\ such as the 50th or the 59th) counting from the 3'- end of SEQ ID NO: 112, (i-3) or a sequence comprising at least one (such as one, two, three, four, five, six, seven, eight, nine, or ten) silent mutation(s) relative to the sequence of (i-1) or (i-2), (ii) the central region has the sequence of 5'-CTCA-3', 5'-TTCA-3', 5'-CTCT-3', 5'-TTCT-3', 5'-CTCC-3', 5'-TTCC-3', 5'-CTCG-3', or 5'-TTCG-3', and (iii) the 3' homology arm comprises or consists of (i-1) the sequence of SEQ ID NO: 122, (i-2) the sequence corresponding to the first nucleotide to at least the 20th nucleotide (e.g., at least the 30th, such as the 37t1,, at least the 40111, such as the 47111, or at least the 50th, such as the 57th) counting from the 5 '-end of SEQ ID NO: 122, (i-3) or a sequence comprising at least one (such as one, two, three, four, five, six, seven, eight, nine, or ten) silent mutation(s) relative to the sequence of (iii-l ) or (iii-2), optionally wherein the ssODN comprises the consists of the sequence of any of SEQ ID NOs: 170, 172, 174, 176, and 101-108; (b) the sequence of the ssODN is fully complementary to any of the ssODNs of(a), optionally wherein the sequence of the ssODN is or comprises any of SEQ ID NOs: 169,171,173, and 175; or (II) a double-stranded DNA molecule, which comprises a first strand comprising any of the ssODN sequences of (I) and a second strand complementary to the first strand; and/or in (E), (i) the target sequence is or comprises SEQ ID NO: 74 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 74, and/or the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 75 or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 75; and/or (ii) the target sequence is SEQ ID NO: 76 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 76, and/or the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 77 or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 77, and/or wherein the gRNA comprises at least one gRNA according to claim 1. 15. The pharmaceutical composition of claim 14, wherein: in (A), (i) the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 71,277, or 279 and the Cas nuclease is Cas12a, optionally AsCasl2a or LbCasl2a, further optionally comprising any one of SEQ ID NOS: 210-215 or 220-221, or (ii) the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 271,273,275, 65, 69, or 67 and the Cas nuclease is Cas9, optionally SpCas9 or SaCas9, further optionally comprising SEQ ID NO: 150, optionally wherein, the pharmaceutical composition or the at least one cargo further comprises an isolated ODN, which optionally comprises: (I) a ssODN, which comprises the nucleic acid sequence of (i) SEQ ID NO: 190 and/or the sequence complementary thereto or (ii) SEQ ID NO: 305, 306,303,304, 301, or 302, and is about 60-120 nt, about 70-90 nt, or about 80 nt in length; or (II) a dsODN, which comprises a first strand comprising a ssODN sequence of (I) and a second strand complementary to the first strand; or in (B) and/or (C), (i) the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 87,287, or 289 and the Cas nuclease is Cas12a, optionally AsCas 12a or LbCas 12a, further optionally comprising any one of SEQ ID NOS: 210-215 or 220-221, or (ii) the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 281,283,285, or 85 and the Cas nuclease is Cas9, optionally SpCas9 or SaCas9, further optionally comprising SEQ ID NO: 150, optionally wherein the pharmaceutical composition or the at least one cargo further comprises an isolated ODN, which optionally comprises: (I) a ssODN comprising or consisting of the sequence of SEQ ID NO: 191 or 192; or (II) which comprises a first strand comprising a ssODN sequence of (I) and a second strand complementary to the first strand. 16. The pharmaceutical composition of any one of claims 7-15, wherein the at least one cargo is capable of altering the expression and comprises, essentially consists of, or consists of a nucleic acid molecule, optionally a ribonucleic acid (RNA), a single or double stranded RNA, a small interfering RNA (siRNA), a short hairpin RNA, a microRNA (miRNA), a messenger RNA (mRNA), a deoxyribonucleic acid (DNA), a double or single stranded DNA, a plasmid DNA, a complementary DNA (cDNA), and/or a locked nucleic acid, optionally wherein the at least one cargo encapsulated in the TCV is obtained by: (i) providing an aqueous solution comprising the TCV, optionally wherein the pH of the aqueous solution is about 3 to about 8, further optionally about 4 to about 7.5; and (ii) mixing the at least one cargo with the aqueous solution, wherein mixing is effected under conditions suitable for the at least one cargo to be encapsulate within the TCV, wherein the mixing comprises gentle mixing ( optionally repeated manual reciprocation of the TCV-generating fluid in a pipette), micromixing optionally using a staggered herringbone micromixer (SHM) or T-junction or Y-junction mixing, or extrusion, optionally wherein the mixing time is about 0.1 second to about 20 minutes; wherein the aqueous solution is substantially, essentially, or entirely free of ethanol, methanol, isopropanol, tetrahydrofuran (THF), dimethyl sulfoxide (DMSO), dimethyl formamide (DMF), and acetonitrile (ACN), optionally substantially, essentially, or entirely free of organic solvents and detergents, further optionally substantially, essentially, or entirely free of destabilizing agents, wherein the mixing is performed substantially, essentially, or entirely free of ethanol, methanol, isopropanol, tetrahydrofuran (THF), dimethyl sulfoxide (DMSO), dimethyl formamide (DMF), and acetonitrile (ACN), optionally substantially, essentially, or entirely free of organic solvents and detergents, further optionally substantially, essentially, or entirely free of destabilizing agents, and optionally wherein the final ethanol concentration after encapsulation is 5% (v/v) or below, preferably 0.5% (v/v) or below.

7. The pharmaceutical composition of any one of claims 7-16, wherein: (A) the pharmaceutical composition comprises at least another cargo, which is encapsulated in the TCV encapsulating said at least one cargo or in a different TCV, optionally wherein the at least another cargo is according to the at least one cargo of any one of claims 7-15; (B) the pharmaceutical composition further comprises at least one agent that promotes stem cell mobilization, optionally selected from the group consisting of granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), Plerixafor, stem cell factor (SCF), CXCR4 antagonists (e.g., POL6326, BKT-140, TG-0054), CXCL12 neutralizers ( e.g., NOX-Al 2), Sphingosine-l-phosphate (SIP) antagonists ( e.g., SEW2871 ), vascular cell adhesion molecule-I/Very Late Antigen 4 (VCAM/VLA-4) inhibitors (e.g., BIO 5192), parathyroid hormone, protease inhibitors (e.g., Bortezomib), Grof (e.g., SB-251353), and hypoxia inducible factor (HIF) stabilizers (e.g., FG-4497); (C) the pharmaceutical composition further comprises at least one agent that promotes erythropoiesis, which optionally comprises: (i) an agent selected from the group consisting ofSCF, GM-CSF, interleukin-3 (IL-3), interleukin-9 (IL-9), erythropoietin (EPO) (or an engineered EPO or EPO mimetic), TGF- beta, growth differentiating factor 11 (GDFl 1), Activin A, Transferrin (Tf), ferritin, ferroportin, hepcidin, vitamin B 12, folic acid, and copper, (ii) an agent selected from the group consisting ofGATA-1, STAT5A, STAT5B, MCL-1, BCL-xL, and HSP70, a RNA or DNA encoding thereof, optionally wherein the agent is encapsulated in the TCV encapsulating said at least one cargo or in a different TCV; and/or (iii) an inhibitor or silencer of a negative regulator of erythropoiesis, optionally wherein the negative regulator is selected from the group consisting of inhibin, TGF-beta, BID (a member of the BCL-2 family), Fas ligand, Fas, and caspases, optionally wherein the agent is encapsulated in the TCV encapsulating said at least one cargo or in a different TCV; and/or (D) the TCV comprises at least one targeting moiety which allows the TCV to carry the at least one cargo preferentially into one or more target cells, optionally wherein the one or more target cells comprise hematopoietic stem cells (HSCs), hematopoietic stem and progenitor cells (HSPCs), multipotent progenitor cells (MPPs), common myeloid progenitors (CMPs), megakaryocyte-erythroid progenitors (MEPs), hematopoietic progenitor cells (HPCs), erythroid progenitors (e.g., burst-forming unit erythroid cells (BFU-Es), colony-forming unit erythroid cells (CFU-Es)), proerythroblasts, erythroblasts (basophilic erythroblasts, early erythroblasts ( e.g., type I, type II), polychromatic erythroblasts, intermediate erythroblasts, acidophilic erythroblasts, late erythroblasts, normoblasts, reticulocytes (before nucleus expulsion), or any combinations thereof, preferably HSCs and/or HSPCs, optionally wherein the targeting moiety targets CD34. 18. A method of effecting gene editing and/or gene expression alteration in one or more target cells in vivo in a subject in need thereof, the method comprising injecting the pharmaceutical composition of any one of claims 7-17 into the bone marrow of the subject, wherein the one or more target cells comprise HSCs, HSPCs, MPPs, CMPs, MEPs, HPCs, erythroid progenitors (e.g., BFU-Es, CFU-Es), proerythroblasts, erythroblasts (basophilic erythroblasts, early erythroblasts (e.g., type I, type II), polychromatic erythroblasts, intermediate erythroblasts, acidophilic erythroblasts, late erythroblasts, normoblasts, reticulocytes (before nucleus expulsion), or any combinations thereof, preferably HSCs and/or HSPCs. optionally the subject has or has a risk of developing SCD, which is optionally SCA, HbSC, or HbS ~- thalassaemia. 19. The method of claim 18, which comprises one or more of the following features: (i) the pharmaceutical composition is according to the pharmaceutical composition of any of one of claims 7-17, and comprises per mL about 300 pmol to about 30000 pmol, optionally about 500 to about 10000 pmol, about 1000 to about 5000 pmol, about 2000 to about 4000 pmol, about 2500 to about 3000 pmol, or about 2700 pmol of the RNP or the nucleic acid molecule; (ii) the injecting comprising injecting the pharmaceutical composition in a continuous flow of about 25 mL to 125 mL per minute, optionally about 25 mL to 50 mL per minute, about 50 mL to 100 mL per minute, about 100 mL to 125 mL per minute, about 40 mL to about 80 mL per minute, or about 50 mL to about 70 mL per minute; (iii) the injecting is a slow bolus push using an instrument with an intraosseous device or needle optionally having a needle length of about 50 to about 100 mm or about 70 to about 80 nm; (iv) the injecting is effected, optionally two or more times, to reach a minimum of about 10%, about 15%, about 20%, about 30%, or an about final 15-30% or about final 20-40% HSCs and HSPCs with successful gene editing and/or gene expression alteration among the total HSCs and HSPCs in the bone marrow; (v) the injecting is effected two or more times, optionally about 3-5 time, optionally about once a week, about every 2 weeks, or about every 3 weeks, about once a month, about every 3 months, about every 6 months, or about once per year; (vi) the bone marrow is of tibia, femur, sternum, skull, ribs, pelvis (e.g., iliac), or any combinations thereof; and/or (vii) the subject (a) is in the immediate post-natal period, optionally about 6 weeks old or younger, (b) is about 3 month old or younger, ( c) still comprises sufficient amount of fetal hemoglobin (HbF) relative to adult hemoglobin (HbA) (e.g., HbF:HbA is about 2:1, about 1: 1, about 1 :2, about 1 :3, about 1:4, about 1 :5, or about 1: 10), and/or (d) has not fully developed SCD and is prior to manifesting a symptom or complication. 20. A method of effecting gene editing and/or gene expression alteration in one or more target cells in vivo in a subject in need thereof, the method comprising: (I) administering, optionally intravenously, to the subject at least one agent that promotes stem cell mobilization (from the bone marrow to the peripheral circulation), optionally selected from the group consisting ofG-CSF (filgrastim), GM-CSF, Plerixafor, SCF, CXCR4 antagonists (e.g., POL6326, BKT-140, TG-0054), CXCL12 neutralizers (e.g., NOX-A12), Sphingosine-1-phosphate (SIP) antagonists (e.g., SEW2871), VCAM/VLA-4 inhibitors (e.g., BIO 5192), parathyroid hormone, protease inhibitors (e.g., Bortezomib), Grof (e.g., SB- 251353), and hypoxia inducible factor (HIF) stabilizers ( e.g., FG-4497), and (II) injecting, optionally intravenously, the pharmaceutical composition of any one of claims 7- 17 into the peripheral circulation of the subject, wherein the one or more target cells comprise HSCs, HSPCs, MPPs, CMPs, MEPs, HPCs, erythroid progenitors (e.g., BFU-Es, CFU-Es), proerythroblasts, erythroblasts (basophilic erythroblasts, early erythroblasts ( e.g., type I, type II), polychromatic erythroblasts, intermediate erythroblasts, acidophilic erythroblasts, late erythroblasts, normoblasts, reticulocytes (before nucleus expulsion), or any combinations thereof, preferably HSCs and/or HSPCs, optionally the subject has or has a risk of developing SCD, which is optionally SCA, HbSC, or HbS ~-thalassaemia. 21. The method of claim 20, which comprises one or more of the following features: (i) said administering comprises intravenous administration of G-CSF followed by intravenous administration of plerixafor prior to said injecting, optionally wherein: (a) the dosing ofG-CSF is about 5-30 µg/kg/day, preferably about 10 µg/kg/day, for about 3- 5 days, preferably 4 days, (b) the dosing of plerixafor starts once the peripheral blood CD34+ cells are <20 cells/µL and/or on the day of the last G-CSF administration or the following day, and/or ( c) the dosing of plerixafor is about 0.1-0.5 mg/kg, preferably about 0.2-0.3 mg/kg or about 0.24 mg/kg; (ii) the pharmaceutical composition is according to the pharmaceutical composition of any one of claims 7-17 and comprises per mL about 300 pmol to about 30000 pmol, optionally about 500 to about l 0000 pmol, about 1000 to about 5000 pmol, about 2000 to about 4000 pmol, about 2500 to about 3000 pmol, or about 2700 pmol of the RNP or the nucleic acid molecule; (iii) said injecting: (a) starts once the peripheral blood CD34+ cells are 60 cells/µL or more, (b) is a single injection, optionally about 3- 7 days, about 4-6 days, or about 5 days after the last plerixafor administration, and/or (c) occurs once daily for one week following the last plerixafor administration; (iv) the injecting comprising injecting the pharmaceutical composition in a continuous flow of about 25 mL to 125 mL per minute, optionally about 25 mL to 50 mL per minute, about 50 mL to 100 mL per minute, about 100 mL to 125 mL per minute, about 40 mL to about 80 mL per minute, or about 50 mL to about 70 mL per minute; (v) the combination of said administering and said injecting is effected, optionally two or more times, to reach a minimum of about l 0%, about 15%, about 20%, about 30%, or an about final 15-30% or about final 20-40% HSCs and HSPCs with successful gene editing and/or gene expression alteration among the total HSCs and HSPCs in the peripheral circulation; ( vi) the combination of said administering and said injecting is effected, optionally two or more times, to reach a minimum of about 10%, about 15%, about 20%, about 30%, or an about final 20-30% increase in the peripheral HSCs and HSPCs expressing HbF, or a minimum of about 10%, about 15%, about 20%, about 30%, or an about final 20-30% increase in the total HbF expression levels in the total HSCs and HSPCs in the peripheral circulation, optionally wherein the SCD-associated gene is BCLJ JA, HBGJ, HBG2, or KLFJ; (vii) the combination of said administering and said injecting is effected two or more times, optionally about 3-5 time, optionally about once a week, about every 2 weeks, or about every 3 weeks, about once a month, about every 3 months, about every 6 months, or about once per year; (viii) the subject (a) is in the immediate post-natal period, optionally about 6 weeks old or younger, (b) is about 3 month old or younger, (c) still comprises sufficient amount ofHbF relative to HbA (e.g., HbF:HbA is about 2:1, about 1:1, about 1:2, about 1:3, about 1:4, about 1 :5, or about 1: 10), and/or (d) has not fully developed SCD and is prior to manifesting a symptom or complication. 22. A method of preventing, ameliorating, or treating SCD, which is optionally SCA, HbSC, or HbS ~-thalassaemia, in a subject in need thereof, the method comprises the method according to claim 18 or 19 and/or the method according to claim 20 or 21, optionally the effect of the method is evaluated based on: (i) % HSCs and HSPCs in the blood with successful gene editing and/or gene expression alteration; (ii) the number of HSCs and HSPCs in the blood with successful gene editing and/or gene expression alteration; (iii) % HSCs and HSPCs expressing HbF, optionally wherein the SCD-associated gene is BCLl IA or KLFl; (iv) the number ofHSCs and HSPCs expressing HbF, optionally wherein the SCD-associated gene is BCLI JA, HBGJ, HBG2, or KLFJ; (v) the expression level of the at least one SCD-associated gene or gene product or molecule, optionally beta-globin, beta-globin variant (HbS variant or HbC variant), gamma-globin 1, gamma-globin 2, HbF, HbA, BCLl lA, and/or KLFl; and/or (iv) changes in the symptom optionally pain, swelling of hands and feet, infection frequency, growth, and/or symptoms associated with vision, and optionally wherein the method further comprises: (A) administering at least one agent that promotes erythropoiesis, which optionally comprises: (a) an agent selected from the group consisting of SCF, GM-CSF, IL-3, IL-9, EPO (or an engineered EPO or EPO mimetic), TGF-beta, GDFl 1, Activin A, Tf, ferritin, ferroportin, hepcidin, vitamin B12, folic acid, and copper, (b) an agent selected from the group consisting of GATA-I, STAT5A, STAT5B, MCL-1, BCL-xL, and HSP70, a RNA or DNA encoding thereof, optionally wherein the agent is encapsulated in the TCV encapsulating said at least one cargo or in a different TCV, and/or ( c) an inhibitor or silencer of a negative regulator of erythropoiesis, optionally wherein the negative regulator is selected from the group consisting of inhibin, TGF-beta, BID (a member of the BCL-2 family), Fas ligand, Fas, and caspases, optionally wherein the agent is encapsulated in the TCV encapsulating said at least one cargo or in a different TCV; and/or (B) administering at least one other agent for treating SCD, which optionally comprises hydroxyurea, L-glutamine oral powder, crizanlizumab, a general pain medication, voxelotor, or any combination thereof. 23. A method of manufacturing the pharmaceutical composition of claim 13(11), 14, or 15, comprising: (a) providing an aqueous solution comprising the TCV, optionally wherein the pH of the aqueous solution is about 3 to about 8, further optionally about 3.5 to about 7.5, about 3.5 to about 5.5, or about4; and (b) mixing the at least one cargo with the aqueous solution under conditions suitable for the at least one cargo to be encapsulate within the TCV, optionally wherein the at least one cargo comprises the RNP of claim 5, and optionally wherein the mixing is for about 0.1 second to about 20 minutes, optionally via gentle mixing (optionally repeated manual reciprocation of the TCV-generating fluid in a pipette), optionally via micromixing, further optionally using a staggered herringbone micromixer (SHM) or T-junction or Y-junction mixing, or extrusion. 24. A composition comprising: (A) a Cas nuclease or a nucleic acid encoding the Cas nuclease, optionally wherein the Cas nuclease is: (i) selected from the group consisting ofCasl2a or Cpfl, Cas9, Cas3, Cas8a2, Cas8b, Cas8c, Casl0, Casl 1, Cas12, Cas12b, Cas12f, Casl2j, Cas13, Cas13a, Cas14, C2cl, C2c3, and C2c2; (ii) a class 2 Cas endonuclease, optionally a type II, type V, or type VI Cas nuclease; (iii) Cas12a of Acidaminococcus sp. (AsCas12a), Lachnospiraceae bacterium (LbCas12a), Francisella novicida (FnCas12a), Moraxella bovoculi (MbCas12a), Coprococcus eutactus ( CeCas 12a ), Butyrivibrio ji.brisolvens (BdCas 12a ); (iv) Cas12a or any engineered variants thereof, optionally comprising any one of SEQ ID NOS: 210-215, 220-221, 230,240,250, and 260; auricularis (SauriCas9), or any engineered variants thereof, including SaCas9-HF, SpCas9- HF1, KKHSaCas9, eSpCas9, HypaCas9, Fokl-Fused dCas9, xCas9, SpRY (variant of SpCas9), and SpG (variant ofSpCas9); and/or (vi) Cas9, optionally comprising any one of SEQ ID NOS: 150-161; (B) a gRNA or a nucleic acid encoding the gRNA, wherein the gRNA comprises a target- complementary sequence of interest such that the target-complementary sequence is capable of binding to a target sequence of interest within a target gene of interest thereby guiding the Cas9 nuclease of (A) to and allowing for cleavage at a target site of interest within said target gene of interest, optionally wherein the target gene of interest is not a human huntingtin gene, further optionally wherein the target gene of interest is not a huntingtin gene, and/or optionally wherein the target sequence is not within an about 120-nt, about 100-nt, about 90-nt, or about 80-nt sequence encompassing the junction between intron 12 and exon 13 of a human huntingtin gene and/or not within intron 12 and/or exon 13 of a human huntingtin gene; and (C) an isolated ODN or a nucleic acid encoding the isolated ODN, wherein the isolated ODN comprises: (I) a ssODN comprising a nucleic acid sequence having less than 90%, less than 85%, less than 80%, less than 75%, less than 70%, less than 65%, less than 60%, less than 55%, less than 50%, less than 45%, less than 40%, less than 35%, less than 30%, less than 30%, less than 25%, less than 20%, less than 15%, or less than 10% complementarity to the sequence of a segment within the target gene of interest, wherein the segment comprises the target sequence and/or the sequence complementary thereto and is about 20-300 nt, about 25-200 nt, about 30-160 nt, about 40-140 nt, about 50-120 nt, about 60-100 nt, about 70-90 nt, about 75- 85 nt, about 20 nt, about 25 nt, about 30 nt, about 35 nt, about 40 nt, about 45 nt, about 50 nt, about 55 nt, about 60 nt, about 65 nt, about 70 nt, about 75 nt, about 80 nt, about 85 nt, about 90 nt, about 95 nt, about 100 nt, about 105 nt, about 120 nt, about 125 nt, about 130 nt, about 135 nt, about 140 nt, about 145 nt, about 150 nt, about 155 nt, about 160 nt, about 175 nt, about 180 nt, about 185 nt, about 190 nt, about 195 nt, about 200 nt, about 205 nt, about 220 nt, about 225 nt, about 230 nt, about 235 nt, about 240 nt, about 245 nt, about 250 nt, about 255 nt, about 260 nt, about 275 nt, about 280 nt, about 285 nt, about 290 nt, about 295 nt, or about 300 nt in length; or (II) a dsODN, which comprises a first strand comprising a ssODN sequence of (I) and a second strand complementary to the first strand, optionally wherein: (i) the isolated ODN is about 30-160 nt, 50-120 nt, about 60-100 nt, about 70-90 nt, about 75- 85 nt, about 80 nt in length; (ii) the nucleic acid sequence of the isolated ODN has at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to SEQ ID NO: 190 or a sequence complementary thereto; (iii) the isolated ODN has no significant alignment (optionally defined bye-value< 0.05) with an about 120-nt, about 100-nt, about 90 nt, about 85-nt, about 80-nt, about 75-nt, or about 70-nt sequence of the target gene of interest encompassing the target sequence or a reverse complement thereof, as analyzed by Blast®N or Nucleotide BLAST®; and/or (iv) the delta G value for a hypothetical dimer of the isolated ODN and an about 120-nt, about 100-nt, about 90 nt, about 85-nt, about 80-nt, about 75-nt, or about 70-nt sequence of the target gene encompassing the target sequence or a reverse complement thereof is about -100 kcal/mol or higher, about -80 kcal/mo! or higher, about -60 kcal/mol or higher, about -50 kcal/mo! or higher, about -40 kcal/mo! or higher, about -30 kcal/mo! or higher, about -20 kcal/mo! or higher, about -18 kcal/mo! or higher, about -16 kcal/mo! or higher, about -15 kcal/mo! or higher, about -14 kcal/mo! or higher, about -12 kcal/mo! or higher, or about -10 kcal/mo! or higher, optionally wherein the Cas nuclease and the gRNA are comprised in a RNP, and optionally wherein one or more of the Cas nuclease, the gRNA, and/or the isolated ODN are encapsulated in a carrier, optionally a lipid-based TCV, further optionally a lipid-based TCV according to any one of claims 8-10. 25. A method of effecting gene editing at a target site of interest within a target gene of interest in a target cell of interest, the method comprising contacting the target cell of interest with the composition of claim 24, optionally wherein the method comprises any one or more of the following features: (i) the gene editing comprises deletion of one or more nucleotides and/or insertion of one or more nucleotides at the target site of interest (e.g., at the cleavage site), optionally thereby deactivating or activating the target gene of interest; (ii) the target gene of interest is a disease-associated or disease-causing gene, optionally a SCD- associated gene or its regulatory (e.g., enhancer or promoter/repressor-binding) region, further optionally wherein the target site of interest is within BCLJ 1 A or the EER of BCLJ 1 A, HBG 1, the promoter region (e.g., BCLl lA-binding motif) of HBGJ, HBG2, and/or the promoter region (e.g., BCLl lA-binding motif) of HBG2; (iii) the contacting occurs in vitro, ex vivo, or in vivo; (iv) the contacting occurs in vivo and effected by administering the composition to a subject in need of the gene editing, optionally wherein the administering comprises injecting the composition into the bone marrow of the subject or intravenously; and/or (v) the method is for preventing or treating a disease, optionally SCD, optionally wherein the target gene of interest is not a human huntingtin gene, further optionally wherein the target gene of interest is not a huntingtin gene, and/or optionally wherein the target sequence is not within an about 120-nt, about 100-nt, about 90-nt, or about 80-nt sequence encompassing the junction between intron 12 and exon 13 of a human huntingtin gene and/or not within intron 12 and/or exon 13 of a human huntingtin gene.

Description:
COMPOSITIONS AND METHODS FOR PREVENTING, AMELIORATING, OR TREATING SICKLE CELL DISEASE AND COMPOSITIONS AND METHODS FOR DISRUPTING GENES AND GENE SEGMENTS RELATED APPLICATIONS [0001] The present disclosure claims priority to United States Provisional Application No. 63/381,989, filed on November 2, 2022 and United States Provisional Application No. 63/517,518, filed on August 3, 2023. The contents of both provisional applications are incorporated by reference in their entirety .. SEQUENCE LISTING The contents of the electronic sequence listing (29574150002013.xml; Size: 261,412 bytes; and Date of Creation: November 2, 2023) is herein incorporated by reference in its entirety. FIELD OF THE INVENTION [0002] The present disclosure relates to compositions and methods for preventing, ameliorating, and/or treating Sickle cell disease (SCD). The present disclosure also relates to compositions and methods for effecting gene editing and/or gene expression alteration in vivo using a lipid-based, transfection competent vesicle (TCV) in cells in the bone marrow or of bone marrow origin. BACKGROUND OF THE INVENTION [0003] SCD is a group of disorders characterized by a mutation(s) in and/or altered expression of HEB, the gene encoding beta-globin, which is hemoglobin (Hb)'s beta subunit (Kato et al., Nat Rev Dis Primers. 2018 Mar 15;4: 18010.). SCD patients carry at least one sickle Hb (HbS) allele of HBB, the ~S allele, containing an adenine-to-thymine substitution relative to the wildtype HBB gene and encodes the sickle Hb (HbS) variant of beta-globin, containing a glutamate-to-valine ("E-to-V" or "E6V") substitution. [0004] Hb expressed during a fetus stage (HbF) is a tetramer of two alpha-globin subunits and two gamma-globin subunits and does not involve beta-globin (Philipsen. Haematologica. 2014 Nov;99(1 l): 1647-9.). SCD patients typically have no gene alterations in the gene encoding alpha- or gamrna-globin and thus are not affected by the ~S allele during the fetus stage. In contrast, hemoglobin that starts increasing its expression post birth (HbA) is a tetramer formed by two alpha- globin subunits and two beta-globin subunits. HbA in SCD patients (HbS) thus contains the beta- globin HbS variant, and deoxygenated HbS can polymerize and HbS polymers can stiffen the erythrocyte, causing an anemic phenotype as HbS starts to dominate. Common complications include acute pain events, acute chest syndrome and stroke, and chronic complications including chronic kidney disease can damage all organs. (0005] Medications available for reduce the frequency of pain crisis, include hydroxyurea, L- glutamine oral powder, and crizanlizumab. General pain medications are also used to alleviate pain. A recently approved voxelotor, a HbS polymerization inhibitor, decreases sickling ofHbS and extends the half-life ofRBCs Hydroxycarbamide, blood transfusions, and hematopoietic stem cell transplantation can reduce the severity of the disease, but currently there is no sufficiently effective or feasible treatment or cure. SUMMARY OF THE INVENTION (0006] In one aspect, the present disclosure provides one or more guide RNAs (gRNAs) for Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-mediated gene editing and compositions containing. The gene editing preferably may be used to effect CRISPR-mediated gene editing of at least one Sickle cell disease (SCD)-associated gene or a variant thereof and/or a promoter or enhancer thereof in vivo in a subject in need thereof. The gRNA may comprise at least one CRISPR RNA (crRNA) sequence comprising a target-complementary sequence comprising at least 17 nucleic acids, optionally comprising 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleic acids. (0007] In some embodiments, the target-complementary sequence may comprise: (i) the polynucleotide sequence of SEQ ID NO: 71,277,279, 87,287, or 289; or (ii) a polynucleotide sequence comprising one or more (optionally one, two, three, four, or five) mutations (e.g., deletions at the 3'-end) relative to the polynucleotide sequence of SEQ ID NO: 71,277,279, 87,287, or 289, respectively. (0008] In some embodiments, the crRNA sequence may further comprises a repeat sequence optionally at the 5' -end of the target-complementary sequence, the repeat sequence comprising (i) an optional 5'-end sequence, optionally comprising UAAUU or AAUU, (ii) a first stem sequence, optionally comprising UCUAC, (iii) a loop sequence, optionally comprising UCUU, UAAGU, UGUU, UUUU, UAUU, UGUUU, UUCG, or UUU, (iv) a second stem sequence which is reverse complementary to the first stem sequence, optionally comprising GUAGA, and (v) an optional 3'-end sequence, optionally comprising U, optionally wherein the repeat sequence comprises any one of SEQ ID NOS: 201-206. (0009] In some embodiments, the target-complementary sequence may comprise: (i) the polynucleotide sequence of SEQ ID NO: 271,273,275,281,283,285, 85, 25, 45, 47, 49, 65, 67, 69, 75, or 77; or (ii) a polynucleotide sequence comprising one or more (optionally one, two, three, four, or five) mutations (e.g., deletions at the 5'-end) relative to the polynucleotide sequence of SEQ ID NO: 271,273,275,281,283,285, 85, 25, 45, 47, 49, 65, 67, 69, 75, or 77. In certain embodiments, the mutations may be at any nucleic acid position(s) other than the 4th to the 7th nucleic acid positions from the 3'-end of the polynucleotide sequence of SEQ ID NO: 271,273,275,281,283, 285, 85, 25, 45, 47, 49, 65, 67, 69, 75, or 77, respectively. (0010] In some embodiments, the gRNA may be a single guide RNA (sgRNA) comprising (i) a crRNA sequence comprising the target-complementary sequence and a crRNA backbone sequence and (ii) a trans-activating CRISPR RNA (tracrRNA) sequence in a single strand. In certain embodiments, the crRNA sequence and the tracrRNA sequence may be linked via a linker optionally comprising SEQ ID NO: 139. In certain embodiments, the gRNA may comprise the target- complementary sequence followed by a sgRNA backbone sequence of any of SEQ ID NOS: 141-144, In certain embodiments, the sgRNA backbone sequence may be followed by one or more uracils, further optionally 1-10 uracils. (0011] In some embodiments, the gRNA may be a dual guide RNA ( dgRNA) formed by hybridization between (i) a crRNA sequence comprising the target-complementary sequence and a crRNA backbone sequence and (ii) a tracrRNA. In certain embodiments, the crRNA backbone sequence and the tracrRNA may comprise SEQ ID NOS: 145 and 146, respectively, or SEQ ID NOS: 147 and 148, respectively. (0012] In some embodiments, the one or more gRNAs may be synthetic or recombinant. (0013] In some embodiments, the one or more gRNAs may be a synthetic sgRNA and may comprise at least one chemical modification. In certain embodiments, the at least one chemical modification may comprise (i) 2'-O-methylation optionally at first three and last three bases and/or (ii) one or more 3' phosphorothioate bonds, optionally between first three and last two bases. (0014] In some embodiments, the composition may comprise any one or more of the gRNAs described above. (0015] In another aspect, the present disclosure provides a polynucleotide or polynucleotides encoding any one or more of the isolated gRNAs described herein and compositions containing. (0016] In some embodiments, the composition may comprise any one or more of such polynucleotides. (0017] In another aspect, the present disclosure provides a vector comprising a polynucleotide or polynucleotides encoding any of the isolated gRNAs described herein and compositions containing. (0018] In some embodiments, the polynucleotide or polynucleotides may optionally be linked to one or more regulatory sequences. (0019] In some embodiments, the composition may comprise any one or more of such vectors. (0020] In another aspect, the present disclosure provides ribonucleoproteins (RNPs), comprising: (a) any one or more isolated gRNAs described herein complexed with (b) a Cas endonuclease. (0021] In some embodiments, the Cas endonuclease may be: (i) selected from the group consisting of Cas12a or Cpfl, Cas9, Cas3, Cas8a2, Cas8b, Cas8c, Casl0, Casl 1, Casl2, Casl2b, Cas12f, Cas12j, Cas13, Casl3a, Cas14, C2cl, C2c3, and C2c2. In some embodiments, the Cas endonuclease may be a class 2 Cas endonuclease, optionally a type II, type V, or type VI Cas nuclease. In some embodiments, the Cas endonuclease may be Cas12a of Acidaminococcus sp. (AsCas12a), Lachnospiraceae bacterium (LbCas12a), Francisella novicida (FnCas12a), Moraxella bovoculi (MbCas 12a), Coprococcus eutactus (CeCas 12a), Butyrivibrio jibrisolvens (BdCas 12a). In certain embodiments, the Cas endonuclease may be Cas12a or any engineered variants thereof, optionally comprising any one of SEQ ID NOS: 210-215, 220-221, 230,240,250, and 260. In some embodiments, the Cas endonuclease may be Cas9 of Streptococcus pyogenes (SpCas9), Staphylococcus aureus Cas9 (SaCas9), Streptococcus thermophilus (StCas9), Neisseria meningitidis (NmCas9), Francisella novicida (FnCas9), Campylobacter jejuni (CjCas9), Streptococcus canis (ScCas9), Staphylococcus auricularis (SauriCas9), or any engineered variants thereof, including SaCas9-HF, SpCas9-HF1, KKHSaCas9, eSpCas9, HypaCas9, Fold-Fused dCas9, xCas9, SpRY (variant of SpCas9), and SpG (variant of SpCas9). In some embodiments, the Cas endonuclease may be Cas9. In certain embodiments, the Cas9 may comprise any one of SEQ ID NOS: 150-161. [0022] In some embodiments, the RNP may optionally be formed by mixing at an approximately equimolar ratio (I) a solution comprising the one or more isolated gRNAs and (II) a solution comprising the Cas endonuclease. In certain embodiments, the pH of the solution comprising the one or more isolated gRNAs may optionally be about 6 to 8, about 6.5 to 7.5, optionally about 7. In certain embodiments, the pH of the solution comprising the Cas endonuclease may be about 6 to 8, about 6.5 to 7.5, optionally about 7. In certain embodiments, the mixing may be for about 5 minutes. [0023] In one aspect, the present disclosure provides an isolated oligodeoxynucleotide (ODN), which may comprise a nucleic acid sequence having at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to SEQ ID NO: 190 or a reverse complement thereof. In certain embodiments, the ODN may be about 30-160 nt, 50-120 nt, about 60-100 nt, about 70-90 nt, about 75-85 nt, about 80 nt or about 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 81, 82, 83, 84, or 85 nt in length. In certain embodiments, the ODN may be a single-stranded oligo DNA nucleotide molecule (ssODN) or a double-stranded oligo DNA nucleotide molecule (dsODN). [0024] In certain embodiments, when the ODN is to be used in Cas nuclease-mediated gene editing (e.g., intended to disrupt a gene or a gene segment), the ssODN may not be complementary to a sequence flanking the target cut site by the Cas nuclease (e.g., the sequence corresponding to about 20-60 nt 5' of the cut site and about 20-60 nt 3' of the cut site). In particular embodiments, the complementarity to a sequence flanking the target cut site by Cas nuclease is less than about 90%, less than about 85%, less than about 80%, less than about 80%, less than about 75, less than about 70%, less than about 65%, less than about 60%, less than about 55%, less than about 50%, less than about 45%, less than about 40%, less than about 35%, less than about 30%, less than about 25%, less than about 20%, less than about 15%, or less than about 10%. Without wishing to be bound by theory, use of such an ODN in gene editing (e.g., intended to disrupt a gene or a gene segment) not complementary or having low complementarity to the sequence flanking the target cut site by the Cas nuclease may enhance the efficiency of the intended gene editing (e.g., enhanced nt deletion). [0025] In one aspect, the present disclosure provides a pharmaceutical composition for effecting gene editing and/or gene expression alteration. In some embodiments, the gene editing and/or gene expression alteration may be effected in vivo. [0026] In some embodiments, the pharmaceutical composition may comprise at least one cargo encapsulated in a canier. In certain embodiments, the canier may be a lipid-based, transfection competent vesicle (TCV). In some embodiments, the at least one cargo may be capable of effecting gene editing of at least one Sickle cell disease (SCD)-associated gene and/or a promoter or enhancer thereof in vivo in a subject in need thereof. In some embodiments, the at least one cargo may be capable of altering the expression, function, and/or effect of at least one SCD-associated gene in vivo in a subject in need thereof. In some embodiments, the at least one cargo may be capable of effecting gene editing of at least one Sickle cell disease (SCD)-associated gene and/or a promoter or enhancer thereof and altering the expression, function, and/or effect of at least one SCD-associated gene in vivo in a subject in need thereof. In some embodiments, the subject may have or may have a risk of developing SCD, which is optionally sickle cell anemia (SCA), Sickle cell-hemoglobin C (HbSC), or HbS ~-thalassaemia. [0027] In some embodiments, the pharmaceutical composition may be for: (I) direct injection into the bone marrow of the subject; and/or (II) intravenous injection into the subject who may optionally be administered at least one agent that promotes stem cell mobilization. [0028] In some embodiments, the carrier may be a lipid-based TCV, and the TCV in the composition may comprise at least one ionizable cationic lipid. In some embodiments, the at least one ionizable cationic lipid may comprise, essentially consist of, or consist of a lipid selected from the group consisting ofN,N-dimethyl-2,3-dioleyloxy)propylamine (DODMA), 1,2-dioleoyl-3- dimethylammonium propane ("DODAP"), 1,2-Dilinoleoyl-3-dimethylaminopropane (DLinDAP), N,N-dimethyl-2,2-di-(9Z, l 2Z)-9, 12-octadecadien-1-yl- l ,3-dioxolane-4-ethanamine (KC2), ( 6Z,9Z,28Z,31 Z)-heptatriaconta-6,9 ,28,31-tetraen-19-yl 4-( dimethylamino )butanoate (MC3), 6-((2- hexyldecanoyl)ox y )-N-( 6-( (2-hexyldecanoyl)oxy )hexyl)-N-( 4-hydroxybutyl)hexan-1-aminium ( ALC- 0315), 8-[(2-hydroxyethyl)[6-oxo-6-(undecyloxy)hexyl]amino]-octanoi c acid, 1-octylnonyl ester (SM-102), N,N-dioleyl-N,N-dimethylammonium chloride (DODAC), N,N-distearyl-N,N- dimethylammonium bromide (DDAB), N-(1-(2,3-dioleoyloxy)propyl)-N,N,N-trimethylammonium chloride (DOT AP), N-(1-(2,3-dioleyloxyl)propyl)-N,N,N-trimethylammonium chloride (DOTMA), 1,2-DiLinoleyloxy-N ,N-dimethylaminopropane (DLinDMA), 1,2-Dilinolenyloxy-N,N- dimethylaminopropane (DLenDMA), 1,2-Dilinoleylcarbamoyloxy-3-dimethylaminopropane (DLin- C-DAP), 1,2-Dilinoleyoxy-3-(dimethylamino )acetoxypropane (DLin-DAC), 1,2-Dilinoleyoxy-3- morpholinopropane (DLin-MA), 1,2-Dilinoleylthio-3-dimethylaminopropane (DLin-S-DMA), l- Linoleoyl-2-linoleyloxy-3-dimethy laminopropane (DLin-2- DMAP), 1,2-Dilinoley loxy-3- trimethylaminopropane chloride salt (DLin-TMA.Cl), 1,2-Dilinoleoyl-3-trimethylaminopropane chloride salt (DLin-T AR.Cl), 1,2-Dilinoleyloxy-3-(N-methylpiperazino )propane (DLin-MPZ), or 3- (N,N-Dilinoleylamino)-1,2-propanediol (DLinAP), 3-(N,N-Dioleylamino)-1,2-propanedio (DOAP), 1,2-Dilinoleyloxo-3-(2-N ,N-dimethylamino )ethoxypropane (DLin-EG-DMA), 1,2-Dilinolenyloxy- N ,N-dimethy laminopropane (D Lin-K-DMA), 2,2-Dilinoleyl-4-dimethylaminomethyl-[ 1,3 ]-dioxolane (DLin-K-DMA) or analogs thereof, (3aR,5s,6aS)-N,N-dimethyl-2,2-di((9Z, 12Z)-octadeca-9, 12- dienyl)tetrahydro-3 aH-cyclopenta[ d][l ,3]dioxol-5-amine (ALNY-100), N-(2,3-dioleyloxyl)propyl- N,N-N-triethylammonium chloride ("DOTMA"); 1,2-Dioleyloxy-3-trimethylaminopropane chloride salt ("DOT AP.Cl"); 3~-(N-(N' ,N' -dimethylaminoethane)-carbamoyl)cholesterol ("DC-Chol"), N- (1-(2,3-dioleyloxyl)propyl)-N-2-(sperminecarboxamido)ethyl)- N,N-dimethyl-ammonium trifluoracetate ("DOSPA"), dioctadecylamidoglycyl carboxyspermine ("DOGS"), and N-(l ,2- dimyristyloxyprop-3-yl)-N,N-dimethyl-N-hydroxyethyl ammonium bromide ("DMRIE"), and any combinations thereof; [0029] In some embodiments, the TCV may further comprise at least one helper lipid. In some embodiments, the helper lipid may comprise, essentially consist of, or consist of a lipid selected from the group consisting of dioleoylphosphatidylethanolamine (DOPE), distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), palmitoyloleoylphosphatidylcholine (POPC), palmitoyloleoyl-phosphatidylethanolamine (POPE), dioleoy 1-phosphatidy lethanolamine 4-(N-maleimidomethy l )-cyclohexane-1-carbox y late (DO PE-mal), dipalmitoyl phosphatidyl ethanolamine (DPPE), dimyristoylphosphoethanolamine (DMPE), distearoyl-phosphatidyl-ethanolamine (DSPE), 16-O-monomethyl PE, 16-O-dimethyl PE, 18-1-trans PE, l-stearoyl-2-oleoyl-phosphatidyethanolamine (SOPE), and any combinations thereof; [0030] In some embodiments, the TCV may further comprise at least one phospholipid. In some embodiments, the phospholipid may comprise, essentially consist of, or consist of a group selected from the group consisting of distearoylphosphatidylcholine (DSPC), dioleoyl phosphatidylethanolamine (DOPE), dipalmitoylphosphatidylcholine (DPPC), phosphocholine (DOPC), dimyristoylphosphatidylcholine (DMPC), phosphatidylcholine (PLPC), 1,2-distearoyl-sn- glycero-3-phosphocholine (DAPC), phosphatidylethanolamine (PE), egg phosphatidylcholine (EPC), dilauryloylphosphatidylcholine (DLPC), dimyristoylphosphatidylcholine (DMPC), 1-myristoyl-2- palmitoyl phosphatidylcholine (MPPC), l-palmitoyl-2-myristoyl phosphatidylcholine (PMPC), l- palmitoyl-2-stearoyl phosphatidylcholine (PSPC), 1,2-diarachidoyl-sn-glycero-3-phosphocholine (DBPC), l-stearoyl-2-palmitoyl phosphatidylcholine (SPPC), l ,2-dieicosenoyl-sn-glycero-3- phosphocholine (DEPC), palmitoyloleoyl phosphatidylcholine (POPC), lysophosphatidyl choline, dilinoleoylphosphatidylcholine distearoylphophatidylethanolamine (DSPE), dimyristoyl phosphatidylethanolamine (DMPE), dipalmitoyl phosphatidylethanolamine (DPPE), palmitoyloleoyl phosphatidylethanolamine (POPE), lysophosphatidylethanolamine, and any combinations thereof; [0031] In some embodiments, the TCV may further comprise at least one cholesterol or cholesterol derivative. In some embodiments, the cholesterol or cholesterol derivative may comprise, essentially consist of, or consist of a cholesterol or cholesterol derivative selected from the group consisting of cholesterol, N,N-dimethyl-N-ethylcarboxamidocholesterol (DC-Chol), 1,4-bis(3-N-oleylamino- propyl)piperazine, imidazole cholesterol ester (ICE), and any combinations thereof. [0032] In some embodiments, the TCV may further comprise at least one PEG-lipid. In some embodiments, the PEG-lipid may comprise, essentially consist of, or consist of a PEG-lipid selected from the group consisting of PEG-myristoyl diglyceride (PEG-DMG) (e.g., 1,2-dimyristoyl-rac- glycero-3-methoxypolyethylene glycol-2000 (Avanti® Polar Lipids (Birmingham, AL)), which is a mixture of 1,2-DMG PEG2000 and 1,3-DMG PEG2000 (e.g., in about 97:3 ratio)), PEG- phosphatidylethanolamine and phosphatidic acid, PEG-ceramide conjugates (e.g., PEG-CerC14 or PEG-CerC20), PEG-modified dialkylamines, PEG-modified 1,2-diacyloxypropan-3-amines, and any combinations thereof. [0033] In some embodiment, the TCV comprising the at least one ionizable cationic lipid as described above may further comprises one or more of: the at least one helper lipid as described above; the at least one phospholipid as described above; the at least one cholesterol or cholesterol derivative as described above; and/or the at least one PEG-lipid as described above. [0034] In some embodiments, the TCV is substantially, essentially, or entirely free of organic solvents and/or detergents. [0035] In some embodiments, the TCV may be formed by: (a) generating a first solution by dissolving all components of the TCV, optionally at about 20-35 mM, in ethanol; (b) providing a second solution, which is aqueous and contains sodium acetate and/or sodium citrate, optionally at about 25 mM, optionally wherein the pH of the solution is about 4; (c) combining the first and second solutions by gentle mixing ( optionally repeated manual reciprocation of the TCV-generating fluid in a pipette), micromixing optionally using a staggered herringbone micromixer (SHM) or T-junction or Y-junction mixing, or extrusion; and ( d) removing ethanol, optionally by dialysis or evaporation. [0036] In some embodiments, the size of the TCV before encapsulation of the at least one cargo may be in a range of about 9 nm to about 80 nm at pH of about 4. [0037] In some embodiments, in any of the pharmaceutical compositions described above, the amount of the at least one ionizable cationic lipid relative to the total lipid components of the TCV may be about 20 mol% to about 30 mo!%, about 20, 21, 22, 23, 24, 25 26, 27, 28, 29 or 30 +/- 0.5 mo!%, about 10 mo!% to about 70 mo!%, about 10 mo!% to about 60 mo!%, about 10 mo!% to about 50 mo!%, about 10 mo!% to about 40 mo!%, about 10 mo!% to about 30 mo!%, about 15 mo!% to about 25 mo!%, about 18 mo!% to about 22 mo!%, about 19 mo!% to about 21 mo!%, about 19.5 mo!% to about 20.5 mo!%, about 19.8 mo!% to about 20.2 mo!%, or about 20 mo!%. In particular embodiments, the amount of the at least one ionizable cationic lipid relative to the total components of the TCV may be about 20 mo!%. [0038] In some embodiments, in any of the pharmaceutical compositions described above, the amount of the at least one ionizable cationic lipid relative to the total lipid components of the TCV may be about 10 mo!% to about 70 mo!%, about 20 mo!% to about 70 mo!%, about 30 mo!% to about 70 mo!%, about 40 mo!% to about 70 mo!%, about 40 mo!% to about 60 mo!%, about 45 mo!% to about 55 mo!%, about 48 mo!% to about 52 mo!%, about 49 mo!% to about 51 mo!%, about 49.5 mo!% to about 50.5 mo!%, about 49.8 mo!% to about 50.2 mo!%, or about 50 mo!%. In particular embodiments, the amount of the at least one ionizable cationic lipid relative to the total components of the TCV may be about 50 mo!%. [0039] In some embodiments, in any of the pharmaceutical compositions described above, the amount of the at least one helper lipid relative to the total lipid components of the TCV may be about 10 mo!% to about 60 mo!%, about IO mo!% to about 50 mo!%, about 10 mo!% to about 40 mo!%, about 20 mo!% to about 40 mo!%, about 25 mo!% to about 35 mo!%, about 28 mo!% to about 32 mo!%, about 29 mo!% to about 31 mo!%, about 28, 29, 30, or 31 +/- 0.5 mo!%, about 29.5 mo!% to about 30.5 mo!%, about 29.8 mo!% to about 30.2 mo!%, or about 30 mo!%. In particular embodiments, the amount of the at least one helper lipid relative to the total components of the TCV may be about 30 mo!%. [0040] In some embodiments, in any of the pharmaceutical compositions described above, the amount of the at least one phospholipid relative to the total lipid components of the TCV may be about 5 mo!% to about 65 mo!%, about 5 mo!% to about 55 mo!%, about 5 mo!% to about 45 mo!%, about 5 mo!% to about 35 mo!%, about 5 mo!% to about 25 mo!%, about 5 mo!% to about 15 mo!%, about 8 mo!% to about 12 mo!%, about 8, 9, 10, 11 or 12 mo!%+/- 0.5 mo!%, about 9 mo!% to about 11 mo!%, about 9.5 mo!% to about 10.5 mo!%, about 9.8 mo!% to about 10.2 mo!%, or about 10 mo!%. In particular embodiments, the amount of the at least one phospholipid relative to the total components of the TCV may be about 10 mo!%. [0041] In some embodiments, in any of the pharmaceutical compositions described above, the amount of the at least one cholesterol or cholesterol derivative relative to the total lipid components of the TCV may be about 20 mo!% to about 60 mo!%, about 25 mo!% to about 55 mo!%, about 30 mo!% to about 50 mo!%, about 35 mo!% to about 45 mo!%, about 38 mo!% to about 42 mo!%, about 35, 36, 37, 38, 39, 40, 41, 42, 43, 44 or 45 mo!%+/- 0.5 mo!%, about 39 mo!% to about 41 mo!%, about 39.5 mo!% to about 40.5 mo!%, about 39.8 mo!% to about 40.2 mo!%, or about 40 mo!%, or about 39%. In particular embodiments, the amount of the at least one cholesterol or cholesterol derivative relative to the total components of the TCV may be about 40 mo!% or about 39%. [0042] In some embodiments, in any of the pharmaceutical compositions described above, the amount of the at least one PEG or PEG-lipid relative to the total lipid components of the TCV may be about 0.1 mo!% to about 5 mo!%, 0.1 mo!% to about 4 mo!%, 0.1 mo!% to about 3 mo!%, 0.1 mo!% to about 2 mol%, about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9 or 2.0 mol%, 0.5 mol% to about 1.5 mol%, 0.8 mo!% to about 1.2 mol%, 0.9 mo!% to about 1.1 mol%, or about 1 mol%. In particular embodiments, the amount of the at least one PEG-lipid relative to the total components of the TCV may be about 1 mol%. (0043] In some embodiments, in any of the pharmaceutical compositions described above, the TCV may comprise, essentially consist of, or consist of: (i) at least one ionizable cationic lipid, which is optionally DODMA, DLinDMA, DLin-KC2-DMA, DLin-MC3-DMA, SM-102, or ALC-0315; (ii) at least one helper lipid, which is optionally DOPE; (iii) at least one phospholipid, which is optionally DSP<;::; and (iv) at least one cholesterol or cholesterol derivative. In particular embodiments, the amounts of the at least one ionizable cationic lipid, the at least one helper lipid, the at least one phospholipid, and the at least one cholesterol or cholesterol derivative, relative to the total components of the TCV, may be about 20 mo!%, about 30 mo!%, about 10 mo!%, and about 40 mol%, respectively. In a particular embodiment, the TCV may comprise, essentially consist of, or consist of, DODMA (and/or another ionizable cationic lipid DLinDMA, DLin-KC2-DMA, DLin- MC3-DMA, SM-102, or ALC-0315), DOPE, DSPC, cholesterol, with amounts (relative to the total components of the TCV) of about 20 mo!%, about 30 mo!%, about IO mol%, and about 40 mol%, respectively. [0044] In some embodiments, in any of the pharmaceutical compositions described above, the TCV may comprise, essentially consist of, or consist of: (i) at least one ionizable cationic lipid, which is optionally DODMA, DLinDMA, DLin-KC2-DMA, DLin-MC3-DMA, SM-102, and/or ALC-0315; (ii) at least one helper lipid, which is optionally DOPE; (iii) at least one phospholipid, which is optionally DSPC; (iv) at least one cholesterol or cholesterol derivative; and (v) at least one PEG or PEG-lipid, which is optionally PEG-DMG. In particular embodiments, the amounts of the at least one ionizable cationic lipid, the at least one helper lipid, the at least one phospholipid, the at least one cholesterol or cholesterol derivative, and the at least one PEG or PEG-lipid, relative to the total components of the TCV, may be about 20 mol%, about 30 mo!%, about 10 mo!%, about 39 mo!%, and about 1 mo!%, respectively. [0045] In some embodiments, the TCV may be substantially, essentially, or entirely free of ethanol, methanol, isopropanol, tetrahydrofuran (THF), dimethyl sulfoxide (DMSO), dimethyl formamide (DMF), and acetonitrile (ACN). In particular embodiments, optionally wherein the TCV is substantially, essentially, or entirely free of organic solvents and detergents. In particular embodiments, the TCV may be substantially, essentially, or entirely free of destabilizing agents. In particular embodiments, the TCV may be stable for prolonged periods of time at about 1 to about 40 °C, about 5 to about 35 °C, about 10 to about 30 °C, or about 15 to about 25 °C. (0046] In some embodiments, the TCV or the pharmaceutical composition may further comprise and/or be stored in the presence of at least one cryoprotectant. In certain embodiments, the cryoprotectant may comprise a sugar-based molecule, which is optionally sucrose, trehalose, or a combination thereof. In particular embodiments, the concentration of the cryoprotectant may be about 1 % to about 40 %, about 3% to about 30%, about 5% to about 30%, about 10% to about 20%, or about 15%. In a particular embodiment, the concentration of the cryoprotectant may be about 10% to about 20%. In certain embodiments, the TCV may be stable at a freezing temperature, optionally at about -20°C or about -80°C. In certain embodiments, the TCV may be stable at a freezing temperature for at least about one week, at least about two weeks, at least about three weeks, at least about a month, at least about two months, at least about four months, at least about five months, at least about six months, at least about nine months, at least about a year, or at least about two years, or longer. In certain embodiments, the TCV may be stable at a freezing temperature for about one week to about two year, about two weeks to about a year, about three weeks to about nine months, about one to about six months, about one to five months, about one to four months, about one to three months, or about two months. In a particular embodiment, the TCV or the pharmaceutical composition may further comprise and/or be stored in the presence of about 10% to about 20% sucrose and may be stable at about -80°C for at least about two months. [0047] In some embodiments, in any of the pharmaceutical composition described above, the at least one SCD-associated gene may comprise one or more genes selected from the group consisting of BCLJ JA, HBGJ, HBG2, HBB (the sickle cell hemoglobin (HbS) variant, also known as the ~S allele), KLFJ, SOX6, GATAJ, NF-E4 (or NFE4), COUP-TF, NR2Cl (also known as TR2), NR2C2 (also known as TR4), genes encoding members of the MBD2 protein complex, IKZFJ (also known as Ikaros), genes encoding other members of PYR complex (CHD4, HDAC2, RBBP7, SMARCBJ, SMARCCJ, BRGJ, and variants of any of the foregoing and genes that directly or indirectly modulate the expression variants of any of the foregoing. [0048] In particular embodiments, the at least one SCD-associated gene may be BCLJ 1 A, optionally encoding the amino acid sequence of SEQ ID NO: 6, and/or a promoter or enhancer region of BCLJ JA, preferably the erythroid-enhancer region (EER) of BCLJ JA. [0049] In particular embodiments, the at least one SCD-associated gene may be HBG 1, optionally encoding the amino acid sequence of SEQ ID NO: 8, and/or a promoter or enhancer region of HBG l .In particular embodiments, the at least one SCD-associated gene may be HBG2, optionally encoding the amino acid sequence of SEQ ID NO: 9, and/or a promoter or enhancer region of HBG2. [0050] In particular embodiments, the at least one SCD-associated gene may be HEB (such as the sickle cell hemoglobin (HbS) variant of HBB, also known as the ~S allele or the hemoglobin C (HbC) variant of HBB), which optionally comprises the polynucleotide sequence of SEQ ID NO: 11, 21, or 31 and/or encoding the amino acid sequence of SEQ ID NO: 1, 2, or 3, and/or a promoter or enhancer region of HBB. [0051] In particular embodiments, the at least one SCD-associated gene may be KLFJ, optionally encoding the amino acid sequence of SEQ ID NO: 7, and/or a promoter or enhancer region of KLF 1. [0052] In some embodiments, in any of the pharmaceutical composition described above, the gene editing may be mediated by a protease, nuclease, endonuclease, meganuclease, zinc finger nuclease (ZFN), transcription activator-like nuclease (T ALEN), or clustered regularly interspaced short palindromic repeats (CRISPR)-associated (Cas) nuclease, optionally resulting in at least one nucleic acid insertion, deletion, or replacement (e.g., resulting in a nonsense, missense, or silent mutation) in the at least one SCD-associated gene. [0053] In some embodiments, in any of the pharmaceutical composition described above, the at least one cargo capable of effecting gene editing may comprise, essentially consist of, or consist of: (a) a Cas nuclease, a RNA encoding a Cas nuclease, or a nucleic acid such as a DNA or RNA encoding a Cas nuclease; and (b) a guide RNA (gRNA) comprising a target-complementary sequence which is complementary to a target sequence within the at least one SCD-associated gene and/or a promoter or enhancer thereof, or a nucleic acid encoding said gRNA. [0054] In some embodiments, the Cas nuclease may be selected from the group consisting ofCas12a or Cpfl, Cas 9, Cas3, Cas8a2, Cas8b, Cas8c, Casl0, Csxl 1, Cas12, Cas12b, Cas12f, Cas12j, Cas13, Cas 13a, Cas 14, C2c 1, C2c3, and C2c2. In some embodiments, the Cas nuclease may be a class 2 Cas nuclease, optionally a type V or type VI Cas nuclease. In particular embodiments, the Cas nuclease may be Cas12a of Acidaminococcus sp. (AsCas12a), Lachnospiraceae bacterium (LbCas12a), Francisella novicida (FnCas12a), Moraxella bovoculi (MbCas12a), Coprococcus eutactus (CeCas12a), Butyrivibrio fibrisolvens (BfCas12a). In particular embodiments, the Cas nuclease may be Cas12a or any engineered variants thereof, optionally comprising any one of SEQ ID NOS: 210- 215, 220-221, 230,240,250, and 260. In particular embodiments, the Cas nuclease may be Cas 9. In particular embodiments, Cas9 may be Cas9 of Streptococcus pyogenes (SpCas9), Staphylococcus aureus Cas9 (SaCas9), Streptococcus thermophilus (StCas9), Neisseria meningitidis (NmCas9), Francisella novicida (FnCas9), Campylobacter jejuni (CjCas9), Streptococcus canis (ScCas9), Staphylococcus auricularis (SauriCas9), or any engineered variants thereof, including SaCas9-HF, SpCas9-HF1, KKHSaCas9, eSpCas9, HypaCas9, Fokl-Fused dCas9, xCas9, SpRY (variant of SpCas9), and SpG (variant of SpCas9). In particular embodiments, the Cas9 may comprise any one of SEQ ID NOS: 150-161. [0055] In some embodiments, the gRNA may comprise a repeat sequence optionally at the 5'-end of the target-complementary sequence, the repeat sequence comprising (i) an optional 5'-end sequence, optionally comprising UAAUU or AAUU, (ii) a first stem sequence, optionally comprising UCUAC, (iii) a loop sequence, optionally comprising UCUU, UAAGU, UGUU, UUUU, UAUU, UGUUU, UUCG, or UUU, (iv) a second stem sequence which is reverse complementary to the first stem sequence, optionally comprising GUAGA, and (v) an optional 3' -end sequence, optionally comprising U. In certain embodiments, optionally wherein the repeat sequence may comprise any one of SEQ ID NOS: 201-206. In certain embodiments, such a gRNA may be used with Cas12a. (0056) In some embodiments, the gRNA may be a single guide RNA (sgRNA) comprising (1) a crRNA sequence comprising the target-complementary sequence and a crRNA backbone sequence and (2) a trans-activating CRISPR RNA (tracrRNA) sequence in a single strand. In certain embodiments, the crRNA sequence and the tracrRNA sequence may be linked via a linker optionally comprising SEQ ID NO: 139. In certain embodiments, the gRNA may comprise the target- complementary sequence followed by a sgRNA backbone sequence of any of SEQ ID NOS: 141-144, optionally wherein the sgRNA backbone sequence may be followed by one or more uracils, further optionally 1-10 uracils. (0057) In some embodiments, the gRNA may be a dual guide RNA ( dgRNA) formed by hybridization between (1) a crRNA sequence comprising the target-complementary sequence and a crRNA backbone sequence and (2) a tracrRNA. In certain embodiments, the crRNA backbone sequence and the tracrRNA may comprise SEQ ID NOS: 145 and 146, respectively, or SEQ ID NOS: 147 and 148, respectively. (0058) In some embodiments, in any of the pharmaceutical composition described above, the at least one cargo may comprise, essentially consist of, or consist of a ribonucleoprotein (RNP), which is a complex of the gRNA and the Cas nuclease. In certain embodiments, the RNP may be any of the RNPs described above or herein. (0059) In some embodiments, the RNP may be formed by mixing Cas9 and gRNA at an approximately equimolar ratio. In some embodiments, the mixing may be for about 5 minutes, (0060) In some embodiments, in any of the pharmaceutical composition described above, the pharmaceutical composition or the at least one cargo may further comprise a DNA repair template, which optionally may be single stranded or double stranded. (0061) In some embodiments, the at least one cargo (which may comprise the RNP or the RNP and the DNA repair template) encapsulated in the TCV may be obtained by: (i) providing an aqueous solution comprising the TCV, optionally wherein the pH of the aqueous solution is about 3 to about 8, further optionally about 4 to about 7.5; and (ii) mixing the at least one cargo with the aqueous solution, wherein mixing is effected under conditions suitable for the at least one cargo to be encapsulate within the TCV. In some embodiments, the mixing may comprise gentle mixing (optionally repeated manual reciprocation of the TCV-generating fluid in a pipette), micromixing optionally using a staggered herringbone micromixer (SHM) or T-junction or Y-junction mixing, or extrusion. In some embodiments, the mixing time may be about 0.1 second to about 20 minutes. (0062) In some embodiments, the aqueous solution of step (i) may be substantially, essentially, or entirely free of ethanol, methanol, isopropanol, tetrahydrofuran (THF), dimethyl sulfoxide (DMSO), dimethyl fonnamide (DMF), and acetonitrile (ACN). In some embodiments, the aqueous solution of step (i) may be substantially, essentially, or entirely free of organic solvents and detergents, further optionally substantially, essentially, or entirely free of destabilizing agents. [0063] In some embodiments, the mixing of step (ii) may be performed substantially, essentially, or entirely free of ethanol, methanol, isopropanol, tetrahydrofuran (THF), dimethyl sulfoxide (DMSO), dimethyl formamide (DMF), and acetonitrile (ACN). In some embodiments, the mixing of step (ii) may be performed substantially, essentially, or entirely free of organic solvents and detergents. In particular embodiments, the mixing of step (ii) may be performed substantially, essentially, or entirely free of destabilizing agents. In a particular embodiment, the final ethanol concentration after encapsulation may be 5% (v/v) or below. In a yet particular embodiment, the final ethanol concentration after encapsulation may be 0.5% (v/v) or below. [0064] In some embodiments, the size of the TCV after encapsulation of the at least one cargo may be in a range of about 80 nm to about 1000 nm and/or in arrange of about 100 nm to about 250 nm, at pH of about 7.5. [0065] In some embodiments, the at least one cargo (which comprises the RNP or the RNP and the DNA repair template) encapsulated in the TCV may be comprised in a matrix vesicle, which is optionally for gradual release of the TCV. [0066] In some embodiments, the at least one SCD-associated gene may comprise or consist of BCLJ IA or a variant thereof and/or the erythroid-enhancer region (EER) of BCLJ IA and/or a promoter or enhancer region of BCLJ IA. In certain embodiments, the gRNA may direct the Cas protein to and hybridize to a target sequence, which may be located between nucleotide positions 60450520 to 60553654 of Chromosome 2 (according to Gene Assembly GRCh38.pl3, positive or negative strand) and/or a promoter or enhancer region of BCLJ IA, optionally within or overlapping with the polynucleotide sequence of SEQ ID NO: 4 or 5. [0067] In some embodiments, the at least one SCD-associated gene may comprise or consist of HBGI or a variant thereof and/or a promoter or enhancer region of HBGI, optionally within or overlapping with the polynucleotide sequence of SEQ ID NO: 95 or 97, preferably within or overlapping with the BCLI IA-binding site thereof. In some embodiments, the gRNA may direct the Cas protein to and hybridize to a target sequence, which may be located between nucleotide positions 5248269 to 5249857 of Chromosome 11 (according to Gene Assembly GRCh38.p14, positive or negative strand), preferably in the BCLl lA-binding site thereof. [0068] In some embodiments, the at least one SCD-associated gene may comprise or consist of HBG2 or a variant thereof and/or a promoter or enhancer region of HBG2, optionally within or overlapping with the polynucleotide sequence of SEQ ID NO: 95 or 97, preferably within or overlapping with the BCLI lA-binding site thereof. In some embodiments, the gRNA may direct the Cas protein to and hybridize to a target sequence, which may be located between nucleotide positions 5253188 to 5254781 of Chromosome 11 (according to Gene Assembly GRCh38.p14, positive or negative strand), preferably in the BCLl lA-binding site thereof. [0069] In some embodiments, in the pharmaceutical composition according to the present disclosure, the at least one SCD-associated gene may comprise or consist of HEB or a variant thereof (such as the sickle cell hemoglobin (HbS) variant of HEB, also known as the ~S allele, or the hemoglobin C (HbC) variant of HEB) and/or a promoter or enhancer region of HEB. In particular embodiments, the gRNA may direct the Cas protein to and hybridize to a target sequence, which may be located between nucleotide positions 5225464 to 5227071 of Chromosome 11 (according to Gene Assembly GRCh38.pl 3, positive or negative strand) and which may optionally be within the polynucleotide sequence of SEQ ID NO: 11, 21, or 31 or the sequence complementary thereto. In particular embodiments, the gRNA may direct the Cas protein to and hybridize to a target sequence, which may be located within or overlapping with exon 1 of HEB. [0070] In some embodiments, the pharmaceutical composition or the at least one cargo may further comprise a DNA repair template which may allow for a knock-in of or correction to the wildtype HEB gene sequence (SEQ ID NO: 11) or the polynucleotide sequence encoding the wildtype beta-globin amino acid sequence (SEQ ID NO: 1). [0071] In some embodiments, the at least one SCD-associated gene may comprise or consist of KLFJ or a variant thereof and/or a promoter or enhancer region of KLF 1. In some embodiments, the gRNA may direct the Cas protein to and hybridize to a target sequence, which may be located between nucleotide positions 12884422 to 12887201 of Chromosome 19 (according to Gene Assembly GRCh38.pl 3, positive or negative strand) and/or a promoter or enhancer region of KLFJ. [0072] In some embodiments, the pharmaceutical composition or the at least one cargo may further comprise an isolated ODN. In certain embodiments, the ODN may comprise a ssODN comprising a nucleic acid sequence having less than 90%, less than 85%, less than 80%, less than 75%, less than 70%, less than 65%, less than 60%, less than 55%, less than 50%, less than 45%, less than 40%, less than 35%, less than 30%, less than 30%, less than 25%, less than 20%, less than 15%, or less than 10% complementarity to the sequence of a segment within the SCD-associated gene , wherein the segment comprises the target sequence and/or the sequence complementary thereto and is about 20- 300 nt, about 25-200 nt, about 30-160 nt, about 40-140 nt, about 50-120 nt, about 60-100 nt, about 70- 90 nt, about 75-85 nt, about 20 nt, about 25 nt, about 30 nt, about 35 nt, about 40 nt, about 45 nt, about 50 nt, about 55 nt, about 60 nt, about 65 nt, about 70 nt, about 75 nt, about 80 nt, about 85 nt, about 90 nt, about 95 nt, about 100 nt, about 105 nt, about 120 nt, about 125 nt, about 130 nt, about 135 nt, about 140 nt, about 145 nt, about 150 nt, about 155 nt, about 160 nt, about 175 nt, about 180 nt, about 185 nt, about 190 nt, about 195 nt, about 200 nt, about 205 nt, about 220 nt, about 225 nt, about 230 nt, about 235 nt, about 240 nt, about 245 nt, about 250 nt, about 255 nt, about 260 nt, about 275 nt, about 280 nt, about 285 nt, about 290 nt, about 295 nt, or about 300 nt in length. In certain embodiments, the ODN may comprise a dsODN comprising a first strand comprising such a ssODN sequence and a second strand complementary to the first strand. In certain embodiments, the ODN may be about 30-160 nt, 50-120 nt, about 60-100 nt, about 70-90 nt, about 75-85 nt, about 80 nt in length. In certain embodiments, the nucleic acid sequence of the ODN may have at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to SEQ ID NO: 190 or a reverse complement thereof. [0073] In some embodiments, in any of the pharmaceutical composition for effecting gene editing to BCLI I A, the target sequence may or may comprise SEQ ID NO: 70 or at least the first 17, 18, or 19 nucleotides from the 3' end of SEQ ID NO: 70, and/or the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 71 , or at least the first 17, 18, or 19 nucleotides thereof from the 5' end of SEQ ID NO: 71. In some embodiments, the target sequence may or may comprise SEQ ID NO: 276 or at least the first 17, 18, or 19 nucleotides from the 3' end of SEQ ID NO: 276, and/or the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 277, or at least the first 17, 18, or 19 nucleotides thereof from the 5' end of SEQ ID NO: 277. In some embodiments, the target sequence may or may comprise SEQ ID NO: 278 or at least the first 17, 18, or 19 nucleotides from the 3' end of SEQ ID NO: 278, and/or the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 279, or at least the first 17, 18, or 19 nucleotides thereof from the 5' end of SEQ ID NO: 279. In some embodiments, the target sequence may be or comprise SEQ ID NO: 270 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 270, and/or the target-complementary sequence may comprise the polynucleotide sequence of SEQ ID NO: 271, or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 271. In some embodiments, the target sequence may be or comprise SEQ ID NO: 272 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 272, and/or the target- complementary sequence may comprise the polynucleotide sequence of SEQ ID NO: 273, or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 273. In some embodiments, the target sequence may be or comprise SEQ ID NO: 274 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 274, and/or the target-complementary may comprise the polynucleotide sequence of SEQ ID NO: 275, or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 275. In some embodiments, the target sequence may be or comprise SEQ ID NO: 64 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 64, and/or the target-complementary sequence may comprise the polynucleotide sequence of SEQ ID NO: 65, or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 65. In some embodiments, the target sequence may be or comprise SEQ ID NO: 66 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 66, and/or the target-complementary sequence may comprise the polynucleotide sequence of SEQ ID NO: 67 or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 67. In some embodiments, the target sequence may be or comprise SEQ ID NO: 68 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 68, and/or the target-complementary sequence may comprise the polynucleotide sequence of SEQ ID NO: 69 or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 69. (0074] In certain embodiments, the pharmaceutical composition or the at least one cargo may further comprise an isolated ODN. In some cases, the isolated ODN may comprise a ssODN comprising or consisting of a nucleic acid sequence having at least 80%, at least 85%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% complementarity to the sequence of a segment within BCLJ 1 A or a variant thereof and/or the erythroid-enhancer region (EER) of BCLJ JA and/or a promoter or enhancer region of BCLJ JA. In certain cases, the segment may comprise the target sequence and/or the sequence complementary thereto and may be about 20-300 nt, about 25-200 nt, about 30-160 nt, about 40-140 nt, about 50-120 nt, about 60-100 nt, about 70-90 nt, about 75-85 nt, about 20 nt, about 25 nt, about 30 nt, about 35 nt, about 40 nt, about 45 nt, about 50 nt, about 55 nt, about 60 nt, about 65 nt, about 70 nt, about 75 nt, about 80 nt, about 85 nt, about 90 nt, about 95 nt, about 100 nt, about 105 nt, about 120 nt, about 125 nt, about 130 nt, about 135 nt, about 140 nt, about 145 nt, about 150 nt, about 155 nt, about 160 nt, about 175 nt, about 180 nt, about 185 nt, about 190 nt, about 195 nt, about 200 nt, about 205 nt, about 220 nt, about 225 nt, about 230 nt, about 235 nt, about 240 nt, about 245 nt, about 250 nt, about 255 nt, about 260 nt, about 275 nt, about 280 nt, about 285 nt, about 290 nt, about 295 nt, or about 300 nt in length. In particular cases, the isolated ODN may be about 30-160 nt, 50-120 nt, about 60-100 nt, about 70-90 nt, about 75-85 nt, about 80 nt or about 75, 76, 77, 78, 79, 81, 82, 83, 84, or 85 nt in length. In particular case, the nucleic acid sequence of the ssODN may have at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to SEQ ID NO: 305, 306,303,304, 301, or 302. In some cases, the isolated ODN may comprise a dsODN, which may comprise a first strand comprising a ssODN sequence described above and a second strand complementary to the first strand. (0075] In some embodiments, in any of the pharmaceutical composition for effecting gene editing to HBGJ and/or HBG2 (or a promoter region thereof), the target sequence may or may comprise SEQ ID NO: 86 or at least the first 17, 18, or 19 nucleotides from the 3' end of SEQ ID NO: 86, and/or the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 87 or at least the first 17, 18, or 19 nucleotides thereof from the 5' end of SEQ ID NO: 87. In some embodiments, the target sequence may or may comprise SEQ ID NO: 286 or at least the first 17, 18, or 19 nucleotides from the 3' end of SEQ ID NO: 286, and/or the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 287 or at least the first 17, 18, or 19 nucleotides thereof from the 5' end of SEQ ID NO: 287. In some embodiments, the target sequence may or may comprise SEQ ID NO: 288 or at least the first 17, 18, or 19 nucleotides from the 3' end of SEQ ID NO: 288, and/or the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 289 or at least the first 17, 18, or 19 nucleotides thereof from the 5' end of SEQ ID NO: 289. In some embodiments, the target sequence may be or comprise SEQ ID NO: 280 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 280, and/or the target- complementary sequence may comprise the polynucleotide sequence of SEQ ID NO: 281 or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 281. In some embodiments, the target sequence may be or comprise SEQ ID NO: 282 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 282, and/or the target-complementary sequence may comprise the polynucleotide sequence of SEQ ID NO: 283 or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 283. In some embodiments, the target sequence may be or comprise SEQ ID NO: 284 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 284, and/or the target-complementary sequence may comprise the polynucleotide sequence of SEQ ID NO: 285 or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 285. In some embodiments, the target sequence may be or comprise SEQ ID NO: 84 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 84, and/or the target-complementary sequence may comprise the polynucleotide sequence of SEQ ID NO: 85 or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 85. [0076] In certain embodiments, in any of the pharmaceutical composition for effecting gene editing to HBGJ and/or HBG2 (or a promoter region thereof), the pharmaceutical composition or the at least one cargo may further comprise an isolated ODN. In certain embodiments, the ODN may comprise (I) a ssODN comprising or consisting of a 5' homology arm, an optional central region, and a 3' homology arm, In certain embodiments, (a) (i) the 5' homology arm comprises or consists of the sequence corresponding to: (i-1) a sequence within SEQ ID NO: 195, which: contains at least 5, least l 0, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50 nucleotides, at least 55, at least 60, at least 65, at least 70, at least 75, or at least 80 nucleotides, about 5-80 nucleotides, about l 0- 70 nucleotides, about 20-60 nucleotides, about 30-50 nucleotides, about 35-45 nucleotides, or about 40 nucleotides; and ends at the first, second, third, fourth, fifth, sixth, seventh, eighth, nineth, 10 111, 11 11', 12 111, 13 1", 14 111, 15 1", 16 1", 17 1", 18 1", 19 111, or 20 1" nucleotide counting from the 3'-end of SEQ ID NO: 195 or a more upstream nucleotide within SEQ ID NO: 195; (i-2) a sequence within SEQ ID NO: 195, which: contains about 35-40 nucleotides or about 40 nucleotides; and ends at the first nucleotide counting from the 3'-end of SEQ ID NO: 195; or (i-3) a sequence comprising at least one (such as one, two, three, four, five, six, seven, eight, nine, or ten) mutation(s) relative to the sequence of(i-1) or (i-2), (ii) the optional central region: (ii-1) is absent; or (ii-2) comprises a sequence of one, two, three, five, six, seven, eight, nine, ten, 11, 12, 13, 14,_15, 16, 17, 18, 19, 20, or more nucleotides optionally not comprising the five consecutive nucleotides of the BCL 11 A-binding motif of 5' -TGACC-3', further optionally comprising or consisting of the sequence of 5' -TGAC-3', and (iii) the 3' homology ann comprises or consists of the sequence corresponding to: (iii-1) a sequence within SEQ ID NO: 196, which: contains at least 5, least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50 nucleotides, at least 55, at least 60, at least 65, at least 70, at least 75, or at least 80 nucleotides, about 5-80 nucleotides, about 10- 70 nucleotides, about 20-60 nucleotides, about 30-50 nucleotides, about 35-45 nucleotides, or about 40 nucleotides; and starts at the first, second, third, fourth, fifth, sixth, seventh, eighth, nineth, 10 111 , 11 1 ", 12th, 13t11, 14t1,, 1st\ 16u', 17t1,, 18u', 19t1,, 2ot1,, 21th, 22t11, 23u,, 24u,, 2sn 1, 26 th , 27t1,, 28u', 29 th , 3ou,, 31 u,, 32t11, 33t1,, 34 th , 35t1,, 36 1 11, 37t1,, 38 th , 39t1,, or 40 th nucleotide counting from the 5'-end of SEQ ID NO: 196 or a more upstream nucleotide within SEQ ID NO: 196; (iii-2) a sequence within SEQ ID NO: 196, which: contains about 35-40 nucleotides or about 40 or 41 nucleotides; and starts at the 13 th nucleotide counting from the 5'-end of SEQ ID NO: 196; (iii-3) or a sequence comprising at least one (such as one, two, three, four, five, six, seven, eight, nine, or ten) mutation(s) relative to the sequence of (iii-1) or (iii-2), optionally wherein the ssODN comprises or consists of the sequence of SEQ ID NO: 191; or (b) the sequence of the ssODN is fully complementary to any of the ssODNs of(a), optionally wherein the sequence of the ssODN comprises or consists of the sequence of SEQ ID NO: 192. In certain embodiments, the ODN may comprise (II) a dsODN, which comprises a first strand comprising any of the ssODN sequences of (I) and a second strand complementary to the first strand. [0077] In some embodiments, in any of the pharmaceutical composition for effecting gene editing to HBB, the target sequence may be or comprise SEQ ID NO: 24 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 24, and/or the target-complementary sequence may comprise the polynucleotide sequence of SEQ ID NO: 25, or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 25. In some embodiments, in any of the pharmaceutical composition for effecting gene editing to HBB, the target sequence may be or comprise SEQ ID NO: 44 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 44 such as SEQ ID NO: 46, and/or the target-complementary sequence may comprise the polynucleotide sequence of SEQ ID NO: 45, or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 45 such as SEQ ID NO: 47. In some embodiments, in any of the pharmaceutical composition for effecting gene editing to HBB, the target sequence may be or comprise SEQ ID NO: 48 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 48, and/or the target-complementary sequence may comprise the polynucleotide sequence of SEQ ID NO: 49, or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 49. [0078] Optionally, such a pharmaceutical composition or the at least one cargo may further comprise a DNA repair template, which optionally comprise: (I) a single-stranded oligo DNA nucleotide molecule (ssODN) comprising or consisting of a 5' homology arm, a central region, and a 3' homology arm, or (II) a double-stranded DNA molecule, which comprises a first strand comprising any of the ssODN sequences of (I) and a second strand complementary to the first strand. [0079] As for ssODNs, in some embodiments, the 5' homology arm may comprise or consist of(i-1) the sequence of SEQ ID NO: 112, (i-2) the sequence corresponding to the first nucleotide to at least the 20th nucleotide ( e.g., at least the 30th, such as to the 39th, at least the 40th, such as to the 49th, or at least the 50th, such as to the 50th or the 59th) counting from the 3 '-end of SEQ ID NO: 112, (i-3) or a sequence comprising at least one (such as one, two, three, four, five, six, seven, eight, nine, or ten) silent mutation(s) relative to the sequence of(i-1) or (i-2). In some embodiments, the central region may have the sequence of5'-CTCA-3', 5'-TTCA-3', 5'-CTCT-3', 5'-TTCT-3', 5'-CTCC-3', 5'-TTCC-3', 5'-CTCG-3', or 5'-TTCG-3'. In some embodiments, the 3' homology arm may comprise or consist of(i-1) the sequence of SEQ ID NO: 122, (i-2) the sequence corresponding to the first nucleotide to at least the 20th nucleotide ( e.g., at least the 30th, such as to the 37th, at least the 40th, such as to the 47th, or at least the 50th, such as to the 57th) counting from the 5 '-end of SEQ ID NO: 122, (i-3) or a sequence comprising at least one (such as one, two, three, four, five, six, seven, eight, nine, or ten) silent mutation(s) relative to the sequence of (iii-1) or (iii-2). In particular embodiments, the ssODN may comprise the consist of the sequence of any of SEQ ID NOs: 170, 172, 174, 176, and 101-108. In a particular embodiment, the ssODN may comprise or consist of the sequence of SEQ ID NO: 101 or 102. Alternatively, the sequence of the ssODN may be fully complementary to the sequence any of the ssODNs described above. In particular embodiments, the sequence of the ssODN may be or may comprise any of SEQ ID NOs: 169, 171, 173, and 175. In particular embodiments, a silent mutation if included may be at the 12th nucleotide of SEQ ID NO: 112 (for exarnple a G-to-C mutation) or the corresponding nucleotide position of a sequence complementary to SEQ ID NO: 112 (for example a C-to-G mutation). [0080] As for the double-stranded DNA molecules, a double-stranded DNA molecule may comprise a first strand comprising any of the ssODN sequences described above and a second strand complementary to the first strand. [0081] In some embodiments, in any of the pharmaceutical composition for effecting gene editing to KLF 1, the target sequence may be or comprise SEQ ID NO: 74 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 74, and/or the target-complementary sequence may comprise the polynucleotide sequence of SEQ ID NO: 75 or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 75. In some embodiments, the target sequence may be or comprise SEQ ID NO: 76 or at least the first 17, 18, or 19 nucleotides from the 5' end of SEQ ID NO: 76, and/or the target-complementary sequence may comprise the polynucleotide sequence of SEQ ID NO: 77 or at least the first 17, 18, or 19 nucleotides thereof from the 3' end of SEQ ID NO: 77.In some embodiments, the pharmaceutical composition may comprise a RNP comprising any of the gRNAs described above or herein. [0082] In certain embodiments, in any of the pharmaceutical compositions for effecting gene editing to BCLJ IA (or an EER thereof), (i) the target-complementary sequence may comprise the polynucleotide sequence of SEQ ID NO: 71,277, or 279 and the Cas nuclease may be Casl2a, optionally AsCas 12a or LbCas 12a, further optionally comprising any one of SEQ ID NOS: 210-215 or 220-221, or (ii) the target-complementary sequence may comprise the polynucleotide sequence of SEQ ID NO: 271,273,275, 65, 69, or 67 and the Cas nuclease may be Cas9, optionally SpCas9 or SaCas9, further optionally comprising SEQ ID NO: 150. In particular embodiments, the pharmaceutical composition or the at least one cargo may further comprise an isolated ODN, which optionally comprises: (I) a ssODN which comprises the nucleic acid sequence of (i) SEQ ID NO: 190 and/or the sequence complementary thereto or (ii) SEQ ID NO: 305, 306, 303, 304, 301, or 302, and is about 60-120 nt, about 70-90 nt, or about 80 nt in length; or (II) a dsODN, which comprises a first strand comprising a ssODN sequence of (I) and a second strand complementary to the first strand. [0083] In certain embodiments, in any of the pharmaceutical compositions for effecting gene editing to HBGJ and/or HBG2 (or a promoter region thereof (e.g., the BCLl lA-binding motif), (i) the target- complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 87,287, or 289 and the Cas nuclease is Casl2a, optionally AsCas12a or LbCas12a, further optionally comprising any one of SEQ ID NOS: 210-215 or 220-221, or (ii) the target-complementary sequence comprises the polynucleotide sequence of SEQ ID NO: 281, 283, 285, or 85 and the Cas nuclease is Cas9, optionally SpCas9 or SaCas9, further optionally comprising SEQ ID NO: 150. In particular embodiments, the pharmaceutical composition or the at least one cargo may further comprise an isolated ODN, which optionally comprises: (I) a ssODN comprising or consisting of the sequence of SEQ ID NO: 191 or 192 ; or (II) which comprises a first strand comprising a ssODN sequence of (I) and a second strand complementary to the first strand. [0084] In some embodiments, the pharmaceutical composition may comprise at least one cargo capable of altering the expression of a target gene. In some embodiments, the cargo may comprise, essentially consist of, or consist of a nucleic acid molecule. In some embodiments, the nucleic acid molecule may be a ribonucleic acid (RNA), a single or double stranded RNA, a small interfering RNA (siRNA), a short hairpin RNA, a microRNA (miRNA), a messenger RNA (mRNA), a deoxyribonucleic acid (DNA), a double or single stranded DNA, a plasmid DNA, a complementary DNA ( cDNA), and/or a locked nucleic acid. [0085] Again, in some embodiments, the at least one cargo encapsulated in the TCV may be obtained by: (i) providing an aqueous solution comprising the TCV, optionally wherein the pH of the aqueous solution is about 3 to about 8, further optionally about 4 to about 7.5; and (ii) mixing the at least one cargo with the aqueous solution. In some embodiments, the mixing may be effected under conditions suitable for the at least one cargo to be encapsulate within the TCV. In some embodiments, the mixing may comprise gentle mixing (optionally repeated manual reciprocation of the TCV-generating fluid in a pipette), micromixing optionally using a staggered herringbone micromixer (SHM) or T-junction or Y-junction mixing, or extrusion. In some embodiments, the mixing time may be about 0.1 second to about 20 minutes. [0086] In some embodiments, the aqueous solution of step (i) may be substantially, essentially, or entirely free of ethanol, methanol, isopropanol, tetrahydrofuran (THF), dimethyl sulfoxide (DMSO), dimethyl formamide (DMF), and acetonitrile (ACN), optionally substantially, essentially, or entirely free of organic solvents and detergents. In particular embodiments, the aqueous solution of step (i) may be substantially, essentially, or entirely free of destabilizing agents. [0087] In some embodiments, the mixing of step (ii) may be performed substantially, essentially, or entirely free of ethanol, methanol, isopropanol, tetrahydrofuran (THF), dimethyl sulfoxide (DMSO), dimethyl formamide (DMF), and acetonitrile (ACN). In some embodiments, the mixing of step (ii) may be performed substantially, essentially, or entirely free of organic solvents and detergents. In particular embodiments, the mixing of step (ii) may be performed substantially, essentially, or entirely free of destabilizing agents. (0088] In particular embodiments, the final ethanol concentration after encapsulation may be 5% (v/v) or below, preferably 0.5% (v/v) or below. [0089] In some embodiments, in any of the above-described pharmaceutical composition, the pharmaceutical composition may comprise at least another cargo. In some instances, the at least another cargo may be encapsulated in the TCV encapsulating at least one cargo capable of effecting gene editing or gene expression alteration as describe above. Or, in some instances, the at least another cargo may be encapsulated in a different or separate TCV. Or, in other instances, the at least another cargo may not be encapsulated in a TCV. Regardless of whether the encapsulation status of the at least another cargo, the at least another cargo may be according to any of the at least one cargo capable of effecting gene editing and/or gene expression alteration. [0090] In some embodiments, in any of the above-described pharmaceutical composition, the pharmaceutical composition may further comprise at least one agent that promotes stem cell mobilization. In some instances, the at least one agent that promotes stem cell mobilization may be selected from the group consisting of granulocyte colony-stimulating factor (G-CSF), granulocyte- macrophage colony-stimulating factor (GM-CSF), Plerixafor, stem cell factor (SCF), CXCR4 antagonists (e.g., POL6326, BKT-140, TG-0054), CXCL12 neutralizers (e.g., NOX-Al2), Sphingosine-l-phosphate (SIP) antagonists (e.g., SEW2871), vascular cell adhesion molecule-1/Very Late Antigen 4 (VCAM/VLA-4) inhibitors (e.g., BIO 5192), parathyroid hormone, protease inhibitors ( e.g., Bortezomib ), Grof (e.g., SB-251353), and hypoxia inducible factor (HIF) stabilizers (e.g., FG- 4497). (0091] In some embodiments, in any of the above-described pharmaceutical composition, the pharmaceutical composition may further comprise at least one agent that promotes erythropoiesis. Om some instances, such an agent may be selected from the group consisting ofSCF, GM-CSF, interleukin-3 (IL-3), interleukin-9 (IL-9), erythropoietin (EPO) (or an engineered EPO or EPO mimetic), TGF-beta, growth differentiating factor 11 (GDFl 1), Activin A, Transferrin (Tf), ferritin, ferroportin, hepcidin, vitamin Bl 2, folic acid, and copper. In some other instances, the at least one agent that promotes erythropoiesis may be selected from the group consisting of GAT A-1, ST AT5A, ST AT5B, MCL-1, BCL-xL, and HSP70, a RNA or DNA encoding thereof, and in some cases the agent may be encapsulated in the TCV encapsulating at least one cargo capable of effecting gene editing or gene expression alteration as describe above, or, alternatively, in a different (separate) TCV. In yet other instances, the at least one agent that promotes erythropoiesis may be an inhibitor or silencer of a negative regulator of erythropoiesis, and in some cases such a negative regulator may be selected from the group consisting ofinhibin, TGF-beta, BID (a member of the BCL-2 family), Fas ligand, Fas, and caspases. In some cases, the negative regulator may be encapsulated in the TCV encapsulating at least one cargo capable of effecting gene editing or gene expression alteration as describe above or, alternatively, in a different (separate) TCV. (0092] In some embodiments, in any of the above-described pharmaceutical composition, the TCV may comprise at least one targeting moiety which may allow the TCV to carry the at least one cargo preferentially into one or more target cells. In some embodiments, the one or more target cells may comprise hematopoietic stem cells (HSCs), hematopoietic stem and progenitor cells (HSPCs), multipotent progenitor cells (MPPs), common myeloid progenitors (CMPs), megakaryocyte-erythroid progenitors (MEPs), hematopoietic progenitor cells (HPCs), erythroid progenitors (e.g., burst-forming unit erythroid cells (BFU-Es), colony-forming unit erythroid cells (CFU-Es)), proerythroblasts, erythroblasts (basophilic erythroblasts, early erythroblasts (e.g., type I, type II), polychromatic erythroblasts, intermediate erythroblasts, acidophilic erythroblasts, late erythroblasts, normoblasts, reticulocytes (before nucleus expulsion), or any combinations thereof, In some preferred embodiments, the one or more target cells may comprise, may be, may essentially consist of, or consist ofHSCs and/or HSPCs. In particular embodiments, the targeting moiety may be specific to, target CD34, and/or target CD34+ cells. (0093] In another aspect, the present disclosure further provides a method for effecting gene editing and/or gene expression alteration in one or more target cells in vivo in a subject in need thereof. (0094] In some embodiments, the pharmaceutical composition may be injected into the bone marrow of the subject. (0095] In certain embodiments, the one or more target cells may comprise HSCs, HSPCs, MPPs, CMPs, MEPs, HPCs, erythroid progenitors (e.g., BFU-Es, CFU-Es), proerythroblasts, erythroblasts (basophilic erythroblasts, early erythroblasts (e.g., type I, type II), polychromatic erythroblasts, intermediate erythroblasts, acidophilic erythroblasts, late erythroblasts, normoblasts, reticulocytes (before nucleus expulsion), or any combinations thereof, preferably HSCs and/or HSPCs. (0096] In certain embodiments, the subject has or has a risk of developing SCD, which may optionally be SCA, HbSC, or HbS ~-thalassaemia. (0097] In certain embodiments, the pharmaceutical composition may comprise, per mL, about 300 pmol to about 30000 pmol of the RNP or the nucleic acid molecule. In particular embodiments, the pharmaceutical composition may comprise, per mL, about 500 to about 10000 pmol, about 1000 to about 5000 pmol, about 2000 to about 4000 pmol, about 2500 to about 3000 pmol, or about 2700 pmol of the RNP or the nucleic acid molecule. (0098] In certain embodiments, the injecting may comprise injecting the pharmaceutical composition in a continuous flow of about 25 mL to 125 mL per minute, In particular embodiments, the injecting may comprise about 25 mL to 50 mL per minute, about 50 mL to 100 mL per minute, about 100 mL to 125 mL per minute, about 40 mL to about 80 mL per minute, or about 50 mL to about 70 mL per minute. [0099] In certain embodiments, the injecting may be a slow bolus push using an instrument with an intraosseous device or intramarrow needle optionally having a needle length of about 50 to about 100 mm or about 70 to about 80 nm. [0100] In certain embodiments, the injecting may be effected, optionally two or more times, to reach a minimum of about 10%, about 15%, about 20%, about 30%, or an about final 15-30% or about final 20-40% HSCs and HSPCs with successful gene editing and/or gene expression alteration among the total HS Cs and HS PCs in the bone marrow. [0101] In certain embodiments, the injecting may be effected two or more times, optionally about 3-5 time, optionally about once a week, about every 2 weeks, or about every 3 weeks, about once a month, about every 3 months, about every 6 months, or about once per year. [0102] In ce1tain embodiments, the bone marrow the composition may be injected into may be the bone of tibia, femur, sternum, skull, ribs, pelvis (e.g., iliac), or any combinations thereof. [0103] In certain embodiments, the subject may be of any age and/or at any stage of the disease. For example, the subject may be in the immediate post-natal period, optionally about 6 weeks old or younger, may be about 3 month old or younger, may still comprises sufficient amount of fetal hemoglobin (HbF) relative to adult hemoglobin (HbA) ( e.g., HbF:HbA is about 2: 1, about 1: 1, about I :2, about 1 :3, about 1 :4, about 1 :5, or about 1: 10), and/or may not have fully developed SCD and may be prior to manifesting a symptom or complication. [0104] In some embodiments, in the method of effecting gene editing and/or gene expression alteration in one or more target cells in vivo in a subject in need thereof, the method may comprise: (I) administering, optionally intravenously, to the subject at least one agent that promotes stem cell mobilization (from the bone marrow to the peripheral circulation); and (II) injecting, optionally intravenously, any of the pharmaceutical compositions described above into the peripheral circulation of the subject. [0105] In some instances, the at least one agent that promotes stem cell mobilization may, for example, selected from the group consisting ofG-CSF (filgrastim), GM-CSF, Plerixafor, SCF, CXCR4 antagonists (e.g., POL6326, BKT-140, TG-0054), CXCL12 neutralizers (e.g., NOX-A12), Sphingosine-1-phosphate (SIP) antagonists (e.g., SEW2871), VCAM/VLA-4 inhibitors (e.g., BIO 5192), parathyroid hormone, protease inhibitors (e.g., Bortezomib), Gro~ (e.g., SB-251353), hypoxia inducible factor (HIF) stabilizers (e.g., FG-4497), and any combinations thereof. [0106] In certain embodiments, the one or more target cells may comprise HSCs, HS PCs, MPPs, CMPs, MEPs, HPCs, erythroid progenitors (e.g., BFU-Es, CFU-Es), proerythroblasts, erythroblasts (basophilic erythroblasts, early erythroblasts ( e.g., type I, type II), polychromatic erythroblasts, intermediate erythroblasts, acidophilic erythroblasts, late erythroblasts, normoblasts, reticulocytes (before nucleus expulsion), or any combinations thereof. In a particular embodiment the one or more target cells may comprise, may be, may essentially consist of, or consist ofHSCs and/or HSPCs. [0107] In some instances, the subject may have or may have a risk of developing SCD, which optionally may be SCA, HbSC, or HbS ~-thalassaemia. [0108] In certain embodiments, the administering at least one agent that promotes stem cell mobilization may comprise intravenous (IV) administration of G-CSF followed by intravenous administration of plerixafor prior to said injecting. In particular embodiments, the dosing of G-CSF may be about 5-30 µg/kg/day, preferably about 10 ug/kg/day, for about 3-5 days, preferably 4 days. In particular embodiments, the dosing of plerixafor may start once the peripheral blood CD34+ cells are <20 cells/µL and/or on the day of the last G-CSF administration or the following day. In particular embodiments, the dosing of plerixafor may be about 0.1-0.5 mg/kg, preferably about 0.2-0.3 mg/kg or about 0.24 mg/kg. [0109] In certain embodiments, the pharmaceutical composition which is to be administered to the peripheral circulation of the subject may comprise, per mL, about 300 pmol to about 30000 pmol of the RNP or the nucleic acid molecule. In particular embodiments, the pharmaceutical composition which is to be administered to the peripheral circulation of the subject may comprise, per mL, about 500 to about 10000 pmol, about 1000 to about 5000 pmol, about 2000 to about 4000 pmol, about 2500 to about 3000 pmol, or about 2700 pmol of the RNP or the nucleic acid molecule. [0110] In certain embodiments, the injecting any of the pharmaceutical compositions may starts once the peripheral blood CD34+ cells are 60 cells/µL or more. In certain embodiments, the injecting may be a single injection, optionally about 3- 7 days, about every 3- 7 days, about 4-6 days, about every 4-6 days, about 5 days, or about every 5 days after the last plerixafor administration. In certain embodiments, the injecting may occur once daily for one week following the last plerixafor administration. [0111] In certain embodiments, the injecting may comprise injecting the pharmaceutical composition in a continuous flow of about 25 mL to 125 mL per minute. In particular embodiments, the injecting may comprise about 25 mL to 50 mL per minute, about 50 mL to 100 mL per minute, about 100 mL to 125 mL per minute, about 40 mL to about 80 mL per minute, or about 50 mL to about 70 mL per minute. [0112] In certain embodiments, the combination of the adm inistration of at least one agent that promotes stem cell mobilization and the injection of any of the pharmaceutical compositions described herein may be effected, optionally two or more times, to reach a minimum of about 10%, about 15%, about 20%, about 30%, or an about final 15-30% or about final 20-40% HSCs and HSPCs with successful gene editing and/or gene expression alteration among the total HSCs and HSPCs in the peripheral circulation. [0113] In certain embodiments, the combination of the administration of at least one agent that promotes stem cell mobilization and the injection of any of the pharmaceutical compositions described herein may be effected, optionally two or more times, to reach a minimum of about 10%, about 15%, about 20%, about 30%, or an about final 20-30% increase in the peripheral HSCs and HS PCs expressing HbF, or a minimum of about 10%, about 15%, about 20%, about 30%, or an about final 20-30% increase in the total HbF expression levels in the total HSCs and HSPCs in the peripheral circulation, optionally wherein the SCD-associated gene is BCLJ JA, HBGJ, HBG2, or KLFJ. [0114) In certain embodiments, the combination of the administration of at least one agent that promotes stem cell mobilization and the injection of any of the pharmaceutical compositions described herein may be effected two or more times. In particular embodiments, the combination may be effected about 3-5 time, about once a week, about every 2 weeks, or about every 3 weeks, about once a month, about every 3 months, about every 6 months, or about once per year. [0115) In certain embodiments, the subject may be at any age and/or at any disease stage. In particular embodiments, the subject may be in the immediate post-natal period, optionally about 6 weeks old or younger, may be about 3 month old or younger, may still comprises sufficient amount of HbF relative to HbA (e.g., HbF:HbA is about 2: 1, about 1: 1, about 1:2, about 1 :3, about 1 :4, about 1 :5, or about 1: 10), and/or may not have fully developed SCD and is prior to manifesting a symptom or complication. [0116) In another aspect, the present disclosure further provides a method for preventing, ameliorating, or treating a disease, which may relate to cells of bone marrow origin and/or cells of the bone marrow. In some embodiments, the disease may be SCD, which optionally may be SCA, HbSC, or HbS ~-thalassaemia, in a subject in need thereof. [0117) In some embodiments, the method for preventing, ameliorating, or treating a disease may comprise any of the in vivo methods described herein in which the pharmaceutical composition is injected into the bone marrow of the subject and/or the in vivo method in which the pharmaceutical composition is injected into the peripheral circulation (e.g., IV) of the subject. [0118) In certain embodiments, the effect of the method may be evaluated based on any appropriate parameters (and/or any combinations thereof) that indicate successful gene editing and/or gene expression alteration and/or the associated improvement in any of the disease symptoms. [0119) In particular embodiments, the method may be evaluated based on % HSCs and HSPCs in the blood with successful gene editing and/or gene expression alteration. In particular embodiments, the method may be evaluated based on the number of HSCs and HSPCs in the blood with successful gene editing and/or gene expression alteration. In particular, the method may be evaluated based on% HSCs and HSPCs expressing HbF (e.g., when the SCD-associated gene is BCLl lA or KLFl). In particular embodiments, the method may be evaluated based on the number ofHSCs and HSPCs expressing HbF, optionally wherein the SCD-associated gene is BCLJ JA, HBGJ, HBG2, or KLFJ. In particular embodiments, the method may be evaluated based on the expression level of the at least one SCD-associated gene or gene product or molecule, such as but not limited to beta-globin, beta-globin (HbS variant), gamma-globin 1, gamma-globin 2, HbF, HbA, BCLl lA, and/or KLFl. In particular embodiments, the method may be evaluated based on changes in the symptom optionally pain, swelling of hands and feet, infection frequency, growth, and/or symptoms associated with vision. [0120] In certain embodiments, the method further comprises administering at least one agent that promotes erythropoiesis. In particular embodiments, the at least one agent that promotes erythropoiesis may be selected from the group consisting of SCF, GM-CSF, IL-3, IL-9, EPO (or an engineered EPO or EPO mimetic), TGF-beta, GDF 11, Activin A, Tf, ferritin, ferroportin, hepcidin, vitamin B12, folic acid, copper, and any combinations thereof. In particular embodiments, the at least one agent that promotes erythropoiesis may be selected from the group consisting of GAT A-1, ST AT5A, ST AT5B, MCL-1, BCL-xL, and HSP70, a RNA or DNA encoding thereof. Such an agent may be encapsulated in the TCV encapsulating the at least one cargo capable of effecting gene therapy and/or gene expression alteration. Alternatively, such an agent may be encapsulated in a TCV different or separate from the TCV encapsulating the at least one cargo capable of effecting gene therapy and/or gene expression alteration. In particular embodiments, the at least one agent that promotes erythropoiesis may be an inhibitor or silencer of a negative regulator of erythropoiesis. In a particular embodiment, optionally the negative regulator may be selected from the group consisting of inhibin, TGF-beta, BID (a member of the BCL-2 family), Fas ligand, Fas, and caspases, and any combinations thereof. Such an agent may be encapsulated in the TCV encapsulating the at least one cargo capable of effecting gene therapy and/or gene expression alteration. Alternatively, such an agent may be encapsulated in a TCV different or separate from the TCV encapsulating the at least one cargo capable of effecting gene therapy and/or gene expression alteration. [0121] In certain embodiments, the method further comprises administering at least one other agent for treating SCD, which optionally comprises hydroxyurea, L-glutamine oral powder, crizanlizumab, a general pain medication, voxelotor, or any combination thereof. [0122] In another aspect, the present disclosure further provides a method of manufacturing any of the pharmaceutical compositions described herein comprising a RNP. [0123] In some embodiments, the method may comprise: (a) providing an aqueous solution comprising the TCV, optionally wherein the pH of the aqueous solution is about 3 to about 8, further optionally about 3.5 to about 7.5, about 3.5 to about 5.5, or about 4; and (b) mixing the at least one cargo with the aqueous solution under conditions suitable for the at least one cargo to be encapsulate within the TCV, [0124] In certain embodiments, the RNP may be any of the RNPs described herein. [0125] In certain embodiments, the mixing may be for about 0.1 second to about 20 minutes. In certain embodiments, the mixing may be via gentle mixing ( optionally repeated manual reciprocation of the TCV-generating fluid in a pipette). In certain embodiments, the mixing may be via micromixing, optionally using a staggered herringbone micromixer (SHM) or T-junction or Y- junction mixing. In certain embodiments, the method may comprise extrusion. [0126] In another aspect, the present disclosure further provides a composition for gene editing ( e.g., disrupting a gene or gene segment of interest) comprising: (A) a Cas nuclease or a nucleic acid encoding the Cas nuclease; (B) a gRNA or a nucleic acid encoding the gRNA; and (C) an isolated ODN or a nucleic acid encoding the isolated ODN. [0127] In some embodiments, in (A), the Cas nuclease may be: (i) selected from the group consisting ofCasl2a or Cpfl, Cas9, Cas3, Cas8a2, Cas8b, Cas8c, Casl0, Casl 1, Casl2, Cas12b, Cas12f, Cas12j, Cas13, Cas13a, Cas14, C2cl, C2c3, and C2c2; (ii) a class 2 Cas endonuclease, optionally a type II, type V, or type VI Cas nuclease; (iii) Cas12a of Acidaminococcus sp. (AsCas12a), Lachnospiraceae bacterium (LbCas12a), Francisella novicida (FnCas12a), Moraxella bovoculi (MbCas12a), Coprococcus eutactus (CeCas12a), Butyrivibrio fibrisolvens (BdCas12a); (iv) Cas12a or any engineered variants thereof, optionally comprising any one of SEQ ID NOS: 210-215, 220-221, 230, 240, 250, and 260; (v) Cas9 of Streptococcus pyogenes (SpCas9), Staphylococcus aureus (SaCas9), Streptococcus thermophilus (StCas9), Neisseria meningitidis (NmCas9), Francisella novicida (FnCas9), Campylobacter jejuni (CjCas9), Streptococcus canis (ScCas9), or Staphylococcus auricularis (SauriCas9), or any engineered variants thereof, including SaCas9-HF, SpCas9-HF1, KKHSaCas9, eSpCas9, HypaCas9, FokI-Fused dCas9, xCas9, SpRY (variant ofSpCas9), and SpG (variant of SpCas9); and/or (vi) Cas9, optionally comprising any one of SEQ ID NOS: 150-161. [0128] In some embodiments, in (B), the gRNA may comprise a target-complementary sequence of interest such that the target-complementary sequence is capable of binding to a target sequence of interest within a target gene of interest thereby guiding the Cas9 nuclease of (A) to and allowing for cleavage at a target site of interest within said target gene of interest. [0129] In some embodiments, in (C), the isolated ODN may comprise: (I) a ssODN comprising a nucleic acid sequence having less than 90%, less than 85%, less than 80%, less than 75%, less than 70%, less than 65%, less than 60%, less than 55%, less than 50%, less than 45%, less than 40%, less than 35%, less than 30%, less than 30%, less than 25%, less than 20%, less than 15%, or less than 10% complementarity to the sequence of a segment within the target gene of interest , wherein the segment comprises the target sequence and/or the sequence complementary thereto and is about 20- 300 nt, about 25-200 nt, about 30-160 nt, about 40-140 nt, about 50-120 nt, about 60-100 nt, about 70- 90 nt, about 75-85 nt, about 20 nt, about 25 nt, about 30 nt, about 35 nt, about 40 nt, about 45 nt, about 50 nt, about 55 nt, about 60 nt, about 65 nt, about 70 nt, about 75 nt, about 80 nt, about 85 nt, about 90 nt, about 95 nt, about 100 nt, about 105 nt, about 120 nt, about 125 nt, about 130 nt, about 135 nt, about 140 nt, about 145 nt, about 150 nt, about 155 nt, about 160 nt, about 175 nt, about 180 nt, about 185 nt, about 190 nt, about 195 nt, about 200 nt, about 205 nt, about 220 nt, about 225 nt, about 230 nt, about 235 nt, about 240 nt, about 245 nt, about 250 nt, about 255 nt, about 260 nt, about 275 nt, about 280 nt, about 285 nt, about 290 nt, about 295 nt, or about 300 nt in length; or (11) a dsODN, which comprises a first strand comprising a ssODN sequence of (I) and a second strand complementary to the first strand. In certain embodiments, (i) the isolated ODN is about 30-160 nt, 50-120 nt, about 60-100 nt, about 70-90 nt, about 75-85 nt, about 80 nt in length; (ii) the nucleic acid sequence of the isolated ODN has at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to SEQ ID NO: 190 or a sequence complementary thereto; (iii) the isolated ODN has no significant alignment ( optionally defined bye-value < 0.05) with an about 120-nt, about 100-nt, about 90 nt, about 85-nt, about 80-nt, about 75-nt, or about 70-nt sequence of the target gene of interest encompassing the target sequence or a reverse complement thereof, as analyzed by Blast®N or Nucleotide BLAST® and/or (iv) the delta G value for a hypothetical dimer of the isolated ODN and an about 120-nt, about 100-nt, about 90 nt, about 85-nt, about 80-nt, about 75-nt, or about 70-nt sequence of the target gene of interest encompassing the target sequence and/or a reverse complement thereof is about -100 kcal/mo! or higher, about -80 kcal/mo! or higher, about -60 kcal/mo! or higher, about -50 kcal/mo! or higher, about -40 kcal/mo! or higher, about -30 kcal/mo! or higher, about -20 kcal/mo! or higher, about -18 kcal/mo! or higher, about -16 kcal/mo! or higher, about -15 kcal/mo! or higher, about -14 kcal/mo! or higher, about -12 kcal/mo! or higher, or about -10 kcal/mo! or higher. [0130] In certain embodiments, the Cas nuclease and the gRNA may be comprised in a RNP. In certain embodiments, one or more of the Cas nuclease, the gRNA, and/or the isolated ODN may be encapsulated in a carrier, optionally a lipid-based TCV, such as any of those described herein. [0131] Without wishing to be bound by theory, use of such an ODN (one that is not complementary to or have low complementarity to the sequence flanking the cut site by the Cas nuclease) may enhance the efficiency of the intended gene editing (e.g., disrupting a gene or gene segment of interest). [0132] In particular embodiments, the target gene of interest is not human huntingtin. In particular embodiments, the target gene of interest is not huntingtin. In particular cases, the target sequence is not within an about 120-nt, about 100-nt, about 90-nt, or about 80-nt sequence encompassing the junction between intron 12 and exon 13 of the human huntingtin gene and/or not within intron 12 and/or exon 13 of the human huntingtin gene. [0133] In another aspect, the present disclosure further provides a method of effecting gene editing ( e.g., disrupting a gene or gene segment of interest) at a target site of interest within a target gene of interest in a target cell of interest. [0134] In some embodiments, the method comprising contacting the target cell of interest with such a composition described above comprising: (A) a Cas nuclease or a nucleic acid encoding the Cas nuclease; (B) a gRNA or a nucleic acid encoding the gRNA; and (C) an isolated ODN or a nucleic acid encoding the isolated ODN. In certain embodiments, the gene editing may comprise deletion of one or more nucleotides and/or insertion of one or more nucleotides at the target site of interest ( e.g., at the cleavage site), optionally thereby deactivating or activating the target gene of interest. In certain embodiments, the target gene of interest may be a disease-associated or disease-causing gene, optionally a SCD-associated gene or its regulatory (e.g., enhancer or promoter/repressor-binding) region, further optionally wherein the target site of interest is within BCLJ J A or the EER of BCLJ 1 A, HBGJ, the promoter region (e.g., BCLl lA-binding motif) of HBGJ, HBG2, and/or the promoter region (e.g., BCLl lA-binding motif) of HBG2. In certain embodiments, the contacting may occur in vitro, ex vivo, or in vivo. In certain embodiments, the contacting occurs in vivo and effected by administering the composition to a subject in need of the gene editing, optionally wherein the administering comprises injecting the composition into the bone marrow of the subject or intravenously. In certain embodiments, the method may be for preventing or treating a disease, optionally SCD. In particular embodiments, the target gene of interest is not human huntingtin. In particular embodiments, the target gene of interest is not huntingtin. In particular cases, the target sequence is not within an about 120-nt, about 100-nt, about 90-nt, or about 80-nt sequence encompassing the junction between intron 12 and exon 13 of the human huntingtin gene. In some particular cases, the target sequence is not within intron 12 and/or exon 13 of the human huntingtin gene. BRIEF DESCRIPTION OF THE DRAWINGS [0135] FIG. lA is a schematic of a transgenic reporter mouse design. Proof-of-concept gRNAs are designed, so that, when successful gene editing occurs, the reporter fluorescent gene will be turned on. The top diagram is a generic schematic of a trans gene construct of reporter mice. The bottom diagram is a schematic of the trans gene construct of Ai 14 mice and approximate locations of the target cut sites of the four gRNAs ("LaRo", "PS2", "PS3", and "LoxP") used in Example 5. Ai14 mice harbor a modification at the Rosa26 locus with a ubiquitous CAG promoter followed by a floxed-stop cassette (three repeats of the SV40 polyadenylation (poly A) sequence) that prevents expression of the tdTomato fluorescent marker protein. The Ai14 strain was originally designed and generated by Madisen et al. (Madisen et al., Nat Neurosci. 2010 January; 13(1): 133-140.), along with several other reporter mouse strains having the same stop cassette, for Cre-mediated recombination at the loxP sites leading to excision of the stop cassette and tdTomato expression. [0136] FIG. 1B provides exemplary results obtained in Example 5. Ai14 reporter mice received direct bone marrow injections with TCVs encapsulating RNP composed ofCas9 and sgRNAs targeting either a reporter gene ("IG-TOM00l ") or a control gene ("IG-Luc00l "). Bone marrow cells were harvested 48 hours after injections and cultured for 72 hours. Cells were then stained for CD34 and with DAPI and analyzed by fluorescent microscopy. The graphs show %reporter+ cells (unpaired t-test: **p=0.0077) (left), %CD34+ cells (unpaired t-test: p=0.8547) (middle), and% double positive cells (i.e., reporter+ CD34+) (Unpaired t-test: **p=0.0040) (right). [0137] FIGS. 2A-2J relate to exemplary disruption of the erythroid-enhancer region (EER) of BCLJJA. [O 138] FIG. 2A shows a schematic of an editing strategy used in Examples 12-13. Numbered boxes represent BCLJ 1 A exons of the indicated exon number, and lines between exons indicate introns. The dashed box represents the EER in its intact form. Each open rectangle above the EER represents a sgRNA portion comprising a target-complementary sequence (e.g., SEQ ID NO: 65, 67, 69,, 71,277 or 279) which may hybridize to the EER. Each filled rectangle adjacent thereto indicates where the PAM sequence is in the target DNA Upon hybridization of a gRNA, Cas nuclease may mediate NHEJ, resulting in a disrupted EER (grid box). Transcription factor(s) may no longer bind to the disrupted EER, and transcription of BCLJ 1 A may be inhibited in cells of the erythroid lineage. BCLl lA is a repressor of HBGJ and HBG2, and inhibition of BCLJ JA transcription may result in increased gamma-globin thereby restoring HbF production. [0139] FIG. 2B provides exemplary results obtained in Example 12 (12-1). HEK293 cells were treated with TCV-encapsulated Cas9-based RNPs comprising sgRNA targeting a control gene (luciferase; using SEQ ID NO: 55) or BCLJ JA EER (using SEQ ID NO: 65 or SEQ ID NO: 69). The graph shows percent editing efficiency at the target sites. N=3 per treatment condition. One way ANOV A: p=0.0010. Dunnett's multiple comparison test: **p=0.0027, ***p=0.0007. [0140] FIG. 2C provides exemplary results obtained in Example 13. HEK293 cells were treated with increasing amounts ofTCV-encapsulated Cas9-based RNPs comprising sgRNA targeting a control gene (luciferase; using SEQ ID NO: 55) or targeting BCLJ JA EER (using SEQ ID NO: 65). The graphs in the top show percent editing efficiency at the target site in BCLJ 1 A. N=3 per treatment condition. Two-way way ANOV A: treatment p<0.0001, dose p<0.0001, interaction p<0.0001. Tukey's multiple comparison test: ****p<0.0001. The graph in the bottom shows dose-dependent changes in the percent editing efficiency at the target site in BCLJ 1 A. (Linear regression: Luciferase: Y = 0.003838*X + 0.4319, BCLl lA EER 1: Y = 0.1170*X + 12.44. Slopes are significantly different p<0.0001). [0141] FIG. 2D provides exemplary results obtained in Example 12 (12-2 and 12-3). In the left graph, HEK293 cells were treated with TCV-encapsulated Cas9-based RNPs comprising sgRNA targeting a control gene (HBGJ and HBG2 promoter) or BCLJ JA (using SEQ ID NO: 65 or SEQ ID NO: 69) or Cas12a-based RNP comprising gRNA targeting BCLJ JA EER (using SEQ ID NO: 71). The left graph shows percent editing efficiency at the target sites. N=3 per treatment condition. One- way ANOV A p<0.0001. Dunnett's multiple comparison test ***p=0.0001, ****p<0.0001). In the right graph, HEK293 cells were treated with TCV-encapsulated Cas 12-based RNPs comprising sgRNA targeting a control gene (HBGJ and HBG2 promoter) or BCLJ JA EER (using SEQ ID NO: 71, 277, or 279). The right graph shows percent editing efficiency at the target sites. N=3 per treatment condition. One-way ANOVA p<0.0001. Turkey's multiple comparison test ****p<0.0001). [0142] FIG. 2E provides exemplary results obtained in Example 12 (12-4). HEK293 cells were treated with TCV-encapsulated Cas9-based RNPs comprising sgRNA targeting a control gene (HBGJ and HBG2 promoter) or BCLJ JA EER (using SEQ ID NO: 65 or SEQ ID NO: 69) or Cas12a-based RNP comprising gRNA targeting a control gene (HBGJ and HBG2 promoter) or BCLJ JA EER (using SEQ ID NO: 71), with or without a ssODN "HTT C680bp_R" (SEQ ID NO: 190) which is not complementary to the sequence proximate to the target site. The left graph shows percent BCLJ 1 A editing efficiency with Cas9-based RNPs (left, One-way ANOV A p<0.0001. Tukey's multiple comparison test: *p=0.0110, ***p=0.0002, ****p<0.0001). The right graph shows percent BCLJ JA editing efficiency with Casl 2a-based RNPs (right, One-way ANOV A p<0.0001. Tukey's multiple comparison test: *p=0.0124, ****p<0.0001) at the target sites. N=3 per treatment condition. [0143) FIG. 2F shows a portion of the human BCLJ JA EER sequence. Specifically, the segment corresponding to the nucleotide positions 60,495,352 to 60,495,172 of Chromosome 2 according to Gene Assembly GRCh38.pl4 having SEQ ID NO: 4 in the coding strand and SEQ ID NO: 5 in the non-coding strand is presented. Some of the potential PAM sequences which may be utilized for Cas- mediated gene editing found in the segment are also shown (solid lines for 5'-NGG-3' and double headed arrows for 5'-TTTN-3'). [0144] FIG. 2G shows an alignment between a non-complementary ssODN sequence (top, SEQ ID NO: 190) and a 123-base, coding-strand sequence of BCLJ JA EER around the target cut site(s) (bottom, SEQ ID NO: 14; coding strand at positions 60,495,317 to 60,495,195 of Chromosome 2 according to Gene Assembly GRCh38.pl4 (bases 36-158 of SEQ ID NO: 4)). "I" represents identical bases between the top and bottom represents mismatches; and"" (space) represents unaligned sequence. A similarity analysis using BlastN found no significant similarity. [0145) In FIG. 2H, the top shows minimal alignment between a non-complementary ssODN sequence (top, SEQ ID NO: 190) and a 123-base, coding-strand (positive strand) sequence of BCLJ JA EER around the target cut site(s) (bottom, SEQ ID NO: 14; coding strand at positions 60,495,317 to 60,495,195 of Chromosome 2 according to Gene Assembly GRCh38.p14 (bases 36-158 of SEQ ID NO: 4)), analyzed using CLUSTAL alignment software. The bottom shows a heterodimer analysis between SEQ ID NOS: 190 and 14. The delta G is calculated by taking into account the longest stretch of complementary bases in the dimer shown. Solid lines represent complementary base bases; and dotted lines represent additional complementary bases for the dimer structure, but their presence does not impact calculated delta G values. The Maximum Delta G value refers to the free energy of the oligo sequence binding to its perfect complement. A delta G value of -10.88 kcal/mo! represents essentially no binding (no formation of the dimer structure). [0146) In FIG. 21, the top shows minimal alignment between a non-complementary ssODN sequence (top, SEQ ID NO: 190) and a 123-base, non-coding-strand (negative strand) sequence of BCLJ JA EER around the target cut site(s) (bottom, SEQ ID NO: 15; non-coding strand at positions 60,495,195 to 60,495,317 of Chromosome 2 according to Gene Assembly GRCh38.p14 (bases 24-146 of SEQ ID NO: 5)), analyzed using CL UST AL alignment software. The bottom shows a heterodimer analysis between SEQ ID NOS: 190 and 15. The delta G is calculated by taking into account the longest stretch of complementary bases in the dimer shown. Solid lines represent complementary base bases; and dotted lines represent additional complementary bases for the dimer structure, but their presence does not impact calculated delta G values. The Maximum Delta G value refers to the free energy of the oligo sequence binding to its perfect complement. A delta G value of -15.42 kcal/mo! represents essentially no binding (no formation of the dimer structure). [0147] FIG. 2J provides exemplary results obtained in Example 12 (12-5). HEK293 cells were treated with TCV-encapsulated Cas9-based RNPs comprising sgRNA targeting a control gene ( GFP) or BCLJ 1 A EER (using SEQ ID NO: 65) or Cas 12a-based RNP comprising gRNA targeting a control gene (GFP) or BCLJ JA EER (using SEQ ID NO: 71), with or without a ssODN "BCLl IA EER_80bp F" or "BCLl lA EER_80bp R" (SEQ ID NO: 305 or 306, respectively) which are either identical or complementary to the coding strand sequence proximate to the target site. The left graph shows percent BCLJ JA editing efficiency with Cas9-based RNPs (N=3 per treatment condition; One-way ANOVA p<0.0001; Tukey's multiple comparison test: **p=0.0034, ****p<0.0001). The right graph shows percent BCLJ JA editing efficiency with Cas12a-based RNPs (N=3 per treatment condition; One-way ANOVA p<0.0001; Tukey's multiple comparison test: *p=0.0373, ***p=0.0009, ****p<0.0001) at the target sites. [0148] FIGS. 3A-3G provide exemplary disruption of the BCLl IA-binding site present in both the promoter of HBGJ and the promoter of HBG2. [0149] FIG. 3A shows a schematic of an editing strategy used in Examples 14-15. Boxes with italic letters indicate the HBE, HBG2, HBGJ, HBD, and HEB genes located in the ~-like globin gene cluster. Dotted boxes are the promoters for HBG2 and HBG 1. Each open rectangle above the promoter represents a gRNA portion comprising a target-complementary sequence (e.g., SEQ ID NO: 85 or 87) which may hybridize to the promoter. Each filled rectangle adjacent thereto indicates where the PAM sequence is in the target DNA. Upon hybridization of a gRNA, Cas nuclease may mediate NHEJ, resulting in a disrupted promoter (grid box). BCLl IA (gamma-globin repressor) may no longer bind to the promoter of HBGJ and/or the promoter of HBG2, leading to de-repression or expression of HBGJ and/or HBG2, thereby restoring HbF production. [0150] FIG. 3B provides exemplary results obtained in Example 14. HEK293 cells were treated with TCV-encapsulated Cas9-based RNPs comprising sgRNA targeting a control gene (luciferase; using SEQ ID NO: 55) or the promoter region of HBGJ and HBG2 (using SEQ ID NO: 85). The graph shows percent editing efficiency at the target promoter site of HBGJ (left) and the target promoter site of HBG2 (right). (N=3 per treatment condition. Two-way ANOV A: HBG promoter p<0.0001, treatment p<0.0001, interaction p=0.0021. Sidak's multiple comparison test: *p=0.0298, ****p<0.0001). [O 151] FIG. 3C provides exemplary editing efficiency results obtained in Example 15. HEK293 cells were treated with increasing amounts ofTCV-encapsulated Cas9-based RNPs comprising sgRNA targeting a control gene (luciferase ; using SEQ ID NO: 55) or targeting the promoter region of HBG 1 and HBG2 (using SEQ ID NO: 85). The left graphs show percent editing efficiency at the target promoter site of HBG 1. N=3 per treatment condition. Two-way ANOV A: treatment p<0.0001, dose p<0.0001, interaction p<0.0001. Tukey's multiple comparison test: **p<0.01, ****p<0.0001. The right graphs show percent editing efficiency at the target promoter site of HBG2. N=3 per treatment condition. Two-way ANOVA: treatment p<0.0001, dose p<0.0001, interaction p<0.0001. Tukey's multiple comparison test: ****p<0.0001. [0152] FIG. 3D provides exemplary editing event results obtained in Example 15. The graph provides a representative histogram from TIDE analyses showing distribution of specific editing events following treatment with the TCV-encapsulated Cas9-based RNP targeting the promoter region of HBGJ and HBG2 at 200 nM ofRNPs. The p values indicate whether the editing event occurred at a frequency higher than what would be expected by chance. P values were determined as described in Brinkman et al., Nucleic Acids Res. 2014 Dec 16;42(22):el68 (see, e.g., pages 2-3 such as "the sequence trace from the mutated DNA sample is assumed to be a linear combination of the wild-type and the modeled indel traces. This combination is then resolved by standard non- negative linear modeling, for which we used the R package nnls. R 2 is calculated to assess the goodness of fit. The p-value associated with the estimated abundance of each indel is calculated by a two tailed t-test of the variance-covariance matrix of the standard errors. In order to account for systematic differences between the sequence trace intensities of the control and mutated DNA, the fitting parameters are then multiplied by a constant factor such that their sum equals R 2 ."). [0153] FIG. 3E exemplary results obtained in Example 14 (14-2 and 14-3). In the top graphs, HEK293 cells were treated with TCV-encapsulated Cas9-based RNPs comprising sgRNA targeting a control gene (GFP) or the promoter region of HBGJ and HBG2 (using SEQ ID NO: 85) or Casl2a- based RNP comprising gRNA targeting a promoter region of HBGJ and HBG2 (using SEQ ID NO: 87). The top left graph shows percent editing efficiency at the target sites within the respective promoter region of HBGJ and HBG2. N=3 per treatment condition. Two-way ANOV A: treatment p<0.0001, HBG p<0.0001, interaction p=0.0027. Tukey's multiple comparison test: ****p<0.0001. The top right graph shows percent 13-bp deletion at the target sites within the respective promoter region of HBGJ and HBG2 as determined by the TIDE analyses. N=3 per treatment condition. Two- way ANOV A: treatment p<0.0001, HBG p<0.0001, interaction p<0.0001. Tukey's multiple comparison test: ****p<0.0001) were assessed using TIDE analytical tool. In the bottom graph, HEK293 cells were treated with TCV-encapsulated Cas12-based RNPs comprising sgRNA targeting a control gene (BCLJ IA) or the promoter region of HBGJ and HBG2 (using SEQ ID NO: 87,287, or 289). The bottom graph shows percent editing efficiency at the target sites within the respective promoter region of HBGJ and HBG2. N=3 per treatment condition. Two-way ANOV A: treatment p<0.0001, HBG p<0.0001, interaction p<0.0001. Tukey's multiple comparison test: **p<0.0014, ***p=0.0007, ****p<0.0001. [0154] FIG. 3F shows a segment of the promoter region of human HBGJ and HBG2. Specifically, the segments corresponding to the nucleotide positions 5250070 to 5249890 (within the promoter region of HBG 1) and positions 5254994 to 5254814 (within the promoter region of HBG2), respectively, of Chromosome 11 according to Gene Assembly GRCh38.p14) are presented. Both segments have the identical sequences of SEQ ID NO: 95 in the coding strand and SEQ ID NO: 97 in the non-coding strand and contain the BCLl lA-binding motif of"TGACC". The sequence span corresponding to the 13-nt deletion commonly found in hereditary persistence of fetal hemoglobin (HPFH) (SEQ ID NO: 99) is also indicated. Some of the potential PAM sequences which may be utilized for Cas-mediated gene editing found in the proximity of the BCLl lA-binding motif are also shown (solid lines for 5'-NGG-3' and double headed arrows for 5'-TTTN-3'). [0155] FIG. 3G provides exemplary results obtained in Example 14 (14-4). HEK293 cells were treated with TCV-encapsulated Cas9-based RNPs comprising sgRNA targeting a control gene (GFP) or the promoter region of HBGJ and HBG2 (using SEQ ID NO: 85) or Cas12a-based RNP comprising gRNA targeting a control gene (GFP) or the promoter region of HBGJ and HBG2 (using SEQ ID NO: 87), with or without the ssODN named "13bp del_80bpF" (SEQ ID NO: 191) or the ssODN named "13bp del_80bpR" (SEQ ID NO: 192). The top graphs show percent editing efficiency (left, N=3-6 per treatment condition: Two-way ANOV A: treatment p<0.0001, HBG p<0.0001, interaction p=0.0027; Tukey's multiple comparison test: ****p<0.0001) and percent 13-bp deletion (right, N=3-6 per treatment condition: Two-way ANOV A: treatment p<0.0001, HBG p<0.0001, interaction p<0.0001; Tukey's multiple comparison test: ****p<0.0001), at the target sites within the respective promoter region of HBGJ and HBG2 using Cas9-based RNPs as determined by TIDE analyses. The bottom graphs show percent editing efficiency (left, N=3-6 per treatment condition: Two-way ANOV A: treatment p<0.0001, HBG p<0.0001, interaction p=0.0159; Tukey's multiple comparison test: *p<0.05, **p<0.0l,.***p<0.001, ****p<0.0001) and percent 13-bp deletion (right, N=3-6 per treatment condition: Two-way ANOV A: treatment p<0.0001, HBG p<0.0001, interaction p=0.0002. Tukey's multiple comparison test: *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001), at the target sites within the respective promoter region of HBGJ and HBG2 using Cas12a-based RNPs as determined by TIDE analyses. [0156] FIG. 3H shows an aligmnent between a non-complementary ssODN sequence (bottom, SEQ ID NO: 190) and a 120-base, coding-strand sequence of HBGJ and HBG2 promoter region around the target cut site(s) (top, SEQ ID NO: 295; coding strand at positions 5,250,033 to 5,249,914 (same as positions 5,254,957 to 5,254,838) of Chromosome 11 according to Gene Assembly GRCh38.p14 (bases 38-157 of SEQ ID NO: 95)). "I" represents identical bases between the top and bottom represents mismatches; and"" (space) represents unaligned sequence. A similarity analyis using BlastN found no significant similarity. [O 157] In FIG. 31, the top shows minimal alignment between a non-complementary ssODN sequence ( top, SEQ ID NO: 190) and a 120-base, coding-strand (positive strand) sequence of HBG 1 and HBG 2 promoter region around the target cut site(s) (bottom, SEQ ID NO: 295; coding strand at positions 5,250,033 to 5,249,914 (same as positions 5,254,957 to 5,254,838) of Chromosome 11 according to Gene Assembly GRCh38.p14 (bases 38-157 of SEQ ID NO: 95)), analyzed using CL UST AL alignment software. The bottom shows a heterodimer analysis between SEQ ID NOS: 190 and 295. The delta G is calculated by taking into account the longest stretch of complementary bases in the dimer shown. Solid lines represent complementary base bases; and dotted lines represent additional complementary bases for the dimer structure, but their presence does not impact calculated delta G values. The Maximum Delta G value refers to the free energy of the oligo sequence binding to its perfect complement. A delta G value of -10.14 kcal/mo! represents essentially no binding (no formation of the dimer structure). [0158] In FIG. 3J, the top shows minimal alignment between a non-complementary ssODN sequence (top, SEQ ID NO: 190) and a 120-base, non-coding-strand sequence of HBGJ and HBG2 promoter region around the target cut site(s) (bottom, SEQ ID NO: 297; non-coding strand at positions 5,249,914 to 5,250,033 (same as positions 5,254,838 to 5,254,957) of Chromosome 11 according to Gene Assembly GRCh38.p14 (bases 25-144 of SEQ ID NO: 97)), analyzed using CLUSTAL aligmnent software. The bottom shows a heterodimer analysis between SEQ ID NOS: 190 and 297. The delta G is calculated by taking into account the longest stretch of complementary bases in the dimer shown. Solid lines represent complementary base bases; and dotted lines represent additional complementary bases for the dimer structure, but their presence does not impact calculated delta G values. The Maximum Delta G value refers to the free energy of the oligo sequence binding to its perfect complement. A delta G value of -13.25 kcal/mol represents essentially no binding (no formation of the dimer structure). [0159] FIGS. 4A-4D provide exemplary editing of and/or correction of mutant HEB exon 1. [O 160] FIG. 4A shows a schematic of strategy for editing and correcting mutant HEB exon l ( e.g., containing a E-to-V mutation). Editing of HEB exon 1 exemplified in Examples 16-17 may be used as part of the strategy. Numbered boxes represent HEB exons of the indicated exon number. The checker box represents a mutation in HEB exon 1. Each open rectangle above HEB exon 1 represents a sgRNA portion comprising a target-complementary sequence (e.g., SEQ ID NO: 25, 45, 47, or 49) which may hybridize to HEB exon 1. Each filled rectangle adjacent thereto indicates where the PAM sequence is in the target DNA. A DNA template (e.g., ssODN) for correcting back to WT or non- disease causing exon 1 may be added. Upon hybridization of a sgRNA, spCas9 may cause cleavage at the target site and the DNA template may mediate HDR, resulting in a correction (slashed box) in HEB exon l . Correction of mutant HEB exon 1 may result in production of WT or corrected beta- globin, thereby restoring normal HbA production. [0161] FIG. 4B provides exemplary results obtained in Example 16. HEK293 cells were treated with TCV-encapsulated Cas9-based RNPs comprising sgRNA targeting a control gene (luciferase; using SEQ ID NO: 55) or targeting HEB exon 1 (using SEQ ID NO: 45or SEQ ID NO: 47). The graph shows percent HBB editing efficiency at the target sites. N=3 per treatment condition. One way ANOVA: p<0.0001 Dunnett's multiple comparison test: ***p=0.0001, ****p<0.0001. [0162] FIG. 4C provides exemplary results obtained in Example 17. HEK293 cells were treated with increasing amounts ofTCV-encapsulated Cas9-based RNPs comprising sgRNA targeting a control gene (luciferase; using SEQ ID NO: 55) or targeting HEB exon I (using SEQ ID NO: 45). The top graphs show percent HEB editing efficiency at the target site. N=3 per treatment condition. Two way ANOV A: treatment p<0.0001, dose p<0.0001, interaction p<0.0001. Tukey's multiple comparison test : ****p<0.0001. The bottom graph shows dose-dependent changes in the percent editing efficiency at the target site in HEB. (Linear regression: Luciferase: Y = 0.003217*X + 0.3775, HBB E6V _IA: Y = 0.1933*X + 11.96. Slopes are significantly different p<0.0001). [0163] FIG. 4D provides exemplary results obtained in Example 16 (16-2). HEK293 cells were treated with TCV-encapsulated Cas9-based RNPs comprising sgRNA targeting a control gene (HBG promoter) or HEB exon 1 (using SEQ ID NO: 45), with or without the ssODN named "HBB_E6V 80bp F" (SEQ ID NO: 181 ), "HBB _ E6V 80bp R" (SEQ ID NO: 182), "HBB _ E6V 100bp F" (SEQ ID NO: 183), "HBB_E6V l00bp R" (SEQ ID NO: 184), or "HBB_E6V _PAM 1 l00bp R" (SEQ ID NO: 185). The left graph shows percent HBB editing efficiency at the target site assessed using the TIDE analytical tool (N=3 per treatment condition: One-way ANOV A: p<0.0001. Dunnett's multiple comparison: ****p<0.0001). The right graph shows percent HDR efficiency assessed using TIDER (N=3 per treatment condition: One-way ANOV A: p=0.0197. Dunnett's multiple comparison: *p=0.0337, **p=0.0077). DETAILED DESCRIPTION OF THE INVENTION [0164] The present disclosure provides, among other things, compositions and methods for preventing, ameliorating, and/or treating SCD. The present disclosure also provides compositions and methods for effecting gene editing and/or gene expression alteration in vivo in cells of bone marrow origin and/or cells in the bone marrow. [0165] Targets [0166] Target diseases [0167] In one aspect, a target disease according to the present disclosure may comprise a disease involving cells of bone marrow. In some embodiments, the disease may comprise SCD. In some embodiments, the disease may comprise sickle cell anemia (SCA), Sickle cell-hemoglobin C (HbSC), and HbS ~-thalassaemia (also called ~-thalassaemia). [0168] Target cells [O 169) In one aspect, a target cell or target cells according to the present disclosure may comprise a cell or cells of bone marrow origin. In some embodiments, the target cell or target cells may comprise a cell or cells in the bone marrow origin. In some embodiments, the target cell or target cells may comprise a cell or cells capable of differentiating into a RBC. In some embodiments, the target cell or target cells may comprise HSCs, HSPCs, MPPs, CMPs, MEPs, HPCs, erythroid progenitors (e.g., BFU-E, CFU-E), proerythroblasts, erythroblasts (basophilic erythroblasts, early erythroblasts (e.g., type I, type 11), polychromatic erythroblasts, intermediate erythroblasts, acidophilic erythroblasts, late erythroblasts, normoblasts, reticulocytes before nucleus expulsion, reticulocytes, or erythrocytes, or any combinations thereof. In particular embodiments, the target cell or target cells may comprise HSCs and/or HSPCs. In particular embodiments, the target cell or target cells may comprise cells that are CD34+. [0170) Target genes [0171) In one aspect, a target gene according to the present disclosure may be a gene associated with a target disease. In some embodiments, a target gene according to the present disclosure may be edited via any appropriate technique. In some embodiments, the expression (e.g., protein and/or mRNA level) of a target gene according to the present disclosure may be modified via any appropriate technique. [O 172] In some embodiments, the target gene may be a SCD-associated gene including a regulatory element thereof, e.g., a region of a promoter or enhancer of such a gene. [0173] In some embodiments, the target gene may comprise a gene encoding a hemoglobin component such as beta-globin (the HEB gene) and/or a regulatory element thereof, e.g., a region of a promoter or enhancer of HEB. The generic sequence of human HEB may comprise the nucleic acid sequence corresponding to the nucleotide positions 5225464 to 5227071 of chromosome 11 (according to Gene Assembly GRCh38.pl3). In some embodiments, the HEB gene may be the HbS variant of HEB, the causative gene ofSCD. The HbS variant of HEB (coding strand) may comprise the nucleic acid sequence of SEQ ID NO: 21, which encodes the HbS variant of beta-globin having the amino acid sequence of SEQ ID NO: 2. In some embodiments, the HEB gene may be the HbC variant of HEB, which in the homozygous state can cause mild chronic hemolysis, splenomegaly, and jaundice. The HbC variant of HEB ( coding strand) may comprise the nucleic acid sequence of SEQ ID NO: 31, which encodes the HbC variant ofbeta-globin having the amino acid sequence of SEQ ID NO: 3. In some embodiments, the target gene may comprise a gene encoding a gene product (e.g., transcription factor) that directly or indirectly regulates or a DNA region (e.g., promoter, enhancer, transcription factor-binding site) that directly or indirectly regulates the expression of the HBB gene. [0174] In some embodiments, the target gene may comprise a gene encoding a gene product that regulates hemoglobin switching and/or erythropoiesis. (0175) In some embodiments, the target gene may comprise a gene encoding BAF chromatin remodeling complex subunit BCLl lA (the BCLJ IA gene) and/or a regulatory element thereof, e.g., a region of a promoter or enhancer of BCLJ I A. The generic sequence of human BCLJ I A may comprise the nucleic acid sequence corresponding to the nucleotide positions 60450520 to 60553654 of chromosome 2 (according to Gene Assembly GRCh38.pl3), which encodes the amino acid sequence of SEQ ID NO: 6. In some embodiments, the target gene may comprise a gene encoding a gene product (e.g., transcription factor) that directly or indirectly regulates or a DNA region (e.g., promoter, enhancer, transcription factor-binding site) that directly or indirectly regulates the expression of the BCLI I A gene. (0176] In some embodiments, the target gene may comprise a gene encoding Kruppel like factor 1 (the KLFI gene) and/or a regulatory element thereof, e.g., a region of a promoter or enhancer of KLFI. The generic sequence of human KLFI may comprise the nucleic acid sequence corresponding to the nucleotide positions 12884422 to 12887201 of chromosome 19 (according to Gene Assembly GRCh38.pl3), which encodes the amino acid sequence of SEQ ID NO: 7. In some embodiments, the target gene may comprise a gene encoding a gene product (e.g., transcription factor) that directly or indirectly regulates or a DNA region (e.g., promoter, enhancer, transcription factor-binding site) that directly or indirectly regulates the expression of the KLF I gene. (0177] In some embodiments, the target gene may comprise a gene encoding human hemoglobin subunit gamma 1 (the HBGI gene) and/or a regulatory element thereof, e.g., a region of a promoter or enhancer of HBGI. The generic sequence of human HBGJ may comprise the nucleic acid sequence corresponding to the nucleotide positions 5248269 to 5249857 of Chromosome 11 ( according to Gene Assembly GRCh38.p14), which encodes the amino acid sequence of SEQ ID NO: 8. In some embodiments, the target gene may comprise a gene encoding a gene product (e.g., transcription factor) that directly or indirectly regulates or a DNA region (e.g., promoter, enhancer, transcription factor-binding site) that directly or indirectly regulates the expression of the HBGI gene, preferably the BCLl lA-binding site in the promoter. [0178] In some embodiments, the target gene may comprise a gene encoding human hemoglobin subunit gamma 2 (the HBG2 gene) and/or a regulatory element thereof, e.g., a region of a promoter or enhancer of HBG2. The generic sequence of human HBG2 may comprise the nucleic acid sequence corresponding to the nucleotide positions 5253188 to 5254781 of Chromosome 11 (according to Gene Assembly GRCh38.p14), which encodes the amino acid sequence of SEQ ID NO: 9. In some embodiments, the target gene may comprise a gene encoding a gene product (e.g., transcription factor) that directly or indirectly regulates or a DNA region (e.g., promoter, enhancer, transcription factor-binding site) that directly or indirectly regulates the expression of the HBGI gene, preferably the BCLl 1 A-binding site in the promoter. (0179] In some embodiments, the target gene may be SOX6, GATA], NF-E4 (or NFE4), COUP-TF, NR2CI (also known as TR2), NR2C2 (also known as TR4), genes encoding members of the MBD2 protein complex, IKZFI (also known as Jkaros), genes encoding other members of PYR complex (CHD4, HDAC2, RBBP7, SMARCBJ, SMARCCJ, SMARCC2, SMARCDJ, and SMARCEJ), or BRGJ, or a gene that directly or indirectly modulate the expression thereof, or any combination thereof. [0180] Target sequences [0181] In one aspect, any parts of a target gene according to the present disclosure may be targeted (i.e., may be a target sequence), and the target sequence may be any parts of the sequence of the coding (sense) strand or the non-coding (antisense) strand of the gene or its transcript (including pre- and post- splicing sequences), any parts of the sequence of the coding region or non-coding region of the gene or its transcripts, or any parts of the sequence of the polynucleotide regions regulating the expression of the gene (e.g., promoter region, enhancer region, transcription factor-binding site). [0182] For example, when the target gene is the HbS variant of human HEB, in some embodiments, the target sequence may be any parts of the nucleic acid sequence corresponding to the nucleotide positions 5225464 to 5227071 of chromosome 11 (according to Gene Assembly GRCh38.pl 3) or its transcript (including pre- and post- splicing sequences). In some embodiments, the target sequence may be any parts of the nucleic acid sequence of SEQ ID NO: 21. In some embodiments, the target sequence may be any parts of the nucleic acid sequence of SEQ ID NO: 22. In some embodiments, the target sequence may be any parts of the nucleic acid sequence of SEQ ID NO: 23. In certain embodiments, the target sequence may be the nucleic acid sequence of SEQ ID NO: 44, 46, 48, or 24 or a variant thereof. [0183] For example, when the target gene is human BCLJ JA, in some embodiments, the target sequence may be any parts of the nucleic acid sequence corresponding to the nucleotide positions 60450520 to 60553654 of chromosome 2 (according to Gene Assembly GRCh38.pl3) or its transcript (including pre- and post- splicing sequences). In certain embodiments, the target sequence may be within or may overlap with intron 2 of human BCLJ JA, preferably within or overlapping with the erythroid-enhancer region (EER) therein, such as but not limited to the EER region shown in FIG. 2F. e.g., SEQ ID NO: 4 or 5 or 14 or 15. In particular embodiments, the target sequence may be the nucleic acid sequence of SEQ ID NO: 70, 276, 278, 270, 272, 274, 64, 68, or 66 or a variant thereof. [0184] For example, when the target gene is human KLFJ, in some embodiments, the target sequence may be any parts of the nucleic acid sequence corresponding to the nucleotide positions 12884422 to 12887201 of chromosome 19 (according to Gene Assembly GRCh38.p13) or its transcript (including pre- and post- splicing sequences). In certain embodiments, the target sequence may be the nucleic acid sequence of SEQ ID NO: 74 or 76 or a variant thereof. [0185] For example, when the target gene is human HBG 1, in some embodiments, the target sequence may be any parts of the nucleic acid sequence corresponding to the nucleotide positions 5248269 to 5249857 of Chromosome 11 (according to Gene Assembly GRCh38.p14) or a promoter region of human HBGJ such as the region shown in FIG. 3F top or its transcript (including pre- and post- splicing sequences), e.g., SEQ ID NO: 95 or 97. In certain embodiments, the target sequence may be the nucleic acid sequence of SEQ ID NO: 86, 286, 288, 280, 282, 284, or 84 or a variant thereof. [0186] For example, when the target gene is human HBG2, in some embodiments, the target sequence may be any parts of the nucleic acid sequence corresponding to the nucleotide positions 5253188 to 5254781 of Chromosome 11 (according to Gene Assembly GRCh38.p14) or a promoter region of human HBG2 such as the region shown in FIG. 3F bottom or its transcript (including pre- and post- splicing sequences), e.g., SEQ ID NO: 95 or 97, or 295 or 297. In certain embodiments, the target sequence may be the nucleic acid sequence of SEQ ID NO: 86, 286, 288, 280, 282, 284, or 84 or a variant thereof. [0187] Gene editing [0189] In one aspect, a target gene according to the present disclosure may be edited in vivo. Gene editing may be effected by any appropriate techniques. In some embodiments, gene editing may be mediated by a nuclease, endonuclease, meganuclease, zinc finger nuclease (ZFN), transcription activator-like nuclease (T ALEN), or Cas nuclease. In some embodiments, gene editing may result in at least one nucleic acid insertion, deletion, or replacement (e.g., resulting in a nonsense, missense, or silent mutation) in the target gene, such as SCD-associated gene, such as BCLJ JA, HBGJ, HBG2, HEB, or KLFJ. [0190] Cas nucleases [0191] When the CRISPR/Cas system is used for gene editing, any appropriate Cas nucleases may mediate gene editing. In some embodiments, a Cas nuclease (as a protein) or a Cas nuclease-encoding polynucleotide (e.g., DNA or RNA) may be used. In some embodiments, the Cas nuclease may be Cas12a or Cpfl, Cas 9, Cas3, Cas8a2, Cas8b, Cas8c, Casl0, Csxl 1, Cas12, Cas12b, Cas12f, Cas12j, Cas13, Cas13a, Cas14, C2cl, C2c3, or C2c2. In some embodiments, the Cas nuclease may be a class 2 Cas nuclease. In some embodiments, the Cas nuclease may be a type V or type VI Cas nuclease. In certain embodiments, the Cas nuclease is Cas 12a. In certain embodiments, the Cas 12a may be of Acidaminococcus sp. (AsCas 12a), Lachnospiraceae bacterium (LbCas 12a), Francisella novicida (FnCas12a), Moraxella bovoculi (MbCas12a), Coprococcus eutactus (CeCas12a), Butyrivibrio jibrisolvens (BfCas12a) or any engineered variants thereof, including AsCas12a Ultra, enAsCas12a, enAsCas12a-Hifi, enAsCas12a Ultra, enAsCas12a-Hifi Ultra, and LbCas12a Ultra (Zhang et al., Nat Commun. 2021 Jun 23;12(1):3908.; Kleinstiver et al., Nat Biotechnol. 2019 March; 37(3): 276-282.). In certain embodiments, the Cas nuclease is Cas9. In certain embodiments, the Cas9 may be Cas9 of Streptococcus pyogenes (SpCas9), Staphylococcus aureus Cas9 (SaCas9), Streptococcus thermophilus (StCas9), Neisseria meningitidis (NmCas9), Francisella novicida (FnCas9), Campylobacter jejuni (CjCas9), Streptococcus canis (ScCas9), Staphylococcus auricularis (SauriCas9), or any engineered variants thereof, including SaCas9-HF, SpCas9-HF1, KKHSaCas9, eSpCas9, HypaCas9, Fokl-Fused dCas9, xCas9, SpRY (variant ofSpCas9), or SpG (variant of SpCas9). Cas nuclease of different bacterial origins often recognize different PAM sequences and/or different cleavage accuracy or specificity. In some cases, the type ofCas nuclease to use may be selected based on the presence or absence or a certain PAM sequence in the target gene. [0192] In certain embodiments, any of the Cas nucleases may be expressed using a nuclear localization signal (NLS) sequence. In some cases, the NLS may be placed at the N-terminal side, the C-terminal side, or both sides of the Cas nuclease. The NLS may have any appropriate sequence which allows expression of the Cas in the nucleus. In some cases, the NLS may have the sequence of SEQ ID NO: 291 or 292, optionally placed at the C-terminal side of the Cas nuclease. [0193] Guide RNA [0194] When the CRISPR/Cas system is used for gene editing, the gRNA may be designed based on the sequence of the target gene and the PAM sequence recognized by the Cas nuclease to be used. [0195] When Cas12a for example of or derived from Acidaminococcus sp. (AsCas12a), Lachnospiraceae bacterium (LbCas12a), Francisella novicida (FnCas12a), Moraxella bovoculi (MbCas12a), Coprococcus eutactus (CeCas12a), or Butyrivibriofibrisolvens (BfCas12a) is used, the target-complementary sequence of a gRNA may be designed, for example, as the 21 ( or alternatively about 17-27 or about 20-24) nucleotides immediately downstream (the 3'-side) of any of the 5'- TTTV-3' (V may be A, C, or G), or in some cases 5'-TTTN-3' (N may be any nucleic acid) or 5'- TTN-3' (N may be any nucleic acid, i.e., A, T, C, or G) PAM sites present in the target gene (the coding strand or non-coding strand). The same target-complementary sequence may also be used with other Cas 12a homologs and orthologs or variants thereof which recognize the same PAM, e.g., Cas12b (such as Cas12b of Bacillus hisashii (BhCasl2b)), Cas12f, Un1Cas12fl (Harrington et al., Science. 2018 Nov 16;362(6416):839-842.), or CasMINI (Xu et al., Mol Cell. 2021 Oct 21 ;81(20):4333-4345.e4.), or another shorter but similar PAM such as 5'-TTN-3' (N may be any nucleic acid), e.g., Cas 12b (such as Cas12b of Alicyclobacillus acidophilus (AacCasb )) or Cas 12j. [0196] When Cas9 of Streptococcus pyogenes (SpCas9) or Francisella novicida (FnCas9) is used, the target-complementary sequence of a gRNA may be designed, for example, as the 20 ( or alternatively about 17-24) nucleotides immediately upstream (the 5'-side) of any of the 5'-NGG-3' (N may be any nucleic acid) PAM sites present in the target gene (the coding strand or non-coding strand). [0197] A desired target-complementary sequence may be selected from all possible sequences, for example based on, the proximity to the desired editing position, the G-C content (e.g., for example in the range of about 40-80%), self-complementarity, the potential editing efficiency, and/or the potential off-target effects. Non-limiting tools for selecting a desired target-complementary sequence include https://chopchop.cbu.uib.no/. [0198] Exemplary target-complementary sequences of a gRNA which may be used for example with Cas 12a nucleases ( e.g., AsCas 12a or LbCas 12a or) or in some cases any other Cas nucleases which recognizes the PAM sequence of 5'-TTTN-3' for targeting human BCLJ JA (or an EER thereof) include but are not limited to: SEQ ID NO: 71, which may target the target sequence of SEQ ID NO: 70 or a variant thereof; SEQ ID NO: 277, which may target the target sequence of SEQ ID NO: 276 or a variant thereof; and SEQ ID NO: 279, which may target the target sequence of SEQ ID NO: 278 or a variant thereof. [0199] Exemplary target-complementary sequences of a gRNA which may be used for example with Cas9 nucleases (e.g., SpCas9) or in some cases any other Cas nucleases which recognizes the PAM sequence of 5'-NGG-3' for targeting human BCLJ JA include but are not limited to SEQ ID NOS: 271,273,275, 65, 69, and 67, which may target the target sequence of SEQ ID NOS: 270,272,274, 64, 68, and 66, respectively, or a variant thereof. [0200] Exemplary target-complementary sequences of a gRNA which may be used for example with Cas9 nucleases (e.g., SpCas9) or in some cases any other Cas nucleases which recognizes the PAM sequence of 5'-NGG-3' for targeting human HBGJ and/or HBG2 (or a promoter region thereof) include but are not limited to: SEQ ID NO: 85, which targets the target sequence of SEQ ID NOS: 84 or a variant thereof; SEQ ID NO: 281, which targets the target sequence of SEQ ID NOS: 280 or a variant thereof; SEQ ID NO: 283, which targets the target sequence of SEQ ID NOS: 282 or a variant thereof; and SEQ ID NO: 285, which targets the target sequence of SEQ ID NOS: 284 or a variant thereof. [0201] Exemplary target-complementary sequences of a gRNA which may be used for example with Cas12a nucleases (e.g., AsCas12a or LbCas12a) or in some cases any other Cas nucleases which recognizes the PAM sequence of5'-TTTN-3' for targeting human HBGJ and/or HBG2 (or a promoter region thereof) include but are not limited to: SEQ ID NO: 87, which targets the target sequence of SEQ ID NOS: 86 or a variant thereof; SEQ ID NO: 287, which targets the target sequence of SEQ ID NOS: 286 or a variant thereof; and SEQ ID NO: 289, which targets the target sequence of SEQ ID NOS: 288 or a variant thereof;. [0202] Exemplary target-complementary sequences of a gRNA which may be used for example with Cas9 nucleases ( e.g., SpCas9) or in some cases any other Cas nucleases which recognizes the PAM sequence of 5'-NGG-3' for targeting human HEB (any variants) include but are not limited to: SEQ ID NOS: 25, 45, 47, and 49, which targets the target sequence of SEQ ID NOS: 24, 44, 46, and 48, respectively, or a variant thereof. [0203] Exemplary target-complementary sequences of a gRNA which may be used for example with Cas9 nucleases ( e.g., SpCas9) or in some cases any other Cas nucleases which recognizes the PAM sequence of 5 '-NGG-3' for targeting the HbS variant of human HEB include but are not limited to SEQ ID NOS: 25, which may target the target sequence of SEQ ID NOS: 24. [0204] Exemplary target-complementary sequences of a gRNA which may be used for example with Cas9 nucleases ( e.g., SpCas9) or in some cases any other Cas nucleases which recognizes the PAM sequence of5'-NGG-3' for targeting human KLFJ include but are not limited to SEQ ID NOS: 75 and 77, which may target the target sequence of SEQ ID NOS: 64 and 76, respectively. [0205] gRNA modifications [0206] In some embodiments, a gRNA according to the present disclosure may comprise one or more modifications. In some embodiments, the modification may be selected from the group consisting of: 2'-O-Cl-4alkyl such as 2'-O-methyl (2'-oMe), 2'-deoxy (2'-H), 2'-O-Cl-3alkyl-O-Cl-3alkyl such as 2'-methoxyethyl (2'-MOE), 2'-fluoro (2'-F), 2'-amino (2'-NH2), 2'-arabinosyl (2'-arabino) nucleotide, 2'-F-arabinosyl (2'-F-arabino) nucleotide, 2'-locked nucleic acid (LNA) nucleotide, 2'- unlocked nucleic acid (ULNA) nucleotide, a sugar in 1 form (I-sugar), and 4'-thioribosyl nucleotide. In some embodiments, the modification is an intemucleotide linkage modification selected from the group consisting of: phosphorothioate, phosphonocarboxylate, thiophosphonocarboxylate, alkylphosphonate, and phosphorodithioate. In some embodiments, the modification is selected from the group consisting of: 2-thiouracil (2-thioU), 2-thiocytosine (2-thioC), 4-thiouracil ( 4-thioU), 6- thioguanine (6-thioG), 2-aminoadenine (2-aminoA), 2-aminopurine, pseudouracil, hypoxanthine, 7- deazaguanine, 7-deaza-8-azaguanine, 7-deazaadenine, 7-deaza-8-azaadenine, 5-methylcytosine (5- methylC), 5-methyluracil (5-methylU), 5-hydroxymethylcytosine, 5-hydroxymethyluracil, 5,6- dehydrouracil, 5-propynylcytosine, 5-propynyluracil, 5-ethynylcytosine, 5-ethynyluracil, 5-allyluracil (5-ally!U), 5-allylcytosine (5-allylC), 5-aminoallyluracil (5-aminoally!U), 5-aminoallyl-cytosine (5- aminoallylC), an abasic nucleotide, Z base, P base, Unstructured Nucleic Acid (UNA), isoguanine (isoG), isocytosine (isoC), and 5-methyl-2-pyrimidine. In particular embodiments, a gRNA may comprise (i-1) 2'-O-methylation further optionally at first three and last three bases and/or (i-2) one or more 3' phosphorothioate bonds, further optionally between first three and last two bases. [0207] DNA templates [0208] In one aspect, when the CRISPR/Cas system is used for gene editing and when a gene knock- in or gene sequence correction (e.g., deletion of one or more particular nucleic acids) is desired, a DNA template (or simply referred to as a template or a repair template) comprising a desired mutation or sequence may further be provided so that a gene knock-in or gene sequence correction is effected based on the template sequence via the cells' endogenous DNA mechanisms. When Casl2a is used, Casl2a provides a staggered double-strand break (DSB) on a PAM distal site (e.g., between the 17 th and 18 th nt, between the 18 th and 19 th nt, or between the 19 th and 20 th nt of the target sequence) on the DNA with a several nt overhang (e.g., 4, 5, or 6 nt) on the target strand and if a template having homology arms is provided, homology-directed repair (HDR) will take place. When Cas9 is used, Cas9 provides a double-strand break (DSB) in the target gene between the third and fourth nucleotides upstream (5' side) of the 5'-NGG-3' and if a template having homology arms is provided, homology- directed repair (HDR) will take place. In some embodiments, a r template may be a single-stranded (ssODN) or double-stranded. [0209] In some embodiments, a template comprises or consists of a 5' homology arm, an optional central region, and a 3' homology arm. In some embodiments, a template may be approximately centered with respect to the DSB position. In some embodiments, the DSB position may be in the optional central region. In some embodiments, a homology arm may be less than about 30, about 25, about 20, about 20, about 15, or about 10 nt away from the DSB position. In particular embodiments, a homology ann may be about 10, about 8, about 6, about 4, about 3, about 2, or about 1 nt away from the DSB position. [021 O] In some embodiments, a DNA template may comprise a total length of approximately 40- 5000 nt. In some embodiments, the total length may be about 40-2000 nt, about 40-1000 nt, about 40- 500 nt, about 40-200 nt, about 60-100 nt, about 80-160 nt, about 100-140 nt, about 110- 130 nt, about 80 nt, about 100 nt, or about 120 nt. In certain embodiments, the total length may be about 70-90 nt or about 75-85 nt in length. In particular embodiments, the total length may be about 80 nt. In particular cases, the total length may be as about 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, or 85 nt in length. [0211] Any appropriate size of a homology arm may be used. In some embodiments, the 5' and 3' homology arms may have the same or similar nucleotide lengths (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nt difference). In some embodiments, the 5' and 3' homology arms may significantly differ in length. In some embodiments, the size of a homology arm may be approximately 20-2500 nucleotides (nt), about 20-1000 nt, about 20-500 nt, or about 20-100 nt. In particular embodiments, the size of a homology arm may be about 40-80 nt, about 50- 70 nt, or about 60 nt. In particular embodiments, the size of each homology arm may be about 40-80 nt, about 50- 70 nt, or about 60 nt. In some embodiments, 5' and/or 3' homology arms may be 100% complementary to the corresponding sequence in the original DNA sequence before gene editing or may have one or more (a few, for example, 2, 3, 4, or 5) mutations (e.g., silent mutation) relative to the corresponding sequence in the original DNA sequence before gene editing. [0212] In some embodiments, a template may have one or more mutations at one or more of the PAM positions. In some embodiments, such a mutation(s) helps prevent or reduce Cas-mediated cleavage of the template itself or of the DNA molecule post HDR. In case of a ssODN, such a mutation may be within the PAM bases or the reverse (or antisense) bases, i.e., the opposite strand) and/or at one or more of the 5 '-neighbouring bases of the PAM ( or the 3 '-neighbouring bases of the reverse (or antisense) sequences corresponding to the PAM). In some embodiments, a ssODN may comprise complementarity to the gRNA strand. [0213] In some embodiments, when the target gene is BCLJ JA and the target sequence is within the EER, such as the region shown in FIG. 2F, any DNA template which promotes mutagenesis at the cut site may be used. In certain embodiments, a DNA template may comprise a ssODN comprising or consisting of a nucleic acid sequence having at least 80%, at least 85%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% complementarity to the sequence of a segment within BCLJ 1 A or a variant thereof and/or the erythroid-enhancer region (EER) of BCLJ JA and/or a promoter or enhancer region of BCLJ JA. In some cases, the segment comprises the target sequence and/or the sequence complementary thereto. In some cases, the segment is about 20-300 nt, about 25-200 nt, about 30-160 nt, about 40-140 nt, about 50-120 nt, about 60-100 nt, about 70-90 nt, or about 75-85 nt in length. In some cases, the segment is about 20 nt, about 25 nt, about 30 nt, about 35 nt, about 40 nt, about 45 nt, about 50 nt, about 55 nt, about 60 nt, about 65 nt, about 70 nt, about 75 nt, about 80 nt, about 85 nt, about 90 nt, about 95 nt, about 100 nt, about 105 nt, about 120 nt, about 125 nt, about 130 nt, about 135 nt, about 140 nt, about 145 nt, about 150 nt, about 155 nt, about 160 nt, about 175 nt, about 180 nt, about 185 nt, about 190 nt, about 195 nt, about 200 nt, about 205 nt, about 220 nt, about 225 nt, about 230 nt, about 235 nt, about 240 nt, about 245 nt, about 250 nt, about 255 nt, about 260 nt, about 275 nt, about 280 nt, about 285 nt, about 290 nt, about 295 nt, or about 300 nt in length. In particular embodiments, the DNA template may comprise a nucleic acid sequence at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to SEQ ID NO: 305, 306, 303, 304, 301, or 302. In particular cases, the DNA template may comprise a nucleic acid sequence at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to SEQ ID NO: 305 or 306. In certain embodiments, a DNA template which comprises a first strand comprising a ssODN sequence described above and a second strand complementary to the first strand may be used. [0214] In some embodiments, when the target gene is HBGJ and/or HBG2 and the target sequence is within the promoter region of HBGJ and/or HBG2, such as the region shown in FIG.3F, which contains the BCLl IA-binding motif 5'-TGACC-3', alteration (e.g., deletion or another type of mutation) of one of more of the five nucleotides in the BCLl 1 A-binding motif may be desired. Without wishing to be bound by theory, such an alteration may block or reduce binding of the BCLl lA (a repressor of HBGI and/or HBG2) thereby recovering the expression offetal globin. [0215] In certain embodiments, the 5' homology arm may comprise part of the sequences present in the 5' side of the BCLl 1 A-binding motif ( e.g., part or all of SEQ ID NO: 195) or comprise one or more mutations relative to such a sequence. [0216] In some cases, the 5' homology arm may comprise at least 5, least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50 nucleotides, at least 55, at least 60, at least 65, at least 70, at least 75, or at least 80 nucleotides, about 5-80 nucleotides, about 10- 70 nucleotides, about 20-60 nucleotides, about 30-50 nucleotides, about 35-45 nucleotides, or about 40 nucleotides. In some cases, the 5' homology arm may end at the first, second, third, fourth, fifth, sixth, seventh, eighth, nineth, 10 1 ", 11 1 \ 12 1 \ 13 1 \ 14 th , 15 th , 16 th , 17 1 ", 18 1 '\ 19 th , or 20 th nucleotide counting from the 3'-end of SEQ ID NO: 195 or a more upstream nucleotide within SEQ ID NO: 195. In some cases, the 5' homology arm may comprise from the l 5', 2 nd, 3 rd, 4 1\ 5 th, or 6 th to the 35 1", 36 th, 37 th, to 38 1 \ 39 th , or 40 th nucleotides counting from the 3 '-end of SEQ ID NO: 195. In particular cases the 5' homology arm may comprise the I" to the 37 111 nucleotides counting from the 3 '-end of SEQ ID NO: 195. [0217] In some cases, the optional central region is absent (i.e., contains zero nt). In some cases, the optional central region comprises a sequence of one, two, three, five, six, seven, eight, nine, ten, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more nucleotides. In some cases, the full sequence of the central region is not identical to 5'-TGACC-3'. In some cases, the central region may lack at least one nucleotide from the 5' -TGACC-3' motif sequence. In some cases, the full sequence of the central region is 5'-TGAC-3', 5'-TGCC-3', 5'-TACC-3', 5'-GACC-3', 5'-TGA-3', 5'-TGC-3', 5'-TAC-3', 5'-GAC-3', 5'-TCC-3', 5'-GCC-3', 5'-ACC-3', 5'-TG-3', 5'-TA-3', 5'-TC-3', 5'-GA-3', 5'-GC-3', 5'-AC-3', 5'-CC-3', 5'-T-3', 5'-G-3', 5'-A-3', or 5'-C-3'. In some cases, the central region may contain one or more nucleotides inserted in between bases within the 5'-TGACC-3' sequence. In particular cases the central region may consist of the sequence of 5'-TGAC-3'. [0218] In some cases, the 3' homology arm may comprise at least 5, least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50 nucleotides, at least 55, at least 60, at least 65, at least 70, at least 75, or at least 80 nucleotides, about 5-80 nucleotides, about 10- 70 nucleotides, about 20-60 nucleotides, about 30-50 nucleotides, about 35-45 nucleotides, or about 40 nucleotides. In some cases, the 3' homology arm may start at the first, second, third, fourth, fifth, sixth, seventh, eighth, nineth, 10 11 1, 11 11 1, 12 11 1, 13 th , 14 11 1, 15 11 1, 16t", 17 th , 18 111 , 19 11 1, or 20 th nucleotide counting from the 5' -end of SEQ ID NO: 196 or a more downstream nucleotide within SEQ ID NO: 196. In some cases, the 3' homology arm may comprise from the l ", 2 nd, 3 rd, 4 1\ 5 th , or 6 th to the 35 th , to 36 th , 37t", 38 11 1, to 39 th , 4ot11, 41 st, 42 nd , 43 rd , 44 th , or 45 th nucleotides counting from the 5' -end of SEQ ID NO: 196. In particular cases, the 3' homology arm may comprise the l " to the 40 th nucleotides counting from the 5'-end of SEQ ID NO: 196. [0219] In certain embodiments, the DNA template may be a ssODN comprising any of the above- mentioned 5' homology arm, any of the optional central region, and any of the 3' homology arm in a direction from the 5' to the 3'. In certain embodiments, the DNA template may be a ssODN which is reverse complement of such a ssODN. In certain embodiments, the DNA template may be a dsODN comprising any of such ssODN and another ssODN reverse complement thereof. [0220] In certain embodiments, the DNA template is designed to delete 13 nucleotides corresponding to the 13-nt HPFH deletion or at least a partial sequence thereof. In particular embodiments, the DNA template may comprise a ssODN of SEQ ID NO: 191 or 192, or at least a partial sequences thereof (e.g., lacking 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides in the 5'- and/or 3'- end thereof). In particular embodiments, the DNA template may comprise a dsODN having a strand of SEQ ID NO: 191 or at least a partial sequences thereof(e.g., lacking 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides in the 5'- and/or 3' - end thereof) and a strand of SEQ ID NO: 192 or at least a partial sequences thereof (e.g., lacking 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides in the 5'- and/or 3'- end thereof), [0221] In some embodiments when the target gene is the HbS variant of HEB, a knock-in of wild type HEB may be desired. [0222] In some embodiments, a ssODN encoding part of the wildtype beta-globin, or a ssODN comprising a sequence complementary thereto may be used. In certain embodiments, the part of the wildtype beta-globin may be encoded by the wildtype nucleic acid sequence. In certain embodiments, the part of the wildtype beta-globin may be encoded by a nucleic acid sequence comprising one or more mutations (e.g., silent mutation(s)) relative to the wildtype nucleic acid sequence. [0223] In some embodiments, the ssODN may comprise a central region having the sequence of 5' - CTCA-3', 5'-TTCA-3', 5'-CTCT-3', 5'-TTCT-3', 5'-CTCC-3', 5'-TTCC-3', 5'-CTCG-3', or 5'- TTCG-3'. Any of these central region sequences, once knocked-in, would con-ect the glutamate-to- valine ("E-to-V") SCD-causing amino acid substitution back to the wild type glutamate. [0224] In some embodiments, the ssODN may comprise a 5' homology arm having the sequence of SEQ ID NO: 112. Alternatively, a 5' homology arm may have part of the 3' sequence of SEQ ID NO: 112. In some embodiments, a 5' homology arm may have any length of20nt or longer counting from the 3' end of SEQ ID NO: 112. In certain embodiments, a 5' homology arm may have a length of at least 20, at least 30 (such as 39), at least the 40 (such as 49), or at least 50 (such as 50 or 59) nt counting from the 3'-end of SEQ ID NO: 112. In some embodiments, the 5' homology arm may further comprise one or more mutations, preferably silent mutation(s), relative to such 5' homology arm sequences. [0225] In some embodiments, the ssODN may comprise a 3' homology arm having the sequence of SEQ ID NO: 122. Alternatively, a 3' homology arm may have part of the 5' sequence of SEQ ID NO: 122. In some embodiments, a 3' homology arm may have any length of20nt or longer counting from the 5' end of SEQ ID NO: 122. In certain embodiments, a 3' homology arm may have a length of at least 20, at least 30 (such as 37), at least the 40 (such as 47), or at least 50 (such as 57) nt counting from the 5'-end of SEQ ID NO: 122. In some embodiments, the 3' homology arm may further comprise one or more mutations, preferably silent mutation(s), relative to such 3' homology arm sequences. [0226] In particular embodiments, the ssODN may comprise, essentially consist of or consist of the sequence of any one of SEQ ID NOS: 101-108. [0227] In further embodiments, the ssODN may comprise a sequence complementary to any of the sequence of ssODNs for correcting back to a wildtype beta globin-encoding DNA sequence described herein. [0228] In some embodiments, when a DNA template, a double-stranded DNA template may also be used instead. In such a case, one of the strands of the template may comprise the same sequence as a desired ssODN and the other strand have a sequence complementary thereto. [0229] In some embodiments, a template may also be used when Cas nucleases are used to disrupt ( e.g., cause deletion of one or more nucleotides, or one or more nucleotide insertions or alterations in the nucleotide sequence) a target gene or a target gene region (including a regulatory region such as a promoter region) without necessarily relying on HDR. [0230] In certain embodiments, a template which is not complementary to or has low complementarity to the target site (the sequences flanking the site to be cleaved by the Cas nuclease used). Without wishing to be bound by theory, use of such a non-complementary template may enhance the efficiency of causing deletion of one or more nucleotides or one or more nucleotide insertions or alterations in the nucleotide sequence (e.g., thereby disrupting the function (e.g., promoter binding) of the nucleotide sequence before editing or causing early appearance of a terminator or stop codon). In certain embodiments, a template may have less than 90%, less than 85%, less than 80%, less than 75%, less than 70%, less than 65%, less than 60%, less than 55%, less than 50%, less than 45%, less than 40%, less than 35%, less than 30%, less than 30%, less than 25%, less than 20%, less than 15%, or less than 10% complementarity to the sequence of a segment within the target gene, wherein the segment comprises the target sequence ( contained in the gRNA to be used with the Cas nuclease) and/or the sequence complementary thereto and is about 20-300 nt, about 25- 200 nt, about 30-160 nt, about 40-140 nt, about 50-120 nt, about 60-100 nt, about 70-90 nt, about 75- 85 nt, about 20 nt, about 25 nt, about 30 nt, about 35 nt, about 40 nt, about 45 nt, about 50 nt, about 55 nt, about 60 nt, about 65 nt, about 70 nt, about 75 nt, about 80 nt, about 85 nt, about 90 nt, about 95 nt, about 100 nt, about 105 nt, about 120 nt, about 125 nt, about 130 nt, about 135 nt, about 140 nt, about 145 nt, about 150 nt, about 155 nt, about 160 nt, about 175 nt, about 180 nt, about 185 nt, about 190 nt, about 195 nt, about 200 nt, about 205 nt, about 220 nt, about 225 nt, about 230 nt, about 235 nt, about 240 nt, about 245 nt, about 250 nt, about 255 nt, about 260 nt, about 275 nt, about 280 nt, about 285 nt, about 290 nt, about 295 nt, or about 300 nt in length. Such a template may be a ssODN or a dsODN. In some cases, the template may be about 30-160 nt, 50-120 nt, about 60-100 nt, about 70-90 nt, about 75-85 nt, about 80 nt in length. [0231] For example, the complementarity may be analyzed by alignining the template sequence with an about 120-nt, about 100-nt, about 90 nt, about 85-nt, about 80-nt, about 75-nt, or about 70-nt sequence of the target gene of interest encompassing the target sequence or a reverse complement thereof. In some cases, complementarity/alignment may be analyzed using Blast®N or Nucleotide BLAST® or CLUSTAL software (e.g., https://www.ebi.ac.uk/Tools/msa/clustalo/). In certain cases, a complementarity/alignment analysis using Blast®N or Nucleotide BLAST®may give "no significant alignment", e.g., defined bye-value< 0.05. In some cases, complementarity may be analyzed by a heterodimer analysis. In some cases, complementarity may be analyzed by culculating the delta G value for a dimer (hypothetical dimer) of the isolated ODN and an about 120-nt, about 100-nt, about 90 nt, about 85-nt, about 80-nt, about 75-nt, or about 70-nt sequence of the target gene of interest encompassing the target sequence or a reverse complement thereof. In certain cases, the delta G value may be about -100 kcal/mo! or higher, about -80 kcal/mo! or higher, about -60 kcal/mo! or higher, about -50 kcal/mo! or higher, about -40 kcal/mo! or higher, about -30 kcal/mo! or higher, about -20 kcal/mo! or higher, about -18 kcal/mo! or higher, about -16 kcal/mo! or higher, about -15 kcal/mo! or higher, about -14 kcal/mo! or higher, about -12 kcal/mo! or higher, or about -10 kcal/mo! or higher, because, without wishing to be bound by theory, such delta G values would indicate that such a hypothetical dimer would unlikely form . [0232] In particular embodiments, the template may be a ssODN which comprises or consists of SEQ ID NO: 190 (a sequence at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, identical thereto) or a sequence reverse complementary thereto or a dsODN comprising a strand of SEQ ID NO: 190 (a sequence at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, identical thereto) and a strand reverse complementary thereto. [0233] In particular embodiments, the target gene may be a SCD-associate gene, such as but not limited to HBGl, HBG2, BCLl IA, and KLFl. [0234] In particular embodiments, the target gene of interest is not human huntingtin. In particular embodiments, the target gene of interest is not huntingtin. In particular cases, the target sequence is not within an about 120-nt, about 100-nt, about 90-nt, or about 80-nt sequence encompassing the junction between intron 12 and exon 13 of human huntingtin. In some particular cases, the target sequence is not within intron 12 and/or exon 13ofhuman huntingtin. [0235] Gene expression alteration [0236] Expression alteration tools [0237] In one aspect, the expression of a target gene according to the present disclosure maybe altered in vivo. Gene expression may be altered by any appropriate techniques. In some embodiments, gene expression may be altered via a nucleic acid molecule, such as but not limited to: a RNA (single or double stranded), a siRNA, a shRNA, a miRNA, a mRNA, a DNA (single or double stranded), a plasmid DNA, a cDNA, and/or a locked nucleic acid. Gene expression alteration may be effected alone or in combination with gene editing described herein. [0238] When reduced expression of a target gene is desired: In some embodiments, the target gene expression may be altered via RNA interference. For example, in some embodiments, an siRNA or shRNA specific to the transcript sequence of a target gene may be used. In some embodiments, a miRNA which negatively modulates the target gene expression may be introduced. In further embodiments, an RNA or DNA molecule encoding a gene that negatively modulates the target gene expression (e.g., repressor of the target gene) may be introduced, via transduction or transfection. [0239] When increased expression of a target gene is desired: In some embodiments, the target gene expression may be altered via forced expression of the gene, via transduction or transfection of a nucleic acid molecule encoding the gene. In some embodiments, the target gene expression may be altered via transduction or transfection of a nucleic acid molecule encoding a gene that positively modulates the target gene expression (e.g., a transcription factor). In further embodiments, a negative regulator of the target gene may be silenced via RNA interference, using e.g., an siRNA or shRNA. [0240] Delivery vehicles [0241] Vehicle type [0242] In one aspect, any components that may be used for effecting gene editing and/or gene expression alteration as described herein may be carried into as a cargo (or cargoes) into a cell by a delivery vehicle. Such a delivery vehicle may be a transfection competent vehicle (TCV). [0243] Lipid-based TCVs [0244] TCVs particularly used in the present disclosure include lipid-based TCVs. Compared to non- lipid-based TCVs such as viral vectors, lipid-based TCVs may have several advantages, e.g., less immunogenicity if needed, no random integration into the target cell genome. [0245] Ionizable cationic lipid [0246] In some embodiments, a lipid-based TCV may comprise at least one ionizable cationic lipid. In some embodiments, the at least one ionizable cationic lipid may comprise DODMA, DODAP, DLinDAP, KC2, MC3, ALC-0315, SM-102, DODAC, DDAB, DOTAP, DOTMA, DLinDMA, DLenDMA, DLin-C-DAP, DLin-DAC, DLin-MA, DLin-S-DMA, DLin-2-DMAP, DLin-TMA.Cl, DLin-T AR.Cl, DLin-MPZ, dLinAP, DOAP, DLin-EG-DMA, DLin-K-DMA, DLin-K-DMA or analogs thereof, ALNY-100, DOTMA, DOT AP.Cl, DC-Chol, DOSPA, DOGS", DMRIE, or any combinations thereof. In particular embodiments, the at least one ionizable cationic lipid may comprise or consist of DODMA. [0247] The amount of the at least one ionizable cationic lipid may be determined as appropriate. In some cases, the amount of the at least one ionizable cationic lipid to be used may be determined based on the type of cargo. [0248] In some embodiments, the amount of ionizable cationic lipid(s) relative to the total amount of TCV components may be about 20 mo!% to about 30 mo!%, about 20 mo!% to about 35 mo!%, about 20 mo!% to about 40 mo!%, or about 10 mo!% to about 70 mo!%. In some embodiments, the total amount of ionizable cationic lipid(s) relative to TCV components may be about 10 mo!% to about 60 mo!%, about 10 mo!% to about 50 mo!%, about 10 mo!% to about 40 mo!%, about 10 mo!% to about 30 mo!%, about 15 mo!% to about 25 mo!%, about 18 mo!% to about 22 mo!%, about 19 mo!% to about 21 mo!%, about 19.5 mo!% to about 20.5 mo!%, about 19.8 mo!% to about 20.2 mo!%, or about 20 mo!% or about 30 mo!%. In particular embodiments, for example when the cargo comprises a protein, or a nucleic acid and a protein (or a RNP), the total amount of ionizable cationic lipid(s) relative to the total amount ofTCV components may be about 20 mo!%. [0249] In a preferred embodiment, a lipid-based TCV according to the present disclosure comprises DODMA, DLinDMA, DLin-KC2-DMA, DLin-MC3-DMA, SM-102, and/or ALC-0315 at about 20 J mo!% to about 30 mo!% or at about 20 mo!% relative to the total amount ofTCV components. so (0250] In some embodiments, the amount of ionizable cationic lipid(s) relative to the total amount of TCV components may be about 10 mo!% to about 70 mo!%, about 20 mo!% to about 70 mo!%, about 30 mo!% to about 70 mo!%, about 40 mo!% to about 70 mo!%, about 40 mo!% to about 60 mo!%, about 45 mo!% to about 55 mo!%, about 48 mo!% to about 52 mo!%, about 49 mo!% to about 51 mo!%, about 49.5 mo!% to about 50.5 mo!%, about 49.8 mo!% to about 50.2 mo!%, or about 50 mo!%. In particular embodiments, for example when the cargo comprises a nucleic acid such as a siRNA, shRNA or miRNA or a RNA or DNA vector, the total amount of ionizable cationic lipid(s) relative to the total amount ofTCV components may be about 50 mo!%. [0251] In a preferred embodiment, a lipid-based TCV according to the present disclosure comprises DODMA, DLinDMA, DLin-KC2-DMA, DLin-MC3-DMA, SM-102, and/or ALC-0315 at 50 mo!% relative to the total amount ofTCV components. [0252] Helper lipid (0253] In some embodiments, a lipid-based TCV may comprise at least one helper lipid in addition to the at least one ionizable cationic lipid. In some embodiments, the at least one helper lipid may comprise DOPE, DSPC, DOPC, DPPC, DOPG, DPPG, POPC, POPE, DOPE-ma!, DPPE, DMPE, DSPE, 16-O-monomethyl PE, 16-O-dimethyl PE, 18-1-trans PE, SOPE, or any combinations thereof. In particular embodiments, the at least one helper lipid may comprise or consist of DOPE. In some cases, the at least one helper lipid to be used may be determined based on the stability of the TCV. (0254] The amount of the at least one helper lipid may be determined as appropriate. [0255] In some embodiments, the total amount of helper lipid(s) relative to the total amount ofTCV components may be about IO mo!% to about 60 mo!%. In some embodiments, the total amount of helper lipid(s) relative to the total amount ofTCV components may be about 20 mo!% to about 30 mo!%, about 20 mo!% to about 40 mo!%, about 10 mo!% to about 50 mo!%, about 10 mo!% to about 40 mo!%, about 25 mo!% to about 35 mo!%, about 28 mo!% to about 32 mo!%, about 29 mo!% to about 31 mo!%, about 29.5 mo!% to about 30.5 mo!%, about 29.8 mo!% to about 30.2 mo!%, or about 20 mo!% or about 30 mo!%. In particular embodiments, the total amount of helper lipid(s) relative to the total amount ofTCV components may be about 30 mo!%. (0256] In some embodiments, the total amount of helper lipid(s) relative to the total amount ofTCV components may be about 20 mo!% to about 60 mo!%, about 30 mo!% to about 50 mo!%, about 35 mo!% to about 45 mo!%, about 38 mo!% to about 42 mo!%, about 39 mo!% to about 41 mo!%, about 39.5 mo!% to about 40.5 mo!%, about 39.8 mo!% to about 40.2 mo!%, or about 40 mo!%. In particular embodiments, the total amount of helper lipid(s) relative to the total amount ofTCV components may be about 40 mo!%. [0257] In a preferred embodiment, a lipid-based TCV according to the present disclosure comprises DOPE at 30 mo!%. Such a TCV may be used, for example when the cargo comprises a nucleic acid and a protein ( or a RNP). [0258} Phospholipid [0259) In some embodiments, a lipid-based TCV may comprise at least one phospholipid in addition to the at least one ionizable cationic lipid. In some embodiments, the at least one phospholipid may comprise DSPC, DOPE, DPPC, DOPC, DMPC, PLPC, DAPC, PE, EPC, DLPC, DMPC, MPPC, PMPC, PSPC, DBPC, SPPC, DEPC, POPC, lysophosphatidyl choline, DSPE, DMPE, DPPE, POPE, lysophosphatidylethanolamine, or any combinations thereof. In particular embodiments, the at least one helper lipid may comprise or consist of DSPC. [0260) In some embodiments, the amount of phospholipid(s) relative to the total amount ofTCV components may be about 20 mo!% to about 30 mo!%, about 20 mo!% to about 40 mo!%, about 5 mo!% to about 65 mo!%, about 5 mo!% to about 55 mo!%, about 5 mo!% to about 45 mo!%, about 5 mo!% to about 35 mo!%, about 5 mo!% to about 25 mo!%, about 5 mo!% to about 15 mo!%, about 8 mo!% to about 12 mo!%, about 9 mo!% to about 11 mo!%, about 9.5 mo!% to about 10.5 mo!%, about 9.8 mo!% to about 10.2 mo!%, or about 10 mo!%. In particular embodiments, the total amount of phospholipid(s) relative to the total amount ofTCV components may be about 10 mo!%. [0261) In some embodiments, the total amount ofphospholipid(s) relative to the total amount of TCV components may be about 5 mo!% to about 65 mo!%, about 15 mo!% to about 65 mo!%, about 25 mo!% to about 55 mo!%, about 35 mo!% to about 45 mo!%, about 38 mo!% to about 42 mo!%, about 39 mo!% to about 41 mo!%, about 39.5 mo!% to about 40.5 mo!%, about 39.8 mo!% to about 40.2 mo!%, or about 40 mo!%. In particular embodiments, the total amount of phospholipid(s) relative to the total amount ofTCV components may be about 40 mo!%. [0262) In a preferred embodiment, a lipid-based TCV according to the present disclosure comprises DSPC at about 5 mo!% to about 15 mo!%, or about 10 mo!% relative to the total amount ofTCV components. Such a TCV may be used, for example when the cargo comprises a nucleic acid molecule or nucleic acid and a protein (or a RNP complex). [0263} Cholesterol or cholesterol derivative [0264) In some embodiments, a lipid-based TCV may comprise at least one cholesterol or cholesterol derivative in addition to the at least one ionizable cationic lipid. In some embodiments, the at least one cholesterol or cholesterol derivative may comprise cholesterol, DC-Chol, l ,4-bis(3-N-oleylamino- propyl)piperazine, ICE, or any combinations thereof. In particular embodiments, the at least one cholesterol or cholesterol derivative may comprise or consist of cholesterol. [0265) In some embodiments, the amount of cholesterol and/or cholesterol derivative(s) relative to the total amount ofTCV components may be about 20 mo!% to about 60 mo!%. some embodiments, the amount of cholesterol and/or cholesterol derivative(s) relative to the total amount ofTCV components may be about 25 mo!% to about 55 mo!%, about 30 mo!% to about 50 mo!%, about 35 mo!% to about 45 mo!%, about 38 mo!% to about 42 mo!%, about 39 mo!% to about 41 mo!%, about 39.5 mo!% to about 40.5 mo!%, about 39.8 mol% to about 40.2 mo!%, or about 40 mo!%, or about 39%. In particular embodiments, the total amount of cholesterol and/or cholesterol derivative(s) relative to the total amount ofTCV components may be about 40 mo!% or about 39 mo!%. [0266] In a preferred embodiment, a lipid-based TCV according to the present disclosure comprises cholesterol at about 40 mo!% or about 39 mo!% relative to the total amount ofTCV components. Such a TCV may be used, for example when the cargo comprises a nucleic acid molecule or a nucleic acid and a protein (or a RNP complex). [0267} PEG-lipid [0268] In some embodiments, a lipid-based TCV may comprise at least one PEG-lipid in addition to the at least one ionizable cationic lipid. In some embodiments, the at least one PEG-lipid may comprise PEG-DMG (e.g., (Avanti® Polar Lipids (Birmingham, AL)), PEG- phosphatidylethanolamine and phosphatidic acid, PEG-ceramide conjugates (e.g., PEG-CerC14 or PEG-CerC20), PEG-modified dialkylarnines, PEG-modified 1,2-diacyloxypropan-3-amines, or any combinations thereof. In particular embodiments, the at least one PEG-lipid may comprise or consist ofPEG-DMG. [0269] In some embodiments, the amount of PEG and/or PEG-lipid(s) relative to the total amount of TCV components may be about 0.1 mol% to about 5 mol%, 0.1 mol% to about 4 mol%, 0.1 mol% to about 3 mol%, 0.1 mo!% to about 2 mol%, 0.5 mol% to about 1.5 mol%, 0.8 mol% to about 1.2 mol%, 0.9 mol% to about 1.1 mol%, or about 1 mol%. In particular embodiments, the total amount of PEG-lipid(s) relative to the total amount ofTCV components may be about I mo!%. [0270] In a preferred embodiment, a lipid-based TCV according to the present disclosure comprises PEG-DMG at 1 mo!% relative to the total amount ofTCV components. [0271] In a preferred embodiment, a lipid-based TCV according to the present disclosure comprises DODMA, DLinDMA, DLin-KC2-DMA, DLin-MC3-DMA, SM-102, and/or ALC-0315 at about 20 mol%, DOPE at about 30 mol%, DSPC at about 10 mo!%, and cholesterol at about 40 mol% relative to the total amount ofTCV components. In another preferred embodiment, a lipid-based TCV according to the present disclosure comprises DOD MA, DLinDMA, DLin-KC2-DMA, DLin-MC3- DMA, SM-102, and/or ALC-0315 at about 30 mol%, DOPE at about 20 mol%, DSPC at about 10 mo!%, and cholesterol at about 40 mol% relative to the total amount ofTCV components. Such a TCV may be used, for example when the cargo comprises a nucleic acid and a protein ( or a RNP complex). [0272] In another preferred embodiment, a lipid-based TCV according to the present disclosure comprises DODMA, DLinDMA, DLin-KC2-DMA, DLin-MC3-DMA, SM-102, and/or ALC-0315 at 50 mol%, DSPC at 10 mo!%, cholesterol at 39 mol%, PEG-DMG at 1 mo!% relative to the total amount ofTCV components. Such a TCV may be used, for example when the cargo comprises a nucleic acid molecule. [0273] Surface modulation [0274] In some embodiments, a TCV according to the present disclosure may comprise a targeting moiety. The targeting moiety to be incorporated may be determined based on the target cell type, so that the TCV may preferentially carry its cargo into the target cells. [0275] The targeting moiety may be any type of materials that allows for specific or preferential binding to a target cell. In some embodiments, the targeting molecule may be a protein (e.g., an antibody or an antibody fragment), a peptide, a nucleic acid (e.g., aptamer), a small molecule, or another material (e.g., a vitamin or a carbohydrate). [0276] In some embodiments, the targeting moiety may be designed to be specific for HSCs, HSPCs, MPPs, CMPs, MEPs, HPCs, erythroid progenitors (e.g., BFU-Es, CFU-Es), proerythroblasts, erythroblasts (basophilic erythroblasts, early erythroblasts ( e.g., type I, type II), polychromatic erythroblasts, intermediate erythroblasts, acidophilic erythroblasts, late erythroblasts, normoblasts, reticulocytes (before nucleus expulsion), or any combinations thereof. [0277] In particular embodiments, the targeting moiety may be designed to be specific for HSCs and/or HSPCs. In certain embodiments, the targeting moiety may specifically or preferentially bind to CD34, which is present on HSCs and/or HSPCs. In certain embodiments, the targeting moiety may be an antibody or an antibody fragment specific to CD34. [0278] TCV size [0279] In some embodiments, the size ofTCVs may be determined by any appropriate techniques. Non-limiting examples of measurement methods include dynamic light chattering, binding assays, surface plasmon resonance (SPR), static image analysis, and dynamic image analysis. An appropriate measurement technique may be selected based on the accuracy and the approximate size range the technique is optimal for. [0280] In some embodiments, the size of the TCV before encapsulation of the at least one cargo may be in a range of about 3 nm to about 240 nm, about 6 nm to about 160 nm, about 9 nm to about 80 run, or about 20 nm to about 40 nm, at pH of about 4. In particular embodiments, the size of the TCV before encapsulation of the at least one cargo may be in a range of about 9 nm to about 80 nm at pH of about 4. [0282] Compositions [0283] In one aspect, a pharmaceutical composition according to the present disclosure comprises at least one cargo which is capable of effecting gene editing. In some embodiments, the gene editing may a SCD-associated gene. In some embodiments, the gene editing takes place in vivo. For example, the gene editing may take place in the bone marrow (in a cell in the bone marrow). Alternatively, the gene editing may take place in the peripheral circulation (in a cell in the peripheral circulation). [0284] In some embodiments, a pharmaceutical composition which may be used for effecting gene editing may comprise a Cas nuclease and a gRNA. Non-limiting examples of Cas proteins and gRNAs that may be contained in a pharmaceutical composition are as described herein. In some embodiments, the Cas nuclease and the gRNA contained in a pharmaceutical composition may be forming a complex, i.e., RNP. [0285] In particular embodiments, the Cas nuclease contained in a pharmaceutical composition may be a Cas 12a nuclease. In a particular embodiment, the Cas 12a nuclease may be AsCas 12a, LbCas 12a, FnCas12a, MbCas12a, CeCasl2a, or BfCasl2a. In particular embodiments, the gRNA contained in a pharmaceutical composition may comprise a target-complementary sequence of SEQ ID NO: 71,277, or279,or87,287,or289. [0286] In particular embodiments, the Cas nuclease contained in a pharmaceutical composition may be a Cas9 nuclease. In a particular embodiment, the Cas9 nuclease may be Cas9 of Streptococcus pyogenes (SpCas9). In particular embodiments, the gRNA contained in a pharmaceutical composition may comprise a target-complementary sequence of any one of SEQ ID NOS: 85, 281, 283, 285, 25, 65, 67, 69,271,273,275, 75, and 77. [0287] In some embodiments, e.g., when a gene knock-in and/or gene correction is desired, a pharmaceutical composition may further comprise a DNA template. In some embodiments, e.g., when disruption of a gene or gene segment is desired, a pharmaceutical composition may further comprise a DNA template. In particular embodiments, the repair DNA template may be a ssODN. In some embodiments, the repair DNA template may be encapsulated in the sarne TCV as the Cas nuclease and gRNA (or RNP) or in a separate TCV. [0288] In some embodiments, a composition may contain the RNP and the template at any appropriate ratio. In certain embodiments, the RNP : template ratio may be about 1 :5, about 1 :4, about 1 :3, about 1 :2, about 1: 1.5, about 1: 1, about 1.5: 1, about 2: 1, about 3: 1, about 4: 1, about 5: 1, about 6: 1, or about 7: 1. In some cases the RNP: template ratio may be about 1 :2, about 1: 1.5, about 1: 1, about 1.5: 1, about 2: 1, about 3: 1, about 4: 1, or about 5: 1. In certain cases the RNP : template ratio may be about 1: 1, about 1.5: 1, about 2: 1, or about 3: 1. In particular cases the RNP : template ratio may be about 1.5: 1, about 1.8: 1, about 2: 1, about 2.5: 1, or about 3: 1. In particular embodiments, the RNP : template ratio may be about 2: 1. [0289] In some embodiments, when disruption of a gene or gene segment is desired, the template may be any of the templates or ODNs described herein as not being complementary to or as having low complementarity to a segment within a target gene. In particular embodiments, the template may have SEQ ID NO: 190 ( or a reverse complementary thereto) or a sequence with at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identity thereto or a reverse complement of any of the foregoing. [0290] In some embodiments, when the gRNA targets the EER of BCLJ I A, , the template may have SEQ ID NO: 305 or 306 ( or 303, 304, 301, or 302) or a sequence with at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identity thereto . [0291] In some embodiments, when editing in the HBG gene promoter (e.g., the region containing the BCLl lA-binding motifor a region in the proximity thereto, such as within about 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 nt from the 5'-end or the 3'-end of the motif) is desired, the template may be any of the DNA template for editing the HBG gene promoter described herein. In particular embodiments, the template may be a ssODN which comprises the nucleic acid sequence of SEQ ID NO: 191 or 192 or a dsODN which comprises a strand of SEQ ID NO: 191 and a strand of SEQ ID NO: 192. [0292] In some embodiments, when correction in the HEB gene back to a gene encoding a wildtype beta-globin is desired, the DNA template may be any of the DNA template for HBB described herein. In particular embodiments, the repair DNA template may be a ssODN comprising the nucleic acid sequence of any one of SEQ ID NOS: 101-108, or a sequence fully complementary thereto. In particular embodiments, the repair DNA template may be a ssODN comprising the nucleic acid sequence of SEQ ID NO: 101 or 102, or a sequence fully complementary thereto. [0293] In some embodiments, a pharmaceutical composition according to the present disclosure may be effecting gene editing thereby disrupting a gene or gene segment and comprises: (A) a Cas nuclease or a nucleic acid encoding the Cas nuclease, wherein the Cas nuclease may be any appropriate Cas nuclease such as those described herein; (B) a gRNA or a nucleic acid encoding the gRNA, wherein the gRNA comprises a target-complementary sequence of interest such that the target-complementary sequence is capable of binding to a target sequence of interest within a target gene of interest thereby guiding the Cas9 nuclease of (A) to and allowing for cleavage at a target site of interest within said target gene of interest; and (C) an isolated ODN or a nucleic acid encoding the isolated ODN, wherein the ODN may any of the ODNs described herein to be not complementary to or to have low complementarity to the target site (the sequences flanking the site to be cleaved by the Cas nuclease used). Without wishing to be bound by theory, such a non-complementary template may enhance the efficiency of causing deletion of one or more nucleotides or one or more nucleotide insertions or alterations in the nucleotide sequence (e.g., thereby disrupting the function (e.g., promoter binding) of the nucleotide sequence before editing or causing early appearance of a terminator or stop codon). [0294] In certain embodiments, a template may have less than 90%, less than 85%, less than 80%, less than 75%, less than 70%, less than 65%, less than 60%, less than 55%, less than 50%, less than 45%, less than 40%, less than 35%, less than 30%, less than 30%, less than 25%, less than 20%, less than 15%, or less than 10% complementarity to the sequence of a segment within the target gene, wherein the segment comprises the target sequence ( contained in the gRNA to be used with the Cas nuclease) and/or the sequence complementary thereto. [0295] In some cases, the segment within the target gene may be from about 100 nt, about 95 nt, about 90 nt, about 85 nt, about 80 nt, about 75 nt, about 70 nt, about 65 nt, about 60 nt, about 55 nt, about 50 nt, about 45 nt, about 40 nt, about 35 nt, about 30 nt, about 25 nt about 20 nt, about 15 nt, about 10 nt, about 5 nt upstream of the cut site (the position to be cleaved by the Cas nuclease) or the 5'-end of the target sequence to about 100 nt, about 95 nt, about 90 nt, about 85 nt, about 80 nt, about 75 nt, about 70 nt, about 65 nt, about 60 nt, about 55 nt, about 50 nt, about 45 nt, about 40 nt, about 35 nt, about 30 nt, about 25 nt about 20 nt, about 15 nt, about 10 nt, about 5 nt downstream of the cut site (the position to be cleaved by the Cas nuclease) or the 3'-end of the target sequence. [0296) In particular embodiments, the template may be a ssODN which comprises or consists of SEQ ID NO: 190 (a sequence at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, identical thereto) or a sequence reverse complementary thereto or a dsODN comprising a strand of SEQ ID NO: 190 (a sequence at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, identical thereto) and a strand reverse complementary thereto. [0297] In particular embodiments, the target gene may be a SCD-associate gene, such as but not limited to HBGI, HBG2, BCLI IA, and KLFI. [0299) In one aspect, a pharmaceutical composition according to the present disclosure comprises at least one cargo which is capable of effecting gene expression alteration. In some embodiments, the expression a SCD-associated gene may be altered, e.g., upregulated or downregulated, or increased or decreased. In some embodiments, the gene expression alteration takes place in vivo. For example, the gene expression alteration ( or modification) may take place in the bone marrow (in a cell in the bone marrow). Alternatively, the gene expression alteration (or modification) may take place in the peripheral circulation (in a cell in the peripheral circulation). [0300) In some embodiments, a pharmaceutical composition which may be used for effecting gene expression alteration ( or modification) may comprise a nucleic acid molecule. Any of the nucleic acid molecules that mediate gene expression alteration ( or modification) disclosed herein may be contained in a pharmaceutical composition. [030 l] Cargo combinations [0302) In some embodiments, a pharmaceutical composition according to the present disclosure may comprise a TCV comprising (i) at least one cargo which is capable of effecting gene editing and (ii) at least one cargo capable of effecting gene expression alteration. In some embodiments, a pharmaceutical composition according to the present disclosure may comprise (i) a TCV comprising at least one cargo which is capable of effecting gene editing and (ii) a separate TCV comprising at least one cargo capable of effecting gene expression alteration. [0303) In some embodiments, a pharmaceutical composition according to the present disclosure may comprise a TCV comprising (i) at least one first cargo which is capable of effecting first gene editing and (ii) at least one second cargo capable of effecting second gene editing. In some embodiments, a pharmaceutical composition according to the present disclosure may comprise (i) a first TCV comprising at least one first cargo which is capable of effecting first gene editing and (ii) a second TCV comprising at least one second cargo capable of effecting second gene editing. [0304] In some embodiments, a pharmaceutical composition according to the present disclosure may comprise a TCV comprising (i) at least one first cargo which is capable of effecting first gene expression alteration and (ii) at least one second cargo capable of effecting second gene expression alteration. In some embodiments, a pharmaceutical composition according to the present disclosure may comprise (i) a first TCV comprising at least one first cargo which is capable of effecting gene expression alteration and (ii) a second TCV comprising at least one second cargo capable of effecting second gene expression alteration. [0305] Organic solvents and detergents [0306] In some embodiments, one characteristic of a pharmaceutical composition is that the composition is substantially, essentially, or entirely free of destabilizing agents, and/or contains significantly lower amounts of destabilizing agents compared to other pharmaceutical compositions comprising a similar type ofTCVs. [0307] In some embodiments, one characteristic of a pharmaceutical composition is that the composition is substantially, essentially, or entirely free of organic solvents and detergents, and/or contains significantly lower amounts of organic solvents and detergents compared to other pharmaceutical compositions comprising a similar type ofTCVs. [0308] In some embodiments, one characteristic of a pharmaceutical composition is that the composition is substantially, essentially, or entirely free of ethanol, methanol, isopropanol, tetrahydrofuran (THF), dimethyl sulfoxide (DMSO), dimethyl formamide (DMF), and acetonitrile (ACN), and/or contains significantly lower amounts of ethanol, methanol, isopropanol, THF, DMSO, DMF, and ACN, compared to other pharmaceutical compositions comprising a similar type ofTCVs. [0309] In particular embodiments, the pharmaceutical composition may be entirely free of methanol, isopropanol, THF, DMSO, DMF, and ACN. [031 O] With regard to ethanol, in some embodiments, the pharmaceutical composition may be substantially free of ethanol, which may mean that the ethanol concentration is 5% (v/v) or below. In particular embodiments, the pharmaceutical composition may be essentially free of ethanol, which may mean that the ethanol concentration is 0.5% (v/v) or below. [0311] In a particular embodiment, the pharmaceutical composition may be entirely free of ethanol, methanol, isopropanol, THF, DMSO, DMF, and ACN. [0312] Additional components [0313] In one aspect, a pharmaceutical composition may comprise an additional component in addition to at least one cargo for effecting gene editing or gene expression alteration. [0314} Stem cell mobilization agents [0315] In some embodiments, the pharmaceutical composition may further comprise at least one stem cell mobilization agent. In some embodiments, the at least one stem cell mobilization agent may be contained in the composition outside the TCVs comprising the at least one cargo for effecting gene editing or gene expression alteration. [0316] In some embodiments, a composition comprising a at least one stem cell mobilization agent may be suited for injection into the peripheral circulation, e.g., IV injection. In such an embodiment, the at least one stem cell mobilization agent may promote exit of stem cells ( e.g., HSPs and/or HSPCs) into the circulation, and this may help TCVs efficiently enter the stem cells in the circulation. In some embodiments, such a composition may be suited for continuous injection for a period of time (e.g., 3 hours, 6 hours, 12 hours, 18 hours, 24 hours) in some instances multiple times. Without wishing to be bound by theory, the continuous and/or multiple loading of at least one stem cell mobilization may provide relatively sustained levels of stem cells in circulation that TCV s may enter. [0317] In some embodiments, the at least one stem cell mobilization agent may comprise G-CSF (filgrastim), GM-CSF, Plerixafor, SCF, CXCR4 antagonists (e.g., POL6326, BKT-140, TG-0054), CXCL12 neutralizers (e.g., NOX-A12), Sphingosine-1-phosphate (SIP) antagonists (e.g., SEW2871), VCAMNLA-4 inhibitors (e.g., BIO 5192), parathyroid hormone, protease inhibitors (e.g., Bortezomib), Grof (e.g., SB-251353), hypoxia inducible factor (HIF) stabilizers (e.g., FG-4497), or any combinations thereof. [0318] In some embodiments, the at least one stem cell mobilization agents may comprise G-CSF (filgrastim). In some embodiments, the at least one stem cell mobilization agents may comprise Plerixafor. In some embodiments, the at least one stem cell mobilization agents may comprise G-CSF (filgrastim) and Plerixafor. [0319 J Erythropoiesis stimulating agents [0320] In some embodiments, the pharmaceutical composition may further comprise at least one erythropoiesis stimulating agent. In some embodiments, the at least one erythropoiesis stimulating agent may be contained in the composition inside or outside a TCV, and if inside, the TCV may be the TCV comprising the at least one cargo for effecting gene editing or gene expression alteration or a separate TCV. In some embodiments, if the agent is an extracellular factor (e.g., growth factor, cytokine), the agent may be contained outside a RCV. In some embodiments, if the agent is an intracellular factor (e.g., transcription factor) or an agent that requires an intracellular machinery (e.g., a nucleic acid that needs to be transcribed and/or translated to function). [0321] In some embodiments, the at least one erythropoiesis stimulating agent may help more of the HSCs and/or HSPCs the TCV comprising the at least one cargo for effecting gene editing or gene expression alteration entered ultimately differentiate into red blood cells. In some embodiments, the at least one erythropoiesis stimulating agent may help more of the cells at the early stages of erythropoiesis (e.g., MMPs, CMPs, MEPs) the TCV comprising the at least one cargo for effecting gene editing or gene expression alteration entered ultimately differentiate into red blood cells. [0322] In some embodiments, the at least one erythropoiesis stimulating agent may comprise SCF, GM-CSF, interleukin-3 (IL-3), interleukin-9 (IL-9), erythropoietin (EPO), TGF-beta, growth differentiating factor 11 (GDFl 1), Activin A, Transferrin (Tf), ferritin, ferroportin, hepcidin, vitamin B 12, folic acid, copper, or any combinations thereof. In some embodiments, such an agent may be encapsulated outside a TCV. [0323] In some embodiments, the at least one erythropoiesis stimulating agent may comprise an agent selected from the group consisting ofGATA-1, STAT5A, STAT5B, MCL-1, BCL-xL, HSP70, or any combinations thereof, or a RNA or DNA encoding thereof. In some embodiments, such an agent may be encapsulated in the TCV encapsulating the at least one cargo for effecting gene editing or gene expression alteration or in a separate TCV. [0324] In some embodiments, the at least one erythropoiesis stimulating agent may comprise an inhibitor or silencer of a negative regulator of erythropoiesis. In some embodiments, such a negative regulator may be inhibin, TGF-beta, BID (a member of the BCL-2 family), Fas ligand, Fas, caspase, or any combinations thereof. In some embodiments, the agent may be encapsulated in the TCV encapsulating said at least one cargo for effecting gene editing or gene expression alteration or in a different TCV. Alternatively, in some embodiments, the agent may be encapsulated outside a TCV. [0325] Additional exemplary erythropoiesis stimulating agents include but are not limited to engineered EPOs (such as Darbepoetin alfa, AMG 205, AMG 114, Pegzyrepoetin alfa, or MK-2578), EPO mimetics (such as EMPl, CNTO 528, CNTO 530, or Peginesatide), or anti-EPO receptor agonistic antibodies. Non-limiting examples of such agents are reviewed in detail in Sinclair. Biologics. 2013;7: 161-74. Epub 2013 Jul 3. [0326} Agents for treating SCD [0327] In some embodiments, the pharmaceutical composition may further comprise at least another agent for treating SCD. In some embodiments, the at least another agent may be hydroxyurea, L- glutamine oral powder, crizanlizumab, a general pain medication, voxelotor, or any combination thereof. [0328] Manufacturing [0329] TCVs [0330] As described above, one characteristic of a pharmaceutical composition according to some embodiments may be that the composition substantially, essentially, or entirely lacks organic solvents and detergents, which may help improve the stability and/or integrity of the TCV and/or its cargo. In some embodiments, the manufacturing method of a TCV according to the present disclosure may contribute to such a characteristic. [0331] In some embodiments, TCVs may be stored at a freezing temperature. In some embodiments, when a TCV is prepared, a cryoprotectant may be added. In some embodiments, a cryoprotectant may comprise a sugar-based molecule. Non-limiting examples of cryoprotectants include sucrose, trehalose, and a combination thereof. [0332] In some embodiments, the TCV may further comprise at least one cryoprotectant. In certain embodiments, the at least one cryoprotectant may be or may comprise a sugar-based molecule, e.g., a sugar molecule or a derivative thereof. In particular embodiments, the at least one cryoprotectant may be sucrose, trehalose, or a combination thereof. In a particular embodiment, the at least one cryoprotectant may be sucrose. [0333] In some embodiments, a pharmaceutical composition according to the present disclosure may comprise at least one cryoprotectant. In certain embodiments, the cryoprotectant in a pharmaceutical composition may be the cryoprotectant comprised in the TCV contained in the pharmaceutical composition. [0334] In some embodiments, the TCV, which may comprise at least one cryoprotectant, may be stable at a freezing temperature, optionally at about -20°C or about -80°C, optionally for at least about one week, at least about two weeks, at least about three weeks, at least about a month, at least about two months, at least about four months, at least about five months, at least about 6 months, at least about 9 months, at least about a year, or at least about two year, or longer, or about one week to about two year, about two weeks to about a year, about three weeks to about nine month, about one to about six months, about one to five months, about one to four months, about one to three months, or about one to two months. [0335] In some embodiments, the concentration of the at least one cryoprotectant contained in the TCV or pharmaceutical composition may be about 1 % to about 40 %, about 3% to about 30%, about 5% to about 30%, about 10% to about 20%, or about 15%. [0336] A TCV according to the present disclosure may be prepared by any appropriate methods. In some embodiments, a TCV may be prepared by (a) generating a first solution by dissolving all components of the TCV in ethanol; (b) providing a second solution, which is aqueous; (c) combining the first and second solutions; and (d) removing ethanol, optionally by dialysis or evaporation. [0337] In some embodiments, the first solution in step (a) may contain the TCV components at about 20-35 mM. In some embodiments, the second solution in step (b) may contain sodium acetate and/or sodium citrate, which optionally may be at about 25 mM. In some embodiments, the pH of the second solution in step (b) may be about 4 In some embodiments, the combining in step (c) may be by gentle mixing (optionally repeated manual reciprocation of the TCV-generating fluid in a pipette), micromixing optionally using a staggered herringbone micromixer (SHM), T-junction or Y-junction mixing, or extrusion. In a particular embodiment, the removing in step ( d) is by dialysis. [0338] RNPs [0339] In some embodiments, wherein the TCV comprise a RNP as a cargo, the RNP may be generated by any appropriate methods. In some embodiments, the RNP may be formed by mixing Cas9 and gRNA at an approximately equimolar ratio, optionally for about 5 minutes. [0341] In some embodiments wherein the TCV comprise a RNP as a cargo, the RNP encapsulation by TCVs may be performed by any appropriate methods. In some embodiments, the encapsulation may be performed by (i) providing an aqueous solution comprising the TCV; and (ii) mixing the at least one RNP with the aqueous solution, wherein mixing is effected under conditions suitable for the at least one RNP to be encapsulate within the TCV. In some embodiments, the aqueous solution in step (i) may have the pH of about 3 to about 8, optionally about 4 to about 7.5. [0342] In some embodiments wherein the TCV comprise a nucleic acid molecule (not RNPs) as a cargo, the nucleic acid molecule encapsulation by TCVs may be performed by any appropriate methods. In some embodiments, the encapsulation may be performed by: (i) providing an aqueous solution comprising the TCV; and (ii) mixing the nucleic acid molecule with the aqueous solution, wherein mixing is effected under conditions suitable for the at least one nucleic acid molecule to be encapsulate within the TCV. In some embodiments, the aqueous solution in step (i) may have the pH of about 3 to about 8, optionally about 4 to about 7.5. [0343] Method of effecting gene editing [0344] In one aspect, a method of effecting gene editing ( e.g., disrupting a gene or gene segment) at a target site of interest within a target gene of interest in a target cell of interest is also provided herein. [0345] In some embodiments, the method may comprise contacting the target cell of interest with a composition comprising (A) a Cas nuclease or a nucleic acid encoding the Cas nuclease, wherein the Cas nuclease may be any appropriate Cas nuclease such as those described herein; (B) a gRNA or a nucleic acid encoding the gRNA, wherein the gRNA comprises a target-complementary sequence of interest such that the target-complementary sequence is capable of binding to a target sequence of interest within a target gene of interest thereby guiding the Cas9 nuclease of (A) to and allowing for cleavage at a target site of interest within said target gene of interest; and (C) an isolated ODN or a nucleic acid encoding the isolated ODN, wherein the ODN may be any of the ODNs described herein to be not complementary to or to have low complementarity to the target site (the sequences flanking the site to be cleaved by the Cas nuclease used). Without wishing to be bound by theory, use of such an ODN increases the efficiency of gene editing (e.g., deleting one or more nucleotides). [0346] The resulting gene editing may for example comprise deletion of one or more nucleotides, insertion of one or more nucleotides, and/or substituting one or more nucleotides at the target site of interest (e.g., at the cleavage site). In certain embodiments, such editing may result in deactivation of the gene or gene segment, for example by blocking or reducing the function of the affected gene segment (e.g., prevents binding by a regulatory element such as a promoter, repressor, and/or enhancer). [0347] In certain embodiments, the target gene of interest is a disease-associated or disease-causing gene, including but not limited to any of the SCD-associated genes described herein. [0348] In certain embodiments, the contacting may occur in vitro. [0349] In certain embodiments, the contacting may occur ex vivo. For example, cells derived from a subject (e.g., human patient) may be contacted with the composition and later the cells may be administered to the same or a different subject. [0350] In certain embodiments, the contacting may occur in vivo. For example, the composition may be administered to a subject, and once in the subject may be contacted to one or more cells of the subject. In particular embodiments, the administration may be into the bone marrow of the subject or intravenously. [0351] In certain embodiments, the method may be for preventing or treating a disease. The disease may be any appropriate disease (e.g., any genetic disease of interest, such as Huntington's disease) and may include but not be limited to SCD. [0352] In vivo methods [0353] In one aspect, a pharmaceutical composition may be used for in vivo purposes. In some embodiments, the pharmaceutical composition may be for effecting gene editing of a target gene such as a SCD-associated gene in vivo. In some embodiments, the pharmaceutical composition may be for effecting gene expression alteration of a target gene such as a SCD-associated gene in vivo. In some embodiments, the pharmaceutical composition may be for preventing, ameliorating, or treating SCD. [0354] In the strategies currently tested in the clinic for providing gene editing or gene expression alteration on a SCD-associated gene for the purpose of treating SCD, gene editing or gene expression alteration occurs in autologous HSCs and/or HSPCs ex vivo. This means that the strategy requires the steps of harvesting ofHSCs and/or HSPCs from a patient, ex vivo modification of the harvested HSCs and/or HSPCs, myeloablation, and administering the modified HSCs and/or HSPCs back to the patient, which requires lengthy steps that require sophisticated facilities and high cost. [0355] In one aspect, one characteristic of the inventive method is that the method addresses such disadvantages and difficulties. In some embodiments, the method involves injection of a pharmaceutical composition according to the present disclosure directly into the bone marrow of a subject, e.g. a subject with SCD, SCA, HbSC, or HbS P-thalassaemia, or other subjects as disclosed in the following section. [0356] . In some embodiments, the method involves injection of a pharmaceutical composition according to the present disclosure into the peripheral circulation of a subject, e.g. a subject with SCD, SCA, HbSC, or HbS P-thalassaemia or other subjects as disclosed in the following section, in which stem cell mobilization is induced. Such methods avoid any of the complicated steps as explained above (steps from stem cell harvest to administering modified stem cells). [0357] Subject/patient population [0358] In one aspect, an in vivo method according to the present disclosure may be applied to any subject who is in need of a pharmaceutical composition according to the present disclosure. [0359] In some embodiments, a subject may have or have a risk of developing a disease involving a cell or bone marrow origin or a cell in the bone marrow. In some embodiments, a subject may have or have a risk of developing SCD. In such an embodiment, the subject may have at least one ps allele, i.e., the subject is in the fate of having SCD from birth. [0360] In some embodiment, the subject may be at any age or at any stage ofSCD. In certain embodiments, the subject is in the immediate post-natal period, optionally about 6 weeks old or younger. In certain embodiments, the subject is about 3 month old or younger. In certain embodiments, the subject still comprises sufficient amount ofHbF relative to HbA. In some embodiments, the subject is has the HbF:HbA ratio of about 2: 1, about 1: 1, about 1:2, about 1:3, about 1 :4, about 1: 5, or about 1: 10. In certain embodiments, the subject has not fully developed SCD and is prior to manifesting a symptom or complication. [0361] Injection into the bone marrow [0362] In some embodiments, the method involves injection of a pharmaceutical composition according to the present disclosure directly into the bone marrow of a patient. [0363] Intramarrow injection [0364] Injection into the bone marrow, intramarrow injection or Intraosseous infusion (IO), may be performed via any appropriate methods. The intramarrow route is currently used in the clinic, e.g., for bone marrow transplant or for acute infusion when peripheral venous access is not available. Also, application for the treatment ofhematopoietic malignancies is being proposed (Islam. Clin Case Rep. 2015 Dec; 3(12): 1026-1029.; Islam. [0365] In some embodiments, intramarrow injection may be performed using any appropriate methods, e.g., by a slow bolus push, e.g., using a standard IO device such as but not limited to First Access for Shock and Trauma (FASTl), the EZ-10, or the Bone Injection Gun (BIG) or a manual device such as the Jamshidi needle or the Diekman modified needle (Dornhofer and Kellar. Intraosseous Vascular Access. [Updated 2021 Jul 26]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2021 Jan-.). Other examples of devices and details on the injection methods may be found in Islam. J Clin Pathol. 2020 Sep;73(9):552-556. or US10265481B2. In particular embodiments, an intramarrow needle comprising the length of about 50 to about 100 mm or about 70 to about 80 nm may be used. [0366] In some embodiments, when the target cells are cells that have a potential to eventually differentiate into red blood cells, the direct injection may be performed into tibia, femur, sternum, skull, ribs, pelvis (e.g., iliac), or any combinations thereof, which are the bones in which erythropoiesis mainly takes place. As erythropoiesis in the tibia and femur declines by about age 25, in some embodiments, these bones may be a suitable injection site for patients up to younger patients up to about age 20 or about 25. [0367] Dosing regimen [0368] The pharmaceutical composition for intramarrow injection may comprise any amounts of cargo sufficient for effecting sufficient gene editing and/or gene expression alteration. In some embodiments, the pharmaceutical composition may comprise per mL about 300 pmol to about 30000 pmol, optionally about 500 to about 10000 pmol, about 1000 to about 5000 pmol, about 2000 to about 4000 pmol, about 2500 to about 3000 pmol, or about 2700 pmol of the RNP or the nucleic acid molecule. In particular embodiments, the pharmaceutical composition may comprise about 2700 pmol of the RNP or the nucleic acid molecule per mL. [0369] The intramarrow injection may be given at any appropriate volume and/or speed suited for effecting sufficient gene editing and/or gene expression alteration. In some embodiments, injection of the pharmaceutical composition may be via a continuous flow of about 25 mL to 125 mL per minute, optionally about 25 mL to 50 mL per minute, about 50 mL to 100 mL per minute, about 100 mL to 125 mL per minute, about 40 mL to about 80 mL per minute, or about 50 mL to about 70 mL per minute. [0370] In some embodiments, the TCVs encapsulating at least one cargo may be comprised in or loaded in a matrix or material that may be injected or implanted in the bone marrow. Any appropriate matrices or materials may be used. In some embodiments, use of such a matrix or material may lead to improved safety, for example by allowing a smaller thus safer dose to provide efficacy and/or lead to improved feasibility, for example by allowing gradual release of the TCVs, thereby offering less frequent need of injections or shorter injection duration. Non-limiting examples of such a matrix or material may be those described in Ho et al., Sci Adv. 2021 May 19;7(21):eabg3217, Lee et al., Proc Natl Acad Sci US A. 2012 Nov 27; 109(48): 19638-43. Epub 2012 Nov 12., and Shah et al., Nat Biotechnol. 2019 Mar;37(3):293-302. [0371] Injection into the peripheral circulation [0372] In some embodiments, the method involves injection of a pharmaceutical composition according to the present disclosure into the peripheral circulation of a patient in which stem cell mobilization is induced. [0373] Stem Cell Mobilization [0374] Stem cell mobilization may be induced in a subject via any appropriate methods. In some embodiments, at least one agent that promotes stem cell mobilization (from the bone marrow to the peripheral circulation) may be administered to the subject. In some embodiments, such at least one agent may be administered prior to or at the same time as injection of a pharmaceutical composition comprising a TCV encapsulating a cargo according to the present disclosure. [0375] In some embodiments, exemplary agents for stem cell mobilization include but are not limited to G-CSF (filgrastim), GM-CSF, Plerixafor, SCF, CXCR4 antagonists (e.g., POL6326, BKT-140, TG-0054), CXCL12 neutralizers (e.g., NOX-A12), Sphingosine-l-phosphate (SIP) antagonists (e.g., SEW2871), VCAM/VLA-4 inhibitors (e.g., BIO 5192), parathyroid hormone, protease inhibitors (e.g., Bortezomib), Grof (e.g., SB-251353), and hypoxia inducible factor (HIF) stabilizers (e.g., FG- 4497). In particular embodiments, the stem cell mobilization agent may be G-CSF (filgrastim). In particular embodiments, the stem cell mobilization agent may be Plerixafor. In particular embodiments, the stem cell mobilization agent may be G-CSF (filgrastim) and Plerixafor. [0376] The at least one stem cell mobilization agents may be administered in any appropriate manner (dose, route, frequency) that would provide sufficient and timely mobilization. Exemplary dosing may be described in the art, for example in Andreola et al., Eur J Haematol. 2012 Feb;88(2): 154-8. Epub 2011 Nov 17. In some embodiments, stem cell mobilization may be induced by intravenous administration of G-CSF and plerixafor prior to injection of a pharmaceutical composition comprising a TCV encapsulating a cargo according to the present disclosure. In some embodiments, stem cell mobilization may be induced by intravenous administration of G-CSF followed by intravenous administration of plerixafor prior to injection of a pharmaceutical composition comprising a TCV encapsulating a cargo according to the present disclosure. [0377] In particular embodiments, the dosing of G-CSF may be about 5-30 ug/kg/day, preferably about 10 µg/kg/day, for about 3-5 days, preferably 4 days. In particular embodiments, the dosing of plerixafor may start once the peripheral blood CD34+ cells are <20 cells/ul, and/or on the day of the last G-CSF administration (e.g., the 4 th day) or the following day. In particular embodiments, the dosing of plerixafor may be about 0.1-0.5 mg/kg, preferably about 0.2-0.3 mg/kg or about 0.24 mg/kg. [0378] Dosing regimen [0379) The pharmaceutical composition for injection into the peripheral circulation (e.g., intravenous (IV) injection) may comprise any amounts of cargo sufficient for effecting sufficient gene editing and/or gene expression alteration. In some embodiments, the pharmaceutical composition may comprise per mL about 300 pmol to about 30000 pmol, optionally about 500 to about 10000 pmol, about 1000 to about 5000 pmol, about 2000 to about 4000 pmol, about 2500 to about 3000 pmol, or about 2700 pmol of the RNP or the nucleic acid molecule. In particular embodiments, the pharmaceutical composition may comprise about 2700 pmol of the RNP or the nucleic acid molecule per mL. [0380) The injection into the peripheral circulation may be given at any appropriate volume and/or speed suited for effecting sufficient gene editing and/or gene expression alteration. In some embodiments, injection of the pharmaceutical composition may be via a continuous flow of about 25 mL to 125 mL per minute, optionally about 25 mL to 50 mL per minute, about 50 mL to 100 mL per minute, about 100 mL to 125 mL per minute, about 40 mL to about 80 mL per minute, or about 50 mL to about 70 mL per minute. [0381) The maximum number of circulating CD34+ cells (or HSCs and/or HPCSs) may be achieved about 5 days after last plerixafor administration, at which point the median number of CD34+ cells ( or HSCs and/or HPCSs) may be about 60 per µL (Andreola et al., Eur J Haematol. 2012 Feb;88(2): 154- 8. Epub 2011 Nov 17.). Therefore, in some embodiments, injection of a pharmaceutical composition comprising a TCV encapsulating a cargo according to the present disclosure may start once the peripheral blood CD34+ cells (or HSCs and/or HPCSs) are 60 cells/µL or more. [0382) In some embodiments, a single injection or a first injection (if injection is to be repeated or continuous injection is intended) may take place about 3- 7 days, about every 3- 7 days, about 4-6 days, about every 4-6 days, about 5 days, or about every 5 days after the last plerixafor administration. In alternative embodiments, a series of injections may be given once daily, e.g., for one week following the last plerixafor administration. [0383] In some embodiments, there may be a prolonged time period between administration of the composition comprising the cargo comprising TCVs into the peripheral circulation or bone marrow, e.g., if it is determined after treatment (see "Monitoring and dosing regimen adjustment" section infra), that the treated subject does not comprise a sufficient number of normal (gene-edited) cells in their peripheral circulation, e.g., HSCs, HSPCs, MPPs, CMPs, MEPs, HPCs, erythroid progenitors (e.g., BFU-E, CFU-E), proerythroblasts, erythroblasts (basophilic erythroblasts, early erythroblasts (e.g., type I, type II), polychromatic erythroblasts, intermediate erythroblasts, acidophilic erythroblasts, late erythroblasts, normoblasts, reticulocytes before nucleus expulsion, reticulocytes, or erythrocytes, or any combination thereof. [0384] Method of preventing, [0385] In one aspect, a pharmaceutical composition comprising a TCV encapsulating a cargo according to the present disclosure may be for preventing, ameliorating, or treating a target disease. In some embodiments, the target disease may be a disease associated with cells of bone marrow origin and/or cells in the bone marrow. In some embodiments, the target disease may be SCD. In particular embodiments, the target disease may be SCA, HbSC, or HbS ~-thalassaemia. [0386] Such a preventative, amelioration, or therapeutic method may comprise any of the methods for effecting gene editing and/or gene expression alteration in one or more target cells in vivo disclosed herein. [0387] In some embodiments, the preventative, amelioration, or therapeutic method may comprise further administering another agent, together with or separately from the pharmaceutical composition according to the present disclosure. In some embodiments, the other agent may be one or more erythropoiesis stimulating agents. In some embodiments, the one or more erythropoiesis stimulating agents may be any of such agents disclosed herein. In some embodiments, the other agent may be another agent for SCD. In some embodiments, such another agent for SCD may be hydroxyurea, L- glutamine oral powder, crizanlizumab, a general pain medication, voxelotor, or any combination thereof. In some embodiments, a synergistic effect may be achieved by combining a pharmaceutical composition according to the present disclosure and at least one other agent for treating SCD. [0388] Monitoring and dosing [0389] The effect of any of the in vivo method may be monitored, and monitoring may be on any appropriate parameters. Non-limiting examples of such parameters include but are not limited to: (i) % HSCs and HSPCs in the blood or bone marrow with successful gene editing and/or gene expression alteration; (ii) the number ofHSCs and HSPCs in the blood or bone marrow with successful gene editing and/or gene expression alteration; (iii)% HSCs and HSPCs expressing HbF in the blood or bone marrow (e.g., the target gene is BCLI IA or KLFI); (iv) the number ofHSCs and HSPCs expressing HbF in the blood or bone marrow (e.g., the target gene is BCLI IA or KLFl); (v) the expression level of the at least one SCD-associated gene or gene product or molecule, optionally beta-globin, beta-globin (HbS variant), gamma-globin, HbF, HbA, BCLl IA, and/or KLFl; and (vi) changes in the symptom, which optionally may be changes in the frequency and/or levels of pain, swelling of hands and feet, infection, growth, and/or symptoms associated with vision. [0390] Monitoring may be effected periodically, e.g., weekly, every 2 weeks, monthly or every 2 months, to assess whether the subject comprises a sufficient number of normal (gene-edited) cells in their peripheral circulation, e.g., HSCs, HSPCs, MPPs, CMPs, MEPs, HPCs, erythroid progenitors (e.g., BFU-E, CFU-E), proerythroblasts, erythroblasts (basophilic erythroblasts, early erythroblasts (e.g., type I, type II), polychromatic erythroblasts, intermediate erythroblasts, acidophilic erythroblasts, late erythroblasts, normoblasts, reticulocytes before nucleus expulsion, reticulocytes, or erythrocytes, or any combination thereof. A sufficient amount refers to the number of gene-edited which is determined to preclude or inhibit the symptoms ofSCD, SCD, SCA, HbSC, or HbS P- thalassaemia, such as sickle cell crisis, vaso-occlusive crisis, acute cell syndrome, aplastic crisis, hemolytic crisis and the like. This will typically involve collecting blood samples from the subject periodically and assaying the genome of the collected peripheral cells thereof in order to determine the approximate number of gene-edited cells therein. [0391] In any of the in vivo methods disclosed herein, the dosing regimen (such as dose, frequency, injection duration, etc) may be adjusted based on such monitoring. [0392] Regardless of the injection route, the injection may be repeated as many times as need to for providing sufficient gene editing and/or gene expression alteration and/or sufficient prevention, amelioration, or treatment outcome. [0393] In some embodiments, the injection may be given two or more times, to reach e.g., a minimum of about 10%, about 15%, about 20%, about 30%, or an about final 15-30% or about final 20-40% HSCs and HSPCs with successful gene editing and/or gene expression alteration among the total HSCs and HSPCs (in the bone marrow or in the peripheral circulation). In some embodiments, the injection may be given two or more times, to reach e.g., a minimum of about 10%, about 15%, about 20%, about 30%, or an about final 15-30% or about final 20-40% HSCs and HSPCs with wildtype beta-globin expression (e.g., when the target gene is the HbS variant of HEB) among the total HSCs and HSPCs (in the bone marrow or in the peripheral circulation). In some embodiments, the injection may be given two or more times, to reach e.g., a minimum of about 10%, about 15%, about 20%, about 30%, or an about final 15-30% or about final 20-40% HSCs and HSPCs with HbF expression (e.g., when the target gene is BCLI IA or KLFJ) among the total HSCs and HSPCs (in the bone marrow or in the peripheral circulation). [0394] Regardless of the injection route, the injection may be repeated at any appropriate frequency for providing sufficient gene editing and/or gene expression alteration. In some embodiments, the injection may be given two or more times, optionally about 3-5 time, optionally about once a week, about every 2 weeks, or about every 3 weeks, about once a month, about every 3 months, about every 6 months, or about once per year. [0395] In some embodiments, the level of successfully modified (by gene editing or gene expression alteration) target cells (or differentiated cells thereof) may be monitored. In some embodiments, the level of cells expressing the intended phenotype ( e.g., expression of wildtype beta-globin or expression of HbF) may be monitored. In some embodiments, the dosing regimen such as injection dose, speed, or frequency may be adjusted based on the observation made during such monitoring. [0396] Definitions [0397] All references cited herein, including patent documents and non-patent documents, are hereby incorporated by reference in their entirety. Nothing herein is to be construed as an admission that the present invention is not entitled to antedate such disclosure by virtue of prior invention. [0398] Although various embodiments and examples of the present invention have been described referring to certain molecules, compositions, methods, or protocols, it is to be understood that the present invention is not limited to the particular molecules, compositions, methods, or protocols described herein, as theses may vary. It is also to be understood that the terminology used in the description is for the purpose of describing the particular versions or embodiments only and is not intended to limit the scope of the present invention which will be limited only by the appended claims. [0399] It should be understood that, unless clearly indicated otherwise, in any methods disclosed or claimed herein that comprise more than one step, the order of the steps to be performed is not restricted by the order of the steps specifically cited. [0400] 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 disclosure belongs. [0401] It must also be noted that, unless the context clearly dictates otherwise, the singular forms "a," "an," and "the" as used herein and in the appended claims include plural refence. Thus, the reference to "a cell" refers to one or more cells and equivalents thereof known to those skilled in the art, and so forth. Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by a person of skilled in the art. [0402] The term "about" or "approximately" means within a statistically meaningful range of a value. Such a range can be within an order of magnitude, preferably within 50%, more preferably within 20%, still more preferably within 10%, and even more preferably within 5% of a given value or range. The allowable variation encompassed by the term "about" or "approximately" depends on the particular system under study, and can be readily appreciated by one of ordinary skill in the art. [0403] In the specification above and in the appended claims, all transitional phrases such as "comprising," "including," "having," "containing," "involving," "composed of," and the like are to be understood to be open-ended, namely, to mean including but not limited to. Only the transitional phrases "consisting of' and "consisting essentially of' shall be closed or semi-closed transitional phrases, respectively. [0404] "BAF chromatin remodeling complex subunit BCLl lA", also referred to herein as "BCLl lA", is known to function as a repressor of the HBGJ and HBG2 genes and thus a key regulator of the switch from HbF to HbA. BCL 11 A may bind to the BCL 11 A-binding motif found in the promoter regions of the HBGI and HBG2 genes (see FIG. 3F). BCLl IA may have an amino acid sequence provided as GenBank: ADL14508.l. In one aspect, human BCLl IA has the amino acid sequence provided as SEQ ID NO: 6 or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like. In humans, BCLl IA is encoded by the BCLJ IA gene on chromosome 2, with gene location 2p16.l at nucleotide positions 60450520 to 60553654 (according to Gene Assembly GRCh38.pl3), which encodes nine exons (NCBI, Gene ID: 53335). In one aspect, the BCLI IA gene may have the polynucleotide sequence provided as NCBI Reference Sequence: NC 000002.12. [0405] The term "cargo" or "cargo molecule" as used herein is one or more materials carried by and/or encapsulated by/in a TCV according to the present disclosure. In some embodiments, the combination of materials carried by a TCV may be collectively referred to as a "cargo". For example, a TCV may carry a combination of a nuclease protein (such as Cas9) and a guide RNA (such as one comprising a sequence complementary to a target sequence) as a cargo. In some embodiments, a TCV may carry as a cargo a siRNA or a shRNA comprising a sequence complementary to a target sequence. In some embodiments, a TCV may carry as a cargo a miRNA comprising a sequence partially complementary (i.e., the percent complementarity is less than 100%) to a target sequence. [0406] The term "cholesterol derivative" as used herein, in its broadest sense, encompasses any derivatives of cholesterol. Non-limiting examples of cholesterol derivatives include: DC-Chol (N,N- dimethyl-N-ethylcarboxamidocholesterol), l ,4-bis(3-N-oleylamino-propyl)piperazine (Gao, et al. Biochem. Biophys. Res. Comm. 179,280 (1991); Wolf et al. Bio Techniques 23, 139 (1997); U.S. Pat. No. 5,744,335), or imidazole cholesterol ester (ICE) (US20210220273Al). [0407] Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated (Cas) systems are a class of genome-editing tools that target desired genomic sites in mammalian cells. A CRISPR/Cas system involves at least one Cas nuclease and a gRNA. Typically, the Cas nuclease may recognize a protospacer adjacent motif (PAM) sequence specific to the Cas nuclease in the target gene (sense or antisense strand) and if the gRNA is able to hybridize with a target sequence in the target gene proximate to the PAM site, the Cas nuclease may mediate cleavage of the target gene at about 2-6 nucleotides upstream of the PAM site. For example, the PAM sequence for Cas9 is 5'-NGG-3'. Type II CRISPR/Cas systems use Cas9 nuclease that is targeted to a genomic site by complexing with a guide RNA that hybridizes to an approximately 17-24-nucleotide DNA sequence immediately preceding an 5'-NGG-3' motif(where "N" can be any nucleotide) recognized by Cas9 (thus, a (N)20NGG target DNA sequence). This results in a double-strand break between the third and fourth nucleotides upstream of the NGG motif. The double strand break instigates either non- homologous end-joining (NHEJ), which typically leads to the introduction of one or more nucleotide insertions or deletions resulting in frameshift mutations that knock out gene alleles (e.g., nonsense- mediated mRNA decay (NMD)), or homology-directed repair (HDR), which can be exploited with the use of an exogenously introduced double-stranded or single-stranded DNA template to knock in or correct a mutation in the genome. [0408] The term "destabilizing agent" as used herein encompasses any agents that destabilizes the cargo of a TCV according to the present disclosure. In some embodiments, a destabilizing agent may destabilize or degrade a nucleic acid cargo such as a gRNA, a protein cargo such as Cas nuclease, and/or a RNP. Exemplary destabilizing agents include but are not limited to: organic solvents such as ethanol and detergents such as sodium dodecyl sulfate. In some embodiments, a TCV may be substantially free of destabilizing agents. In some embodiments, such a TCV may be [0409] The term "disease-associated gene" as used herein refers to a gene that is involved in and/or contributes to the pathogenesis or pathology of a disease or condition. Disease may be any disease such as but not limited to hematological diseases and cancer. In some embodiments, the disease is SCD. [0410] The term "erythropoiesis" as used herein refers to the general process in which hematopoietic stem cells (HSCs) or hematopoietic stem and progenitor cells (HSCs) develop into mature erythrocytes (Zivot et al., Mo! Med. 2018 Mar 23;24(1 ): 11.). HSCs or HS PCs, both of which have self-renewal capacity, give rise to multipotent progenitors (MPPs, also called short-term HSCs), which may develop directly into megakaryocyte-erythroid progenitors (MEPs) or first into common myeloid progenitors (CMPs) and then into MEPs (Erasmus et al., Cold Spring Harb Perspect Med 2013;3:a0l 1601). Cells in the stages from MPPs to MEPs belong to hematopoietic progenitor cells (HPCs), which do not have self-renewal capacity (Ferrari et al., Nat Rev Genet. 2021 Apr;22(4):216- 234.). Subsequently, MEPs may develop into erythroid progenitors of different differentiation levels (burst-forming unit erythroid cells (BFU-E); then colony-forming unit erythroid cells (CFU-E)), followed by proerythroblasts, and then erythroblasts of different differentiation levels (basophilic erythroblasts, also called early erythroblasts (further classified into type I and type II); polychromatic erythroblasts, also called intermediate erythroblasts; and acidophilic erythroblasts, also called late erythroblasts) (Valent et al., Haematologica. 2018 Oct;103(10):1593-1603.). Healthy erythroblasts may also be referred to as normoblasts. Late erythroblasts then go through expulsion of the nucleus, which leads to the formation ofreticulocytes. Reticulocytes exits the site of erythropoiesis ( e.g., yolk sac, liver, spleen, or bone marrow) and enter the bloodstream. RNA and micro-organelles contained in reticulocytes mediate further protein synthesis, which promotes maturation of reticulocytes into erythrocytes, i.e., mature red blood cells (RBCs) (Lee et al., Blood Cells Mo! Dis. Jun-Aug 2014;53(1- 2): 1-10.). [0411] Various pathways involving various factors (e.g., cytokines, cell surface receptors, transcription factors, etc) mediate erythropoiesis (Valent et al., Haematologica. 2018 Oct;l 03(10): 1593-1603; Sinclair. Biologics. 2013;7: 161-74. Epub 2013 Jul 3.). For example, interleukin-3 (IL-3), granulocyte-macrophage colony-stimulating factor (GM-CSF), and granulocyte colony-stimulating factor (G-CSF) mediate development from CMPs and MEPs into BFU-E. IL-3, GM-CSF, IL-9, and insulin-like growth factor-I (IGF-1) promote development during the BFU-E stage (early stage to late stage). IL-3, GM-CSF, IL-9, and erythropoietin (EPO) promote development from BFU-E to CFU-E, and EPO further promotes development ofCFU-E into proerythroblasts. Effects mediated by EPO via EPO receptor are dependent on iron-metabolism and the interaction between the death receptor FAS and its ligand (FAS-L). Development during the erythroid progenitor up to proerythroblast stages are also regulated by broadly acting hematopoietic cytokines including stem cell factor (SCF) and the SCF receptor KIT. Transferrin (Tf) and Tf-receptor-1 (TfR-1) are additional major regulators of erythropoiesis, and growth differentiating factor 11 (GDFl 1) and polymeric immunoglobulin A (IgA) are considered to be involved in the regulation of certain stages of erythropoiesis. FAS-Land GDFl 1 seem to be particularly involved in the final maturation stage that leads to the generation of erythrocytes. As for transcription factors, GAT A-1 triggers erythropoiesis by regulating the transcription of several erythroid differentiation-related genes, including genes involved in heme and/or globin synthesis, glycophorins, anti-apoptotic genes of the BH-3 family, genes involved in cell cycle regulation, and the gene for EPO receptor and therefor is the main regulator of lineage commitment, differentiation and survival of erythroid progenitors. Caspase is activated during the erythroblast stages and various caspase targets are affected, including Rock-1, Lamin Band Acinus. However, GATA-1 is protected from caspase cleavage by heat shock protein 70 (HSP70). Other factors (e.g., Activin A, TGF-beta, MCL-1, BCL-xL, HSP70, vitamin B 12, folic acid, copper, ferritin, ferroportin, hepcidin) and transcription factors (e.g., STAT5A and STAT5B) and negative regulators (e.g., inhibin, TGF-beta, BID, caspases) also play a role in erythropoiesis regulation, which is reviewed in detail in, e.g., Valent et al., Haematologica. 2018 Oct; 103(10): 1593-1603. [0412] The location that erythropoiesis takes place in depends on the developmental stage of an organism. In humans, during the fetal stage, erythropoiesis occurs in the blood islands of the yolk sac in the first 8 weeks of gestation and then the fetal liver between 8 and 32 weeks of gestation (Philipsen. Haematologica. 2014 Nov;99(11): 1647-9.; Sankaran et a., Br J Haematol. 2010 Apr; 149(2): I 81-94. Epub 2010 Mar 1.). The majority of erythropoiesis starts occurring in the bone marrow at around 32 weeks of gestation. Around birth the spleen serves as a transient erythropoietic organ, and the bone marrow takes over the majority erythropoiesis in about three months. The sites of erythropoiesis further change over time; red blood cell production recedes in the long bones (tibia, femur) by about age 25 and persists in the flat bones (sternum, skull, ribs, pelvis (e.g., iliac)), while minor contributions from the liver and less so from the spleen (Hom et al., Immunol Res. 2015 Dec;63(1-3):75-89.). Once reticulocytes are formed, reticulocytes enter into the circulation. The site of erythropoiesis within the fetal liver, spleen (specifically the red pulp), and bone marrow is called erythroblastic islands (Wanwani and Bieker. Curr Top Dev Biol. 2008; 82: 23-53.). [0413] "Guide RNA" or "gRNA", as used herein in relation to the CRISPR/Cas gene editing, refers to a piece of a RNA fragment that binds to a target DNA sequence and guide a Cas nuclease protein to the specific site of gene editing. In CRISPR/Cas gene editing, a gRNA may comprise or consist of: (A) a crispr RNA (crRNA), which comprises a crRNA repeat sequence and a target-complementary sequence or (B) a crispr RNA ( crRNA), which comprises a target-complementary sequence of about 15- 75 nucleotides that is complementary to the target DNA sequence; and a trans-activating crispr RNA (tracrRNA), which serves as a binding scaffold for the Cas nuclease. In (11), a gRNA may be comprise the two parts ( crRNA and tracrRNA) linked forming a single molecule, or a gRNA may be a complex of a crRNA molecule and a trcrRNA molecule. In some embodiments, the target- complementary sequence may comprise a GC content in the range of 40-80%, and in some embodiments, and the target-complementary sequence may have a length of 17-24 nucleotides. A gRNA may be a single-stranded gRNA (sgRNA) molecule. [0414] "Guide RNA" or "gRNA", as used herein in relation to the CRISPR/Cas gene editing (also referred to as "CRISPR-mediated gene editing), refers to a RNA fragment (e.g., single guide RNA ("sgRNA")) or a hybrid of two RNA fragments (e.g., dual guide RNA ("dgRNA")) that binds to a target DNA sequence and guide a Cas endonuclease protein to the specific site ofa DNA (e.g., in a genome) to allow for Cas-mediated cleavage of a DNA molecule. [0415] In some embodiments, a gRNA (e.g., one which may be used with Casl2a or Casl2a variants) may comprise a crispr RNA (crRNA), which comprises (I) a crRNA repeat sequence linked to (II) a target-complementary sequence of about 15- 75 nucleotides that is complementary to ( or comprising some mismatches relative to) the target DNA sequence. In certain embodiments, the gRNA may comprise the crRNA repeat sequence and the target-complementary sequence in the direction from the 5' to the 3'. In certain embodiments, the crRNA may comprise (i) an optional 5' - end sequence, (ii) a first stem sequence, (iii) a loop sequence, (iv) a second stem sequence, and (v) an optional 3 '-end sequence. The first and second stem sequences may be reverse complementary to each other and with the loop sequence may form a hairpin-like structure (which may be recognized, e.g., by Casl2a or Cas12a variants). In certain embodiments, the first stem sequence may comprise UCUAC, and the second stem sequence may comprise GUAGA. In certain embodiments, the loop sequence may comprise UCUU, UAAGU, UGUU, UUUU, UAUU, UGUUU, UUCG, or UUU. In certain embodiments, the optional 5'-end sequence may comprise UAAUU or AAUU. In certain embodiments, the optional 3' -end sequence may comprise U. In particular embodiments, the repeat sequence may comprise (i) an optional 5'-end sequence comprising UAAUU, (ii) a first stem sequence comprising UCUAC, (iii) a loop sequence comprising UCUU, (iv) a second stem sequence comprising GUAGA, and (v) an optional 3'-end sequence comprising U. In particular embodiments, the repeat sequence may comprise any one of SEQ ID NOS: 201-206. In particular cases, the repeat sequence may comprise SEQ ID NO: 201. [0416] In some embodiments, a gRNA (e.g., one which may be used with Cas9 or Cas9 variants) may be dgRNA comprising: (I) a crispr RNA (crRNA), which comprises (i) a target-complementary sequence of about 15- 75 nucleotides that is complementary to ( or comprising some mismatches relative to) the target DNA sequence and (ii) a crRNA flagpole sequence; and (II) a trans-activating crispr RNA (tracrRNA), which comprises (i) a tracrRNA flagpole sequence and (ii) tracrRNA endonuclease binding domain, which serves as a binding scaffold for the Cas endonuclease, wherein the crRNA and tracrRNA hybridize with each other via the flagpole sequences. In some embodiments, a gRNA may be sgRNA comprising (I) a crRNA sequence linked to (II) a trarRNA sequence as a single polynucleotide. [0417] In some embodiments, the dgRNA and sgRNA ( e.g., ones which may be used with Cas9 or Cas9 variants) may have the following formats: dgRNA crRNA (polynucleotide 1 having a crRNA sequence): [target-complementary sequence]-[crRNA flagpole sequence]-[(optional) crRNA first flagpole extension]-[(optional) crRNA second flagpole extension] * the sequence of[crRNA flagpole sequence]-[(optional) crRNA first flagpole extension]-[(optional) crRNA second flagpole extension] may be referred to herein as "crRNA backbone sequence". tracrRNA (polynucleotide 2 having a tracrRNA): [(optional) tracrRNA first extension]-[tracrRNA flagpole sequence]-[tracrRNA endonuclease binding domain] sgRNA (having a crRNA sequence linked to a tracrRNA sequence) [target-complementary sequence]-[crRNA flagpole sequence]-[(optional) crRNA first flagpole extension]-[(optional) linker]-[(optional) tracrRNA first extension]-[tracrRNA flagpole sequence]- [tracrRNA endonuclease binding domain] * the sequence of [ crRNA flagpole sequence]-[( optional) crRNA first flagpole extension]-[( optional) linker]-[(optional) tracrRNA first extension]-[tracrRNA flagpole sequence]-[tracrRNA endonuclease binding domain] may be referred to herein as "sgRNA backbone sequence". [0418] In some embodiments, the crRNA flagpole sequence may comprise SEQ ID NO: 131 or 132. In some embodiments, the optional crRNA first flagpole extension may comprise SEQ ID NO: 133. In some embodiments, the optional crRNA second flagpole extension may comprise SEQ ID NO: 134. In some embodiments, the optional tracrRNA first extension may comprise SEQ ID NO: 135. In some embodiments, the tracrRNA flagpole sequence may comprise SEQ ID NO: 136 or 137. In some embodiments, the tracrRNA endonuclease binding domain may comprise SEQ ID NO: 138. In some embodiments, the tracrRNA endonuclease binding domain may further comprise or may be followed by one or more uracil based, e.g., 5'-U-3', 5'-UU-3', 5'-UUU-3', 5'-UUUU-3', 5'-UUUUU-3', 5'- UUUUUU-3', 5'-UUUUUUU-3', or 5'-UUUUUUUU-3'. [0419] In certain embodiments, the crRNA flagpole sequence may comprise SEQ ID NO: 131 and the tracrRNA flagpole sequence may comprise SEQ ID NO: 136. In certain embodiments, the crRNA flagpole sequence may comprise SEQ ID NO: 132 and the tracrRNA flagpole sequence may comprise SEQ ID NO: 137. In some embodiments, the optional linker which links a crRNA and tracrRNA in a sgRNA may comprise or consist of SEQ ID NO: 139. [0420] In some embodiments, a sgRNA may comprise a sgRNA backbone sequence (the sequence which is placed 3' to a target-complementary sequence in a sgRNA) of any of SEQ ID NOS: 141-144. In certain embodiments, the sgRNA backbone sequence may be followed by one or more uracils. In particular embodiments, the sgRNA backbone sequence may be followed by 1-10 uracils, such as 3 uracils, 4 uracils, 5 uracils, 6 uracils, 7 uracils, or 8 uracils. [0421] In some embodiments, a dgRNA may comprise (I) a crRNA sequence comprising a crRNA backbone sequence (the sequence which is placed 3' to a target-complementary sequence in a crRNA) comprising SEQ ID NO: 145 and (II) a tracrRNA sequence comprising SEQ ID NO: 146. In some embodiments, a dgRNA may comprise (I) a crRNA sequence comprising a sgRNA backbone sequence (the sequence which is placed 3' to a target-complementary sequence in a crRNA) comprising SEQ ID NO: 147 and (II) a tracrRNA sequence comprising SEQ ID NO: 148. [0422] When Cas9 is used, in some embodiments, the target-complementary sequence may comprise a GC content in the range of 40-80%, and in some embodiments, and the target-complementary sequence may have a length of 17-24 nucleotides. [0423] A target-complementary sequence of a gRNA may be any appropriate length. While the most frequently used target-complementary sequence length is 20 nt, a longer or shorter target- complementary sequence may also be used. In some embodiments, a gRNA longer than 20 nt may be used. For example, Ran et al. demonstrated that longer gRNAs are commonly cleaved to a shorter length so that the target-complementary sequence is e.g., 20 nt and thus the complementarity in the segment in excess of20 nt may not be important, i.e., may or may not be complementary to a target sequence (Ran et al., Cell. 2013 Sep 12; 154(6): 1380-9.). In some embodiments, a gRNA shorter than 20 nt may be used. For example, Fu et al. demonstrated that truncated (i.e.,< 20 nt) gRNAs, which is as short as 17, 18, or 19 nt, may also target the same target as a corresponding 20 nt-long gRNA and perhaps even may have decreased off-target effects (Fu et al. Nat Biotechnol. 2014 March; 32(3): 279-284.). [0424] A target-complementary sequence of a gRNA may or may not comprise a mismatch relative to the target sequence. In some cases, a mismatch at a particular position may reduce gRNA specificity to the target sequence. For example, in the context of SpCas9, Cong et al demonstrated that complementarity at up to 11 nt from the 3'-end of a target-complementary sequence is more important than that at a more upstream region (Cong et al., Science. 2013 February 15; 339(6121): 819-823.). Again in the context of SpCas9, Zheng et al demonstrated that the core sequence which is from the 4 th to the 7 th nt from the 3' -end is more sensitive to target mismatch compared to the rest of the target- complementary sequence (Zheng et al., Sci Rep. 2017 Jan 18;7:40638.). Therefore, in some embodiments, a gRNA target-complementary sequence may comprise a mismatch relative to its target sequence outside of such a core position. (0425] The term "helper lipid" or "structural lipid" as used herein refers to a type of lipid that may be comprised in a TCV in addition to an ionizable cationic lipid. In some embodiments, a helper lipid may be a non-cationic lipid and may be neutral, zwitterionic, or anionic lipid. In some embodiments, a helper lipid may be a lipid that carries a net negative charge at a selected pH, such as physiological pH. Without wishing to be bound by theory, helper lipids in TCVs in general are used to provide particle stability and/or biocompatibility and/or to enhance cargo delivery efficiency. Non-limiting helper lipids include, but are not limited to dioleoylphosphatidylethanolamine (DOPE), distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), palmitoyloleoylphosphatidylcholine (POPC), palmitoyloleoyl-phosphatidylethanolamine (POPE), dioleoyl-phosphatidylethanolamine 4-(N- maleimidomethyl)-cyclohexane-1-carboxylate (DOPE-ma!), dipalmitoyl phosphatidyl ethanolamine (DPPE), dimyristoylphosphoethanolamine (DMPE), distearoyl-phosphatidyl-ethanolamine (DSPE), 16-O-monomethyl PE, 16-O-dimethyl PE, 18-1-trans PE, l-stearoyl-z-oleoyl- phosphatidyethanolamine (SOPE), or a mixture thereof. In some embodiments, a helper lipid is dioleoylphosphatidylethanolamine (DOPE). (0426] "Hemoglobin", "haemoglobin", "Hb", or "Hgb" is a tetrameric protein composed of both alpha-like and beta-like globin subunits (Sankaran et a., Br J Haematol. 2010 Apr; 149(2): 181-94. Epub 2010 Mar 1.). Each globin subunit is associated with the cofactor hem (also called haem), which can carry an oxygen molecule. In humans, genes encoding alpha-like globin subunits are located in the alpha-globin locus on Chromosome 16 and are under the control of a set of distal enhancers referred to as the multispecies conserved sequences (MCS); and genes encoding beta-like globin subunits are located in the beta-globin locus on Chromosome 11 and are under the control of a set of distal enhancers referred to as the locus control region (LCR) (Barbarani et al., Front Cell Dev Biol. 2021 Apr 1;9:640060.). The alpha-globin locus contains three functional alpha-like globin genes: the embryonic HBZ gene, which encodes zeta-globin, and the two fetal/adult HBA2 and HBAJ duplicated genes, which encode alpha2-globin and alpha l-globin, respectively. The beta-globin locus contains five functional beta-like globin genes: the embryonic HBE gene, which encodes epsilon-globin, the two highly homologous fetal HBG2 and HBGJ genes, which encode Gvgamma-globin (hemoglobin subunit gamma 2) and Avgamma-globin (hemoglobin subunit gamma 1) (which only differ by a single amino acid), respectively, and the two adult HBD and HBB genes, which encode delta-globin and beta-globin, respectively. Beta-globin accounts for about 98% of adult beta-like globin. The genes contained in the alpha- and beta-globin loci are sequentially expressed in a stage-specific manner that maintains the 1: 1 ratio between the alpha-like and beta-like globin chains, in a process known as "hemoglobin switching". Hb is mostly found in cells of the erythrocyte lineage. Hb synthesis starts at around the proerythroblast stage, and Hb continues to accumulate as proerythroblasts develop into basophilic, polychromatophilic, and orthochromatic erythroblasts (Zivot et al., Mo! Med. 2018 Mar 23;24(1):11.). [0427] "Hemoglobin subunit beta", also referred to herein as "beta-globin", "beta-globin subunit", "~-globin", "~-globin subunit", "HBB", or the like, is a component of the adult hemoglobin (HbA). Human beta-globin may have an amino acid sequence provided as NCBI Reference Sequence: NP_ 000509.1. In one aspect, human beta-globin has the amino acid sequence provided as SEQ ID NO: 1 or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like. In humans, beta-globin is encoded by the HEB gene on chromosome 11, with gene location 1 lp15.4 (Gene Assembly GRCh38.p13) (NCBI, Gene ID: 3043). In one aspect, the wildtype HEB gene may have the polynucleotide sequence provided as SEQ ID NO: 11 ( corresponding to nucleotide positions 5225464 to 5227071 of Chromosome 11 (according to Gene Assembly GRCh38.p13)), with the open reading frame sequence provided as SEQ ID NO: 12 (corresponding to positions 5225601 to 5227021 of Chromosome 11 ), encoding three exons. In some embodiments, human beta-globin may be encoded by a cDNA comprising the polynucleotide sequence of SEQ ID NO: 13. [0428] Several diseases and/or phenotypes are caused by one or more alterations in the HEB gene. According to the OMIM® database (https://www.omim.org/), such diseases and/or phenotypes include: Delta-beta thalassemia (Phenotype MIM number 141749, autosomal dominant); Erythrocytosis 6 (Phenotype MIM number 617980, autosomal dominant); Heinz body anemia (Phenotype MIM number 140700, autosomal dominant); Hereditary persistence offetal hemoglobin (Phenotype MIM number 141749, autosomal dominant); Methemoglobinemia, beta type (Phenotype MIM number 617971, autosomal dominant); Sickle cell anemia (Phenotype MIM number 603903, autosomal recessive); Thalassemia, beta (Phenotype MIM number 613985); Thalassemia-beta, dominant inclusion-body beta (Phenotype MIM number 603902, autosomal recessive; and resistance to Malaria, resistance to (Phenotype MIM number 611162). [0429] "Hemoglobin subunit gamma", also referred to herein as "gamma-globin", "gamma-globin subunit", "y-globin", "y-globin subunit", "HBG", or the like, is a component of the fetal hemoglobin (HbF). Human hemoglobin subunit gamma 1 may have an amino acid sequence provided as GenBank: EA W68804. l. In one aspect, human hemoglobin subunit gamma 1 has the amino acid sequence provided as SEQ ID NO: 8 or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like. In humans, hemoglobin subunit gamma 1 is encoded by the HBGJ gene on chromosome 11, with gene location 1 lp15.4 (Gene Assembly GRCh38.p14) (NCBI, Gene ID: 3047). In one aspect, the wildtype HBGJ gene may have the polynucleotide sequence corresponding to nucleotide positions 5248269 to 5249857 of Chromosome 11 (according to Gene Assembly GRCh38.p14)), encoding three exons. Human hemoglobin subunit gamma 2 may have an amino acid sequence provided as GenBank: AAI30460.1. In one aspect, human hemoglobin subunit gamma 2 has the amino acid sequence provided as SEQ ID NO: 9 or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like. In humans, hemoglobin subunit gamma 2 is encoded by the HBG2 gene on chromosome 11, with gene location 1 lp15.4 (Gene Assembly GRCh38.pl4) (NCBI, Gene ID: 3048). In one aspect, the wildtype HBG2 gene may have the polynucleotide sequence corresponding to nucleotide positions 5253188 to 5254781 of Chromosome 11 (according to Gene Assembly GRCh38.p14)), encoding three exons. [0430] "Hemoglobin switch" or "hemoglobin switching" is the process of developmental stage- specific expression of different globin genes. In humans, for beta-like globins, around week 6 of gestation, embryonic globin (epsilon-globin) is silenced and fetal globin (gamma-globin, which is G- gamma-globin or A-gamma-globin) starts to be expressed. Perinatally, the switch to adult globin (beta-globin) occurs; and for the alpha-like globins, a single switch from the embryonic globin (zeta- globin) to the adult globin (alpha-globin, which is alpha2 or alphal globin) occurs (Philipsen. Haematologica. 2014 Nov;99( 11 ): 1647-9. ). Therefore, around birth, the most abundant Hb switches from the fetal hemoglobin (HbF), which is a tetramer of two alpha and two gamma globins (a2y2), to the adult hemoglobin (HbA) ((>90%) form of adult Hb), which is a tetramer of two alpha and two beta globins (a2~2) (Kato et al., Nat Rev Dis Primers. 2018 Mar 15;4: 18010.). This switch is normally completed during infancy and typically lasts until approximately 6 months of age (Sankaran et a., BrJ Haematol. 2010 Apr;149(2):181-94. Epub 2010 Mar 1.). Notably, HbF binds oxygen with greater affinity than HbA, being functional when reactivated in adults (Lamsfus-Calle et al., Sci Rep. 2020 Jun 23; 10(1): 10133.). [0431] A variety of nuclear factors involved in transcriptional regulation have been suggested to be involved in globin gene regulation and switching. Such nuclear factors include but are not limited to: BCLl lA, KLFl, SOX6, GATAl, NF-E4, COUP-TF, DRED/TR2/TR4, MBD2, Ikaros-PYR complex, and BRG 1 (the catalytic subunit of the SWI/SNF complex) (Sankaran et a., Br J Haematol. 2010 Apr; 149(2): 181-94. Epub 2010 Mar 1.). For example, BCLl lA is a repressor of the HBGI and HBG2 genes and thus a key regulator of the switch from HbF to HbA and is crucial for the maintenance ofHbF silencing in humans; and KLFl was discovered as an activator of the HEB gene. In fact, transduction ofK562 cells with BCLl lA or KLFl increased the HEB transcript about 5.9- and 7.5- fold, respectively, and transduction ofK562 cells with both BCLI IA and KLFI increased the HEB transcript about 300-890-fold (Trakamsanga et al., Haematologica. 2014 Nov;99(11): 1677-85. Epub 2014 Aug 8.). When erythroid cells differentiated from human iPS cell-derived erythroid progenitor-I cells (HiDEP-1 cells, which express endogenous KLFl at a level similar to adult erythroid cells) were transduced with BCLI IA, a robust increase in the beta-globin protein expression was observed. As reviewed in Sankaran et al. (Sankaran et a., Br J Haematol. 2010 Apr; 149(2): 181- 94. Epub 2010 Mar 1.): SOX6 seems to have a role in repressing HbF; GATAl seems to repress HBGJ and/or HBG2 gene expression and have a direct role in Hb switching; NF-E4 increases HBGJ and HBG2 gene expression; COUP-TF is a repressor of HBGJ and HBG2 genes; DRED complex (heterodimer of nuclear orphan receptors TR2 and TR4) seems to repress expression of HBEJ, HBG 1 and HBG2 genes; MBD2 is a group of proteins (part of the methyl-CpG binding protein complex 1 (MeCPl), which contains the proteins Mi-2, MT Al, MTA2, MBD3, HDACl, HDAC2, RbAp46 and RbAp48) and is a repressor of HBGJ and HBG2 genes; and Ikaros-PYR complex appears to promote Hb switching; and BRG 1 (the catalytic subunit of the SWI/SNF complex) appears to active transcription of HBB. [0432] The term "intra man-ow", "intraosseous", "intraosseously", "IO" as used herein the administration route involving direct injection into the bone marrow. [0433] The term "ionizable cationic lipid" as used herein, refers to any lipid that can-ies a net neutral charge at about physiological pH but is capable of becoming positively charged at a lower pH, e.g., pH below about 7, below about 6.5, below about 6, below about 5.5, below about 5, or below about 4.5, typically below about 6, or between about 5 and 6.5, between about 5 and 6, or between about 5.5 and 6. Without wishing to be bound by theory, a net neutral charge helps toxicity, and positive charges under a low pH may be useful in forming a complex with a negatively charged cargo such as a nucleic acid molecule and/or protein. Becoming positive charges under as the pH decreases may also help release of the cargo from an endosome once in a cell (endosomal escape), e.g., by taking protons in an endosome thereby destabilizing and bursting the endosome. Examples of ionizable cationic lipids may include, for example, N,N-dimethyl-2,3-dioleyloxy)propylamine (DODMA), 6- ( (2-hexyldecanoyl )oxy )-N-( 6-( (2-hexyldecanoyl)oxy )hex yl)- N-( 4-hydroxybutyl)hexan-1-aminium (ALC-0315), 8-[(2-hydroxyethyl)[6-oxo-6-(undecyloxy)hexyl]amino [-octanoic acid, l-octylnonyl ester (SM-102), N,N-dioleyl-N,N-dimethylammonium chloride (DODAC), N,N-distearyl-N,N- dimethylammonium bromide (DDAB), N-( 1-(2,3-dioleoyloxy)propyl)-N ,N,N-trimethylammonium chloride (DOT AP), N-( 1-(2,3-dioleyloxyl)propyl)-N ,N,N-trimethylammonium chloride (DOTMA), 1,2-DiLinoleyloxy-N,N-dimethylaminopropane (DLinDMA), 1,2-Dilinolenyloxy-N,N- dimethylaminopropane (DLenDMA), 1,2-Dilinoleylcarbamoyloxy-3-dimethylaminopropane (DLin- C-DAP), l ,2-Dilinoleyoxy-3-(dimethylamino )acetoxypropane (DLin-DAC), 1,2-Dilinoleyoxy-3- morpholinopropane (DLin-MA), 1,2-Dilinoleoyl-3-dimethylaminopropane (DLinDAP), 1,2- Dilinoleyl thio-3-dimethylaminopropane (D Lin-S-DMA), l -Linoleoyl-2-linoleyloxy-3- dimethylaminopropane (DLin-2-DMAP), 1,2-Dilinoleyloxy-3-trimethylaminopropane chloride salt (DLin-TMA.Cl), 1,2-Dilinoleoyl-3-trimethylaminopropane chloride salt (DLin-T AR.Cl), 1,2- Dilinoleyloxy-3-(N-methylpiperazino )propane (DLin-MPZ), or 3-(N,N-Dilinoleylamino)-1,2- propanediol (DLinAP), 3-(N,N-Dioleylamino )-1,2-propanedio (DOAP), l ,2-Dilinoleyloxo-3-(2-N,N- dimethylamino )ethoxypropane (DLin-EG-DMA), 1,2-Dilinolenyloxy- N ,N-dimethylaminopropane (DLin-K-DMA), 2,2-Dilinoleyl-4-dimethylaminomethyl-[ 1,3]-dioxolane (DLin-K-DMA) or analogs thereof, (3aR,5s,6aS)-N,N-dimethyl-2,2-di((9Z,12Z)-octadeca-9,12-dien yl)tetrahydro-3 aH- cyclopenta[ d] [ 1,3]dioxol-5-amine (ALNY-100), N ,N-dimethyl-2,2-di-(9Z, 12Z)-9, 12-octadecadien- l- yl- l ,3-dioxolane-4-ethanamine (KC2), (6Z,9Z,28Z,31Z)-heptatriaconta-6,9,28,31-tetraen-19-yl 4- ( dimethylamino)butanoate (MC3), or a mixture thereof. [0434] Additional examples of ionizable cationic lipids include, but are not limited to, N-(2,3- dioleyloxyl)propyl-N,N-N-triethylammonium chloride ("DOTMA"); 1,2-Dioleyloxy-3- trimethylaminopropane chloride salt ("DOT AP. Cl"); 3 ~-(N-(N' ,N' -dimethylaminoethane )- carbamoyl)cholesterol ("DC-Chol"), N-(1-(2,3-dioleyloxyl)propyl)-N-2- ( sperminecarboxamido )ethyl)-N,N-dimethyl-ammonium trifl uoracetate ("DOSP A"), dioctadecylamidoglycyl carboxyspermine ("DOGS"), 1,2-dioleoyl-3-dimethylammonium propane ("DODAP"), and N-(l ,2-dimyristyloxyprop-3-yl)-N,N-dimethyl-N-hydroxyethyl ammonium bromide ("DMRIE"), and mixtures thereof. Additionally, a number of commercial preparations of cationic lipids can be used, such as, e.g., LIPOFECTIN (available from GIBCO/BRL), and LIPOFECTAMINE (available from GIBCO/BRL). [0435] The term "complementary" or "complementarity" means that a nucleic acid can form hydrogen bond(s) with another nucleic acid sequence by either traditional Watson-Crick or other non- traditional types of interactions such as Wobble-base pairing which permits binding of guanine and uracil. A percent complementarity indicates the percentage of residues in a nucleic acid molecule that can form hydrogen bonds with a second nucleic acid sequence. [0436] "Kruppel like factor l ", also referred to herein as "KLFl ", is known to function as an activator of the HEB gene and thus also a key regulator of the switch from HbF to HbA. KLFl may have an amino acid sequence provided as GenBank: AHA61454.1. In one aspect, human KLFl has the amino acid sequence provided as SEQ ID NO: 7 or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like. In humans, KLFl is encoded by the KLFJ gene on chromosome 19, with gene location 19pl 3.13 at nucleotide positions 12884422 to 12887201 (according to Gene Assembly GRCh38.pl 3), which encodes three exons (NCBI, Gene ID: 10661). In one aspect, the KLFJ gene may have the polynucleotide sequence provided as NCBI Reference Sequence: NC_000019.10. [0437] The term "mutation" <_:>r "point mutation" as used herein in relation to nucleic acid or nucleotide sequence means a change in a nucleotide in a DNA or RNA molecule. A mutation may be a change from a nucleotide to another nucleotide or deletion of a nucleotide or an insertion of a nucleotide. When a mutation causes replacement of a nucleotide with another nucleotide in an open reading frame, the mutation may cause an amino acid substitution ("missense mutation") or appearance of an early stop codon ("nonsense mutation") leading to a shorter protein product or may not cause any changes in the protein product ("silent mutation"). When a mutation causes insertion or deletion of a nucleotide in an open reading frame, unless the number of insertion or deletion is divisible by three, the mutation changes the grouping of the codons to be read ("frame shift mutation"), causing dramatic changes in the protein sequence. [0438] "Lipid-based TCVs" as used in are TCVs that comprise at least one lipid and encompass lipid nanoparticles. In some embodiments, a lipid-based TCV may comprise at least one ionizable cationic lipid. In some embodiments, a lipid-based TCV may comprise at least one helper lipid. In some embodiments, a lipid-based TCV may comprise at least one phospholipid. In some embodiments, a lipid-based TCV may comprise at least one cholesterol (or cholesterol derivative). In some embodiments, a lipid-based TCV may comprise, essentially consist of, or consist of at least one ionizable cationic lipid, at least one helper lipid, at least one phospholipid, and at least one cholesterol (or cholesterol derivative), and optionally polyethyleneglycol (PEG) or PEG-lipid. Exemplary TCVs include but not are limited to those described in Applicant's WO2020077007 Al. In some embodiments, a lipid-based TCV may comprise, essentially consist of, or consist of an ionizable cationic lipid, one or more phospholipids, and cholesterol, the ratio of which are about 20:30: 10:40 in mo! %. In some embodiments, a lipid-based TCV may comprise, essentially consist of, or consist of an ionizable cationic lipid, one or more phospholipids, cholesterol, and PEG-lipid, the ratio of which are about 20:30: 10:39: 1 in mo!%. TCVs may be generated using gentle mixing such as repeated manual reciprocation of the TCV-generating fluid in a pipette, micromixing optionally using staggered herringbone micromixer (SHM) or T-junction or Y-junction mixing, or extrusion methods, or other TCV-mixing methods as desired. [0439] The term "nuclease" as used herein refers to an enzyme capable of catalyzing the cleavage of phosphodiester bonds between nucleotides of nucleic acids. In the CRISPR/Cas system, which involves a gRNA and a CRISPR-associated (Cas) nuclease, the Cas nuclease recognizes a PAM sequence in the target gene (sense or antisense) and if the gRNA is able to hybridize with a target sequence of the target gene proximate to the PAM sequence, the Cas nuclease may mediate cleavage of the target gene at about 2-6 nucleotides upstream of the PAM. The PAM sequence is specific to the Cas nuclease. Any appropriate Cas nucleases may be used in the invention disclosed herein. Appropriate Cas nucleases include but are not limited to Casl2a including Cas12a of different bacterial species such as Acidaminococcus sp. (AsCasl2a, which recognizes the PAM sequence of 5'- TTTN-3 '), Lachnospiraceae bacterium (LbCas 12a, which recognizes the PAM sequence of 5' - TTTN-3 '), Francisella novicida (FnCas 12a, which recognizes the PAM sequence of 5 '-TTTN-3 '), Moraxella bovoculi (MbCasl2a, which recognizes the PAM sequence of 5'-TTTN-3'), Coprococcus eutactus (CeCasl2a, which recognizes the PAM sequence of 5'-TTTN-3'), or Butyrivibrio fibrisolvens (BfCas 12a, which recognizes the PAM sequence of 5' -TTTN-3 '), Cas 12b of different bacterial species such as of Bacillus hisashii (BhCas 12b, which recognizes the PAM sequence of 5 ' - TTTN-3', 5' -ATTN' 3', 5 '-GTTN-3 ') or of Alicyclobacillus acidophilus ( AacCasb, which recognizes the PAM sequence of5'-TTN-3'), Cas12f, which recognizes the PAM sequence of5'-TTTN-3' or its variants such as Un1Casl2fl, which recognizes the PAM sequence of 5'-TTTN-3' or CasMINI, which recognizes the PAM sequence of 5 '-TTTR-3', or Cas l 2j, which recognizes the PAM sequence of 5' -TTN-3', Cas9 including Cas9 of different bacterial species such as Streptococcus pyogenes (SpCas9, which recognizes the PAM sequence of 5' -NGG-3 '), Staphylococcus aureus Cas9 (SaCas9, which recognizes the PAM sequence of 5 '-NNGRR T-3 '), Streptococcus thermophilus (StCas9, which recognizes the PAM sequence of5'-NGGNG-3'), Neisseria meningitidis (NmCas9, which recognizes the PAM sequence of 5' -NNNNGATT-3 '), Francisella novicida (FnCas9, which recognizes the PAM sequence of 5 '-NG-3 '), Campylobacter jejuni (CjCas9, which recognizes the PAM sequence of 5' - NNNNACA-3'), Streptococcus canis (ScCas9, which recognizes the PAM sequence of5'-NNGG-3'), Staphylococcus auricularis (SauriCas9, which recognizes the PAM sequence of 5' -NNG-3 '), or any engineered variants thereof, including but not limited to SaCas9-HF, SpCas9-HF1, KKHSaCas9, eSpCas9, HypaCas9, Fokl-Fused dCas9, xCas9, SpRY (variant ofSpCas9), SpG (variant ofSpCas9), which are collectively referred to as Cas9 herein. Other Cas nuclease examples include Cas3, Cas8a2, Cas8b, Cas8c, Casl0, Csxl 1, Cas12, Cas13, Cas13a, Cas14, C2cl, C2c3, and C2c2. [0440] The terms "nucleic acid", "nucleic acid molecule", and "polynucleotide" are used interchangeably herein and encompass any compounds that comprise a polymer of nucleotides linked via a phosphodiester bond. Exemplary nucleic acids include but are not limited to RNA and DNA molecules, including molecules comprising cDNA, genomic DNA, synthetic DNA, and DNA or RNA molecules containing nucleic acid analogs. Nucleic acid molecules can have any three-dimensional structure. A nucleic acid molecule can be double-stranded or single-stranded (e.g., a sense strand or an antisense strand). Other non-limiting examples of nucleic acid molecules include genes, gene fragments, exons, introns, messenger RNA (mRNA), transfer RNA, ribosomal RNA, siRNA, micro- RNA, tracrRNAs, crRNAs, guide RNAs, ribozymes, cDNA, recombinant polynucleotides, branched polynucleotides, nucleic acid probes and nucleic acid primers. A nucleic acid molecule may contain unconventional or modified nucleotides. The terms "polynucleotide sequence" and "nucleic acid sequence" as used herein interchangeably refer to the sequence of a polynucleotide molecule. The nomenclature for nucleotide bases as set forth in 37 CFR § 1.822 is used herein. [0441] The term "phospholipid" as used herein refers to any lipid comprising a phosphate group. Non-limiting examples of suitable phospholipids include: distearoylphosphatidylcholine (DSPC), dioleoyl phosphatidylethanolamine (DOPE), dipalmitoylphosphatidylcholine (DPPC), phosphocholine (DOPC), dimyristoylphosphatidylcholine (DMPC), phosphatidylcholine (PLPC), 1,2- distearoyl-sn-glycero-3-phosphocholine (DAPC), phosphatidylethanolamine (PE), egg phosphatidylcholine (EPC), dilauryloylphosphatidylcholine (DLPC), dimyristoylphosphatidylcholine (DMPC), l-myristoyl-Z-palmitoyl phosphatidylcholine (MPPC), 1-palmitoyl-2-myristoyl phosphatidylcholine (PMPC), l-palmitoyl-z-stearoyl phosphatidylcholine (PSPC), 1,2-diarachidoyl- sn-glycero-S-phosphocholine (DBPC), l-stearoyl-2-palmitoyl phosphatidylcholine (SPPC), 1,2- dieicosenoyl-sn-glycero-3-phosphocholine (DEPC), palmitoyloleoyl phosphatidylcholine (POPC), lysophosphatidyl choline, dilinoleoylphosphatidylcholine distearoylphophatidylethanolamine (DSPE), dimyristoyl phosphatidylethanolamine (DMPE), dipalmitoyl phosphatidylethanolamine (DPPE), palmitoyloleoyl phosphatidylethanolamine (POPE), lysophosphatidylethanolamine, and combinations thereof. In one embodiment, the phospholipid is distearoylphosphatidylcholine (DSPC). [0442] The term "polyethyleneglycol-lipid" or "PEG-lipid" as used herein refers to any lipid modified or conjugated to one or more polyethyleneglycol (PEG) molecules. Without wishing to be bound by theory, containing PEG or a PEG-lipid in a TCV may help maintain TCV particle size (keep a TCV from getting too big) and/or help maintain particle stability in vivo. Some examples of PEG- lipids that are useful in the present invention may have a variety of "anchoring" lipid portions to secure the PEG to the surface of the lipid-based TCVs. Non-limiting examples of suitable PEG-lipids include PEG-myristoyl diglyceride (PEG-DMG) (e.g., 1,2-dimyristoyl-rac-glycero-3- methoxypolyethylene glycol-2000 (Avanti® Polar Lipids (Birmingham, AL)), which is a mixture of 1,2-DMG PEG2000 and 1,3-DMG PEG2000 (e.g., in about 97:3 ratio)), PEG- phosphatidylethanolamine and phosphatidic acid, PEG-ceramide conjugates (e.g., PEG-CerC14 or PEG-CerC20) which are described in U.S. Pat. No. 5,820,873, incorporated herein by reference, PEG- modified dialkylamines, and PEG-modified 1,2-diacyloxypropan-3-amines. Particularly examples include PEG-modified diacylglycerols and dialkylglycerols. [0443] The phrase "pharmaceutically acceptable" refers to molecular entities and compositions th/it are physiologically tolerable and do not typically produce an unintended and intolerable response such as an allergic response, when administered to a human. In some embodiments, the term "pharmaceutically acceptable", as used herein, means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans. [0444] The term "ribonucleoprotein", "RNP", or "RNP complex" as used herein refers to a complex of one or more RNA molecules and an RNA-binding protein. In the context of the CRISPR/Cas system, an RNP may be a complex of a gRNA and a Cas nuclease. The gRNA may be a RNA fragment containing a crRNA portion and a tracrRNA portion linked to each other or be a complex formed between a crRNA molecule and a tracrRNA molecule. [0445] "Sickle cell disease" or "SCD", as used herein, refers to a group of disorders caused by a mutation(s) in and/or altered expression of HBB, the gene encoding beta-globin (i.e., hemoglobin subunit beta) and may also be referred to as "~-hemoglobinopathies" (Kato et al., Nat Rev Dis Primers. 2018 Mar 15;4: 18010.). Exemplary SCDs include but are not limited to sickle cell anemia (SCA), Sickle cell-hemoglobin C (HbSC), and HbS ~-thalassaemia. HbA is a tetramer formed by two alpha-globin subunits and two beta-globin subunits, the latter of which are encoded by HBB. The sickle Hb (HbS) allele, ~S, is an HBB allele in which an adenine-to-thymine (A-to-T) substitution (HBB wild type (SEQ ID NO: 11) to HBB ~S allele (SEQ ID NO: 21)) results in the replacement of glutamic acid with valine at position 7 (Glu7Val) (HBB wild type (SEQ ID NO: 1) to HBB HbS variant (SEQ ID NO: 2)) in mature beta-globin (when the first methionine is counted as position 1). SCD occurs when both HBB alleles are mutated and at least one of them is the ~S allele. Hemoglobin S (also referred to as sickle Hb or HbS) (containing beta-globin subunits encoded by the ps allele) that is deoxygenated (not bound to oxygen) can polymerize, and HbS polymers can stiffen the erythrocyte. Individuals with one ps allele have the sickle cell trait (HbAS) but not SCD; individuals with SCA, the most common SCD genotype, have two ps alleles (PS/PS). [0446] Other relatively common SCD genotypes are also possible. Individuals with the HbSC genotype have one ps allele and one HEB allele with a different nucleotide substitution, pc allele, that generates another structural variant of Hb, hemoglobin C variant (HbC). pc is an HEB allele in which nucleic acid substitution (e.g., G to A; HEB wild type (SEQ ID NO: 11) to HEB pc allele (SEQ ID NO: 31)) results in the replacement ofglutamic acid with lysine at position 7 (Glu7Lys) (HBB wild type (SEQ ID NO: 1) to HBB HbC variant (SEQ ID NO: 3)) in mature beta-globin (when the first methionine is counted as position 1). The pc allele is mostly prevalent in West Africa or in individuals with ancestry from this region. HbSC disease ( caused by ps;pq is a condition with generally milder hemolytic anemia and less frequent acute and chronic complications than SCA, although retinopathy and osteonecrosis (also known as bone infarction, in which bone tissue is lost owing to interruption of the blood flow) are common occurrences. The ps allele combined with a null HEB allele (HbP0) that results in no protein translation causes HbSP0-thalassaemia, a clinical syndrome indistinguishable from SCA except for the presence of microcytosis ( a condition in which erythrocytes are abnormally small). The ps allele combined with a hypomorphic HBB allele (Hbji+; with a decreased amount of normal P-globin protein) results in HbSP+- thalassaemia, a clinical syndrome generally milder than SCA owing to low-level expression of normal HbA. Severe and moderate forms of Hbxb-thalassaemia are most prevalent in the eastern Mediterranean region and parts of India, whereas mild forms are common in populations of African ancestry. [0447] One proposed strategy for treating SCD is to correct a mutant HBB gene back to encode wild- type beta-globin or force express wild-type beta-globin. For example, Park et al. (Park et al. Nucleic Acids Res. 2019 Sep 5;47(15):7955-7972.) used CRISPR/Cas9 targeting the ps allele and a short single-stranded oligonucleotide template to correct the sickle mutation in PS in hematopoietic stem and progenitor Cells (hHSPCs) from peripheral blood or bone marrow ofSCD patients. The CRISPR treatment markedly increased normal HbA and reduced sickle cells. [0448] An alternative approach may involve reversing Hb switching by suppressing beta-globing expression and/or enhancing gamma-globin expression via modifying the enhance and/or repressor of a beta-like globin gene. For example, when the GATAl-binding site of the BCLl IA gene's enhancer was edited via the CRISPR/Cas9 system in SCD patient CD34+ HSCs, erythroid cells derived from such HSCs showed increased gamma-globin expression, and the sickling morphology was prevented (Wu et al., Nat Med. 2019 May;25(5):776-783. Epub 2019 Mar 25.). Similarly, targeting of the KLFJ gene via the CRISPR/Cas9 system in K562 cells also increased the HBG transcript levels and the HbF protein expression levels (Shariati et al., J Gene Med. 2016 Oct; 18(10):294-301.). Analogous observation was also made by other studies, such as Lamsfus-Calle et al., Sci Rep. 2020 Jun 23;10(1):10133. [0449] Currently, several gene therapy strategies for treating ~-hemoglobinopathies are being tested in the clinic or are about to enter the clinical stage. For example, Vertex Pharmaceuticals and CRISPR Therapeutics recently tested safety and efficacy of their CTX001, autologous CD34+ human HS PCs modified via the CRISPR/Cas9 system targeting the erythroid-specific enhancer region of BCLJ 1 A, in subjects with transfusion-dependent ~-thalassemia (TDT) or severe SCD (ClinicalTrials.gov Identifiers: NCT03655678 (CLIMB THAL-111) and NCT03745287 (CLIMB SCD-121); Frangoul et al., N Engl J Med 2021 ;384:252-60.). CD34+ HSPCs were collected from patients by apheresis after mobilization with either filgrastim and plerixafor or plerixafor alone. CTX00l was manufactured by performing gene editing ex vivo using the CRISPR/Cas system on the CD34+ HSPCs. Patients received busulfan myeloablation, followed the infusion of CTX001. Bluebird bio has been testing their LentiGlobin ("bb 1111 "), autologous CD34+ HSCs transduced ex vivo with the recombinant lentiviral vector encoding ~A-T87Q-globin, in patients with TDT or SCD (e.g., Clinical Trials.gov Identifiers: NCT02140554 (HGB-206) and NCT04293185 (HGB-210)). However, trials have been suspended after finding cancer cases. All these strategies involve harvesting HSCs, modifying HSCs ex vivo, myeloablation, and putting the modified HSCs back into the patients, which creates various difficulties and disadvantages. The lengthy treatment processes not only require an extremely high cost and a dedicated facility but also impose patients a physiological burden. Furthermore, because of such issues, especially considering SCDs are more prevalent in regions with less access to high- quality healthcare such as West Africa (Kato et al., Nat Rev Dis Primers. 2018 Mar 15;4:18010.), these therapeutic strategies would unlikely solve the global health problem. The current disclosure provides a method that circumvents the need ofHSC harvesting, myeloablation, and infusion of ex vivo edited HSCs. [0450] The tenn "Sickle cell disease-associated gene" or "SCD-associated gene" as used herein refers to any genes and their mutant forms involved in or associated with the pathogenesis and/or pathology of SCD, including both coding and noncoding sequences (e.g., exons and introns) and regulatory elements for the gene such as promoters and enhancers. SCD-associated genes include genes involved in Hb switching. Non-limiting examples ofSCD-associated genes include: HEB (e.g., the HbS variant), BCLJ JA, KLFJ, SOX6, GATAJ, NF-E4 ( or NFE4), COUP-TF, NR2Cl (also known as TR2), NR2C2 (also known as TR4), genes encoding members of the MBD2 protein complex, IKZFJ (also known as Ikaros), genes encoding other members of PYR complex (CHD4, HDAC2, RBBP7, SMARCBJ, SMARCCJ, SMARCC2, SMARCDJ, and SMARCEJ), and BRGJ (Sankaran et a., Br J Haematol. 2010 Apr;149(2): 181-94. Epub 2010 Mar 1.), and also genes that directly or indirectly regulate expression thereof. [0451] "Single-stranded oligo DNA nucleotides" or "ssODN" as used herein refers to a short DNA fragment of a single strand comprising a particular polynucleotide sequence that may be useful for some of the embodiments disclosed herein. In one aspect, ssODN may be used as part of CRISPR/Cas-mediated gene editing disclosed herein and may function as a DNA template (may also referred to as a DNA repair template, a repair template, or a template) to mediate a knock-in of a sequence of interest through the Cas9-mediated double-strand break site. Such a knock-in may be via homology-directed repair (HDR). In some embodiments, a ssODN may have homology to the strand that initiates repair in the direction of a desired modification. In some embodiments, a ssODN may comprise (i) a central region comprising one or more desired nucleic acids, sandwiched by (ii) a 5' homology arm and (iii) a 3' homology mm. Such a homology aim may comprise approximately 20- 2500 nucleotides (nt). 5' and 3' homology arms often have the same or similar nucleotide lengths (e.g., 0 or 1 to 10 nt difference), but 5' and 3' homology arms that significantly differ in length may also be used as long as the ssODN mediate an intended gene repair. 5' and/or 3' homology arms may be 100% complementary to the corresponding sequence in the original DNA sequence before gene editing or may have one or more (a few) mutations ( e.g., silent mutation) relative to the corresponding sequence in the original DNA sequence before gene editing. In some embodiments, ssODN may have one or more mutations at the PAM sequence ( or its reverse ( or antisense) sequence of to the PAM sequence, i.e., the opposite strand) and/or at one or more of the 5' -neighbouring bases of the PAM ( or the 3 '-neighbouring bases of the reverse ( or antisense) sequence corresponding to the PAM). In some cases, such a mutation(s) helps prevent or reduce Cas-mediated cleavage of the ssODN itself or of a gene-edited DNA molecule. In some embodiments, a ssODN may comprise complementarity to the gRNA strand. In some embodiments, a ssODN may comprise a total length of approximately 40-5000 nucleotides (nt). As a DNA repair template, a double-stranded DNA template may also be used instead. In such a case, one of the strands of the template may comprise the same sequence as a desired ssODN and the other strand have a sequence complementary thereto. [0452] The term "stem cell mobilization" as used herein refers to a process in which the movement of stem cells from the bone marrow into the blood is stimulated. In some embodiments, the stem cells mobilized may be HSCs and/or HSPCs. Exemplary agents that promote stem cell mobilization include G-CSF, GM-CSF, Plerixafor, and SCF (Hopman and DiPerio. Blood Rev. 2014 Jan; 28(1): 31--40. ). Other exemplary agents that promote stem cell mobilization include but are not limited to CXCR4 antagonists (e.g., POL6326, BKT-140, TG-0054), CXCL12 neutralizers (e.g., NOX-Al2), Sphingosine-1-phosphate (SIP) antagonists ( e.g., SEW2871), vascular cell adhesion molecule-I/Very Late Antigen 4 (VCAM/VLA-4) inhibitors (e.g., BIO 5192), parathyroid hormone, protease inhibitors (e.g., Bortezomib), Grof (e.g., SB-251353), hypoxia inducible factor (HIF) stabilizers (e.g., FG- 4497). [0453] A "subject" as used herein, which may be interchangeably referred to as "patient", "individual", or "animal", refers to a vertebrate including members of the mammalian species, such as canine, feline, lupine, mustela, rodent (racine, murine, etc.), equine, bovine, ovine, caprine, porcine species, and primates including humans. In specific embodiments, the subject is a human. In some embodiments, a subject may have or have a risk of developing a target disease. In specific embodiments, a subject may have or have a risk of developing SCD. [0454] The term "target cell" or "host cell" as used herein refers to a cell in which the cargo of a TCV according to the present disclosure is intended to function. A TCV according to the present disclosure may be engineered to specifically carry its cargo in a target cell, for example by comprising one or more targeting moiety on the surface. [0455] The term "target disease", as used herein, which may be used interchangeably with "target disorder" or "target condition", refers to a disease, disease, or condition that a TCV containing a cargo or a composition containing such a TCV according to the present disclosure is intended to treat, prevent, or ameliorate. A TCV according to the present disclosure may carry its cargo into a target cell, thereby altering a target gene or target gene expression and thus prevent, treat, or ameliorate a target disease. [0456] The term "target gene" or "target gene of interest" as used herein is a gene (including the gene itself and in some cases a polynucleotide region that regulates the expression of the gene such as a promoter and/or an enhancer of the gene) whose sequence is to be altered (e.g., disrupted, partially or entirely removed, or partially or entirely replaced with an intended sequence, for example by a nuclease (such as Cas9) and a guide RNA) or whose expression is to be altered (e.g., reduced or diminished or, in some cases, completely abrogated, for example by a siRNA, shRNA, or miRNA) by a cargo of a TCV according to the present disclosure. In general, "target gene" may be any gene of interest in a target cell. The sequence of "target gene" encompasses the sense antisense strand sequences of the gene. [0457] The term "target sequence" or "target polynucleotide sequence" as used herein is the sequence of a polynucleotide that a cargo of a TCV according to the present disclosure may interact with in a target cell to alter the target gene and/or target gene expression. [0458] The term "target site" as used herein refers to a sequence span within a target gene of about 1- 120 nt ( or about 1-100 nt or about 1-80 nt) which includes the nucleotide position(s) to be cleaved ("cut site(s)") by a given Cas nuclease (one position in case of a blunt-ended DBS (e.g., by Cas9) and two positions in case of a staggered DBS (e.g., by Cas12a)) and the sequence upstream of the cut site (e.g., about 0-60 nt upstream) and the sequence downstream of the cut site (e.g., about 0-60 nt downstream). The target sequence (to be targeted by a gRNA used with the Cas nuclease) may be fully included in the target site of about 20-120 nt. In some embodiments the target site may be comprised in at least one Sickle cell disease (SCD)-associated gene or a variant thereof and/or a promoter or enhancer thereof. [0459] The term "therapeutically effective amount/dose" refers to the quantity of a TCV or a pharmaceutical composition comprising such a TCV or its cargo that is sufficient to provide a therapeutic effect (which may be based on, e.g., the number or percentage of target cells in which the intended target gene alteration occurred, the overall change in the target gene expression, the amelioration of one or more symptom, the number or percentage of target cells exhibiting an intended phenotype such as morphology, etc) upon administration to a subject. [0460] The term "transfection competent vesicle" or "TCV" as used herein, in its broadest sense, encompasses any materials capable of carrying one or more cargoes, such as but not limited to a nucleic acid molecule (e.g., a DNA or a RNA) and/or a nucleic acid molecule complexed with a protein or peptide, into a cell. Examples ofTCVs include but are not limited to: compounds, such as calcium phosphate, polycations, cationic lipids, phospholipids, organic and nonorganic polymers, dendrimers, organic and nonorganic nanoparticles and nanobeads, and any combinations thereof; lipid-based compositions capable of carrying a nucleic acid molecule, such as liposomes and lipid nanoparticles (LNPs); plasmids; virus-like particles (VLPs); and viral vectors, such as retroviral, lentiviral, and adenoviral vectors. In some embodiments, a TCV may comprise a targeting moiety (e.g., antibody or antibody fragment such as a Fab fragment), which allows the TCV to carry its cargo preferentially into a target cell. In some embodiments, such a targeting moiety may be specific to HSCs, HSCPs, MPPs, CMPs, MEPs, HPCs, erythroid progenitors (e.g., BFU-E, CFU-E), proerythroblasts, erythroblasts (basophilic erythroblasts, early erythroblasts (e.g., type I, type 11), polychromatic erythroblasts, intermediate erythroblasts, acidophilic erythroblasts, late erythroblasts, nonnoblasts, or reticulocytes (before nucleus expulsion). [0461] As used herein, the term "treat," "treatment," or "treating" generally refers to the clinical procedure for reducing or ameliorating the progression, severity, and/or duration of a disease or of a condition, or for ameliorating one or more conditions or symptoms (preferably, one or more discernible ones) of a disease. In specific embodiments, the effect of the "treatment" may be evaluated by the amelioration of at least one measurable physical parameter of a disease, resulting from the administration of one or more therapies. The parameter may be, for example, gene expression profiles, the number of disease-affected cells, the percentage or frequency of disease-affected cells among the cells of the same lineage, disease-associated marker levels, and/or the presence or absence or levels of certain cytokines or chemokines or other disease-associated molecules and may not necessarily discernible by the patient. In some embodiments "treat", "treatment," or "treating" may result in and/or be evaluated based on the inhibition of the progression of a disease, either physically by, e.g., stabilization of a discernible symptom, physiologically by, e.g., stabilization of a physical parameter, or both. In some embodiments the terms "treat", "treatment" and "treating" refer to the reduction or stabilization of cancerous tissue or cells. Additionally, the terms "treat," and "prevent" as well as words stemming therefrom, as used herein, do not necessarily imply 100% or complete cure or prevention. Rather, there are varying degrees of treatment effects or prevention effects of which one of ordinary skill in the art recognizes as having a potential benefit or therapeutic effect. In this respect, the inventive methods can provide any amount of any level of treatment or prevention effects of a disease in a mammal. Furthermore, the treatment or prevention provided by the inventive method can include treatment or prevention of one or more conditions or symptoms of the disease being treated or prevented. Also, for purposes herein, "prevention" can encompass delaying the onset of the disease, or a symptom or condition thereof. [0462] As will be understood by one having ordinary skill in the art, for any and all purposes, such as in terms of providing a written description, all ranges disclosed herein also encompass any and all possible sub-ranges and combinations of sub-ranges thereof. Any listed range can be easily recognized as sufficiently describing and enabling the same range being broken down into at least equal halves, thirds, quarters, fifths, tenths, etc. As a non-limiting example, each range discussed herein can be readily broken down into a lower third, middle third and upper third, etc. As will also be understood by one skilled in the art all language such as "up to," "at least," "greater than," "less than," and the like include the number recited and refer to ranges which can be subsequently broken down into sub-ranges as discussed above. Finally, as will be understood by one skilled in the art, a range includes each individual member. Thus, for example, a group having 1-3 articles refers to groups having 1, 2, or 3 articles. Similarly, a group having 1-5 articles refers to groups having 1, 2, 3, 4, or 5 articles, and so forth. [0463] While in the above set forth preferred construction, specific elements have been recited in order to adequately illustrate the principles of this invention, it will be apparent to those skilled in the art that alterations and modifications in the construction and arrangement of the system may be made without thereby departing from the spirit of said invention. Changes of form, of details of construction and materials may be made without thereby departing from the spirit of invention set forth, which shall be limited only by the scope of the appended claims/ Examples are provided below to illustrate the present invention. These examples are not meant to constrain the present invention to any particular application or theory of operation. [0464) EXAMPLES [0465) Example 1: Preparation of transfection competent vesicles (TCVs) [0466] Materials [0467] 1,2-Dioleyloxy-3-dimethylamino-propane (DODMA) was purchased from Cayman Chemical (Ann Arbor, MI). 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) and 1,2-distearoyl-sn- glycero-3-phosphocholine (DSPC) were purchased from Avanti Polar Lipids (Alabaster, AL). Cholesterol was purchased from Sigma Aldrich (St. Louis, MO). All lipids were maintained as ethanol stocks. [0468] TCV formation [0469] Lipid components (ionizable cationic lipid, helper lipid, phospholipid, and cholesterol) were dissolved in ethanol at appropriate ratios to achieve a final concentration of about 20-35 mM total lipid. Unless otherwise noted, the lipid ratio ofDODMA:DOPE: DSCP:cholesterol = 20:30:10:40 mo!% was used. An aqueous phase was prepared containing about 25 mM sodium acetate (approximately pH 4) buffer. The two solutions were combined via rapid-mixing. Specifically, the organic phase containing lipids was mixed with the aqueous phase through a T-junction mixer (Ignite microfluidic mixer (Cat#NINO00l, Precision Nanosystems, Vancouver, Canada) fabricated to meet the specifications of the PEEK Low Pressure Tee Assembly (1/16, 0.02 in thru hole, Part# P-712) at a final flow rate of about 20 mL/min with about 1:3 organic:aqueous (v/v) ratio (Jeffs, Palmer, et al. Pharm Res. 2005;22(3): 362-372.; Kulkarni et al. Nanoscale. 2017 Sep 21 ;9(36): 13600-13609; Kulkarni et al. ACS Nano. 2018 May 22;12(5):4787-4795.). The resulting suspension was dialyzed against 1000-fold volume of 25 mM sodium acetate (approximately pH 4) buffer overnight at 4°C to remove ethanol. Following dialysis, empty TCVs were concentrated using the Amicon (Sigma- Aldrich) centrifugation filter. [0470] Analysis ofTCV lipid concentrations [0471] Lipid concentrations were adjusted to 10 mM, as determined by assaying for the cholesterol content using a T-Cholesterol Assay Kit (Waka Chemicals, Mountain View, CA) and extrapolating total lipid concentration as described elsewhere (Chen et al. J Control Release. 2014 Dec 28; 196: 106- 12.). [0472) Example 2: Preparation ofRNP [0473] Recombinant Cas9 nuclease protein and recombinant Cas12a nuclease protein were obtained from IDT (San Jose, CA). Single-stranded gRNAs (sgRNAs) designed by Applicant to be used with Cas9 were ordered from Synthego (Redwood City, CA) or IDT (Coralville, IA). gRNA designed by Applicant to be used as gRNAs with Cas12a were ordered from IDT (San Jose, CA). RNP formation was performed by combining a gRNA solution of about 10 µM with a Cas9 or Cas 12a solution of about 10 µM ( at an approximately equimolar ratio between the total gRNA and Cas) and allowing to stand at room temperature for about 5 minutes. [0474) Example 3: Preparation ofRNP-TCV with or without DNA repair template Encapsulation of RNP (no DNA template) [0475] An about 0.5-20 mM (e.g., 10 mM) TCV solution (about pH 4) and an about 0.5-20 µM RNP solution ( about pH7) were combined and mixed at a 467: 1 to 5000: 1 molar ratio (molar concentration for TCV is the concentration of total lipid components of the TCV). Typically, 8.33 µL ofa 10 mM TCV solution and 10 µL of about 5 µM RNP were combined. The mixture was then allowed to incubate at room temperature for 5 minutes. Co-encapsulation of RNP and DNA template [0476] An about 0.5-20 mM TCV solution (about pH 4), an about 0.5-20 µM RNP solution (about pH7), and an about 0.5-20 µM DNA template (e.g., ssODN) solution (about pH7) were combined at a 467: 1: 1 to 5000: 1: 1 molar ratio (molar concentration for TCV is the concentration of total lipid components of the TCV). Typically, 8.33 µL of a 10 mM TCV solution (molar of total lipid components) (about pH 4), 10 µL of a mixture containing about 5 µM RNP (about pH 7), and 5 µL of a 10 µM solution ofssODN (about pH 7) were combined. The mixture was then allowed to incubate at room temperature for 5 minutes. Encapsulation of DNA template separate from RNP [0477] An about 0.5-20 mM TCV solution (about pH 4) and an about 0.5-20 DNA template (e.g., ssODN) solution (about pH7) were combined at a 467: 1 to 5000: 1 molar ratio (molar concentration for TCV is the concentration of total lipid components of the TCV). Typically, 8.33 µL ofa 10 mM TCV solution (molar of total lipid components) (about pH 4) and 5 µL ofa 10 µM solution ofssODN (about pH 7) were combined. The mixture was then allowed to incubate at room temperature for 5 minutes. [0478] Example 4: Use of TCV s comprising gene editing constituents for effecting gene editing in HSCs, HSPCs, and/or bone marrow cells [0479] To first confirm that the TCVs according to the present disclosure are compatible with and are able to deliver a cargo to effect gene editing in HSCs, HSPCs, and/or bone marrow cells are harvested from a CRISPR/Cas gene editing reporter mouse, such as the "Ail4" mouse (FIG. lA). For example, the femur and tibia will be harvested, and bone marrow cells will be collected by pushing saline through the marrow using a syringe and a needle, followed by several cycles of washing. For HSCs and/or HSPCs, CD34+ cells may be isolated from the collected bone marrow cells via anti-CD34 staining followed by F ACS sorting or via a CD34+ cell magnetic isolation kit. [0480] TCVs are generated according to Example 1. The TCV components ofDODMA:DOPE: DSCP:cholesterol=20:30: 10:40 may be used. Any of the gRNAs designed to effect interruption of the terminators and/or stop codons is complexed with Cas nuclease (e.g., Cas9, for example of Streptococcus pyogenes (SpCas9)) to generate RNPs according to Example 2. RNP encapsulation by the TCV will be performed as in Example 3. [0481] The HSCs, HSPCs, and/or bone marrow cells are incubated with the RNP-TCV complex. Exemplary incubation protocol may be, for example, in Applicant's WO2020077007 Al. Successful gene editing is confirmed via expression of the reporter gene, measured e.g., by flow cytometry and/or fluorescence microscopy. [0482] Example 5: Use of TCV s comprising gene editing constituents for effecting gene editing via intramarrow injection in reporter mice [0483] Next, Applicant tested whether intramarrow injection of the phannaceutical compositions according to the present disclosure allows the TCV to carry its cargo for gene editing into HSCs, HSPCs, and/or bone marrow cells and provide intended gene editing. The same reporter mouse ("Ai14" mouse) as in Example 4 was used. [0484] About 30 µL ofa composition comprising the RNP-TCV complex (comprising about 2700 pmol Cas9-based RNPs per mL) comprising gRNAs targeting either the floxed-stop cassette ("IG- TOM00l" containing gRNAs "PS2", "PS3", "LaRo", and "LoxP", which comprise the target- complementary sequences of SEQ ID NOS: 56, 57, 58, and 59; the gRNAs when used with Cas9 allow for disruption of three repeats ofSV40 polyA, resulting in TdTomato expression (see FIG. lA bottom)) or the control gene luciferase ("IG-Luc00 l ") ("IG-LUC001 ") was injected into the bone marrow of the femur of both legs of Ai mice (2 month old) via intramarrow injection. Mice were monitored for signs of pain, abnormal gait, or distress. Two days (48 hours) later, mice were sacrificed, and the femurs and quadriceps were collected (blood samples may be also harvested at various timepoints, such as one, two, three, five, seven, 14, or 30 days after injection and analyzed in the same manner). Quadriceps muscles were snap frozen in dry ice and stored in the -80°C freezer. The femurs were cleaned and placed separately in a 6-well plate containing 3 mL of HBB medium placed on ice. The bone marrow cells were harvested, filtered using a 70 µm cell strainer, seeded at 3x 10 6 cells/ mL in a 24-wel plate, and cultured for 72 hours. [0485] The bone marrow cells were stained with eFluor® 660-conjugated anti-CD34 antibody (Invitrogen, used at 1: 100) and DAPI (used at l :50,000), mounted ion slides, and analyzed for the reporter gene expression using the fluorescence microscope Zeiss Axio. Images were taken at 20x magnification in three different channels for DAPI, reporter gene, and CD34 and analyzed using the "multi-point" tool of the Fiji software. Percentage of cells expressing the reporter gene among the total bone marrow cells and CD34+ cells in the bone marrow cells were calculated to evaluate successful gene editing in the cells. [0486] Results [0487] Exemplary results are shown in FIG. lB. [0488] Example 6: Use ofTCVs comprising gene editing constituents for effecting gene editing via IV injection following stem cell mobilization in reporter mice [0489] Applicant will confirm that injection of the pharmaceutical compositions according to the present disclosure to the peripheral circulation, following stem cell mobilization, allows the TCV to carry its cargo for gene editing into HSCs and/or HSPCs cells in the peripheral circulation and provide intended gene editing. The same RNP-TCV complex and the same reporter mouse as in Example 4 will be used. [0490] Mice will receive G-CSF (Filgrastim) at a dose of about 10 µg/kg/day for 4 days. On day 4, plerixafor is also administered at a dose of about 0.24 mg/kg body weight. About 5 days after last plerixafor administration, a composition comprising the RNP-TCV complex (comprising about 2700 pmol RNPs per mL) will be IV injected to the repo1ier mice at up to about 50 µl per minute for 5, 10, 20, 30, or 60 minutes. One, two, three, five, seven, or 14 days later, the whole blood will be harvested, and mice will be sacrificed. The bone marrow cells may also be harvested and analyzed similarly, as some fraction of peripheral stem cells can return to the bone marrow. [0491] The blood cells (and bone marrow cells) will be washed and stained with anti-CD34 antibody and analyzed for the expression of the reporter gene, e.g., by flow cytometry . Percentage of cells expressing the reporter gene among the total blood cells, (total bone marrow cells,) CD34+ cells in the blood, (CD34+ cells in the bone marrow cells) will be calculated to confirm successful gene editing in the cells. (0492] Example 7: SCD-associated gene editing in SCD HSCs, HSPCs, or bone marrow cells [0493] To confirm that the gRNAs targeting SCD-associated genes according to the present disclosure are compatible with and are able to induce intended gene editing in HSCs, HSPCs, and/or bone marrow cells are harvested from mice ( or another model animal) carrying at least one ~S allele (SCD mice, for example, Noguchi et al. Blood Cells Mol Dis. Nov-Dec 2001 ;27(6):971-7.). For example, the femur and tibia will be harvested, and bone marrow cells will be collected by pushing saline through the marrow using a syringe and a needle, followed by several cycles of washing. For HSCs and/or HSPCs, CD34+ cells may be isolated from the collected bone marrow cells via anti- CD34 staining followed by FACS sorting or via a CD34+ cell magnetic isolation kit. Alternatively, equivalent cells from SCD patients may be used. [0494] TCVs are generated according to Example I. The TCV components ofDODMA:DOPE: DSCP:cholesterol=20:30:10:40 may be used. Any of the gRNAs designed to target a SCD-associated gene may be prepared. [0495] In some experiments, for example, for targeting the ~S allele, gRNA may comprise the target- complementary sequence of SEQ ID NO: 25, 45, 47, or 49; for targeting BCLJ JA, gRNA may comprise the target-complementary sequence of SEQ ID NO: 71,271,273,275,277,279, 65, 67, or 69; for targeting KLFJ, gRNA may comprise the target-complementary sequence of SEQ ID NO: 75 or 77; and for targeting HBGJ and/or HBG2, gRNA may comprise the target-complementary sequence of SEQ ID NO: 87,281,283,285,287,289, or 85. The gRNA is complexed with Cas nuclease (e.g., Cas9, for example of Streptococcus pyogenes (SpCas9) or any other Cas which recognizes the PAM sequence of 5' -NGG-3 ') to generate RNPs according to Example 2. [0496] In some experiments, for example, for targeting BCLJ JA, gRNA may comprise the target- complementary sequence of SEQ ID NO: 71,277, or 279; and for targeting the promoter region of HBGJ and/or HBG2, gRNA may comprise the target-complementary sequence of SEQ ID NO: 87, 287, or 289. The gRNA is complexed with Cas nuclease (e.g., Cas12a, for example AsCas12 or LbCas12a, or any other Cas which recognizes the PAM sequence of5'-TTN-3' or 5'-TTTN-3') to generate RNPs according to Example 2. [0497] In some experiments, for example, for targeting BCLJ JA, gRNA may comprise the target- complementary sequence of SEQ ID NO: 271, 273, or 275; and for targeting the promoter region of HBGJ and/or HBG2, gRNA may comprise the target-complementary sequence of SEQ ID NO: 281, 283, or 285. The gRNA is complexed with Cas nuclease (e.g., Cas9, for example SpCas12, or any other Cas which recognizes the PAM sequence of 5'-NGG-3') to generate RNPs according to Example 2. [0498] For disrupting the EER of BCLJ JA or disrupting BCLl lA-binding to the promoter region of HBGJ and/or HBG2, an ODN, such as a ssODN or dsODN, having the sequence of SEQ ID NO: 190 or any other ODN sequences described herein for this purpose may also be prepared. For promoting mutation in the (BCLl lA-binding) promoter region of HBGJ and/or HBG2, an ODN, such as a ssODN or dsODN, having the sequence of SEQ ID NO: 191 and/or 192 or any other ODN sequences described herein for this purpose may also be prepared. For correcting a mutant HEB gene, a DNA repair template may also be prepared. For example, such a template may comprise any of SEQ ID NOS: 169-176 and 101-108. [0499] RNP (and optionally DNA template) encapsulation by the TCV will be performed as in Example 3. [0500] The HSCs, HSPCs, and/or bone marrow cells are incubated with the RNP-TCV or RNP-DNA template-TCV complex. Exemplary incubation protocol may be, for example, in Applicant's WO2020077007 Al. Successful gene editing is confirmed based on (i) the absence of the original gene and/or (ii) the presence of the corrected gene when a DNA template was used, using PCR. [0501] Example 8: SCD-associated gene editing via intramarrow injection in SCD mice [0502] Next, Applicant will confirm that intramarrow injection of the pharmaceutical compositions according to the present disclosure allows the TCV to cany it's CRISPR/Cas-effecting cargo for gene editing into HSCs, HSPCs, and/or bone marrow cells and provide gene editing (optionally including correction) to the SCD-associated gene(s). The same RNP-TCV or RNP-DNA template-TCV complex and the same SCD mice as in Example 7 ( or mice of another strain, such as wild-type mice (with gRNAs designed to target the ortholog of the corresponding human SCD-associated gene) or humanized mice) will be used. Treatment and analyses may be performed for example as described in Example 5. [0503] For example, a composition comprising the RNP-TCV or RNP-DNA template-TCV complex (comprising about 2700 pmol RNPs per mL) as in Example 7 will be injected into the bone marrow (e.g., of the femur) of the SCD mice at up to about 50 µI per minute for 5, 10, 20, 30, or 60 minutes. One, two, three, five, seven, 14, or 30 days later, the whole blood will be harvested, mice will be sacrificed, and the bone marrow cells will be harvested and washed. [0504] A portion of the blood cells and bone marrow cells will be stained with an anti-CD34 antibody to isolate CD34+ cells by sorting. Successful gene editing is confirmed based on (i) the absence of the original gene and/or (ii) the presence of the corrected gene when a DNA template was used, using PCR. Percentage of gene-edited cells among the total blood cells, total bone marrow cells, CD34+ cells in the blood, CD34+ cells in the bone marrow cells will be calculated to confirm successful gene editing in the cells. In some cases, the expression of HBG (gene and/or protein) and/or the amount of HbF will also be measured. (0505] Example 9: SCD-associated editing via IV injection following stem cell mobilization in SCD mice [0506] Applicant will then confirm that injection of the pharmaceutical compositions according to the present disclosure to the peripheral circulation, following stem cell mobilization, successfully lead to gene editing ( optionally including correction) of SCD-associated genes in HSCs and/or HS PCs cells in the peripheral circulation. The same RNP-TCV or RNP-DNA template-TCV complex and the same SCD mice as in Example 7 will be used. Treatment and analyses may be performed for example as described in Example 6. [0507] For example, mice will receive G-CSF (Filgrastim) at a dose of about l O µg/kg/day for 4 days. On day 4, plerixafor is also administered at a dose of about 0.24 mg/kg body weight. About 5 days after last plerixafor administration, a composition comprising the RNP-TCV or RNP-DNA template-TCV complex (comprising about 2700 pmol RNPs per mL) will be IV injected to the reporter mice at up to about 50 µl per minute for 5, 10, 20, 30, or 60 minutes. One, two, three, five, seven, or 14 days later, the whole blood will be harvested, and mice will be sacrificed. The bone marrow cells may also be harvested and analyzed similarly, as some fraction of peripheral stem cells can return to the bone marrow. [0508] The blood cells (and bone marrow cells) will be washed. A portion of the blood cells (and bone marrow cells) will be stained with an anti-CD34 antibody to isolate CD34+ cells by sorting. Successful gene editing is confirmed based on (i) the absence of the original gene and/or (ii) the presence of the corrected gene when a DNA template was used, using PCR. Percentage of gene-edited cells among the total blood cells, total bone marrow cells, CD34+ cells in the blood, CD34+ cells in the bone marrow cells will be calculated to confirm successful gene editing in the cells. [0509] Example 10: HBB gene correction via intramarrow injection in SCD mice [051 O] Applicant will further confirm that addition of a DNA repair template according to the present disclosure in the pharmaceutical composition successfully corrects a HEB gene by intramarrow injection. [0511] TCVs are generated according to Example 1. The TCV components ofDODMA:DOPE: DSCP:cholesterol=20:30: I 0:40 may be used. A gRNA may comprise the target-complementary sequence of SEQ ID NO: 25, 45, 47, or 49. The gRNA is complexed with Cas nuclease (e.g., Cas9, for example of Streptococcus pyogenes (SpCas9)) to generate RNPs according to Example 2. A ssODN according to the present disclosure (e.g., for correction to wildtype beta-globin-encoding sequence, any of SEQ ID NOS: 169-176 and 101-108 or a sequence complementary thereto) or a double stranded DNA template having such a ssODN sequence will be encapsulated in TCVs in a similar manner to Example 3 (template may be encapsulated in TCVs separately from RNPs or together with RNPs). Treatment ofSCD mice will be performed in a similar manner to Example 8. [0512] The blood cells and bone marrow cells will be harvested and washed. A portion of the blood cells and bone marrow cells will be stained with an anti-CD34 antibody to isolate CD34+ cells by sorting. Successful gene correction is confirmed based on (i) the absence of the original gene and/or (ii) the presence of the corrected sequence using PCR followed by sequencing. Percentage of gene- corrected cells among the total blood cells, total bone marrow cells, CD34+ cells in the blood, CD34+ cells in the bone marrow cells will be calculated to confirm successful gene correction in the cells. [0513] Example 11: HBB gene correction via IV injection following stem cell mobilization in SCD mice [0514] Applicant will further confirm that addition of a DNA repair template according to the present disclosure in the pharmaceutical composition successfully corrects a SCD-associated gene by IV injection following stem cell mobilization. [0515] The pharmaceutical composition comprising the repair DNA template used in Example 10 will be used. Treatment of SCD mice will be performed in a similar manner to Example 9. [0516] The blood cells (and bone marrow cells) will be washed. A portion of the blood cells (and bone marrow cells) will be stained with an anti-CD34 antibody to isolate CD34+ cells by sorting. Successful gene correction is confirmed based on (i) the absence of the original gene and/or (ii) the presence of the corrected sequence using PCR followed by sequencing. Percentage of gene-corrected cells among the total blood cells, total bone marrow cells, CD34+ cells in the blood, CD34+ cells in the bone marrow cells will be calculated to confirm successful gene correction in the cells. [0517] Example 12: BCLJ JA gene editing in human cells. [0518] In this Example, the ability to disrupt the erythroid-enhancer region (EER) in intron 2 of BCLJ IA in human cells was tested. The editing strategy is visualized in FIG. 2A. 12-1: Using Cas9 [0519] TCVs comprising DODMA:DOPE: DSCP:cholesterol = 20:30:10:40 mo!% were generated according to Example 1. RNPs for targeting the EER of BCLJ 1 A were generated by complexing a sgRNA comprising the target-complementary sequence of SEQ ID NO: 65 (complementary to region 1 ofEER (EER 1) comprising SEQ ID NO: 64) or SEQ ID NO: 69 (complementary to region 2 of EER (EER 2) comprising SEQ ID NO: 68) with spCas9, as described in Example 2. Control RNPs for targeting luciferase were generated in the same manner using a sgRNA comprising the target- complementary sequence of SEQ ID NO: 55. RNPs were encapsulated by the TCVs as described in Example 3. [0520) HEK293 cells were seeded at a density of 80,000 cells/mL on Day 0. Twenty-four hours after seeding (Day 1), the TCV-encapsulated RNPs targeting either luciferase or BCLI IA EER were added to culture media at a final RNP concentration of 50 nM. Two hours after treatment (Day 1 ), culture media was changed, and cells were incubated for 46hours prior to harvest. On Day 2, cells were harvested, and genomic DNA was extracted. A 788 base pair region of DNA flanking the two target sites of BCLI IA EER was amplified using the forward and reverse primers of SEQ ID NOS: 61 and 62, respectively, and sent for Sanger sequencing. Percent editing efficiency at the target site was assessed using the Tracking oflndels by Decomposition (TIDE) analytical tool. [0521) Exemplary results are shown in FIG. 2B. As shown in the graph, successful editing was observed at the respective target sites, BCLI IA EER 1 and BCLI IA EER 2. [0522) Additionally, the same study may be performed using a sgRNA comprising the target- complementary sequence of SEQ ID NO: 271,273, or 275. 12-2: Comparing Cas9 and Cas12a [0523) TCVs comprising DODMA:DOPE: DSCP:cholesterol = 20:30:10:40 mo!% were generated according to Example 1. Cas12a-based RNPs for targeting the EER of BCLI IA were generated by complexing a gRNA comprising the target-complementary sequence of SEQ ID NO: 71 ( complementary to SEQ ID NO: 70) with AsCas 12a, as described in Example 2. Cas9-based RNPs for targeting the EER of BCLI I A were generated by complexing a gRNA comprising the target- complementary sequence of SEQ ID NO: 65 or 69 (complementary to EER I and EER2, respectively) comprising SEQ ID NO: 64) with SpCas9, as described in Example 2. Control RNPs for targeting an irrelevant gene (HBGI promoter) were also generated in the same manner. RNPs were encapsulated by the TCVs as described in Example 3. [0524) HEK293 cells were seeded at a density of 40,000 cells/mL on Day 0. Twenty-four hours after seeding (Day 1), the TCV-encapsulated RNPs targeting either the irrelevant gene or BCLI IA EER were added to culture media at a final RNP concentration of I 00 nM. Two hours after treatment (Day 1), culture media was changed, and cells were incubated for 46 hours prior to harvest. On Day 2, cells were harvested, and genomic DNA was extracted. A 788 base pair region of DNA flanking the target sites of BCLI IA EER was amplified using the forward and reverse primers of SEQ ID NOS: 61 and 62, respectively, and sent for Sanger sequencing. Percent editing efficiency at the target site was assessed using the TIDE analytical tool. [0525) Exemplary results are shown in FIG. 2D (left). As shown in the graph, successful editing was observed at the respective target sites with all three RNPs and the Cas 12a-based RNP provided the highest editing efficiency. [0526) Additionally, the same study may be performed using a sgRNA comprising the target- complementary sequence of SEQ ID NO: 271,273, or 275 with Cas9 and a sgRNA comprising the target-complementary sequence of SEQ ID NO: 277 or 279 with Cas12a. 12-3: Comparing dif erent gRNAs with Cas12a [0527] TCVs comprising DODMA:DOPE: DSCP:cholesterol = 20:30: 10:40 mo!% were generated according to Example l. Cas l 2a-based RNPs for targeting the EER of BCLJ 1 A were generated by complexing a gRNA comprising the target-complementary sequence of SEQ ID NO: 71 ("BCLl IA EER_Casl2a", complementary to SEQ ID NO: 70), SEQ ID NO: 277 ("BCLl lA EER_Cas12a_2", complementary to SEQ ID NO: 276), or SEQ ID NO: 279 ("BCLl lA EER_Cas12a_3", complementary to SEQ ID NO: 278) with AsCas12a, as described in Example 2. Control RNPs for targeting an irrelevant gene (HBGJ and HBG2 promoter, "HBG promoter_Casl2a ("-'ve control") were also generated in the same manner. RNPs were encapsulated by the TCVs as described in Example 3. [0528] HEK293 cells were seeded at a density of 40,000 cells/mL on Day 0. Twenty-four hours after seeding (Day 1 ), the TCV-encapsulated RNPs targeting either the irrelevant gene or BCLI 1 A EER were added to culture media at a final RNP concentration of 100 nM. Two hours after treatment (Day 1), culture media was changed, and cells were incubated for 46 hours prior to harvest. On Day 2, cells were harvested, and genomic DNA was extracted. A 788 base pair region of DNA flanking the target sites of BCLJ JA EER was amplified using the forward and reverse primers of SEQ ID NOS: 61 and 62, respectively, and sent for Sanger sequencing. Percent editing efficiency at the target site was assessed using the TIDE analytical tool. [0529] Exemplary results are shown in FIG. 2D (right). As shown in the graph, successful editing was observed at the respective target sites with the gRNAs "BCLl 1 A EER _ Cas 12a" and "BCLl l A [0530] TCVs comprising DODMA:DOPE: DSCP:cholesterol = 20:30: I 0:40 mo!% were generated according to Example 1. Cas12a-based RNPs for targeting the EER of BCLI JA were generated by complexing a gRNA comprising the target-complementary sequence of SEQ ID NO: 71 (complementary to SEQ ID NO: 70) with AsCas12a, as described in Example 2. Cas9-based RNPs for targeting the EER of BCLJ 1 A were generated by complexing a gRNA comprising the target- complementary sequence of SEQ ID NO: 65 or 69 ( complementary to EER 1 or EER2, respectively, comprising SEQ ID NO: 64 or 68, respectively) with SpCas9, as described in Example 2. Control RNPs for targeting an irrelevant gene (HBGJ promoter) were also generated in the same manner. ssODN ("HTT C680p_R") comprising SEQ ID NO: 190 which is not complementary to the sequence encompassing and/or proximate to the target sequence of the EER of BCLI JA (see, e.g., FIG. 2G, which shows below 50% sequence match between "HTT C680_R" and a 123-base sequence of BCLl lA EER around the target cut site(s), even when many gaps (63 gaps are shown) (shown as "" (space, no mark) are allowed) was prepared. RNPs were encapsulated by the TCVs as described in Example 3, and ssODN was encapsulated by the TCVs as described in Example 3. [0531] HEK293 cells were seeded at a density of 40,000 cells/mL on Day 0. Twenty-four hours after seeding (Day 1), the TCV-encapsulated RNPs targeting either the irrelevant gene or BCLI JA EER and the TCV-encapsulated ssODN were added to culture media at a final RNP concentration of 100 nM and a final ssODN concentration of 50 nM. Two hours after treatment (Day 1 ), culture media was changed, and cells were incubated for 46 hours prior to harvest. On Day 2, cells were harvested, and genomic DNA was extracted. A 788 base pair region of DNA flanking the target sites of BCLJ IA EER was amplified using the forward and reverse primers of SEQ ID NOS: 61 and 62, respectively, and sent for Sanger sequencing. Percent editing efficiency at the target site was assessed using the TIDE analytical tool. [0532] Exemplary results with the Cas9-based and Cas12a-based RNPs are shown in FIG. 2E left and right, respectively. As shown in the graph, editing efficiency with all of the three RNPs was enhanced by the ssODN that is not complementary to and thus is unlikely to form a dimer with the sequence of the target site or of the region (e.g., from about 40 nt to about 140, 130, 125, 123, or 120 nt, about 60-125 nt, about 70-125 nt, about 80-125 nt, about 90-125 nt, about 100-125 nt, about 110- 125 nt, about 45 nt, about 55 nt, about 60 nt, about 70 nt, about 80 nt, about 90, about 100 nt, about 110 nt, about 120 nt, or about 123 nt) encompassing the target site as shown in FIG. 2G-2I. [0533] Additionally, the same study may be performed using a sgRNA comprising the target- complementary sequence of SEQ ID NO: 271, 273, or 275 with Cas9 and a sgRNA comprising the target-complementary sequence of SEQ ID NO: 277 or 279 with Cas12a. 12-5: Using complementary ssODN [0534] TCVs comprising DODMA:DOPE: DSCP:cholesterol = 20:30: 10:40 mol% were generated according to Example 1. Cas9-based RNPs for targeting the EER of BCLJ I A were generated by complexing a gRNA comprising the target-complementary sequence of SEQ ID NO: 65 (complementary to EER 1 comprising SEQ ID NO: 64) with SpCas9, as described in Example 2. Control Cas9-based RNPs for targeting an irrelevant gene ( GFP) were also generated in the same manner. Cas12a-based RNPs for targeting the EER of BCLI IA were generated by complexing a gRNA comprising the target-complementary sequence of SEQ ID NO: 71 ( complementary to SEQ ID NO: 70) with AsCas12a, as described in Example 2. Control Cas12a-based RNPs for targeting an irrelevant gene ( GFP) were also generated in the same manner. An 80-base long ssODN ("BCLl 1 A EER_80bp F" or "BCLl lA EER_80bp R" comprising SEQ ID NO: 305 or 306, respectively) which is complementary to the sequence encompassing the target sequence of the EER of BCLI I A was prepared. RNPs were encapsulated by the TCVs as described in Example 3, and ssODN was encapsulated by the TCVs as described in Example 3. [0535] HEK293 cells were seeded at a density of 40,000 cells/mL on Day 0. Twenty-four hours after seeding (Day 1 ), the TCV-encapsulated RNPs targeting either the irrelevant gene or BCLJ I A EER and the TCV-encapsulated ssODN were added to culture media at a final RNP concentration of 100 nM and a final ssODN concentration of 50 nM (RNP:ssODN = 2: 1 molar ratio). Two hours after treatment (Day 1 ), culture media was changed, and cells were incubated for 46 hours prior to harvest. On Day 2, cells were harvested, and genomic DNA was extracted. A 788 base pair region of DNA flanking the target sites of BCLJ 1 A EER was amplified using the forward and reverse primers of SEQ ID NOS: 61 and 62, respectively, and sent for Sanger sequencing. Percent editing efficiency at the target site was assessed using the TIDE analytical tool. [0536] Exemplary results with the Cas9-based and Cas12a-based RNPs are shown in FIG. 2J left and right, respectively. As shown in the graph, editing efficiency by Cas9-based RNPs was enhanced by "BCLl IA EER_80bp F" and "BCLl IA EER_80bp R" and editing efficiency by Cas12a-based RNPs was enhanced with "BCLl IA EER_80bp F". (0537) Example 13: Dose-dependent BCLllA gene editing in human cells. [0538] In this Example, the effect of different doses of RNPs on editing of BCLJ 1 A EER in human cells was tested. [0539] TCVs comprising DODMA:DOPE: DSCP:cholesterol = 20:30: 10:40 mo!% were generated according to Example 1. Cas9-based RNPs for targeting the EER of BCLJ JA were generated by complexing a sgRNA comprising the target-complementary sequence of SEQ ID NO: 65 with spCas9, as described in Example 2. Control Cas9-based RNPs for targeting luciferase were generated in the same manner using a sgRNA comprising the target-complementary sequence of SEQ ID NO: 55. RNPs were encapsulated by the TCVs as described in Example 3. [0540] HEK293 cells were seeded at a density of 40,000 cells/mL on Day 0. Twenty-four hours after seeding (Day 1), the TCV-encapsulated Cas9-based RNPs targeting either luciferase or BCLJ JA EER 1 were added to culture media at a final RNP concentration of either 25, 50, 100 or 200nM (same stoichiometry ofTCV: RNP as in Example 12-1 but added at increasing volumes). Two hours after treatment (Day 1 ), culture media was changed, and cells were incubated for 46 hours prior to harvest (Day 3). On Day 3, cells were harvested, and genomic DNA was extracted. A 788 base pair region of DNA flanking the BCLJ JA EER I target site was amplified and sequenced as described in Example 12. Percent editing efficiency at the target site was assessed using the TIDE analytical tool. [0541] Exemplary results are shown in FIG. 2C. As shown in the graph, dose-dependent editing was observed at the target site (BCLJ JA EER 1). [0542) Example 14: Gene editing in the BCLllA-binding site in the promoter region of HBGJ and HBG2 in human cells. [0543] In this Example, the ability to disrupt the BCLl lA-binding site in the promoter region of HBGJ and HBG2 in human cells was tested. The editing strategy is visualized in FIG. 3A. 14-1: Using Cas9 [0544] TCVs comprising DODMA:DOPE: DSCP:cholesterol = 20:30:10:40 mo!% were generated according to Example 1. RNPs for targeting the BCLl lA-binding site in the promoter region of HBGJ and HBG2 were generated by complexing a sgRNA comprising the target-complementary sequence of SEQ ID NO: 85 ( complementary to the promoter region comprising SEQ ID NO: 84, designed to hybridize to both the BCLl !A-binding promoter region of HBGJ and the BCLl lA- binding promoter region of HBG2) with spCas9, as described in Example 2. Control RNPs for targeting luciferase were generated in the same manner using a sgRNA comprising the target- complementary sequence of SEQ ID NO: 55. RNPs were encapsulated by the TCVs as described in Example 3. [0545] HEK293 cells were seeded at a density of 80,000 cells/mL on Day 0. Twenty-four hours after seeding (Day 1), TCV-encapsulated RNP targeting either luciferase or HBGJ and HBG2 promoter regions were added to culture media at a final RNP concentration of 50 nM. Two hours after treatment (Day 1 ), culture media was changed, and cells were incubated for 22 hours prior to harvest (Day 2). On Day 2, cells were harvested, and genomic DNA was extracted. A DNA region flanking the target HBG promoter site of HBG 1 was amplified using the forward and reverse primers of SEQ ID NOS: 81 and 82, respectively, and sent for Sanger sequencing. A DNA region flanking the target HBG promoter site of HBG2 was amplified using the forward and reverse primers of SEQ ID NOS: 91 and 92, respectively, and sent for Sanger sequencing. Percent editing efficiency at HBGJ and HBG2 promoters was assessed using the TIDE analytical tool. [0546] Exemplary results are shown in FIG. 3B. As shown in the graph, successful editing was observed in both of the target sites (the promoter of HBGJ and the promoter of HBG2). [0547] Additionally, the same study may be performed using a sgRNA comprising the target- complementary sequence of SEQ ID NO: 281, 283, or 285. 14-2: Comparing Cas9 and Cas12a [0548] TCVs comprising DODMA:DOPE: DSCP:cholesterol = 20:30: 10:40 mol% were generated according to Example 1. Cas 12a-based RNPs for targeting the BCL 11 A-binding site in the promoter region of HBGJ and HBG2 were generated by complexing a gRNA comprising the target- complementary sequence of SEQ ID NO: 87 ("HBG promoter_Cas12a", complementary to SEQ ID NO: 86) with AsCas 12a, as described in Example 2. Cas9-based RNPs for targeting the BCLl 1 A- binding site in the promoter region of HBGJ and HBG2 were generated by complexing a sgRNA comprising the target-complementary sequence of SEQ ID NO: 85 ("HBG promoter_ 1 ", complementary to SEQ ID NO: 84) with SpCas9, as described in Example 2. Control RNPs for targeting an irrelevant gene ( GFP) were also generated in the same manner. RNPs were encapsulated by the TCVs as described in Example 3. [0549] HEK293 cells were seeded at a density of 40,000 cells/mL on Day 0. Twenty-four hours after seeding (Day 1), the TCV-encapsulated RNPs targeting either the irrelevant gene or HBGJ and HBG2 promoter were added to culture media at a final RNP concentration of 100 nM. Two hours after treatment (Day 1 ), culture media was changed, and cells were incubated for 46 hours prior to harvest. On Day 2, cells were harvested, and genomic DNA was extracted. A DNA region flanking the target HBG promoter site of HBGJ was amplified using the forward and reverse primers of SEQ ID NOS: 81 and 82, respectively, and sent for Sanger sequencing. A DNA region flanking the target HBG promoter site of HBG2 was amplified using the forward and reverse primers of SEQ ID NOS: 91 and 92, respectively, and sent for Sanger sequencing. Percent editing efficiency at HBGJ and HBG2 promoters was assessed using the TIDE analytical tool. [0550] Exemplary results are shown in FIG. 3E (top). As shown in the graph, successful editing was observed at the respective target sites with both Cas12a-based and Cas9-based RNPs and the Cas12a- based RNP provided higher editing efficiency. (0551] Additionally, the same study may be performed using a sgRNA comprising the target- complementary sequence of SEQ ID NO: 281, 283, or 285 with Cas9 and a sgRNA comprising the target-complementary sequence of SEQ ID NO: 287 or 289 with Cas12a. [0552] 14-3: Comparing dif erent gRNAs with Cas12a (0553] TCVs comprising DODMA:DOPE: DSCP:cholesterol = 20:30: 10:40 mo!% were generated according to Example 1. Cas12a-based RNPs for targeting the BCLI lA-binding site in the promoter region of HBGJ and HBG2 were generated by complexing a gRNA comprising the target- complementary sequence of SEQ ID NO: 87 ("HBG promoter_Cas12a", complementary to SEQ ID NO: 86), SEQ ID NO: 287 ("HBG promoter_Cas12a_2", complementary to SEQ ID NO: 286), or SEQ ID NO: 289 ("HBG promoter_Cas12a_3", complementary to SEQ ID NO: 288) with AsCas12a, as described in Example 2. Control RNPs for targeting a control gene (BCLJ JA, "-'ve control") were also generated in the same manner. RNPs were encapsulated by the TCVs as described in Example 3. (0554] HEK293 cells were seeded at a density of 40,000 cells/mL on Day 0. Twenty-four hours after seeding (Day 1), the TCV-encapsulated RNPs targeting either the irrelevant gene or HBGJ and HBG2 promoter were added to culture media at a final RNP concentration of 100 nM. Two hours after treatment (Day 1 ), culture media was changed, and cells were incubated for 46 hours prior to harvest. On Day 2, cells were harvested, and genomic DNA was extracted. A DNA region flanking the target HBG promoter site of HBGJ was amplified using the forward and reverse primers of SEQ ID NOS: 81 and 82, respectively, and sent for Sanger sequencing. A DNA region flanking the target HBG promoter site of HBG2 was amplified using the forward and reverse primers of SEQ ID NOS: 91 and 92, respectively, and sent for Sanger sequencing. Percent editing efficiency at HBGJ and HBG2 promoters was assessed using the TIDE analytical tool. (0555] Exemplary results are shown in FIG. 3E (bottom). As shown in the graph, successful editing was observed at the respective target sites with all of the tested Cas 12a-based RNPs in the promoter region of HBGJ and also with the gRNAs "HBG promoter_Cas12a" and "HBG promoter_Cas12a _3" in the promoter region of HBG2. With regard to editing with the gRNA "HBG promoter_Cas12a_2" in the promoter region of HBG2, without wishing to be bound by theory, it is possible that the cells used in this study had a mutation within the gRNA's target site (e.g., the target sequence and/or the PAM sequence) in the promoter region of HBG2. 14-4: Using ssODN [0556] TCVs comprising DODMA:DOPE: DSCP:cholesterol = 20:30:10:40 mol% were generated according to Example 1. Cas9-based RNPs for targeting the BCL 11 A-binding site in the promoter region of HBGJ and HBG2 were generated by complexing a sgRNA comprising the target- complementary sequence of SEQ ID NO: 85 ("HBG promoter_!", complementary to SEQ ID NO: 84) with SpCas9, as described in Example 2. Casl2a-based RNPs for targeting the BCLl IA-binding site in the promoter region of HBGJ and HBG2 were generated by complexing a gRNA comprising the target-complementary sequence of SEQ ID NO: 87 ("HBG promoter_Casl2a", complementary to SEQ ID NO: 86) with AsCasl2a, as described in Example 2. Control Casl2a-based and Cas9-based RNPs for targeting an irrelevant gene ( GFP) were also generated in the same manner. A ssODN comprising SEQ ID NO: 191 ("HBG_l3bp del_80pF", having a 5' homology rum corresponding to the 40-nt sequence immediately upstream of the 13-nt HPFH deletion sequence span and a 3' homology arm corresponding to the 40-nt sequence immediately downstream of the 13-nt HPFH deletion sequence span) and a ssODN comprising SEQ ID NO: 192 ("HBG_l3bp del_80pR", reverse complement of SEQ ID NO: 191) were prepared. RNPs were encapsulated by the TCVs as described in Example 3, and ssODNs were encapsulated by the TCVs as described in Example 3. (0557] HEK293 cells were seeded at a density of 40,000 cells/mL on Day 0. Twenty-four hours after seeding (Day 1), the TCV-encapsulated RNPs targeting either the irrelevant gene or the BCLl IA- binding site in the promoter region of HBGJ and HBG2 with or without the TCV-encapsulated ssODN were added to culture media at a final RNP concentration of 100 nM and a final ssODN concentration of 50 nM (when present). Two hours after treatment (Day 1), culture media was changed, and cells were incubated for 46 hours prior to harvest. On Day 2, cells were harvested, and genomic DNA was extracted. A DNA region flanking the target HBG promoter site of HBG 1 was amplified using the forward and reverse primers of SEQ ID NOS: 81 and 82, respectively, and sent for Sanger sequencing. A DNA region flanking the target HBG promoter site of HBG2 was amplified using the forward and reverse primers of SEQ ID NOS: 91 and 92, respectively, and sent for Sanger sequencing. Percent editing efficiency at HBGJ and HBG2 promoters was assessed using the TIDE analytical tool. (0558] Exemplary results with the Cas9-based and Casl2a-based RNPs are shown in FIG. 3G top and bottom, respectively. (0559] Additionally, the same study may be performed using a sgRNA comprising the target- complementary sequence of SEQ ID NO: 281, 283, or 285 with Cas9 and a sgRNA comprising the target-complementary sequence of SEQ ID NO: 287 or 289 with Casl2a. (0560] In some cases, a DNA template (e.g., a ssODN or dsODN), which is not complementary to the sequence of the target site, e.g., a site which encompasses the target sequence, may be used in addition or instead. In certain cases, the sequence of the target site may be, e.g., from about 40 nt to about 140, 130, 125, 123, or 120 nt, about 60-125 nt, about 70-125 nt, about 80-125 nt, about 90-125 nt, about 100-125 nt, about 110-125 nt, about 45 nt, about 55 nt, about 60 nt, about 70 nt, about 80 nt, about 90, about 100 nt, about 110 nt, or about 120 nt in length. For example, a ssODN ("HTT C6 80p_R") comprising SEQ ID NO: 190 which is not complementary to and thus is unlikely to form a dimer with the sequence encompassing and/or proximate to the target sequence of the HBG promoter region (see, e.g., FIG. 3H-3J, which shows below 50% sequence match between "HTT C680_R" and a 120-base sequence of the HBGJ and HBG2 promoter region around the target cut site(s), even when many gaps (48 gaps are shown) (shown as"" (space, no mark) are allowed). [0561] Example 15: Dose-dependent gene editing in the BCLllA-binding site in the promoter region of HBGJ and HBG2 in human cells. (0562] In this Example, the effect of different doses of RNPs on editing of the BCLl 1 A-binding site in the promoter region of HBGI and HBG2 in human cells was tested. (0563] TCVs comprising DODMA:DOPE: DSCP:cholesterol = 20:30: 10:40 mo!% were generated according to Example 1. Cas9-based RNPs for targeting the BCLl 1 A-binding site in the promoter region of HBGI and HBG2 were generated by complexing a sgRNA comprising the target- complementary sequence of SEQ ID NO: 85 with spCas9, as described in Example 2. Control Cas9- based RNPs for targeting luciferase were generated in the same manner using a sgRNA comprising the target-complementary sequence of SEQ ID NO: 55. RNPs were encapsulated by the TCVs as described in Example 3. (0564] HEK293 cells were seeded at a density of 80,000 cells/mL on Day 0. Twenty-four hours after seeding (Day 1), the TCV-encapsulated Cas9-based RNP targeting either luciferase or HBGI and HBG2 promoter regions were added to culture media at a final RNP concentration of either 25, 50, 100 or 200 nM (same stoichiometry ofTCV: RNP as in Example 14-1 but added at increasing volumes). Two hours after treatment (Day 1), culture media was changed, and cells were incubated for 46 hours prior to harvest (Day 3). On Day 3, cells were harvested, and genomic DNA was extracted. DNA regions flanking the target HBG promoter site of HBGJ or of HBG2 were amplified and sequenced as described in Example 14. Percent editing efficiency at HBGJ and HBG2 promoters and editing events were assessed using the TIDE analytical tool. (0565] Exemplary editing efficiency results are shown in FIG. 3C. As shown in the graphs, dose- dependent editing was observed in both of the target sites (the promoter of HBGI (left) and the promoter of HBG2 (right)). A representative histogram from the TIDE analysis showing distribution of specific editing events following treatment with the TCV-encapsulated RNP targeting the promoter region of HBGI and HBG2 at 200 nM is shown in FIG. 3D. As shown in the graph, the most common editing event was a 13-nucleotide deletion, which was the same as the one known to be a naturally occurring mutation that leads to hereditary persistence of fetal hemoglobin (HPFH), that disrupts the BCLl IA-binding site within the HBG promoter. The 13-nucleotide deletion occurred at a frequency higher than would be expected by chance, as indicated by the p value of <0.001 calculated by TIDE. [0566] Example 16: Gene editing in HBB exon 1 in human cells. 16-1: Editing at a HBB exon 1 position corresponding to E6 V site [0567] In this Example, the ability to edit or disrupt HEB exon 1 in human cells was tested. HEB exon 1 is the exon which contains the E-to-V mutation in the disease-causing HbS and HbC variants. Editing of HEB exon 1 as in this Example may be used as part of the gene correction strategy visualized in FIG. 4A. [0568] TCVs comprising DODMA:DOPE: DSCP:cholesterol = 20:30: 10:40 mo!% were generated according to Example 1. Cas9-based RNPs for targeting HEB exon 1 were generated by complexing a sgRNA comprising the target-complementary sequence of SEQ ID NO: 45 (complementary to HEB exon 1 region A ("E6V IA") comprising SEQ ID NO: 44) or SEQ ID NO: 47 (complementary to HEB exon 1 region B ("E6V 1 B") comprising SEQ ID NO: 46) with spCas9, as described in Example 2. Control Cas9-based RNPs for targeting luciferase were generated in the same manner using a sgRNA comprising the target-complementary sequence of SEQ ID NO: 55. RNPs were encapsulated by the TCVs as described in Example 3. [0569] HEK293 cells were seeded at a density of 80,000 cells/mL on Day 0. Twenty-four hours after seeding (Day 1), TCV-encapsulated RNP targeting either luciferase, HEB E6V IA, or HEB E6V 1B were added to culture media at a final RNP concentration of 50 nM. Two hours after treatment (Day 1), culture media was changed, and cells were incubated for 22 hours prior to harvest (Day 2). On Day 2, cells were harvested, and genomic DNA was extracted. A 746 base-pair region of DNA flanking the HEB E6V IA and 1B target sites were amplified using the forward and reverse primers of SEQ ID NOS: 41 and 42, respectively, and sent for Sanger sequencing. Percent editing efficiency at the target sites was assessed using the TIDE analytical tool. [0570] Exemplary results are shown in FIG. 4B. As shown in the graph, successful editing was observed at the respective target sites, HEB E6V IA and HEB E6V lB. 1 [0571] In this Example, the ability to introduce the disease-causing GAG>GTG (E6V) mutation at HEB exon 1 using a ssODN template containing the mutation (i.e., via homology-directed repair (HDR)) in human cells harboring WT HEB was tested. This serves as a proof-of-concept that an intended mutation can be introduced at this site of HEB exon 1 as part of the gene correction strategy visualized in FIG. 4A. [0572] TCVs comprising DODMA:DOPE: DSCP:cholesterol = 20:30: 10:40 mo!% were generated according to Example 1. Cas9-based RNPs for targeting HEB exon 1 were generated by complexing a sgRNA comprising the target-complementary sequence of SEQ ID NO: 45 (complementary to HEB exon 1 region A ("E6V IA") comprising SEQ ID NO: 44) with spCas9, as described in Example 2. Control Cas9-based RNPs for targeting an irrelevant gene (a promoter region of HBG, using SEQ ID NO: 85) were also generated in the same manner. ssODNs comprising SEQ ID NO: 181 ("HBB_E6V _80bp F"), 182 ("HBB_E6V _80bp R"), 183 ("HBB_E6V _l00bp F"), 184 ("HBB_E6V _l00bp R"), and 185 ("HBB_E6V _PAM l_l00bp R", one G-to-T mutation relative to SEQ ID NO: 184) were also prepared. RNPs were encapsulated by the TCVs as described in Example 3, and ssODNs were encapsulated by the TCVs as described in Example 3. (0573] HEK293 cells were seeded at a density of 40,000 cells/mL on Day 0. Twenty-four hours after seeding (Day 1), the TCV-encapsulated Cas9-based RNPs targeting either the irrelevant gene or HEB exon 1 with or without one of the TCV-encapsulated ssODNs were added to culture media at a final RNP concentration of 100 nM and a final ssODN concentration of 50 nM (when present). Two hours after treatment (Day 1), culture media was changed, and cells were incubated for 70 hours prior to harvest. On Day 2, cells were harvested, and genomic DNA was extracted. A 746 base-pair region of DNA flanking the HEB E6V lA and 1B target sites were amplified using the forward and reverse primers of SEQ ID NOS: 41 and 42, respectively, and sent for Sanger sequencing. Percent editing efficiency and percent HDR efficiency at the target sites was assessed using the TIDE and TIDER analytical tool, respectively. (0574] Exemplary results with the RNPs and different ssODNs are shown in FIG. 4D. [0575] Example 17: Dose-dependent gene editing in HBB exon 1 in human cells. (0576] In this Example, the effect of different doses ofRNPs on editing of HEB exon 1 in human cells was tested. (0577] TCVs comprising DODMA:DOPE: DSCP:cholesterol = 20:30: 10:40 mol% were generated according to Example 1. Cas9-based RNPs for targeting HEB exon 1 were generated by complexing a sgRNA comprising the target-complementary sequence of SEQ ID NO: 45 (complementary to HEB E6V lA) with spCas9, as described in Example 2. Control Cas9-based RNPs for targeting luciferase were generated in the same manner using a sgRNA comprising the target-complementary sequence of SEQ ID NO: 55. RNPs were encapsulated by the TCVs as described in Example 3. (0578] HEK293 cells were seeded at a density of 80,000 cells/mL on Day 0. Twenty-four hours after seeding (Day 1), TCV-encapsulated RNP targeting either luciferase or HEB exon 1 were added to culture media at a final RNP concentration of either 25, 50, 100 or 200 nM (same stoichiometry of TCV: RNP as in Example 16 but added at increasing volumes). Two hours after treatment (Day 1), culture media was changed, and cells were incubated for 46 hours prior to harvest (Day 3). On Day 3, cells were harvested, and genomic DNA was extracted. A 746 base-pair region of DNA flanking the HEB E6V 1 A target site was amplified and sequenced as described in Example 16. Percent editing efficiency at the target site was assessed using the TIDE analytical tool. (0579] Exemplary results are shown in FIG. 4C. As shown in the graph, dose-dependent editing was observed at the target site (HEB E6V lA). [0580) Example 18: Use of TCVs comprising gene editing constituents for effecting a gene correction in HBB exon 1 in human cells. [0581] HBB exon 1 is the exon which contains the E-to-V mutation in the disease-causing HbS and HbC variants. To confirm the ability of providing gene correction at the nucleic acid position corresponding to the E-to-V mutation, in this Example, a E-to-V mutation will be introduced into human cells possessing WT HBB. [0582] TCVs comprising DODMA:DOPE: DSCP:cholesterol = 20:30: 10:40 mol% are generated according to Example 1. RNPs for targeting HBB exon 1 are generated by complexing a sgRNA comprising the target-complementary sequence of SEQ ID NO: 45, 47, or 49 with spCas9, as described in Example 2. Control RNPs for targeting luciferase may be generated in the same manner using a sgRNA comprising the target-complementary sequence of SEQ ID NO: 55. RNPs are encapsulated by the TCVs as described in Example 3. A ssDNA such as one comprising any of SEQ ID NOS: 181-184 or a sequence complementary thereto or a double stranded DNA template having such a ssODN sequence will be encapsulated in TCVs in a similar manner to Example 3 (such a template may be encapsulated in TCVs separately from RNPs or together with RNPs). [0583] HEK293 cells are seeded ( e.g., at a density of 80,000 cells/mL) on Day 0. For example, twenty-four hours after seeding (Day 1 ), the RNP-DNA template-TCV complex ( or a combination of the RNA-TCV complex and the DNA template- TCV complex) is added to culture media (e.g., at a final RNP concentration of 50 nM). For example, two hours after treatment (Day 1 ), culture media is changed and cells are incubated (e.g., for 22 hours prior to harvest (Day 2)). For example, on Day 2, cells are harvested, and genomic DNA is extracted. For example, a 746 base-pair region of DNA flanking the HBB target sites is amplified using the forward and reverse primers of SEQ ID NOS: 41 and 42, respectively, and sent for sequencing to confirm gene correction as intended. Percent editing efficiency at the target sites is assessed using the TIDE analytical tool. [0584) Example 19: SCD-associated gene editing via intramarrow injection in reporter mice [0585] That intramarrow injection of the pharmaceutical compositions according to the present disclosure allows the TCV to carry it's CRISPR/Cas-effecting cargo to facilitate gene editing into HSCs, HSPCs, and/or bone marrow cells and provide for effective gene editing ( optionally including correction) to the SCD-associated gene(s) can be further confirmed, e.g., using methods disclosed in the present example. In particular this can be confirmed using the same reporter mouse as in Example 4. [0586] In such experiments the same reporter gene targeting RNP as in Example 4 and a BCLl IA- targeting RNP (e.g., targeting the mouse ortholog ofBCLl lA) will be prepared and both RNPs will be encapsulated in TCVs as described in Example 3. For example, a composition comprising the RNP-TCV or RNP-DNA template-TCV complex ( comprising about 2700 pmol RNPs per mL) will be injected into the bone marrow (e.g., of the femur) of the reporter mice at up to about 50 µI per minute for 5, 10, 20, 30, or 60 minutes. One, two, three, five, seven, 14, or 30 days later, the whole blood will be harvested, mice will be sacrificed, and the bone marrow cells will be harvested and washed. [0587] A portion of the blood cells and bone marrow cells will be stained with an anti-CD34 antibody to isolate CD34+ cells by sorting. Reporter gene-positive cells may be further sorted. Successful gene editing is confirmed based on the absence of the original gene (the mouse ortholog of human BCLJ JA), using PCR. Percentage of gene-edited cells among the total blood cells, total bone marrow cells, CD34+ cells in the blood, CD34+ cells in the bone marrow cells will be calculated to confirm successful gene editing in the cells. The expression ofHBG (transcript by qPCR and/or protein) in the cell will also be measured. Additionally, the amount ofHbF and/or the HbF/HbA ratio in the blood will also be measured by HPLC. [0588] Example 20: Gene editing in the BCLllA gene and the BCLllA-binding site in the promoter region of HBGJ and HBG2 in human erythroid progenitor cell lines. [0589] The ability to disrupt the EER in intron 2 of BCLJ J A and the BCLl lA-binding site in the promoter region ofHBGl and HBG2 in human erythroid progenitor cells (e.g., HUDEP-2 cells) according to the present invention can further be confirmed, e.g., using methods disclosed in the present example. The editing strategy is visualized in FIG. 2A. [0590] TCVs comprising DODMA:DOPE: DSCP:cholesterol = 20:30: 10:40 mo!% will be generated according to Example I. RNPs for targeting the EER of BCLJ J A will be generated by complexing a sgRNA comprising the target-complementary sequence of SEQ ID NO: 271 (complementary to SEQ ID NO: 270), SEQ ID NO: 273 (complementary to SEQ ID NO: 272), SEQ ID NO: 275 (complementary to SEQ ID NO: 274), SEQ ID NO: 65 (complementary to region 1 ofEER (EER I) comprising SEQ ID NO: 64), or SEQ ID NO: 69 (complementary to region 2 ofEER (EER 2) comprising SEQ ID NO: 68) with spCas9, or complexing a gRNA comprising the target- complementary sequence of SEQ ID NO: 277 (complementary to SEQ ID NO: 276) or SEQ ID NO: 279 (complementary to SEQ ID NO: 278) with AsCasl2a, as described in Example 2. RNPs for targeting the BCLl IA-binding site in the promoter region of HBGJ and HBG2 will be generated by complexing a sgRNA comprising the target-complementary sequence of SEQ ID NO: 85 (complementary to the promoter region comprising SEQ ID NO: 84, designed to hybridize to both the BCLl lA-binding promoter region of HBGJ and the BCLl lA-binding promoter region of HBG2) with spCas9, or complexing a gRNA comprising the target-complementary sequence of SEQ ID NO: 287 (complementary to SEQ ID NO: 286) or SEQ ID NO: 289 (complementary to SEQ ID NO: 288) with AsCasl2a, as described in Example 2. Control RNPs for targeting luciferase will be generated in the same manner using a sgRNA comprising the target-complementary sequence of SEQ ID NO: 55. RNPs will be encapsulated by the TCVs as described in Example 3. [0591] Human erythroid progenitor cells (e.g., HUDEP-2 cells) will be seeded on Day 0. On Day 1, the TCV-encapsulated RNPs will be added to culture media at a final RNP concentration of 50 nM. On Day 1, culture media will be changed, and cells will be incubated for about 48 hours prior to harvest. On Day 3, cells will be harvested, and genomic DNA will be extracted. A 788 base pair region of DNA flanking the two target sites of BCLl l A EER will be amplified using the forward and reverse primers of SEQ ID NOS: 61 and 62, respectively, and will be sent for Sanger sequencing. A DNA region flanking the target HBG promoter site of HBG 1 was amplified using the forward and reverse primers of SEQ ID NOS: 81 and 82, respectively. A DNA region flanking the target HBG promoter site ofHBG2 will be amplified using the forward and reverse primers of SEQ ID NOS: 91 and 92, respectively. Amplified DNAs will be sent for Sanger sequencing. Percent editing efficiency at each target site will be assessed using the TIDE analytical tool. The expression of BCLl l A and HBG (transcript by qPCR and/or protein) in the cell will be measured. Additionally, the amount of HbF and/or the HbF/HbA ratio in the blood will also be measured by HPLC. [0592] Example 21: Gene editing in HBB exon 1 in SCD patient-derived lymphoblastoid cells. [0593] The ability to edit or disrupt HEB exon l in human SCD patient-derived lymphoblastoid cells (e.g., GM16265 lymphoblastoid cells homozygous for the E6V mutation in HBB) according to the present invention can additionally be confirmed, e.g., using methods disclosed in the present example. The gene editing may be used as part of the gene correction strategy visualized in FIG. 4A. [0594] TCVs comprising DODMA:DOPE: DSCP:cholesterol = 20:30: 10:40 mo!% will be generated according to Example 1. RNPs for targeting HEB exon 1 will be generated by complexing a sgRNA comprising the target-complementary sequence of SEQ ID NO: 25 (complementary to HEB exon 1 region comprising SEQ ID NO: 24), the target-complementary sequence of SEQ ID NO: 45 (complementary to HEB exon 1 region A ("E6V lA") comprising SEQ ID NO: 44), SEQ ID NO: 47 ( complementary to HEB exon 1 region B ("E6V 1 B") comprising SEQ ID NO: 46), or SEQ ID NO: 49 (complementary to HEB exon 1 region B ("E6V lB") comprising SEQ ID NO: 48) with spCas9, as described in Example 2. Control RNPs for targeting luciferase will be generated in the same manner using a sgRNA comprising the target-complementary sequence of SEQ ID NO: 55. RNPs will be encapsulated by the TCV s as described in Example 3. [0595] Cells will be seeded on Day 0. On Day 1, TCV-encapsulated RNP targeting either luciferase or HEB will be added to culture media at a final RNP concentration of 50 nM. On Day 1, culture media was changed, and cells were incubated for about 48 hours prior to harvest (Day 2). On Day 2, cells will be harvested, and genomic DNA will be extracted. A DNA fragment flanking the target site (e.g., flanking the HEB E6V lA and 1B target sites) will be amplified using primers (e.g., the forward and reverse primers of SEQ ID NOS: 41 and 42, respectively) and sent for Sanger sequencing. Percent editing efficiency at the target sites will be assessed using the TIDE analytical tool. [0596] Example 22: Gene correction in HBB exon 1 in SCD patient-derived lymphoblastoid cells. [0597] The ability to correct the E6V mutation in HEB exon l in human SCD patient-derived lymphoblastoid cells (e.g., GM16265 lymphoblastoid cells homozygous for the E6V mutation in HBB) according to the invention can also be confirmed, e.g., using methods disclosed in the present example. The gene correction strategy is visualized in FIG. 4A. [0598] TCVs comprising DODMA:DOPE: DSCP:cholesterol = 20:30: l 0:40 mo!% are generated according to Example l. RNPs for targeting HEB exon l will be generated by complexing a sgRNA comprising the target-complementary sequence of SEQ ID NO: 25 (complementary to HEB exon l region comprising SEQ ID NO: 24), the target-complementary sequence of SEQ ID NO: 45 (complementary to HEB exon l region A ("E6V lA") comprising SEQ ID NO: 44), SEQ ID NO: 47 (complementary to HEB exon 1 region B ("E6V 1B") comprising SEQ ID NO: 46), or SEQ ID NO: 49 (complementary to HEB exon 1 region B ("E6V 1B") comprising SEQ ID NO: 48) with spCas9, as described in Example 2. Control RNPs for targeting luciferase may be generated in the same manner using a sgRNA comprising the target-complementary sequence of SEQ ID NO: 55. A ssDNA for correcting the E6V mutation back to wild-type HEB-encoding sequence such as one comprising any of SEQ ID NOS: 101-108 and 169-176 or a sequence complementary thereto or a double stranded DNA template having such a ssODN sequence will be encapsulated in TCVs in a similar manner to Example 3 (such a template may be encapsulated in TCVs separately from RNPs or together with RNPs). [0599] Cells will be seeded (e.g., at a density of80,000 cells/mL) on Day 0. On Day 1, the RNP- DNA template-TCV complex (or a combination of the RNA-TCV complex and the DNA template- TCV complex) will be added to culture media ( e.g., at a final RNP concentration of 50 nM). For example, two hours after treatment (Day 1), culture media is changed and cells will be incubated (e.g., for 48 hours prior to harvest (Day 3)). For example, on Day 3, cells will be harvested, and genomic DNA will be extracted. A DNA fragment flanking the target site (e.g., flanking the HEB E6V lA and 1B target sites) will be amplified using primers (e.g., the forward and reverse primers of SEQ ID NOS: 41 and 42, respectively) and sent for Sanger sequencing. Percent editing efficiency at the target sites (e.g., temp lated mutations as well as non-templated indels around the cut site) will be assessed using the TIDE analytical tool.

APPENDIX: AMINO ACID AND NUCLEIC ACID SEQUENCES HBB wild type: SEQ IDNO:1 Proteinsequence Humanhemoglobinsubunitbeta("beta-globin", "~-globin",or "HBB"),wild-type MVHLTPEEKSAVTALWGKVNVDEVGGEALGRLLVVYPWTQRFFESFGDLSTPDAVMGNPK VKAHGKKVLGAFS DGLAHLDNLKGTFATLSELHCDKLHVDPENFRLLGNVLVCVLAHHFGKEFTPPVQAAYQK VVAGVANALAHKYH SEQ ID NO: 11 Nucleicacidsequence Humanhemoglobinsubunitbeta,wild-type(lsequencelcorrespondsto aPAM sequence),encoded onChromosome11(llp15.4;AssemblyGRCh38.p13)from positions5225464to5227071of Chromosome11(1608ntincludingtheUTRs) ACATTTGCTTCTGACACAACTGTGTTCACTAGCAACCTCAAACAGACACCATGGTGCATC TGAC@ G~ GAAGTCTG~ ACTGCCCTGTGGGGCAAGGTGAACGTGGATGAAGTTGGTGGTGAGGCCCTGGGCAGGT TGGTATCAAGGTTACAAGACAGGTTTAAGGAGACCAATAGAAACTGGGCATGTGGAGACA GAGAAGACTCTT GGGTTTCTGATAGGCACTGACTCTCTCTGCCTATTGGTCTATTTTCCCACCCTTAGGCTG CTGGTGGTCTACCCT TGGACCCAGAGGTTCTTTGAGTCCTTTGGGGATCTGTCCACTCCTGATGCTGTTATGGGC AACCCTAAGGTGA AGGCTCATGGCAAGAAAGTGCTCGGTGCCTTTAGTGATGGCCTGGCTCACCTGGACAACC TCAAGGGCACCTT TGCCACACTGAGTGAGCTGCACTGTGACAAGCTGCACGTGGATCCTGAGAACTTCAGGGT GAGTCTATGGGA CGCTTGATGTTTTCTTTCCCCTTCTTTTCTATGGTTAAGTTCATGTCATAGGAAGGGGAT AAGTAACAGGGTAC AGTTTAGAATGGGAAACAGACGAATGATTGCATCAGTGTGGAAGTCTCAGGATCGTTTTA GTTTCTTTTATTTG CTGTTCATAACAATTGTTTTCTTTTGTTTAATTCTTGCTTICII 111111 ICTTCTCCGCAAm ACTATTATACT TAATGCCTTAACATTGTGTATAACAAAAGGAAATATCTCTGAGATACATTAAGTAACTTA AAAAAAAACTTTAC ACAGTCTGCCTAGTACATTACTATTTGGAATATATGTGTGCTTATTTGCATATTCATAAT CTCCCTACTTTATTTT CTTTTAm AATTGATACATAATCATTATACATATTTATGGGTTAAAGTGTAATGTTTTAATATGTGTA CACAT ATTGACCAAATCAGGGTAATTTTGCATTTGTAATTTTAAAAAATGCTTTCTTCTTTTAAT ATACTTTTTTGTTTAT CTTATTTCTAATACTTTCCCTAATCTCTTTCTTTCAGGGCAATAATGATACAATGTATCA TGCCTCTTTGCACCAT TCTAAAGAATAACAGTGATAATTTCTGGGTTAAGGCAATAGCAATATCTCTGCATATAAA TATTTCTGCATATA AATTGTAACTGATGTAAGAGGTTTCATATTGCTAATAGCAGCTACAATCCAGCTACCATT CTGCTTTTATTTTAT GGTTGGGATAAGGCTGGATTATTCTGAGTCCAAGCTAGGCCCTTTTGCTAATCATGTTCA TACCTCTTATCTTCC TCCCACAGCTCCTGGGCAACGTGCTGGTCTGTGTGCTGGCCCATCACTTTGGCAAAGAAT TCACCCCACCAGT GCAGGCTGCCTATCAGAAAGTGGTGGCTGGTGTGGCTAATGCCCTGGCCCACAAGTATCA CTAAGCTCGCTTT CTTGCTGTCCAATTTCTATTAAAGGTTCCTTTGTTCCCTAAGTCCAACTACTAAACTGGG GGATATTATGAAGG GCCTTGAGCATCTGGATTCTGCCTAATAAAAAACATTTATTTTCATTGCAA SEQ ID NO:12 Nucleicacidsequence HumanhemoglobinsubunitbetaORF,wild-type(isequencelcorrespond stoaPAM sequence),from positions5225601to5227021ofChromosome11;containingpositions5 226929-5227021(Exon1 5226798 5225726 GGTGGTGAGGCCCTGGGCAGGTTGGTATCAAGGTTACAAGACAGGTTTAAGGAGACCAAT AGAAACTGGGC ATGTGGAGACAGAGAAGACTCTTGGGTTTCTGATAGGCACTGACTCTCTCTGCCTATTGG TCTATTTTCCCACC CTTAGGCTGCTGGTGGTCTACCCTTGGACCCAGAGGTTCTTTGAGTCCTTTGGGGATCTG TCCACTCCTGATGC TGTTATGGGCAACCCTAAGGTGAAGGCTCATGGCAAGAAAGTGCTCGGTGCCTTTAGTGA TGGCCTGGCTCA CCTGGACAACCTCAAGGGCACCTTTGCCACACTGAGTGAGCTGCACTGTGACAAGCTGCA CGTGGATCCTGAG AACTTCAGGGTGAGTCTATGGGACGCTTGATGTTTTCTTTCCCCTTCTTTTCTATGGTTA AGTTCATGTCATAGG AAGGGGATAAGTAACAGGGTACAGTTTAGAATGGGAAACAGACGAATGATTGCATCAGTG TGGAAGTCTCA GGATCGTTTTAGTTTCTTTTATTTGCTGTTCATAACAATTGTTTTCTTTTGTTTAATTCT TGCTTICII 111111 ICT TCTCCGCAATTTTTACTATTATACTTAATGCCTTAACATTGTGTATAACAAAAGGAAATA TCTCTGAGATACATT AAGTAACTTAAAAAAAAACTTTACACAGTCTGCCTAGTACATTACTATTTGGAATATATG TGTGCTTATTTGCAT ATTCATAATCTCCCTACTTTATTTTCTTTTATTTTTAATTGATACATAATCATTATACAT ATTTATGGGTTAAAGTG TAATGTTTTAATATGTGTACACATATTGACCAAATCAGGGTAATTTTGCATTTGTAATTT TAAAAAATGCTTTCT ATGTATCATGCCTCTTTGCACCATTCTAAAGAATAACAGTGATAATTTCTGGGTTAAGGC AATAGCAATATCTC TGCATATAAATATTTCTGCATATAAATTGTAACTGATGTAAGAGGTTTCATATTGCTAAT AGCAGCTACAATCC AGCTACCATTCTGCTTTTATTTTATGGTTGGGATAAGGCTGGATTATTCTGAGTCCAAGC TAGGCCCTTTTGCTA ATCATGTTCATACCTCTTATCTTCCTCCCACAGCTCCTGGGCAACGTGCTGGTCTGTGTG CTGGCCCATCACTTT GGCAAAGAATTCACCCCACCAGTGCAGGCTGCCTATCAGAAAGTGGTGGCTGGTGTGGCT AATGCCCTGGCC CACAAGTATCAC SEQ ID NO: 13 Nucleic acid sequence Human hemoglobin subunit beta cDNA, wild-type (!sequence[ corresponds to a PAM sequence) ATGGTGCATCTGACT@G[AGG~GAAGTCTG~ ACTGCCCTGTGGGGCAAGGTGAACGTGGATGAAGTT GGTGGTGAGGCCCTGGGCAGGCTGCTGGTGGTCTACCCTTGGACCCAGAGGTTCTTTGAG TCCTTTGGGGAT CTGTCCACTCCTGATGCTGTTATGGGCAACCCTAAGGTGAAGGCTCATGGCAAGAAAGTG CTCGGTGCCTTTA GTGATGGCCTGGCTCACCTGGACAACCTCAAGGGCACCTTTGCCACACTGAGTGAGCTGC ACTGTGACAAGCT GCACGTGGATCCTGAGAACTTCAGGCTCCTGGGCAACGTGCTGGTCTGTGTGCTGGCCCA TCACTTTGGCAAA GAATTCACCCCACCAGTGCAGGCTGCCTATCAGAAAGTGGTGGCTGGTGTGGCTAATGCC CTGGCCCACAAGT ATCACTAA

HBB HbS variant: SEQ IDNO:2 Proteinsequence Humanhemoglobinsubunitbeta,SickleHb(HbS)variant MVHLTPVEKSAVTALWGKVNVDEVGGEALGRLLVVYPWTQRFFESFGDLSTPDAVMGNPK VKAHGKKVLGAFS DGLAHLDNLKGTFATLSELHCDKLHVDPENFRLLGNVLVCVLAHHFGKEFTPPVQAAYQK VVAGVANALAHKYH GGGTTTCTGATAGGCACTGACTCTCTCTGCCTATTGGTCTATTTTCCCACCCTTAGGCTG CTGGTGGTCTACCCT TGGACCCAGAGGTTCTTTGAGTCCTTTGGGGATCTGTCCACTCCTGATGCTGTTATGGGC AACCCTAAGGTGA AGGCTCATGGCAAGAAAGTGCTCGGTGCCTTTAGTGATGGCCTGGCTCACCTGGACAACC TCAAGGGCACCTT TGCCACACTGAGTGAGCTGCACTGTGACAAGCTGCACGTGGATCCTGAGAACTTCAGGGT GAGTCTATGGGA CGCTTGATGTTTTCTTTCCCCTTCTTTTCTATGGTTAAGTTCATGTCATAGGAAGGGGAT AAGTAACAGGGTAC AGTTTAGAATGGGAAACAGACGAATGATTGCATCAGTGTGGAAGTCTCAGGATCGTTTTA GTTTCTTTTATTTG CTGTTCATAACAATTGTTTTCTTTTGTTTAATTCTTGCTTICIIIII Ill ICTTCTCCGCAATTTTTACTATTATACT TAATGCCTTAACATTGTGTATAACAAAAGGAAATATCTCTGAGATACATTAAGTAACTTA AAAAAAAACTTTAC ACAGTCTGCCTAGTACATTACTATTTGGAATATATGTGTGCTTATTTGCATATTCATAAT CTCCCTACTTTATTTT CTTTTATTTTTAATTGATACATAATCATTATACATATTTATGGGTTAAAGTGTAATGTTT TAATATGTGTACACAT ATTGACCAAATCAGGGTAATTTTGCATTTGTAATTTTAAAAAATGCTTTCTTCTTTTAAT ATACTTTTTTGTTTAT CTTATTTCTAATACTTTCCCTAATCTCTTTCTTTCAGGGCAATAATGATACAATGTATCA TGCCTCTTTGCACCAT TCTAAAGAATAACAGTGATAATTTCTGGGTTAAGGCAATAGCAATATCTCTGCATATAAA TATTTCTGCATATA AATTGTAACTGATGTAAGAGGTTTCATATTGCTAATAGCAGCTACAATCCAGCTACCATT CTGCTTTTATTTTAT GGTTGGGATAAGGCTGGATTATTCTGAGTCCAAGCTAGGCCCTTTTGCTAATCATGTTCA TACCTCTTATCTTCC TCCCACAGCTCCTGGGCAACGTGCTGGTCTGTGTGCTGGCCCATCACTTTGGCAAAGAAT TCACCCCACCAGT GCAGGCTGCCTATCAGAAAGTGGTGGCTGGTGTGGCTAATGCCCTGGCCCACAAGTATCA CTAAGCTCGCTTT CTTGCTGTCCAATTTCTATTAAAGGTTCCTTTGTTCCCTAAGTCCAACTACTAAACTGGG GGATATTATGAAGG GCCTTGAGCATCTGGATTCTGCCTAATAAAAAACATTTATTTTCATTGCAA GGTGGTGAGGCCCTGGGCAGGTTGGTATCAAGGTTACAAGACAGGTTTAAGGAGACCAAT AGAAACTGGGC ATGTGGAGACAGAGAAGACTCTTGGGTTTCTGATAGGCACTGACTCTCTCTGCCTATTGG TCTATTTTCCCACC CTTAGGCTGCTGGTGGTCTACCCTTGGACCCAGAGGTTCTTTGAGTCCTTTGGGGATCTG TCCACTCCTGATGC TGTTATGGGCAACCCTAAGGTGAAGGCTCATGGCAAGAAAGTGCTCGGTGCCTTTAGTGA TGGCCTGGCTCA CCTGGACAACCTCAAGGGCACCTTTGCCACACTGAGTGAGCTGCACTGTGACAAGCTGCA CGTGGATCCTGAG AACTTCAGGGTGAGTCTATGGGACGCTTGATGTTTTCTTTCCCCTTCTTTTCTATGGTTA AGTTCATGTCATAGG AAGGGGATAAGTAACAGGGTACAGTTTAGAATGGGAAACAGACGAATGATTGCATCAGTG TGGAAGTCTCA GGATCGTTTTAGTTTCTTTTATTTGCTGTTCATAACAATTGTTTTCTTTTGTTTAATTCT TGcm c11111IIIICT TCTCCGCAATTTTTACTATTATACTTAATGCCTTAACATTGTGTATAACAAAAGGAAATA TCTCTGAGATACATT AAGTAACTTAAAAAAAAACTTTACACAGTCTGCCTAGTACATTACTATTTGGAATATATG TGTGCTTATTTGCAT ATTCATAATCTCCCTACTTTATTTTCTTTTATTTTTAATTGATACATAATCATTATACAT ATTTATGGGTTAAAGTG TAATGTTTTAATATGTGTACACATATTGACCAAATCAGGGTAATTTTGCATTTGTAATTT TAAAAAATGCTTTCT ATGTATCATGCCTCTTTGCACCATTCTAAAGAATAACAGTGATAATTTCTGGGTTAAGGC AATAGCAATATCTC TGCATATAAATATTTCTGCATATAAATTGTAACTGATGTAAGAGGTTTCATATTGCTAAT AGCAGCTACAATCC AGCTACCATTCTGCTTTTATTTTATGGTTGGGATAAGGCTGGATTATTCTGAGTCCAAGC TAGGCCCTTTTGCTA ATCATGTTCATACCTCTTATCTTCCTCCCACAGCTCCTGGGCAACGTGCTGGTCTGTGTG CTGGCCCATCACTTT GGCAAAGAATTCACCCCACCAGTGCAGGCTGCCTATCAGAAAGTGGTGGCTGGTGTGGCT AATGCCCTGGCC CACAAGTATCAC SEQ IDNO:23 Nucleicacidsequence HumanhemoglobinsubunitbetacDNA,SickleHbvariant(alsoreferredt oas "~S"allele)(lsequencel correspondstoaPAM sequence) ATGGTGCATCTGAc:@G~ GAAGTCTG~ ACTGCCCTGTGGGGCAAGGTGAACGTGGATGAAGTT GGTGGTGAGGCCCTGGGCAGGCTGCTGGTGGTCTACCCTTGGACCCAGAGGTTCTTTGAG TCCTTTGGGGAT CTGTCCACTCCTGATGCTGTTATGGGCAACCCTAAGGTGAAGGCTCATGGCAAGAAAGTG CTCGGTGCCTTTA GTGATGGCCTGGCTCACCTGGACAACCTCAAGGGCACCTTTGCCACACTGAGTGAGCTGC ACTGTGACAAGCT GCACGTGGATCCTGAGAACTTCAGGCTCCTGGGCAACGTGCTGGTCTGTGTGCTGGCCCA TCACTTTGGCAAA GAATTCACCCCACCAGTGCAGGCTGCCTATCAGAAAGTGGTGGCTGGTGTGGCTAATGCC CTGGCCCACAAGT ATCACTAA SEQ ID NO:24 Nucleicacidsequence HumanHBBHbSvariantCRISPR/Castargetsequence 1 GTGGAGAAGTCTGCCGTTAC SEQ ID NO:25 Nucleicacidsequence gRNAtarget-complementarysequence,complementarytoSEQ IDNO:24 GTAACGGCAGACTTCTCCAC

HBB HbC variant: SEQ IDNO:3 Proteinsequence Humanhemoglobinsubunitbeta,HbC(HbC)variant MVHLTPKEKSAVTALWGKVNVDEVGGEALGRLLVVYPWTQRFFESFGDLSTPDAVMGNPK VKAHGKKVLGAFS DGLAHLDNLKGTFATLSELHCDKLHVDPENFRLLGNVLVCVLAHHFGKEFTPPVQAAYQK VVAGVANALAHKYH SEQ ID NO: 31 Nucleicacidsequence Humanhemoglobinsubunitbeta,HbCvariant(alsoreferredtoas "~C"allele)(lsequencel correspondstoaPAM sequence),encodedonChromosome11(11p15.4;AssemblyGRCh38.p13) from positions5225464to5227071ofChromosome11(1608ntincludingtheUT Rs) ACATTTGCTTCTGACACAACTGTGTTCACTAGCAACCTCAAACAGACACCATGGTGCATC TGAC@ AAGGAG AAGTCTG~ ACTGCCCTGTGGGGCAAGGTGAACGTGGATGAAGTTGGTGGTGAGGCCCTGGGCAGGTT GGTATCAAGGTTACAAGACAGGTTTAAGGAGACCAATAGAAACTGGGCATGTGGAGACAG AGAAGACTCTT GGGTTTCTGATAGGCACTGACTCTCTCTGCCTATTGGTCTATTTTCCCACCCTTAGGCTG CTGGTGGTCTACCCT TGGACCCAGAGGTTCTTTGAGTCCTTTGGGGATCTGTCCACTCCTGATGCTGTTATGGGC AACCCTAAGGTGA AGGCTCATGGCAAGAAAGTGCTCGGTGCCTTTAGTGATGGCCTGGCTCACCTGGACAACC TCAAGGGCACCTT TGCCACACTGAGTGAGCTGCACTGTGACAAGCTGCACGTGGATCCTGAGAACTTCAGGGT GAGTCTATGGGA CGCTTGATGTTTTCTTTCCCCTTCTTTTCTATGGTTAAGTTCATGTCATAGGAAGGGGAT AAGTAACAGGGTAC AGTTTAGAATGGGAAACAGACGAATGATTGCATCAGTGTGGAAGTCTCAGGATCGTTTTA GTTTCTTTTATTTG CTGTTCATAACAATTGTTTTCTTTTGTTTAATTCTTGCTTICIII 11111 ICTTCTCCGCAAm ACTATTATACT TAATGCCTTAACATTGTGTATAACAAAAGGAAATATCTCTGAGATACATTAAGTAACTTA AAAAAAAACTTTAC ACAGTCTGCCTAGTACATTACTATTTGGAATATATGTGTGCTTATTTGCATATTCATAAT CTCCCTACTTTATTTT CTTTTAm AATTGATACATAATCATTATACATATTTATGGGTTAAAGTGTAATGTTTTAATATGTGTA CACAT ATTGACCAAATCAGGGTAATTTTGCATTTGTAATTTTAAAAAATGCTTTCTTCTTTTAAT ATACTTTTTTGTTTAT CTTATTTCTAATACTTTCCCTAATCTCTTTCTTTCAGGGCAATAATGATACAATGTATCA TGCCTCTTTGCACCAT TCTAAAGAATAACAGTGATAATTTCTGGGTTAAGGCAATAGCAATATCTCTGCATATAAA TATTTCTGCATATA AATTGTAACTGATGTAAGAGGTTTCATATTGCTAATAGCAGCTACAATCCAGCTACCATT CTGCTTTTATTTTAT GGTTGGGATAAGGCTGGATTATTCTGAGTCCAAGCTAGGCCCTTTTGCTAATCATGTTCA TACCTCTTATCTTCC TCCCACAGCTCCTGGGCAACGTGCTGGTCTGTGTGCTGGCCCATCACTTTGGCAAAGAAT TCACCCCACCAGT GCAGGCTGCCTATCAGAAAGTGGTGGCTGGTGTGGCTAATGCCCTGGCCCACAAGTATCA CTAAGCTCGCTTT CTTGCTGTCCAATTTCTATTAAAGGTTCCTTTGTTCCCTAAGTCCAACTACTAAACTGGG GGATATTATGAAGG GCCTTGAGCATCTGGATTCTGCCTAATAAAAAACATTTATTTTCATTGCAA SEQ IDNO:32 Nucleicacidsequence HumanhemoglobinsubunitbetaORF,HbCvariant(alsoreferredtoas "~C"allele)(lsequencel correspondstoaPAM sequence),from positions5225601to5227021ofChromosome11 ATGGTGCATCTGACT@AAGGAGAAGTCTG~ ACTGCCCTGTGGGGCAAGGTGAACGTGGATGAAGTT GGTGGTGAGGCCCTGGGCAGGTTGGTATCAAGGTTACAAGACAGGTTTAAGGAGACCAAT AGAAACTGGGC ATGTGGAGACAGAGAAGACTCTTGGGTTTCTGATAGGCACTGACTCTCTCTGCCTATTGG TCTATTTTCCCACC CTTAGGCTGCTGGTGGTCTACCCTTGGACCCAGAGGTTCTTTGAGTCCTTTGGGGATCTG TCCACTCCTGATGC TGTTATGGGCAACCCTAAGGTGAAGGCTCATGGCAAGAAAGTGCTCGGTGCCTTTAGTGA TGGCCTGGCTCA CCTGGACAACCTCAAGGGCACCTTTGCCACACTGAGTGAGCTGCACTGTGACAAGCTGCA CGTGGATCCTGAG AACTTCAGGGTGAGTCTATGGGACGCTTGATGTTTTCTTTCCCCTTCTTTTCTATGGTTA AGTTCATGTCATAGG AAGGGGATAAGTAACAGGGTACAGTTTAGAATGGGAAACAGACGAATGATTGCATCAGTG TGGAAGTCTCA GGATCGTTTTAGTTTCTTTTATTTGCTGTTCATAACAATTGTTTTCTTTTGTTTAATTCT TGCTTTCIIIII Ill ICT TCTCCGCAATTTTTACTATTATACTTAATGCCTTAACATTGTGTATAACAAAAGGAAATA TCTCTGAGATACATT AAGTAACTTAAAAAAAAACTTTACACAGTCTGCCTAGTACATTACTATTTGGAATATATG TGTGCTTATTTGCAT ATTCATAATCTCCCTACTTTATTTTCTTTTATTTTTAATTGATACATAATCATTATACAT ATTTATGGGTTAAAGTG TAATGTTTTAATATGTGTACACATATTGACCAAATCAGGGTAATTTTGCATTTGTAATTT TAAAAAATGCTTTCT TCTTTTAATATACTTTTTTGTTTATCTTATTTCTAATACTTTCCCTAATCTCTTTCTTTC AGGGCAATAATGATACA ATGTATCATGCCTCTTTGCACCATTCTAAAGAATAACAGTGATAATTTCTGGGTTAAGGC AATAGCAATATCTC TGCATATAAATATTTCTGCATATAAATTGTAACTGATGTAAGAGGTTTCATATTGCTAAT AGCAGCTACAATCC AGCTACCATTCTGCTTTTATTTTATGGTTGGGATAAGGCTGGATTATTCTGAGTCCAAGC TAGGCCCTTTTGCTA ATCATGTTCATACCTCTTATCTTCCTCCCACAGCTCCTGGGCAACGTGCTGGTCTGTGTG CTGGCCCATCACTTT GGCAAAGAATTCACCCCACCAGTGCAGGCTGCCTATCAGAAAGTGGTGGCTGGTGTGGCT AATGCCCTGGCC CACAAGTATCAC SEQ ID NO: 33 Nucleic acid sequence Human hemoglobin subunit beta cDNA, Sickle Hb C variant (also referred to as "~C" allele) (/sequence/ corresponds to a PAM sequence) ATGGTGCATCTGAc@AAGGAGAAGTCTG~ ACTGCCCTGTGGGGCAAGGTGAACGTGGATGAAGTT GGTGGTGAGGCCCTGGGCAGGCTGCTGGTGGTCTACCCTTGGACCCAGAGGTTCTTTGAG TCCTTTGGGGAT CTGTCCACTCCTGATGCTGTTATGGGCAACCCTAAGGTGAAGGCTCATGGCAAGAAAGTG CTCGGTGCCTTTA GTGATGGCCTGGCTCACCTGGACAACCTCAAGGGCACCTTTGCCACACTGAGTGAGCTGC ACTGTGACAAGCT GCACGTGGATCCTGAGAACTTCAGGCTCCTGGGCAACGTGCTGGTCTGTGTGCTGGCCCA TCACTTTGGCAAA GAATTCACCCCACCAGTGCAGGCTGCCTATCAGAAAGTGGTGGCTGGTGTGGCTAATGCC CTGGCCCACAAGT ATCACTAA

HBB exon 1 (any appropriate variant): SEQ ID NO: 41 Nucleic acid sequence Human HBB exon 1 forward primer TGCTTACCAAGCTGTGATTCC SEQ ID NO: 42 Nucleic acid sequence Human HBB exon 1 reverse primer CACTCAGTGTGGCAAAGGTG SEQ ID NO: 44 Nucleic acid sequence Human HBB exon 1 CRISPR/Cas target sequence 1 TTACTGCCCTGTGGGGCAAG SEQ ID NO: 45 Nucleic acid sequence gRNA target-complementary sequence, complementary to SEQ ID NO: 44 CTTGCCCCACAGGGCAGTAA SEQ ID NO: 46 Nucleic acid sequence Human HBB exon 1 CRISPR/Cas target sequence 2 TTACTGCCCTGTGGGGCA SEQ ID NO: 47 Nucleic acid sequence gRNA target-complementary sequence, complementary to SEQ ID NO: 46 TGCCCCACAGGGCAGTAA SEQ ID NO: 48 Nucleic acid sequence Human HBB exon 1 CRISPR/Cas target sequence 3 CAGGAGTCAGATGCACCATG SEQ ID NO: 49 Nucleic acid sequence gRNA target-complementary sequence, complementary to SEQ ID NO: 48 CATGGTGCATCTGACTCCTG

BCLllA: SEQ ID NO: 4 Nucleic acid sequence Human BCL11A EER (partial, coding strand at positions 60,495,352 to 60,495,172 of Chromosome 2 according to Gene Assembly GRCh38.p14) AGATATGGCATCTACTCTTAGACATAACACACCAGGGTCAATACAACTTTGAAGCTAGTC TAGTGCAAGCTAACAGT TGcr§~jTCA~CTCCAGGAAGGG~CCTCTGATTAGGGTGGGGGCGTGGGTGGGGTAGAA GAGGACT GGCAGACCTCTCCATCGGTGGCCGTTTGCC TCTGATTAGGGTGGGGGCGTGGGTGGGGTAGAAGAGGACTGGCAGAC SEQ ID NO: 5 Nucleic acid sequence Human BCLllA EER (partial, non-coding strand at positions 60,495,172 to 60,495,352 of Chromosome 2 according to Gene Assembly GRCh38.p14) GGCAAACGGCCACCGATGGAGAGGTCTGCCAGTCCTCTTCTACCCCACCCACGCCCCCAC CCTAATCAGAGGCCAA ACCCTTCCTGGAGCCTGTGATAAAAGCAACTGTTAGCTTGCACTAGACTAGCTTCAAAGT TGTATTGACCCTGGTGT GTTATGTCTAAGAGTAGATGCCATATCT SEQ ID NO: 15 Nucleic acid sequence Human BCLllA EER (partial, non-coding strand at positions 60,495,195 to 60,495,317 of Chromosome 2 according to Gene Assembly GRCh38.pl4, bases 24-146 of SEQ ID NO: 5 GTCTGCCAGTCCTCTTCTACCCCACCCACGCCCCCACCCTAATCAGAGGCCAAACCCTTC CTGGAGCCTGTGAT AAAAGCAACTGTTAGCTTGCACTAGACTAGCTTCAAAGTTGTATTGACC SEQ ID NO: 6 Protein sequence Human BAF chromatin remodeling complex subunit BCLllA ("BCL11A") MSRRKQGKPQHLSKREFSPEPLEAILTDDEPDHGPLGAPEGDHDLLTCGQCQMNFPLGDI LIFIEHKRKQCNGSLCL EKAVDKPPSPSPIEMKKASNPVEVGIQVTPEDDDCLSTSSRGICPKQEHIADKLLHWRGL SSPRSAHGALIPTPGMS AEYAPQGICKDEPSSYTCTTCKQPFTSAWFLLQHAQNTHGLRIYLESEHGSPLTPRVGIP SGLGAECPSQPPLHGIHI ADNNPFNLLRIPGSVSREASGLAEGRFPPTPPLFSPPPRHHLDPHRIERLGAEEMALATH HPSAFDRVLRLNPMAM EPPAMDFSRRLRELAGNTSSPPLSPGRPSPMQRLLQPFQPGSKPPFLATPPLPPLQSAPP PSQPPVKSKSCEFCGKT FKFQSNLVVHRRSHTGEKPYKCNLCDHACTQASKLKRHMKTHMHKSSPMTVKSDDGLSTA SSPEPGTSDLVGSAS SALKSVVAKFKSENDPNLIPENGDEEEEEDDEEEEEEEEEEEEELTESERVDYGFGLSLE AARHHENSSRGAVVGVGD ESRALPDVMQGMVLSSMQHFSEAFHQVLGEKHKRGHLAEAEGHRDTCDEDSVAGESDRID DGTVNGRGCSPGE SASGGLSKKLLLGSPSSLSPFSKRIKLEKEFDLPPAAMPNTENVYSQWLAGYAASRQLKD PFLSFGDSRQSPFASSSE HSSE NGSLRFSTPPG ELDGG ISG RSGTGSGGSTPH ISG PG PG RPSSKE G RRS DTCEYCG KVFKNCS NL TVH RRSHTG ERPYKCELCNYACAQSSKL TRH M KTHGQVGKDVYKCE ICKM PFSVYSTLEKH M KKWHSDRVLN N DI KTE SEQ ID NO: 61 Nucleic acid sequence Human BCLllA EER forward primer TCCAAACTCTCAAACCACAGG SEQ ID NO: 62 Nucleic acid sequence Human BCL11A EER reverse primer GGCAAGTCAGTTGGGAACAC SEQ ID NO: 64 Nucleic acid sequence Human BCL11A EER CRISPR/Cas target sequence 1 (BCLllA genomic antisense sequence) GTGATAAAAGCAACTGTTAG SEQ ID NO: 65 Nucleic acid sequence gRNA target-complementary sequence, complementary to SEQ ID NO: 64 CTAACAGTTGCTTTTATCAC SEQ ID NO: 66 Nucleic acid sequence Human BCLllA EER CRISPR/Cas target sequence 3 (BCLllA genomic antisense sequence) GGAGCCTGTGATAAAAGCAA SEQ ID NO: 67 Nucleic acid sequence gRNA target-complementary sequence, complementary to SEQ ID NO: 66 TTGCTTTTATCACAGGCTCC SEQ ID NO: 68 Nucleic acid sequence Human BCL11A EER CRISPR/Cas target sequence 2 (BCLllA genomic antisense sequence) AACCCTTCCTGGAGCCTGTG SEQ ID NO: 69 Nucleic acid sequence gRNA target-complementary sequence, complementary to SEQ ID NO: 68 CACAGGCTCCAGGAAGGGTT SEQ ID NO: 70 Nucleic acid sequence Human BCLllA EER CRISPR/Cas target sequence 4 (BCL11A genomic antisense sequence) AACCCTTCCTGGAGCCTGTGA SEQ ID NO: 71 Nucleic acid sequence gRNA target-complementary sequence, complementary to SEQ ID NO: 70 (may be used with Cas12a) TCACAGGCTCCAGGAAGGGTT SEQ ID NO: 270 Nucleic acid sequence Human BCL11A EER CRISPR/Cas target sequence 5 (BCLllA genomic antisense sequence) AATCAGAGGCCAAACCCTTC SEQ ID NO: 271 Nucleic acid sequence gRNA target-complementary sequence, complementary to SEQ ID NO: 270 GAAGGGTTTGGCCTCTGA TT SEQ ID NO: 272 Nucleic acid sequence Human BCLllA EER CRISPR/Cas target sequence 6 (BCLllA genomic sense sequence) GGAAGGGTTTGGCCTCTGAT SEQ ID NO: 273 Nucleic acid sequence gRNA target-complementary sequence, complementary to SEQ ID NO: 272 ATCAGAGGCCAAACCCTTCC SEQ ID NO: 274 Nucleic acid sequence Human BCLllA EER CRISPR/Cas target sequence 7 (BCLllA genomic antisense sequence) TAATCAGAGGCCAAACCCTT SEQ ID NO: 275 Nucleic acid sequence gRNA target-complementary sequence, complementary to SEQ ID NO: 274 AAGGGTTTGGCCTCTGA TT A SEQ ID NO: 276 Nucleic acid sequence Human BCLllA EER CRISPR/Cas target sequence 8 (BCLllA genomic antisense sequence) TAGCTTGCACTAGACTAGCTT SEQ ID NO: 277 Nucleic acid sequence gRNA target-complementary sequence, complementary to SEQ ID NO: 276 (may be used with Cas12a) AAGCTAGTCTAGTGCAAGCTA SEQ ID NO: 278 Nucleic acid sequence Human BCLllA EER CRISPR/Cas target sequence 9 (BCLllA genomic antisense sequence) GCCCCCACCCTAATCAGAGGC SEQ ID NO: 279 Nucleic acid sequence gRNA target-complementary sequence, complementary to SEQ ID NO: 278 (may be used with Cas12a) GCCTCTGATTAGGGTGGGGGC KLFl: SEQ ID NO: 7 Protein sequence Human Kruppel like factor 1 ("KLFl") MATAETALPSISTLTALGPFPDTQDDFLKWWRSEEAQDMGPGPPDPTEPPLHVKSEDQPG EEEDDERGADATW DLDLLL TN FSGPEPGGAPQTCALAPSEAPGAQYPPPPETLGA YAGG PGL VAG LLGSEDHSGWVRPALRARAPDAF VG P ALAP AP APE P KALALQPVYPG PGAGSSG GYFP RTG LSVP AASGAPYG LLSRYP AMYPAPQYQG H FQLF RG LQ GPAPGPATSPSFLSCLGPGTVGTGLGGTAEDPGVIAETAPSKRGRRSWARKRQAAHTCAH PGCGKSYTKSSHLKA H LRTHTG EKPYACTWEGCGWRFARSDEL TRHYRKHTGQRPFRCQLCPRAFSRSDH LAL HM KRHL SEQ ID NO: 74 Nucleic acid sequence Human KLFl CRISPR/Cas target sequence 1 (KLFl genomic antisense sequence) CTTGCGCGCCCACGAACGTC SEQ ID NO: 75 Nucleic acid sequence gRNA target-complementary sequence, complementary to SEQ ID NO: 74 GACGTTCGTGGGCGCGCAAG SEQ ID NO: 76 Nucleic acid sequence Human KLFl CRISPR/Cas target sequence 2 (KLFl genomic antisense sequence) AGCGCGCGAATCTCCAGCCG SEQ ID NO: 77 Nucleic acid sequence gRNA target-complementary sequence, complementary to SEQ ID NO: 76 CGGCTGGAGATTCGCGCGCT

HBGl and HBG2: SEQ ID NO: 8 Protein sequence Human hemoglobin subunit gamma 1 ("A-gamma-globin" or "HBGl") MGHFTEEDKATITSLWGKVNVEDAGGETLGRLLVVYPWTQRFFDSFGNLSSASAIMGNPK VKAHGKKVLTSLGDA I KHLDDLKGTFAQLSELHCDKLHVDPEN FKLLGNVL VTVLAIHFGKEFTPEVQASWQKMVTAVASALSSRYH SEQ ID NO: 9 Protein sequence Human hemoglobin subunit gamma 2 ("G-gamma-globin" or "HBG2") MGHFTEEDKATITSLWGKVNVEDAGGETLGRLLVVYPWTQRFFDSFGNLSSASAIMGNPK VKAHGKKVLTSLGDA IKHLDDLKGTFAQLSELHCDKLHVDPENFKLLGNVLVTVLAIHFGKEFTPEVQASWQKMV TGVASALSSRYH SEQ ID NO: 95 Nucleic acid sequence Human HBG1/HBG2 promoter region (partial, coding strand at positions 5,250,070 to 5,249,890 (same as positions 5,254,994 to 5,254814) of Chromosome 11 according to Gene Assembly GRCh38.p14) CTCTTGGGGGCCCCTTCCCCACACTATCTCAATGCAAATATCTGTCTGAAACGGTCCCTG GCTAAACTCCACCC ATGGGTTGGCCAGCCTTGC AATAGCCTTGACAAGGCAAACTTGACCAATAGTCTTAGAGTATCCAG TGAGGCCAGGGGCCGGCGGCTGGCTAGGGATGAA SEQ ID NO: 295 Nucleic acid sequence Human HBG1/HBG2 promoter region (partial, coding strand at positions 5,250,033 to 5,249,914 (same as positions 5,254,957 to 5,254,838} of Chromosome 11 according to Gene Assembly bases 38-157 of SEQ ID NO: CAAGGCAAACTTGACCAATAGTCTTAGAGTATCCAGTGAGGCCAGG SEQ ID NO: 97 Nucleic acid sequence Human HBG1/HBG2 promoter region (partial, non-coding strand at positions 5,249,890 to 5,250,070 (same as positions 5,254814 to 5,254,994) of Chromosome 11 according to Gene Assembly GCCAACCCATGGGTGGAGTTTAGCCAGGGACCGTTTCAGACAGATAT SEQ ID NO: 99 Nucleic acid sequence Human HBG1/HBG2 promoter region 13-nt HPFH .deletion jgAATAGCCTTGAC SEQ ID NO: 81 Nucleic acid sequence Human HBGl promoter region forward primer TCTCCCAAGGAAGTCAGCAC SEQ ID NO: 82 Nucleic acid sequence Human HBGl promoter region reverse primer CTTAGAAACCACTGCTAACTGAAAGAG SEQ ID NO: 91 Nucleic acid sequence Human HBG2 promoter region forward primer CAGAGGACAGGTTGCCAAAG SEQ ID NO: 92 Nucleic acid sequence Human HBG2 promoter region reverse primer CCAATGCTTACTAAATGAGACTAAGACG SEQ ID NO: 84 Nucleic acid sequence Human HBGl and HBG2 promoter region CRISPR/Cas target sequence 1 TGACCAATAGCCTTGACAAG SEQ ID NO: 85 Nucleic acid sequence gRNA target-complementary sequence, complementary to SEQ ID NO: 84 CTTGTCAAGGCTATTGGTCA SEQ ID NO: 86 Nucleic acid sequence Human HBGl and HBG2 promoter region CRISPR/Cas target sequence 2 TGACCAATAGCCTTGACAAGG SEQ ID NO: 87 Nucleic acid sequence gRNA target-complementary sequence, complementary to SEQ ID NO: 86 (may be used with Cas12a) CCTTGTCAAGGCTATTGGTCA SEQ ID NO: 280 Nucleic acid sequence Human HBGl and HBG2 promoter region CRISPR/Cas target sequence 3 TGTCAAGGCTATTGGTCAAG SEQ ID NO: 281 Nucleic acid sequence gRNA target-complementary sequence, complementary to SEQ ID NO: 280 CTTGACCAATAGCCTTGACA SEQ ID NO: 282 Nucleic acid sequence Human HBGl and HBG2 promoter region CRISPR/Cas target sequence 4 ACCCATGGGTGGAGTTTAGC SEQ ID NO: 283 Nucleic acid sequence gRNA target-complementary sequence, complementary to SEQ ID NO: 282 GCTAAACTCCACCCATGGGT SEQ ID NO: 284 Nucleic acid sequence Human HBGl and HBG2 promoter region CRISPR/Cas target sequence 5 ATAGTCTTAGAGTATCCAGT SEQ ID NO: 285 Nucleic acid sequence gRNA target-complementary sequence, complementary to SEQ ID NO: 284 ACTGGATACTCTAAGACTAT SEQ ID NO: 286 Nucleic acid sequence Human HBGl and HBG2 promoter region CRISPR/Cas target sequence 6 CTGTCTGAAACGGTCCCTGGC SEQ ID NO: 287 Nucleic acid sequence gRNA target-complementary sequence, complementary to SEQ ID NO: 286 (may be used with Cas12a) GCCAGGGACCGTTTCAGACAG SEQ ID NO: 288 Nucleic acid sequence Human HBGl and HBG2 promoter region CRISPR/Cas target sequence 7 CGGCTGGCTAGGGATGAAGAA SEQ ID NO: 289 Nucleic acid sequence gRNA target-complementary sequence, complementary to SEQ ID NO: 288 (may be used with Cas12a) TTCTTCATCCCTAGCCAGCCG

Luciferase (control): SEQ ID NO: 54 Nucleic acid sequence Luciferase CRISPR/Cas target sequence 1 ACCCAACGGACATTTCGAAG SEQ ID NO: 55 Nucleic acid sequence gRNA luciferase target-complementary sequence CTTCGAAATGTCCGTTGGGT Ai14 reporter mouse floxed-stop cassette (for TdTomato expression): SEQ ID NO: 56 Nucleic acid sequence PS2 gRNA target-complementary sequence gtctggatct gcgactctag SEQ ID NO: 57 Nucleic acid sequence PS3 gRNA target-complementary sequence tcaatgtatc ttatcatgtc SEQ ID NO: 58 Nucleic acid sequence La Ro gRNA target-complementary sequence aaagaattga tttgataccg SEQ ID NO: 59 Nucleic acid sequence LoxP gRNA target-complementary sequence gtatgctata cgaagttatt

ssODN for correcting to HBB wild type: SEQ ID NO: 101 Nucleic acid sequence Human beta-globin (WT)-encoding ssODN sequence 1 TCACCACCAACTTCATCCACGTTCACCTTGCCCCACAGGGCAGTAACGGCAGACTTCTCc t GTCAGATG CACCATGGTGTCTGTTTGAGGTTGCTAGTGAACACAGTTGTGTCAGA SEQ ID NO: 102 Nucleic acid sequence Human beta- - ssODN 2 CACCATGGTGTCTGTTTGAGGTTGCTAGTGAACACAGTTGTGTCAGA SEQ ID NO: 104 Nucleic acid sequence Human beta-globin (WT)-encoding ssODN sequence 4 TCACCACCAACTTCATCCACGTTCACCTTGCCCCACAGGGCAGTAACGGCAGACTTCTCt tcjTGG!AGTCAGATG CACCATGGTGTCTGTTTGAGGTTGCTAGTGAACACAGTTGTGTCAGA SEQ ID NO: 105 Nucleic acid sequence Human beta-globin (WT)-encoding ssODN sequence 5 TCACCACCAACTTCATCCACGTTCACCTTGCCCCACAGGGCAGTAACGGCAGACTTCTCc t GTCAGATG CACCATGGTGTCTGTTTGAGGTTGCTAGTGAACACAGTTGTGTCAGA SEQ ID NO: 106 Nucleic acid sequence Human beta-globin (WT)-encoding ssODN sequence 6 TCACCACCAACTTCATCCACGTTCACCTTGCCCCACAGGGCAGTAACGGCAGACTTCTCt t GTCAGATG CACCATGGTGTCTGTTTGAGGTTGCTAGTGAACACAGTTGTGTCAGA SEQ ID NO: 107 Nucleic acid sequence - CACCATGGTGTCTGTTTGAGGTTGCTAGTGAACACAGTTGTGTCAGA SEQ ID NO: 108 Nucleic acid sequence Human beta-globin (WT)-encoding ssODN sequence 8 TCACCACCAACTTCATCCACGTTCACCTTGCCCCACAGGGCAGTAACGGCAGACTTCTCt t GTCAGATG CACCATGGTGTCTGTTTGAGGTTGCTAGTGAACACAGTTGTGTCAGA SEQ ID NO: 169 Nucleic acid sequence Human beta-globin (WT)-encoding ssODN sequence 9 (80 bp Forward) AGCAACCTCAAACAGACACCATGGTGCATCTGACTCCTGAGGAGAAGTCTGCCGTTACTG CCCTGTGGGGCAA GGTGAAC SEQ ID NO: 170 Nucleic acid sequence Human beta-globin (WT)-encoding ssODN sequence 10 (80 bp Reverse) GTTCACCTTGCCCCACAGGGCAGTAACGGCAGACTTCTCCTCAGGAGTCAGATGCACCAT GGTGTCTGm GA GGTTGCT SEQ ID NO: 171 Nucleic acid sequence Human beta-globin (WT)-encoding ssODN sequence 11 (100 bp Forward) TGTGTTCACTAGCAACCTCAAACAGACACCATGGTGCATCTGACTCCTGAGGAGAAGTCT GCCGTTACTGCCCT GTGGGGCAAGGTGAACGTGGATGAAG SEQ ID NO: 172 Nucleic acid sequence Human beta-globin (WT)-encoding ssODN sequence 12 (100 bp Reverse) CTTCATCCACGTTCACCTTGCCCCACAGGGCAGTAACGGCAGACTTCTCCTCAGGAGTCA GATGCACCATGGT GTCTGm GAGGTTGCTAGTGAACACA SEQ ID NO: 173 Nucleic acid sequence Human beta-globin (WT)-encoding ssODN sequence 13 (120 bp Forward) CTGACACAACTGTGTTCACTAGCAACCTCAAACAGACACCATGGTGCATCTGACTCCTGA GGAGAAGTCTGCC GTTACTGCCCTGTGGGGCAAGGTGAACGTGGATGAAGTTGGTGGTGA SEQ ID NO: 174 Nucleic acid sequence Human beta-globin (WT)-encoding ssODN sequence 14 (120 bp Reverse) TCACCACCAACTTCATCCACGTTCACCTTGCCCCACAGGGCAGTAACGGCAGACTTCTCC TCAGGAGTCAGATG CACCATGGTGTCTGm GAGGTTGCTAGTGAACACAGTTGTGTCAG SEQ ID NO: 175 Nucleic acid sequence - SEQ ID NO: 176 Nucleic acid sequence Human beta-globin (WT)-encoding ssODN sequence 16 (with one missense; 111 bp Reverse) CTTCATCCACGTTCACCTTGCCCCACAGGGCAGTA~CCG!CAGACTTCTCCTCAGGAGTC AGATGCACCATGGT GTCTGm GAGGTTGCTAGTGAACACAGTTGTGTCAG ssODN for HBB, parts: SEQ ID NO: 111 Nucleic acid sequence 5' homology arm (60 nt) of SEQ ID NO: 101, 103, 105, and 107 TCACCACCAACTTCATCCACGTTCACCTTGCCCCACAGGGCAGTAACGGCAGACTTCTCc SEQ ID NO: 112 Nucleic acid sequence 5' homology arm (59 nt) of SEQ ID NO: 102, 104, 106, and 108 TCACCACCAACTTCATCCACGTTCACCTTGCCCCACAGGGCAGTAACGGCAGACTTCTC SEQ ID NO: 121 Nucleic acid sequence 3' homology arm (60 nt) of SEQ ID NO: 101, 102 GTCAGATGCACCATGGTGTCTGTTTGAGGTTGCTAGTGAACACAGTTGTGTCAGA SEQ ID NO: 122 Nucleic acid sequence 3' homology arm (58 nt) of SEQ ID NO: 103, 104, 105, 106, 107, and 108 ~ GTCAGATGCACCATGGTGTCTGTTTGAGGTTGCTAGTGAACACAGTTGTGTCAGA

ssODN for introducing HBB EGV substitution: SEQ ID NO: 181 Nucleic acid sequence Human beta-globin (E6V)-encoding ssODN sequence 1 (80 bp F) AGCAACCTCAAACAGACACCATGGTGCATCTGACTCCTGTGGAGAAGTCTGCCGTTACTG CCCTGTGGGGCAA GGTGAAC SEQ ID NO: 182 Nucleic acid sequence Human beta-globin (E6V)-encoding ssODN sequence 2 (80 bp R) GTTCACCTTGCCCCACAGGGCAGTAACGGCAGACTTCTCCACAGGAGTCAGATGCACCAT GGTGTCTGTTTGA GGTTGCT SEQ ID NO: 183 Nucleic acid sequence Human beta-globin (E6V)-encoding ssODN sequence 3 (100 bp F) TGTGTTCACTAGCAACCTCAAACAGACACCATGGTGCATCTGACTCCTGTGGAGAAGTCT GCCGTTACTGCCCT GTGGGGCAAGGTGAACGTGGATGAAG SEQ ID NO: 184 Nucleic acid sequence Human beta-globin (E6V)-encoding ssODN sequence 4 (100 bp R) CTTCATCCACGTTCACCTTGCCCCACAGGGCAGTAACGGCAGACTTCTCCACAGGAGTCA GATGCACCATGGT GTCTGTTTGAGGTTGCTAGTGAACACA SEQ ID NO: 185 Nucleic acid sequence Human beta-globin (E6V)-encoding ssODN sequence 5 (PAM 1_100 bp R) CTTCATCCACGTTCACCTTGCCCCACAGGGCAGTAACTGCAGACTTCTCCACAGGAGTCA GATGCACCATGGT GTCTGTTTGAGGTTGCTAGTGAACACA

ssODN for editing human BCL11A EER: SEQ ID NO: 301 Nucleic acid sequence Human BCL11A EER ssODN sequence 1(120bp F) TCAATACAACTTTGAAGCTAGTCTAGTGCAAGCTAACAGTTGCTTTTATCACAGGCTCCA GGAAGGGTTTGGC CTCTGATTAGGGTGGGGGCGTGGGTGGGGTAGAAGAGGACTGGCAGA S EQ ID NO: 302 Nucleic acid sequence H uman BCL11A EER ssODN sequence 2(120bp R) GGTGGGGGCGTGGGTGGGGTAGAAGAG S EQ ID NO: 304 Nucleic acid sequence H uman BCL11A EER ssODN sequence 4 (l00bp R) CTCTTCTACCCCACCCACGCCCCCACCCTAATCAGAGGCCAAACCCTTCCTGGAGCCTGT GATAAAAGCAACTG TTAGCTTGCACTAGACTAGCTTCAAA SEQ ID NO: 305 Nucleic acid sequence Human BCL11A EER ssODN sequence 5 (80bp F) GTCTAGTGCAAGCTAACAGTTGCTTTTATCACAGGCTCCAGGAAGGGTTTGGCCTCTGAT TAGGGTGGGGGC GTGGGTGG S EQ ID NO: 306 Nucleic acid sequence H uman BCL11A EER ssODN 6

Non-complement ssODN for enhancing editing: SEQ ID NO: 190 Nucleic acid sequence Non-complementary ssODN HTT C6 sequence 1 (80 bp R) CTGTCCAATCTGCAGGCCCAAATACTGGTTGTCGGTACCGGCTAACACCTAAACGGTTCA AGGGGGGCTGTGA GAATTTT ssODN for introducing HBG 13-nt HPFH deletion: SEQ ID NO: 191 Nucleic acid sequence Human HBG 13-nt HPFH-introducing ssODN sequence 1 (80 bp F) GGCTAAACTCCACCCATGGGTTGGCCAGCCTTGCCTTGACAAGGCAAACTTGACCAATAG TCTTAGAGTATCC AGTGAGG SEQ ID NO: 192 Nucleic acid sequence Human HBG 13-nt HPFH-introducing ssODN sequence 1 (80 bp R) ssODN for HBG1/HBG2 promoter for preventing BCLllA-binding, parts: SEQ ID NO: 195 Nucleic acid sequence Human HBG1/HBG2 promoter BCL11A-binding-blocking ssODN (coding) 5' homology arm template, based on which 5' homology arm may be designed CTCTTGGGGGCCCCTTCCCCACACTATCTCAATGCAAATATCTGTCTGAAACGGTCCCTG GCTAAACTCCACCC ATGGGTTGGCCAGCCTTGCCT SEQ ID NO: 196 Nucleic acid sequence Human HBG1/HBG2 promoter BCL11A-binding-blocking ssODN (coding) 3' homology arm template, based on which 5' homology arm may be designed AATAGCCTTGACAAGGCAAACTTGACCAATAGTCTTAGAGTATCCAGTGAGGCCAGGGGC CGGCGGCTGGCT AGGGATGAA SEQ ID NO: 197 Nucleic acid sequence Human HBG1/HBG2 promoter BCL11A-binding-blocking ssODN (non-coding) 5' homology arm template, based on which 5' homology arm may be designed TTCATCCCTAGCCAGCCGCCGGCCCCTGGCCTCACTGGATACTCTAAGACTATTGGTCAA GTTTGCCTTGTCAA GGCTATT SEQ ID NO: 198 Nucleic acid sequence Human HBG1/HBG2 promoter BCL11A-binding-blocking ssODN (non-coding) 3' homology arm template, based on which 5' homology arm may be designed AGGCAAGGCTGGCCAACCCATGGGTGGAGTTTAGCCAGGGACCGTTTCAGACAGATATTT GCATTGAGATAG TGTGGGGAAGGGGCCCCCAAGAG gRNA subparts (e.g., for Cas9): SEQ ID NO: 131 Nucleic acid sequence crRNA flagpole sequence (option 1) GUUUUAGAGCUA SEQ ID NO: 132 Nucleic acid sequence crRNA flagpole sequence (option 2) GUUUAAGAGCUA SEQID NO: NA Nucleic acid sequence crRNA first flagpole extension (optional) UGCUG SEQ ID NO: NA Nucleic acid sequence crRNA second flagpole extension (optional) UUUUG SEQ ID NO: NA Nucleic acid sequence (optional) tracrRNA first extension CAGCA SEQ ID NO: 136 Nucleic acid sequence tracrRNA flagpole (option 1) UAGCAAGUUAAAA SEQ ID NO: 137 Nucleic acid sequence tracrRNA flagpole (option 2) UAGCAAGUUUAAA SEQID NO: 138 Nucleic acid sequence tracrRNA nuclease binding domain (may be followed by multiple uracils) UAAGGCUAGUCCGUUAUCAACUUGAAAAAGUGGCACCGAGUCGGUGC SEQ ID NO: NA Nucleic acid sequence Linker (linker between crRNA sequence and tracrRNA sequence) GAAA SEQ ID NO: 141 Nucleic acid sequence sgRNA backbone (3' to targeting sequence) (option 1) (may be followed by multiple uracils) GUUUUAGAGCUAGAAAUAGCAAGUUAAAAUAAGGCUAGUCCGUUAUCAACUUGAAAAAGU GGCACCGA GUCGGUGC SEQ ID NO: 142 Nucleic acid sequence sgRNA backbone (3' to targeting sequence) (option 2) (may be followed by multiple uracils) GUUUAAGAGCUAGAAAUAGCAAGUUUAAAUAAGGCUAGUCCGUUAUCAACUUGAAAAAGU GGCACCGA GUCGGUGC SEQ ID NO: 143 Nucleic acid sequence sgRNA backbone (3' to targeting sequence) (option 3) (may be followed by multiple uracils) GUUUUAGAGCUAUGCUGGAAACAGCAUAGCAAGUUAAAAUAAGGCUAGUCCGUUAUCAAC UUGAAAAA GUGGCACCGAGUCGGUGC SEQ ID NO: 144 Nucleic acid sequence sgRNA backbone (3' to targeting sequence) (option 4) (may be followed by multiple uracils) GUUUAAGAGCUAUGCUGGAAACAGCAUAGCAAGUUUAAAUAAGGCUAGUCCGUUAUCAAC UUGAAAAA GUGGCACCGAGUCGGUGC SEQ ID NO: 145 Nucleic acid sequence dgRNA crRNA backbone (option 1) GUUUUAGAGCUAUGCUGUUUUG SEQ ID NO: 146 Nucleic acid sequence dgRNA tracrRNA (option 1) AACAGCAUAGCAAGUUAAAAUAAGGCUAGUCCGUUAUCAACUUGAAAAAGUGGCA CCGAGUCGGUGCU uuuuuu SEQ ID NO: 147 Nucleic acid sequence dgRNA crRNA backbone (option 2) GUUUAAGAGCUAUGCUGUUUUG SEQ ID NO: 148 Nucleic acid sequence dgRNA tracrRNA (option 2) AACAGCAUAGCAAGUUUAAAUAAGGCUAGUCCGUUAUCAACUUGAAAAAGUGGCA CCGAGUCGGUGCU uuuuuu

SpCas9 and Cas9 variants: SEQ ID NO: 150 Protein sequence SpCas9 WT MDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGALLFDSGETAE ATRLKRTARRRYTRRKNR ICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYPTIYHL RKKLVDSTDKADLRLIYLAL AHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSK SRRLENLIAQLPGEKKNG LFG NLIALSLGL TPNFKSN FDLAEDAKLQLSKDTYDDDLDN LLAQIGDQYADLFLAAKN LSDAILLSDILRVNTE ITKAP LSASM IKRYDEHHQDL TLLKAL VRQQLPEKYKEIFFDQSKNGYAGYIDGGASQEEFYKFI KPILEKM DGTEELL VKLN REDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIPYYVGP LARGNSRFAWMTRKSEETI TPWN FE EVVDKGASAQSFI E RMTN FDKN LPN E KVLPKHSLL YEYFTVYN EL TKVKYVTEG M RKP AFLSGEQKKAIV D LLFKTN RKVTVKQLKEDYFKKI ECFDSVE ISGVED RFNASLGTYH DLLKI I KDKDFLDN EE NED I LED IVL TL TLFE DRE MIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIRDKQSGKTILDFLKSDGFAN RNFMQLIHDDSLTFKE DIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENIVIEMARE NQTTQKGQKNSRERM KRIEEGIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDHI VPQSFLKDDSIDNKVLTR SDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLITQRKFDNLTKAERGGLSELDKAGFIKR QLVETRQITKHVAQIL DSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNYHHAHDAYLNAVVGT ALIKKYPKLESEFVYGDY KVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIV WDKGRDFATVRKVLSMP QVN IVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFDSPTVA YSVL VVAKVEKGKSKKLKSVKELLG ITI MERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRMLASAGELQKGNELAL PSKYVNFLYLASHYEKLKG SPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKHRDKPIREQAE NIIHLFTLTNLGAPAAFKYF DTTIDRKRYTSTKEVLDATLIHQSITG L YETRIDLSQLGGD SEQ ID NO: 151 Protein sequence SpCas9 Variant 1 MAPKKKRKVDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGALL FDSGETAEATRLKRTARR RYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHE KYPTIYHLRKKLVDSTDKA DLRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVDAK AILSARLSKSRRLENLIAQ LPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYAD LFLAAKNLSDAILLSDILR VNTEITKAPLSASM IKRYDEHHQDL TLLKAL VRQQLPEKYKEIFFDQSKNGYAGYIDGGASQEEFYKFI KPILEKM DG TEELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILT FRIPYYVGPLARGNSRFAW MTRKSEETITPWNFEEVVDKGASAQSFIERMTNFDKNLPNEKVLPKHSLLYEYFTVYNEL TKVKYVTEGMRKPAFLS GEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLLKII KDKDFLDNEENEDILEDIVL TLTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIRDKQSGKTILD FLKSDGFANRNFMQLIH DDSLTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPEN IVIEMARENQTTQKGQ KNSRERM KRIEEG IKELGSQILKEH PVENTQLQN EKLYL YYLQNG RDMYVDQELDI NRLSDYDVDHIVPQSFLKDDS IDN KVL TRSDKN RGKSDNVPSEEVVKKM KNYWRQLLNAKLITQRKFDN L TKAERGGLSELDKAGFI KRQL VETRQIT KHVAQILDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNYHHAHDAY LNAVVGTALIKKYPKLES EFVYGDYKVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETN GETGEIVWDKGRDFATVR KVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVL VVAKVEKGKSKKLKSVK ELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRMLASAGELQ KGNELALPSKYVNFLYLASHY EKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKHRDKPI REQAENIIHLFTLTNLGAP AAFKYFDTTI D RKRYTSTKEVLDATLI HQSITG L YETRIDLSQLGG DSRAD PKKKR KVH H H H H H SEQ ID NO: 152 Protein sequence SpCas9 Variant 2 MAPKKKRKVDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGALL FDSGETAEATRLKRTARR RYTRRKNRILYLQEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHE KYPTIYHLRKKLVDSTDKA DLRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVDAK AILSARLSKSRRLENLIAQ LPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYAD LFLAAKNLSDAILLSDILR VNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGA SQEEFYKFIKPILEKMDG TEELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILT FRIPYYVGPLARGNSRFAW MTRKSEET ITPWN FEEVVDKGASAQSFIERMTN FDKN LPNEKVLPKHSLL YEYFTVYNEL TKVKYVTEGM RKPAFLS G EQKKAIVDLLFKTN RKVTVKQLKEDYFKKI EE FDSVE ISGVE DRFNASLGTYH DLLKI I KDKDFLDN EE NED I LED IVL T LTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIRDKQSGKTILDF LKSDGFANRNFMQLIHD DSLTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENI VIEMARENQTTQKGQK NSRERM KRIEEGIKE LGSQILKEH PVENTQLQN EKL YL YYLQNG RDMYVDQELDI NRLSDYDVDH IVPQSFLKDDSI DNKVL TRSDKNRGKSDNVPSEEVVKKM KNYWRQLLNAKLITQRKFDNL TKAERGGLSELDKAGFI KRQLVETRQIT KHVAQILDSRM NTKYDE NDKLI REVKVITLKSKL VSDFRKDFQFYKVREI N NYH HAH DA YLNAVVGTALIKKYPKLES EFVYGDYKVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETN GETGEIVWDKGRDFATVR KVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVL VVAKVEKGKSKKLKSVK ELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRMLASAGELQ KGNELALPSKYVNFLYLASHY EKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKHRDKPI REQAENIIHLFTLTNLGAP AAFKYFDTTIDRKRYTSTKEVLDATLIHQSITGLYETRIDLSQLGGDSRADPKKKRKVHH HHHH SEQ ID NO: 153 Protein sequence SpCas9 Variant 3 MGSSHH H H H H H HEN L YFQGSM DKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLG NTDRHSI KKN LIGALLFDSG ETAEATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERH PIFGNIVDEVAYHEKYPTI YHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQL FEENPINASGVDAKAILSA RLSKSRRLENLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDD LDNLLAQIGDQYADLFLA AKNLSDAILLSDILRVNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFF DQSKNGYAGYIDGGASQEE FYKFI KPI LE KM DGTEE LL VKLN RE DLLRKQRTFD N GSI PH QI H LG E LHAI LRRQED FYP FLKDN REKI EKI L TFRIPYYV G PLARG NSRFAWMTRKSEETITPWN FEEVVDKGASAQSFI ERM TN FD KN LPN E KVLP KHSLL YEYFTVYN EL TKVK YVTEGMRKPAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFN ASLGTYHDLLKIIKDKDFL DNEENEDILED IVLTLTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIRDKQSGKT ILDFLKSD GFANRNFMQLIHDDSLTFKED IQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENIVIE MARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDM YVDQELDINRLSDYD VDH IVPQSFLKDDSIDNKVL TRSDKN RGKSDNVPSEEVVKKM KNYWRQLLNAKLITQRKFDNL TKAERGGLSELDK AGFIKRQL VETRQITKHVAQILDSRM NTKYDENDKLIREVKVITLKSKL VSDFRKDFQFYKVREIN NYHHAH DA YLNA VVGTALIKKYPKLESEFVYGDYKVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEIT LANGEIRKRPLIETNGETGE IVWDKG RDFA TVRKVLSM PQVNIVKKTEVQTGGFSKESI LP KRNSDKLIARKKDWDPKKYGG FDSPTVA YSVL VVA KVEKGKSKKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELE NGRKRMLASAGELQKGNELA LPSKYVNFL YLASHYEKLKGSPEDNEQKQLFVEQH KHYLDEIIEQISEFSKRVI LADANLDKVLSA YN KH RDKPIREQA ENIIHLFTLTNLGAPAAFKYFDTT IDRKRYTSTKEVLDATLIHQSITGLYETRIDLSQLGGDGGGSPKKKRKV SEQ ID NO: 154 Protein sequence SpCas9 Variant 4 MAHHHHHHGGSPKKKRKVDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSI KKNLIGALLFDSGETA EATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIF GNIVDEVAYHEKYPTIYHL RKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEE NPINASGVDAKAILSARLS KSRRLENLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDN LLAQIGDQYADLFLAAKN LSDAILLSDILRVNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQS KNGYAGYIDGGASQEEFYKF IKPILEKMDGTEELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKD NREKIEKILTFRIPYYVGPLA RGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIERMTNFDKNLPNEKVLPKHSLLYE YFTVYNELTKVKYVTE GMRKPAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLG TYHDLLKIIKDKDFLDNEE N EDILED IVL TL TLFE DREMIEERLKTYAH LFDDKVM KQLKRRRYTGWGRLSRKLINGIRDKQSGKTI LDFLKSDGFA NRNFMQLIHDDSLTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVK VMGRHKPENIVIEMAR ENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDMYVD QELDINRLSDYDVDHI VPQSFLKD DSI D N KVL TRSDKN RG KSDNVPSE EVVKKM KNYWRQLLNAKLITQR KFDN L TKAE RGG LSE LD KAG Fl KRQL VETRQITKHVAQILDSRM NTKYDENDKLI REVKVITLKSKLVSDFRKDFQFYKVREIN NYH HAH DA YLNAVVG TALIKKYPKLESEFVYGDYKVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEITLAN GEIRKRPLIETNGETGEIVW DKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGF DSPTVAYSVLVVAKVE KGKSKKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELENGR KRMLASAGELQKGNELALPS KYVN FL YLASHYE KLKGSP EDN EQKQLFVEQH KHYLDE 11 EQISE FSKRVI LA DAN LDKVLSA YN KH RDKP I REQAE N 11 H LFTL TNLGAPAAFKYFDTTIDRKRYTSTKEVLDATLIHQSITG L YETRIDLSQLGG DSRADP KKKRKV SEQ ID NO: 155 Protein sequence SpCas9 Variant 5 MAPKKKRKVDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGALL FDSGETAEATRLKRTARR RYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHE KYPTIYHLRKKLVDSTDKA DLRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVDAK AILSARLSKSRRLENLIAQ LPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYAD LFLAAKNLSDAILLSDILR VNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGA SQEEFYKFIKPILEKMDG TEELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILT FRIPYYVGPLARGNSRFAW MTRKSEETITPWNFEEVVDKGASAQSFIERMTNFDKNLPNEKVLPKHSLLYEYFTVYNEL TKVKYVTEGMRKPAFLS GEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLLKII KDKDFLDNEENEDILEDIVL TLTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIRDKQSGKTILD FLKSDGFANRNFMQLIH DDSLTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPEN IVIEMARENQTTQKGQ KNSRERMKRIEEGIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDMYVDQELDINRLS DYDVDHIVPQSFLKDDS IDN KVL TRSDKN RG KSDNVPSEEVVKKM KNYWRQLLNAKLITQRKFDN L TKAERGG LSELDKAGFIKRQL VETRQIT KHVAQILDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNYHHAHDAY LNAVVGTALIKKYPKLES EFVYGDYKVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETN GETGEIVWDKGRDFATVR KVLSM PQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFDSPTVA YSVL VVAKVEKG KSKKLKSVK ELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRMLASAGELQ KGNELALPSKYVNFLYLASHY EKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKHRDKPI REQAENIIHLFTLTNLGAP AAFKYFDTTIDRKRYTSTKEVLDATLIHQSITGLYETRIDLSQLGGDSRADHHHHHH SEQ ID NO: 156 Protein sequence SpCas9 Variant 6 MAH H H H H HGGSDKKYSIG LDIGTNSVGWA VITDEYKVPSKKFKVLG NTDRHSI KKN LIGALLFDSG ET AEATR LKRT ARRRYTRRKNRICYLQEIFSN EMAKVDDSFFHRLEESFL VEEDKKH ERH PIFG NIVDEVA YHEKYPTIYH LRKKL VDST DKADLRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGV DAKAILSARLSKSRRLENL IAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQ YADLFLAAKNLSDAILLS DILRVNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKNGYAGYI DGGASQEEFYKFIKPILEK MDGTEELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEK ILTFRIPYYVGPLARGNSR FAWMTRKSEETITPWN FEEVVDKGASAQSFIERMTN FDKNLPN EKVLPKHSLL YEYFTVYN EL TKVKYVTEG M RKP AFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDL LKIIKDKDFLDNEENEDILE DIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIRDKQSGK TILDFLKSDGFANRNFM QLIHDDSLTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRH KPENIVIEMARENQTT QKGQKNSRE RM KRI EEG I KELGSQI LKE H PVE NTQLQN EKL YL YYLQNG RD MYVDQE LD IN RLSDYDVD H IVPQSF LKDDS I DN KVL TRSDKN RG KSDNVPSEEVVKKM KNYWRQLLNAKLITQRKFDN L TKAE RGG LSE LDKAG Fl KRQL V ETRQITKHVAQILDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNYH HAHDAYLNAVVGTALIKK YPKLESEFVYGDYKVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKR PLIETNGETGEIVWDKGRD FATVRKVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFDSPTV AYSVLVVAKVEKGKSKK LKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRMLAS AGELQKGNELALPSKYVNFLY LASHYEKLKGSPEDN EQKQLFVEQH KHYLDEII EQISEFSKRVILADAN LDKVLSA YN KH RDKPIREQAENIIH LFTL TN LGAP AAFKYFDTT I D RKRYTSTKEVLDATLI H QSITG L VETRI DLSQLGG DSRADP KKKRKV SEQ ID NO: 157 Protein sequence SpCas9 Variant 7 MG SSH H H H H HH HE NL YFQGSM DKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLG NTDRHSI KKN LIGALLFDSG ETAEATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERH PIFGNIVDEVAYHEKYPTI YH LRKKL VDSTDKADLRLIYLALAH MI KFRGH FLIEGDLN PDNSDVDKLFIQL VQTYNQLFE EN Pl NASGVDAKAILSA RLSKSRRLENLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDD LDNLLAQIGDQYADLFLA AKNLSDAILLSDILRVNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFF DQSKNGYAGYIDGGASQEE FYKFIKPILEKMDGTEELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYP FLKDNREKIEKILTFRIPYYV GPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIERMTNFDKNLPNEKVLPKHS LLYEYFTVYNELTKVK YVTEGMRKPAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFN ASLGTYHDLLKIIKDKDFL DNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLI NGIRDKQSGKTILDFLKSD GFANRNFMQLIHDDSLTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDE LVKVMGRHKPENIVIE MARENQTTQKGQKNSRERM KRIEEG I KELGSQILKEHPVENTQLQN EKL YL YYLQNGRDMYVDQELDI N RLSDYD VDH IVPQSFLKDDSIDNKVL TRSDKN RGKSDNVPSEEVVKKM KNYWRQLLNAKLITQRKFDN L TKAERGGLSELDK AGFI KRQL VETRQITKHVAQILDSRM NTKYDENDKLIREVKVITLKSKL VSDFRKDFQFYKVREIN NYHHAHDA YLNA VVGTALIKKYPKLESEFVYGDYKVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEIT LANGEIRKRPLIETNGETGE IVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKY GGFDSPTVAYSVLVVA KVEKGKSKKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELE NGRKRMLASAGELQKGNELA LPSKYVNFLYLASHYEKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANL DKVLSAYNKHRDKPIREQA ENIIHLFTLTNLGAPAAFKYFDTT IDRKRYTSTKEVLDATLIHQSITGLYETRIDLSQLGGDGGGSPKKKRKV SEQ ID NO: 158 Protein sequence SpCas9 Variant 8 MAPKKKRKVDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGALL FDSGETAEATRLKRTARR RYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHE KYPTIYHLRKKLVDSTDKA DLRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVDAK AILSARLSKSRRLENLIAQ LPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYAD LFLAAKNLSDAILLSDILR VNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGA SQEEFYKFIKPILEKMDG TEELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILT FRIPYYVGPLARGNSRFAW MTRKSEETITPWNFEEVVDKGASAQSFIERMTNFDKNLPNEKVLPKHSLLYEYFTVYNEL TKVKYVTEGMRKPAFLS GEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLLKII KDKDFLDNEENEDILEDIVL TLTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIRDKQSGKTILD FLKSDGFANRNFMQLIH DDSLTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPEN IVIEMARENQTTQKGQ KNSRE RM KR IE EG I KE LGSQI LKE H PVE NTQLQN E KL YL YYLQNG R DMYVDQE LDI N RLSDYDVDH IVPQSFLKDDS I DNAVL TRSDKNRGKSDNVPSEEVVKKM KNYWRQLLNAKLITQRKFDNL TKAE RGG LSELDKAGFI KRQL VETRQI TKHVAQILDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNYHHAHDA YLNAVVGTALIKKYPKLE SEFVYGDYKVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTE ITLANGEIRKRPLIETNGETGEIVWDKGRDFATV RKVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSV LVVAKVEKGKSKKLKSV KELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRMLASAGEL QKGNELALPSKYVNFLYLAS HYEKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKHRDK PIREQAENIIHLFTLTNLG AP AAFKYFDTTIDR KRYTSTKEVLDATLI HQS ITG L YETRIDLSQLGGDSRADP KKKRKVH H H H H H SEQ ID NO: 159 Protein sequence SpCas9 Variant 9 MAPKKKRKVDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGALL FDSGETAEATRLKRTARR RYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHE KYPTIYHLRKKLVDSTDKA DLRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVDAK AILSARLSKSRRLENLIAQ LPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYAD LFLAAKNLSDAILLSDILR VNTEITKAPLSASM IKRYDEH HQDL TLLKAL VRQQLPEKYKEIFFDQSKNGYAGYIDGGASQEEFYKFIKPILEKM DG TEELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILT FRIPYYVGPLARGNSRFAW MTRKSEET ITPWNFEEVVDKGASAQSFIERMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTE GMRKPAFLS GEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLLKII KDKDFLDNEENEDILEDIVL TLTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIRDKQSGKTILD FLKSDGFANRNFMQLIH DDSLTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPEN IVIEMARENQTTQKGQ KNSRERMKRIEEGIKELGSQILKEHPVENTQLQNEALYLYYLQNGRDMYVDQELDINRLS DYDVDHIVPQSFLKDDS IDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLITQRKFDNLTKAERGGLSEL DKAGFIKRQLVETRQIT KHVAQILDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNYHHAHDAY LNAVVGTALIKKYPALES EFVYGDYKVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKAPLIETN GETGEIVWDKGRDFATVR KVLSM PQVNIVKKTEVQTGGFSKESI LPKRNSDKLIARKKDWDPKKYGGFDSPTVA YSVL VVAKVEKGKSKKLKSVK ELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRMLASAGELQ KGNELALPSKYVNFLYLASHY EKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKHRDKPI REQAENIIHLFTLTNLGAP AAFKYFDTTIDRKRYTSTKEVLDATLIHQSITGLYETRIDLSQLGGDSRADPKKKRKVHH HHHH SEQ ID NO: 160 Protein sequence SpCas9 Variant 10 MAPKKKRKVDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGALL FDSGETAEATRLKRTARR RYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHE KYPTIYHLRKKLVDSTDKA DLRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVDAK AILSARLSKSRRLENLIAQ LPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYAD LFLAAKNLSDAILLSDILR VNTEITKAPLSASM IKRYDEH HQDL TLLKAL VRQQLPEKYKEIFFDQSKNGYAGYIDGGASQEEFYKFIKPI LEKMDG TEELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILT FRIPYYVGPLARGNSRFAW MTRKSEETITPWNFEEVVDKGASAQSFIERMTNFDKNLPNEKVLPKHSLLYEYFTVYNEL TKVKYVTEGMRKPAFLS GEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLLKII KDKDFLDNEENEDILEDIVL TLTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIRDKQSGKTILD FLKSDGFANRNFMQLIH DDSLTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPEN IVIEMARENQTTQKGQ KNSRE RM KRI EEG I KE LGSQI LKEH PVE NTQLQN E KL YL YYLQNG RDMYVDQELD IN RLSDYDVD H IVPQS FLADDS IDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLITQRKFDNLTKAERGGLSEL DKAGFIKRQLVETRQIT KHVAQILDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNYHHAHDAY LNAVVGTALIKKYPALES EFVYGDYKVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKAPLIETN GETGEIVWDKGRDFATVR KVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVL VVAKVEKGKSKKLKSVK ELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRMLASAGELQ KGNELALPSKYVNFLYLASHY EKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKHRDKPI REQAENIIHLFTLTNLGAP AAFKYFDTTIDRKRYTSTKEVLDATLIHQSITGLYETRIDLSQLGGDSRADPKKKRKVHH HHHH SEQ ID NO: 161 Protein sequence SpCas9 Variant 11 MAPKKKRKVDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGALL FDSGETAEATRLKRTARR RYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHE KYPTIYHLRKKLVDSTDKA DLRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVDAK AILSARLSKSRRLENLIAQ LPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYAD LFLAAKNLSDAILLSDILR VNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGA SQEEFYKFIKPILEKMDG TEELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILT FRIPYYVGPLARGNSRFAW MTRKSEETITPWNFEEVVDKGASAQSFIERMTAFDKNLPNEKVLPKHSLLYEYFTVYNEL TKVKYVTEGMRKPAFLS GEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLLKII KDKDFLDNEENEDILEDIVL TLTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGALSRKLINGIRDKQSGKTILD FLKSDGFANRNFMALIH DDSLTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPEN IVIEMARENQTTQKGQ KNSRERMKRIEEGIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDMYVDQELDINRLS DYDVDHIVPQSFLKDDS I DN KVL TRSDKN RG KSDNVPSEEVVKKM KNYWRQLLNAKLITQRKFDN L TKAERGGLSELDKAGFI KRQL VETRAIT KHVAQILDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNYHHAHDAY LNAVVGTALIKKYPKLES EFVYGDYKVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETN GETGEIVWDKGRDFATVR KVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVL VVAKVEKGKSKKLKSVK ELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRMLASAGELQ KGNELALPSKYVNFLYLASHY EKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKHRDKPI REQAENIIHLFTLTNLGAP AAFKYFDTT IDRKRYTSTKEVLDATLIHQSITGLYETRIDLSQLGGDSRADPKKKRKVHHHHHH

gRNA subparts (e.g., for Cas12a): SEQ ID NO: 201 Nucleic acid sequence crRNA repeat sequence (option 1) UAAUUUCUACUCUUGUAGAU SEQ ID NO: 202 Nucleic acid sequence crRNA repeat sequence (option 2) AAUUUCUACUCUUGUAGAU SEQ ID NO: 203 Nucleic acid sequence crRNA repeat sequence (option 3) UAAUUUCUACUAAGUGUAGAU SEQ ID NO: 204 Nucleic acid sequence crRNA repeat sequence (option 4) AAUUUCUACUAAGUGUAGAU SEQ ID NO: 205 Nucleic acid sequence crRNA repeat sequence (option 5) UAAUUUCUACUGUUGUAGAU SEQ ID NO: 206 Nucleic acid sequence crRNA repeat sequence (option 6) AAUUUCUACUGUUGUAGAU

AsCas12a and AsCas12a variants: SEQ ID NO: 210 Protein sequence AsCas9 WT MTQFEGFTN L YQVSKTLRFELIPQGKTLKHIQEQG FIEEDKARNDHYKELKPIIDRIYKTY ADQCLQL VQLDWENLSA AIDSYRKEKTEETRNALIEEQATYRNAIHDYFIGRTDNLTDAINKRHAEIYKGLFKAELF NGKVLKQLGTVTTTEHENA LLRSFDKFTTYFSGFYENRKNVFSAEDISTAIPHRIVQDNFPKFKENCHIFTRLITAVPS LREHFENVKKAIGIFVSTSIEE VFSFPFYN QLL TQTQI D LYN QLLGG ISREAGTE Kl KG LN EVLN LAIQKN DET AH I IASLPH RFI PLFKQI LSDRNTLSF I LE EFKSDEEVIQSFCKYKTLLRNENVLETAEALFN ELNSIDL THI FISH KKLETISSALCDHWDTLRNAL YERRISEL TGKITK SAKEKVQRSLKHEDINLQEIISAAGKELSEAFKQKTSEILSHAHAALDQPLPTTLKKQEE KEILKSQLDSLLGLYHLLDW FAVDESNEVDPEFSARLTGIKLEMEPSLSFYNKARNYATKKPYSVEKFKLNFQMPTLASG WDVNKEKNNGAILFVK NGLYYLGIMPKQKGRYKALSFEPTEKTSEGFDKMYYDYFPDAAKMIPKCSTQLKAVTAHF QTHTTPILLSNNFIEPLE ITKEIYDLNNPEKEPKKFQTAYAKKTGDQKGYREALCKWIDFTRDFLSKYTKTTSIDLSS LRPSSQYKDLGEYYAELNPL LYHISFQRIAEKEIMDAVETGKLYLFQIYNKDFAKGHHGKPNLHTLYWTGLFSPENLAKT SIKLNGQAELFYRPKSRM KR MAH RLGEKM LN KKLKDQKTP IPDTLYQEL YDYVN H RLSH DLSDEARALLPNVITKEVSH El I KDRRFTSDKFFFHV PITLNYQAANSPSKFNQRVNAYLKEHPETPIIGIDRGERNLIYITVIDSTGKILEQRSLN TIQQFDYQKKLDNREKERVA ARQAWSVVGTIKDLKQGYLSQVIHEIVDLMIHYQAVVVLENLNFGFKSKRTGIAEKAVYQ QFEKMLIDKLNCLVLKD YPAEKVGGVLN PYQL TDQFTSFAKMGTQSG FLFYVPAPYTSKIDPL TG FVDPFVWKTIKN H ESRKH FLEG FDFLHYD VKTGDFILHFKMNRNLSFQRGLPGFMPAWDIVFEKNETQFDAKGTPFIAGKRIVPVIENH RFTGRYRDLYPANELIA LLEEKGIVFRDGSNILPKLLENDDSHAIDTMVALIRSVLQMRNSNAATGEDYINSPVRDL NGVCFDSRFQNPEWPM DADANGAYHIALKGQLLLNHLKESKDLKLQNGISNQDWLAYIQELRN SEQ ID NO: 211 Protein sequence AsCas12a Ultra (M537R, F870L relative to WT) MTQFE G FTN L YQVSKTLRFELI P QG KTLKH I QEQG FIE ED KARN D HYKELKPI I DRIYKTY ADQCLQL VQLDWEN LSA Al DSYRKEKTEETRNALIE EQA TYRNAI H DYF IG RTD NL TDAIN KRHAEIYKGLFKAE LF NG KVLKQLGTVTTTEH ENA LLRSFDKFTTYFSGFYENRKNVFSAEDISTAIPHRIVQDNFPKFKENCHIFTRLITAVPS LREHFENVKKAIGIFVSTSIEE VFSFPFYNQLLTQTQIDLYNQLLGGISREAGTEKIKGLNEVLNLAIQKNDETAHIIASLP HRFIPLFKQILSDRNTLSFILE EFKSDEEVIQSFCKYKTLLRNENVLETAEALFNELNSIDLTHIFISHKKLETISSALCDH WDTLRNALYERRISELTGKITK SAKEKVQRSLKHEDINLQEIISAAGKELSEAFKQKTSEILSHAHAALDQPLPTTLKKQEE KEILKSQLDSLLGLYHLLDW FAVDESNEVDPEFSARLTGIKLEMEPSLSFYNKARNYATKKPYSVEKFKLNFQRPTLASG WDVNKEKNNGAILFVKN GLYYLGIMPKQKGRYKALSFEPTEKTSEGFDKMYYDYFPDAAKMIPKCSTQLKAVTAHFQ THTTPILLSNNFIEPLEIT KEIYDLNNPEKEPKKFQTAYAKKTGDQKGYREALCKWIDFTRDFLSKYTKTTSIDLSSLR PSSQYKDLGEYYAELNPLL YHISFQRIAEKEIMDAVETGKLYLFQIYNKDFAKGHHGKPNLHTLYWTGLFSPENLAKTS IKLNGQAELFYRPKSRMK RMAH RLG EKM LN KKLKDQKTPIPDTLYQE LYDYVN H RLSHDLSDEARALLPNVITKEVSH Ell KDRRFTSDKFLFHVP ITLNYQAANSPSKFNQRVNAYLKEHPETPIIGIDRGERNLIYITVIDSTGKILEQRSLNT IQQFDYQKKLDNREKERVAA RQAWSVVGTIKDLKQGYLSQVIHEIVDLMIHYQAVVVLENLNFGFKSKRTGIAEKAVYQQ FEKMLIDKLNCLVLKDY PAEKVGGVLNPYQLTDQFTSFAKMGTQSGFLFYVPAPYTSKIDPLTGFVDPFVWKTIKNH ESRKHFLEGFDFLHYDV KTGDFILH FKM NRN LSFQRGLPGFM PAWDIVFEKN ETQFDAKGTPFIAGKRIVPVIENH RFTGRYRDL YPAN ELIAL LEEKGIVFRDGSNILPKLLENDDSHAIDTMVALIRSVLQMRNSNAATGEDYINSPVRDLN GVCFDSRFQNPEWPM DADANGA YH IALKGQLLLNHLKESKDLKLQNG ISNQDWLA YIQE LRN SEQ ID NO: 212 Protein sequence enAsCas12a (E174R, S542R, and K548R relative to WT) MTQFEGFTNLYQVSKTLRFELIPQGKTLKHIQEQGFIEEDKARNDHYKELKPIIDRIYKT YADQCLQLVQLDWENLSA Al DSYRKE KTE ETRNALI EE QA TYRNAI H DYFIG RTD NL TDAIN KRHAEIYKGLFKAE LFNG KVLKQLGTVTTTE HE NA LLRSFDKFTTY FSGFYRNRKNVFSAEDISTAIPHRIVQDNFPKFKENCHIFTRLITAVPSLREHFENVKKA IGIFVSTSIEE VFSFPFYN QLL TQTQI DL YNQLLGG ISREAGTEKI KG LN EVLN LAIQKN DET AH I IASLPH RFI PLFKQI LSDRNTLSF I LE EFKSDEEVIQSFCKYKTLLRNENVLET AEALFN ELNSIDL THI FISHKKLETISSALCDHWDTLRNAL YERRISEL TGKITK SAKE KVQRSLKH EDIN LQE I ISAAG KE LSEAFKQKTSE I LSHAHAALDQPLPTTLKKQEE KE ILKSQLDSLLG L YH LLDW FAVDESNEVDPEFSARLTGIKLEMEPSLSFYNKARNYATKKPYSVEKFKLNFQMPTLARG WDVNREKNNGAILFVK NGLYYLGIMPKQKGRYKALSFEPTEKTSEGFDKMYYDYFPDAAKMIPKCSTQLKAVTAHF QTHTTPILLSNNFIEPLE ITKEIYDLNNPEKEPKKFQTAYAKKTGDQKGYREALCKWIDFTRDFLSKYTKTTSIDLSS LRPSSQYKDLGEYYAELNPL LYHISFQRIAEKEIMDAVETGKLYLFQIYNKDFAKGHHGKPNLHTLYWTGLFSPENLAKT SIKLNGQAELFYRPKSRM KR MAH RLGEKMLN KKLKDQKTPIPDTL YQEL YDYVNH RLSH DLSDEARALLPNVITKEVSH EIIKDRRFTSDKFFFHV PITLNYQAANSPSKFNQRVNAYLKEHPETPIIGIDRGERNLIYITVIDSTGKILEQRSLN TIQQFDYQKKLDNREKERVA ARQAWSVVGTIKDLKQGYLSQVIHEIVDLMIHYQAVVVLENLNFGFKSKRTGIAEKAVYQ QFEKMLIDKLNCLVLKD YPAEKVGGVLNPYQLTDQFTSFAKMGTQSGFLFYVPAPYTSKIDPLTGFVDPFVWKTIKN HESRKHFLEGFDFLHYD VKTGDFILHFKMNRNLSFQRGLPGFMPAWDIVFEKNETQFDAKGTPFIAGKRIVPVIENH RFTGRYRDLYPANELIA LLEEKGIVFRDGSNILPKLLENDDSHAIDTMVALIRSVLQMRNSNAATGEDYINSPVRDL NGVCFDSRFQNPEWPM DADANGA YH IALKGQLLLNH LKESKDLKLQNG ISNQDWLAYIQE LRN SEQ ID NO: 213 Protein sequence enAsCas12a-Hifi (E174R, S542R, and K548R + N282A relative to WT) MTQFEGFTNLYQVSKTLRFELIPQGKTLKHIQEQGFIEEDKARNDHYKELKPIIDRIYKT YADQCLQLVQLDWENLSA AIDSYRKE KTEETRNALIE EQA TYRNAI H DYF IG RTD NL TDAIN KRHAEIYKG LFKAE LFNG KVLKQLGTVTTTE HE NA LLRSFDKFTTYFSGFYRNRKNVFSAEDISTAIPHRIVQDNFPKFKENCHIFTRLITAVPS LREHFENVKKAIGIFVSTSIEE VFSFPFYNQLLTQTQIDLYNQLLGGISREAGTEKIKGLNEVLALAIQKNDETAHIIASLP HRFIPLFKQILSDRNTLSFILE EFKSDEEVIQSFCKYKTLLRN ENVLETAEALFN ELNSIDL THI FISH KKLETISSALCDHWDTLRNAL YERRISEL TGKITK SAKEKVQRSLKHEDINLQEIISAAGKELSEAFKQKTSEILSHAHAALDQPLPTTLKKQEE KEILKSQLDSLLGLYHLLDW FAVDESNEVDPEFSARLTGIKLEMEPSLSFYNKARNYATKKPYSVEKFKLNFQMPTLARG WDVNREKNNGAILFVK NG L YYLG IMP KQKG RYKALSFE PTEKTSE G FD KMYYDYFPDAAKM I PKCSTQLKA VT AH FQTHTTP I LLSN N FIE P LE ITKEIYDLNNPEKEPKKFQTAYAKKTGDQKGYREALCKWIDFTRDFLSKYTKTTSIDLSS LRPSSQYKDLGEYYAELNPL LYHISFQRIAEKEIMDAVETGKLYLFQIYNKDFAKGHHGKPNLHTLYWTGLFSPENLAKT SIKLNGQAELFYRPKSRM KRMAHRLGEKMLNKKLKDQKTPIPDTLYQELYDYVNHRLSHDLSDEARALLPNVITKEVS HEIIKDRRFTSDKFFFHV PITLNYQAANSPSKFNQRVNAYLKEHPETPIIGIDRGERNLIYITVIDSTGKILEQRSLN TIQQFDYQKKLDNREKERVA ARQAWSVVGTIKDLKQGYLSQVIHEIVDLMIHYQAVVVLENLNFGFKSKRTGIAEKAVYQ QFEKMLIDKLNCLVLKD YPAEKVGGVLNPYQLTDQFTSFAKMGTQSGFLFYVPAPYTSKIDPLTGFVDPFVWKTIKN HESRKHFLEGFDFLHYD VKTGDFILHFKMNRNLSFQRGLPGFMPAWDIVFEKNETQFDAKGTPFIAGKRIVPVIENH RFTGRYRDLYPANELIA LLEEKGIVFRDGSNILPKLLENDDSHAIDTMVALIRSVLQMRNSNAATGEDYINSPVRDL NGVCFDSRFQNPEWPM DADANGAYHIALKGQLLLNHLKESKDLKLQNGISNQDWLAYIQELRN SEQ ID NO: 214 Protein sequence enAsCas12a Ultra (E174R, S542R, and K548R + M537R, F870L relative to WT) MTQFEGFTNLYQVSKTLRFELIPQGKTLKHIQEQGFIEEDKARNDHYKELKPIIDRIYKT YADQCLQLVQLDWENLSA Al DSYRKEKTEETRNALIE EQA TYRNAI H DYF IG RTD NL TDAIN KR HAE IYKGLFKAE LF NG KVLKQLGTVTTTE HE NA LLRSFDKFTTYFSGFYRNRKNVFSAEDISTAIPHRIVQDNFPKFKENCHIFTRLITAVPS LREHFENVKKAIGIFVSTSIEE VFSFP FYNQLL TQTQI DL YNQLLGG ISREAGTE Kl KG LN EVLN LAIQKN DET AH I IASLP HR Fl PLFKQI LSD R NTLSFI LE EFKSDEEVIQSFCKYKTLLRNENVLETAEALFN ELNSIDL THI FISH KKLETISSALCDHWDTLRNAL YERRISEL TGKITK SAKEKVQRSLKHEDINLQEIISAAGKELSEAFKQKTSEILSHAHAALDQPLPTTLKKQEE KEILKSQLDSLLGLYHLLDW FAVDESNEVDPEFSARLTGIKLEMEPSLSFYNKARNYATKKPYSVEKFKLNFQRPTLARG WDVNREKNNGAILFVKN GLYYLGIMPKQKGRYKALSFEPTEKTSEGFDKMYYDYFPDAAKMIPKCSTQLKAVTAHFQ THTTPILLSNNFIEPLEIT KEIYDLNN PEKEPKKFQTA YAKKTGDQKGYREALCKWI DFTRDFLSKYTKTTSIDLSSLRPSSQYKDLGEYYAELNPLL YHISFQRIAEKEIMDAVETGKLYLFQIYNKDFAKGHHGKPNLHTLYWTGLFSPENLAKTS IKLNGQAELFYRPKSRMK RMAH RLGEKM LN KKLKDQKTPIPDTL YQELYDYVNH RLSHDLSDEARALLPNVITKEVSH Ell KDRRFTSDKFLFHVP ITLNYQAANSPSKFNQRVNAYLKEHPETPIIGIDRGERNLIYITVIDSTGKILEQRSLNT IQQFDYQKKLDNREKERVAA RQAWSVVGTIKDLKQGYLSQVIHEIVDLMIHYQAVVVLENLNFGFKSKRTGIAEKAVYQQ FEKMLIDKLNCLVLKDY PAEKVGGVLNPYQLTDQFTSFAKMGTQSGFLFYVPAPYTSKIDPLTGFVDPFVWKTIKNH ESRKHFLEGFDFLHYDV KTGDFILHFKMNRNLSFQRGLPGFMPAWDIVFEKNETQFDAKGTPFIAGKRIVPVIENHR FTGRYRDLYPANELIAL LEEKGIVFRDGSNILPKLLENDDSHAIDTMVALIRSVLQMRNSNAATGEDYINSPVRDLN GVCFDSRFQNPEWPM DADANGAYHIALKGQLLLNHLKESKDLKLQNGISNQDWLAYIQELRN SEQ ID NO: 215 Protein sequence enAsCas12a-Hifi Ultra (E174R, S542R, and K548R + N282A + M537R, F870L relative to WT) MTQFE G FTN L YQVSKTLRFE LI PQG KTLKH IQEQG Fl EE D KARN D HYKELKP I IDRIYKTY ADQCLQL VQLDWE N LSA AIDSYRKE KTEETRNALI EE QATYRNAI H DYF IG RTD NL TDAI N KRHAEIYKG LFKAELF NG KVLKQLGTVTTTE HE NA LLRSFDKFTTYFSG FYRN RKNVFSAE DIST Al PH RIVQD N FP KFKE NCH I FTRLIT AVPSLRE H FE NVKKAIG I FVSTSI EE VFSFPFYNQLLTQTQIDLYNQLLGGISREAGTEKIKGLNEVLALAIQKNDETAHIIASLP HRFIPLFKQILSDRNTLSFILE E FKSDEEVIQSFCKYKTLLRN E NV LET AEALFN ELNSI DL TH IF IS H KKLETISSALCDHWDTLRNAL YE RRISEL TGKITK SAKEKVQRSLKHEDINLQEIISAAGKELSEAFKQKTSEILSHAHAALDQPLPTTLKKQEE KEILKSQLDSLLGLYHLLDW FAVDESNEVDPEFSARLTGIKLEMEPSLSFYNKARNYATKKPYSVEKFKLNFQRPTLARG WDVNREKNNGAILFVKN GLYYLGIMPKQKGRYKALSFEPTEKTSEGFDKMYYDYFPDAAKMIPKCSTQLKAVTAHFQ THTTPILLSNNFIEPLEIT KEIYDLNNPEKEPKKFQTAYAKKTGDQKGYREALCKWIDFTRDFLSKYTKTTSIDLSSLR PSSQYKDLGEYYAELNPLL YHISFQRIAEKEIMDAVETGKLYLFQIYNKDFAKGHHGKPNLHTLYWTGLFSPENLAKTS IKLNGQAELFYRPKSRMK RMAHRLGEKMLNKKLKDQKTPIPDTLYQELYDYVNHRLSHDLSDEARALLPNVITKEVSH EIIKDRRFTSDKFLFHVP ITLNYQAANSPSKFNQRVNAYLKEHPETPIIGIDRGERNLIYITVIDSTGKILEQRSLNT IQQFDYQKKLDNREKERVAA RQAWSVVGTIKDLKQGYLSQVIHEIVDLMIHYQAVVVLENLNFGFKSKRTGIAEKAVYQQ FEKMLIDKLNCLVLKDY PAEKVGGVLNPYQLTDQFTSFAKMGTQSGFLFYVPAPYTSKIDPLTGFVDPFVWKTIKNH ESRKHFLEGFDFLHYDV KTGDFILHFKMNRNLSFQRGLPGFMPAWDIVFEKNETQFDAKGTPFIAGKRIVPVIENHR FTGRYRDLYPANELIAL LEEKGIVFRDGSNILPKLLENDDSHAIDTMVALIRSVLQMRNSNAATGEDYINSPVRDLN GVCFDSRFQNPEWPM DADANGAYHIALKGQLLLNHLKESKDLKLQNGISNQDWLAYIQELRN

LbCas12a and LbCas12a variants: SEQ ID NO: 220 Protein sequence LbCas12a WT MSKLEKFTNCYSLSKTLRFKAIPVGKTQENIDNKRLLVEDEKRAEDYKGVKKLLDRYYLS FINDVLHSIKLKNLNNYISL FRKKTRTEKENKELENLEINLRKEIAKAFKGNEGYKSLFKKDIIETILPEFLDDKDEIAL VNSFNGFTTAFTGFFDNREN MFSEEAKSTSIAFRCINENLTRYISNMDIFEKVDAIFDKHEVQEIKEKILNSDYDVEDFF EGEFFNFVLTQEGIDVYNAII GG FVTESG E Kl KG LN EYI NL YNQKTKQKLP KFKPL YKQVLSDRESLSFYG E GYTSDE EVLEVFRNTLN KN SE I FSSI KKL EKLFKNFDEYSSAGIFVKNGPAISTISKDIFGEWNVIRDKWNAEYDDIHLKKKAVVTEKY EDDRRKSFKKIGSFSLEQL QEYADADLSVVEKLKEIIIQKVDEIYKVYGSSEKLFDADFVLEKSLKKNDAVVAIMKDLL DSVKSFENYIKAFFGEGKET N RDESFYGDFVLA YDILLKVDH IYDAIRNYVTQKPYSKDKFKL YFQN PQFMGGWDKDKETDYRATILRYGSKYYLAI M DKKYAKCLQKIDKDDVNGNYEKINYKLLPGPN KM LPKVFFSKKWMA YYN PSEDIQKIYKNGTFKKGDM FN LND CH KLIDFFKDSISRYPKWSNA YDFN FSETEKYKDIAGFYREVEEQGYKVSFESASKKEVDKL VEEGKL YM FQIYN KDFS DKSHGTPNLHTMYFKLLFDENNHGQIRLSGGAELFMRRASLKKEELVVHPANSPIANKNP DNPKKTTTLSYDVYKD KRFSEDQYELHIPIAINKCPKNIFKINTEVRVLLKHDDNPYVIGIDRGERNLLYIVVVDG KGNIVEQYSLNEIINNFNGIR I KTDYHSLLD KKE KE RF EARQNWTSI EN I KE LKAGYISQVVH KICEL VEKYDA VIALEDLNSG FKNS RVKVE KQVYQKF EKMLIDKLNYMVDKKSNPCATGGALKGYQITNKFESFKSMSTQNGFIFYIPAWLTSKIDP STGFVNLLKTKYTSIADS KKFISSFDRIMYVPEEDLFEFALDYKN FSRTDADYIKKWKLYSYGN RI RIFRN PKKN NVFDWEEVCL TSA YKE LFN KYG INYQQGDIRALLCEQSDKAFYSSFMALMSLMLQMRNSITGRTDVDFLISPVKNSDGIFYD SRNYEAQENAILPKNA DAN GA YN IARKVLWAIGQFKKAEDEKLDKVKIAISN KEWLEYAQTSVKH SEQ ID NO: 221 Protein sequence LbCas12a Ultra (E795L relative to WT) MSKLEKFTNCYSLSKTLRFKAIPVGKTQENIDNKRLLVEDEKRAEDYKGVKKLLDRYYLS FINDVLHSIKLKNLNNYISL FRKKTRTEKENKELENLEINLRKEIAKAFKGNEGYKSLFKKDIIETILPEFLDDKDEIAL VNSFNGFTTAFTGFFDNREN MFSEEAKSTSIAFRCINENLTRYISNMDIFEKVDAIFDKHEVQEIKEKILNSDYDVEDFF EGEFFNFVLTQEGIDVYNAII GGFVTESGEKIKG LN EVIN L YNQKTKQKLPKFKPL YKQVLSDRESLSFYGEGYTSDEEVLEVFRNTLN KNSEI FSSIKKL EKLFKNFDEYSSAGIFVKNGPAISTISKDIFGEWNVIRDKWNAEYDDIHLKKKAVVTEKY EDDRRKSFKKIGSFSLEQL QEYADADLSVVEKLKEIIIQKVDEIYKVYGSSEKLFDADFVLEKSLKKNDAVVAIMKDLL DSVKSFENYIKAFFGEGKET NRDESFYGDFVLAYDILLKVDHIYDAIRNYVTQKPYSKDKFKLYFQNPQFMGGWDKDKET DYRATILRYGSKYYLAI M DKKYAKCLQKIDKDDVNGNYEKI NYKLLPG PN KMLPKVFFSKKWMA YYNPSEDIQKIYKNGTFKKGDM FN LND CH KU DFFKDSISRYPKWSNA YDFNFSETEKYKDIAGFYREVEEQGYKVSFESASKKEVDKL VEEGKL YM FQIYN KDFS DKSHGTPNLHTMYFKLLFDENNHGQIRLSGGAELFMRRASLKKEELVVHPANSPIANKNP DNPKKTTTLSYDVYKD KRFSEDQYLLHIPIAINKCPKNIFKINTEVRVLLKHDDNPYVIGIDRGERNLLYIVVVDG KGNIVEQYSLNEIINNFNGIR IKTDYHSLLDKKEKERFEARQNWTSIENIKELKAGYISQVVHKICELVEKYDAVIALEDL NSGFKNSRVKVEKQVYQKF EKM LID KLNYMVD KKSN PCATGGALKGYQITN KF ESF KSM STQNG Fl FYI PAWL TSKI DPSTG FVN LLKTKYTSIADS KKFISSFDRIMYVPEEDLFEFALDYKNFSRTDADYIKKWKLYSYGNRIRIFRNPKKNNVF DWEEVCLTSAYKELFNKYG INYQQGDIRALLCEQSDKAFYSSFMALMSLMLQMRNSITGRTDVDFLISPVKNSDGIFYD SRNYEAQENAILPKNA DANG A YNIARKVLWAIGQFKKAEDEKLDKVKIAISN KEWLEYAQTSVKH Other Cas12a orthologs: SEQ ID NO: 230 Protein sequence FnCas12a WT MSIYQEFVNKYSLSKTLRFELIPQGKTLENIKARGLILDDEKRAKDYKKAKQIIDKYHQF FIEEILSSVCISEDLLQNYSDV YFKLKKSDDDNLQKDFKSAKDTIKKQISEYIKDSEKFKNLFNQNLIDAKKGQESDLILWL KQSKDNGIELFKANSDITDI DEALE 11 KS FKGWTTYFKG FH EN R KNVYSSN DIPTSI IYRIVDD N LP KFLE N KAKYESLKD KAP EAi NYE QI KKDLAE EL T FDIDYKTSEVNQRVFSLDEVFEIANFNNYLNQSGITKFNTIIGGKFVNGENTKRKGINEY INLYSQQINDKTLKKYKMS VLFKQILSDTESKSFVIDKLEDDSDVVTTMQSFYEQIAAFKTVEEKSIKETLSLLFDDLK AQKLDLSKIYFKNDKSLTDLS QQVFDDYSVIGTAVLEYITQQIAPKNLDNPSKKEQELIAKKTEKAKYLSLETIKLALEEF NKHRDIDKQCRFEEILANFA AIPMIFDEIAQNKDNLAQISIKYQNQGKKDLLQASAEDDVKAIKDLLDQTNNLLHKLKIF HISQSEDKANILDKDEHF YL VFE ECYFELANIVPL YN KIRNYITQKPYSDEKFKLN FE NSTLANGWDKNKEPDNT AILFI KDDKYYLGVM N KKN N K IFDDKAIKENKGEGYKKIVYKLLPGANKMLPKVFFSAKSIKFYNPSEDILRIRNHSTHTK NGSPQKGYEKFEFNIEDCRK FIDFYKQSISKHPEWKDFGFRFSDTQRYNSI DEFYREVENQGYKL TFEN ISESYI DSVVNQGKL YLFQIYNKDFSA YSK G RPN LHTL YWKALFDE RN LQDVVYKLNG EAELFYR KQSI P KKITH P AKEAIAN KN KD N PKKESVFEYD LI KDKRFTE D KFFFHCPITINFKSSGANKFNDEINLLLKEKANDVHILSIDRGERHLAYYTLVDGKGNII KQDTFNIIGNDRMKTNYHD KLAAIEKDRDSARKDWKKINNIKEMKEGYLSQVVHEIAKLVIEYNAIVVFEDLNFGFKRG RFKVEKQVYQKLEKMLIE KLNYLVFKDN EFDKTGGVLRA YQL TAPFETFKKMGKQTGIIYYVPAG FTSKICPVTG FVNQL YPKYESVSKSQEFFSK FDKICYNLDKGYFEFSFDYKNFGDKAAKGKWTIASFGSRLINFRNSDKNHNWDTREVYPT KELEKLLKDYSIEYGHG ECI KAAI CG ESDKKFFAKL TSVLNTI LQM RNSKTGTELDYLISPVADVNG N FFDSRQAPKN M PQDADANGAYH IGL KGLMLLGRIKNNQEGKKLNLVIKNEEYFEFVQNRNN SEQ ID NO: 240 Protein sequence MbCas12a WT M LFQDFTH L YPLSKTVR FE LKP I DRTLE HI HAKN FLSQDETMAD M H QKVKVI LDDYH RDFIADM MG EVKL TKLAEF YDVYLKFRKNPKDDELQKQLKDLQAVLRKEIVKPIGNGGKYKAGYDRLFGAKLFKDGKEL GDLAKFVIAQEGESSPK LAHLAHFEKFSTYFTGFHDNRKNMYSDEDKHTAIAYRLIHENLPRFIDNLQILTTIKQKH SALYDQIINELTASGLDVSL ASHLDGYHKLLTQEGITAYNTLLGGISGEAGSPKIQGINELINSHHNQHCHKSERIAKLR PLHKQILSDGMSVSFLPSK FADDSEMCQAVNEFYRHYADVFAKVQSLFDGFDDHQKDGIYVEHKNLNELSKQAFGDFAL LGRVLDGYYVDVVN PEFNERFAKAKTDNAKAKLTKEKDKFIKGVHSLASLEQAIEHYTARHDDESVQAGKLGQY FKHGLAGVDNPIQKIH NNHSTIKGFLERERPAGERALPKIKSGKNPEMTQLRQLKELLDNALNVAHFAKLLTTKTT LDNQDGNFYGEFGVLYD ELAKIPTLYNKVRDYLSQKPFSTEKYKLNFGNPTLLNGWDLNKEKDNFGVILQKDGCYYL ALLDKAHKKVFDNAPNT G KSIYQKM IYKYLEVRKQFPKVF FS KEAIAI NYH PS KEL VE I KD KG RQRSD DE RLKL YRF I LE CLKI H PKYD KKF EGAIG D IQLFKKDKKG REVPISE KDLFDKI NGIFSSKPKLEM EDFFIG EFKRYN PSQDL VDQYN IYKKI DSN DN RKKEN FYN NH P KFKKDL VRYYYESMCKH E EWE ESFE FSKKLQDI GCYVDVN ELFTE I ETRRLNYKISFCN I NADYI DEL VE QGQL YLFQIY NKDFSPKAHGKPNLHTLYFKALFSEDNLADPIYKLNGEAQIFYRKASLDMNETTIHRAGE VLENKNPDNPKKRQFVY D 11 KDKRYTQDKF M LHVP ITM N FGVQG MTI KE FN KKVNQSIQQYDEVNVIG I DRG E RH LL YL TVI NS KG EI LEQCSL NDITTASANGTQMTTPYHKILDKREIERLNARVGWGEIETIKELKSGYLSHVVHQISQLM LKYNAIVVLEDLNFGFKR GRFKVEKQIYQNFENALIKKLNHLVLKDKADDEIGSYKNALQLTNNFTDLKSIGKQTGFL FYVPAWNTSKIDPETGFV DLLKPRYENIAQSQAFFGKFDKICYNADKDYFEFHIDYAKFTDKAKNSRQIWTICSHGDK RYVYDKTANQNKGAAK GINVNDELKSLFARHHINEKQPNLVMDICQNNDKEFHKSLMYLLKTLLALRYSNASSDED FILSPVANDEGVFFNSA LADDTQPQNADANGA)'HIALKGLWLLNELKNSDDLNKVKLAIDNQTWLNFAQNR SEQ ID NO: 250 Protein sequence KSNKVSKELDKIQESTRKEIVKIFSSDPIYKDMLKADMISKILPEYIVDKYGDAASRIEA VKVFYGFSGYFIDFWASRKN VFSDKNIASAIPHRIVNVNARIHLDNITAFNRIAEIAGDEVAGIAEDACAYLQNMSLEDV FTGACYGEFICQKDIDRY NNICGVINQHMNQYCQNKKISRSKFKMERLHKQILCRSESGFEIPIGFQTDGEVIDAINS FSTILEEKDILDRLRTLSQE VTGYDMERIYVSSKAFESVSKYIDHKWDVIASSMYNYFSGAVRGKDDKKDAKIQTEIKKI KSCSLLDLKKLVDMYYK MDGMCLEHEATEYVAGITEILVDFNYKTFDMDDSVKMIQNEHMINEIKEYLDTYMSIYHW AKDFMIDELVDRDM E FYSE LOE IYYDLSD IVP LYN KVRNYVTQKPYSQD KIKLN FGSPTLANGWS KSKE FON NVVVLLRDE KIYLAI LNVG N K PSKDIMAGEDRRRSDTDYKKMNYYLLPGASKTLPHVFISSNAWKKSHGIPDEIMYGYNQN KHLKSSPNFDLEFCRK LIDYYKECIDSYPNYQIFNFKFAATETYNDISEFYKDVERQGYKIEWSYISEDDINQMDR DGQIYLFQIYNKDFAPNSK GMQNLHTLYLKNIFSEENLSDVVIKLNGEAELFFRKSSIQHKRGHKKGSVLVNKTYKTTE KTENGQGEIEVIESVPDQ CYLELVKYWSEGGVGQLSEEASKYKDKVSHYAATMDIVKDRRYTEDKFFIHMPITINFKA DNRNNVNEKVLKFIAEN DDLHVIGIDRGERNLLYVSVIDSRGRIVEQKSFNIVENYESSKNVIRRHDYKGKLVNKEH YRNEARKSWKEIGKIKEIK EGYLSQVIHEISKLVLKYNAIIVMEDLNYGFKRGRFKVERQVYQKFETMLINKLAYLVDK SRAVDEPGGLLKGYQLTY VPDNLGELGSQCGIIFYVPAAYTSKIDPVTGFVDVFDFKAYSNAEARLDFINKLDCIRYD ASRNKFEIAFDYGNFRTH HTTLAKTSWTIFIHGDRIKKERGSYGWKDEIIDIEARIRKLFEDTDIEYADGHNLIGDIN ELESPIQKKFVGELFDIIRFTV QLRNSKSEKYDGTEKEYD Kl ISPVM DEE GVFFTTDSYI RADGTE LP KDADAN GA YCIALKGL YDVLAVKKYW KEG EK FDRKLLAITNYNWFDFIQNRRF SEQ ID NO: 260 Protein sequence BfCas12a WT MYYESLTKLYPIKKTIRNELVPIGKTLENIKKNNILEADEDRKIAYIRVKAIMDDYHKRL INEALSGFALIDLDKAANLYL SRSKSAD DIESFSRFQDKLRKAIAKRLRE HEN FG Kl G N KD 11 PLLQKLSEN ED DYNALESFKN FYTYFESYN DVR LN L VS DKEKSSTVAYRLINENLPRFLDNIRAYDAVQKAGITSEELSSEAQDGLFLVNTFNNVLIQ DGINTYNEDIGKLNVAINL YNQKNASVQGFRKVPKMKVLYKQILSDREESFIDEFESDTELLDSLESHYANLAKYFGSN KVQLLFTALRESKGVNVY VKNDIAKTSFSNVVFGSWSRIDELINGEYDDNNNRKKDEKYYDKRQKELKKNKSYTIEKI ITLSTEDVDVIGKYIEKLES DIDDI RFKGKN FYEAVLCG H DRSKKLSKN KGAVEAI KGYLDSVKDFERDLKLINGSGQELEKNL VVYGEQEAVLSELS GIDSLYNMTRNYLTKKPFSTEKIKLNFNKPTFLDGWDYGNEEAYLGFFMIKEGNYFLAVM DANWNKEFRNIPSVDK SDCYKKVIYKQISSPEKSIQNLMVIDGKTVKKNGRKEKEGIHSGENLILEELKNTYLPKK INDIRKRRSYLNGDTFSKKD L TEFIGYYKQRVI EYYNGYSFYFKSDDDYASFKEFQEDVGRQA YQISYVDVPVSFVDDLINSGKLYLFRVYN KDFSEYS KGRLNLHTLYFKMLFDERNLKNVVYKLNGQAEVFYRPSSIKKEELIVHRAGEEIKNKNPK RAAQKPTRRLDYDIVKDR RYSQDKFMLHTSIIMNFGAEENVSFNDIVNGVLRNEDKVNVIGIDRGERNLLYVVVIDPE GKILEQRSLNCITDSNLD IETDYHRLLDEKESDRKIARRDWTTIENIKELKAGYLSQVVHIVAELVLKYNAIICLEDL NFGFKRGRQKVEKQVYQKF EKMLIDKLNYLVMDKSREQLSPEKISGALNALQLTPDFKSFKVLGKQTGIIYYVPAYLTS KIDPMTGFANLFYVKYENV DKAKEFFSKFDSIKYNKDGKNWNTKGYFEFAFDYKKFTDRAYGRVSEWTVCTVGERIIKF KNKEKNNSYDDKVIDLT NSLKELFDSYKVTYESEVDLKDAILAIDDPAFYRDLTRRLQQTLQMRNSSCDGSRDYIIS PVKNSKGEFFCSDNNDDT TPN DADANGAFN IARKG LWVLN EIRNSEEGSKI N LAMSNAQWLEYAQDNTI

Nuclear Localization Signal (NLS): SEQ ID NO: 291 Protein sequence NLS sequence 1 SEQ ID NO: 292 Protein sequence NLS sequence 2 KRPAATKKAGQAKKKK