Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
IN VIVO LENTIVIRAL GENE THERAPY FOR THE TREATMENT OF PRIMARY HYPEROXALURIA TYPE 1
Document Type and Number:
WIPO Patent Application WO/2022/229223
Kind Code:
A1
Abstract:
The present invention relates to a lentivirus vector (LV) comprising a nucleic acid, wherein the nucleic acid comprises a transcriptional unit which comprises, from 5' to 3', an hepatocyte-specific promoter, preferably a ET promoter, a nucleotide sequence encoding the optimized Alanine-Glyoxylate and Serine-Pyruvate Aminotransferase protein (AGXT- RHEAM), a woodchuck hepatitis virus posttranscriptional regulatory element (WPRE), preferably a mutated WPRE, and at least one copy of at least one miRNA target sequence, preferably miRNA-142. The lentiviral vector of the present invention further comprises the backbone elements needed for the LV to carry out its function, which are at least one of the following: a 5' long terminal repeat (5´ LTR), a primer binding site (PBS), a psi packaging signal, the Stem-loop 4 (SL4) region of wild-type HIV virus, a Rev responsive element (RRE), and a DNA flap central polypurine tract (cPPT), and a 3´ long terminal repeat (3´ LTR), or any combination thereof.

Inventors:
SEGOVIA SANZ JOSÉ CARLOS (ES)
GARCÍA BRAVO MARÍA (ES)
MOLINOS VICENTE ANDREA (ES)
GARCÍA TORRALBA AIDA (ES)
NIETO ROMERO VIRGINIA (ES)
Application Number:
PCT/EP2022/061107
Publication Date:
November 03, 2022
Filing Date:
April 26, 2022
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
CENTRO DE INVESTIG ENERGETICAS MEDIOAMBIENTALES Y TECNOLOGICAS O A M P CIEMAT (ES)
CONSORCIO CENTRO DE INVESTIG BIOMEDICA EN RED (ES)
FUNDACION INSTITUTO DE INVESTIG SANITARIA FUNDACION JIMENEZ DIAZ FIIS FJD (ES)
International Classes:
C12N15/867; A61K48/00; A61P1/16; C12N9/10
Domestic Patent References:
WO2019152692A12019-08-08
WO2018106724A12018-06-14
Foreign References:
EP2384200A12011-11-09
Other References:
ESTÈVE JULIE ET AL: "Generation of induced pluripotent stem cells-derived hepatocyte-like cells forex vivogene therapy of primary hyperoxaluria type 1", STEM CELL RESEARCH, vol. 38, 101467, 21 May 2019 (2019-05-21), XP085726998, ISSN: 1873-5061, DOI: 10.1016/J.SCR.2019.101467
KUKREJA ANJLI ET AL: "Systemic Alanine Glyoxylate Aminotransferase mRNA Improves Glyoxylate Metabolism in a Mouse Model of Primary Hyperoxaluria Type 1", vol. 29, no. 2, 1 April 2019 (2019-04-01), US, pages 104 - 113, XP055849763, ISSN: 2159-3337, Retrieved from the Internet DOI: 10.1089/nat.2018.0740
MESA-TORRES NOEL: "The consensus-based approach for gene/enzyme replacement therapies and crystallization strategies: the case of human alanine-glyoxylate aminotransferase", BIOCHEMICAL JOURNAL, vol. 462, no. Part 3, 15 September 2014 (2014-09-15), pages 453 - 463, XP009530546, DOI: 10.1042/BJ20140250
MOLINOS-VICENTE ANDREA ET AL: "New Gene Therapy Approach for the Treatment of Primary Hyperoxaluria Type 1 (PH1) Patients. Proof of Concept in a Mouse Model of PH1", MOLECULAR THERAPY, vol. 29, no. 4 Suppl. 1, 27 April 2021 (2021-04-27), pages 246, XP055944354
ALTSCHUL ET AL., J MOL BIOL., vol. 215, no. 3, 1990, pages 403 - 10
AGUDO ET AL., MOLECULAR THERAPY, vol. 20, no. 12, 2012, pages 2257 - 2267
Attorney, Agent or Firm:
HOFFMANN EITLE S.L.U. (ES)
Download PDF:
Claims:
CLAIMS

1. A lentiviral vector comprising a nucleic acid, wherein the nucleic acid comprises the following: an engineered hepatocyte-specific promoter with at least 98% sequence identity over the full length of SEQ ID NO: 16, a nucleotide sequence encoding for an optimized Alanine-Glyoxylate and Serine- Pyruvate Aminotransferase (AGXT-RHEAM), wherein the AGXT-RHEAM protein has at least 98% amino acid sequence identity over the full length of SEQ ID NO: 52, a mutated optimized woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) with at least 98% sequence identity over the full length of SEQ ID NO: 21, and at least a copy of at least one miRNA target sequence, wherein the miRNA target sequence is selected from the group consisting of miRNA-142-3, miR-181, miR-223, or miR-30b, wherein the engineered hepatocyte-specific promoter, the WPRE and the at least a copy of at least one miRNA target sequence are operably linked to and regulate the expression of the AGXT-RHEAM protein.

2. The lentiviral vector according to claim 1, wherein the amino acid sequence of the Alanine- Glyoxylate and Serine-Pyruvate Aminotransferase (AGXT-RHEAM) consists of SEQ ID NO: 52.

3. The lentiviral vector according to claim 1 or 2, wherein the nucleotide sequence of the engineered hepatocyte-specific promoter consists of SEQ ID NO: 16.

4. The lentiviral vector according to any of claims 1 to 3, wherein the nucleotide sequence of the mutated optimized woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) consists of SEQ ID NO: 21.

5. The lentiviral vector according to any of claims 1 to 4, wherein the nucleotide sequence encoding for the optimized Alanine-Glyoxylate and Serine-Pyruvate Aminotransferase (AGXT-RHEAM) protein consists of SEQ ID NO: 29.

6. The lentiviral vector according to any of claims 1 to 5, wherein the at least a copy of at least one miRNA target sequence consists of four copies of the target sequence of miRNA- 142-3 with at least 95%, preferably 98%, sequence identity over the full length of SEQ ID NO 22.

7. The lentiviral vector according to any of claims 1 to 6, wherein the nucleic acid further comprises the following: a) a 5’ long terminal repeat (LTR), b) a primer binding site (PBS) c) a psi packaging signal, d) the Stem-loop 4 (SL4) region of wild-type HIV virus, e) a Rev responsive element (RRE), f) a DNA flap central polypurine tract (cPPT), and g) a 3’ long terminal repeat (LTR).

8. The lentiviral vector according to any of claims 1 to 7, wherein the nucleotide comprised in said lentiviral vector further comprises: h) a DenvRFI region located between d) and e), and i) a DenvRF2 region located between e) and f).

9. The lentiviral vector according to any of claims 1 to 8, wherein the full-length nucleotide of said lentiviral has at least 95%, preferably 98% sequence identity with SEQ ID NO 30 (LV. ET. AGXT-RH EAM .142-3pT) .

10. The lentiviral vector according to claim 9, wherein the sequence identity with SEQ ID NO 30 is 100%.

11. A substantially pure population of cells transduced with the lentiviral vector as defined in any of claims 1 to 10.

12. A pharmaceutical composition comprising the lentiviral vector as defined in any of claims 1 to 10, or the substantially pure population of cells as defined in claim 11, and a pharmaceutically acceptable carrier or diluent.

13. The lentiviral vector according to any of claims 1 to 10, the substantially pure population of cells according to claim 11, or the pharmaceutical composition according to claim 12, for use as a medicament.

14. The lentiviral vector according to any of claims 1 to 10, the substantially pure population of cells according to claim 11, or the pharmaceutical composition according to claim 12, for use in the treatment of Primary Hyperoxaluria in a subject in need thereof, the method comprising administering to the subject said lentiviral vector, said population of cells, or said pharmaceutical composition.

15. The lentiviral vector, the substantially pure population of cells, or the pharmaceutical composition for use according to claim 14, wherein the Primary Hyperoxaluria is type 1. 16. The lentiviral vector, the substantially pure population of cells, or the pharmaceutical composition, for use according to any of claims 13 to 15, wherein the use is in combination with an immune response inhibitor, preferably dexamethasone, ciclosporin A, ciclosporin H and/or rapamycin, or any combination thereof.

Description:
IN VIVO LENTIVIRAL GENE THERAPY FOR THE TREATMENT OF PRIMARY HYPEROXALURIA TYPE 1

TECHNICAL FIELD OF THE INVENTION

The present invention can be included in the field of gene therapy, particularly in the treatment of Primary Hyperoxaluria type 1. Specifically, the present invention relates to a lentiviral vector for gene therapy, capable of treating Primary Hyperoxaluria type 1.

BACKGROUND OF THE INVENTION

Primary Hyperoxaluria Type 1 (PH1) (OMIM#259900) is an inherited rare autosomal recessive metabolic disease. It is caused by a deficiency in the alanine:glyoxylate aminotransferase (AGT; EC 2.6.1.44) enzyme, encoded by the AGXT gene. AGT activity is limited to hepatic peroxisomes in humans. This enzyme is only active intracellularly and is not secreted into the extracellular space. PH1 is characterized by an overproduction of oxalate in the liver. Oxalate is an end product of metabolism in mammals that is normally excreted by the urine. When produced in large amounts, the kidney is the first organ affected due to the aggregates of calcium oxalate (CaOx) that are formed in the urinary space (urolithiasis) and renal tissue (nephrocalcinosis), causing interstitial fibrosis and renal failure. As a result of renal damage, the glomerular filtration rate (GFR) decreases and chronic kidney disease rises. Ultimately, around 60% of PH1 patients progress to end-stage renal disease by the age of 40. Subsequent systemic involvement, with widespread CaOx precipitation, called oxalosis, put life at risk for PH1 patients (Cochat et al., 2012; Sas et al., 2020). Current guidelines for patients with PH1 given by the European Consortium of Hyperoxaluria (http://www.oxaleurope.com/) recommend preventive organ transplantation when renal function is already damaged and before systemic oxalosis appears. Thus, there is a need for new therapeutic approaches that, ideally, could be applied as soon as possible, just after the disease is diagnosed. Because of the absence of an accurate test to predict the decline in renal function and the available genetic diagnosis, new treatments should be introduced at an early stage of the disease aiming to reduce oxalate levels and to preserve kidney function.

Gene therapy has the potential to be curative with one-time dosing, by restoring AGT activity through delivery of a functional copy of the AGXT gene. Currently, gene therapy for inherited metabolic liver diseases is mostly based on in vivo approaches. Liver sinusoidal endothelial cells have a fenestrated structure, facilitating access and gene transfer to hepatocytes (Piccolo & Brunetti-Pierri, 2015). AAV vectors are widely adopted for in vivo liver-directed gene therapy, given their remarkable safety and efficacy profile shown in pre-clinical models and clinical trials. AAV-vector based gene transfer to the liver of clotting factors for the treatment of the coagulation disorder hemophilia is among the most successful applications of gene therapy (George et al. , 2017; Pasi et al. , 2020). In the particular case of PH1, because is an autonomous-cell disease in which the functional cells can not compensate the oxalate overproduction from the deficient ones, the percentage of corrected cells that should be achieved after therapy is thought to be high, in order to revert hyperoxaluric phenotype and correct the disease (Danpure, 2009). Preclinical proof of concept of gene therapy efficacy has been provided using adenoviral or adenoassociated viral vectors in PH1 mice (Salido et al., 2006; Salido et al., 2011). Despite this success, the non-integrating nature of AAV vectors challenges their application to paediatric patients, as the transgene would be diluted during liver growth and the immune response currently hampers effective re administration. Moreover, the widespread pre-existing immunity against AAV capsid reduces the fraction of eligible patients. Since around 30% of PH1 patients have an infantile onset (Sas et al., 2019; Mandrile et al., 2014) it becomes crucial to develop a gene therapy strategy with the potential to persist life-long following a single administration to paediatric patients.

Lentiviral vectors (LV) are attractive vehicles for liver gene therapy, by virtue of their ability to stably integrate in the genome of target cells and the low prevalence of pre-existing immunity against vector components in humans. In the last years, the use of a LV harbouring FIX for the treatment of Haemophilia B has shown promising results in mice, dogs and non-human primates (Annoni et al., 2013; Cantore et al., 2015; Milani et al., 2019). In the present invention, LV based on the vector used for the treatment of Haemophilia B will be used. These LV are designed to target transgene expression to hepatocytes by a combination of transcriptional (synthetic liver-specific promoter) and post-transcriptional control (microRNAs 142 target sequences, specific for hematopoietic cells, responsible for the degradation of any mRNA from the vector that are ectopically expressed in antigen presenting cells in liver and spleen) (Annoni et al., 2013). In addition, the proposed LV includes an enhanced version of AGXT that confers a higher stability to the protein (Mesa-Torres et al., 2014).

DESCRIPTION OF THE FIGURES

Figure 1. Lentiviral vector constructs. A. Reporter lentiviral vector: LV.ET.eGFP.142-3pT. The developed reporter lentiviral vector expresses an eGFP gene which codifies for Green Fluorescence Protein (GFP). Hepatocyte-specific expression of this transgene is achieved due to the presence of two different transcriptional control elements, a specific hepatic promoter (TTR) composed of a synthetic enhancer, a murine TTR enhancer and a part of the murine TTR promoter, and four target sequences of a hematopoietic specific miRNA-142-3 to block the transgene expression in Antigen Presenting Cells (APCs) and, therefore, the development of an immune response. B. Therapeutic lentiviral vector: LV.ET.AGXT- RHEAM.142-3pT. It was developed from the same lentiviral vector structure, as the reporter lentiviral vector, and expresses an improved version of the AGXT gene, AGXT-RHEAM, with five amino acid changes, which codifies for a more active form of AGT protein As well as in reporter LV, hepatocyte-specific expression of the therapeutic transgene is achieved due to the presence of two different transcriptional control elements, a specific hepatic promoter (TTR) and four repetitions of a target sequence of a hematopoietic specific miRNA-142-3.

Figure 2. In vivo testing protocol of the gene therapy approach efficacy study. To study the efficacy of the proposed strategy two kinds of experiments were performed. A. C57BL/6 adult male mice were intravenously injected with a single dose of reporter lentiviral vector and were sacrificed four weeks later. Afterwards, the number of genome copies of the lentiviral vector integrated, as well as the percentage of transduced cells were analysed in the target tissue, the liver, to determine the transduction efficacy. Moreover, a biodistribution study was performed to determine the security of the procedure, by analysis of vector copy number (VCN) in spleen, lungs, bone marrow, lymph nodes, thymus, brain, testicles, pancreas, and kidneys. B. For the analysis of the AGXT-expressing lentiviral vector therapeutic effect in a mouse model of PH1 the following protocol was performed. Adult Agxtt 1 male mice were intravenously injected with different doses of the therapeutic lentiviral vector. Moreover, three different control groups were included, two of them composed of Agxtt 1 mice non-injected or injected with the reporter lentiviral vector, and the other one composed of non-injected C57BL/6 wild-type mice. Three weeks after the treatment, all mice groups were subjected to an ethylene glycol challenge, in which they were supplemented with 0.5% ethylene glycol in drinking water, to induce overloading in oxalate production. During the challenge, urine oxalate concentration and weight, two different PH1 end-points, were measured. After the treatment, mice were sacrificed, and kidney damage was defined. After the sacrifice, the transduction efficacy of the therapeutic lentiviral vector was analyzed by VCN and AGXT expression in liver. In reporter lentiviral vector injected Agxtt 1 mice percentage of transduced cells was also analysed to complete the results obtained with the therapeutic lentiviral vector.

Figure 3. Number of viral genomes integrated into the target tissue of lentiviral vector injected mice. Integration of lentiviral vector in the liver, the target tissue, was analysed four weeks after the lentiviral vector injection by quantification of VCN by qPCR. Data from experiments performed in C57BL/6 mice as well as in Agxtt 1 are represented. Non-injected control mice (n=3) did not show lentiviral vector integration in any mouse strains. Regarding experiments performed in C57BL/6 mice, two different reporter lentiviral vector doses (T10 8 TU/mouse and 2.5- 10 8 TU/mouse) (n=3) were tested. We observed 0.55 (0.34-0.56) lentiviral vector copies per diploid genome in the lower one, whereas in the high-dose 1.03 (0.94-1.56) lentiviral vector copies per diploid genome were obtained. Furthermore, Agxtt 1 mice were intravenously injected with two different doses of LV.ET.eGFP.142-3pT resulting in a VCN of 0.11 (0.09-0.49) in the liver of mice injected with 1.4- 10 s TU/mouse (n=4) and 0.84 (0.73- 1.58) in mice injected with 5- 10 8 TU/mouse (n=5); and with three different doses of LV.ET.AGXT-RHEAM.142-3pT (2.5- 10 8 TU/mouse (n=6); 5- 10 8 TU/mouse (n=10) and 1 10 9 TU/mouse (n=6)) resulting in a VCN of 0.615 (0.39-1.46), 0.765 (0.45-1.19) and 1.165 (0.85- 0.32) respectively.

Figure 4. AGXT expression analysis in the liver of Agxtt 1 injected mice after the in vivo lentiviral vector treatment. Comparison with physiological mouse Agxt expression in C57BL/6 wild-type mice. AGXT expression in therapeutic lentiviral vector injected Agxtt 1 mice was analysed by RT-qPCR. Representation of AGXT expression representation was referred to a housekeeping gene. Two different housekeeping genes, with different expression levels were used for this analysis ( msTbp , low level of expression, and msActb, high level of expression) to determine differences between a physiological Agxt expression in a wild-type mouse (higher expression level) compared with AGXT expression induced by lentiviral vector integration after the in vivo gene therapy strategy (lower expression level). A. AGXT expression referred to msTbp expression in the liver of LV.ET.AGXT-RHEAM.142-3pT injected Agxtt 1 mice and non-injected wild-type mice. Therapeutic lentiviral injected mice received a dose of 2.5- 10 8 TU/mouse (n=6); 5- 10 8 TU/mouse (n=10) and T 10 9 TU/mouse (n=6), respectively, resulting in an AGXT expression of 10.88 (5.62-16.79), 12.11 (4.38-14.48) and 19.88 (18.52-28.55), expressed as % of msTbp expression, which correlates with the received dose. Non-injected Agxtt 1 mice (n=8) did not show AGXT expression. Moreover, wild-type C57BL/6 mice (n=4) showed an Agxt expression of 8563 % of msTbp expression (6996-8939), significantly higher than the one achieved in Agxtt 1 treated mice. B. AGXT expression referred to ms Actb expression in the liver of LV.ET.AGXT-RHEAM.142-3pT injected Agxtt 1 mice and non-injected wild-type mice. AGXT expression, expressed as % of msActb housekeeping gene expression, was of 0.1041 (0.06007-0.2452) in Agxtt 1 mice which received a therapeutic lentiviral vector dose of 2.5-10 8 TU/mouse (n=6); 0.1857 (0.08878-0.2966) in mice injected with 5- 10 8 TU/mouse (n=10) and 0.3088 (0.1707-0.4418) in mice injected with T10 9 TU/mouse (n=6). Agxt physiological expression referred to ms Actb was 340.5 % (141-374.5). Although the differences in Agxt expression levels depending on the housekeeping gene, differences among groups were maintained. Figure 5. Percentage of transduced cells in reporter lentiviral vector C57BL/6 and Agxtt 1 injected mice. Percentage of eGFP expressing hepatocytes in liver sections of reporter lentiviral vector injected wild-type C57BL/6 and Agxtt 1 mice was defined by immunostaining and following image analysis by QuPath ® software. A-E. Representative images of immunostaining for eGFP in liver sections of all mice groups. Green: eGFP; blue: nuclei stained with DAPI. F-H. Representative images of double immunostaining of eGFP and mouse Albumin, a hepatocyte-specific marker, in liver sections of C57BL/6 mice. Red: eGFP: green: mouse Albumin ; blue: nuclei stained with DAPI. I. Percentage of transduced hepatocytes. Regarding experiments performed in C57BL/6 mice, two different reporter lentiviral vector doses (1 10 8 TU/mouse (n=3) and 2.5- 10 8 TU/mouse (n=3)) were tested. This corresponds to 1.751 (1.132-5.556) % positive hepatocytes and 12.92 (10.72-13.81) % positive hepatocytes respectively. Furthermore, in Agxtt 1 two different doses were tested (1.4-10 s TU/mouse (n=4) and 5-10 8 TU/mouse (n=5)) resulting in 1.777 (0.399-4.572) % eGFP positive hepatocytes and 5.5059 (2.109-8.754) % eGFP positive hepatocytes respectively.

Figure 6. Biodistribution analysis. To determine the safety of the proposed therapeutic strategy, the number of lentiviral vector integrated genomes was analysed in C57BL/6 wild type mice injected with two different doses of the reporter lentiviral vector. The number of LV genome copies integrated into different tissues of injected wild-type C57BL/6 mice was analysed by qPCR. Integration of reporter lentiviral vector genome was only observed in liver, the target tissue, and in lungs, spleen, and bone marrow. C57BL/6 mice injected with T10 8 TU/mouse (n=3) showed off-target transduction with a VCN value of 2.72 (2.33-6.2) in lungs, 0.21 (0.14-0.32) in spleen and 0.05 (0.04-0.09) in bone marrow; whereas mice injected with a higher dose of 2.5- 10 8 TU/mouse (n=3) showed a VCN value of 4.94 (0.185- 7.79) in lungs, 0.35 (0.25-0.55) in spleen and 0.13 (0.09-0.18) in bone marrow.

Figure 7. PH1 end-points. Oxalate urine concentration during the ethylene glycol challenge. Quantification of urine oxalate (pmol/24 h) at a basal time point and on days 3 and 7 of ethylene glycol challenge. All represented mice were subjected to a 7-days ethylene glycol challenge in which 0.5% of ethylene glycol was added into the drinking water. Two different negative control groups are represented, one composed of Agxtt 1 non-injected mice (n=8) and the other one composed of Agxtt 1 mice injected at two different doses of LV.ET.GFP.142-3pT (n=9) (1.4-10 8 TU/mouse and 5-10 8 TU/mouse). It is also included a positive control group composed of wild-type C57BL/6 non-injected mice (n=5). Therapeutic lentiviral vector injected Agxtt 1 mice are divided into three different groups according to the dose received (2.5-10 8 (n=6), 5-10 8 (n=10) or 10- 10 s (n=6) TU/mouse). Basal data shows a higher oxalate concentration in treated and non-treated Agxtt 1 mice compared with wild-type ones. During the ethylene glycol challenge an increase in urine oxalate levels was observed in all groups, however, this increase was higher in the case of untreated Agxtt 1 mice (non- injected and LV.ET.eGFP.142-3pT injected Agxt1 /_ mice) compared with Agxtt 1 treated mice at different doses of LV.ET.AGXT-RHEAM.142-3pT (2.5-10 8 , 5-10 8 or 10-10 8 TU/mouse) and wild-type C57BL/6 mice group. This difference was much higher in the case of Agxtt 1 mice which received 5- 10 8 TU/mouse of LV.ET.AGXT-RHEAM.142-3pT at day 7 of the ethylene glycol challenge compared with untreated groups. Moreover, at day 7 of ethylene glycol treatment non-significant differences were observed between Agxtt 1 treated with 5- 10 8 TU/mouse of therapeutic LV and wild-type C57BL/6 mice.

Figure 8. PH1 end-points: Weight evolution during ethylene glycol challenge. Weight progress was analysed during the ethylene glycol challenge. Animal weight was measured at a basal time point before the challenge and on days 3 and 7 of the challenge. In this figure, the gain or loss in weight of each animal to the basal weight is represented. Two different negative control groups are represented, one composed of Agxtt 1 non-injected mice and the other one composed of Agxtt 1 mice injected at two different doses of LV.ET.GFP.142-3pT (1.4-10 8 TU/mouse and 5- 10 8 TU/mouse). It is also included a positive control group composed of wild-type C57BL/6 non-injected mice. Therapeutic lentiviral vector injected Agxtt 1 mice are divided into three different groups according to the dose received (2.5- 10 8 , 5-10 8 or 10- 10 s TU/mouse). In the case of therapeutic lentiviral vector treated mice, there was not observed a decrease in body weight and an increase was observed on day 7 of the challenge whereas in the case of untreated mice (the non-injected or reporter lentiviral vector injected Agxtt 1 mice) they experienced a weight loss during the challenge. Additionally, non significant differences were observed between wild-type C57BL/6 mice and Agxtt 1 mice injected with the therapeutic lentiviral vector at a dose of 5- 10 8 TU/mouse or 10- 10 8 TU/mouse, meanwhile significant differences were observed among these groups and non- treated mice.

Figure 9. PH1 end-points. Kidney damage Score. Kidney damage development after ethylene glycol challenge. To determine kidney damage in treated and non-treated mice subjected to ethylene glycol challenge, kidney histological samples were classified according to the “kidney damage score” as 0: no damage; 1 : normal architecture, but the presence of some damage signs such as dilated ducts; 2: architecture partially affected and an increase in the presence of dilated ducts; 3: tissue architecture highly affected and the presence of calcium oxalate crystals can be observed, indicating a nephrocalcinosis stage. A. Representative images of haematoxylin-eosin kidney sections of each mice group. From each group, there is a representative image of samples with a kidney damage score of 0-1 , which indicates none or mild kidney damage, and a representative image of samples with a kidney damage score of 2-3, which indicates moderate or severe kidney damage. The number of mice that developed that level of kidney damage to the total number of mice in each group is specified in each image. According to the different mice groups, two different negative control groups are represented, one composed of Agxtt 1 non-injected mice (n=8) and the other one composed of Agxtt 1 mice injected at two different doses of LV.ET.GFP.142-3pT (1.4-10 8 TU/mouse and 5- 10 8 TU/mouse) (n=9). It is also included a positive control group composed of wild-type C57BL/6 non-injected mice (n=5). Therapeutic lentiviral vector injected Agxtt 1 mice are divided into three different groups according to the dose received (2.5- 10 8 TU/mouse (n=6), 5-10 8 TU/mouse (n=10) or 10- 10 s TU/mouse (n=6)). B. Representation of kidney damage stage of each animal. Grey band represents the development of nephrocalcinosis. Among different treated Agxtt 1 mice, none of the mice injected with the middle dose of therapeutic lentiviral vector developed nephrocalcinosis.

Figure 10. Transduction enhancers: Cyclosporin H increases reporter lentiviral vector transduction in HepG2 cell line. HepG2 cells were transduced with different administration regimens of LV.ET.eGFP.142-3pT and cyclosporine H (CsH) combinations. Fold increase of the percentage of eGFP positive cells between the different CsH treated conditions compared to reporter lentiviral vector transduced cells without CsH is represented. In the first transduction protocol, HepG2 cells were subjected to different concentrations of CsH (4, 8 or, 16 mM) during lentiviral vector transduction. Cells were transduced with a lentiviral vector MOI of 0.3. Three technical replicates of each condition were performed. Basal transduction levels were 36.67% and in the different CsH treated samples a transduction percentage of 37.67%, 44.3% and, 51.5% in 4, 8 and, 16 mM CsH conditions were observed, which means a fold-increase of 1.03, 1.2 and, 1.4 respectively. In addition, another CsH administration regimen was tested. HepG2 cells were pre-treated with CsH for 16 hours before transduction, and CsH was added together with LV.ET.eGFP.142-3pT during lentiviral transduction. The only CsH concentration tested was 16 mM. In this CsH administration regimen, a fold-increase in eGFP positive cells of 2.35 to the control group was observed. Cells were transduced with a MOI of 0.3 of LV.ET.eGFP.142-3pT and the percentage of transduced cells achieved were 13.2% in the control group and 31.03% in the CsH treated cells. The differences observed in transduction levels at the same MOI are probably due to different viral stocks used.

Figure 11. LV.ET.eGFP.142-3pT construct. Figure 12. LV.ET.AGXT-RHEAM.142-3pT construct.

Figure 13. Comparison of LV.ET.eGFP.142-3pT transduction efficacy in vitro and in vivo. For in vitro transduction efficacy determination, HepG2 cells were transduced with LV.ET.eGFP.142-3pT at different Multiplicity of Infection (MOI). Three days after transduction, the percentage of eGFP positive cells was determined by flow cytometry. Using MOIs of about 1, we achieved a transduction percentage of 50%, whereas higher MOIs achieved 100% transduction. For in vivo transduction efficacy determination, adult C57BL/6 (filled circle) or Agxtt 1 mice (open circle) were intravenously injected with LV. ET. eGFP.142- 3pT at different doses (T10 8 TU/mouse, 1.4- 10 s TU/mouse, 2-10 8 TU/mouse and 5-10 8 TU/mouse). Corresponding MOIs were calculated only according to the estimated number of hepatocytes in the liver of these mice as previously described (Park et al., 2000; Schmitt et al., 2010). Therefore, the mice were injected with the following MOIs: 0.8, 1.1, 2, and 4. Percentage of transduced hepatocytes was determined 4 weeks later in liver histology samples.

Figure 14. Dexamethasone treatment improves the transduction efficacy in the liver of in vivo reporter lentiviral vector treated C57BL/6 mice

To test the use of dexamethasone as a transduction enhancer in the context of the proposed strategy an in vivo experiment was performed. A. C57BL/6 adult male mice were intravenously injected with a single dose of LV.ET.eGFP.142-3pT (5-10 8 TU/mouse) by intravenous injection in combination or not with dexamethasone (Fortecortin®). Dexamethasone was administered by intraperitoneal injection at a dose of 5 mg/kg body weight in three consecutive administrations: 12 hours and 2 hours before lentiviral vector injection and 4 hours after. Four weeks later mice were sacrificed. Afterwards, the number of genome copies of the lentiviral vector integrated, as well as the percentage of transduced cells were analysed in the target tissue, the liver, to determine the transduction efficacy. B. Integration of the lentiviral vector in the liver was analysed by quantification of VCN by qPCR. Two different mice-treated groups are represented, one that received the treatment only with the lentiviral vector (n=5) and another one that received the combined treatment (n=5). In mice treated only with the reporter lentiviral vector, we observed 1.48 (1.2-1.76) copies per diploid genome whereas in mice also treated with dexamethasone 0.92 (0.38-1.4) copies per diploid genome were detected, a significant reduction (p<0.05) compared with mice treated only with the lentiviral vector. C. Percentage of eGFP positive hepatocytes in liver sections was defined by immunostaining and following image analysis by QuPath® software. In mice treated only with the reporter lentiviral vector we observed 4.434 (2.167-7.55) % eGFP positive hepatocytes, whereas in mice treated with the combination of the lentiviral vector and dexamethasone we obtained 18.11 (6.381-19.79) % eGFP positive hepatocytes, a significant increase (p<0.05) with respect to the mice treated only with the lentiviral vector. D- E. Representative images of immunostaining for eGFP in liver sections of all mice groups.

DESCRIPTION OF THE INVENTION

In a first aspect, the present invention provides a lentivirus vector (LV) comprising a nucleic acid, wherein the nucleic acid comprises a transcriptional unit, wherein the transcriptional unit is defined herein as comprising, from 5’ to 3’, an hepatocyte-specific promoter, preferably a ET promoter, a nucleotide sequence encoding optimized Alanine-Glyoxylate and Serine-Pyruvate Aminotransferase (hereinafter referred to as AGXT-RHEAM) protein, a woodchuck hepatitis virus posttranscriptional regulatory element (hereinafter referred to as WPRE), preferably a mutated WPRE, and at least one copy of at least one miRNA target sequence; wherein the hepatocyte-specific promoter, the WPRE and the at least one copy of at least one miRNA target sequence are operably linked to and regulate the expression of AGXT-RHEAM.

Preferably, the nucleic acid comprises the following: an engineered hepatocyte-specific promoter with at least 98% sequence identity over the full length of SEQ ID NO: 16, a nucleotide sequence encoding for an optimized Alanine-Glyoxylate and Serine- Pyruvate Aminotransferase (AGXT-RHEAM), wherein the AGXT-RHEAM protein has at least 98% amino acid sequence identity over the full length of SEQ ID NO: 52, a mutated optimized woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) with at least 98% sequence identity over the full length of SEQ ID NO: 21, and at least a copy of at least one miRNA target sequence, wherein the miRNA target sequence is selected from the group consisting of miRNA-142-3, miR-181, miR-223, or miR-30b, wherein the engineered hepatocyte-specific promoter, the WPRE and the at least a copy of at least one miRNA target sequence are operably linked to and regulate the expression of the AGXT-RHEAM protein. Preferably, the amino acid sequence of the Alanine-Glyoxylate and Serine-Pyruvate Aminotransferase (AGXT-RHEAM) consists of SEQ ID NO: 52. Preferably, the nucleotide sequence of the engineered hepatocyte-specific promoter consists of SEQ ID NO: 16. Preferably, the nucleotide sequence of the mutated optimized woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) consists of SEQ ID NO: 21. Preferably, the nucleotide sequence encoding for the optimized Alanine-Glyoxylate and Serine-Pyruvate Aminotransferase (AGXT-RHEAM) protein consists of SEQ ID NO: 29. Preferably the at least a copy of at least one miRNA target sequence consists of four copies of the target sequence of miRNA-142-3 with at least 95%, preferably 98%, sequence identity over the full length of SEQ ID NO 22.

In a preferred embodiment, the nucleic acid comprised in the lentiviral vector further comprises, preferably from 5’ to 3’, the following: a) a 5’ long terminal repeat (LTR), b) a primer binding site (PBS) c) a psi packaging signal, d) the Stem-loop 4 (SL4) region of wild-type HIV virus, e) a Rev responsive element (RRE), f) a DNA flap central polypurine tract (cPPT), and g) a 3’ long terminal repeat (LTR), wherein, preferably, the nucleotide further comprises a DenvRFI region located between d) and e), and a DenvRF2 region located between e) and f).

In a preferred embodiment of the first aspect or any of its embodiments, the lentiviral vector comprises a nucleic acid, wherein the nucleic acid comprises from 5’ to 3’ the following nucleotides: a) a 5’ long terminal repeat (LTR), b) a primer binding site (PBS) c) a psi packaging signal, d) the Stem-loop 4 (SL4) region of wild-type HIV virus, e) a Rev responsive element (RRE), f) a DNA flap central polypurine tract (cPPT), g) an engineered hepatocyte-specific promoter with at least 95%, preferably 98%, sequence identity over the full length of SEQ ID NO 16, h) a nucleotide sequence encoding optimized Alanine-Glyoxylate and Serine- Pyruvate Aminotransferase (AGXT-RHEAM) protein with at least 95%, preferably 98%, sequence identity over the full length of SEQ ID NO 29, i) a mutated optimized woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) with at least 95%, preferably 98%, sequence identity over the full length of SEQ ID NO 21, j) at least a copy of at least one miRNA target sequence, and k) a 3’ long terminal repeat (LTR), wherein the long terminal repeat regions of a) and k) have been rendered substantially transcriptionally inactive by virtue of total or partial deletion in the U3 region of the LTR, and wherein g), i) and j) are operably linked to and regulate the expression of h). In a preferred embodiment of the first aspect or any of its embodiments, the sequence identity of g), h), i) to SEQ ID NOs 16, 29, and 21, respectively, is 100%.

In a preferred embodiment of the first aspect or any of its embodiments, the at least a copy of at least one miRNA target sequence of j) consists of four copies of the target sequence of miRNA-142-3 with at least 95%, preferably 98%, most preferably 100%, sequence identity over the full length of SEQ ID NO 22.

In a preferred embodiment of the first aspect or any of its embodiments, the nucleotide comprised in said lentiviral vector further comprises:

L) a DenvRFI region located between d) and e), and m) a DenvRF2 region located between e) and f).

In a preferred embodiment of the first aspect or any of its embodiments, the 5 ' long terminal repeat comprised in the nucleotide of said lentiviral vector has at least 95%, preferably 98%, most preferably 100%, sequence identity over the full length of SEQ ID NO 4.

In a preferred embodiment of the first aspect or any of its embodiments, the primer binding site comprised in the nucleotide of said lentiviral vector has at least 95%, preferably 98%, most preferably 100%, sequence identity over the full length of SEQ ID NO 9.

In a preferred embodiment of the first aspect or any of its embodiments the psi packaging signal comprised in the nucleotide of said lentiviral vector has at least 95%, preferably 98%, most preferably 100%, sequence identity over the full length of SEQ ID NO 10.

In a preferred embodiment of the first aspect or any of its embodiments, the Stem-loop 4 (SL4) comprised in nucleotide of said lentiviral vector has at least 95%, preferably 98%, most preferably 100%, sequence identity over the full length of SEQ ID NO 11.

In a preferred embodiment of the first aspect or any of its embodiments, the Rev responsive element (RRE) comprised in the nucleotide of said lentiviral vector has at least 95%, preferably 98%, most preferably 100%, sequence identity over the full length of SEQ ID NO 13.

In a preferred embodiment of the first aspect or any of its embodiments, the DNA flap central polypurine tract (cPPT) comprised in the nucleotide of said lentiviral vector has at least 95%, preferably 98%, most preferably 100%, sequence identity over the full length of SEQ ID NO 15.

In a preferred embodiment of the first aspect or any of its embodiments, the 3 ' long terminal repeat comprised in the nucleotide of said lentiviral vector has at least 95%, preferably 98%, most preferably 100%, sequence identity over the full length of SEQ ID NO 23.

In a preferred embodiment of the first aspect or any of its embodiments, the DenvRFI comprised in the nucleotide of said lentiviral vector has at least 95%, preferably 98%, most preferably 100%, sequence identity over the full length of SEQ ID NO 12.

In a preferred embodiment of the first aspect or any of its embodiments, the DenvRF2 comprised in the nucleotide of said lentiviral vector has at least 95%, preferably 98%, most preferably 100%, sequence identity over the full length of SEQ ID NO 14.

In a preferred embodiment of the first aspect or any of its embodiments, the full-length nucleotide of said lentiviral has at least 95%, preferably 98%, most preferably 100% sequence identity with SEQ ID NO 30 (LV.ET.AGXT-RHEAM.142-3pT).

In a second aspect, the present invention relates to a host cell or a substantially pure population of cells comprising (e.g., transduced) the lentiviral vector as defined in the first aspect or in any of its embodiments. In an embodiment of the second aspect, the host cell or the substantially pure population of cells are hepatic cells.

In a third aspect, the present invention relates to an isolated nucleic acid comprising the nucleotide comprised in the lentiviral vector as defined in the first aspect or any of its embodiments.

In a fourth aspect, the present invention provides a pharmaceutical composition comprising the lentiviral as defined in the first aspect or in any of its embodiments, the host cell or the substantially pure population of cells according to the second aspect or any of its embodiments, and/or the nucleic acid as defined in the third aspect or any of its embodiments and a pharmaceutically acceptable carrier or diluent.

In a fifth aspect, the present invention provides the lentiviral vector according to the first aspect or any of its embodiment, the host cell or the substantially pure population of cells according to the second aspect or any of its embodiments, the isolated nucleic acid according to the third aspect or any of its embodiments, or the pharmaceutical composition of the fourth aspect or any of its embodiments for use as a medicament.

In a sixth aspect, the present invention provides the lentiviral vector according to the first aspect or any of its embodiment, the host cell or the substantially pure population of cells according to the second aspect or any of its embodiments, the isolated nucleic acid according to the third aspect or any of its embodiments, or the pharmaceutical composition of the fourth aspect or any of its embodiments for use in the treatment of Primary Hyperoxaluria, preferably Primary Hyperoxaluria type 1, the use comprising administering to the subject said lentiviral, said host cell or population of cells, or said composition.

Preferably, the use according to the fifth and sixth aspect is in combination with an immune response inhibitor, preferably dexamethasone, ciclosporin A, ciclosporin H and/or rapamycin, or any combination thereof.

GENERAL DEFINITIONS

It must be noted that, as used herein, the singular forms "a", "an", and "the", include plural references unless the context clearly indicates otherwise. Further, unless otherwise indicated, the term "at least" preceding a series of elements is to be understood to refer to every element in the series. Those skilled in the art will recognize or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the present invention.

The term “about” is used herein to mean approximately, roughly, around, or in the regions of. When the term "about" is used in conjunction with a numerical range, it modifies that range by extending the boundaries above and below the numerical values set forth. In general, the term "about" is used herein to modify a numerical value above and below the stated value by a variance of 10 percent, up or down (higher or lower).

As used herein, the conjunctive term "and/or" between multiple recited elements is understood as encompassing both individual and combined options. For instance, where two elements are conjoined by "and/or", a first option refers to the applicability of the first element without the second. A second option refers to the applicability of the second element without the first. A third option refers to the applicability of the first and second elements together. Any one of these options is understood to fall within the meaning, and therefore satisfy the requirement of the term "and/or" as used herein. Concurrent applicability of more than one of the options is also understood to fall within the meaning, and therefore satisfy the requirement of the term "and/or."

Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and variations such as "comprises" and "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integer or step. When used herein the term "comprising" can be substituted with the term "containing" or "including" or sometimes when used herein with the term "having". Any of the aforementioned terms (comprising, containing, including, having), whenever used herein in the context of an aspect or embodiment of the present invention may be substituted with the term "consisting of", though less preferred.

When used herein "consisting of" excludes any element, step, or ingredient not specified in the claim element.

A protein or nucleotide that "consists essentially of" a protein or nucleotide is a protein or nucleotide that has considerably the same amino acid or nucleotide sequence as the specified protein or nucleotide.

A protein or nucleotide that has "essentially the same amino acid or nucleotide sequence" as a protein or nucleotide, respectively, typically has more than 90% amino acid or nucleotide identity with this protein or nucleotide. Included in this definition are conservative amino acid substitutions.

The term "isolated" for the purposes of the present invention designates a biological material (cell, polypeptide, polynucleotide, or a fragment, variant, or derivative thereof) that has been removed from its original environment (the environment in which it is naturally present). For example, a polynucleotide present in the natural state in a plant or an animal is not isolated, however the same polynucleotide separated from the adjacent nucleic acids in which it is naturally present, is considered "isolated."

"Nucleic acids," "nucleic acid molecules," "oligonucleotide," and "polynucleotide" are used interchangeably and refer to the phosphate ester polymeric form of ribonucleosides (adenosine, guanosine, uridine or cytidine; "RNA molecules") or deoxyribonucleosides (deoxyadenosine, deoxyguanosine, deoxythymidine, or deoxycytidine; "DNA molecules"), or any phosphoester analogs thereof, such as phosphorothioates and thioesters, in either single strandedform, or a double-stranded helix. The term nucleic acid molecule, and in particular DNA or RNA molecule, refers only to the primary and secondary structure of the molecule, and does not limit it to any particular tertiary forms. Thus, this term includes double-stranded DNA found, inter alia, in linear or circular DNA molecules (e.g., restriction fragments), plasmids, supercoiled DNA and chromosomes.

A "coding region" or "coding sequence" is a portion of polynucleotide which consists of codons translatable into amino acids.

The term "downstream" refers to a nucleotide sequence that is located 3’ to a reference nucleotide sequence. In certain embodiments, downstream nucleotide sequences relate to sequences that follow the starting point of transcription.

The term "upstream" refers to a nucleotide sequence that is located 5’ to a reference nucleotide sequence. In certain embodiments, upstream nucleotide sequences relate to sequences that are located on the 5’ side of a coding region or starting point of transcription.

As used herein, the term "gene regulatory region" or "regulatory region" refers to nucleotide sequences located upstream (5' sequences), within, or downstream (3' sequences) of a coding region, and which influence the transcription, RNA processing, stability, or translation of the associated coding region. Regulatory regions can include promoters, translation leader sequences, introns, polyadenylation recognition sequences, RNA processing sites, effector binding sites and stem-loop structures. If a coding region is intended for expression in a eukaryotic cell, a polyadenylation signal and transcription termination sequence will usually be located 3’ to the coding sequence.

The term “posttranscriptional regulatory element” refers to a DNA sequence that when transcribed creates a tertiary structure which enhances or inhibits the expression of a protein.

"Transcriptional control sequences" refer to DNA regulatory sequences, such as promoters, enhancers, terminators, and the like, that provide for the expression of a coding sequence in a host cell.

The term “promoter” refers to a DNA sequence to which RNA polymerase can bind to in order to initiate transcription. The sequence may further contain binding sites for various proteins that regulate transcription, such as transcription factors. The promoter sequence may be composed of different promoter fragments (either different or the same fragments) that are localized closely in the DNA sequence and may be separated by linkers or spacers. Such promoters are referred to as chimeric promoters.

The terms “treatment” and “therapy”, as used in the present application, refer to a set of hygienic, pharmacological, surgical and/or physical means used with the intent to cure and/or alleviate a disease and/or symptoms with the goal of remediating the health problem. The terms “treatment” and “therapy” include preventive and curative methods, since both are directed to the maintenance and/or reestablishment of the health of an individual or animal. Regardless of the origin of the symptoms, disease and disability, the administration of a suitable medicament to alleviate and/or cure a health problem should be interpreted as a form of treatment or therapy within the context of this application.

The term “therapeutically effective amount” refers to an amount of matter which has a therapeutic effect, and which is able to treat PH1.

The terms “individual”, “patient” or “subject” are used interchangeably in the present application and are not meant to be limiting in any way. The “individual”, “patient” or “subject” can be of any age, sex and physical condition.

As used herein, "pharmaceutically acceptable carrier" or “pharmaceutically acceptable diluent” means any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is well known in the art. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed and, without limiting the scope of the present invention, include: additional buffering agents; preservatives; co-solvents; antioxidants, including ascorbic acid and methionine; chelating agents such as EDTA; metal complexes (e.g., Zn-protein complexes); biodegradable polymers, such as polyesters; salt-forming counterions, such as sodium, polyhydric sugar alcohols; amino acids, such as alanine, glycine, glutamine, asparagine, histidine, arginine, lysine, ornithine, leucine, 2-phenylalanine, glutamic acid, and threonine; organic sugars or sugar alcohols, such as lactitol, stachyose, mannose, sorbose, xylose, ribose, ribitol, myoinisitose, myoinisitol, galactose, galactitol, glycerol, cyclitols (e.g., inositol), polyethylene glycol; sulphur containing reducing agents, such as urea, glutathione, thioctic acid, sodium thioglycolate, thioglycerol, [alpha]- monothioglycerol, and sodium thio sulphate; low molecular weight proteins, such as human serum albumin, bovine serum albumin, gelatin, or other immunoglobulins; and hydrophilic polymers, such as polyvinylpyrrolidone. As used herein, the phrase "subject in need thereof includes subjects, such as mammalian subjects, that would benefit from administration of a nucleic acid molecule, a polypeptide, or vector provided herein, e.g., to improve hemostasis.

As used herein, the term "optimized," with regard to nucleotide sequences, refers to a polynucleotide sequence that encodes a polypeptide, wherein the polynucleotide sequence has been mutated to enhance a property of that polynucleotide sequence.

The term “lentivirus” (LV) refers to a genus of retroviruses that cause chronic and deadly diseases characterized by long incubation periods, in the human and other mammalian species. A “vector” is any vehicle which can be used to artificially carry foreign genetic material into a cell. Thus, a “lentiviral vector” refers to a recombinant lentivirus which carries a nucleic acid into a cell.

By “transcriptional unit” as used herein is meant the nucleotide sequence comprised in the LV vector of the present invention that comprises from 5 ' to 3 ' an hepatocyte-specific promoter, preferably the ET promoter, a nucleotide sequence encoding optimized Alanine- Glyoxylate and Serine-Pyruvate Aminotransferase (AGXT-RHEAM) protein, a woodchuck hepatitis virus posttranscriptional regulatory element (WPRE), preferably a mutated WPRE, and at least one copy of at least one miRNA target sequence; wherein the hepatocyte- specific promoter, the WPRE and the at least one copy of at least one miRNA target sequence are operably linked to and regulate the expression of AGXT-RHEAM.

DESCRIPTION

Although lentiviral vectors represent an attractive tool for gene delivery, the efficiency of transduction is key to ensure that enough cells are genetically corrected in order to obtain the desired therapeutic effect. Unfortunately, it is common to find that the rate of transduction observed in vitro does not correspond to the transduction efficiency achieved in vivo, and this is especially accentuated when the vectors are not or cannot be delivered directly to the tissue where the genetic correction is desired (for instance, in the case of liver-specific diseases), but they are delivered by other routes, such as parenteral route, where the vector is incorporated into blood stream and is targeted by host ' s immune response. The authors of the present invention present in Figure 13 the transduction efficiency of the same lentiviral vector used in vitro (in HepG2 cells) and in vivo (C57BL/6 mice), demonstrating that there is a significant reduction in the percentage of transduced cells when the virus is in an in vivo context. Additionally, the transduction efficiency does not only vary from in vitro/in vivo, or from local delivery versus systemically delivery, but it also varies depending on the type of the lentivirus used and its ability to transduce the cells and to integrate into the host cell genome. Particularly, the ability to transduce the target cell is clearly influenced by the lentiviral backbone (Johnson et al., 2021, Mol Therapy). Often, small changes in the lentiviral backbone are translated into big changes in its transduction efficiency. In view of all of this, it can be asserted that it is difficult to determine whether a specific lentiviral vector comprising a gene of interest (transgene) would efficiently work in vivo for a specific disease.

Further, some diseases, such as hemophilia (e.g., hemophilia A), can be treated with lentiviral vectors even though their transduction efficiency in vivo does not even reach 10% of cells. This is because, in these cases, an in vivo transduction efficacy of as low as 5% can be enough to obtain a therapeutic effect that is sufficient to revert the phenotype of the disease. However, in the particular case of PH1, it is widely accepted that at least 40% of hepatic cells need to be corrected, that is, need to efficiently produce AGXT protein, in order to revert, at least partially, the PH1 phenotype. Castello et al. have previously disclosed that hepatocyte transplantation strategies are unlikely to be successful due to the high number of engrafted hepatocytes needed for correction of PH1, which is highly likely to be beyond the capacity of this procedure (Castello et al. 2015. doi: 10.1038/gt.2015.107). Further, Jiang and colleagues also disclose that some authorities estimate that at least 75% of the overproducing mutant hepatocyte mass must be replaced with wild-type hepatocytes to achieve a clinical cure (Jiang et al. 2008. DOI: 10.1097/TP.0b013e31816de49e). However, the authors of the present invention have developed a lentiviral vector that is able to correct the deficiency in the expression of AGXT protein in hepatic cells causing a decrease in the oxalate accumulation and thus partially reverting the PH1 phenotype, as shown in Fig. 7-9. Surprisingly, this effect provided by the lentiviral vector described herein was achieved despite of the fact that, as shown in Fig. 5, said lentiviral vector was only able to transduce about 5% of the cells in vivo.

Thus, the present invention meets an important need in the art by providing lentiviral vectors comprising a codon optimized AGXT sequence that, upon transduction into a hepatic cell, increases the intracellular levels of AGXT protein and results in a great therapeutic efficacy to treat PH1. A schematic representation of the nucleic acid sequence comprised in the lentiviral vector provided herein is presented in Fig. 12.

Thus, in a first aspect, the present invention provides a lentivirus vector (LV) comprising a nucleic acid, wherein the nucleic acid comprises a transcriptional unit, wherein the transcriptional unit is defined herein as comprising, from 5’ to 3’, an hepatocyte-specific promoter, preferably a ET promoter, a nucleotide sequence encoding optimized Alanine- Glyoxylate and Serine-Pyruvate Aminotransferase (hereinafter referred to as AGXT-RHEAM) protein, a woodchuck hepatitis virus posttranscriptional regulatory element (hereinafter referred to as WPRE), preferably a mutated WPRE, and at least one copy of at least one miRNA target sequence; wherein the hepatocyte-specific promoter, the WPRE and the at least one copy of at least one miRNA target sequence are operably linked to and regulate the expression of AGXT-RHEAM. A coding sequence and a gene expression control sequence are said to be operably linked when they are linked in such a way as to place the expression or transcription and/or translation of the coding sequence under the influence or control of the gene expression control sequence. For example, the hepatocyte-specific promoter, the WPRE and the at least one copy of at least one miRNA target sequence are operably linked to nucleotide sequence encoding optimized AGXT-RHEAM protein so that the expression levels of AGXT are regulated by the promoter and WPRE.

In an embodiment, the nucleic acid comprised in the LV vector according to the first aspect further comprises one or more lentiviral backbone elements, which are defined as nucleic acids encoding for the necessary proteins for the LV to carry out its function (infect the cell and integrate into the viral genome to persist in the cell) and nucleic acids encoding for the necessary regulatory sequences for the LV to carry out its functions.

Thus, the LV vector of the first aspect comprises a nucleic acid, wherein the nucleic acid comprises 1) a transcriptional unit and 2) one or more LV vector backbone elements. It is noted that all the nucleic acid comprised in the transcriptional unit and in the LV vector backbone can be combined. Each of these two components are characterized in detail below:

1) The transcriptional unit

As defined above, by transcriptional unit is referred herein as to the nucleic acid sequence comprised in a LV vector of the present invention that comprises from 5 ' to 3 ' an hepatocyte- specific promoter, a nucleotide sequence encoding optimized AGXT-RHEAM protein, a WPRE element, and at least one copy of at least one miRNA target sequence; wherein the hepatocyte-specific promoter, the WPRE and the at least one copy of at least one miRNA target sequence are operably linked to and regulate the expression of AGXT-RHEAM.

• Hepatocyte-specific promoter

The hepatocyte-specific promoter leads the AGXT-RHEAM expression after lentiviral vector genome integration into the target cells. In some embodiments, the nucleic acid sequence comprised in the lentiviral vector comprises at least one tissue specific promoter, i.e., a promoter that will regulate the expression of the optimized AGXT protein in a particular tissue or cell type. In some embodiments, a tissue specific promoter in the lentiviral vector selectively enhances expression of the optimized AGXT protein in a target liver cell. In some embodiments, the target liver cell is a hepatocyte or a precursor cell thereof. In some embodiments, the isolated nucleic acid molecule comprised in the lentiviral vector is stably integrated into the genome of the target cell or target tissue, for example, in the genome of a hepatocyte.

In some embodiments, the lentiviral vector comprises a hepatocyte-specific promoter for transgene expression. In an embodiment, the nucleotide comprising the hepatocyte-specific promoter is an engineered hepatocyte-specific promoter. In some embodiments, the tissue specific promoter that selectively enhances expression of the optimized AGXT protein in a target liver cell comprises a transthyretin promoter (TTRp). In an embodiment, the hepatocyte-specific promoter is a chimeric promoter. In some embodiments, the chimeric hepatocyte-specific promoter is the ET promoter, which consists of a synthetic enhancer, a transthyretin enhancer (hereinafter referred to as TTR enhancer), a transthyretin promoter (hereinafter referred to as TTRp), and a transthyretin 5 ' UT (hereinafter referred to as mTTR 5’ UT).

In a preferred embodiment, the synthetic enhancer of the ET promoter comprises SEQ ID NO: 17 or a sequence which is at least 98% identical over the full length of SEQ ID NO: 17. Preferably the synthetic enhancer of the ET promoter comprises SEQ ID NO: 17 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 17. More preferably, the synthetic enhancer of the ET promoter is SEQ ID NO: 17 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 17.

In some embodiments, the nucleotide sequence of the synthetic enhancer of the ET promoter comprised in the lentiviral vector comprises, consists, or consists essentially of SEQ ID NO:17.

In a preferred embodiment, the TTR enhancer of the ET promoter is a murine TTR enhancer (mTTR enhancer) and comprises SEQ ID NO: 18 or a sequence which is at least 98% identical over the full length of SEQ ID NO: 18. Preferably the mTTR enhancer of the ET promoter comprises SEQ ID NO: 18 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 18. More preferably, the mTTR enhancer of the ET promoter is SEQ ID NO: 18 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 18.

In some embodiments, the nucleotide sequence of the mTTR enhancer of the ET promoter comprised in the lentiviral vector comprises, consists, or consists essentially of SEQ ID NO:18.

In a preferred embodiment, the transthyretin promoter (TTRp) of the ET promoter is a murine TTRp (mTTRp) and comprises SEQ ID NO: 19 or a sequence which is at least 98% identical over the full length of SEQ ID NO: 19. Preferably the mTTRp of the ET promoter comprises SEQ ID NO: 19 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 19. More preferably, the mTTRp of the ET promoter is SEQ ID NO: 19 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 19.

In some embodiments, the nucleotide sequence of the mTTRp of the ET promoter comprised in the lentiviral vector comprises, consists, or consists essentially of SEQ ID NO: 19.

In a preferred embodiment, the transthyretin 5 ' UT (TTR 5’ UT) of the ET promoter is a murine TTR 5 ' UT (mTTR 5 ' UT) and comprises SEQ ID NO: 20 or a sequence which is at least 98% identical over the full length of SEQ ID NO: 20. Preferably the mTTR 5’ UT of the ET promoter comprises SEQ ID NO: 20 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 20. More preferably, the mTTR 5’ UT of the ET promoter is SEQ ID NO: 20 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 20.

In some embodiments, the nucleotide sequence of the mTTR 5’ UT of the ET promoter comprised in the lentiviral vector comprises, consists, or consists essentially of SEQ ID NO:20.

The sequence identity between two sequences can be determined through conventional methods. For example, by using standard alignment logarithms known in the state of the art such as BLAST (Altschul etal., 1990. J Mol Biol. 215(3): 403-10). In a preferred embodiment, the sequence identity between two sequences is determined using BLAST.

In a preferred embodiment, the hepatocyte-specific promoter is the ET promoter which consists of a synthetic enhancer of SEQ ID NO: 17, an mTTR enhancer of SEQ ID NO: 18, a mTTRp of SEQ ID NO: 19, and a mTTR 5 ' UT of SEQ ID NO: 20, or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 17, 18, 19, or 20, respectively.

In a preferred embodiment, the nucleotide sequence of the hepatocyte-specific ET promoter comprises SEQ ID NO: 16 or a sequence, which is at least 98% identical over the full length of SEQ ID NO: 16. Preferably the nucleotide sequence of the hepatocyte-specific ET promoter comprises SEQ ID NO: 16 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 16. More preferably, the nucleotide sequence of the hepatocyte-specific ET promoter is SEQ ID NO: 16 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 16.

In some embodiments, the nucleotide sequence comprised in the lentiviral vector and that is a liver-specific promoter that selectively enhances expression of the optimized AGXT protein in a target liver cell comprises, consists, or consists essentially of ET promoter of SEQ ID NO:16.

• The nucleotide sequence encoding for the optimized Alanine-Glyoxylate and Serine- Pyruvate Aminotransferase (AGXT-RHEAM) protein.

Lentiviral vectors provided herein comprise codon optimized polynucleotides encoding the optimized AGXT protein. In one embodiment, the codon optimized polynucleotides AGXT is operably linked to at least one, preferably two, expression control sequence, including a promoter and a posttranscriptional element. In some embodiments, the present invention provides a lentiviral vector comprising an isolated nucleic acid molecule comprising a nucleotide sequence encoding a polypeptide with similar activity to AGXT. In an embodiment, the nucleotide sequence comprised in the lentiviral vector and encoding for AGXT-RHEAM encodes for a signal peptide that locates the newly synthetized protein in the peroxisome of the cell.

In a preferred embodiment, the nucleotide sequence encoding for AGXT-RHEAM comprises SEQ ID NO: 29 or a sequence, which is at least 98% identical over the full length of SEQ ID NO: 29. Preferably the nucleotide sequence encoding for AGXT-RHEAM comprises SEQ ID NO: 29 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 29. More preferably, the nucleotide sequence encoding for AGXT-RHEAM is SEQ ID NO: 29 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 29.

In some embodiments, the nucleotide sequence comprised in the lentiviral vector and encoding for AGXT-RHEAM comprises, consists, or consists essentially of SEQ ID NO:29.

In a preferred embodiment, the amino acid sequence of AGXT-RHEAM comprises SEQ ID NO: 52 or a sequence which is at least 98% identical over the full length of SEQ ID NO: 52. Preferably the amino acid sequence of AGXT-RHEAM comprises SEQ ID NO: 52 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 52. More preferably, the amino acid sequence of AGXT-RHEAM is SEQ ID NO: 52 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 52.

In some embodiments, the AGXT-RHEAM protein encoded by the nucleotide sequence comprised in the lentiviral vector comprises, consists, or consists essentially of SEQ ID NO: 52.

• WHV (woodchuck hepatitis virus) post-transcriptional regulatory element (WPRE)

Lentiviral vectors provided herein also comprise at least one posttranscriptional regulatory element operably linked to the nucleotide encoding for the optimized AGXT. In an embodiment, said posttranscriptional regulatory element is the WHV (woodchuck hepatitis virus) post-transcriptional regulatory element (WPRE). In an embodiment, the posttranscriptional regulatory element is mutated to increase the safety of the vector. In a preferred embodiment, the mutated WPRE contains a mutation in gene X frame.

In a preferred embodiment, the nucleotide sequence comprising the mutated WPRE comprises SEQ ID NO: 21 or a sequence which is at least 98% identical over the full length of SEQ ID NO: 21. Preferably the nucleotide sequence comprising the mutated WPRE comprises SEQ ID NO: 21 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 21. More preferably, the nucleotide sequence comprising the mutated WPRE is SEQ ID NO: 21 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 21.

In some embodiments, the nucleotide sequence WPRE comprised in the lentiviral vector comprises, consists, or consists essentially of SEQ ID NO:21.

• The at least one copy of at least one miRNA target sequence.

The present invention also provides at least one copy of at least one miRNA target sequence operably linked to the optimized AGXT-RHEAM nucleotide sequence. More than one copy of a miRNA target sequence may be included in the transcription unit to increase the effectiveness of the system. The one or more miRNA target sequences can be the same or different. In an embodiment, the transcription unit comprised in the lentiviral vector comprises 1, 2, 3, 4, 5, 6, 7 or 8 copies of the same or different miRNA target sequence.

In an embodiment, the target sequence is a miRNA-142-3 target. In some embodiments, the lentiviral vector comprises one or more target sequences for miRNA-142-3 to reduce immune response to the transgene product. In one embodiment, the lentiviral vector comprisesl, 2, 3, preferably 4, 5, 6, 7 or 8 repetitions or copies of miRNA-142-3 target sequences. In some embodiments, incorporating one or more target sequences for miRNA-142-3 into the transcription unit of the lentivirus of the present invention allows for a desired transgene expression profile and/or a reduced immunogenicity. In particular, incorporating one or more target sequences for miRNA-142-3 may suppress transgene expression in intravascular and extravascular hematopoietic lineages, whereas transgene expression is maintained in non- hematopoietic cells. In certain embodiments, the complementary sequence of hematopoietic- specific microRNAs, such as miRNA-142-3, is incorporated into the 3' untranslated region of a lentiviral vector, making the transgene-encoding transcript susceptible to miRNA-mediated down-regulation.

The target sequence can be fully or partially complementary to the miRNA. The term "fully complementary" means that the target sequence has a nucleic acid sequence which is 100 % complementary to the sequence of the miRNA which recognizes it. In an embodiment, the nucleic acid sequence comprised in the transcription unit comprises four repetitions of the target sequence of miRNA-142-3.

In a preferred embodiment, the nucleotide sequence comprising the miRNA-142-3 comprises SEQ ID NO: 22 or a sequence which is at least 98% identical over the full length of SEQ ID NO: 22. Preferably the nucleotide sequence comprising the miRNA-142-3 comprises SEQ ID NO: 22 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 22. More preferably, the nucleotide sequence comprising the miRNA-142-3 is SEQ ID NO: 22 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 22.

In some embodiments, the nucleotide sequence of miRNA-142-3 comprised in the lentiviral vector comprises, consists, or consists essentially of SEQ ID NO: 22.

In some embodiments, the target sequence is an miR-181 target. In some embodiments, the target sequence is an miR-223 target. In some embodiments, the target sequence is an miR- 122 target. In some embodiments, the target sequence is an miR-30b target. In a preferred embodiment, the miRNA target sequence is selected from the group consisting of miRNA- 142-3, miR-181, miR-223, or miR-30b.

It is to be understood that all the embodiments described above for the different elements of the transcription unit (the hepatocyte-specific promoter, the nucleotide sequence encoding optimized AGXT-RHEAM protein, the WPRE element, and the at least one copy of at least one miRNA target sequence) can be combined with each other. In a preferred embodiment, the transcription unit comprised in the nucleic acid of the lentiviral vector comprises or consists of: a) an engineered hepatocyte-specific promoter with at least 95%, preferably 98%, most preferably 100% sequence identity over the full length of SEQ ID NO 16, b) a nucleotide sequence encoding optimized Alanine-Glyoxylate and Serine- Pyruvate Aminotransferase (AGXT-RHEAM) protein with at least 95%, preferably 98%, most preferably 100% sequence identity over the full length of SEQ ID NO 29, c) a mutated optimized woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) with at least 95%, preferably 98%, most preferably 100% sequence identity over the full length of SEQ ID NO 21, and d) at least a copy of at least one miRNA target sequence, and wherein a), c) and d) are operably linked to and regulate the expression of b). In a further preferred embodiment, the d) at least a copy of at least one miRNA target sequence consists of four copies of the target sequence of miRNA-142-3, preferably with at least 95%, preferably 98%, most preferably 100% sequence identity over the full length of SEQ ID NO 22. In a preferred embodiment, the transcription unit comprised in the nucleic acid of the lentiviral vector comprises or consists of: a) an engineered hepatocyte-specific promoter with at least 95%, preferably 98%, most preferably 100% sequence identity over the full length of SEQ ID NO 16, b) a nucleotide sequence encoding optimized Alanine-Glyoxylate and Serine- Pyruvate Aminotransferase (AGXT-RHEAM) protein, wherein the AGXT-RHEAM protein has at least 95%, preferably 98%, most preferably 100%, amino acid sequence identity over the full length of SEQ ID NO: 52, c) a mutated optimized woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) with at least 95%, preferably 98%, most preferably 100% sequence identity over the full length of SEQ ID NO 21, and d) at least a copy of at least one miRNA target sequence, wherein the miRNA target sequence is selected from the group consisting of miRNA-142-3, miR-181, miR-223, or miR-30b, and wherein a), c) and d) are operably linked to and regulate the expression of b). In a further preferred embodiment, the d) at least a copy of at least one miRNA target sequence consists of four copies of the target sequence of miRNA-142-3, preferably with at least 95%, preferably 98%, most preferably 100% sequence identity over the full length of SEQ ID NO 22.

In a preferred embodiment, the transcription unit comprised in the nucleic acid of the lentiviral vector comprises consists of: a) an engineered hepatocyte-specific promoter consisting of SEQ ID NO 16, b) a nucleotide sequence encoding optimized Alanine-Glyoxylate and Serine- Pyruvate Aminotransferase (AGXT-RHEAM) protein, wherein the AGXT-RHEAM protein consists of SEQ ID NO: 52, c) a mutated optimized woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) consisting of SEQ ID NO 21, and d) at least a copy of at least one miRNA target sequence, wherein the miRNA target sequence is selected from the group consisting of miRNA-142-3, miR-181, miR-223, or miR-30b, and wherein a), c) and d) are operably linked to and regulate the expression of b). In a further preferred embodiment, the d) at least a copy of at least one miRNA target sequence consists of four copies of the target sequence of miRNA-142-3, preferably with at least 95%, preferably 98%, most preferably 100% sequence identity over the full length of SEQ ID NO 22. In a preferred embodiment, the transcription unit comprised in the nucleic acid of the lentiviral vector comprises or consists of: a) an engineered hepatocyte-specific promoter with at least 95%, preferably 98%, most preferably 100% sequence identity over the full length of SEQ ID NO 16, b) a nucleotide sequence encoding optimized Alanine-Glyoxylate and Serine- Pyruvate Aminotransferase (AGXT-RHEAM) protein with at least 95%, preferably 98%, most preferably 100% sequence identity over the full length of SEQ ID NO 29, c) a mutated optimized woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) with at least 95%, preferably 98%, most preferably 100% sequence identity over the full length of SEQ ID NO 21, and d) at least a copy of at least one miRNA target sequence, wherein the miRNA target sequence is selected from the group consisting of miRNA-142-3, miR-181, miR-223, or miR-30b, and wherein a), c) and d) are operably linked to and regulate the expression of b). In a further preferred embodiment, the d) at least a copy of at least one miRNA target sequence consists of four copies of the target sequence of miRNA-142-3, preferably with at least 95%, preferably 98%, most preferably 100% sequence identity over the full length of SEQ ID NO 22.

In a preferred embodiment, the transcription unit comprised in the nucleic acid of the lentiviral vector comprises or consists of: a) an engineered hepatocyte-specific promoter consisting of SEQ ID NO 16, b) a nucleotide sequence encoding optimized Alanine-Glyoxylate and Serine- Pyruvate Aminotransferase (AGXT-RHEAM) protein consisting of SEQ ID NO 29, c) a mutated optimized woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) consisting of SEQ ID NO 21, and d) at least a copy of at least one miRNA target sequence, wherein the miRNA target sequence is selected from the group consisting of miRNA-142-3, miR-181, miR-223, or miR-30b, and wherein a), c) and d) are operably linked to and regulate the expression of b). In a further preferred embodiment, the d) at least a copy of at least one miRNA target sequence consists of four copies of the target sequence of miRNA-142-3, preferably with at least 95%, preferably 98%, most preferably 100% sequence identity over the full length of SEQ ID NO 22. 2) Lentiviral vector backbone elements

As defined above, the lentiviral vector backbone elements are defined as nucleic acids sequences needed for the LV to carry out its function (infect the cell and integrate into the viral genome to persist in the cell).

Lentiviruses include members of the bovine lentivirus group, equine lentivirus group, feline lentivirus group, ovinecaprine lentivirus group, and primate lentivirus group.

In some embodiments, the lentiviral vector provided herein is "third-generation" lentiviral vector. As used herein, the term "third-generation" lentiviral vector refers to a lentiviral packaging system that has the characteristics of a second-generation vector system, and that further lacks a functional tat gene, such as one from which the tat gene has been deleted or inactivated. Typically, the gene encoding rev is provided on a separate expression construct. As used herein, a "second-generation" lentiviral vector system refers to a lentiviral packaging system that lacks functional accessory genes, such as one from which the accessory genes vif, vpr, vpu, and nef have been deleted or inactivated. As used herein, "packaging system" refers to a set of viral constructs comprising genes that encode viral proteins involved in packaging a recombinant virus. Typically, the constructs of the packaging system will ultimately be incorporated into a packaging cell.

In some embodiments, the third-generation lentiviral vector provided herein is a self inactivating lentiviral vector. In some embodiments, the lentiviral vector is a VSV.G pseudo type lentiviral vector.

In certain embodiments, the lentiviral vector is a vector of a recombinant lentivirus capable of infecting non-dividing cells. In certain embodiments, the lentiviral vector is a vector of a recombinant lentivirus capable of infecting liver cells (e.g., hepatocytes). The lentiviral genome and the proviral DNA typically have the three genes found in retroviruses: gag, pol and env, which are flanked by two long terminal repeat (LTR) sequences. The gag gene encodes the internal structural (matrix, capsid and nucleocapsid) proteins; the pol gene encodes the RNA-directed DNA polymerase (reverse transcriptase), a protease and an integrase; and the env gene encodes viral envelope glycoproteins. The 5' and 3' LTR's serve to promote transcription and polyadenylation of the virion RNA's. The LTR contains all other cis-acting sequences necessary for viral replication. In some embodiments the lentiviruses have additional genes including vif, vpr, tat, rev, vpu, net and vpx (in HIV-I, HIV-2 and/or SIV) In certain embodiments, the lentiviral vector has the HIV virulence genes env, vif, vpr, vpu and net deleted without compromising the ability of the vector to transduce non-dividing cells.

Adjacent to the 5' LTR are sequences necessary for reverse transcription of the genome (the tRNA primer binding site) and for efficient encapsidation of viral RNA into particles (the Psi site). The sequences necessary for encapsidation (or packaging of retroviral RNA into infectious virions) may be missing from the viral genome, the cis defect prevents encapsidation of genomic RNA. However, the resulting mutant remains capable of directing the synthesis of all virion proteins.

Thus, as defined above, the lentiviral vector provided herein comprises a nucleic acid, wherein the nucleic acid comprises a transcription unit. Further, in another embodiment, said nucleic acid comprised in the lentiviral vector comprises upstream, that is, at the 5 ' region of the transcriptional unit as defined above, and in order from 5 ' to 3 ' , at least one of the following: a) a 5’ long terminal repeat (5 ' LTR), b) a primer binding site (PBS), c) a psi packaging signal, d) the Stem-loop 4 (SL4) region of wild-type HIV virus, e) a Rev responsive element (RRE), and f) a DNA flap central polypurine tract (cPPT) or any combination thereof. In a preferred embodiment, said nucleic acid comprised in the lentiviral vector further comprises upstream, that is, at the 5 ' region of the transcriptional unit as defined above, I) a DenvRFI region located between d) and e), and m) a DenvRF2 region located between e) and f), or any combination thereof. Each of these nucleotides are further defined below.

In another embodiment, said nucleic acid comprised in the lentiviral vector further comprises downstream, that is, at the 3 ' region of the transcriptional unit as defined above, a k) 3’ long terminal repeat (LTR). In some embodiments, the lentiviral vector comprises a deletion of the U3 region of the 5' LTR. The deletion of the U3 region of the 5' LTR can be the complete deletion or a partial deletion.

It is to be understood that any combination possible between the nucleotides a) to f) and j) to k) is comprised in the present invention and in combination with the transcription unit described in the section above. Each of the nucleotides a) to f) and j) to k) and their preferred embodiments are further defined below.

Further, in another embodiment, said nucleic acid comprised in the lentiviral vector further comprises, preferably upstream (that is, at the 5 ' region) of the transcriptional unit as defined above, and preferably in order from 5 ' to 3 ' , at least one of the following: a) a 5’ long terminal repeat (5 ' LTR), b) a primer binding site (PBS), c) a psi packaging signal, d) the Stem-loop 4 (SL4) region of wild-type HIV virus, e) a Rev responsive element (RRE), and f) a DNA flap central polypurine tract (cPPT) or any combination thereof. In a preferred embodiment, said nucleic acid comprised in the lentiviral vector further comprises, preferably upstream (that is, at the 5 ' region) of the transcriptional unit as defined above h) a DenvRFI region located between d) and e), and i) a DenvRF2 region located between e) and f), or any combination thereof. In a further preferred embodiment, the nucleic acid comprised in the lentiviral vector further comprises, preferably downstream (that is, at the 3 ' region) of the transcriptional unit as defined above a g) 3’ long terminal repeat (LTR). Similar as above, it is to be understood that any combination possible between the nucleotides a), b), c), d), e), f), h) and i) and g) is comprised in the present invention and in combination with the transcription unit described in the section above.

In a preferred embodiment, the nucleotide sequence comprising the 5’ LTR comprises SEQ ID NO: 4 or a sequence which is at least 98% identical over the full length of SEQ ID NO: 4. Preferably the nucleotide sequence comprising the 5’ LTR comprises SEQ ID NO: 4 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 4. More preferably, the nucleotide sequence comprising the 5’ LTR is SEQ ID NO: 4 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 4.

In some embodiments, the nucleotide sequence of 5’ LTR comprised in the lentiviral vector comprises, consists, or consists essentially of SEQ ID NO:4.

In an embodiment, the 5 ' LTR region comprises a CMV promoter, preferably a chimeric CMV promoter, and/or a RU5 element. In an embodiment, the chimeric CMV promoter comprises or consists of a CMV enhancer and a CMV promoter. In a preferred embodiment, the 5 ' LTR region comprises or consists of a CMV enhancer, a CMV promoter, a RU5 element, and/or any combination thereof.

In a preferred embodiment, the nucleotide sequence comprising the chimeric CMV promoter comprises SEQ ID NO: 5 or a sequence which is at least 98% identical over the full length of SEQ ID NO: 5. Preferably the nucleotide sequence comprising the Chimeric CMV promoter comprises SEQ ID NO: 5 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 5. More preferably, the nucleotide sequence comprising the chimeric CMV promoter is SEQ ID NO: 5 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 5.

In some embodiments, the nucleotide sequence of chimeric CMV promoter comprised in the lentiviral vector comprises, consists, or consists essentially of SEQ ID NO:5.

In a preferred embodiment, the nucleotide sequence comprising the CMV enhancer comprises SEQ ID NO: 6 or a sequence which is at least 98% identical over the full length of SEQ ID NO: 6. Preferably the nucleotide sequence comprising the CMV enhancer comprises SEQ ID NO: 6 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 6. More preferably, the nucleotide sequence comprising the CMV enhancer is SEQ ID NO: 6 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 6.

In some embodiments, the nucleotide sequence of CMV enhancer comprised in the lentiviral vector comprises, consists, or consists essentially of SEQ ID NO: 6.

In a preferred embodiment, the nucleotide sequence comprising the CMV promoter comprises SEQ ID NO: 7 or a sequence which is at least 98% identical over the full length of SEQ ID NO: 7. Preferably the nucleotide sequence comprising the CMV promoter comprises SEQ ID NO: 7 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 7. More preferably, the nucleotide sequence comprising the CMV promoter is SEQ ID NO: 7 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 7.

In some embodiments, the nucleotide sequence of CMV promoter comprised in the lentiviral vector comprises, consists, or consists essentially of SEQ ID NO:7.

In a preferred embodiment, the nucleotide sequence comprising the RU5 element comprises SEQ ID NO: 8 or a sequence which is at least 98% identical over the full length of SEQ ID NO: 8. Preferably the nucleotide sequence comprising the RU5 element comprises SEQ ID NO: 8 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 8. More preferably, the nucleotide sequence comprising the RU5 element is SEQ ID NO: 8 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 8.

In some embodiments, the nucleotide sequence of RU5 element comprised in the lentiviral vector comprises, consists, or consists essentially of SEQ ID NO: 8.

In an embodiment, adjacent to the 5' LTR are sequences necessary for reverse transcription of the genome, which are the tRNA primer binding site (hereinafter referred to as PBS or PBS SL23).

In a preferred embodiment, the nucleotide sequence comprising the PBS SL23 comprises SEQ ID NO: 9 or a sequence which is at least 98% identical over the full length of SEQ ID NO: 9. Preferably the nucleotide sequence comprising the PBS SL23 comprises SEQ ID NO: 9 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 9. More preferably, the nucleotide sequence comprising the PBS SL23 is SEQ ID NO: 9 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 9.

In some embodiments, the nucleotide sequence of the PBS SL23 comprised in the lentiviral vector comprises, consists, or consists essentially of SEQ ID NO: 9.

In some embodiments, the lentiviral vector provided herein comprises at least a psi (Y) packaging signal, also called the psi site, for efficient encapsulation of viral RNA into particles.

In a preferred embodiment, the nucleotide sequence comprising the psi packaging signal comprises SEQ ID NO: 10 or a sequence which is at least 98% identical over the full length of SEQ ID NO: 10. Preferably the nucleotide sequence comprising the psi packaging signal comprises SEQ ID NO: 10 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 10. More preferably, the nucleotide sequence comprising the psi packaging signal is SEQ ID NO: 10 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 10.

In some embodiments, the nucleotide sequence of the psi packaging signal comprised in the lentiviral vector comprises, consists, or consists essentially of SEQ ID NO: 10. In an embodiment, the nucleotide of the PSI packaging signal partially overlaps with the PBS SL23 and with the Stem-loop 4 (SL4) region of wild-type HIV virus, which is defined below.

In certain embodiments, the lentiviral vector provided herein further comprises one or more nucleotide sequences encoding for a gag protein, a DenvRFI, a Rev-response element (hereinafter referred to as RRE), a DenVRF2, a central polypurine track (hereinafter referred to as cPPT), and/or any combination thereof.

In some embodiments, the lentiviral vector provided herein comprises at least a Stem-loop 4 (SL4) region of wild-type HIV virus (hereinafter referred to as SL4mgag).

In a preferred embodiment, the nucleotide sequence comprising the SL4mgag comprises SEQ ID NO: 11 or a sequence which is at least 98% identical over the full length of SEQ ID NO: 11. Preferably the nucleotide sequence comprising the SL4mgag comprises SEQ ID NO: 11 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 11. More preferably, the nucleotide sequence comprising the SL4mgag is SEQ ID NO: 11 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 11.

In some embodiments, the nucleotide sequence of the SL4mgag comprised in the lentiviral vector comprises, consists, or consists essentially of SEQ ID NO: 11.

In some embodiments, the lentiviral vector provided herein comprises at least a DenvRFI element.

In a preferred embodiment, the nucleotide sequence comprising the DenvRFI comprises SEQ ID NO: 12 or a sequence which is at least 98% identical over the full length of SEQ ID NO: 12. Preferably the nucleotide sequence comprising the DenvRFI comprises SEQ ID NO: 12 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 12. More preferably, the nucleotide sequence comprising the DenvRFI is SEQ ID NO: 12 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 12.

In some embodiments, the nucleotide sequence of the DenvRFI comprised in the lentiviral vector comprises, consists, or consists essentially of SEQ ID NO: 12. In an embodiment, the nucleotide of the DenvRFI partially overlaps with the nucleotide of the SL4mgag defined above and with the nucleotide of the RRE, which is defined below.

In a preferred embodiment, the nucleotide sequence comprising the RRE comprises SEQ ID NO: 13 or a sequence which is at least 98% identical over the full length of SEQ ID NO: 13. Preferably the nucleotide sequence comprising the RRE comprises SEQ ID NO: 13 or a sequence which is at least 75%, 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 13. More preferably, the nucleotide sequence comprising the RRE is SEQ ID NO: 13 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 13.

In some embodiments, the nucleotide sequence of RRE comprised in the lentiviral vector comprises, consists, or consists essentially of SEQ ID NO: 13.

In some embodiments, the lentiviral vector provided herein comprises at least a DenvRF2 element.

In a preferred embodiment, the nucleotide sequence comprising the DenvRF2 comprises SEQ ID NO: 14 or a sequence which is at least 98% identical over the full length of SEQ ID NO: 14. Preferably the nucleotide sequence comprising the DenvRF2 comprises SEQ ID NO: 14 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 14. More preferably, the nucleotide sequence comprising the DenvRF2 is SEQ ID NO: 14 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 14.

In some embodiments, the nucleotide sequence of the DenvRF2 comprised in the lentiviral vector comprises, consists, or consists essentially of SEQ ID NO: 14.

In a preferred embodiment, the nucleotide sequence comprising the cPPT comprises SEQ ID NO: 15 or a sequence which is at least 98% identical over the full length of SEQ ID NO: 15. Preferably the nucleotide sequence comprising the cPPT comprises SEQ ID NO: 15 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 15. More preferably, the nucleotide sequence comprising the cPPT is SEQ ID NO: 15 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 15.

In some embodiments, the nucleotide sequence of cPPT comprised in the lentiviral vector comprises, consists, or consists essentially of SEQ ID NO: 15.

In some embodiments the lentiviral vector comprises a nucleic acid, wherein the nucleic acid comprises a 3 ' long terminal repeat (hereinafter referred to as 3 ' LTR). 3’ LTR is involved in mRNAs polyadenylation during lentiviral vector production.

In a preferred embodiment, the 3 ' LTR comprises a deletion of the U3 region of the 3' LTR. The deletion of the U3 region of the 3' LTR can be the complete deletion or a partial deletion.

In a preferred embodiment, the nucleotide sequence comprising the 3 ' LTR comprises SEQ ID NO: 23 or a sequence which is at least 98% identical over the full length of SEQ ID NO: 23. Preferably the nucleotide sequence comprising the 3 ' LTR comprises SEQ ID NO: 23 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 23. More preferably, the nucleotide sequence comprising the 3 ' LTR is SEQ ID NO: 23 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 23.

In some embodiments, the nucleotide sequence of 3 ' LTR comprised in the lentiviral vector comprises, consists, or consists essentially of SEQ ID NO: 23.

In a preferred embodiment, the 3’ LTR is composed of a truncated U3 element (hereinafter referred to as AU3) and the elements R and U5 (hereinafter referred to as RU5) from wild- type HIV-1, being a self-inactivating lentiviral vector.

In a preferred embodiment, the nucleotide sequence comprising the AU3 comprises SEQ ID NO: 24 or a sequence which is at least 98% identical over the full length of SEQ ID NO: 24. Preferably the nucleotide sequence comprising the AU3 comprises SEQ ID NO: 24 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 24. More preferably, the nucleotide sequence comprising the AU3 is SEQ ID NO: 24 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 24. In some embodiments, the nucleotide sequence of DII3 comprised in the lentiviral vector comprises, consists, or consists essentially of SEQ ID NO: 24.

In a preferred embodiment, the nucleotide sequence comprising the RU5 comprises SEQ ID NO: 25 or a sequence which is at least 98% identical over the full length of SEQ ID NO: 25. Preferably the nucleotide sequence comprising the RU5 comprises SEQ ID NO: 25 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 25. More preferably, the nucleotide sequence comprising the RU5 is SEQ ID NO: 25 or a sequence which is at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% identical over the full length of SEQ ID NO: 25.

In some embodiments, the nucleotide sequence of RU5 comprised in the lentiviral vector comprises, consists, or consists essentially of SEQ ID NO: 25.

In an embodiment, the RU5 nucleotide is located between the chimeric CMV promoter and the PBS SL123 nucleotide. In an embodiment, the PBS SL123 nucleotide is located between the 5 ' LTR nucleotide and the Psi packaging signal. In an embodiment, the Psi packaging signal nucleotide is located between the PBS SL123 nucleotide and the Stem-loop 4 (SL4) region of wild-type HIV virus nucleotide. In an embodiment, the DenvRFI nucleotide is located between the SL4mgag nucleotide and the RRE nucleotide. In an embodiment, the DenvRF2 nucleotide is located between the RRE nucleotide and the cPPT nucleotide. In an embodiment, the cPPT nucleotide is located between the RRE nucleotide and the ET promoter nucleotide. In an embodiment, the at least one copy of at least one miRNA target sequence is located between the mutated WPRE nucleotide and the 3 ' LTR nucleotide.

In a further preferred embodiment of the first aspect or any of its embodiment, the lentiviral vector comprises a nucleic acid, wherein the nucleic acid comprises from 5’ to 3’ the following nucleotides: a) a 5’ long terminal repeat (LTR), b) a primer binding site (PBS) c) a psi packaging signal, d) the Stem-loop 4 (SL4) region of wild-type HIV virus e) a Rev responsive element (RRE), f) a DNA flap central polypurine tract (cPPT), g) an engineered hepatocyte-specific promoter with at least 95%, preferably 98%, sequence identity over the full length of SEQ ID NO 16, h) a nucleotide sequence encoding optimized Alanine-Glyoxylate and Serine- Pyruvate Aminotransferase (AGXT-RHEAM) protein with at least 95%, preferably 98%, sequence identity over the full length of SEQ ID NO 29, i) a mutated optimized woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) with at least 95%, preferably 98%, sequence identity over the full length of SEQ ID NO 21, j) at least a copy of at least one miRNA target sequence, and k) a 3’ long terminal repeat (LTR), wherein the long terminal repeat regions of a) and k) have been rendered substantially transcriptionally inactive by virtue of total or partial deletion in the U3 region of the LTR, and wherein g), i) and j) are operably linked to and regulate the expression of h). In a preferred embodiment, the at least a copy of at least one miRNA target sequence consists of four repetitions of the target sequence of miRNA-142-3.

In some embodiments, the nucleotide comprised in said lentiviral vector further comprises:

L) a DenvRFI region located between d) the Stem-loop 4 (SL4) region of wild- type HIV virus and e) a Rev responsive element (RRE), and m) a DenvRF2 region located between e) a Rev responsive element (RRE) and f) a DNA flap central polypurine tract (cPPT).

In a most preferred embodiment of the first aspect or any of its embodiment, the lentiviral vector comprises a nucleic acid, wherein the nucleic acid comprises from 5’ to 3’ the following nucleotides: a) a 5’ long terminal repeat (LTR) with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 5, b) a primer binding site (PBS) with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 9, c) a psi packaging signal with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 10, d) a Stem-loop 4 (SL4) region of wild-type HIV virus with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 11, e) a Rev responsive element (RRE) with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 13, f) a DNA flap central polypurine tract (cPPT) with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 15, g) an engineered hepatocyte-specific promoter with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 16, h) a nucleotide sequence encoding optimized Alanine-Glyoxylate and Serine- Pyruvate Aminotransferase (AGXT-RHEAM) protein with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 29, i) a mutated optimized woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 21, j) at least one copy of at least one miRNA target sequence, and k) a 3’ long terminal repeat (LTR) with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 3, wherein the long terminal repeat regions of a) and k) have been rendered substantially transcriptionally inactive by virtue of total or partial deletion in the U3 region of the LTR, and wherein g), i) and j) are operably linked to and regulate the expression of h). In a preferred embodiment, the at least a copy of at least one miRNA target sequence consists of four repetitions of the target sequence of miRNA-142-3 with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 22.

In some embodiments, the nucleotide comprised in said lentiviral vector further comprises:

L) a DenvRFI region with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 12 and located between d) the Stem-loop 4 (SL4) region of wild-type HIV virus and e) a Rev responsive element (RRE), and m) a DenvRF2 region with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 14 and located between e) a Rev responsive element (RRE) and f) a DNA flap central polypurine tract (cPPT),

In a further preferred embodiment of the first aspect, the lentiviral vector comprises a nucleic acid, wherein the nucleic acid comprises from 5’ to 3’ the following nucleotides: a) a 5’ long terminal repeat (LTR), b) a primer binding site (PBS), c) a psi packaging signal, d) a Stem-loop 4 (SL4) region of wild-type HIV virus, e) a Rev responsive element (RRE), f) a DNA flap central polypurine tract (cPPT), g) an engineered hepatocyte-specific promoter, with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 16, h) a nucleotide sequence encoding optimized Alanine-Glyoxylate and Serine- Pyruvate Aminotransferase (AGXT-RHEAM) protein with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 29, i) a mutated optimized woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 21 , j) at least one copy of at least one miRNA target sequence, k) a 3’ long terminal repeat (LTR),

L) a DenvRFI region located between d) and e), and m) a DenvRF2 region located between e) and f), and wherein the long terminal repeat regions of a) and k) have been rendered substantially transcriptionally inactive by virtue of total or partial deletion in the U3 region of the LTR, and wherein g), i) and j) are operably linked to and regulate the expression of h). In a preferred embodiment, the at least a copy of at least one miRNA target sequence consists of four repetitions of the target sequence of miRNA-142-3.

In a further preferred embodiment of the first aspect, the lentiviral vector comprises a nucleic acid, wherein the nucleic acid comprises from 5’ to 3’ the following nucleotides: a) a 5’ long terminal repeat (LTR) with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 5, b) a primer binding site (PBS) with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 9, c) a psi packaging signal with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 10, d) a Stem-loop 4 (SL4) region of wild-type HIV virus with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 11, e) a Rev responsive element (RRE) with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 13, f) a DNA flap central polypurine tract (cPPT) with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 15, g) an engineered hepatocyte-specific promoter with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 16, h) a nucleotide sequence encoding optimized Alanine-Glyoxylate and Serine- Pyruvate Aminotransferase (AGXT-RHEAM) protein with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 29, i) a mutated optimized woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 21, j) at least one copy of at least one miRNA target sequence, and k) a 3’ long terminal repeat (LTR) with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 3,

L) a DenvRFI region with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 12 and located between d) and e), and m) a DenvRF2 region with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 14 and located between e) and f), and wherein the long terminal repeat regions of a) and k) have been rendered substantially transcriptionally inactive by virtue of total or partial deletion in the U3 region of the LTR, and wherein g), i) and j) are operably linked to and regulate the expression of h). In a preferred embodiment, the at least a copy of at least one miRNA target sequence consists of four repetitions of the target sequence of miRNA-142-3 with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 22.

In another embodiment, the lentiviral vector, wherein the lentiviral vector comprises a nucleic acid, wherein the nucleic acid comprises from 5’ to 3’ the following nucleotides: a) a 5’ long terminal repeat (LTR), b) a primer binding site (PBS) c) a psi packaging signal, d) the Stem-loop 4 (SL4) region of wild-type HIV virus e) a Rev responsive element (RRE), f) a DNA flap central polypurine tract (cPPT), g) an engineered hepatocyte-specific promoter with at least 95%, preferably 98%, sequence identity over the full length of SEQ ID NO 16, h) a nucleotide sequence encoding optimized Alanine-Glyoxylate and Serine- Pyruvate Aminotransferase (AGXT-RHEAM) protein with at least 95%, preferably 98%, sequence identity over the full length of SEQ ID NO 29, i) a mutated optimized woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) with at least 95%, preferably 98%, sequence identity over the full length of SEQ ID NO 21, j) four copies of the target sequence of miRNA-142-3 with at least 95%, preferably 98%, sequence identity over the full length of SEQ ID NO 22, and k) a 3’ long terminal repeat (LTR), wherein the long terminal repeat regions of a) and k) have been rendered substantially transcriptionally inactive by virtue of total or partial deletion in the U3 region of the LTR, and wherein g), i) and j) are operably linked to and regulate the expression of h).

In some embodiments, the nucleotide comprised in said lentiviral vector further comprises:

L) a DenvRFI region located between d) the Stem-loop 4 (SL4) region of wild- type HIV virus and e) a Rev responsive element (RRE), and m) a DenvRF2 region located between e) a Rev responsive element (RRE) and f) a DNA flap central polypurine tract (cPPT).

In a preferred embodiment, the lentiviral vector comprises a nucleic acid, wherein the nucleic acid comprises, preferably from 5’ to 3’, the following: a) a 5’ long terminal repeat (LTR), b) a primer binding site (PBS) c) a psi packaging signal, d) the Stem-loop 4 (SL4) region of wild-type HIV virus, e) a Rev responsive element (RRE), f) a DNA flap central polypurine tract (cPPT), g) an engineered hepatocyte-specific promoter with at least 95%, preferably 98%, most preferably 100% sequence identity over the full length of SEQ ID NO 16, h) a nucleotide sequence encoding optimized Alanine-Glyoxylate and Serine- Pyruvate Aminotransferase (AGXT-RHEAM) protein, wherein the AGXT- RHEAM protein has at least 95%, preferably 98%, most preferably 100%, amino acid sequence identity over the full length of SEQ ID NO: 52, i) a mutated optimized woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) with at least 95%, preferably 98%, most preferably 100% sequence identity over the full length of SEQ ID NO 21, j) at least a copy of at least one miRNA target sequence, wherein the miRNA target sequence is selected from the group consisting of miRNA-142-3, miR- 181, miR-223, or miR-30b, k) a 3’ long terminal repeat (LTR), and wherein the long terminal repeat regions of a) and k) have been rendered substantially transcriptionally inactive by virtue of total or partial deletion in the U3 region of the LTR, and wherein g), i) and j) are operably linked to and regulate the expression of h). In a further preferred embodiment, the j) at least a copy of at least one miRNA target sequence consists of four copies of the target sequence of miRNA-142-3, preferably with at least 95%, preferably 98%, most preferably 100% sequence identity over the full length of SEQ ID NO 22.

In some embodiments, the nucleotide comprised in said lentiviral vector further comprises:

L) a DenvRFI region located between d) the Stem-loop 4 (SL4) region of wild- type HIV virus and e) a Rev responsive element (RRE), and m) a DenvRF2 region located between e) a Rev responsive element (RRE) and f) a DNA flap central polypurine tract (cPPT).

In a preferred embodiment, the lentiviral vector comprises a nucleic acid, wherein the nucleic acid comprises, preferably from 5’ to 3’, the following: a) a 5’ long terminal repeat (LTR), b) a primer binding site (PBS) c) a psi packaging signal, d) the Stem-loop 4 (SL4) region of wild-type HIV virus, e) a Rev responsive element (RRE), f) a DNA flap central polypurine tract (cPPT), g) an engineered hepatocyte-specific promoter consisting of SEQ ID NO 16, h) a nucleotide sequence encoding optimized Alanine-Glyoxylate and Serine- Pyruvate Aminotransferase (AGXT-RHEAM) protein, wherein the protein consists of SEQ ID NO: 52, i) a mutated optimized woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) consisting of SEQ ID NO 21 , j) at least a copy of at least one miRNA target sequence, wherein the miRNA target sequence is selected from the group consisting of miRNA-142-3, miR- 181, miR-223, or miR-30b, k) a 3’ long terminal repeat (LTR), and wherein the long terminal repeat regions of a) and k) have been rendered substantially transcriptionally inactive by virtue of total or partial deletion in the U3 region of the LTR, and wherein g), i) and j) are operably linked to and regulate the expression of h). In a further preferred embodiment, the j) at least a copy of at least one miRNA target sequence consists of four copies of the target sequence of miRNA-142-3, preferably with at least 95%, preferably 98%, most preferably 100% sequence identity over the full length of SEQ ID NO 22.

In some embodiments, the nucleotide comprised in said lentiviral vector further comprises:

L) a DenvRFI region located between d) the Stem-loop 4 (SL4) region of wild- type HIV virus and e) a Rev responsive element (RRE), and m) a DenvRF2 region located between e) a Rev responsive element (RRE) and f) a DNA flap central polypurine tract (cPPT).

In a further preferred embodiment of the first aspect, the lentiviral vector comprises a nucleic acid, wherein the nucleic acid comprises, preferably from 5’ to 3’, the following nucleotides: a) a 5’ long terminal repeat (LTR) with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 5, b) a primer binding site (PBS) with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 9, c) a psi packaging signal with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 10, d) a Stem-loop 4 (SL4) region of wild-type HIV virus with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 11, e) a Rev responsive element (RRE) with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 13, f) a DNA flap central polypurine tract (cPPT) with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 15, g) an engineered hepatocyte-specific promoter with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 16, h) a nucleotide sequence encoding optimized Alanine-Glyoxylate and Serine- Pyruvate Aminotransferase (AGXT-RHEAM) protein, wherein the AGXT- RHEAM protein has at least 95%, preferably 98%, most preferably 100%, amino acid sequence identity over the full length of SEQ ID NO: 52, i) a mutated optimized woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 21, j) at least one copy of at least one miRNA target sequence, wherein the miRNA target sequence is selected from the group consisting of miRNA-142-3, miR- 181, miR-223, or miR-30b, k) a 3’ long terminal repeat (LTR) with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 3,

L) a DenvRFI region with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 12 and located between d) and e), and m) a DenvRF2 region with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 14 and located between e) and f), and wherein the long terminal repeat regions of a) and k) have been rendered substantially transcriptionally inactive by virtue of total or partial deletion in the U3 region of the LTR, and wherein g), i) and j) are operably linked to and regulate the expression of h). In a preferred embodiment, the at least a copy of at least one miRNA target sequence consists of four repetitions of the target sequence of miRNA-142-3 with at least 95%, preferably 98%, more preferably 100%, sequence identity over the full length of SEQ ID NO: 22.

In a preferred embodiment, the full-length nucleotide comprised in the lentiviral provided herein has at least 75%, 80%, 85%, 89%, 90%, 91%, 92%, 95%, 97%, 98%, 99%, or 100% sequence identity over the full length of SEQ ID NO 30 (LV.ET.AGXT-RHEAM.142-3pT). In another preferred embodiment, the full-length nucleotide comprised in the lentiviral provided comprises, consists, or consists essentially of SEQ ID NO: 30.

In an embodiment, the lipid coat of the viral particle can include membrane bound polypeptides that were previously present on the surface of the host cell.

In some embodiments, the lentiviral vector has a modified expression of one or more polypeptides on its surface that inhibit an immune response to the lentiviral vector following administration to a human subject. In some embodiments, the surface of the lentiviral vector comprises one or more CD47 molecules. CD47 is a "marker of self-protein, which is ubiquitously expressed on human cells. Surface expression of CD47 inhibits macrophage- induced phagocytosis of endogenous cells through the interaction of CD47 and macrophage expressed-SIRPa. Cells expressing high levels of CD47 are less likely to be targeted and destroyed by human macrophages in vivo. In some embodiments, the lentiviral vector comprises a high concentration of CD47 polypeptide molecules on its surface. In some embodiments, the lentiviral vector is produced in a cell line that has a high expression level of CD47. In certain embodiments, the lentiviral vector is produced in a cell line, wherein the cell line has high expression of CD47 on the cell membrane. In particular embodiments, the lentiviral vector is produced in a CD47high HEK 293T cell, wherein the HEK 293T is has high expression of CD47 on the cell membrane. In some embodiments, the HEK 293T cell is modified to have increased expression of CD47 relative to unmodified HEK 293T cells. In certain embodiments, the CD47 is human CD47. In an embodiment, the surface of the lentiviral vector lacks expression or has a reduced expression of one or more major histocompatibility complex of class I (MHCI). In certain embodiment, the MHCI is human MHCI.

In a second aspect, the present invention relates to a host cell (e.g., a hepatocyte) or a substantially pure population of cells comprising (e.g., transduced) the lentiviral vector as defined in the first aspect or in any of its embodiments. In an embodiment of the second aspect, the host cell or the substantially pure population of cells are hepatic cells.

In a third aspect, the present invention relates to an isolated nucleic acid comprising the nucleotide comprised in the lentiviral vector as defined in the first aspect or any of its embodiments.

Pharmaceutical compositions

In a fourth aspect, the present invention provides a pharmaceutical composition comprising the lentiviral as defined in the first aspect or in any of its embodiments, the host cell or the substantially pure population of cells according to the second aspect or any of its embodiments, and/or the nucleic acid as defined in the third aspect or any of its embodiments and a pharmaceutically acceptable carrier or diluent.

In some embodiments, the substantially pure population of cells according to the second aspect or any of its embodiments, or the pharmaceutical composition of the fourth aspect or any of its embodiments the composition for administration are contacted or transduced with the lentiviral vector according to the first aspect or any of its embodiments, either in vivo, in vitro, or ex vivo.

A pharmaceutical composition as described herein may also contain other substances. These substances include, but are not limited to, cryoprotectants, lyoprotectants, surfactants, bulking agents, anti-oxidants, and stabilizing agents. In some embodiments, the pharmaceutical composition may be lyophilized.

The term "cryoprotectant' as used herein, includes agents which provide stability to the lentiviral vector against freezing-induced stresses, by being preferentially excluded from the lentiviral vector’s surface. Cryoprotectants may also offer protection during primary and secondary drying and long-term product storage. Non-limiting examples of cryoprotectants include sugars, such as sucrose, glucose, trehalose, mannitol, mannose, and lactose; polymers, such as dextran, hydroxyethyl starch and polyethylene glycol; surfactants, such as polysorbates (e.g., PS-20 or PS-80); and amino acids, such as glycine, arginine, leucine, and serine. A cryoprotectant exhibiting low toxicity in biological systems is generally used.

In one embodiment, a lyoprotectant is added to a pharmaceutical composition described herein. The term "lyoprotectant' as used herein, includes agents that provide stability to the lentiviral vector during the freeze-drying or dehydration process (primary and secondary freeze- drying cycles), by providing an amorphous glassy matrix and by binding with the lentiviral vector’s surface through hydrogen bonding, replacing the water molecules that are removed during the drying process. This helps to minimize product degradation during the lyophilization cycle, and improve the long-term product stability. Non-limiting examples of lyoprotectants include sugars, such as sucrose or trehalose; an amino acid, such as monosodium glutamate, non-crystalline glycine or histidine; a methylamine, such as betaine; a lyotropic salt, such as magnesium sulphate; a polyol, such as trihydric or higher sugar alcohols, e.g., glycerin, erythritol, glycerol, arabitol, xylitol, sorbitol, and mannitol; propylene glycol; polyethylene glycol; pluronics; and combinations thereof. The amount of lyoprotectant added to a pharmaceutical composition is generally an amount that does not lead to an unacceptable amount of degradation of the strain when the pharmaceutical composition is lyophilized.

In some embodiments, a bulking agent is included in the pharmaceutical composition. The term "bulking agent' as used herein, includes agents that provide the structure of the freeze- dried product without interacting directly with the pharmaceutical product. In addition to providing a pharmaceutically elegant cake, bulking agents may also impart useful qualities in regard to modifying the collapse temperature, providing freeze-thaw protection, and enhancing the strain stability over long-term storage. Non-limiting examples of bulking agents include mannitol, glycine, lactose, and sucrose. Bulking agents may be crystalline (such as glycine, mannitol, or sodium chloride) or amorphous (such as dextran, hydroxyethyl starch) and are generally used in formulations in an amount from 0.5% to 10%. Other pharmaceutically acceptable carriers, excipients, or stabilizers, such as those described in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980) may also be included in a pharmaceutical composition described herein, provided that they do not adversely affect the desired characteristics of the pharmaceutical composition. As used herein, " harmaceutically acceptable carrier " means any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is well known in the art. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed and include: additional buffering agents; preservatives; co-solvents; antioxidants, including ascorbic acid and methionine; chelating agents such as EDTA; metal complexes (e.g., Zn- protein complexes); biodegradable polymers, such as polyesters; salt-forming counterions, such as sodium, polyhydric sugar alcohols; amino acids, such as alanine, glycine, glutamine, asparagine, histidine, arginine, lysine, ornithine, leucine, 2-phenylalanine, glutamic acid, and threonine; organic sugars or sugar alcohols, such as lactitol, stachyose, mannose, sorbose, xylose, ribose, ribitol, myoinisitose, myoinisitol, galactose, galactitol, glycerol, cyclitols (e.g., inositol), polyethylene glycol; sulfur containing reducing agents, such as urea, glutathione, thioctic acid, sodium thioglycolate, thioglycerol, [alpha]-monothioglycerol, and sodium thio sulphate; low molecular weight proteins, such as human serum albumin, bovine serum albumin, gelatin, or other immunoglobulins; and hydrophilic polymers, such as polyvinylpyrrolidone.

The pharmaceutical composition may be prepared for oral, sublingual, buccal, intravenous, intramuscular, subcutaneous, intraperitoneal, conjunctival, rectal, transdermal, intrathecal, topical and/or inhalation-mediated administration. In a preferred embodiment, the pharmaceutical composition may be a solution which is suitable for intravenous, intramuscular, conjunctival, transdermal, intraperitoneal and/or subcutaneous administration. In another embodiment, the pharmaceutical composition may be a solution which is suitable for sublingual, buccal and/or inhalation-mediated administration routes. In an alternative embodiment, the pharmaceutical composition may be a gel or solution which is suitable for intrathecal administration. In an alternative embodiment, the pharmaceutical composition may be an aerosol which is suitable for inhalation-mediated administration. In a preferred embodiment, the pharmaceutical composition may be prepared for intrathecal administration.

The pharmaceutical composition may further comprise common excipients and carriers which are known in the state of the art. For solid pharmaceutical compositions, conventional nontoxic solid carriers may be used which include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like. For solution for injection, the pharmaceutical composition may further comprise cryoprotectants, lyoprotectants, surfactants, bulking agents, anti-oxidants, stabilizing agents and pharmaceutically acceptable carriers. For aerosol administration, the pharmaceutical compositions are generally supplied in finely divided form along with a surfactant and propellant. The surfactant must, of course, be nontoxic, and is generally soluble in the propellant. Representative of such agents are the esters or partial esters of fatty acids containing from 6 to 22 carbon atoms, such as caproic, octanoic, lauric, palmitic, stearic, linoleic, linolenic, olesteric and oleic acids with an aliphatic polyhydric alcohol or its cyclic anhydride. Mixed esters, such as mixed or natural glycerides may be employed. A carrier can also be included, as desired, as with, e.g., lecithin for intranasal delivery. For suppositories, traditional binders and carriers may include, for example, polyalkalene glycols or triglycerides.

Therefore, a suitable pharmaceutical composition for injection can comprise a buffer (e.g. acetate, phosphate or citrate buffer), a surfactant (e.g. polysorbate), optionally a stabilizer agent (e.g. human albumin), etc. However, in other embodiments compatible with the teachings herein, the lentiviral vector can be delivered directly to the site of the adverse cellular population, that is the hepatocytes, thereby increasing the exposure of the diseased tissue to the therapeutic agent.

The lentiviral vector provided herein can optionally be administered in combination with other agents that are effective in treating the disorder or condition in need of treatment (e.g., prophylactic or therapeutic). The administration of lentiviral vectors provided herein in conjunction or combination with an adjunct therapy means the sequential, simultaneous, coextensive, concurrent, concomitant or contemporaneous administration or application of the therapy and the disclosed polypeptides. In an embodiment, the lentiviral vector is administered in combination with other agents that are able to induce hepatic cell proliferation, such as, but not limited to, triiodothyronine hormone, human-interleukin 6, and/or derivatives thereof. In another embodiment, the lentiviral vector is administered in combination with other agents that are able to inhibit the immune response against the lentiviral vector, such as, but not limited to, dexamethasone, ciclosporin A, ciclosporin H, rapamycin, and/or derivatives thereof. In another embodiment, the lentiviral vector is administered in combination with other agents that are able to increase the number of specific receptor in target cells for VSV-G pseudotyped lentivirus, such as, but not limited to, statins, iron quelators, and/or derivatives thereof. In another embodiment, the lentiviral vector is administered in combination with other agents that are able to block apoptosis in transduced hepatocytes, such as, but not limited to, antioxidants such as N-acetylcysteine, and/or derivatives thereof.

In a particular embodiment, the lentiviral vector according to the first aspect or any of its embodiments is administrated in combination with Cyclosporin H (CsH), which acts as a transduction enhancer. In a preferred embodiment, the CsH is administrated in a dose that ranges from 20-1, 15-1, 10-1, 5-1, or 5-2.5 mg/kg. In an embodiment, the CsH is administered before, at the same time, or after the lentivirus’s administration. Doses intermediate in the above ranges are also intended to be within the scope of the invention. In some embodiments, the CsH is administered once. In other embodiments, the CsH is administered more than once, at least twice, at least three times, at least four times, at least five times, at least six times, at least seven times, at least eight times, at least nine times, or at least ten times. The repetitive administrations of CsH may be performed in the same day or separated in time. In an embodiment, intervals between single dosages of CsH can be daily, weekly, monthly or yearly.

Medial uses and Schedule, route and dose of administration

In a fifth aspect, the present invention provides the lentiviral vector according to the first aspect or any of its embodiment, the host cell or the substantially pure population of cells according to the second aspect or any of its embodiments, the isolated nucleic acid according to the third aspect or any of its embodiments, or the pharmaceutical composition of the fourth aspect or any of its embodiments for use as a medicament. In a sixth aspect, the present invention provides the lentiviral vector according to the first aspect or any of its embodiment, the host cell or the substantially pure population of cells according to the second aspect or any of its embodiments, the isolated nucleic acid according to the third aspect or any of its embodiments, or the pharmaceutical composition of the fourth aspect or any of its embodiments for use in the treatment of Primary Hyperoxaluria, preferably Primary Hyperoxaluria type 1, the use comprising administering to the subject said lentiviral, said host cell or population of cells, or said composition.

Preferably, the use of the fifth and sixth aspect is in combination with an immune response inhibitor, preferably dexamethasone, ciclosporin A, ciclosporin H and/or rapamycin, or any combination thereof.

The present invention also provides methods of treating, preventing, or ameliorating the PH1 disorder in a subject in need thereof comprising administering to the subject a therapeutically effective amount of the lentiviral vector according to the first aspect or any of its embodiment, the host cell or the substantially pure population of cells according to the second aspect or any of its embodiments, the isolated nucleic acid of the third aspect or any of its embodiments, or the pharmaceutical composition of the fourth aspect or any of its embodiments.

In another embodiment, the administration of a lentiviral vector, the host cell or the substantially pure population of cells, or the composition provided herein does not induce an immune response in a subject or induces a very low immune response in said subject.

In some embodiments, the lentiviral vector, the host cell or the substantially pure population of cells, or the composition of the present invention are administered as a single dose or multiple doses. In some embodiments, the lentiviral vector, the host cell or the substantially pure population of cells, or the composition of the present invention dose are administered at once or divided into multiple sub-dose, e.g., two sub-doses, three sub-doses, four sub-doses, five sub-doses, six sub-doses, or more than six sub-doses. In some embodiments, more than one lentiviral vector is administered. Most preferably, the lentiviral vector, the host cell or the substantially pure population of cells, or the composition of the present invention are administered as a single dose.

In some embodiments, the dose of lentiviral vector of the present invention, the host cell or the substantially pure population of cells of the present invention or the pharmaceutical composition is administered repeated at least twice, at least three times, at least four times, at least five times, at least six times, at least seven times, at least eight times, at least nine times, or at least ten times.

The lentiviral vector of the present invention, the host cell or the substantially pure population of cells of the present invention or the pharmaceutical composition can be administered locally or systemically. In one embodiment, the route of administration of the lentiviral vectors is parenteral. The term parenteral as used herein includes intravenous, intraarterial, intraperitoneal, intramuscular, subcutaneous, rectal or vaginal administration. In an embodiment, said lentiviral vector can be administrated intravenously, subcutaneously, intramuscularly, or via any mucosal surface, e.g., orally, sublingually, buccally, sublingually, nasally, rectally, vaginally or via pulmonary route. The intravenous form of parenteral administration is preferred. In an embodiment, a form for administration would be a solution for injection, in particular for intravenous or intraarterial injection or drip. Effective doses of the compositions provided herein, for the treatment of PH1 vary depending upon many different factors, including means of administration, target site, physiological state of the subject, whether the subject is a human being or an animal, other medications administered, and whether treatment is prophylactic or therapeutic. Usually, the subject is a human but non-human mammals including transgenic mammals can also be treated. Treatment dosages can be titrated using routine methods known to those of skill in the art to optimize safety and efficacy. In one embodiment, the subjects include, but are not limited to, individuals with PH1. In some embodiments, the subject is a pediatric subject, whereas in other aspects, the subject is an adult subject.

The lentiviral vector of the present invention, the host cell or the substantially pure population of cells of the present invention or the pharmaceutical composition can be administered as a single dose or as multiple doses, wherein the multiple doses can be administered continuously or at specific timed intervals. In vitro or in vivo assays can be employed to determine optimal dose ranges and/or schedules for administration. Additionally, effective doses can be extrapolated from dose-response curves obtained from animal models.

In a preferred embodiment, the lentiviral vector of the present invention, the host cell or the substantially pure population of cells of the present invention or the pharmaceutical composition is administered at a dose of at least 1 x 10 9 vector genomes/Kg body weight, preferably 1 x 10 10 , 1 x 10 11 or 1 x 10 12 vector genomes/Kg body weight. More preferably, at least or about 4.6 x 10 12 vector genomes/Kg body weight are administered.

In an embodiment, the dose of lentiviral is of at least 5 x 10 2 , 5 x 10 3 , 5 x 10 4 , 5 x 10 5 , 5 x

10 6 , 5 x 10 7 , 5 x 10 8 , 5 x 10 9 , 5 x 10 10 , 5 x 10 11 , 5 x 1012, 5 x 10 13 , 5 x 10 14 , 5 x 10 15 transducing units/kg (TU/kg). In an embodiment, the dose of lentiviral is between 5 x 10 2 to 5 x 10 3 ; 5 x 10 3 to 5 x 10 4 ; 5 x 10 4 to 5 x 10 5 ; 5 x 10 5 to 5 x 10 6 ; 5 x 10 6 to 5 x 10 7 ; 5 x 10 8 to 5 x 10 9 ; 5 x 10 9 to 5 x 10 10 ; 5 x 10 10 to 5 x 10 11 ; 5 x 10 11 to 5 x 10 12 transducing units/kg (TU/kg). In an embodiment, the dose of lentiviral is about 1 x 10 2 , 1 x 10 3 , 1 x 10 4 , 1 x 10 5 , 1 x 10 6 , 1 x

10 7 , 1 x 10 8 , 1 x 10 9 , 1 x 10 10 , 1 x 10 11 , 1 x 10 12 , 1 x 10 13 , 1 x 10 14 , 1 x 10 15 transducing units/kg (TU/kg) or about 5 x 10 2 , 5 x 10 3 , 5 x 10 4 , 5 x 10 s , 5 x 10 6 , 5 x 10 7 , 5 x 10 s , 5 x 10 9 , 5 x 10 10 , 5 x 10 11 , 5 x 1012, 5 x 10 13 , 5 x 10 14 , 5 x 10 15 transducing units/kg (TU/kg), or about 7.5 x 10 2 , 7.5 x 10 3 , 7.5 x 10 4 , 7.5 x 10 s , 7.5 x 10 6 , 7.5 x 10 7 , 7.5 x 10 s , 7.5 x 10 ® , 7.5 x 10 10 , 7.5 x 10 11 , 7.5 x 1012, 7.5 x 10 13 , 7.5 x 10 14 , 7.5 x 10 15 transducing units/kg (TU/kg).

Doses intermediate in the above ranges are also intended to be within the scope of the invention. The lentiviral vector of the present invention, the host cell or the substantially pure population of cells of the present invention or the pharmaceutical composition provided herein can be administered on multiple occasions. Intervals between single dosages can be daily, weekly, monthly or yearly. Intervals can also be irregular depending on the development of the disease or the curative effects of said lentivirals.

The dosage and frequency of administration of the lentiviral vector of the present invention, the host cell or the substantially pure population of cells of the present invention or the pharmaceutical composition can vary depending on whether the treatment is prophylactic or therapeutic. In prophylactic applications, compositions containing the lentiviral vector provided herein are administered to a subject not already in the disease state to enhance the subject’s resistance or minimize effects of disease. Such an amount is defined to be a "prophylactic effective dose." A relatively low dosage is administered at relatively infrequent intervals over a long period of time. Some subjects may continue to receive treatment for the rest of their lives.

CLAUSES

The present invention further provides the following clauses:

1. A lentiviral vector, wherein the lentiviral vector comprises a nucleic acid, wherein the nucleic acid comprises from 5’ to 3’ the following nucleotides: a) a 5’ long terminal repeat (LTR), b) a primer binding site (PBS) c) a psi packaging signal, d) the Stem-loop 4 (SL4) region of wild-type HIV virus e) a Rev responsive element (RRE), f) a DNA flap central polypurine tract (cPPT), g) an engineered hepatocyte-specific promoter with at least 95%, preferably 98%, sequence identity over the full length of SEQ ID NO 16, h) a nucleotide sequence encoding optimized Alanine-Glyoxylate and Serine- Pyruvate Aminotransferase (AGXT-RHEAM) protein with at least 95%, preferably 98%, sequence identity over the full length of SEQ ID NO 29, i) a mutated optimized woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) with at least 95%, preferably 98%, sequence identity over the full length of SEQ ID N021, j) at least a copy of at least one miRNA target sequence, and k) a 3’ long terminal repeat (LTR), wherein the long terminal repeat regions of a) and k) have been rendered substantially transcriptionally inactive by virtue of total or partial deletion in the U3 region of the LTR, and wherein g), i) and j) are operably linked to and regulate the expression of h).

2. The lentiviral vector according to clause 1, wherein the sequence identity of g), h), i) to SEQ ID NOs 16, 29, and 21, respectively, is 100%.

3. The lentiviral vector according to any of clauses 1 and 2, wherein the at least a copy of at least one miRNA target sequence of j) consists of four copies of the target sequence of miRNA-142-3 with at least 95%, preferably 98%, sequence identity over the full length of SEQ ID NO 22.

4. The lentiviral vector according to any of clauses 1 to 3, wherein the nucleotide comprised in said lentiviral vector further comprises:

L) a DenvRFI region located between d) and e), and m) a DenvRF2 region located between e) and f).

5. The lentiviral vector according to any of clauses 1 to 4, wherein the full-length nucleotide of said lentiviral has at least 95%, preferably 98% sequence identity with SEQ ID NO 30 (LV. ET. AGXT-RH EAM .142-3pT) .

6. The lentiviral vector according to clause 5, wherein the sequence identity with SEQ ID NO 30 is 100%.

7. A substantially pure population of cells transduced with the lentiviral vector as defined in any of clauses 1 to 6.

8. A pharmaceutical composition comprising the lentiviral vector as defined in any of clauses 1 to 6, or the substantially pure population of cells as defined in clause 7, and a pharmaceutically acceptable carrier or diluent.

9. The lentiviral vector according to any of clauses 1 to 6, the substantially pure population of cells according to clause 7, or the pharmaceutical composition according to clause 8, for use as a medicament.

10. The lentiviral vector according to any of clauses 1 to 6, the substantially pure population of cells according to clause 7, or the pharmaceutical composition according to clause 8, for use in the treatment of Primary Hyperoxaluria in a subject in need thereof, the method comprising administering to the subject said lentiviral vector, said population of cells, or said pharmaceutical composition.

11. The lentiviral vector, the substantially pure population of cells, or the pharmaceutical composition for use according to clause 10, wherein the Primary Hyperoxaluria is type 1.

EXAMPLES

EXAMPLE 1 : Materials, methods and sequences 1.1 Lentiviral vector construction.

Reporter lentiviral vector (LV) construct, which expresses an enhanced version of Green Fluorescent Protein (eGFP), was developed from a LV construct previously described by Naldini and Cantore’s group (LV.ET.FIX.142-3pT) (Brown et ai, 2007). eGFP expressing LV was developed by classic cloning techniques. LV.ET.FIX.142-3pT was digested by Nhel and Sail restriction enzymes (New England Biolabs®) to remove FIX sequence. Target sequences of Nhel and Sail are in 5’ and 3’ ends of FIX gene, respectively. After digestion of the plasmid with both enzymes, LV structure was isolated by electrophoresis and band purification.

Furthermore, we amplified eGFP sequence from #241.pCCL.sin.PPT.TTR.GFP.Wpre plasmid by PCR. Primers were designed to add Nhel (SEQ ID NO: 1 Fw: 5’-

ATTGGCTAGCATGGTGAGCAAGGGCGAGGA -3’) and Sail (SEQ ID NO: 2 Rv: 5’- CATTGTCGACTTACTTGTACAGCTCGTCCA -3’) restrictions enzymes target sequences at 5’ and 3’ ends respectively. Finally, both sequences were ligated and LV.ET.eGFP.142-3pT was obtained.

The expression cassette of LV.ET.eGFP.142-3pT contains an engineered hepatocyte- specific promoter (modified from Enhanced Transthyretin, GenBank accession number AY661265) and target sequences for the hematopoietic-lineage specific microRNA 142 (mature human microRNA 142 SEQ ID NO: 3 - miRNA-142-3p:

UGUAGUGUUUCCUACUUUAUGGA), with the aim of avoiding off-target transgene expression in Antigen Presenting Cells and, therefore, the development of an immune response against the transduced cells. LV. ET. EGFP.142-3PT SEQUENCES:

5’ LTR (SEQ ID NO: 4): it is responsible for provirus transcription during lentiviral vector production. In this lentiviral vector construct 5’ LTR is composed of a chimeric form of the CMV promoter and a part of wild-type HIV-1 5’ LTR, RU5. The lack of U3 sequence of wild- type 5’ LTR involves that this lentiviral vector construct is Tat-independent and, therefore, it will be produced in third generation.

TGGCC ATTGCAT ACGTT GT ATCC AT AT CAT AAT ATGT AC ATTT AT ATTGGCT CAT GT C CAACATT ACCGCCAT GTT G ACATT GATT ATT GACT AGTT ATT AAT AGT AAT CAATT AC GGGGTCATT AGTTCAT AGCCCAT AT ATGGAGTTCCGCGTT ACAT AACTT ACGGT AAA TGGCCCGCCTGGCTGACCGCCCAACGACCCCCGCCCATTGACGTCAATAATGACGT AT GTTCCCAT AGT AACGCCAAT AGGGACTTTCCATT GACGTCAAT GGGT GGAGT ATT T ACGGT AAACTGCCCACTTGGCAGT ACATCAAGT GT ATCAT AT GCCAAGT ACGCCCC CT ATT GACGTCAAT GACGGT AAATGGCCCGCCTGGCATT ATGCCCAGT ACAT GACCT T AT GGGACTTTCCT ACTTGGCAGT ACATCT ACGT ATT AGTCATCGCT ATT ACCATGGT GATGCGGTTTTGGCAGTACATCAATGGGCGTGGATAGCGGTTTGACTCACGGGGAT TTCCAAGTCTCCACCCCATTGACGTCAATGGGAGTTTGTTTTGGCACCAAAATCAAC GGGACTTTCCAAAATGTCGTAACAACTCCGCCCCATTGACGCAAATGGGCGGTAGG CGTGTACGGTGGGAGGTCTATATAAGCAGAGCTCGTTTAGTGAACCGGGGTCTCTC TGGTTAGACCAGATCTGAGCCTGGGAGCTCTCTGGCTAACTAGGGAACCCACTGCT T AAGCCTCAAT AAAGCTTGCCTT GAGTGCTTCAAGT AGT GT GTGCCCGTCT GTT GT G TGACTCTGGTAACTAGAGATCCCTCAGACCCTTTTAGTCAGTGTGGAAAATCTCTAG CAG

Chimeric CMV promoter (SEQ ID NO: 5): This sequence leads the transcription of provirus during the lentiviral vector production. It is composed of two different sequences, one with enhancer activity and the other one with promoter activity.

TGGCC ATTGCAT ACGTT GT ATCC AT AT CAT AAT ATGT AC ATTT AT ATTGGCT CAT GT C CAACATT ACCGCCAT GTT G ACATT GATT ATT GACT AGTT ATT AAT AGT AAT CAATT AC GGGGTCATT AGTTCAT AGCCCAT AT ATGGAGTTCCGCGTT ACAT AACTT ACGGT AAA TGGCCCGCCTGGCTGACCGCCCAACGACCCCCGCCCATTGACGTCAATAATGACGT AT GTTCCCAT AGT AACGCCAAT AGGGACTTTCCATT GACGTCAAT GGGTGGAGT ATT T ACGGT AAACTGCCCACTTGGCAGT ACATCAAGT GT ATCAT AT GCCAAGT ACGCCCC CT ATT GACGTCAAT GACGGT AAATGGCCCGCCTGGCATT ATGCCCAGT ACAT GACCT T AT GGG ACTTTCCT ACTT GGCAGT ACAT CTACGT ATT AGTCATCGCT ATT ACCAT GGT GATGCGGTTTTGGCAGTACATCAATGGGCGTGGATAGCGGTTTGACTCACGGGGAT

TTCCAAGTCTCCACCCCATTGACGTCAATGGGAGTTTGTTTTGGCACCAAAATCAAC

GGGACTTTCCAAAATGTCGTAACAACTCCGCCCCATTGACGCAAATGGGCGGTAGG

CGTGTACGGTGGGAGGTCTATATAAGCAGAGCTCGTTTAGTGAACC o CMV enhancer (SEQ ID NO: 6):

G ACATT GATT ATT G ACT AGTT ATT AAT AGT AAT CAATT ACGGGGTCATT AGTT CAT AGCCCAT AT ATGGAGTTCCGCGTT ACAT AACTT ACGGT AAATGGCCCGCCTGGC T GACCGCCCAACGACCCCCGCCCATT GACGTCAAT AAT GACGT AT GTTCCCAT A GTAACGCCAATAGGGACTTTCCATTGACGTCAATGGGTGGAGTATTTACGGTAAA CTGCCCACTTGGCAGT ACATCAAGT GT ATCAT ATGCCAAGTACGCCCCCT ATT GA CGTCAAT GACGGT AAATGGCCCGCCTGGCATT AT GCCCAGT ACAT GACCTT AT G GGACTTTCCT ACTTGGCAGT ACAT CTACGT ATT AGTCATCGCT ATT ACCAT G o CMV promoter (SEQ ID NO: 1Y

GTGATGCGGTTTTGGCAGTACATCAATGGGCGTGGATAGCGGTTTGACTCACGG GGATTTCCAAGTCTCCACCCC ATT GACGT CAAT GGG AGTTT GTTTT GGCACCAAA AT CAACGGG ACTTTCCAAAAT GTCGT AACAACTCCGCCCCATT GACGCAAATGG GCGGTAGGCGTGTACGGTGGGAGGTCTATATAAGCAGAGCT

RU5 (SEQ ID NO: 8): Truncated 5’ LTR without RU3. This sequence is necessary for retrotranscription and integration of proviral genome into transduced cells.

GGGTCTCTCTGGTTAGACCAGATCTGAGCCTGGGAGCTCTCTGGCTAACTAGGGAA CCCACTGCTT AAGCCTCAAT AAAGCTTGCCTT GAGTGCTTCAAGT AGT GT GTGCCCG TCTGTTGTGTGACTCTGGTAACTAGAGATCCCTCAGACCCTTTTAGTCAGTGTGGAA AATCTCTAGCAG

PBS SL23 SL123 (Primer Binding Site) (SEQ ID NO: 9): tRNA is merged to PBS SL23 element during reverse transcription of proviral genome after target cell transduction and before integration into the cell genome.

TGGCGCCCGAACAGGGACTTGAAAGCGAAAGGGAAACCAGAGGAGCTCTCTCGAC GCAGGACTCGGCTTGCTGAAGCGCGCACGGCAAGAGGCGAGGGGCGGCGACTGG T GAGT ACGCCAAAAATTTT GACT AGCGGAGGCT AGAAGGAGAGAG Y (packaqinq siqnal) (SEQ ID NO 10) This element is responsible for dimerization and packaging of lentiviral vector particles.

CTCTCTCGACGCAGGACTCGGCTTGCTGAAGCGCGCACGGCAAGAGGCGAGGGGC GGCGACTGGT GAGT ACGCCAAAAATTTT GACT AGCGGAGGCT AGAAGGAGAGAGAT GGGTGCGAGAGCGTC

SL4mqaq (SEQ ID NO 11) This element is a part of wild-type mgag HIV-1 gene which is involved in provirus packaging.

AT GGGT GCGAGAGCGTCAGT ATT AAGCGGGGGAGAATT AGATCGCGATGGGAAAAA ATTCGGTTAAGGCCAGGGGGAAAGAAAAAATATAAATTAAAACATATAGTATGGGCA AGCAGGGAGCT AGAACGATTCGCAGTT AATCCTGGCCT GTT AGAAACATCAGAAGG CTGT AG ACAAAT ACTGGG ACAGCT ACAACCATCCCTT CAGACAGG AT CAG AAG AACT T AGATCATT AT AT AAT ACAGT AGCAACCCTCT ATT GT GTGCATCAAAGGAT AGAGAT A AAAGACACCAAGGAAGCTTT AG ACAAGAT AG AGG AAG AGCAAAACAAAAGT AAG AC CACCGCACAGCAAGCGGCCGCTGAT

DenvRFI (SEQ ID NO: 12):

TCTTCAGACCTGGAGGAGGAGAT AT GAGGGACAATTGGAGAAGT GAATT AT AT AAAT AT AAAGT AGT AAAAATT G AACCATT AGGAGT AGCACCCACCAAGGCAAAG AG AAG AG TGGTGCAGAGAGAAAAAAGAGCAGTGGGAATA

RRE (Rev-response element) (SEQ ID NO: 13): Rev will merge to this sequence in lentiviral vector transcripts to help nuclear exportation of provirus during lentiviral vector production.

AGGAGCTTTGTTCCTTGGGTTCTTGGGAGCAGCAGGAAGCACTATGGGCGCAGCGT CAAT GACGCT GACGGT ACAGGCCAGACAATT ATT GTCTGGT AT AGTGCAGCAGCAG AACAATTTGCT GAGGGCT ATT GAGGCGCAACAGCATCT GTTGCAACTCACAGTCT G GGGCATCAAGCAGCT CCAGGCAAGAATCCTGGCT GTGGAAAGAT ACCT AAAGGATC AACAGCTCCT

DenvRF2 (SEQ ID NO: 14):

GGGTTGCTCTGGAAAACTCATTTGCACCACTGCTGTGCCTTGGAATGCTAGTTGGA GT AAT AAAT CT CT GG AACAG ATTTGG AAT CACACG ACCT GG AT GG AGTGGGACAGA G AAATT AACAATT ACACAAGCTT AAT ACACTCCTT AATT G AAGAATCGC AAAACCAGC AAGAAAAGAAT GAACAAGAATT ATT GGAATT AGAT AAATGGGCAAGTTT GT GGAATT GGTTT AACAT AACAAATT GGCT GTGGT ATAT AAAATT ATTCAT AAT GAT AGT AGGAGG CTTGGT AGGTTT AAGAAT AGTTTTT GCT GT ACTTTCT AT AGT GAAT AGAGTT AGGCAG GGATATTCACCATTATCGTTTCAGACCCACCTCCCAACCCCGAGGGGACCCGACAG GCCCGAAGGAATAGAAGAAGAAGGTGGAGAGAGAGACAGAGACAGATCCATTCGAT TAGTGAACGGATC cPPT (central polypurin tract) (SEQ ID NO: 15): This element act as a primer for positive DNA chain transcription during reverse transcription of lentiviral vectors after transduction of target cells. It also increases proviral DNA nuclear import and, therefore, lentiviral vector transduction efficacy. Moreover, it increases lentiviral vector titles.

TTTTAAAAGAAAAGGGGGGATTGGGGGGTACAGTGCAGGGGAAAGAATAGTAGACA T AAT AGCAACAGACAT ACAAACT AAAGAATT ACAAAAACAAATT ACAAAAATT CAAAAT

ET Promoter (SEQ ID NO: 16): It is an engineered hepatocyte-specific promoter. This sequence leads the transgene expression after lentiviral vector genome integration into the target cells. It is a chimeric promoter composed of a synthetic enhancer, an mTTR enhancer, and an mTTR promoter (Enhanced Transthyretin, GenBank accession number AY661265).

CGCGAGTTAATAATTACCAGCGCGGGCCAAATAAATAATCCGCGAGGGGCAGGTGA

CGTTTGCCCAGCGCGCGCTGGTAATTATTAACCTCGCGAATATTGATTCGAGGCCG

CGATTGCCGCAATCGCGAGGGGCAGGTGACCTTTGCCCAGCGCGCGTTCGCCCCG

CCCCGGACGGTATCGATAAGCTTAGGAGCTTGGGCTGCAGGTCGAGGGCACTGGG

AGGAT GTT GAGT AAGATGGAAAACT ACT GAT GACCCTTGCAGAGACAGAGT ATT AGG

ACATGTTTGAACAGGGGCCGGGCGATCAGCAGGTAGCTCTAGAGGATCCCCGTCT

GTCTGCACATTTCGTAGAGCGAGTGTTCCGATACTCTAATCTCCCTAGGCAAGGTTC

AT ATTT GTGT AGGTT ACTT ATT CTCCTTTT GTT G ACT AAGT CAAT AAT CAGAAT CAGCA

GGTTTGGAGTCAGCTTGGCAGGGATCAGCAGCCTGGGTTGGAAGGAGGGGGTATA

AAAGCCCCTTCACCAGGAGAAGCCGTCACACAGATCCACAAGCTCCTG

ET promoter components: o Synthetic enhancer (SEQ ID NO: 17): CGCGAGTTAATAATTACCAGCGCGGGCCAAATAAATAATCCGCGAGGGG CAGGTGACGTTTGCCCAGCGCGCGCTGGTAATTATTAACCTCGCGAATA TTGATTCGAGGCCGCGATTGCCGCAATCGCGAGGGGCAGGTGACCTTT GCCCAGCGCGCG o mTTR enhancer (SEQ ID NO: 18):

CACT GGGAGGAT GTT GAGT AAGATGGAAAACT ACT GAT GACCCTT GCAG

AGACAGAGTATTAGGACATGTTTGAACAGGGGCCGGGCGATCAGCAGGT

AG o mTTR promoter (SEQ ID NO: 19):

GTCTGTCTGCACATTTCGTAGAGCGAGTGTTCCGATACTCTAATCTCCCT AGGCAAGGTT CAT ATTT GTGT AGGTT ACTT ATT CTCCTTTT GTT GACT AAG TCAATAATCAGAATCAGCAGGTTTGGAGTCAGCTTGGCAGGGATCAGCA GCCTGGGTTGGAAGGAGGGGGTATAAAAGCCCCTTCACCAGGAGAAGC CGTC o mTTR 5’ UT (SEQ ID NO: 20): ACACAG ATCCACAAGCTCCT G eGFP sequence (SEQ ID NO: 51): This sequence codifies for an enhanced version of Green Fluorescent Protein gene.

TGGTGAGCAAGGGCGAGGAGCTGTTCACCGGGGTGGTGCCCATCCTGGTCGAGCT

GGACGGCGACGTAAACGGCCACAAGTTCAGCGTGTCCGGCGAGGGCGAGGGCGA

TGCCACCTACGGCAAGCTGACCCTGAAGTTCATCTGCACCACCGGCAAGCTGCCCG

TGCCCTGGCCCACCCTCGTGACCACCCTGACCTACGGCGTGCAGTGCTTCAGCCG

CTACCCCGACCACATGAAGCAGCACGACTTCTTCAAGTCCGCCATGCCCGAAGGCT

ACGTCCAGGAGCGCACCATCTTCTTCAAGGACGACGGCAACTACAAGACCCGCGCC

GAGGTGAAGTTCGAGGGCGACACCCTGGTGAACCGCATCGAGCTGAAGGGCATCG

ACTTCAAGGAGGACGGCAACATCCTGGGGCACAAGCTGGAGTACAACTACAACAGC

CACAACGTCTATATCATGGCCGACAAGCAGAAGAACGGCATCAAGGTGAACTTCAA

GATCCGCCACAACATCGAGGACGGCAGCGTGCAGCTCGCCGACCACTACCAGCAG

AACACCCCCATCGGCGACGGCCCCGTGCTGCTGCCCGACAACCACTACCTGAGCA

CCCAGTCCGCCCTGAGCAAAGACCCCAACGAGAAGCGCGATCACATGGTCCTGCT

GGAGTTCGTGACCGCCGCCGGGATCACTCTCGGCATGGACGAGCTGTACAAGTAA

A

Mutated WPRE (WHV (woodchuck hepatitis virus) post-transcriptional regulatory element)

(SEQ ID NO: 21) It increases transgene expression in transduced cells due to improvements in polyadenylation, RNA export from the nuclei and protein synthesis. It contains a mutation in gene X frame to increase security.

CAACCTCT GG ATT ACAAAATTT GT G AAAG ATT GACTGGT ATT CTT AACT ATGTTGCTC

CTTTTACGCTATGTGGATACGCTGCTTTAATGCCTTTGTATCATGCTATTGCTTCCC G

T AT GGCTTT CATTTT CTCCTCCTT GTAT AAATCCT GGTTGCT GTCTCTTT AT G AGG AG

TTGTGGCCCGTTGTCAGGCAACGTGGCGTGGTGTGCACTGTGTTTGCTGACGCAAC

CCCCACTGGTTGGGGCATTGCCACCACCTGTCAGCTCCTTTCCGGGACTTTCGCTT

TCCCCCTCCCTATTGCCACGGCGGAACTCATCGCCGCCTGCCTTGCCCGCTGCTGG

ACAGGGGCTCGGCTGTTGGGCACTGACAATTCCGTGGTGTTGTCGGGGAAATCATC

GTCCTTTCCTTGGCTGCTCGCCTGTGTTGCCACCTGGATTCTGCGCGGGACGTCCT

TCTGCTACGTCCCTTCGGCCCTCAATCCAGCGGACCTTCCTTCCCGC

142-3pT (SEQ ID NO: 22): Target sequence of 142-3 micro-RNA, which is specifically expressed in hematopoietic cells. This element increases the expression specificity of the transgene in transduced cells being a transcriptional control element.

TCCAT AAAGT AGGAAACACT ACA

3’ LTR: DR3RU5 (SEQ ID NO: 23): 3’ LTR is involved in mRNAs polyadenylation during lentiviral vector production. In this case, 3’ LTR is composed of a truncated U3 element and the elements R and U5 from wild-type HIV-1, being a self-inactivating lentiviral vector. The integrated viral genome will contain a deletion in viral promoter region (U3), resulting in the transcriptional inactivation of potentially packable viral genomes in the transduced cells.

TGGAAGGGCT AATTCACTCCCAACGAAGACAAGATCTGCTTTTTGCTT GT ACTGGGT CTCTCTGGTTAGACCAGATCTGAGCCTGGGAGCTCTCTGGCTAACTAGGGAACCCA CTGCTT AAGCCTCAAT AAAGCTT GCCTT GAGTGCTTCAAGT AGT GT GT GCCCGTCT G TTGTGTGACTCTGGTAACTAGAGATCCCTCAGACCCTTTTAGTCAGTGTGGAAAATC TCTAGCA o AU3 (SEQ ID NO: 24):

TGGAAGGGCT AATTCACTCCCAACGAAGACAAGATCTGCTTTTT GCTT GT ACT o RU5 (SEQ ID NO: 25):

GGGTCTCTCTGGTTAGACCAGATCTGAGCCTGGGAGCTCTCTGGCTAAC T AGGGAACCCACTGCTT AAGCCTCAAT AAAGCTT GCCTT GAGT GCTT CAA GTAGTGTGTGCCCGTCTGTTGTGTGACTCTGGTAACTAGAGATCCCTCA G ACCCTTTT AGTCAGT GTGG AAAAT CT CT AGC AG

LV.ET.eGFP.142-3pT sequence (SEQ ID NO: 26):

TGGCC ATTGCAT ACGTT GT ATCC AT AT CAT AAT ATGT AC ATTT AT ATTGGCT CAT GT C CAACATT ACCGCCAT GTT G ACATT GATT ATT GACT AGTT ATT AAT AGT AAT CAATT AC GGGGTCATT AGTTCAT AGCCCAT AT ATGGAGTTCCGCGTT ACAT AACTT ACGGT AAA TGGCCCGCCTGGCTGACCGCCCAACGACCCCCGCCCATTGACGTCAATAATGACGT AT GTTCCCAT AGT AACGCCAAT AGGGACTTTCCATT GACGTCAAT GGGT GGAGT ATT T ACGGT AAACTGCCCACTTGGCAGT ACATCAAGT GT ATCAT AT GCCAAGT ACGCCCC CT ATT GACGTCAAT GACGGT AAATGGCCCGCCTGGCATT ATGCCCAGT ACAT GACCT T AT GGGACTTTCCT ACTTGGCAGT ACATCT ACGT ATT AGTCATCGCT ATT ACCATGGT GATGCGGTTTTGGCAGTACATCAATGGGCGTGGATAGCGGTTTGACTCACGGGGAT TTCCAAGTCTCCACCCCATTGACGTCAATGGGAGTTTGTTTTGGCACCAAAATCAAC GGGACTTTCCAAAATGTCGTAACAACTCCGCCCCATTGACGCAAATGGGCGGTAGG CGTGTACGGTGGGAGGTCTATATAAGCAGAGCTCGTTTAGTGAACCGGGGTCTCTC TGGTTAGACCAGATCTGAGCCTGGGAGCTCTCTGGCTAACTAGGGAACCCACTGCT T AAGCCTCAAT AAAGCTTGCCTT GAGTGCTTCAAGT AGT GT GTGCCCGTCT GTT GT G TGACTCTGGTAACTAGAGATCCCTCAGACCCTTTTAGTCAGTGTGGAAAATCTCTAG CAGTGGCGCCCGAACAGGGACCTGAAAGCGAAAGGGAAACCAGAGCTCTCTCGAC GCAGGACTCGGCTTGCTGAAGCGCGCACGGCAAGAGGCGAGGGGCGGCGACTGG T GAGT ACGCCAAAAATTTT GACT AGCGGAGGCT AGAAGGAGAGAGATGGGTGCGAG AGCGTCAGT ATT AAGCGGGGGAGAATT AGATCGCGAT GGGAAAAAATTCGGTT AAG GCCAGGGGGAAAGAAAAAATATAAATTAAAACATATAGTATGGGCAAGCAGGGAGC T AGAACGATTCGCAGTT AATCCTGGCCT GTT AGAAACATCAGAAGGCT GT AGACAAA TACT GGG ACAGCT AC AACCATCCCTT CAGACAGG AT CAG AAG AACTT AGAT CATT AT AT AAT ACAGT AGCAACCCTCT ATT GTGT GCAT CAAAGGAT AGAG AT AAAAGACACCA AGGAAGCTTTAGACAAGATAGAGGAAGAGCAAAACAAAAGTAAGACCACCGCACAG CAAGCGGCCGCT GATCTTCAGACCTGGAGGAGGAGAT AT GAGGGACAATT GGAGA AGT G AATT AT AT AAAT AT AAAGT AGT AAAAATT G AACCATT AGG AGT AGC ACCC ACCA AGGCAAAGAGAAGAGTGGTGCAGAGAGAAAAAAGAGCAGTGGGAATAGGAGCTTT GTTCCTT GGGTTCTTGGGAGCAGCAGGAAGCACT AT GGGCGCAGCCTCAAT GACGC T GACGGT ACAGGCCAGACAATT ATT GTCTGGT AT AGTGCAGCAGCAGAACAATTT GC T GAGGGCT ATT GAGGCGCAACAGCATCT GTTGCAACTCACAGTCT GGGGCATCAAG CAGCT CCAGGCAAGAATCCTGGCT GTGGAAAGAT ACCT AAAGGATCAACAGCTCCT GGGGATTTGGGGTTGCTCTGGAAAACTCATTTGCACCACTGCTGTGCCTTGGAATG CT AGTTGGAGT AAT AAATCTCTGGAACAGATTTGGAATCACACGACCT GGAT GGAGT

GGGACAGAGAAATT AACAATT ACACAAGCTT AAT ACACTCCTT AATT GAAGAATCGCA

AAACCAGCAAGAAAAGAAT GAACAAGAATT ATTGGAATT AGAT AAATGGGCAAGTTT

GT GGAATTGGTTT AACAT AACAAATT GGCT GT GGT AT AT AAAATT ATTCAT AAT GAT A

GT AGGAGGCTTGGT AGGTTT AAGAAT AGTTTTT GCT GT ACTTTCT AT AGT GAAT AGAG

TTAGGCAGGGATATTCACCATTATCGTTTCAGACCCACCTCCCAACCCCGAGGGGA

CCCGACAGGCCCGAAGGAAT AGAAGAAGAAGGTGGAGAGAGAGACAGAGACAGAT

CCATTCG ATT AGT G AACGG AT CTCG ACGGT ATCGGTT AACTTTT AAAAG AAAAGGGG

GGATTGGGGGGTACAGTGCAGGGGAAAGAATAGTAGACATAATAGCAACAGACATA

CAAACT AAAGAATT ACAAAAACAAATT ACAAAAATT CAAAATTTT ATCG AT CACG AG A

CTAGCCTCGATCGAGGTCAATTCACGCGAGTTAATAATTACCAGCGCGGGCCAAAT

AAATAATCCGCGAGGGGCAGGTGACGTTTGCCCAGCGCGCGCTGGTAATTATTAAC

CTCGCGAATATTGATTCGAGGCCGCGATTGCCGCAATCGCGAGGGGCAGGTGACC

TTTGCCCAGCGCGCGTTCGCCCCGCCCCGGACGGTATCGATAAGCTTAGGAGCTT

GGGCTGCAGGTCGAGGGCACTGGGAGGATGTTGAGTAAGATGGAAAACTACTGAT

GACCCTT GCAGAGACAGAGT ATT AGGACAT GTTT GAACAGGGGCCGGGCGATCAGC

AGGTAGCTCTAGAGGATCCCCGTCTGTCTGCACATTTCGTAGAGCGAGTGTTCCGA

TACTCTAATCTCCCTAGGCAAGGTTCATATTTGTGTAGGTTACTTATTCTCCTTTTG TT

GACTAAGTCAATAATCAGAATCAGCAGGTTTGGAGTCAGCTTGGCAGGGATCAGCA

GCCTGGGTTGGAAGGAGGGGGTATAAAAGCCCCTTCACCAGGAGAAGCCGTCACA

CAGATCCACAAGCTCCTGCCACCATGGTGAGCAAGGGCGAGGAGCTGTTCACCGG

GGTGGTGCCCATCCTGGTCGAGCTGGACGGCGACGTAAACGGCCACAAGTTCAGC

GTGTCCGGCGAGGGCGAGGGCGATGCCACCTACGGCAAGCTGACCCTGAAGTTCA

TCTGCACCACCGGCAAGCTGCCCGTGCCCTGGCCCACCCTCGTGACCACCCTGAC

CTACGGCGTGCAGTGCTTCAGCCGCTACCCCGACCACATGAAGCAGCACGACTTCT

TCAAGTCCGCCATGCCCGAAGGCTACGTCCAGGAGCGCACCATCTTCTTCAAGGAC

GACGGCAACTACAAGACCCGCGCCGAGGTGAAGTTCGAGGGCGACACCCTGGTGA

ACCGCATCGAGCTGAAGGGCATCGACTTCAAGGAGGACGGCAACATCCTGGGGCA

CAAGCTGGAGT ACAACT ACAACAGCCACAACGTCTAT ATCATGGCCGACAAGCAGA

AGAACGGCATCAAGGTGAACTTCAAGATCCGCCACAACATCGAGGACGGCAGCGTG

CAGCTCGCCGACCACTACCAGCAGAACACCCCCATCGGCGACGGCCCCGTGCTGC

TGCCCGACAACCACT ACCT GAGCACCCAGTCCGCCCT GAGCAAAGACCCCAACGA

GAAGCGCGATCACATGGTCCTGCTGGAGTTCGTGACCGCCGCCGGGATCACTCTC

GGCATGGACGAGCTGTACAAGTAAAGCGGCCTCGACGGGCCCGCGGAATTTCGAC

AAT CAACCT CT GG ATT ACAAAATTT GT GAAAG ATT G ACT GGT ATT CTT AACT AT GTT G

CTCCTTTT ACGCT AT GTGGAT ACGCTGCTTT AATGCCTTT GT ATCATGCT ATT GCTTC

CCGTAT GGCTTT CATTTT CTCCTCCTT GTAT AAATCCT GGTTGCTGT CT CTTT AT GAG GAGTTGTGGCCCGTTGTCAGGCAACGTGGCGTGGTGTGCACTGTGTTTGCTGACGC

AACCCCCACTGGTTGGGGCATTGCCACCACCTGTCAGCTCCTTTCCGGGACTTTCG

CTTTCCCCCTCCCTATTGCCACGGCGGAACTCATCGCCGCCTGCCTTGCCCGCTGC

TGGACAGGGGCTCGGCTGTTGGGCACTGACAATTCCGTGGTGTTGTCGGGGAAGC

TGACGTCCTTTCCATGGCTGCTCGCCTGTGTTGCCACCTGGATTCTGCGCGGGACG

TCCTTCTGCTACGTCCCTTCGGCCCTCAATCCAGCGGACCTTCCTTCCCGCGGCCT

GCTGCCGGCTCTGCGGCCTCTTCCGCGTCTTCGCCTTCGCCCTCAGACGAGTCGG

ATCTCCCTTTGGGCCGCCTCCCCGCCTGGAATTCGAGCTCCACCGCGGTGGCGGC

CGCTCTAGAGTCGACTCCATAAAGTAGGAAACACTACACGATTCCATAAAGTAGGAA

ACACT ACAACCGGTTCCAT AAAGT AGG AAACACT ACAT CACTCCAT AAAGT AGG AAA

CACTACACTCGAGGGGGGGCCCGGTACCTTTAAGACCAATGACTTACAAGGCAGCT

GT AGATCTT AGCCACTTTTT AAAAGAAAAGGGGGGACTGGAAGGGCT AATTCACTCC

CAACGAAGACAAGATCTGCTTTTTGCTTGTACTGGGTCTCTCTGGTTAGACCAGATC

TGAGCCTGGGAGCTCTCTGGCTAACTAGGGAACCCACTGCTTAAGCCTCAATAAAG

CTTGCCTT G AGTGCTT CAAGT AGT GTGTGCCCGTCTGTTGTGT G ACT CTGGT AACT A

GAGATCCCTCAGACCCTTTTAGTCAGTGTGGAAAATCTCTAGCA

LV.ET.eGFP.142-3pT map: LV.ET.eGFP.142-3pT construct. Different elements previously described are represented in Figure 11.

At the same time, we developed a therapeutic LV expressing an improved version of AGXT gene (AGXT-RHEAM). We used the isolated structure of LV.ET.FIX.142-3pT, obtained as explained above. We amplified the AGXT-RHEAM sequence, kindly provided by Eduardo Salido’s group (Mesa-Torres et a!., 2014) by PCR. Primers were designed to add Nhel (SEQ ID NO: 27 Fw: 5’ CTA GOT AGO ATG GCC TCT CAC AAG CTG CT 3’) and Sail (SEQ ID NO: 28 Rv: 5’ CAA GTC GAC TCA CAG CTT CTT CTT GGG GC 3’) target sequences in 5’ and 3’ ends respectively, to use them in the cloning. Finally, LV.ET.FIX.142-3pT structure and AGXT- RHEAM sequence were ligated and LV.ET. AGXT-RHEAM.142-3pT vector was obtained.

LV. ET. AGXT-RH EAM.142-3PT SEQUENCES:

LV.ET. AGXT-RHEAM.142-3pT sequence contains the same elements as LV.EG.eGFP.142- 3pT except eGFP, which has been replaced by AGXT-RHEAM coding sequence. AGXT-RHEAM (SEQ ID NO: 29): This sequence, developed by Eduardo Salido’s group

(Mesa-Torres et a!., 2014), codifies for an improved version of AGXT gene with 5 amino acid changes to increase protein stability and half-life.

ATGGCCTCTCACAAGCTGCTGGTGACCCCCCCCAAGGCCCTGCTCAAGCCCCTCTC

CATCCCCAACCGTCTCCTGCTGGGGCCTGGTCCTTCCAACCTGCCTCCTCGCATCA

TGGCAGCCGGGGGGCTGCAGATGATCGGGCACATGAGCAAGGAAATGTACCAGAT

CATGGACGAGATCAAGGAAGGCATCCAGTACGTGTTCCAGACCAGGAACCCACTCA

CACTGGTCATCTCCGGCTCGGGACACTGTGCCCTGGAGGCCGCCCTGGTCAATGT

GCTGGAGCCTGGGGACTCCTTCCTGGTTGGGGCCAATGGCATTTGGGGGCAGCGA

GCCGCGGACATCGGGGAGCGCATAGGAGCCCGAGTGCACCCGATGACCAAGGAC

CCCGGAGGCCACTACACACTGCAGGAGGTGGAGGAGGGCCTGGCCCAGCACAAG

CCAGTGCTGCTGTTCTTAACCCACGGGGAGTCGTCCACCGGCGTGCTGCAGCCCC

TTGATGGCTTCGGGGAACTCTGCCACAGGTACAAGTGCCTGCTCCTGGTGGATTCG

GTGGCATCCCTGGGCGGGACCCCCCTTTACATGGACCGGCAAGGCATCGACATCC

TGTACTCGGGCTCCCAGAAGGCCCTGAACGCCCCTCCAGGGACCTCGCTCATCTCC

TTCAGTGACAAGGCCAAAAAGAAGATGTACTCCCGCAAGACGAAGCCCTTCTCCTTC

TACCTGGACATCAAGTGGCTGGCCAACTTCTGGGGCTGTGACGACCAGCCCAGGAT

GTACCATCACACAATCCCCGTCATCAGCCTGTACAGCCTGAGAGAGAGCCTGGCCC

TCATTGCGGAACAGGGCCTGGAGAACAGCTGGCGCCAGCACCGCGAGGCCGCGG

CGTATCTGCATGGGCGCCTGCAGGCACTGGGGCTGCAGCTCTTCGTGAAGGACCC

GGCGCTCCGGCTTCCCACAGTCACCACTGTGGCTGTACCCGCTGGCTATGACTGGA

GAGACATCGTCAGCTACGTCATGGACCACTTCGACATTGAGATCATGGGTGGCCTT

GGGCCCTCCACGGGGAAGGTGCTGCGGATCGGCCTGCTGGGCTGCAATGCCACC

CGCGAGAATGTGGACCGCGTGACGGAGGCCCTGAGGGCGGCCCTGCAGCACTGC

CCCAAGAAGAAGCT GT G A

LV.ET.AGXT-RHEAM.142-3PT (SEQ ID NO: 30):

TGGCC ATTGCAT ACGTT GT ATCC AT AT CAT AAT ATGT AC ATTT AT ATTGGCT CAT GT C CAACATT ACCGCCAT GTT G ACATT GATT ATT GACT AGTT ATT AAT AGT AAT CAATT AC GGGGTCATT AGTTCAT AGCCCAT AT ATGGAGTTCCGCGTT ACAT AACTT ACGGT AAA TGGCCCGCCTGGCTGACCGCCCAACGACCCCCGCCCATTGACGTCAATAATGACGT AT GTTCCCAT AGT AACGCCAAT AGGGACTTTCCATT GACGTCAAT GGGT GGAGT ATT T ACGGT AAACTGCCCACTTGGCAGT ACATCAAGT GT ATCAT AT GCCAAGT ACGCCCC CT ATT GACGTCAAT GACGGT AAATGGCCCGCCTGGCATT ATGCCCAGT ACAT GACCT T AT GGGACTTTCCT ACTTGGCAGT ACATCT ACGT ATT AGTCATCGCT ATT ACCATGGT GATGCGGTTTTGGCAGTACATCAATGGGCGTGGATAGCGGTTTGACTCACGGGGAT

TTCCAAGTCTCCACCCCATTGACGTCAATGGGAGTTTGTTTTGGCACCAAAATCAAC

GGGACTTTCCAAAATGTCGTAACAACTCCGCCCCATTGACGCAAATGGGCGGTAGG

CGTGTACGGTGGGAGGTCTATATAAGCAGAGCTCGTTTAGTGAACCGGGGTCTCTC

TGGTTAGACCAGATCTGAGCCTGGGAGCTCTCTGGCTAACTAGGGAACCCACTGCT

T AAGCCTCAAT AAAGCTTGCCTT GAGTGCTTCAAGT AGT GT GTGCCCGTCT GTT GT G

TGACTCTGGTAACTAGAGATCCCTCAGACCCTTTTAGTCAGTGTGGAAAATCTCTAG

CAGTGGCGCCCGAACAGGGACTTGAAAGCGAAAGGGAAACCAGAGGAGCTCTCTC

GACGCAGGACTCGGCTTGCTGAAGCGCGCACGGCAAGAGGCGAGGGGCGGCGAC

TGGT GAGT ACGCCAAAAATTTT GACT AGCGGAGGCT AGAAGGAGAGAGAT GGGT GC

GAGAGCGTCAGTATTAAGCGGGGGAGAATTAGATCGCGATGGGAAAAAATTCGGTT

AAGGCCAGGGGGAAAGAAAAAATATAAATTAAAACATATAGTATGGGCAAGCAGGG

AGCT AGAACGATTCGCAGTT AATCCT GGCCT GTT AGAAACATCAGAAGGCT GT AGAC

AAAT ACTGGGACAGCT ACAACC ATCCCTT CAG AC AGG AT CAGAAGAACTT AG AT CAT

T AT AT AAT ACAGT AGCAACCCT CT ATT GT GTGCAT CAAAGG AT AG AGAT AAAAG ACAC

CAAGGAAGCTTTAGACAAGATAGAGGAAGAGCAAAACAAAAGTAAGACCACCGCAC

AGCAAGCGGCCGCTGATCTTCAGACCTGGAGGAGGAGATATGAGGGACAATTGGA

G AAGT GAATT AT AT AAAT AT AAAGT AGT AAAAATT G AACC ATT AGG AGT AGCACCC AC

CAAGGCAAAGAGAAGAGTGGTGCAGAGAGAAAAAAGAGCAGTGGGAATAGGAGCT

TTGTTCCTTGGGTTCTTGGGAGCAGCAGGAAGCACTATGGGCGCAGCGTCAATGAC

GCTGACGGTACAGGCCAGACAATTATTGTCTGGTATAGTGCAGCAGCAGAACAATTT

GCTGAGGGCTATTGAGGCGCAACAGCATCTGTTGCAACTCACAGTCTGGGGCATCA

AGCAGCTCCAGGCAAGAATCCTGGCT GT GGAAAGAT ACCT AAAGGATCAACAGCTC

CTGGGGATTTGGGGTTGCTCTGGAAAACTCATTTGCACCACTGCTGTGCCTTGGAAT

GCT AGTTGGAGT AAT AAATCTCTGGAACAGATTT GGAATCACACGACCTGGATGGAG

TGGGACAGAGAAATT AACAATT ACACAAGCTT AAT ACACTCCTT AATT GAAGAATCGC

AAAACCAGCAAGAAAAGAAT GAACAAGAATT ATTGGAATT AGAT AAATGGGCAAGTT

T GT GGAATTGGTTT AACAT AACAAATT GGCT GT GGT AT AT AAAATT ATTCAT AAT GAT A

GT AGGAGGCTTGGT AGGTTT AAGAAT AGTTTTT GCT GT ACTTTCT AT AGT GAAT AGAG

TTAGGCAGGGATATTCACCATTATCGTTTCAGACCCACCTCCCAACCCCGAGGGGA

CCCGACAGGCCCGAAGGAAT AGAAGAAGAAGGTGGAGAGAGAGACAGAGACAGAT

CCATTCG ATT AGT G AACGG AT CTCG ACGGT ATCGGTT AACTTTT AAAAG AAAAGGGG

GGATTGGGGGGTACAGTGCAGGGGAAAGAATAGTAGACATAATAGCAACAGACATA

CAAACT AAA GAATT ACAAAAACAAATT ACAAAAATT CAAAATTTT ATCG AT CACG AG A

CTAGCCTCGAGCACGCGAGTTAATAATTACCAGCGCGGGCCAAATAAATAATCCGC

GAGGGGCAGGTGACGTTTGCCCAGCGCGCGCTGGTAATTATTAACCTCGCGAATAT

TGATTCGAGGCCGCGATTGCCGCAATCGCGAGGGGCAGGTGACCTTTGCCCAGCG CGCGTTCGCCCCGCCCCGGACGGTATCGATAAGCTTAGGAGCTTGGGCTGCAGGT

CGAGGGCACTGGGAGGAT GTT GAGT AAGATGGAAAACT ACT GAT GACCCTTGCAGA

GACAGAGT ATT AGGACAT GTTT GAACAGGGGCCGGGCGATCAGCAGGT AGCTCT AG

AGGATCCCCGTCTGTCTGCACATTTCGTAGAGCGAGTGTTCCGATACTCTAATCTCC

CT AGGCAAGGTT CAT ATTT GTGTAGGTT ACTT ATT CTCCTTTT GTT G ACT AAGTCAAT

AATCAGAATCAGCAGGTTTGGAGTCAGCTTGGCAGGGATCAGCAGCCTGGGTTGGA

AGGAGGGGGTATAAAAGCCCCTTCACCAGGAGAAGCCGTCACACAGATCCACAAGC

TCCTGGCTAGCTAGCATGGCCTCTCACAAGCTGCTGGTGACCCCCCCCAAGGCCCT

GCTCAAGCCCCTCTCCATCCCCAACCGTCTCCTGCTGGGGCCTGGTCCTTCCAACC

TGCCTCCTCGCATCATGGCAGCCGGGGGGCTGCAGATGATCGGGCACATGAGCAA

GGAAAT GT ACCAGATCATGGACGAGATCAAGGAAGGCATCCAGT ACGT GTTCCAGA

CCAGGAACCCACTCACACTGGTCATCTCCGGCTCGGGACACTGTGCCCTGGAGGC

CGCCCTGGTCAATGTGCTGGAGCCTGGGGACTCCTTCCTGGTTGGGGCCAATGGC

ATTTGGGGGCAGCGAGCCGCGGACATCGGGGAGCGCATAGGAGCCCGAGTGCAC

CCGATGACCAAGGACCCCGGAGGCCACTACACACTGCAGGAGGTGGAGGAGGGC

CTGGCCCAGCACAAGCCAGTGCTGCTGTTCTTAACCCACGGGGAGTCGTCCACCG

GCGTGCTGCAGCCCCTTGATGGCTTCGGGGAACTCTGCCACAGGTACAAGTGCCT

GCTCCTGGTGGATTCGGTGGCATCCCTGGGCGGGACCCCCCTTTACATGGACCGG

CAAGGCATCGACATCCTGTACTCGGGCTCCCAGAAGGCCCTGAACGCCCCTCCAG

GGACCTCGCTCATCTCCTTCAGTGACAAGGCCAAAAAGAAGATGTACTCCCGCAAG

ACGAAGCCCTTCTCCTTCTACCTGGACATCAAGTGGCTGGCCAACTTCTGGGGCTG

T G ACG ACCAGCCCAGGAT GT ACC AT CACACAATCCCCGTCAT CAGCCT GT ACAGCC

TGAGAGAGAGCCTGGCCCTCATTGCGGAACAGGGCCTGGAGAACAGCTGGCGCCA

GCACCGCGAGGCCGCGGCGTATCTGCATGGGCGCCTGCAGGCACTGGGGCTGCA

GCTCTTCGTGAAGGACCCGGCGCTCCGGCTTCCCACAGTCACCACTGTGGCTGTAC

CCGCTGGCTAT G ACT GG AGAG ACATCGTCAGCT ACGTCATGG ACCACTTCG ACATT

GAGATCATGGGTGGCCTTGGGCCCTCCACGGGGAAGGTGCTGCGGATCGGCCTGC

TGGGCTGCAATGCCACCCGCGAGAATGTGGACCGCGTGACGGAGGCCCTGAGGG

CGGCCCTGCAGCACT GCCCCAAGAAGAAGCT GT GAGTCGACAATCAACCTCTGGAT

T ACAAAATTT GT GAAAG ATT G ACT GGT ATT CTT AACT AT GTTGCTCCTTTT ACGCTAT

GT GGAT ACGCTGCTTT AATGCCTTT GT ATCAT GCT ATTGCTTCCCGT ATGGCTTTCAT

TTTCTCCTCCTTGTATAAATCCTGGTTGCTGTCTCTTTATGAGGAGTTGTGGCCCGT T

GTCAGGCAACGTGGCGTGGTGTGCACTGTGTTTGCTGACGCAACCCCCACTGGTTG

GGGCATTGCCACCACCTGTCAGCTCCTTTCCGGGACTTTCGCTTTCCCCCTCCCTAT

TGCCACGGCGGAACTCATCGCCGCCTGCCTTGCCCGCTGCTGGACAGGGGCTCGG

CTGTTGGGCACTGACAATTCCGTGGTGTTGTCGGGGAAATCATCGTCCTTTCCTTGG

CTGCTCGCCTGTGTTGCCACCTGGATTCTGCGCGGGACGTCCTTCTGCTACGTCCC TTCGGCCCTCAATCCAGCGGACCTTCCTTCCCGCGGCCTGCTGCCGGCTCTAGATA ATCCAT AAAGT AGG AAACACT ACACG ATTCCAT AAAGT AGGAAACACT AC AACCGGT TCCAT AAAGT AGG AAACACT ACAT CACTCCAT AAAGT AGG AAACACT ACACCCGGTC GAGCTCGGT ACCTTT AAGACCAAT GACTT ACAAGGCAGCT GT AGATCTT AGCCACTT TTTAAAAGAAAAGGGGGGACTGGAAGGGCTAATTCACTCCCAACGAAGACAAGATC TGCTTTTTGCTTGTACTGGGTCTCTCTGGTTAGACCAGATCTGAGCCTGGGAGCTCT CTGGCT AACT AGGGAACCCACT GCTT AAGCCTCAAT AAAGCTT GCCTT GAGTGCTTC AAGTAGTGTGTGCCCGTCTGTTGTGTGACTCTGGTAACTAGAGATCCCTCAGACCCT TTT AGT C AGT GT GG AAAATCTCT AGCA

LV.ET.AGXT-RHEAM.142-3PT map. LV.ET.AGXT-RHEAM.142-3pT construct. Different elements previously described are represented in Figure 12.

AGXT-RHEAM. Amino acid sequence. SEQ ID NO: 52

MASHKLLVTPPKALLKPLSIPNRLLLGPGPSNLPPRIMAAGGLQMIGHMSKEMYQIM DEIKEG

IQYVFQTRNPLTLVISGSGHCALEAALVNVLEPGDSFLVGANGIWGQRAADIGERIG ARVHP

MTKDPGGHYTLQEVEEGLAQHKPVLLFLTHGESSTGVLQPLDGFGELCHRYKCLLLV DSVA

SLGGTPLYMDRQGIDILYSGSQKALNAPPGTSLISFSDKAKKKMYSRKTKPFSFYLD IKWLAN

FWGCDDQPRMYHHTIPVISLYSLRESLALIAEQGLENSWRQHREAAAYLHGRLQALG LQLF

VKDPALRLPTVTTVAVPAGYDWRDIVSYVMDHFDIEIMGGLGPSTGKVLRIGLLGCN ATREN

VDRVTEALRAALQHCPKKKL*

1.2 LV production

Laboratory-grade VSV.G-pseudotyped third-generation SIN LVs were produced by calcium phosphate transient transfection into HEK293T cells. Ten million HEK293T cells were seeded per 150 cm 2 cell culture dish and 24 hours later cells were transfected with a solution containing a mix of LV genome transfer plasmid (LV.ET.AGXT-RHEAM.142-3pT or LV.ET.eGFP.142- 3pT) and packaging plasmids pMDLg/pRRE,pRSV.Rev, and pMD2.VSV.G. The medium was changed 6 hours after transfection and supernatant was collected 2 days later. Usually, a second collection was performed 24 hours after the first one. The collected supernatants were clarified by filtration (0.22 pm PES) and concentrated by centrifugation at 20,000 g for 120 minutes at 16°C. LV pellets were resuspended in an appropriate volume of sodium chloride 0,9 % and preserved at -80°C. 1.3 LV titration

LV.ET.AGXT-RHEAM.142-3pT titration was performed in HepG2, a hepatic cell line, due to the specificity of the target tissue in this strategy. For LV titration, 2- 10 5 cells were seeded in 24 well plates. Twenty-four hours after the seed, cells were transduced with serial dilutions of the LV in culture media and the number of cells at day 0 was determined. Five days after transduction, cells were collected, and genomic DNA (gDNA) was extracted using NucleoSpin® Tissue Kit (Macherey Nagel®) following manufacturer’s instructions.

The number of lentiviral vector genomes (VCN) integrated into transduced cells was determined by qPCR. To analyse the copies of the lentiviral vector genome we used primers (SEQ ID NO: 31 Fw: 5’ CAGGACTCGGCTTGCT GAAG 3’; SEQ ID NO: 32 Rv: 5’ TCCCCCGCTT AAT ACT G ACG 3’) and a probe (SEQ ID NO: 33 5’- CGCACGGCAAGAGGCGAGG-3’) (Taqman®, Thermo Fisher Scientific) designed on the y sequence of the lentiviral vector. To determine the amount of endogenous DNA we used primers (SEQ ID NO: 34 Fw: 5’ GCTGTCATCTCTTGTGGGCTG 3’; SEQ ID NO: 35 Rv: 5’ ACTCATGGGAGCTGCTGGTTC 3’) and a probe (SEQ ID NO: 36 5’

CCTGTCATGCCCACACAAATCTCTCC 3’) (Taqman®, Thermo Fisher Scientific) against the human albumin gene. We also used a standard curve to determine the number of copies of y LV specific sequence and the number of diploid genomes per sample, which allows us to calculate the number of LV copies per cell (VCN = number of copies of y/number of diploid genomes). Finally, based on this parameter we calculated LV title, defined as the number of transducing units per millilitre, with the following formula: number of cells at day 0 · VCN per diploid genome

Title (TU/mL) =

LV volume (mL)

Standard curve to extrapolate qPCR data was performed using serial dilutions of a DNA fragment which contains y and human albumin sequence. All reactions were carried out in duplicate in 7500 Fast Real-Time PCR System (Applied Biosystems®).

For LV.ET.eGFP.142-3pT titration we also transduced HepG2 as well as above. However, at day 3 post-transduction the percentage of GFP-expressing positive cells was analysed by Flow Cytometry. This parameter was used to calculate LV title with the following formula: number of cells at day 0 · % of fluorescence Dilution factor

Title (TU /ml)

100 LV volume (ml) 1.4 Experimental animals

Agxtt 1 mice (B6.129SvAgxt tm1U11 ) were developed by targeted mutagenesis in embryonic stem cells (Salido et al., 2006). Wild type C57BL/6 mice were purchased from The Jackson Laboratory®.

1.5 Animal procedures

LV.ET.AGXT-RHEAM.142-3pT or LV.ET.GFP.142-3pT was administered in adult male mice (12-14 weeks old) by tail vein injection. According to the experiments performed in Agxtt 1 mice, they received a single dose of LV.ET.AGXT-RHEAM.142-3pT (2.5- 10 8 TU/mouse; 5-10 8 TU/mouse; 1 10 9 TU/mouse) or LV.ET.eGFP.142-3pT (1.4-10 8 TU/mouse; 5-10 8 TU/mouse). LV was injected in a total volume of 200 mI/mouse. Wld-type C57BL/6 mice received a single dose of LV.ET.eGFP.142-3pT (T10 8 TU/mouse; 2.5- 10 8 TU/mouse). LV was injected in a total volume of 200 mI/mouse.

Three weeks after LV injection, Agxtt 1 mice were subjected to an ethylene glycol challenge. It has been reported that overloading of oxalate is needed to develop clinical signs of Primary Hyperoxaluria Type 1 in this mouse model. For this reason, 0.5% of ethylene glycol (Ethylene glycol Reagent Plus®, Sigma-Aldrich, REF 102466), a precursor of glyoxylate metabolism, was administered in drinking water for 7 days. During the ethylene glycol challenge, mice were isolated in metabolic cages and 24 hours urine was collected. Moreover, animal body weight was monitored during the whole treatment.

Seven days after the beginning of the ethylene glycol challenge in the case of Agxtt 1 mice or 4 weeks after the LV injection in the case of wild-type C57BL/6 mice, they were sacrificed.

For necropsy mice were anesthetised with ketamine (2.5 mg/20 g mouse) + medetomidine (0.2 mg/20 g mouse). Once unconscious, mice were sacrificed by diaphragm disruption and, therefore, they were infused with 20 ml of PBS1X to remove blood from the tissues to facilitate the following analysis. In sacrificed animals, different tissues were collected (liver, spleen, lungs, bone marrow, lymph nodes, thymus, brain, testicles, pancreas, and kidneys).

All animal procedures in which Agxtt 1 mice were involved were performed at CIMA (Centro de Investigation Medica Aplicada) in Pamplona. All experimental procedures were approved by the Ethics Committee of the University of Navarra and the Institute of Public Health of Navarra according to European Council Guidelines. All the experiments using animal models complied with all relevant ethical regulations. All animal procedures in which C57BL/6 mice were involved were performed at CIEMAT (Centro de Investigaciones Energeticas y Medioambientales y Tecnologicas) in Madrid. All experimental procedures were approved in PROEX 165-18 by Direccion General de Medio Ambiente of Comunidad de Madrid.

1.6 Determination of VCN in LV injected mice

After animals’ sacrifice, different tissues were collected (liver, spleen, lungs, bone marrow, lymph nodes, thymus, brain, testicles, pancreas, and kidneys) and preserved at -80°C. For the determination of LV genomes integrated into these tissues, genomic DNA was extracted with the NucleoSpin® Tissue Kit (Macherey Nagel) following the manufacturer’s instructions.

The number of integrated lentiviral vector genomes (VCN) in the injected mice tissues was determined by qPCR. To analyse the number of lentiviral vector genomes we used primers (SEQ ID NO: 37 Fw: 5’ CAGGACTCGGCTTGCT GAAG 3’; SEQ ID NO: 38 Rv: 5’ TCCCCCGCTT AAT ACT G ACG 3’) and a probe (SEQ ID NO: 39 5’-

CGCACGGCAAGAGGCGAGG-3’) (Taqman®, Thermo Fisher Scientific) designed on the y sequence of the lentiviral vector. To determine the amount of endogenous DNA we used primers (SEQ ID NO: 40 Fw: 5’ AAAACGAGCAGT GACGT GAGC 3’; SEQ ID NO: 41 Rv: 5’ TTCAGTCATGCTGCTAGCGC 3’) and a probe (SEQ ID NO: 42 5’-

TGCACGGAAGCGTCTCGTCTCAGTC-3’) against the mouse titin gene. Once determined the number of LV copies and the number of diploid genomes per sample, VCN was calculated (VCN = number of copies of y/number of diploid genomes).

A standard curve was performed using serial dilutions of a DNA fragment that contains y and the titin sequences. All reactions were carried out in duplicate in 7500 Fast Real-Time PCR System (Applied Biosystems®).

1.7 Determination of AGXT expression

To analyse the functionality of therapeutic lentiviral vector in injected Agxtt 1 mice, we determined transgene expression in the liver of these mice. To define this parameter, RNA from the liver was extracted by Trizol protocol. Briefly, liver fragments were homogenized in Trizol, after which we added chloroform to separate the aqueous phase. RNA was precipitated with isopropanol and glycogene, cleaned with ethanol 70%, and finally resuspended in free nucleases water. To assure the lack of genomic DNA in RNA samples, they were digested with DNase using DNase Max® Kit (Qiagen) following the manufacturer’s instructions. RNA samples were maintained at -80°C.

Afterward, RT-PCR technique was performed and cDNA was synthesized from RNA samples by RETROscript® Kit (Ambion, USA) following the manufacturer’s instructions. Transgene expression was determined by qPCR. We designed primers against the AGXT- RHEAM (SEQ ID NO: 43 Rv: 5’ GTCTTGCGGGAGTACATCTT 3’; SEQ ID NO: 44 Fw: 5’ CAAGGCATCGACATCCTGTA 3’) and two housekeeping genes, mouse Tbp (SEQ ID NO: 45 Rv: 5’ GATGGGAATTCCAGGAGTCA 3’; SEQ ID NO: 46 Fw: 5’ GGGAGAATCATGGACCAGA 3’) and mouse Actb (SEQ ID NO: 47 Rv: 5’ OTA AGG CCA ACC GTG AAA AG 3’; SEQ ID NO: 48 Fw: 5’ ACC AGA GGC ATA CAG GGA CA 3’). All reactions were carried out in duplicate in Fast Real-Time PCR System (Applied Biosystems®). Quantification of relative expression of the transgene was performed by AACt analysis (Schefe et al., 2006) referenced to two housekeeping genes, mouse Tbp and mouse b-actin, separately.

We also analysed mouse Agxt physiological expression in the liver of non-injected C57BL/6 mice. RNA was extracted from liver samples of non-injected wild-type mice and RT-PCR was performed as explained above. To analyse gene expression, primers against mouse Agxt gene were designed (SEQ ID NO: 49 Rv: 5’ GACAAAGCCAGTCTCCTTCTAC 3’; SEQ ID NO: 50 Fw: 5’ GTGACAGGTGTGGTATGATGAA 3’). For the expression analysis, mouse Tbp and mouse Actb were also measured by RT-qPCR. All reactions were carried out in duplicate in Fast Real-Time PCR System (Applied Biosystems®). Quantification of relative expression of the transgene was performed by AACt analysis (Schefe et at., 2006) referenced to two housekeeping genes, mouse Tbp and mouse Actb, separately.

1.8 Determination of the percentage of transduced cells by immunostaining.

To analyse the percentage of transduced cells in the target tissue we performed an immunostaining assay to detect eGFP positive cells in the liver of LV.ET.eGFP.142-3pT injected mice.

From paraffin sections, samples were deparaffinized by incubation in xylene and decreased ethanol dilutions. Afterward, samples were incubated in blocking solution (10% of donkey serum in PBS 1X) for one hour and overnight with Goat-aeGFP (ab6673 abeam®) 1:100 and Rabbit- amsALB (AHP1478) 1:500 in blocking solution. The following day, samples were incubated with secondary antibodies (Donkey-aGoat AF ® 488 (A11055 invitrogen ® ), Donkey- aRabbit AF ® 488 (A21206 invitrogen ® ), Donkey-aGoat AF ® 594 (A11058 invitrogen ® )) 1 :1000 and DAPI 1:1000 in blocking solution for 1 hour. Subsequently, samples were mounted with Mowiol® and visualized in a Zeiss Axiolmager microscope. Images of each sample were acquired. Determination of eGFP positive cells in liver immunostaining samples was performed by QuPath® software. The percentage of eGFP expressing cells with respect to the total of hepatocytes in each image was determined. At least ten different fields per sample were analysed (400x magnification).

1.9 Urine oxalate determination

Twenty four-hour urine from mice was collected to measure the total amount of oxalate removed from the organism in this period. Individual 24-h urines were collected in 50 pi of 5 N HCI and their volume was measured. Urine samples were clarified with activated charcoal and oxalate was measured by enzymatic determination by oxalate oxidation using Oxalate Kit (Trinity Biotech, REF 591 -D) following the manufacturer’s instructions. To determine the concentration of oxalate (pmol) in each sample, a reference curve of known concentrations of oxalate was performed (Oxalate Standard 0.5 mmol/l REF 591-3 Trinity Biotech). Finally, oxalate quantification (pmol/24 h) was calculated using the oxalate concentration and the urine volume.

1.10 Kidney Damage determination: Kidney Damage Score

After animals’ sacrifice, kidneys were fixed in formalin and embedded in paraffin. Then, 5pm sections were obtained and stained with haematoxylin-eosin. To determine the damage produced we developed a “Kidney Damage Score” in which the stage of each sample was classified into 4 different stages according to the features observed:

• 0: no damage

• 1: Normal architecture, but the presence of some damage signs such as dilated ducts.

• 2: The architecture is partially affected and there is an increase in the presence of dilated ducts.

• 3: Tissue architecture is highly affected, and we can observe the presence of calcium oxalate crystals, indicating a nephrocalcinosis stage.

1.11 Transduction enhancers: Cyclosporin H in vitro testing.

To increase in vivo transduction efficacy after the treatment with the developed lentiviral vector we tested the effect of a transduction enhancer, Cyclosporin H (CsH), during the in vitro transduction protocol. Two different regimens of CsH were tested. Due to the important relevance of the target tissue origin, HepG2 cells, a hepatic cell line, were used. For the first CsH regimen, 2-10 5 cells were seeded in a 24-well plate. One day later, cells were transduced with a culture media solution containing LV.ET.eGFP.142-3pT at a MOI of 0.3 and CsH. Different CsH concentrations were tested (4, 8, and 16 mM). Three days after transduction, the percentage of eGFP positive cells was analysed by Flow Cytometry. All conditions were performed as triple experimental replicates.

For the second CsH regimen, 2-10 5 cells were seeded in a 24-well plate. One day later, culture media was changed and cells were pre-incubated with culture media supplemented with 16 pM CsH. 16 hours later, cells were transduced with a solution containing LV.ET.eGFP.142-3pT at an MOI of 0.3 and 16 pM CsH. Three days after transduction, the percentage of eGFP positive cells was analysed by Flow Cytometry. All conditions were performed as triple experimental replicates.

1.12 Statistical analysis

Significant differences between groups were determined by the Mann-Whitney test, a non- parametric test that is used to compare two independent groups without Gaussian distribution. Analyses were performed with Graphpad Prism ® software. Significative values were considered when p<0.05.

1.13 Transduction enhancers: Dexamethasone in vivo testing

To increase the transduction efficacy of the lentiviral vector of the invention, we tested the combination of the lentiviral vector administration with an inhibitor of the innate immune response, dexamethasone. The analysis was performed in C57BL/6 male adult mice.

C57BL/6 mice received a single dose of LV.ET.eGFP.142-3pT (5-10 8 TU/mouse) by tail vein injection. Additionally, they were treated with 5 mg/kg of body weight of dexamethasone (Fortecortin®), intraperitoneally injected in three consecutive administrations: twelve and two hours before the lentiviral vector injection and four hours after. A control group only injected with the lentiviral vector was also included in the experiment.

Four weeks later mice were sacrificed and the transduction efficacy analysis (lentiviral vector integrations per diploid genome and percentage of eGFP positive cells in the liver) was performed. EXAMPLE 2: Lentiviral vector efficiently transduces the liver in C57BL/6 mice and Agxt1 ~ ' ~ mouse model after intravenous injection.

First, as a proof of concept to study the feasibility of the developed gene therapy tool and to perform a transduction percentage and biodistribution analysis, we carried out an in vivo experiment in which adult C57BL/6 male mice were injected with a single dose of LV.ET.GFP.142-3pT (Figure 1a, Figure 2a). We tested two different doses (1 10 s TU/mouse; 2.5- 10 8 TU/mouse). We observed 0.55 (0.34-0.56) lentiviral vector copies per diploid genome in C57BL/6 injected mice with a dose of T10 8 TU/mouse, whereas in C57BL/6 mice injected with a dose of 2.5- 10 8 TU/mouse, 1.03 (0.94-1.56) lentiviral vector copies per diploid genome were obtained (Figure 3). Immunostaining analysis reveals that this corresponds to 1.751 (1.13-5.55) % eGFP positive hepatocytes and 12.92 (10.72-13.81) % eGFP positive hepatocytes in the low and high dose, respectively (Figure 5). Representative images of these mice can be observed in Figure 5 (A-C; F-H).

Next, in order to further characterize the in vivo transduction efficiency of the lentiviral vector LV.ET.GFP.142-3pT and compare it with the transduction efficiency of the same lentiviral vector in vitro, HepG2 cells were transduced with LV.ET.eGFP.142-3pT at different Multiplicity of Infection (MOI). Three days after transduction, the percentage of eGFP positive cells was determined by flow cytometry. Using MOIs of about 1 , a transduction percentage of 50% was achieved, whereas higher MOIs achieved 100% transduction in vitro. For in vivo transduction efficacy determination, adult C57BL/6 (Fig. 13, filled circle) or Agxtt 1 mice (Fi. 13, open circle) were intravenously injected with LV.ET.eGFP.142-3pT at different doses (T10 8 TU/mouse, 1.4-10 8 TU/mouse, 2- 10 8 TU/mouse and 5-10 8 TU/mouse). Corresponding MOIs were calculated only according to the estimated number of hepatocytes in the liver of these mice as previously described (Park et al., 2000; Schmitt et al., 2010). Therefore, the mice were injected with the following MOIs: 0.8, 1.1, 2, and 4. Percentage of transduced hepatocytes was determined 4 weeks later in liver histology samples. The results depicted in Fig. 13 show that there is a significant reduction in the transduction efficiency of this lentiviral vector when it is used in vivo in comparison with in vitro experiments.

Therefore, to test the feasibility of an in vivo administration of a therapeutic lentiviral vector as a gene therapy strategy to treat Primary Hyperoxaluria Type 1, adult male Agxtl^ mice were intravenously injected with a single dose of LV.ET.AGXT-RHEAM.142-3pT lentiviral vector (Figure 1B). Three different lentiviral vector doses (2.5- 10 8 TU/mouse; 5- 10 8 TU/mouse; T10 9 TU/mouse) were tested. Three weeks after injection, mice were subjected to an ethylene glycol challenge for 7 days (Figure 2B) and then sacrificed. The number of genome lentiviral vector copies integrated in the liver was determined at sacrifice. There was a dose-related efficacy in liver transduction. Liver VCN achieved in the three different groups was 0.615 (0.39-1.46) in Agxtt 1 mice which received 2.5- 10 8 TU/mouse; 0.765 (0.45-1.19) in Agxtt 1 mice which received 5-10 8 TU/mouse and 1.165 (0.85-0.32) in Agxtt 1 mice which received 10-10 8 TU/mouse (Figure 3). We also analysed this parameter in non-injected mice and, as expected, we did not detect any copy of the lentiviral vector genome in those animals.

As control groups, we also injected Agxtt mice with a reporter lentiviral vector. Two different LV doses were tested (1.4-10 8 TU/mouse; 5-10 8 TU/mouse). We also observed a dose- related transduction efficacy of the liver. In low-dose injected mice we obtained a VCN in the liver of 0.11 (0.09-0.49); meanwhile, in the high-dose, we observed a liver VCN of 0.84 (0.73- 1.58) (Figure 3). Furthermore, reporter LV transduction efficacy was comparable to the efficacy achieved with the therapeutic lentiviral vector in Agxtt 1 mice.

Intending to define the percentage of transduced cells achieved after lentiviral vector injection, we also analysed the percentage of positive cells in liver sections of LV.ET.eGFP.142-3pT Agxtt 1 injected mice (Figure 5). In Agxtt mice which received 1.4-10 8 TU/mouse we observed 1.777 (0.399-4.572) % eGFP positive hepatocytes, whereas in Agxtt mice which received 5-10 8 TU/mouse we observed 5.5059 (2.109-8.754) % eGFP positive hepatocytes. Representative images of these mice are provided in Figure 5 (D, E). Since reporter lentiviral vector and therapeutic lentiviral vector seems to function in a similar way, it was expected that in Agxtt 1 mice which received 5- 10 8 TU/mouse of the therapeutic lentiviral vector we had achieved a transduction percentage similar to the observed in Agxtt 1 mice which received the same dose of reporter lentiviral vector.

Differences in terms of transduction efficacy between Agxtt 1 and wild-type C57BL/6 injected mice could be due to the genetic background of the mice or to the variability in title determination of LV lab-grade batches.

EXAMPLE 3: Therapeutic lentiviral vector is efficiently expressed in treated Agxtt 1 mice liver.

Although we checked the presence of therapeutic lentiviral vector in the target tissue of therapeutic lentiviral vector Agxtt 1 injected mice, we wanted to test if this construct was expressing efficiently. We analysed transgene expression in treated Agxtt 1 mouse livers by RT-qPCR. As reference, two different housekeeping genes were used for this analysis (ms Tbp and msActb) to determine properly the differences between a physiological mouse Agxtl expression in C57BL/6 wild-type mice compared with the AGXT expression induced by lentiviral vector integration after the in vivo gene therapy strategy in Agxtl 1 mice.

Therapeutic lentiviral injected mice which received a dose of 2.5- 10 8 TU/mouse showed an AGXT expression of 10.88 % (5.62-16.79) referred to ms Tbp expression and of 0.1041 % (0.06007-0.2452) referred to msActb. Agxtl 1 mice which received a dose of 5- 10 8 TU/mouse resulted in an AGXT expression of 12.11 % (4.38-14.48) referred to ms Tbp and of 0.1857 % (0.08878-0.2966) referred to msActb. Agxtl 1 therapeutic lentiviral vector injected mice with T10 9 TU/mouse showed an AGXT expression of 19.88 % (18.52-28.55) referred to ms Tbp and of 0.3088 % (0.1707-0.4418) referred to msActb (Figure 4). Thus, differences in AGXT expression levels among the different therapeutic lentiviral vector Agxtl 1 injected mice correlated with the dose received. Non-injected Agxtl 1 mice did not show AGXT expression.

Moreover, to study the differences between the achieved AGXT expression after therapeutic lentiviral vector injection and the physiological mouse Agxtl expression, we analysed this expression in wild-type C57BL/6 mice. Wild type C57BL/6 mice showed a mouse Agxtl expression of 8563 (6996-8939) referred to ms Tbp and of 340.5 (141-374.5) referred to msActb, strikingly higher than the one achieved in Agxtl 1 treated mice.

EXAMPLE 4: In vivo lentiviral vector injection into C57BL/6 mice has a reduced off- target transduction.

To determine the security of the proposed therapeutic strategy, the number of lentiviral vector integrated genomes was analysed in C57BL/6 wild type mice injected with two different doses of the reporter lentiviral vector. Number of LV genome copies integrated in different tissues of injected wild-type C57BL/6 mice was analysed by qPCR. Integration of reporter lentiviral vector genome was only observed in liver, the target tissue, and in lungs, spleen and bone marrow, that could occur due to the route of administration used in this strategy. C57BL/6 mice injected with T10 8 TU/mouse showed a VCN of 2.72 (2.33-6.2) in lungs, 0.21 (0.14-0.32) in spleen, and 0.05 (0.04-0.09) in bone marrow. Mice injected with a higher dose of 2.5- 10 8 TU/mouse showed a VCN value of 4.94 (0.185-7.79) in lungs, 0.35 (0.25-0.55) in spleen and 0.13 (0.09-0.18) in bone marrow (Figure 6). Although the presence of integrated vector genomes in these tissues was observed, reporter gene expression was not observed by immunofluorescence analysis, indicating a reduction in the derived risk from off-target transduction. Further, no signs of tumor development were observed. EXAMPLE 5: Therapeutic lentiviral vector treated Agxtt 1 mice experience a reduction in urine oxalate levels.

To get a closer phenotype to the human one, Primary Hyperoxaluria Type 1 pathological phenotype analysis in Agxtt 1 mouse model requires an ethylene glycol challenge. This challenge generates overloading in oxalate production, which is needed to analyse the pathological phenotype of these mice.

Over the seven days of ethylene glycol challenge, mice were isolated in periods of 24 hours and 24-hour urine was collected. Oxalate concentration in urine was measured at three different time points, before the beginning of the treatment (basal measurement) and at days 3 and 7 of the treatment.

To analyse the pathological phenotype, three different therapeutic lentiviral vector doses were used, 2.5-10 8 TU/mouse, 5-10 8 TU/mouse and 10-10 8 TU/mouse. In addition, we included three different control groups: two negative control groups composed of Agxtt 1 mice injected with PBS or with a reporter lentiviral vector (LV.ET.GFP.142-3pT), and a positive control group composed of C57BL/6 wild type mice. All mice groups were subjected to ethylene glycol challenge.

Before treatment, urine oxalate concentration was low in all the conditions. After the beginning of the ethylene glycol challenge an increase in oxalate urine concentration was observed in all the groups. However, the increase observed was higher in untreated Agxtt 1 group than in the groups treated with the therapeutic lentiviral vector. Moreover, the dispersion of the data related to this value, a specific feature of this mouse model, was higher in the untreated Agxtt 1 mice than in the LV treated Agxtt 1 mice. Urine oxalate concentration in Agxtt 1 mice which received 5-10 8 TU/mouse of the therapeutic lentiviral vector was significantly lower at days 3 and 7 than urine oxalate concentration in untreated Agxtt 1 mice groups. Significantly, at day 7 there were no significant differences between the therapeutic LV injected Agxtt 1 mice with 5-10 8 TU/mouse and the wildtype C57BL/6 group (Figure 7).

EXAMPLE 6: Treated mice gain weight during ethylene glycol challenge.

Another important parameter to follow the pathological phenotype in Agxtt 1 mice is the gain or loss of animals’ weight during the ethylene glycol challenge. Regarding this parameter, we analysed the weight development in all the mice at the two referred time points and we compared it with the weight measurement at a basal point. We observed a reduction in animal body weight of untreated control groups. This reduction was not observed in the therapeutic LV treated group. Moreover, a gain of weight was achieved (Figure 8). On day 7 after ethylene glycol challenge, the increase in weight in therapeutic LV injected Agxtt 1 mice with 5-10 8 TU/mouse and with 10- 10 s TU/mouse was not significantly different to the C57BL/6 wild-type mice.

Example 5 and 6 showed that, although the AGT expression observed in the corrected cells is much lower than the observed in normal hepatocytes, the phenotype correction obtained in the treated animals is unexpectedly much higher. Thus, an unexpected phenotype reversion degree was found considering the hypothesis of “more than 40% cells should be corrected to achieve a therapeutic effect”. In the most efficient group, percentage of transduction is not higher than 20% (see figure 5 with reporter LV).

EXAMPLE 7: There is a reduction in nephrocalcinosis development in treated mice.

One of the most important clinical signs of Primary Hyperoxaluria Type 1 patients is the development of nephrocalcinosis due to the calcium oxalate crystals formation in kidney parenchyma. To determine the damage in the animals’ kidneys, we developed a kidney damage score, where 0 represented no damage, 1 represents a mild damage in kidney parenchyma, 2 represents moderate damage which can turn into stage 3 in which we can observe clear signs of nephrocalcinosis, to classify the damage generated because of the ethylene glycol treatment. We classified each animal regarding this score. Regarding this classification, there was a higher percentage of mice, which develop nephrocalcinosis in non- treated groups compared to the treated ones. In therapeutic LV injected Agxtt 1 mice with 5-10 8 TU/mouse we did not observe any mouse which develops nephrocalcinosis (Figure 9A).

If we simplify this score into none or mild kidney damage (score 0-1) and moderate or severe kidney damage (score 2-3), which represents mice in which kidney damage is progressing due to the ethylene glycol challenge, we could observe than in untreated Agxtt 1 mice, the kidney damage progression occurs in a quarter of the half of the cases, as well as it occurs in therapeutic lentiviral vector injected Agxtt 1 mice with doses of 2.5- 10 8 TU/mouse or 10- 10 s TU/mouse. Meanwhile in therapeutic lentiviral vector injected Agxtt 1 mice with 5-10 8 TU/mouse only one out of ten treated mice developed kidney damage progression, and none of the mice injected with the middle dose (5-10 8 ) developed nephrocalcinosis (Figure 9B). Table 1 : Summary of values obtained in Fig. 9B

EXAMPLE 8: The use of Cyclosporin H in lentiviral transduction protocol increases the transduction efficacy in a hepatic cell line in vitro.

In order to increase the amount of corrected hepatocytes without increasing amount of virus dose, that could be not effective as observed in the previous experiments, we looked for potential transduction enhancers to optimize LV transduction. Among others Cyclosporin H had been described to facilitate the in vitro LV transduction in hematopoietic stem cells (HSCs), in which IFITM3, the target protein of CsH, is highly expressed under basal condition. However, no data related to hepatocytes as target cells had been suggested or tested. With the objective of testing this possibility, hepatic HepG2 cells were transduced with two different administration regimens of LV.ET.eGFP.142-3pT and at different CsH concentrations.

In the first transduction protocol, HepG2 cells were subjected to different concentrations of CsH (4, 8, or 16 mM) during the lentiviral vector transduction protocol. Cells were transduced with a lentiviral vector MOI of 0.3. Three technical replicates of each condition were performed. Basal transduction levels were 36.67% and in the different CsH treated samples a transduction percentage of 37.67%, 44.3%, and 51.5% in 4, 8, and 16 mM CsH conditions were observed, which means a fold-increase of control samples of 1.03, 1.2, and 1.4 respectively (Figure 10).

In the second CsH administration regimen, HepG2 cells were subjected to 16 hours of CsH pre-treatment before transduction, in which CsH was also included. The only CsH concentration tested was 16 mM. In this CsH administration regimen, a fold-increase in eGFP positive cells of 2.35 to the control sample was observed (Figure 10). Cells were transduced with an MOI of 0.3 of LV.ET.eGFP.142-3pT and the percentage of transduced cells achieved were 13.2% in the control condition and 31.03% in the CsH treated cells. The differences observed in transduction levels between the control conditions at the same MOI are due to a probable reduction in LV title due to thaw-freeze cycles. The two different administration regimens were independent experiments.

Thus, remarkably, addition of the CsH as transduction enhancer increases the transduction efficiency of the developed lentiviral vectors (in vitro) in a hepatic cell line such as HepG2 even though CsH target gene, IFITM3, is not supposed to be expressed in hepatic lineage.

EXAMPLE 9: Dexamethasone treatment improves the transduction efficacy in the liver of in vivo reporter lentiviral vector treated C57BL/6 mice.

The use of additional compounds may be a valuable tool to increase the amount of transduced hepatocytes, which would in turn improve the therapeutic efficacy of the treatment with the lentiviral of the invention. Further, the immune response mediated by type 1 interferon (IFN-1) that lentiviral vectors can trigger can also hamper the transduction efficacy, and thus the use of inhibitors of said immune response also represents a potential combined treatment after the in vivo administration of the lentivial vector. For this reason, we carried out an experiment in which C57BL/6 mice were treated with dexamethasone (following the protocol described by Agudo et al. 2012, Molecular Therapy vol. 20 no. 12, 2257-2267) in combination with the lentiviral vector.

C57BL/6 mice were treated with a single dose of LV.ET.eGFP.142-3pT (5-10 8 TU/mouse) by tail vein injection. Additionally, they were intraperitoneally injected with 5 mg/kg of body weight of dexamethasone (Fortecortin®), in three consecutive administrations: twelve and two hours before the lentiviral vector injection and four hours after. Four weeks after the combined treatment mice were sacrificed (Figure 14A).

In mice treated with the combination of dexamethasone and the reporter lentiviral vector, we observed 0.92 (0.38-1.4) integrated copies per diploid genome whereas in mice treated only with the lentiviral vector 1.48 (1.2-1.76) integrated copies per diploid genome were detected, a significant increase compared with mice treated with the combined treatment (Figure 14B). However, immunostaining analysis reveals a 4.08 fold increase in the percentage of eGFP positive hepatocytes in mice treated with the combined treatment with respect to the mice treated only with the lentiviral vector with 18.11 (6.381-19.79) % eGFP positive hepatocytes and 4.434 (2.167-7.55) % eGFP positive hepatocytes, respectively. Representative images of these mice can be observed in Figure 14 D-E. These results suggest that the combination of dexamethasone and LV.ET.AGXT- RHEAM.142-3pT in the context of Agxtl-/- mice would improve the transduction efficacy and, plausibly, the therapeutic effect, supporting the feasibility of the lentiviral vector based-in vivo strategy for the treatment of PH1 patients.