Abstract
Mucopolysaccharidosis II (MPS II) is a rare lysosomal storage disease characterized by deficient activity of iduronate-2-sulfatase (I2S), leading to pathological accumulation of glycosaminoglycans (GAGs) in tissues. We used iduronate-2-sulfatase knockout (Ids KO) mice to investigate if liver-directed recombinant adeno-associated virus vectors (rAAV8-LSP-hIDSco) encoding human I2S (hI2S) could cross-correct I2S deficiency in Ids KO mouse tissues, and we then assessed the translation of mouse data to non-human primates (NHPs). Treated mice showed sustained hepatic hI2S production, accompanied by normalized GAG levels in somatic tissues (including critical tissues such as heart and lung), indicating systemic cross-correction from liver-secreted hI2S. Brain GAG levels in Ids KO mice were lowered but not normalized; higher doses were required to see improvements in brain histology and neurobehavioral testing. rAAV8-LSP-hIDSco administration in NHPs resulted in sustained hepatic hI2S production and therapeutic hI2S levels in cross-corrected somatic tissues but no hI2S exposure in the central nervous system, perhaps owing to lower levels of liver transduction in NHPs than in mice. Overall, we demonstrate the ability of rAAV8-LSP-hIDSco to cross-correct I2S deficiency in mouse somatic tissues and highlight the importance of showing translatability of gene therapy data from rodents to NHPs, which is critical for supporting translation to clinical development.
Keywords: gene therapy, AAV, MPS II, cross-correction, non-human primates, iduronate-2-sulfatase, Hunter syndrome, lysosome
Graphical abstract

Chen and colleagues show that gene therapy for mucopolysaccharidosis II through administration of a liver-directed recombinant adeno-associated virus vector encoding human iduronate-2-sulfatase (rAAV8-LSP-hIDSco) is able to cross-correct iduronate-2-sulfatase deficiency in somatic tissues of Ids knockout mice and demonstrate promising translational potential from the mouse model to non-human primates.
Introduction
Mucopolysaccharidosis II (MPS II; or Hunter syndrome [OMIM: 309900]) is a rare, X-linked, progressive, lysosomal storage disease.1 The disease is caused by pathogenic variants of the iduronate-2-sulfatase gene (IDS), which encodes iduronate-2-sulfatase (I2S), a lysosomal enzyme responsible for catalyzing the hydrolysis of C2-sulfate groups of the glycosaminoglycans (GAGs) dermatan sulfate (DS) and heparan sulfate (HS). Deficient activity of I2S leads to pathological accumulation of GAGs throughout the body.2
Although a continuum of different phenotypes can be observed, MPS II is generally classified into two clinical forms: neuronopathic and non-neuronopathic.1 Somatic clinical disease manifestations are present in all patients and can include organ failure, coarsening of facial features, bone and joint abnormalities, and short stature.3 Neuronopathic disease affects approximately two-thirds of patients with MPS II; in addition to somatic disease manifestations, these patients also experience profound central nervous system (CNS) involvement and cognitive impairment.1,3
The current standard of care for MPS II is enzyme replacement therapy (ERT) with intravenous recombinant human I2S (hI2S; idursulfase, Takeda Pharmaceuticals USA, Lexington, MA, USA).1 ERT with intravenous idursulfase is effective at improving or stabilizing many somatic disease manifestations4; however, it has some limitations, including fluctuating enzyme levels, the patient burden of weekly infusions, and limited penetration into some deep somatic tissues such as heart, lung, and bone.5,6,7 Moreover, the high molecular weight (76 kDa) of hI2S and the absence of an active mechanism to cross the blood-brain barrier means that the clinical dose given systemically does not provide exposure to the brain to address neurological symptoms.5,6
MPS II is a monogenic disease with relatively well-known pathological mechanisms, making it a potential candidate for gene replacement therapy.8 Recombinant adeno-associated virus (rAAV) is considered the delivery tool of choice for in vivo gene therapy because it is generally well tolerated, can provide long-term expression, and shows low immunogenicity.9 Gene therapy with rAAV-based vectors offers the possibility of long-term sustained expression of hI2S and the potential to normalize GAG levels in certain deep somatic tissues in patients with MPS II.
The successful development of intravenous ERT for MPS II included use of an integrated approach involving pharmacokinetic, biodistribution, and safety studies in rodents and non-human primates (NHPs).10 We developed a similarly integrated approach to investigate gene therapy for MPS II, using appropriate mouse models together with NHP studies to support the identification of a robust preclinical pharmacology framework for gene therapy optimization and translation into the clinic.
Here, we describe rAAV8-LSP-hIDSco, a gene therapy vector comprising an adeno-associated virus serotype 8 capsid and a genome containing a liver-specific expression cassette encoding hI2S, using the liver as the in vivo hI2S production center (Figure 1A). We evaluated the ability of rAAV8-LSP-hIDSco gene therapy to cross-correct I2S deficiency in somatic and CNS tissues of Ids knockout (Ids KO) mice and the translation of these mice data to NHPs.
Figure 1.
Vector schematic and Ids KO mice pharmacology 3 months after treatment with rAAV8-LSP-hIDSco
(A) rAAV8-LSP-hIDSco vector schematic. (B–G) Ids KO mice were treated with a single intravenous injection of rAAV8-LSP-hIDSco at four dose levels ranging from 2.5 × 108 vg/kg to 2.5 × 1011 vg/kg and monitored for 3 months. An additional cohort was dosed with 2.5 × 1011 vg/kg of a null vector, rAAV8-MY011, as a negative control. (B) Serum and (C) somatic tissues and the CNS were assayed for I2S activity. (D) Somatic tissues and the CNS were assayed for HS content and (E) DS content. (F) Somatic tissues and (G) the CNS were assayed by IHC for LAMP1 positivity. On all plots, data are presented as mean ± SD, n = 8 per group, except for the naive WT group (n = 5). p values for rAAV8-LSP-hIDSco-treated animals compared with naive Ids KO were generated by one-way analysis of variance (ANOVA) with multiple comparisons using Dunnett’s correction. ∗p < 0.05; ∗∗p < 0.01; ∗∗∗p < 0.001; ns, not significant versus naive Ids KO. ANOVA, analysis of covariance; CNS, central nervous system; DS, dermatan sulfate; I2S, iduronate-2-sulfatase; hI2S, human iduronate-2-sulfatase; hIDSco, codon-optimized human iduronate-2-sulfatase gene; HS, heparan sulfate; Ids KO, iduronate-2-sulfatase gene knockout; IHC, immunohistochemistry; ITR, inverted terminal repeat; LAMP1, lysosome-associated membrane protein 1; LSP, liver-specific promoter; Poly A, polyadenylation; rAAV8, recombinant adeno-associated virus 8; vg, vector genome; WPRE, woodchuck hepatitis virus post-transcriptional regulatory element; WT, wild-type.
Results
Effect of rAAV8-LSP-hIDSco on hI2S expression and disease-associated parameters at 3 months after dosing in Ids KO mice
To assess the ability of rAAV8-LSP-hIDSco to cross-correct I2S deficiency, Ids KO mice (5–7 weeks of age) were treated with four rAAV8-LSP-hIDSco doses ranging from 2.5 × 108 vector genome (vg)/kg to 2.5 × 1011 vg/kg via tail vein injection. An additional Ids KO mice cohort was dosed with a null vector, rAAV8-MY011 (2.5 × 1011 vg/kg), as a negative control. Age-matched naive Ids KO and wild-type (WT) mice were also included as controls.
At 3 months after dosing, I2S activity and hI2S concentration in serum and tissues had a dose-dependent increase in mice treated with rAAV8-LSP-hIDSco (Figures 1B, 1C, S1, and S2). As expected, tissue I2S activity in rAAV8-MY011-treated KO mice (<1 nmol/h/mg) was similar to that in naive Ids KO mice. I2S activity in the group treated with the lowest dose (2.5 × 108 vg/kg) was similar to that in naive Ids KO mice in all tissues except for liver. The rAAV8-LSP-hIDSco dose groups had increasing I2S activity with increasing dose (2.5 × 109 vg/kg to 2.5 × 1011 vg/kg) in liver, spleen, kidney, heart, lung, bone marrow, quadriceps, skin, and brain tissues (Figure 1C). Differences in I2S activity between naive Ids KO mice and rAAV-LSP-hIDSco-treated mice reached significance at different dose levels in tissues, namely at 2.5 × 108 vg/kg or higher in the liver, at 2.5 × 109 vg/kg or higher in the spleen, and at 2.5 × 1010 vg/kg or higher for all other somatic tissues except the lung, in which significance was achieved only with the 2.5 × 1011 vg/kg dose. In Ids KO mice treated with the 2.5 × 1011 vg/kg dose, I2S activity was greater in all somatic tissues than in WT mice, except for the brain, where activity was 10% of that in WT mice.
To quantify HS and DS, GAG levels were measured in tissues using a liquid chromatography with tandem mass spectrometry (LC-MS/MS) assay non-conforming with Good Laboratory Practice (GLP) guidelines. HS and DS concentrations saw similar patterns of decrease with increasing rAAV-LSP-hIDSco dose across all tissues except for the kidney (Figures 1D and 1E). In the kidney, DS concentrations for the 2.5 × 1010 vg/kg and 2.5 × 1011 vg/kg doses decreased to a greater extent than HS; however, this did not affect statistical significance, which was equivalent for HS and DS. Significant HS and DS reductions were observed in all somatic tissues at 2.5 × 1010 vg/kg and 2.5 × 1011 vg/kg doses compared with naive Ids KO mice. The 2.5 × 1010 vg/kg dose achieved over 90% HS and DS reduction in somatic tissues and the 2.5 × 1011 vg/kg dose normalized levels to WT. In mice treated with the 2.5 × 1011 vg/kg dose, brain HS and DS levels were approximately 60% and 20%, respectively, of those recorded in naive Ids KO mice (Figure S3). Immunohistochemistry (IHC) analyses were performed to assess changes in lysosome-associated membrane protein 1 (LAMP1), a lysosomal membrane marker that is elevated in lysosomal disease model animals.11 In rAAV8-LSP-hIDSco-treated mice, there was a significant dose-dependent reduction in detectable LAMP1 compared with naive Ids KO mice in somatic tissues (Figure 1F) but not in the brain (Figure 1G).
Effect of rAAV8-LSP-hIDSco on hI2S expression and disease-associated parameters at 12 months after dosing in Ids KO mice
To evaluate the long-term effects of rAAV8-LSP-hIDSco over 12 months, Ids KO mice were treated with three dose levels: 2.5 × 1011 vg/kg, 1.25 × 1012 vg/kg, and 6.25 × 1012 vg/kg. Vector administration partially rescued the diminished weight gain seen in Ids KO mice (Figure S4). At 12 months after dosing, a similar dose-dependent pattern to that seen in animals at 3 months after treatment was observed for I2S activity in serum and tissue (liver, spleen, kidney, heart, lung, and brain). After a peak at 14 days after the injection, serum hI2S activity gradually decreased over 12 months (Figure 2A). In treated Ids KO mice, increases in I2S activity levels in somatic tissues and the brain were directly proportional to the dose received (Figure 2B). I2S activity levels in somatic tissues of treated Ids KO mice were greater than in WT mice at all doses (Figure 2C), and somatic tissue GAG levels were normalized to WT levels (Figure 2D). Interestingly, brain I2S activity levels in the 1.25 × 1012 vg/kg and 6.25 × 1012 vg/kg dose groups were 20% and 30% of those in WT mice, which resulted in significant decreases in HS and DS GAG brain levels compared with Ids KO mice. Cerebrospinal fluid (CSF) HS GAG levels were measured, and reductions compared with Ids KO mice were observed for all rAAV8-LSP-hIDSco groups (Figure 2E). Reduction in LAMP1 in heart sections correlated with the presence of hI2S (Figures S5 and S6). Significant LAMP1 reduction was detected in all brain regions of Ids KO mice treated with 1.25 × 1012 vg/kg and 6.25 × 1012 vg/kg doses (Figures 2F and S7 for immunostaining in the cortex) compared with naive Ids KO mice, and, histologically, I2S was visible in the choroid plexus and meninges (Figures 2G and S8).
Figure 2.
Ids KO mice pharmacology 12 months after treatment with rAAV8-LSP-hIDSco
(A–G) Ids KO mice were treated with a single intravenous injection of rAAV8-LSP-hIDSco at three dose levels ranging from 2.5 × 1011 vg/kg to 6.25 × 1012 vg/kg and monitored for 12 months. (A) Serum and (B) somatic tissues and the CNS were assayed for I2S activity. (C) I2S enzyme activity fold change above WT levels in serum, somatic tissues, and the CNS 12 months after treatment. (D) Somatic tissues and the CNS were assayed for HS and DS content. (E) CSF was assayed for HS content. Bars represent pooled CSF from all mice in each dose group and lack error bars due to pooling. Due to lack of sufficient sample volume, the WT group could not be tested. (F) CNS assayed by IHC for LAMP1 positivity. (G) Naive Ids KO and high rAAV8-LSP-hIDSco dose assayed by IHC for hI2S in the choroid plexus and meninges. On all plots, data are presented as mean ± SD, n = 9–12 per group, except for the naive WT group (n = 6). p values for rAAV8-LSP-hIDSco-treated animals compared with naive Ids KO were generated by one-way ANOVA with multiple comparisons using Dunnett’s correction.∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001 versus naive Ids KO. CSF, cerebral spinal fluid; GAG, glycosaminoglycan.
Effect of rAAV8-LSP-hIDSco on functional parameters in Ids KO mice
For imaging and behavioral experiments in Ids KO mice, rAAV8-LSP-hIDSco was evaluated at doses of 2.5 × 1011 vg/kg, 1.25 × 1012 vg/kg, and 6.25 × 1012 vg/kg. Three-dimensional surface visualizations of micro-computed tomography (CT) images of the zygomatic arch showed similarities in bone structure and width between Ids KO animals treated with rAAV8-LSP-hIDSco and WT animals (Figure 3A). For dense bone volume measurements of the femur, tibia, humerus, radius, and zygomatic arch, vehicle-treated Ids KO mice displayed larger volumes than WT animals. At 13.5 months after dosing, there were no significant differences between animals treated with rAAV8-LSP-hIDSco and WT animals, indicating a normalization of bone-related changes for dense bone volumes even at the lowest dose (2.5 × 1011 vg/kg) (Figure 3B).
Figure 3.
Ids KO mice functional evaluations after treatment with rAAV8-LSP-hIDSco
(A–E) rAAV8-LSP-hIDSco was evaluated at single intravenous doses of 2.5 × 1011 vg/kg, 1.25 × 1012 vg/kg, and 6.25 × 1012 vg/kg in Ids KO mice. An additional cohort was dosed with 6.25 × 1012 vg/kg of a null vector (rAAV8-MY011) as a negative control, and age-matched vehicle-treated Ids KO and naive WT mice were included as controls. (A) Three-dimensional surface visualization of micro-CT images of the mouse skull, with zygomatic arch highlighted in yellow from each group 13.5 months after rAAV8-LSP-hIDSco treatment. Scale bar, 2 mm. (B) Micro-CT measurements of mineralized dense bone volume 13.5 months after rAAV8-LSP-hIDSco treatment. (C) Left ventricular mass measured by echocardiography 12.5 months after rAAV8-LSP-hIDSco treatment. (D) Latency to fall assessed using the accelerating RotaRod motor coordination test in treated Ids KO mice up to 12 months. (E) Y-maze neurobehavioral assessment in treated Ids KO mice up to 12 months. All data are presented as mean ± SD with n = 7–10 animals per group for (B) and (C) and n = 10–12 animals per group for (D) and (E). p values for rAAV8-LSP-hIDSco-treated animals compared with naive WT mice were generated by one-way ANOVA with multiple comparisons using Dunnett’s correction (B and C). Two-way ANOVAs with multiple comparisons using Dunnett’s correction were used to assess differences in behavioral performance between rAAV8-LSP-hIDSco-treated animals and naive WT mice (D and E). ∗p < 0.05; ∗∗∗p < 0.001; ns, not significant versus naive WT.
Echocardiography was performed at 12.5 months after dosing. The cardiac phenotype of the Ids KO control group was mild, and no statistically significant differences were observed between Ids KO and WT mice in functional and morphometric analyses (data not shown). Treatment effects were also not statistically significant. For example, in the rAAV8-LSP-hIDSco groups, there were reductions in left ventricular mass compared with Ids KO mice; however, reduction was also seen for the null vector-treated group (Figure 3C).
Surprisingly, motor performance testing found that naive Ids KO mice and null vector-treated mice stayed longer on the RotaRod than WT mice, which was the opposite of the expected phenotype.12,13 Mice in our study were older than previous reports and we speculate that additional deficits may have contributed to inability of Ids KO mice to leave the rod voluntarily without penalty. The longer latency to fall was reversed at weeks 33, 45, and 57 in mice treated with the 1.25 × 1012 vg/kg and 6.25 × 1012 vg/kg doses (Figure 3D). The Y-maze test, used to assess exploratory behavior and working memory, reflected no meaningful difference in the percentage alternation among all groups at any time point, including WT and Ids KO controls (Figure 3E).
Lung function was measured with the flexiVent system in Ids KO mice at 4 months from treatment initiation with a single intravenous injection of rAAV8-LSP-hIDSco at three dose levels ranging from 2.5 × 1011 vg/kg to 6.25 × 1012 vg/kg. An additional cohort was dosed with 6.25 × 1012 vg/kg of the null vector, rAAV8-MY011, or with intravenous idursulfase 1 mg/kg once a week. The vehicle Ids KO group displayed statistically significant differences from WT mice in three of the five parameters measured (tissue elastance, quasi-static compliance, and inspiration capacity). Neither rAAV8-LSP-hIDSco nor weekly ERT with intravenous idursulfase normalized any of these three parameters to WT levels with statistical significance (Figure S9). HS levels in the trachea were decreased but not normalized for the rAAV8-LSP-hIDSco groups and the idursulfase-treated group, although the magnitude of decrease was greater for all rAAV-treated groups (Figure S10). Trachea DS concentrations decreased to WT levels in the rAAV-treated groups but not in the idursulfase-treated group (Figure S11).
Effect of rAAV8-LSP-hIDSco on hI2S concentrations and activity in NHPs
Evidence from mice provided a framework for investigating the pharmacokinetic and biodistribution of rAAV8-LSP-hIDSco in large animals. We administered rAAV8-LSP-hIDSco to healthy male cynomolgus monkeys at low (1.25 × 1012 vg/kg; n = 3) or high (6.25 × 1012 vg/kg; n = 6) doses to investigate hI2S concentration and I2S activity in serum and multiple tissues over time (up to 3 or 9 months). These two doses were chosen for NHPs to match the same amount per kilogram of body weight given to Ids KO mice in the 12-month long-term study to understand species translation for the same vector genome based on body weight.
Comparing NHPs treated with the low dose with Ids KO mice treated with 2.5 × 1010 vg/kg, where more than 90% of HS reduction was observed in the somatic tissues, serum hI2S concentration was approximately five times higher and I2S activity nearly three times higher, with sustained levels up to 3 months (Figures 4A and 4B). Meanwhile, compared with Ids KO mice administered the same dose (1.25 × 1012 vg/kg), NHPs had a serum hI2S concentration approximately 50 times lower and a serum I2S activity about 30 times lower. A peak in I2S activity was observed at 20 days, followed by an initial drop and stabilization. Anti-hI2S anti-drug antibodies (ADAs) were either transient or undetectable at 3 months in NHPs treated with the low dose (Figure S12). NHPs treated with the high dose showed a variable profile of hI2S concentrations (Figures 4C and 4D). In general, the decline in hI2S serum levels in these NHPs was associated with the presence of anti-hI2S ADAs (Figure S12). The rapid decline in I2S activity was only observed after 3 months, whereas the decline in hI2S concentration was observed as early as 1 month after dosing. For animal #3003, which lacked anti-hI2S ADAs, anti-AAV8 ADAs, and anti-AAV8 neutralizing antibodies (Nabs; Figure S12), the durability of hI2S expression and hI2S exposure in the serum was at least 9 months (planned sacrifice time point). As an additional investigation into the catalytic nature of the enzymatic activity, the levels of formylglycine on the animal-produced enzymes isolated from NHP serum were similar to those on the recombinant hI2S enzyme (Table S1).
Figure 4.
NHP serum transgene product concentration and enzyme activity profiles in individual animals at different time points after treatment with rAAV8-LSP-hIDSco
(A) hI2S serum concentration and (B) I2S serum enzyme activity over time up to 3 months after administration with a low dose of rAAV8-LSP-hIDSco (1.25 × 1012 vg/kg). (C) hI2S serum concentration and (D) I2S serum enzyme activity over time up to 9 months after administration with a high dose of rAAV8-LSP-hIDSco (6.25 × 1012 vg/kg). Animals 2001, 2002, and 2003 were administered 1.25 × 1012 vg/kg of rAAV8-LSP-hIDSco; animals 3001, 3002, 3003, 4001, 4002, and 4003 were administered 6.25 × 1012 vg/kg of rAAV8-LSP-hIDSco. Animals 1001, 1002, 1003, and 1004 were administered formulation buffer. Cavg, average concentration; IV, intravenous.
Translation from mouse to NHPs requires comparison of tissue exposure and tissue activity of I2S between NHPs and Ids KO mice treated with rAAV8-LSP-hIDSco (Table 1). In NHP deep tissues (lung, heart, and bone marrow), hI2S concentration in the low-dose cohort (in the absence of anti-hI2S ADAs) was sufficient to exceed the hI2S concentrations in mice that had normalized GAGs in these tissues at 2.5 × 1010 vg/kg. In the high-dose cohort, hI2S concentrations in NHP deep tissues were affected by the presence of anti-hI2S ADAs in the serum, meaning that hI2S concentrations were numerically lower in NHPs with anti-hI2S ADAs than in those without (Figure S13; Table S2).
Table 1.
Tissue exposure of transgene product in NHPs and Ids KO mice treated with rAAV8-LSP-hIDSco
| Tissue (dose) | hI2S concentration in NHPs without ADAs (above to vehicle control) | hI2S concentration (2.5 × 1010 vg/kg) in Ids KO micea | hI2S activity in NHPs without ADAs (% of WT mice) | hI2S activity (2.5 × 1010 vg/kg) in Ids KO mice (% of WT mice) | HS reduction (2.5 × 1010 vg/kg) in Ids KO mouse tissues (% of Ids KO vehicle) | hI2S activity (2.5 × 1011 vg/kg) in Ids KO mice (% of WT mice) | HS reduction (2.5 × 1011 vg/kg) in Ids KO mouse tissues (% of Ids KO vehicle) |
|---|---|---|---|---|---|---|---|
| Lung (low) | 6.0 ng/mg | 2.0 ng/mg | 40 | 30 | 97 | 270 | 100 |
| Lung (high) | 35.0 ng/mgb | 1.35a,c | |||||
| Heart (low) | 12.5 ng/mg | 1.4 ng/mg | 232 | 210 | 97 | 2,480 | 100 |
| Heart (high) | 23.2 ng/mg | 442c | |||||
| Kidney (low) | 4.4 ng/mg | 3.0 ng/mg | 10 | 90 | 91 | 1,380 | 100 |
| Kidney (high) | 27.3 ng/mg | 439c | |||||
| Muscle (low) | all values < LLOQ |
1.0 ng/mg | 87 | 50 | 100 | 1,000 | 100 |
| Muscle (high) | 5.4 ng/mg | 244c | |||||
| Bone marrow (low) | 29.5 ng/mg | 15.1 ng/mg | 210 | 310 | 100 | 46,000 | 100 |
| Bone marrow (high) | 198.0 ng/mg | 1,574c | |||||
| Spleen (low) | 36.1 ng/mg | 17.0 ng/mg | 110 | 140 | 98 | 1,200 | 100 |
| Spleen (high) | 281.4 ng/mg | 560c |
ADA, anti-drug antibody; hI2S; human iduronate-2-sulfatase; HS, heparan sulfate; Ids KO, iduronate-2-sulfatase gene knockout; LLOQ, lower limit of quantification; NHP, non-human primate; rAAV8, recombinant adeno-associated virus 8; vg, vector genome; WT, wild-type.
hI2S concentration was evaluated at 3-month necropsy.
The lack of correspondence between hI2S concentration and enzyme activity in lung tissues for the high-dose rAAV8-LSP-hIDSco group was probably due to technical challenges involving the enzyme assay used to detect rAAV8-LSP-hIDSco-mediated hI2S increase.
Tissues from animals in the high-dose group at 9 months are normalized to the respective formulation-buffer-treated animals euthanized at 9 months; values obtained from formulation-buffer-treated animals at 9 months may vary from those obtained at 3 months.
There was no detectable hI2S above the NHP endogenous I2S in the CSF, in the spinal cord, and in different brain regions (data not shown) in either cohort throughout the study.
Liver expression profile of rAAV8-LSP-hIDSco in NHPs and mice
To determine the liver expression profile of rAAV8-LSP-hIDSco, liver biopsies were acquired from NHPs over time; vector copy number, hIDS messenger RNA (mRNA) transcripts, and liver hI2S concentrations were measured for the low- and high-dose groups (Figure 5).
Figure 5.
NHP liver vector genome, mRNA transcript, and transgene product concentration in individual animals at different time points after treatment with rAAV8-LSP-hIDSco
(A) Analysis of the liver profile of vector genome, (B) mRNA transcript, and (C) transgene product concentration in NHPs treated with a high or low dose of rAAV8-LSP-hIDSco for up to 3 or 9 months. Animals 2001, 2002, and 2003 were administered 1.25 × 1012 vg/kg of rAAV8-LSP-hIDSco; animals 3001, 3002, 3003, 4001, 4002, and 4003 were administered 6.25 × 1012 vg/kg of rAAV8-LSP-hIDSco.
After administration, a decreasing trend in vector copies was observed in all NHPs by day 50 (Figure 5A). In the high-dose group, the observed decline was slower after 3 months than in the first 90 days. Some animals showed a steady mRNA transcript profile, whereas others showed a decreasing trend in expression after vector administration, except for animal #4002 (administered with 6.25 × 1012 vg/kg of rAAV8-LSP-hIDSco), which experienced a rapid reduction trend in mRNA expression (Figure 5B). Hepatic hI2S concentrations mirrored the mRNA expression profile after administration in animals treated with either dose, including in animal #4002, in which a rapid reduction trend in protein concentration was also observed (Figure 5C).
Microscopic evaluation of hI2S by IHC in the liver of both Ids KO mice and NHPs at 1.25 × 1012 and 6.25 × 1012 vg/kg was conducted to characterize the distribution of hI2S protein expression (Figure 6). The expression of hI2S detected in the hepatocytes of Ids KO was widespread and dose dependent, with accumulations of hI2S in pericanalicular lysosomes and Kupffer cells (Figures 6A–6C). In NHPs, strong hI2S expression was detected in fewer, more scattered individual hepatocytes, with less dense accumulation of hI2S in the remaining hepatocytes and Kupffer cells than that seen in mice. In NHPs, there was a slight increase in the number of hI2S-expressing hepatocytes in the 6.25 × 1012 vg/kg dose group compared with the 1.25 × 1012 vg/kg group (Figures 6E and 6F). hI2S expression at the cellular level showed a marked difference in distribution between mouse and NHP livers. Scattered random individual cells were observed throughout the parenchyma in NHPs, in contrast with the relatively homogeneous dense distribution in all hepatocytes in mice.
Figure 6.
IHC of hI2S transgene product in Ids KO mice and NHP liver
Distribution of hI2S evaluated by IHC in the liver in both Ids KO mice and NHPs. Ids KO mice that received (A) vehicle control or (B) 1.25 × 1012 vg/kg, or (C) 6.25× 1012 vg/kg of rAAV8-LSP-hIDSco 12 months after administration. NHPs that received (D) the vehicle control, or (E) 1.25 × 1012 vg/kg, or (F) 6.25 × 1012 vg/kg of rAAV8-LSP-hIDSco 3 months after administration. Scale bar, 200 μm.
Vector genome levels in the liver and other tissues in NHPs were assessed together with those in treated WT mice (Figures 7A and 7B). In NHPs, non-liver tissues had a numerically lower number of viral vector copies than the liver at 3 months in animals in the low-dose group and at 3 and 9 months in animals in the high-dose group. A trend toward a dose-dependent increase in viral vector copies in all tissues at 3 months was observed. However, at 9 months, the viral vector copy number in the high-dose cohort showed a trend toward a decrease in all non-liver tissues (Figure 7A). Similarly, in mice, higher vector doses were associated with viral vector copy number, and non-liver tissues generally showed a lower vector copy number than the liver (Figure 7B). When each dose was evaluated across species, there were lower levels of vector genome, mRNA transcript, and hI2S in the livers of NHPs than in those of mice, in which the degree of difference was more evident in the mRNA transcript and hI2S levels (Figure 7C).
Figure 7.
Comparison of vector genome, mRNA, and transgene product in WT mice and NHPs
Vector genome in tissues of (A) NHPs treated with rAAV8-LSP-hIDSco for up to 3 or 9 months and (B) WT mice treated with rAAV8-LSP-hIDSco for up to 3 months. (C) Comparison of vector genome, transgene mRNA, and transgene product of liver in WT mice and NHPs treated with rAAV8-LSP-hIDSco. Data are presented as mean ± SD of n = 6 animals.
GLP toxicology study in mice
The aim of this study was to determine any potential toxicity of rAAV8-LSP-hIDSco and to assess the pharmacokinetics, tissue exposure of hI2S, and tissue vector genome concentrations during a 13-week observation period. rAAV8-LSP-hIDSco administered to WT mice at 1.25 × 1012 vg/kg, 6.25 × 1012 vg/kg, and 3.0 × 1013 vg/kg was well tolerated at all dose levels. All in-life, clinical, and anatomical pathology parameters were within normal limits. The no-observed-adverse-effect level (NOAEL) was 3.0 × 1013 vg/kg, the highest dose level in the study. Pharmacokinetic parameters for hI2S in serum are summarized in Table S3. Serum hI2S levels were similar among WT and Ids KO male mice (data not shown). Sex-dependent differences in serum hI2S concentrations were observed; namely, male mice tended to have hI2S concentrations more than twice as high as those of female mice (Table S3).
Discussion
In contrast to current treatments for MPS II, gene therapy with IDS could potentially lead to long-term sustained and constant vector-mediated I2S production, allowing penetration into deep tissues not adequately addressed by intravenous ERT. Using the liver as a depot organ for a systemically delivered rAAV-based vector can lead to cross-correction of other organs via the systemic circulation while also potentially providing immunotolerance to the transgene product.14
In the MPS II mouse model, we demonstrated that, when rAAV8-LSP-hIDSco was administered to young mice, hepatic hI2S production was sufficient to normalize GAG burden in somatic tissues at doses below 1 × 1012 vg/kg. Doses higher than 2.5 × 1010 vg/kg partially reduced brain GAG levels and improved histopathology. We observed normalization of bone density, suggesting that deep tissue exposure of liver-produced hI2S could have translational benefits to skeletal and joint clinical pathology. In RotaRod motor behavioral testing, the reduced latency to fall phenotype expected for Ids KO mice was not observed, possibly owing to age-related mice vision deficits. Nevertheless, the longer-latency knockout phenotype was corrected. Moreover, cardiorespiratory and neurobehavioral functional measurements were uninformative owing to the weak phenotype of the mouse model.
Although several rAAV-based gene therapies have shown promising results in knockout mice models,15,16,17 side-by-side evaluations in larger species remain unfortunately rare in preclinical publications. Knowing that rAAV-driven protein delivery is characterized by lower expression efficiency in larger species,18,19,20,21 we believe that cross-species studies are critical to evaluate whether rAAV-based treatments reach desirable therapeutic levels. To increase the translational understanding of rAAV8-LSP-hIDSco from Ids KO male mouse studies, the transduction efficiency of rAAV8-LSP-hIDSco gene therapy in the liver of NHPs was assessed together with its ability to cross-correct deep tissues (lung, heart, kidney, brain, and bone marrow), where the therapeutic hI2S levels in somatic tissues, but not in the CNS, were observed. At similar doses to those evaluated in a mouse toxicology/biodistribution study (1.25 × 1012 to 6.25 × 1012 vg/kg), NHPs treated with rAAV8-LSP-hIDSco showed sustained hI2S production in the liver. The comparison of the measurements of vector genome, mRNA, and hI2S levels in a mouse toxicology/biodistribution study with those observed in NHPs revealed a lower efficiency in one or multiple steps, from receptor binding and transgene production and secretion to tissue cross-correction in NHPs, than in mice: this may have contributed to the lower transgene product exposure levels observed in NHPs. The observation of the species differences is not unique to rAAV8-LSP-hIDSco but has also been reported by other investigators.22,23 As a result, we speculate that the partial GAG correction observed may be explained by the greater supraphysiological amount in mice than in NHPs of circulating transgene product, which enters the brain by diffusion crossing the blood-CSF barrier through the choroid plexus as suggested by IHC detection of hI2S in mouse choroid plexus and meninges. Although rAAV8-LSP-hIDSco gene therapy has only demonstrated partial CNS GAG normalization in Ids KO mice models and a lack of CNS uptake in NHPs, it has shown promising, translatable therapeutic effects in the cardiorespiratory, skeletal, and bone systems, addressing unmet medical needs.
Safety evaluation of rAAV8-LSP-hIDSco in NHPs was integrated into the study design. With this objective, the animals were not treated with immunosuppressants to avoid potential confounding factors. Without immunosuppressants, anti-hI2S ADAs were only observed in the high-dose cohort, suggesting that there is a circulating level of transgene product above which ADA generation is triggered due to breakthrough of the tolerization threshold that can be species dependent. Given that rAAV8-LSP-hIDSco was designed to have the liver secrete the therapeutic protein into the systemic circulation and cross-correct other tissues, the level of hI2S in the circulation determines its amount in cross-corrected tissues. In animals with anti-hI2S ADAs, hI2S exposure in cross-corrected tissues (e.g., lung, heart, and bone marrow) correlated with the low circulating hI2S level. In contrast, animal #3003 had sustained systemic hI2S levels throughout the study in the absence of ADAs and had consistent hI2S exposure and enzyme activity in somatic tissues. In addition, the impact of anti-capsid NAbs in the absence of anti-hI2S ADAs was observed in animal #4002, which belonged to the high-dose group. This animal was pre-screened for anti-AAV8 NAbs and was confirmed to be seronegative before dosing. Owing to the lack of liver samples before 3 months, it is difficult to know if the vector had a rapid decline of transduction (i.e., vector genome levels) immediately after dosing or if it started with low baseline transduction in this animal. However, based on data obtained at 3 months, we speculate that T cell-mediated cytotoxicity may have inhibited or reduced the hepatocyte transduction in this animal, leading to low mRNA and protein levels. These observations further confirm the need to include immunosuppressants as part of rAAV-based gene delivery studies, should transgene product durability be measured.
In summary, liver-directed rAAV8-LSP-hIDSco gene therapy demonstrated significant reduction of GAG levels in difficult-to-treat systemic tissues and partially reduced CNS GAG levels in Ids KO mice. Although meaningful hI2S levels were achieved in almost all measured somatic organs in NHPs, the level of circulating hI2S was not sufficient to enable adequate amounts of enzyme to cross passively into the CNS. The current study established a foundation for a correlation between dose, circulating hI2S level, and tissue exposure levels (somatic and CNS tissues) in rodents and large animal models, enabling translational understanding of gene therapy development for MPS II. The difference in CNS exposure between rodents and NHPs, probably due to circulating hI2S levels being lower in NHPs than in rodents at the same dose, further emphasizes the importance of evaluating gene therapy products in NHPs.
Overall, these results demonstrate that appropriately designed preclinical rodent and NHP studies are critical to support the translation, including dose selection, of gene therapies into the clinic. Further studies with higher doses, improvement of vector potency, or proteins using an active mechanism to access the CNS would be required to investigate the feasibility of systemic gene therapy for both somatic and CNS benefits for patients with MPS II.
Materials and methods
Animals
Animal welfare complied with all applicable regulations, and all in vivo procedures described were approved by the Takeda Pharmaceuticals Institutional Animal Care and Use Committee.
Hemizygous Ids KO male (B6N.Cg-Idstm1Muen) mice containing an X-linked mutation and recapitulating several biological and pathological aspects of MPS II disease and C57BL/6 WT mice were bred at Taconic Laboratories.24,25 Upon arrival at Takeda, animals were housed with their littermates. C57BL/6 WT mice for the GLP toxicology study were received from Charles River Laboratories, Raleigh, NC, USA and housed at the testing facility (Charles River Laboratories Ashland, Ashland, OH, USA). All mice were maintained on a 12-h light/dark cycle, and water and food were provided ad libitum.
Cynomolgus monkeys (Macaca fascicularis) were supplied by Worldwide Primates, Miami, FL, USA. Blood samples (500 μL of serum) from 100 animals were screened and evaluated for antibodies against the adeno-associated virus serotype 8 (AAV8) vector. In total, 25 animals that tested negative for AAV8 Nabs were shipped to Charles River Laboratories, Reno, NV, USA for study activities and were acclimated to laboratory housing for at least 7 days before dosing initiation. Animals were housed singly upon arrival until completion of dose administration, then in groups of up to three animals of the same dosing group for the remainder of the study.
Vector structure and production
The vector genome for rAAV8-LSP-hIDSco contains a 5′ inverted terminal repeat (ITR), three hepatocyte-specific cis-regulatory modules, a liver-specific transthyretin promoter, a minute virus of mice intron, a codon-optimized human IDS (hIDSco) coding sequence, a woodchuck hepatitis virus post-transcriptional regulatory element (WPRE), a bovine growth hormone polyadenylation sequence, and a 3′ ITR (Figure 1A). The IDS coding sequence was optimized based on codon-usage frequency in the liver, a reduced number of CpG sites, and additional modifications to improve expression (e.g., removing repeat and inverted repeat sequences and potential splicing sites).
The rAAV8-LSP-hIDSco vector was produced using standard triple-transfection methods. In brief, HEK293 cells were co-transfected with three plasmids: the ITR plasmid containing the IDS transgene, a RepCap expression cassette, and an adenoviral helper gene expression cassette. After 72–96 h, the cells were chemically lysed and the cell pellet and medium were collected. The cell lysate was clarified and treated with benzonase. The clarified lysate was loaded to a POROS CaptureSelect AAVX column connected to an AKTA purification system, and AAV vector-containing fractions of the elute were collected according to the manufacturer’s recommended conditions. Fractions containing AAV vectors were pooled and subjected to a cesium chloride continuous gradient to enrich for full capsids. Fractions containing mostly full capsids were pooled and subjected to buffer exchange. The AAV vectors were formulated in phosphate buffered saline with 0.001% Pluronic F-68 and were subjected to standard characterization, including a quantitative polymerase chain reaction (qPCR) for titrating, silver staining for purity, an amebocyte lysate assay (Endosafe) for endotoxin measurement, and an in vitro transduction assay to determine biological activity. Characterized vectors were aliquoted and frozen at −80°C.
Study design and vector administration
All mice for pharmacology experiments were dosed at Takeda in Cambridge, MA, USA; samples were tested at the same facilities or transferred to external contract research organizations (CROs), depending on the experiment.
A single intravenous rAAV8-LSP-hIDSco injection via tail vein was administered to Ids KO mice at different doses ranging from 2.5 × 108 to 6.25 × 1012 vg/kg, depending on the experiment. Ids KO mice were dosed at 5–7 weeks of age (16 weeks of age for the respiratory function studies). In some experiments, an additional Ids KO mice cohort was dosed with a null vector (rAAV8-MY011, vector genome with no promoter and transgene sequences) as a negative control; age-matched naive Ids KO and WT mice were also included as controls. For the respiratory function study, an Ids KO mice cohort was also dosed with intravenous idursulfase 1 mg/kg once a week. The Ids KO mice cohort used for the imaging measurements was assessed at Invicro (San Diego, CA, USA) at different time points up to 13.5 months after dosing. For the functional studies, mice were tested at PsychoGenics (Paramus, NJ, USA) for behavioral assessments beginning at 21 weeks after dosing.
A GLP toxicology study was also performed in WT mice to assess the tolerability of rAAV8-LSP-hIDSco and to identify the NOAEL. For this study, rAAV8-LSP-hIDSco was evaluated at doses of 1.25 × 1012 vg/kg, 6.25 × 1012 vg/kg, and 3.0 × 1013 vg/kg administered via a single intravenous injection to WT mice at Charles River Laboratories Ashland. An additional WT mouse cohort was dosed with formulation buffer as a control.
Data from the mice studies were used to identify the most appropriate vector dose to treat NHPs and, therefore, to assess serum and tissue transgene product concentrations, enzyme activity profiles, and vector biodistribution in large animals. Healthy male cynomolgus monkeys aged 2.1–2.9 years were administered with rAAV8-LSP-hIDSco at two different doses, 1.25 × 1012 vg/kg and 6.25 × 1012 vg/kg, through a single 30-min intravenous infusion via a suitable peripheral vein using a calibrated infusion pump. Formulation buffer was administered to the control animals.
Necropsy and sample collection
Mice were euthanized using carbon dioxide inhalation at different time points after vector administration depending on the experiment (see “results” section). Blood was collected via cardiac puncture and mice were perfused with 10 mL of saline. Serum was obtained by centrifuging blood at 13,200 revolutions per minute (rpm) for 3 min at room temperature. Collected brain and somatic tissues (liver, kidneys, spleen, heart, lung, bone marrow, quadriceps, skin) were divided into multiple sections, placed in 10% formalin for histology analysis, or frozen at −80°C for further analyses.
At 3 or 9 months after AAV injection, NHPs were humanely euthanized by induction into deep, unrecoverable anesthesia with ketamine-pentobarbital administration. This was followed by blood collection for analysis of parameters, and saline perfusion to clear assay-interfering circulating transgene proteins and endogenous serum proteins from the tissues to be evaluated. Serial CSF and blood samples were collected throughout the duration of the study. Following collection of CSF samples, total-body perfusion was performed with ice-cold 0.9% saline for approximately 5–10 min at a rate of 250 mL/min until no blood was visible in the perfusate. Selected peripheral tissues (liver, kidney, spleen, heart, lung, skin, bone marrow, spinal cord, and quadricep) and prespecified brain regions were collected for IHC analysis or frozen at −80°C for further analyses.
hI2S serum and tissue ELISA
hI2S concentrations from serum and tissue were analyzed by ELISA using a purified goat anti-I2S capture antibody (B852) and a horseradish peroxidase-conjugated goat anti-I2S detection antibody (B852). Samples exceeding the high end of the calibration curve were further diluted and retested. Concentrations from serum were expressed as ng/mL, and results from tissues were normalized to total protein in extracts as determined by bicinchoninic acid (BCA) assay (Pierce Biotechnology, Rockford, IL, USA) and were expressed as ng/mg protein.
I2S activity assay
The fluorometric assay for I2S enzymatic activity is a two-step reaction under acidic conditions.26 During the first step, tissue or serum samples and controls were diluted, mixed with 4-methylumbelliferyl α-L-iduronide-2-sulfate (4-MUS) (Santa Cruz Biotechnology, Dallas, TX, USA; or Biosynth, San Diego, CA, USA) and added on to black microtiter 96-well plates (Sigma-Aldrich, St Louis, MO, USA). Following 1 h of incubation at 37°C, I2S hydrolyzed 4-MUS to 4-methylumbelliferyl α-L-iduronide (MUBI). In the second step, the enzyme alpha-L-iduronidase was added to the plate and incubated at 37°C to hydrolyze MUBI to the final product, 4-methylumbelliferone (4-MU). After 4 h of incubation, the reaction was stopped and a standard curve of 4-MU sodium salt (Sigma-Aldrich, St Louis, MO, USA) ranging from 3.91 to 4,000 nM for mice and from 1.47 to 6,000 nM for NHPs was added to the plate. Fluorescence was read at 365-nm excitation, 450-nm emission, and 435-nm cutoff. Serum data were expressed as nmol/h/mL; results from tissues were normalized to total protein in tissue extracts as determined by BCA assay (Pierce Biotechnology, Rockford, IL, USA) and expressed as nmol/h/mg protein.
GAG quantification
For HS and DS quantification, mouse liver, kidney, spleen, heart, lung, bone marrow, quadriceps, skin, and brain tissues were homogenized in 25 mM ammonium formate and 1 mM calcium acetate buffer followed by digestion with 12 mU/μL heparinase I, 4 mU/μL heparinase II, and 1.4 mU/μL heparinase III (all from New England Biolabs, Ipswich, MA, USA) at 30°C for 16 h for HS or 0.1 μg/μL chondroitinase B (R&D Systems, Minneapolis, MN, USA) at 35°C for 16 h for DS to degrade the polysaccharide forms to HS or DS disaccharides. Digestion reactions were precipitated with chilled methanol and the supernatant dried and resuspended in 20 μL of methanol and 150 μL of 5 mM tributylamine, 5 mM ammonium formate, and 0.025% formic acid in 95:5 water:acetonitrile. Ten microliters was injected on a Sciex API5000 mass spectrometer (AB Sciex, Framingham, MA, USA) containing a Waters ACQUITY UPLC BEH C18 column (130Å, 1.7 μm, 2.1 × 50 mm) at 50°C. Following gradient liquid chromatography separation,27 turbo ion spray ionization in negative-ion mode was coupled with multiple reaction monitoring for disaccharides. Intact polysaccharides of HS from porcine intestinal mucosa (Calbiochem, San Diego, CA, USA) and DS from porcine mucosa (Iduron, Alderley Park, Alderley Edge, Cheshire, UK) were used as reference standards. Four major disaccharides degraded from HS (ΔUA, GlcNAc,6S [II-A]; ΔUA, GlcNS [IV-S]; ΔUA, GlcNS,6S [II-S]; and ΔUA, GlcNAc [IV-A]) were selected as signature disaccharides monitored in HS quantitation. Two major disaccharide isomers, DS4S (DS disaccharide UA, GalNAc,4S) and DS6S (DS disaccharide UA, GalNAc,6S), resulting from specific digestion of DS were used in DS quantitation. Calibration standards were used for quality control (QC) and acceptance criteria set at back-calculated concentration not deviating from more than ±25.0%. Lower limit of quantification for HS and DS was 50 ng/mL. Tissue homogenates were measured for protein concentration using BCA assay (Pierce), GAG quantifications were normalized to the tissue protein levels, and concentrations expressed as μg GAG/mg protein.
Functional studies in mice
Imaging measurements
Micro-CT imaging of whole-body skeletal anatomy was performed using an Inveon CT scanner. For CT imaging, mice were anesthetized with 2%–3% inhaled isoflurane and maintained with 1%–2% inhaled isoflurane in oxygen throughout image acquisition. Each mouse was positioned in the scanner such that the left hindlimb was tucked under the animal and the right hindlimb was extended behind, making sure all limbs and the head were in the field of view. Phantom scans were performed with a mineral density phantom (three densities) immediately before in vivo image acquisition. Scans were set to medium magnification (79.66-μm pixels) using standard X-ray tube settings and exposure (80 kV, 500 μA, 275 ms) and acquiring two bed positions in step + shoot mode with 360° projections. The femur, tibia, humerus, radius, cranium, and zygomatic arch were segmented from the whole-body CT scans. The dense bone volume was calculated for each bone using a threshold-based approach. A voxel Hounsfield unit (HU) value was chosen as a low threshold corresponding to dense bone; then, all voxels with HU values above this threshold were segmented, counted, and converted to a volume measurement of dense bone. Test article identities were blinded to investigators for CT and echocardiography measurements.
For echocardiography, mice were anesthetized with 2%–3% inhaled isoflurane and maintained with 1%–2% inhaled isoflurane in oxygen throughout imaging. Isoflurane was adjusted as necessary to maintain a target heart rate of 400–600 beats per minute in each animal. Following induction of anesthesia, animals were placed supine on a heated stage (41°C–42°C) and images were obtained using a Visual Sonics Vevo 2100 ultrasound system with MS550S 32- to 56-MHz linear echocardiography transducer. Measurements acquired were ejection fraction, fractional shortening, stroke volume, cardiac output, heart rate, left ventricular wall thickness in systole and diastole, myocardial area change, left ventricle mass, and left ventricular mass corrected.
RotaRod behavioral test
All mice were tested on a RotaRod (Ugo Basile Instrument, Varese, Italy) over 3 days. Test article identities were blinded to investigators. An accelerating RotaRod testing program was selected based on previous Ids KO mouse phenotyping.13,28 Mice were trained on the RotaRod on day 1 with two training trials. Trial 1 was a habituation session at 5 rpm for 60 s. If mice fell during trial 1, they were replaced on the rod until 60 s was reached. After a 30-min inter-trial interval, mice began trial 2, a 5-min session with an accelerating speed of 5–40 rpm. Both training trials were unrecorded. All mice were then tested in six testing trials, with two trials per day for 3 days. All testing trials were recorded and conducted over 5 min with a speed of 5–40 rpm and an inter-trial interval of 5 min. Latency to fall was recorded for all testing trials.
Y-maze behavioral test
Spontaneous alternation was evaluated in a single 8-min trial in a Y-maze consisting of three identical arms (40 × 10 × 20 cm) and each mouse was allowed to see distal spatial landmarks. Test article identities were blinded to investigators. The test mouse was placed in the middle of the three arms and allowed to explore freely. Arm entry was considered successful when hind paws were placed in the arm in full. Spontaneous alternation was described as successive entries into three arms, in overlapping triplet sets. The effect was calculated as percentage alternation = [number of alternations/(total number of arm entries −2)] × 100. Total entries were recorded as an indication of ambulatory activity, and mice that performed fewer than 12 entries in 10 min were excluded from the analysis.
Respiratory function measurements
Respiratory mechanics were assessed by the flexiVent (SCIREQ, Montreal, QC Canada) system in mice. An endotracheal tube was inserted and connected to a flexiVent ventilator. Before transfer and connection, the flexiVent circuit was primed with isoflurane to minimize potential for animal recovery. Maintenance anesthesia was achieved with 1.0%–3.5% isoflurane. Respiratory parameters, including elastance, resistance, and compliance, were collected via forced oscillation techniques. The CRO conducting the flexiVent measurements (QTest Labs, Columbus, OH, USA) was not blinded to the experimental groups. If measurements could not be accurately recorded, individual mice were excluded from the analysis (n = 2 omitted from the 6.25 × 1012 vg/kg rAAV8-LSP-hIDSco group; n = 1 omitted from the naive Ids KO group; n = 4 omitted from the idursulfase-treated group). Throughout each terminal experiment, analog signals were digitally sampled (500–2,000 Hz) and recorded continuously with a data acquisition system (IOX, emka TECHNOLOGIES, Paris, France; and flexiWare, SCIREQ Scientific Respiratory Equipment, Montreal, QC, Canada). Ids KO animals treated with idursulfase were terminated approximately 7 days after the last idursulfase infusion.
Vector biodistribution and hIDS mRNA
Vector biodistribution and hIDS mRNA quantification were analyzed using qPCR and reverse transcription qPCR (RT-qPCR). Briefly, genomic DNA and total RNA were extracted from RNAprotect Tissue Reagent (Qiagen, Valencia, CA, USA) from preserved mouse and NHP tissues using a QIAsymphony DSP DNA Mini Kit (Qiagen, Valencia, CA, USA) or a QIAsymphony RNA Kit (Qiagen, Valencia, CA, USA).
For vector biodistribution analysis, purified genomic DNA was analyzed using a qPCR assay, a set of transgene-specific primer/probe for vector genome copy number quantification. An endogenous single-copy gene (transketolase gene) on mouse and NHP chromosomes was also measured by qPCR to quantify the copy number of haploid genome for normalizing the genomic DNA concentration. The measured vector genome copy number and the normalized genomic DNA concentration were then used to calculate the copy/μg genomic DNA or copy/diploid genome units.
For hIDS mRNA quantification, total RNA was analyzed by RT-qPCR for the mRNA of hIDS transgene and three housekeeping genes, namely the actin γ1 gene (ACTG1), the ribosomal protein S18 gene (RPS18), and the ribosomal protein S27 gene (RPS27), separately with the corresponding primers and TaqMan probes. Relative hIDS mRNA expression was calculated using the average of the three housekeeping gene measurements and the method.29
Histology
For mouse studies, somatic tissues and left hemispheres of brains were fixed in 10% neutral buffered formalin for 24 h. Brain was trimmed into four slices from anterior to posterior, namely striatum, thalamus, hippocampus, and cerebellum. All tissues were processed for paraffin block and 5-μm sections were prepared for I2S and LAMP1 IHC staining. IHC staining for hI2S and LAMP1 detection was performed with BondRX Stainer. Briefly, after antigen retrieval with BOND epitope retrieval solution 1 (pH 6.0), the following primary antibodies were applied: rabbit anti-hI2S polyclonal antibody (1:500; LSBio, Seattle, WA, USA) for detection of hI2S expression and distribution, and rabbit anti-LAMP1 polyclonal antibody (Abcam, Cambridge, UK; ab24170; 1:2,000 for brain and 1:3,000 for other tissues) to detect LAMP1, a marker of lysosomes. A BOND Polymer Refine kit (Leica Biosystems, Buffalo Grove, IL, USA) containing a peroxide block, post primary, polymer reagent, DAB chromogen, and hematoxylin counterstain was supplied as ready-to-use detection system for the automated BOND stainer. All stained slides were dehydrated with gradient alcohol, cleared with xylene, and mounted with mounting medium, then scanned with an Aperio ScanScope AT2 scanner (Leica Biosystems, Lincolnshire, IL, USA). The whole digital slide for each tissue or the main brain regions, including cortex, striatum, thalamus, hippocampus, and cerebellum, were viewed and analyzed, and representative images were taken by ImageScope (Leica Biosystems, Lincolnshire, IL, USA). For LAMP1 quantitative analysis, the Aperio Positive Pixel Count algorithm (Leica Biosystems, Lincolnshire, IL, USA) was selected and adjusted to cover LAMP1-positive staining accurately. The data were presented as LAMP1 positivity percentage (positivity percentage = positive area [pixels]/total analyzed area [pixels] × 100%).
For the NHP IHC study, formalin-fixed, paraffin-embedded male cynomolgus monkey liver tissue was cut at 3 μm and kept at room temperature before staining. Following deparaffinization and rehydration, the slides were incubated in 1× Target Retrieval Solution pH 6 (Agilent, Dako, Santa Clara, CA, USA) for 20 min at 97°C and then placed in a Thermo Autostainer (Agilent, Santa Clara, CA, USA). Endogenous peroxidase activity was blocked by using 0.3% hydrogen peroxide for 20 min. Slides were then incubated in either 1.0 μg/mL of rabbit anti-I2S antibody (LSBio, Seattle, WA, USA; catalog number LS-C341717) or an isotype control rabbit immunoglobulin G (Vector Laboratories, Burlingame, CA, USA; catalog number I-1000) for 60 min followed by the secondary antibody, Dako EnVision+ System labeled polymer (goat anti-rabbit; Agilent, Santa Clara, CA, USA; catalog number K400311-2) for 30 min. Antibody binding was detected using 3,3′-diaminobenzidine tetrahydrochloride as the chromogen. Slides were then counterstained in hematoxylin and dehydrated through a progressive series of alcohols and xylene before cover slipping.
I2S ADA assay
The I2S ADA assay comprised a homogeneous “bridging” electrochemiluminescence (ECL) format with biotin and ruthenium-labeled recombinant hI2S (idursulfase, Takeda Pharmaceuticals USA, Lexington, MA, USA) bound together (bridged) by ADAs in test samples for detection.
rAAV8 capsid ADA assay
For the rAAV8 capsid ADA assay (a sandwich ECL assay), serum samples were diluted to minimum required dilution and added to an AAV8-coated plate; after incubation for 1 h, the plate was washed and the rAAV8 ADA bound to the AAV8 capsid was detected with SULFO-TAG-labeled Protein A/G/L (Meso Scale Diagnostics, Rockville, MD, USA).
rAAV8 capsid NAb assay
The rAAV8 capsid NAb assay was a cell-based assay that used a rAAV8 vector coding for luciferase as a reporter. Briefly, serum samples were first incubated with a rAAV8-CMV-Luc vector for 1 h, then the mixture was added to wells containing pre-seeded HEK293T cells pretreated with etoposide. The cells were lysed after 2 days and the luciferase in the cell lysate was measured. The luciferase signal intensity inversely correlates with the concentration of NAb in the serum sample. The assay threshold was established at a 50% reduction in luciferase signal, and any sample with a signal reduction of more than 50% was determined as NAb positive.
GLP toxicology study in mice
Potential toxicity of rAAV8-LSP-hIDSco, pharmacokinetics, tissue exposure of transgene product hI2S, and tissue vector genome concentrations were evaluated across a 13-week observation period. Standard histopathology and safety assessments were performed on 10 animals/sex/group at necropsy on days 30 and 91.
Statistical analysis
All data were analyzed with Prism 7 (GraphPad Software, San Diego, CA, USA) unless otherwise noted. Data are presented as either individual values or mean ± standard deviation (SD). Data were evaluated using one-way or two-way analysis of variance (ANOVA) with multiple comparisons using Dunnett’s correction. p < 0.05 was considered statistically significant.
Acknowledgments
This study was sponsored and funded by Takeda Pharmaceutical Company Limited. Data analyses were performed by Takeda Pharmaceutical Company Limited under the direction of the authors. Medical writing support under the direction of the authors was provided by Giovanni Ferrati, PhD, of Oxford PharmaGenesis, Oxford, UK and was funded by Takeda Development Center Americas, Inc.
The authors would like to thank Madhusudan Natarajan and Jens Wendland for their valuable contributions to this study.
All mouse experiments were performed at Takeda Pharmaceutical Company Limited, 125 Binney Street, Cambridge, MA, 02142, USA, except the following: the GLP toxicology study was performed at Charles River Laboratories Ashland, LLC, 1407 George Road Ashland, OH, 44805, USA; mice behavioral studies were performed at PsychoGenics, Inc., 215 College Road, Paramus, NJ, 07652, USA; the mouse imaging study was conducted at Invicro, San Diego, CA, 92137, USA; and respiratory tests were performed at QTest Labs, Columbus, OH, 43235, USA.
All in-life aspects of the non-human primate study were performed at Charles River Laboratories, Inc., 6995 Longley Lane, Reno, NV, 89511, USA.
Author contributions
N.C., K.D., D.E.E., B.F., X.F., C.G., H.G., Y.H., X.L., J.S., S.T., C.T.W., and V.W.C. contributed material and intellectual input into study designs, analysis, and interpretation of data. R.C., E.J.G., C.L., N.L., K.P., and D.S. made substantial contributions to the acquisition, analysis, and interpretation of data. All authors were involved in drafting or revising the manuscript.
Declaration of interests
N.C., R.C., K.D., B.F., E.J.G., C.L., N.L., K.P., J.S., S.T., C.T.W., and V.W.C. are employees of Takeda Development Center Americas, Inc., Cambridge, MA, USA. D.E.E., X.F., C.G., H.G., Y.H., X.L., and D.S. were employees of Takeda Development Center Americas, Inc., Cambridge, MA, USA while contributing to this study. All authors may hold stock or equity interests in Takeda Pharmaceuticals Company Limited. V.W.C. and X.L. are authors on a patent application related to this work.
Footnotes
Supplemental information can be found online at https://doi.org/10.1016/j.omtm.2023.03.014.
Supplemental information
Data availability
Restrictions apply to the availability of information and materials related to the viral vectors (cis-regulatory modules), which were used under license for this study. Please contact the corresponding author with any inquiries.
References
- 1.D'Avanzo F., Rigon L., Zanetti A., Tomanin R. Mucopolysaccharidosis type II: one hundred years of research, diagnosis, and treatment. Int. J. Mol. Sci. 2020;21 doi: 10.3390/ijms21041258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Muenzer J. Overview of the mucopolysaccharidoses. Rheumatology. 2011;50(Suppl 5):v4–v12. doi: 10.1093/rheumatology/ker394. [DOI] [PubMed] [Google Scholar]
- 3.Young I.D., Harper P.S. The natural history of the severe form of Hunter's syndrome: a study based on 52 cases. Dev. Med. Child Neurol. 1983;25:481–489. doi: 10.1111/j.1469-8749.1983.tb13794.x. [DOI] [PubMed] [Google Scholar]
- 4.Muenzer J., Wraith J.E., Beck M., Giugliani R., Harmatz P., Eng C.M., Vellodi A., Martin R., Ramaswami U., Gucsavas-Calikoglu M., et al. A phase II/III clinical study of enzyme replacement therapy with idursulfase in mucopolysaccharidosis II (Hunter syndrome) Genet. Med. 2006;8:465–473. doi: 10.1097/01.gim.0000232477.37660.fb. [DOI] [PubMed] [Google Scholar]
- 5.Parini R., Deodato F. Intravenous enzyme replacement therapy in mucopolysaccharidoses: clinical effectiveness and limitations. Int. J. Mol. Sci. 2020;21 doi: 10.3390/ijms21082975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Concolino D., Deodato F., Parini R. Enzyme replacement therapy: efficacy and limitations. Ital. J. Pediatr. 2018;44:120. doi: 10.1186/s13052-018-0562-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Brooks D.A., Kakavanos R., Hopwood J.J. Significance of immune response to enzyme-replacement therapy for patients with a lysosomal storage disorder. Trends Mol. Med. 2003;9:450–453. doi: 10.1016/j.molmed.2003.08.004. [DOI] [PubMed] [Google Scholar]
- 8.Massaro G., Geard A.F., Liu W., Coombe-Tennant O., Waddington S.N., Baruteau J., Gissen P., Rahim A.A. Gene Therapy for Lysosomal Storage Disorders: Ongoing Studies and Clinical Development. Biomolecules. 2021;11 doi: 10.3390/biom11040611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Kaufmann K.B., Büning H., Galy A., Schambach A., Grez M. Gene therapy on the move. EMBO Mol. Med. 2013;5:1642–1661. doi: 10.1002/emmm.201202287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Whiteman D.A., Kimura A. Development of idursulfase therapy for mucopolysaccharidosis type II (Hunter syndrome): the past, the present and the future. Drug Des. Devel. Ther. 2017;11:2467–2480. doi: 10.2147/DDDT.S139601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Meikle P.J., Brooks D.A., Ravenscroft E.M., Yan M., Williams R.E., Jaunzems A.E., Chataway T.K., Karageorgos L.E., Davey R.C., Boulter C.D., et al. Diagnosis of lysosomal storage disorders: evaluation of lysosome-associated membrane protein LAMP-1 as a diagnostic marker. Clin. Chem. 1997;43:1325–1335. doi: 10.1093/clinchem/43.8.1325. [DOI] [PubMed] [Google Scholar]
- 12.Polito V.A., Abbondante S., Polishchuk R.S., Nusco E., Salvia R., Cosma M.P. Correction of CNS defects in the MPSII mouse model via systemic enzyme replacement therapy. Hum. Mol. Genet. 2010;19:4871–4885. doi: 10.1093/hmg/ddq420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Gleitz H.F.E., O'Leary C., Holley R.J., Bigger B.W. Identification of age-dependent motor and neuropsychological behavioural abnormalities in a mouse model of Mucopolysaccharidosis Type II. PLoS One. 2017;12 doi: 10.1371/journal.pone.0172435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Colella P., Ronzitti G., Mingozzi F. Emerging issues in AAV-mediated in vivo gene therapy. Mol. Ther. Methods Clin. Dev. 2018;8:87–104. doi: 10.1016/j.omtm.2017.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Zapolnik P., Pyrkosz A. Gene therapy for mucopolysaccharidosis type II-A review of the current possibilities. Int. J. Mol. Sci. 2021;22 doi: 10.3390/ijms22115490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Sawamoto K., Chen H.H., Alméciga-Díaz C.J., Mason R.W., Tomatsu S. Gene therapy for mucopolysaccharidoses. Mol. Genet. Metab. 2018;123:59–68. doi: 10.1016/j.ymgme.2017.12.434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Homology Medicines Adeno-associated virus compositions for IDS gene transfer and methods of use thereof. 2021. https://patentscope.wipo.int/search/en/detail.jsf?docId=US342806608&_cid=P10-KYBRJV-57744-1
- 18.Samaranch L., Salegio E.A., San Sebastian W., Kells A.P., Foust K.D., Bringas J.R., Lamarre C., Forsayeth J., Kaspar B.K., Bankiewicz K.S. Adeno-associated virus serotype 9 transduction in the central nervous system of nonhuman primates. Hum. Gene Ther. 2012;23:382–389. doi: 10.1089/hum.2011.200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Hordeaux J., Wang Q., Katz N., Buza E.L., Bell P., Wilson J.M. The neurotropic properties of AAV-PHP.B are limited to C57BL/6J mice. Mol. Ther. 2018;26:664–668. doi: 10.1016/j.ymthe.2018.01.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hocquemiller M., Hemsley K.M., Douglass M.L., Tamang S.J., Neumann D., King B.M., Beard H., Trim P.J., Winner L.K., Lau A.A., et al. AAVrh10 vector corrects disease pathology in MPS IIIA mice and achieves widespread distribution of SGSH in large animal brains. Mol. Ther. Methods Clin. Dev. 2020;17:174–187. doi: 10.1016/j.omtm.2019.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Gray S.J., Matagne V., Bachaboina L., Yadav S., Ojeda S.R., Samulski R.J. Preclinical differences of intravascular AAV9 delivery to neurons and glia: a comparative study of adult mice and nonhuman primates. Mol. Ther. 2011;19:1058–1069. doi: 10.1038/mt.2011.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Puzzo F., Colella P., Biferi M.G., Bali D., Paulk N.K., Vidal P., Collaud F., Simon-Sola M., Charles S., Hardet R., et al. Rescue of Pompe disease in mice by AAV-mediated liver delivery of secretable acid α-glucosidase. Sci. Transl. Med. 2017;9 doi: 10.1126/scitranslmed.aam6375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Hurlbut G.D., Ziegler R.J., Nietupski J.B., Foley J.W., Woodworth L.A., Meyers E., Bercury S.D., Pande N.N., Souza D.W., Bree M.P., et al. Preexisting immunity and low expression in primates highlight translational challenges for liver-directed AAV8-mediated gene therapy. Mol. Ther. 2010;18:1983–1994. doi: 10.1038/mt.2010.175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Garcia A.R., Pan J., Lamsa J.C., Muenzer J. The characterization of a murine model of mucopolysaccharidosis II (Hunter syndrome) J. Inherit. Metab. Dis. 2007;30:924–934. doi: 10.1007/s10545-007-0641-8. [DOI] [PubMed] [Google Scholar]
- 25.Muenzer J., Lamsa J.C., Garcia A., Dacosta J., Garcia J., Treco D.A. Enzyme replacement therapy in mucopolysaccharidosis type II (Hunter syndrome): a preliminary report. Acta Paediatr. Suppl. 2002;91:98–99. doi: 10.1111/j.1651-2227.2002.tb03115.x. [DOI] [PubMed] [Google Scholar]
- 26.Azadeh M., Pan L., Qiu Y., Boado R. A rapid two-step iduronate-2-Sulfatatse enzymatic activity assay for MPSII pharmacokinetic assessment. JIMD Rep. 2018;38:89–95. doi: 10.1007/8904_2017_34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Pan P., Chen M., Zhang Z., Corte A.D., Souza C., Giugliani R., Pan L., Qiu Y., Amaravadi L., Wu J. A novel LC-MS/MS assay to quantify dermatan sulfate in cerebrospinal fluid as a biomarker for mucopolysaccharidosis II. Bioanalysis. 2018;10:825–838. doi: 10.4155/bio-2018-0025. [DOI] [PubMed] [Google Scholar]
- 28.Gleitz H.F., Liao A.Y., Cook J.R., Rowlston S.F., Forte G.M., D'Souza Z., O'Leary C., Holley R.J., Bigger B.W. Brain-targeted stem cell gene therapy corrects mucopolysaccharidosis type II via multiple mechanisms. EMBO Mol. Med. 2018;10 doi: 10.15252/emmm.201708730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Livak K.J., Schmittgen T.D. Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods. 2001;25:402–408. doi: 10.1006/meth.2001.1262. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
Restrictions apply to the availability of information and materials related to the viral vectors (cis-regulatory modules), which were used under license for this study. Please contact the corresponding author with any inquiries.







