throbber
Original Article
`
`Safety Evaluation of Lipid Nanoparticle–
`Formulated Modified mRNA in the Sprague-
`Dawley Rat and Cynomolgus Monkey
`
`Veterinary Pathology
`1-14
`ª The Author(s) 2017
`Reprints and permission:
`sagepub.com/journalsPermissions.nav
`DOI: 10.1177/0300985817738095
`journals.sagepub.com/home/vet
`
`Maja Sedic1, Joseph J. Senn1, Andy Lynn1, Michael Laska1, Mike Smith1,
`Stefan J. Platz2, Joseph Bolen3, Stephen Hoge1, Alex Bulychev1,
`Eric Jacquinet1, Victoria Bartlett4, and Peter F. Smith5
`
`Abstract
`The pharmacology, pharmacokinetics, and safety of modified mRNA formulated in lipid nanoparticles (LNPs) were evaluated after
`repeat intravenous infusion to rats and monkeys. In both species, modified mRNA encoding the protein for human erythropoietin
`(hEPO) had predictable and consistent pharmacologic and toxicologic effects. Pharmacokinetic analysis conducted following the
`first dose showed that measured hEPO levels were maximal at 6 hours after the end of intravenous infusion and in excess of 100-
`fold the anticipated efficacious exposure (17.6 ng/ml) at the highest dose tested.24 hEPO was pharmacologically active in both the
`rat and the monkey, as indicated by a significant increase in red blood cell mass parameters. The primary safety-related findings
`were caused by the exaggerated pharmacology of hEPO and included increased hematopoiesis in the liver, spleen, and bone
`marrow (rats) and minimal hemorrhage in the heart (monkeys). Additional primary safety-related findings in the rat included
`mildly increased white blood cell counts, changes in the coagulation parameters at all doses, as well as liver injury and release of
`interferon g–inducible protein 10 in high-dose groups only. In the monkey, as seen with the parenteral administration of cationic
`LNPs, splenic necrosis and lymphocyte depletion were observed, accompanied with mild and reversible complement activation.
`These findings defined a well-tolerated dose level above the anticipated efficacious dose. Overall, these combined studies indicate
`that LNP-formulated modified mRNA can be administered by intravenous infusion in 2 toxicologically relevant test species and
`generate supratherapeutic levels of protein (hEPO) in vivo.
`
`Keywords
`modified mRNA, lipid nanoparticle, toxicology, pharmacokinetics, drug discovery
`
`The promise of mRNA as a novel modality to deliver thera-
`peutic proteins in humans is vast, as evidenced by the growth
`and success of recombinant human therapeutic proteins over
`the last 3 decades, such as recombinant human insulin for the
`treatment of diabetes mellitus.17 Protein expression directed by
`exogenous mRNA offers many advantages over other nucleic
`acid–based concepts, as well as recombinant proteins. Potential
`advantages of mRNA over DNA-based technology include (1)
`no integration into the host genome thereby circumventing the
`risk of deleterious chromosomal changes, and (2) faster and
`more efficient expression with proper modifications, since
`mRNA therapeutics only require access to the cytoplasm. In
`comparison with recombinant proteins, mRNA would have
`lower manufacturing costs and could enable access to intracel-
`lular as well as cell membrane–bound therapeutic targets. The
`biggest challenges of mRNA technology are its potential for
`immunogenicity and its relatively poor in vivo stability. These
`challenges have been addressed through progress in chemistry
`and sequence engineering (eg, optimization of the 50 cap, 50-,
`
`and 30-untranslated regions and coding sequences) and through
`the use of specific nucleotide modifications.16,21,29
`Nucleotide-modified mRNA is nearly identical to naturally
`occurring mammalian mRNA, with the exception that certain
`nucleotides, normally present in mammalian mRNA, are
`partially or fully replaced with nucleosides, such as pyrimidine
`nucleosides—specifically, pseudouridine, 2-thiouridine,
`
`1Moderna Therapeutics, Cambridge, MA
`2AstraZeneca, Melbourn, Royston, UK
`3PureTech Health, Boston, MA
`4Akcea Therapeutics, Cambridge, MA
`5Alnylam Pharmaceuticals Inc, Cambridge, MA
`
`Supplementary material for this article is available online.
`
`Corresponding Author:
`Joseph J. Senn, Moderna Therapeutics, 200 Technology Square, Cambridge,
`MA 02139, USA.
`Email: Joe.Senn@modernatx.com
`
`ARBUTUS - EXHIBIT 2026
`Moderna Therapeutics, Inc. v. Arbutus Biopharma Corporation
`IPR2019-00554
`
`

`

`2
`
`Veterinary Pathology XX(X)
`
`5-methyl cytosine, or N1-methyl-pseudouridine.2,14,16,21,29
`These naturally occurring pyrimidine nucleotides are present
`in mammalian tRNA, rRNA, and small nuclear RNAs.20 Incor-
`poration of these nucleotides in place of the normal pyrimidine
`base has been shown to minimize the indiscriminate recogni-
`tion of exogenous mRNA by pathogen-associated molecular
`pattern receptors, such as toll-like receptors, retinoic acid–
`inducible gene 1, melanoma differentiation-associated protein
`5, nucleotide-binding oligomerization domain-containing pro-
`tein 2, and protein kinase R.7
`Given the lability of a naked mRNA molecule, the devel-
`opment of mRNA therapeutics has been further hampered by
`the lack of appropriate formulations for delivery and poten-
`tially as a targeting mechanism to a diseased organ or tissue.12
`However, the application of lipid-based nanoparticle delivery
`systems, initially developed for the in vivo delivery of siRNA,
`has enabled systemic administration of modified mRNA.22
`Adequate delivery of mRNA with lipid nanoparticles (LNPs)
`has been demonstrated for mRNA-based vaccines, where
`intramuscular injection of low doses of mRNA formulated
`in either LNPs or nanoemulsion induced immune protection
`from influenza and respiratory syncytial virus in mice, as well
`as cytomegalovirus and respiratory syncytial virus in mon-
`keys.9 Furthermore, a single administration of modified
`mRNA-LNP complexes in mice by various routes resulted
`in high, sustained protein production.19 Finally, Thess
`et al25 reported that repeated administration of unmodified
`mRNA in combination with the nonliposomal polymeric
`delivery system (TransIT) induced high systemic protein lev-
`els and strong physiologic responses in mice. These authors
`also noted similar observations following single-dose admin-
`istration of erythropoietin (EPO)–mRNA in LNPs to pigs and
`monkeys.
`LNPs have been reported to be clinically effective for the
`delivery of siRNA.6 The LNP vehicle is currently in late-phase
`clinical trials of a synthetic siRNA in patients suffering from
`transthyretin amyloidosis and has been well tolerated in this
`population.6 Therefore, considerable work has been done to
`understand the safety profile of systemic administration of
`siRNA-LNPs.3 Here, we set out to describe, for the first time,
`the pharmacology and toxicologic effects of repeated adminis-
`tration of hEPO-mRNA in LNPs in male Sprague-Dawley rats
`and female cynomolgus monkeys.
`
`Materials and Methods
`
`Animals and Husbandry
`
`The study plan and any amendments or procedures involving
`the care and use of animals in these studies were reviewed and
`approved by the Institutional Animal Care and Use Committee
`of Charles River Laboratories Preclinical Services (Montreal
`and Sherbrooke, Canada). During the study, the care and use of
`animals were conducted according to the guidelines of the US
`National Research Council and the Canadian Council on Ani-
`mal Care.
`
`Male Sprague-Dawley rats (Charles River Laboratories)
`were 11 to 12 weeks old and weighed between 390 and 497
`g at dose initiation. Animals were group housed in polycarbo-
`nate bins and separated during designated procedures. The
`temperature of the animal room was kept between 19C and
`25C, with humidity between 30% and 70%. The light cycle
`was 12 hours light and 12 hours dark, except during designated
`procedures. Animals were fed PMI Nutrition International Cer-
`tified Rodent Chow No. 5CR4 (14% protein) ad libitum
`throughout the in-life studies, except during designated proce-
`dures. Municipal tap water treated by reverse osmosis and
`ultraviolet irradiation was freely available to each animal via
`an automatic watering system. Environmental enrichment was
`provided to animals per standard operating procedures of
`Charles River Laboratories (Montreal, Canada), except during
`study procedures and activities.
`Female cynomolgus monkeys (Charles River Laboratories)
`were 1.5 to 6 years old and weighed 2.5 to 5.1 kg at the initia-
`tion of dosing. Animals were housed in stainless-steel cages
`and separated during designated procedures. The temperature
`of the animal room was kept between 20C and 26C, with
`humidity between 30% and 70%. The light cycle was 12 hours
`light and 12 hours dark except during designated procedures.
`Animals were fed PMI Nutrition International Certified Pri-
`mate Chow No. 5048 (25% protein). Municipal tap water
`treated by reverse osmosis and ultraviolet irradiation was freely
`available to each animal via an automatic watering system.
`Psychological and environmental enrichment was provided to
`animals per standard operating procedures of Charles River
`Laboratories (Montreal, Canada) except during study proce-
`dures and activities.
`
`Control, Test, and Reference Items
`
`An 850-nucleotide messenger RNA was prepared by in vitro
`transcription from a linearized DNA template with T7 RNA
`Polymerase. The DNA template encoded the T7 promoter, a 50
`untranslated region, the 579-nucleotide open reading frame
`encoding human EPO (hEPO) mature protein with signal
`sequence, a 30untranslated region, and a polyadenylated tail.
`The in vitro transcription was performed with the canonical
`nucleotides adenosine triphosphate and guanosine triphosphate
`and the modified nucleotides 1-methylpseudouridine tripho-
`sphate and 5-methylcytidine triphosphate. The mRNA contains
`a 50 Cap 1 structure, which consisted of 7-methylguanosine
`linked to the 50 nucleoside of the mRNA chain through a 50–
`50 triphosphate bridge and 20-O-methyl group present on the
`first nucleotide of the mRNA.23 The messenger RNA was pur-
`ified and buffer exchanged into low ionic strength buffer for
`formulation.18 The final mRNA had a calculated molecular
`weight of 277 786 Da.
`The mRNA-loaded LNPs were generated via stepwise etha-
`nol dilution, with an approach adapted from previously demon-
`strated methods.13,30 The LNP formulation was prepared by
`dissolving the lipids (6Z,9Z,28Z,31Z)-heptatriaconta-
`6,9,28,31-tetraen-19-yl 4-(dimethylamino)butanoate (MC3),
`
`

`

`Sedic et al
`
`3
`
`Table 1. Experimental Design for Safety Study: Rat.a
`
`Table 2. Experimental Design for Safety Study: Monkey.a
`
`Dose
`Level,
`mg/kgb
`
`Test
`Material
`
`Intravenous
`Administration
`10-min infusion, 2/wk
`PBS
`0
`mRNA EPO 0.03 10-min infusion, 2/wk
`10-min infusion, 2/wk
`mRNA EPO 0.1
`10-min infusion, 2/wk
`mRNA EPO 0.3
`10-min infusion, 1/wk
`mRNA EPO 0.3
`10-min infusion, 2/wk
`Empty LNP
`0.3
`
`Dose
`Concentration
`mg/ml
`
`Group
`No.
`
`Test
`Material
`
`Dose
`Level
`(mg/kg)b
`
`0
`0.006
`0.02
`0.06
`0.06
`0.06
`
`1
`2
`3
`4
`5
`6
`
`0
`PBS
`mRNA EPO 0.03
`mRNA EPO 0.1
`mRNA EPO 0.3
`mRNA EPO 0.3
`Empty LNP
`0.3
`
`Intravenous
`Administration
`60-min infusion (2/wk)
`60-min infusion (2/wk)
`60-min infusion (2/wk)
`60-min infusion (2/wk)
`60-min infusion (1/wk)
`60-min infusion (2/wk)
`
`Dose
`Concentration
`(mg/mL)
`
`0
`0.006
`0.02
`0.06
`0.06
`0.06
`
`Group
`No.
`
`1
`2
`3
`4
`5
`6
`
`Abbreviations: EPO, erythropoietin; LNP, lipid nanoparticle; PBS, phosphate-
`buffered saline.
`aNo. of males per group, n ¼ 24. Dose volume per group, 5 ml/kg. Dose rate
`per group, 30 ml/kg/h.
`bDose levels in terms of mRNA content. For group No. 6, the dose level is
`listed in terms of the same amount of lipid:mRNA ratio (by weight).
`
`Abbreviations: EPO, erythropoietin; LNP, lipid nanoparticle; PBS, phosphate-
`buffered saline.
`aNo. of females per group, n ¼ 3. Dose volume per group, 5 ml/kg. Dose rate
`per group, 5 ml/kg/h.
`bDose levels in terms of mRNA content. For group No. 6, the dose level is
`listed in terms of the same amount of lipid to mRNA ratio (by weight).
`
`1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), choles-
`terol, and 1,2-dimyristoyl-rac-glycerol, methoxypolyethylene
`glycol (PEG2000-DMG) in ethanol. The 4 lipids were prepared
`as a combined stock, with a total concentration of 12.5 mM
`(molar ratio of 50:10:38.5:1.5, MC3:DSPC:cholesterol:
`PEG2000-DMG). In brief, the solution containing lipids was
`mixed with an acidic aqueous buffer containing mRNA (0.18
`mg/ml, pH 4.0) in a T-mixer device. The resulting LNP
`dispersion was diluted and subsequently purified and concen-
`trated by tangential flow filtration. The formulation was
`filtered through a clarification filter (0.8/0.2 mm nominal).
`Prior to storage, the formulation was additionally filtered
`through 2 in-line sterile filters (0.2 mm) and aseptically filled
`into sterilized vials, stoppered, and capped. Empty LNPs were
`generated with a similar approach, whereby mRNA was
`excluded from the process.
`The final LNP lipid concentration was determined with an
`ultraperformance liquid chromatography system with online
`charged aerosol detection. The total concentrations of lipids
`in the final mRNA-LNPs and empty LNPs were 22.0 and
`13.1 mg/ml, respectively. The final mRNA content in hEPO
`LNPs was quantified by ultraviolet analysis, resulting in an
`mRNA concentration of 1.2 mg/ml. Measured lipid and mRNA
`concentration values enabled dilution with phosphate-buffered
`saline (PBS) to target levels for dosing (Tables 1, 2). Particle
`hydrodynamic diameters were determined by dynamic light
`scattering. Resulting diameters for mRNA and empty LNPs
`were 81 nm (0.08 polydispersity index) and 61 nm (0.10 poly-
`dispersity index), respectively. Total mRNA encapsulation was
`quantified with the Ribogreen assay (ThermoFisher Scientific).
`The final value for hEPO-mRNA in LNP encapsulation was
`97%. Additional information for the control, test, and reference
`items is provided in Supplemental Table 1.
`
`Male Rat Study Design
`
`Only male rats were used for this study, as there was no
`expected sex-specific differences in metabolism, distribution,
`or toxicity. The negative control, test, or reference items were
`
`administered over the course of 2 weeks in a 10-minute intra-
`venous (IV) infusion via a caudal vein at a dose level, dose
`volume, and frequency listed in Table 1. Dose levels for each
`study were based on previous pharmacology data demonstrat-
`ing production of efficacious levels of hEPO in the rat and
`cynomolgus monkey at doses 0.03 mg/kg of mRNA. Based
`on pharmacokinetic (PK) data indicating predictable increases
`in protein expression with dose, the mid- and high doses for
`these studies were selected to achieve significant multiples of
`the efficacious dose level. Since PK behavior and physiologic
`consequences are well defined for EPO therapy, we employed a
`similar approach in our study of hEPO-mRNA in LNPs.15,16,25
`Each infused dose was administered with a temporary indwel-
`ling catheter inserted in a caudal vein connected to an injection
`set and infusion pump. The animals were temporarily
`restrained for the dose administration and not sedated. The dose
`volume for each animal was based on the most recent body
`weight measurement. The first day of dosing was designated as
`day 1. Six males per group were used for toxicity assessment,
`12 males per group for immunology assessment, and 6 males
`per group for PK / pharmacodynamic (PD) assessment. The
`following end points were evaluated: clinical signs (including
`observations of the infusion sites), body weights, food con-
`sumption, PK/PD, clinical pathology (hematology, coagula-
`tion, and clinical chemistry), macro- and microscopic
`examination of tissues, and immunotoxicology markers: hista-
`mine, interleukin 6 (IL-6), interferon g–induced protein 10 (IP-
`10), tumor necrosis factor a (TNF-a), interferon a (IFN-a), and
`complement (C3).
`Blood samples were collected from nonfasted animals and
`analyzed for hematology on day 9 and from fasted animals for
`hematology, coagulation, and clinical chemistry on day 16 (at
`necropsy). For PD (hEPO) or PK (hEPO-mRNA), blood sam-
`ples were collected and processed to plasma prestudy and at 2,
`6, 24, and 48 hours after the end of injection/infusion on days 1
`and 15. After processing, the plasma samples were stored in a
`freezer set to maintain 80C until analyzed. For cytokines (ie,
`IL-6, IP-10, TNF-a), histamine, and complement (C3) analysis,
`blood samples were collected prestudy and at 5 minutes and 2,
`
`

`

`4
`
`Veterinary Pathology XX(X)
`
`6, and 24 hours after the end of injection/infusion on days 1 and
`15 in K3EDTA tubes and processed to plasma or serum (no
`anticoagulant) for IFN-a analysis.
`
`Female Monkey Study Design
`
`Female monkeys were used for this study, as there was no
`expected sex-specific differences in metabolism, distribu-
`tion, or toxicity. The negative control, test, and reference
`items were administered over the course of 2 weeks in a 60-
`minute IV infusion via an appropriate peripheral vein (eg,
`saphenous or brachial) at the dose level, dose volume, and
`frequency listed in Table 2. The dose volume for each ani-
`mal was based on the most recent body weight measure-
`ment. The animals were temporarily restrained (on a sling
`or a chair) for the dose administration and not sedated. Each
`infused dose was administered with a temporary indwelling
`catheter inserted in a peripheral vein connected to an injec-
`tion set and infusion pump. The first day of dosing was
`designated as day 1. The end points in this study included
`clinical signs (including observation of the infusion sites),
`body weights, food consumption, PK/PD, clinical pathology
`(hematology, coagulation, and serum chemistry), macro-
`and microscopic examination of tissues, and selected cyto-
`kines (interleukin 1 b [IL-1b], IL-6, TNF-a, and IP-10) and
`complement (C3a and C5b-9).
`Blood samples were collected from overnight-fasted ani-
`mals for hematology, coagulation, and clinical chemistry
`parameters at predose (baseline) and on day 16. Addition-
`ally, blood was analyzed on day 8 for hematology para-
`meters only. For PK/PD assessments, blood samples were
`collected and processed to plasma at the following time
`points: predose; 2, 6, 24, and 48 hours after the first dose;
`and 6 hours after subsequent dosing occasions. After pro-
`cessing, the plasma samples were stored in a freezer set to
`80C until analyzed. Blood samples were collected in
`K3EDTA tubes and processed to plasma for analysis of
`cytokines (ie, IL-1b, IL-6, TNF-a) and complement (ie,
`C3a and C5b-9) or to serum for analysis of IFN-a and IP-
`10 at the following time points for all groups: predose; at 2,
`6, and 24 hours after the end of infusion on day 1; and at 2,
`6, and 24 hours after the end of infusion on day 15. Addi-
`tionally, for complement analysis only, blood samples were
`collected 2, 6, and 24 hours after the end of infusion on day
`4 (groups 1–4 and 6).
`
`Histamine, Cytokine and Complement Levels
`
`Histamine levels in the rat plasma were determined with the
`Histamine EIA Kit (IM-2015; Immunotech). Serum levels of
`IFNa were determined with the Rat IFNa ELISA Kit (KT-
`60242; Kamiya Biomedical Company) and the Human IFNa
`Multi-subtype ELISA Kit (41105-1 or 41105-2; PBL Biome-
`dical Laboratories). IL-6, IP-10, and TNFa in rat plasma were
`determined with the Rat Cytokine/Chemokine Magnetic Panel
`Kit (RECYMAG-65K; Millipore). IL1b, IL-6, and TNFa in
`monkey plasma were determined with the Non-Human Primate
`Cytokine/Chemokine Magnetic Panel Kit (PRCYTOMAG-40K;
`Millipore). IP-10 in the monkey serum was determined with
`the Monkey IP-10 Singleplex Magnetic Kit (LHB0001;
`Invitrogen). C3 levels in the rat plasma were determined with
`the Rat C3 ELISA Kit (GWB-A8B8AF; Genway). C3a levels
`in the monkey plasma were determined with the Human C3a
`EIA Kit (A031; Quidel). C5b-9 levels in the monkey plasma
`were determined with the Human C5b-9 ELISA Kit (558315;
`BD Bioscience).
`
`Histopathology
`
`Representative samples of the following tissues from all ani-
`mals were preserved in 10% neutral buffered formalin: bone
`marrow (sternum), heart, infusion site (last dose), kidney, liver,
`lung, spleen, and thymus. Tissues were embedded in paraffin,
`sectioned, mounted on glass slides, and stained with hematox-
`ylin and eosin. The histopathologic evaluation was internally
`peer reviewed.
`
`hEPO bDNA
`
`The bioanalysis of plasma samples for quantification of hEPO-
`mRNA levels was conducted at AxoLabs according to the
`bDNA method for mRNA detection developed by QuantiGene
`(Affymetrix).26 Briefly, plasma samples were directly diluted
`in lysis buffer. On each bDNA plate, including a customized
`assay-specific set of probes, a dilution curve was pipetted with
`spiked standards into untreated plasma. Signal amplification
`was carried out with oligonucleotides bound to the enzyme
`alkaline phosphatase. The calculated amount in picograms was
`normalized to the amount of plasma in the lysate and to the
`amount of lysate applied to the plate. Since measurements in
`the PBS-treated control group were within the background
`level range, cross-reactivity of hEPO-mRNA to rat or monkey
`EPO mRNA was considered negligible.
`
`Clinical Pathology
`
`hEPO ELISA
`
`Hematology parameters were measured with Bayer Advia 120
`Automated Hematology Analyzer (Siemens Healthcare). Stan-
`dard coagulation parameters were measured on a START 4
`Compact Stago Analyzer (Diagnostica Stago). Standard clini-
`cal chemistry parameters were measured with Modular Analy-
`tics (Roche/Hitachi).
`
`hEPO levels were measured with a human EPO Sandwich
`ELISA Kit (01630; Stemcell Technologies). For this assay, the
`lower and upper limits of quantitation were 12 and 800 pg/ml,
`respectively. Since predose measurements were within the
`background level range, cross-reactivity of hEPO to rat or
`monkey EPO was considered negligible.
`
`

`

`Sedic et al
`
`5
`
`Data Analysis and Reporting
`
`The toxicokinetic parameters of human modified hEPO-
`mRNA and its expressed protein in plasma were calculated
`with a noncompartmental approach in WinNonlin Phoenix
`64, version 6.3 (Pharsight). Dose-normalized maximum serum
`concentration (Cmax/dose) and area under the curve (AUC/
`dose) were determined by dividing the respective parameters
`by dose and calculated by either WinNonlin or Excel. The
`mean, standard deviation, and percentage coefficient of varia-
`tion of the toxicokinetic parameters were calculated in Win-
`Nonlin. All reported values were rounded to either 3 significant
`figures or 1 decimal place (time to reach maximum serum
`concentration [Tmax], half-life [t1/2]).
`
`Results
`
`Administration of hEPO-mRNA in LNP Results in
`Detection of Significant Serum hEPO Levels and
`Corresponding PD Effects in the Rat and Monkey
`
`Toxicokinetic analysis in the rat revealed that hEPO-mRNA
`had a moderate half-life (2.9–5.7 hours) and low clearance
`(49.0–97.2 ml/h/kg; Fig. 1, Table 3). The Cmax/dose values
`were consistent among the 4 dose groups, ranging from 2270
`to 3320 ng/ml/mg/kg (Table 3). Measured hEPO levels were
`maximal approximately 6 hours after the 10-minute infusion
`(Fig. 2, Table 4). The AUC values (for hEPO-mRNA and
`hEPO) increased in more than a dose-proportional manner,
`between 0.03 and 0.3 mg/kg (Table 4). Plasma samples col-
`lected at 6 hours after each dose indicated that hEPO levels
`were constant at Cmax at all dose levels until day 15, when
`measured hEPO levels were significantly decreased in the mid-
`and high-dosed groups (Fig. 3). Consistent with literature data,1
`peak reticulocytosis (PD marker described later) was observed
`by day 9, and levels remained elevated during the 15-day
`period. Overall, these results indicate that plasma concentra-
`tions of hEPO were mostly consistent throughout the study and
`exhibited greater-than-dose-proportional increases in AUC
`after IV administration.
`Significant increases were noted in red blood cell and asso-
`ciated parameters (hemoglobin, hematocrit) in all male rat
`groups dosed with hEPO-mRNA in LNPs as compared with
`the PBS group and the group dosed with empty LNPs. Inter-
`estingly, the changes in red blood cell parameters (except mean
`corpuscular volume) were similar across all hEPO-mRNA-
`dosed groups and did not seem to be dose related (Fig. 4, Suppl.
`Fig. 1). In addition, dose-dependent increases in platelet counts
`and reticulocytes were noted, particularly at the highest doses
`administered twice weekly (Suppl. Fig. 1). Overall, these
`results indicate that repeated administration of hEPO-mRNA
`in LNPs achieves physiologically relevant and persistent hEPO
`levels that result in significant changes in precursor cells and
`mature red blood cell count at doses as low as 0.03 mg/kg.
`Like in male rats, toxicokinetic findings in female monkeys
`indicated that the total exposure (AUC) to hEPO-mRNA and
`
`Figures 1–3. Plasma concentration of hEPO-mRNA (ng/ml; Fig. 1)
`and hEPO (ng/ml; Figs. 2, 3) in rats. Graphs represent mean values (n ¼
`6); error bars indicate SD. Following a 10-minute infusion, peak plasma
`concentrations of hEPO-mRNA appear to occur at approximately 2
`hours, while peak plasma concentrations of hEPO are approximately
`at 6 hours. Note that hEPO levels appear constant at all dose levels
`until day 15. hEPO, human erythropoietin; Q7D, 1 dose per week.
`
`

`

`6
`
`Veterinary Pathology XX(X)
`
`Table 3. Toxicokinetic Values for hEPO-mRNA in the Rat.a
`
`Dose, mg/kg
`
`t1/2, h
`
`Cmax, ng/ml
`
`Cmax/Dose, ng/ml
`
`AUC, h  ng/ml
`
`AUC/Dose, h  ng/ml
`
`Cl, ml/h/kg
`
`0.03
`0.1
`0.3
`0.3b
`
`5.7
`4.3
`4
`2.9
`
`92.6
`227
`902
`995
`
`3090
`2270
`3010
`3320
`
`309
`1460
`5450
`6120
`
`10,300
`14,600
`18,200
`20,400
`
`97.2
`68.5
`55.1
`49
`
`Abbreviations: AUC, area under the curve; Cl, clearance; Cmax, maximum serum concentration; hEPO, human erythropoietin; t1/2, half-life; Tmax, time to reach
`maximum serum concentration.
`aTmax per dose, 2 hours.
`bOne dose per week.
`
`Table 4. Toxicokinetic Values for hEPO in the Rat.a
`AUC, h  ng/ml
`
`Cmax, ng/ml
`
`Dose, mg/kg
`
`0.03
`0.1
`0.3
`0.3b
`
`77.1
`154
`3540
`2340
`
`1590
`4480
`44 400
`34 100
`
`t1/2, h
`
`6.4
`8.7
`6.1
`6.6
`
`Abbreviations: AUC, area under the curve; Cmax, maximum serum
`concentration; hEPO, human erythropoietin; t1/2, half-life; Tmax, time to reach
`maximum serum concentration.
`aTmax per dose, 6 hours.
`bOne dose per week.
`
`hEPO increased in a dose-related manner after IV administra-
`tion of 0.1 to 0.3 mg/kg/d of hEPO-mRNA in LNPs (Figs. 5, 6;
`Tables 5, 6). There was a notable difference in hEPO-mRNA
`exposure after the first dose between the groups administered
`0.3 mg/kg twice and once per week, which could be due to the
`small sample size and large intragroup variability (Table 5). In
`addition, the results showed that hEPO-mRNA had a relatively
`long half-life (5.9–9.3 hours) and low clearance (9.03–27.0 ml/
`h/kg; Table 5). After a 60-minute infusion of hEPO-mRNA in
`LNPs, maximum plasma concentration of hEPO was estimated
`to occur between 6 and 24 hours (Table 6). The delay in esti-
`mated Tmax observed in group 5 could be a consequence of
`
`Figure 4. Red blood cell (RBC) mass parameters in rats: RBCs (106 cells/ml), hemoglobin (HGB; g/dl), hematocrit (HCT; %), and erythrocyte
`distribution width (RDW; %). Graphs represent mean values (n ¼ 6); error bars indicate SD. Ordinary one-way analysis of variance (multiple
`comparisons) was used to calculate the P values. *P < .05. LNP, lipid nanoparticle; PBS, phosphate-buffered saline; Q7D, 1 dose per week.
`
`

`

`Sedic et al
`
`7
`
`increased hEPO-mRNA exposure in this particular group or, as
`mentioned before, could be an artifact of high variability within
`that group. Also, as in male rats, serum hEPO concentrations at
`Cmax were well maintained at all dose levels throughout the
`study until day 15, when measured hEPO levels significantly
`decreased in the mid- and high-dose groups (Fig. 7).
`As expected, hEPO expression in monkeys led to significant
`changes in red blood cell mass. By day 8 of the study, absolute
`reticulocyte counts increased in all dose groups (Fig. 8). By the
`end of the study, a significant spike was noted in all other red
`blood cell parameters, such as absolute red blood cells, hema-
`tocrit, hemoglobin, and erythrocyte distribution width, whereas
`reticulocytes returned to baseline levels. Like in rats, the
`increases in red blood cell parameters were similar across all
`groups and did not seem to be dose related. These changes were
`not observed in the group dosed with empty LNPs.
`
`Tolerability of hEPO-mRNA in LNPs in Male Rats
`
`There were no notable clinical observations or body weights/
`food consumption changes in any group (data not shown).
`Slight changes were observed in coagulation parameters. Spe-
`cifically, activated partial thromboplastin time was prolonged
`in all animals dosed with hEPO-mRNA in LNPs, and pro-
`thrombin time was prolonged in all animals dosed with
`hEPO-mRNA twice per week. In addition, fibrinogen levels
`were elevated for animals receiving 0.3 mg/kg of hEPO-
`mRNA once and twice per week (Suppl. Fig. 2).
`Assessment of hematologic parameters on days 9 and 16 of
`the study indicated that white blood cells increased at doses
`0.1 mg/kg of hEPO-mRNA in LNPs given twice weekly.
`Consistent with this, neutrophil, monocyte, and atypical lym-
`phocyte counts were elevated across all groups dosed with
`hEPO-mRNA in LNPs. Interestingly, administration of
`hEPO-mRNA in LNPs once weekly or empty LNPs did not
`elicit the same increase in white blood cells (Fig. 9).
`In addition, histamine release and serum levels of cytokines
`were evaluated. IP-10 was elevated 6 to 24 hours postdose on
`days 1 and 15 only in the groups dosed with 0.3 mg/kg of
`hEPO-mRNA in LNPs once and twice weekly (Fig. 10). No
`changes in IP-10 or histamine were seen in the group given
`empty LNPs (Fig. 10 and data not shown). No changes in other
`cytokines (IL-6, TNF-a, IFN-a) or complement (C3) were
`observed in any study group (data not shown).
`Administration of hEPO-mRNA in LNPs at doses 0.03
`mg/kg resulted in several macro- and microscopic findings in
`the spleen, bone marrow, liver, lungs, and stomach. Primary
`findings were considered to be related to increased hEPO
`expression and included an increase in extramedullary hema-
`topoiesis in the spleen, liver, and bone marrow (Figs. 11–16).1
`In addition, macroscopic enlargement of the spleen was noted.
`These findings correspond to the hematologic changes (reticu-
`locyte counts, red blood cells, and red cell mass parameters).
`Minimal hemorrhage in the lung and glandular stomach was
`noted at all doses of hEPO-mRNA in LNPs, which corresponds
`to macroscopic observations of dark foci in these tissues.
`
`Figures 5–7. Plasma concentrations of hEPO-mRNA (ng/ml; Fig. 5)
`and hEPO (ng/ml; Figs. 6, 7) in monkeys. Graphs represent mean values
`(n ¼ 3); error bars indicate SD. Following a 60-minute infusion, peak
`plasma concentrations of hEPO-mRNA appear to be at 2 hours, while
`peak plasma concentrations of hEPO are approximately at 6 hours.
`Note that hEPO levels appear relatively constant at all dose levels until
`day 15. hEPO, human erythropoietin; Q7D, 1 dose per week.
`
`

`

`8
`
`Veterinary Pathology XX(X)
`
`Table 5. Toxicokinetic Values for hEPO-mRNA in the Monkey.a
`AUC, h  ng/ml
`
`Cmax, ng/mL
`
`Dose, mg/kg Mean
`
`SD
`
`Mean Cmax/Dose, ng/mL Mean
`
`SD
`
`t1/2, h
`Mean AUC/Dose, h  ng/ml Mean
`
`SD
`
`Cl, mL/h/kg
`
`Mean
`
`SD
`
`0.03
`0.1
`0.3
`0.3b
`
`715
`882
`3270
`9240
`
`379
`904
`2620
`1860
`
`23 800
`8820
`10 900
`30 800
`
`4090
`4440
`19 900
`49 900
`
`1870
`3140
`15 400
`13 600
`
`136 000
`44 400
`66 300
`166 000
`
`5.89
`9.31
`6.99
`7.36
`
`0.554
`8.12
`1.22
`6.51
`
`9.03
`27
`21.3
`6.33
`
`5.57
`13
`12.8
`1.87
`
`Abbreviations: AUC, area under the curve; Cl, clearance; Cmax, maximum serum concentration; hEPO, human erythropoietin; t1/2, half-life; Tmax, time to reach
`maximum serum concentration.
`aMean Tmax per dose, 2 hours.
`bOne dose per week.
`
`Table 6. Toxicokinetic Values for hEPO in the Monkey.
`AUC, h  ng/ml
`
`Cmax, ng/ml
`
`Dose, mg/kg Mean Tmax, h Mean
`
`SD
`
`Mean
`
`SD
`
`0.03
`0.1
`0.3
`0.3a
`
`6
`6
`6
`24
`
`30.6
`210
`283
`253
`
`7.42
`187
`111
`49.4
`
`600
`4240
`6660
`8170
`
`39.8
`2530
`915
`1550
`
`Abbreviations: AUC, area under the curve; Cmax, maximum serum
`concentration; hEPO, human erythropoietin; Tmax, time to reach maximum
`serum concentration.
`aOne dose per week.
`
`Additional findings observed in groups dosed with hEPO-
`mRNA in LNPs or empty LNPs included increased mononuc-
`lear cell infiltration and a minimal to moderate extent of
`single-cell necrosis in the liver at doses 0.1 mg/kg (Figs. 17,
`18). At 0.3 mg/kg per dose of hEPO-mRNA, minimal to mild
`hypertrophy/hyperplasia of the sinusoidal endothelial cells was
`observed in the liver. These liver findings were accompanied
`with mild elevations in alanine aminotransferase (ALT) and
`aspartate aminotransferase (AST; Fig. 19). Overall, these data
`suggest that doses 0.1 mg/kg result in minor liver injury that
`seems to be primarily driven by the vehicle (LNPs).
`
`Tolerability of hEPO-mRNA in LNPs in Female Monkey
`
`There were no clinical signs or effects on body

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket