Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters

Publication year range
1.
N Engl J Med ; 387(12): 1099-1110, 2022 09 22.
Article in English | MEDLINE | ID: mdl-36129998

ABSTRACT

BACKGROUND: The intrathecally administered antisense oligonucleotide tofersen reduces synthesis of the superoxide dismutase 1 (SOD1) protein and is being studied in patients with amyotrophic lateral sclerosis (ALS) associated with mutations in SOD1 (SOD1 ALS). METHODS: In this phase 3 trial, we randomly assigned adults with SOD1 ALS in a 2:1 ratio to receive eight doses of tofersen (100 mg) or placebo over a period of 24 weeks. The primary end point was the change from baseline to week 28 in the total score on the ALS Functional Rating Scale-Revised (ALSFRS-R; range, 0 to 48, with higher scores indicating better function) among participants predicted to have faster-progressing disease. Secondary end points included changes in the total concentration of SOD1 protein in cerebrospinal fluid (CSF), in the concentration of neurofilament light chains in plasma, in slow vital capacity, and in handheld dynamometry in 16 muscles. A combined analysis of the randomized component of the trial and its open-label extension at 52 weeks compared the results in participants who started tofersen at trial entry (early-start cohort) with those in participants who switched from placebo to the drug at week 28 (delayed-start cohort). RESULTS: A total of 72 participants received tofersen (39 predicted to have faster progression), and 36 received placebo (21 predicted to have faster progression). Tofersen led to greater reductions in concentrations of SOD1 in CSF and of neurofilament light chains in plasma than placebo. In the faster-progression subgroup (primary analysis), the change to week 28 in the ALSFRS-R score was -6.98 with tofersen and -8.14 with placebo (difference, 1.2 points; 95% confidence interval [CI], -3.2 to 5.5; P = 0.97). Results for secondary clinical end points did not differ significantly between the two groups. A total of 95 participants (88%) entered the open-label extension. At 52 weeks, the change in the ALSFRS-R score was -6.0 in the early-start cohort and -9.5 in the delayed-start cohort (difference, 3.5 points; 95% CI, 0.4 to 6.7); non-multiplicity-adjusted differences favoring early-start tofersen were seen for other end points. Lumbar puncture-related adverse events were common. Neurologic serious adverse events occurred in 7% of tofersen recipients. CONCLUSIONS: In persons with SOD1 ALS, tofersen reduced concentrations of SOD1 in CSF and of neurofilament light chains in plasma over 28 weeks but did not improve clinical end points and was associated with adverse events. The potential effects of earlier as compared with delayed initiation of tofersen are being further evaluated in the extension phase. (Funded by Biogen; VALOR and OLE ClinicalTrials.gov numbers, NCT02623699 and NCT03070119; EudraCT numbers, 2015-004098-33 and 2016-003225-41.).


Subject(s)
Amyotrophic Lateral Sclerosis , Oligonucleotides, Antisense , Superoxide Dismutase-1 , Adult , Amyotrophic Lateral Sclerosis/blood , Amyotrophic Lateral Sclerosis/cerebrospinal fluid , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Double-Blind Method , Humans , Injections, Spinal , Neurofilament Proteins/blood , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , Recovery of Function/drug effects , Superoxide Dismutase-1/cerebrospinal fluid , Superoxide Dismutase-1/genetics
2.
N Engl J Med ; 383(2): 109-119, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32640130

ABSTRACT

BACKGROUND: Tofersen is an antisense oligonucleotide that mediates the degradation of superoxide dismutase 1 (SOD1) messenger RNA to reduce SOD1 protein synthesis. Intrathecal administration of tofersen is being studied for the treatment of amyotrophic lateral sclerosis (ALS) due to SOD1 mutations. METHODS: We conducted a phase 1-2 ascending-dose trial evaluating tofersen in adults with ALS due to SOD1 mutations. In each dose cohort (20, 40, 60, or 100 mg), participants were randomly assigned in a 3:1 ratio to receive five doses of tofersen or placebo, administered intrathecally for 12 weeks. The primary outcomes were safety and pharmacokinetics. The secondary outcome was the change from baseline in the cerebrospinal fluid (CSF) SOD1 concentration at day 85. Clinical function and vital capacity were measured. RESULTS: A total of 50 participants underwent randomization and were included in the analyses; 48 participants received all five planned doses. Lumbar puncture-related adverse events were observed in most participants. Elevations in CSF white-cell count and protein were reported as adverse events in 4 and 5 participants, respectively, who received tofersen. Among participants who received tofersen, one died from pulmonary embolus on day 137, and one from respiratory failure on day 152; one participant in the placebo group died from respiratory failure on day 52. The difference at day 85 in the change from baseline in the CSF SOD1 concentration between the tofersen groups and the placebo group was 2 percentage points (95% confidence interval [CI], -18 to 27) for the 20-mg dose, -25 percentage points (95% CI, -40 to -5) for the 40-mg dose, -19 percentage points (95% CI, -35 to 2) for the 60-mg dose, and -33 percentage points (95% CI, -47 to -16) for the 100-mg dose. CONCLUSIONS: In adults with ALS due to SOD1 mutations, CSF SOD1 concentrations decreased at the highest concentration of tofersen administered intrathecally over a period of 12 weeks. CSF pleocytosis occurred in some participants receiving tofersen. Lumbar puncture-related adverse events were observed in most participants. (Funded by Biogen; ClinicalTrials.gov number, NCT02623699; EudraCT number, 2015-004098-33.).


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides/administration & dosage , Superoxide Dismutase-1/cerebrospinal fluid , Adult , Amyotrophic Lateral Sclerosis/cerebrospinal fluid , Amyotrophic Lateral Sclerosis/genetics , Disease Progression , Dose-Response Relationship, Drug , Double-Blind Method , Female , Headache/chemically induced , Humans , Injections, Spinal/adverse effects , Intermediate Filaments , Leukocytosis/chemically induced , Male , Middle Aged , Mutation , Oligonucleotides/adverse effects , Oligonucleotides/pharmacokinetics , Oligonucleotides, Antisense/adverse effects , Oligonucleotides, Antisense/pharmacokinetics , Superoxide Dismutase-1/genetics , Vital Capacity
3.
J Pharmacokinet Pharmacodyn ; 50(1): 45-62, 2023 02.
Article in English | MEDLINE | ID: mdl-36600109

ABSTRACT

Clinical Dementia Rating-Sum of Boxes (CDR-SB) assessments from two Phase 3 studies (ENGAGE and EMERGE) of aducanumab in subjects with early Alzheimer's disease (AD) were pooled to develop an exposure-response (ER) model. A linear model in the logit-transformed scaled CDR-SB best characterized the time profile for placebo- and aducanumab-treated subjects, with concentration as the exposure metric. The model allowed delineation of slow (4%), typical (86%), and fast (10%) progressing subpopulations in the data. The estimated drug effect on the disease progression rate was significant, 2.05 L/(g·year), with a 95% confidence interval (1.60, 2.50) that did not include zero. Following an evaluation of a series of ER model forms including differential drug and null effects either between the studies or among the three progression classes, the final ER model with a common (pooled) estimate of the drug effect between the studies and among the three progression classes was considered parsimonious. The final model provides supportive evidence that the two studies demonstrate a common intrinsic pharmacology. None of the identified covariates (Mini-Mental State Examination-BL score and Asian race) were clinically meaningful. Finally, simulations demonstrated that the intrinsic pharmacology remained consistent between the two Phase 3 studies.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/drug therapy , Mental Status and Dementia Tests , Disease Progression
4.
J Pharmacokinet Pharmacodyn ; 48(5): 639-654, 2021 10.
Article in English | MEDLINE | ID: mdl-33991294

ABSTRACT

Antisense oligonucleotides (ASOs) are promising therapeutic agents for a variety of neurodegenerative and neuromuscular disorders, e.g., Alzheimer's, Parkinson's and Huntington's diseases, spinal muscular atrophy (SMA) and amyotrophic lateral sclerosis (ALS), caused by genetic abnormalities or increased protein accumulation. The blood-brain barrier (BBB) represents a challenge to the delivery of systemically administered ASOs to the relevant sites of action within the central nervous system (CNS). Intrathecal (IT) delivery, in which drugs are administered directly into the cerebrospinal fluid (CSF) space, enables to bypass the BBB. Several IT-administered ASO therapeutics have already demonstrated clinical effect, e.g., nusinersen (SMA) and tofersen (ALS). Due to novelty of IT dosing for ASOs, very limited pharmacokinetic (PK) data is available and only a few modeling reports have been generated. The objective of this work is to advance fundamental understanding of whole-body distribution of IT-administered ASOs. We propose a physiologically-based pharmacokinetic modeling approach to describe the distribution along the neuroaxis based on PK data from non-human primate (NHP) studies. We aim to understand the key processes that drive and limit ASO access to the CNS target tissues. To elucidate the trade-off between parameter identifiability and physiological plausibility of the model, several alternative model structures were chosen and fitted to the NHP data. The model analysis of the NHP data led to important qualitative conclusions that can inform projection to human. In particular, the model predicts that the maximum total exposure in the CNS tissues, including the spinal cord and brain, is achieved within two days after the IT injection, and the maximum amount absorbed by the CNS tissues is about 4% of the administered IT dose. This amount greatly exceeds the CNS exposures delivered by systemic administration of ASOs. Clearance from the CNS is controlled by the rate of transfer from the CNS tissues back to CSF, whereas ASO degradation in tissues is very slow and can be neglected. The model also describes local differences in ASO concentration emerging along the spinal CSF canal. These local concentrations need to be taken into account when scaling the NHP model to human: due to the lengthier human spinal column, inhomogeneity along the spinal CSF may cause even higher gradients and delays potentially limiting ASO access to target CNS tissues.


Subject(s)
Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/pharmacokinetics , Animals , Biological Transport/physiology , Blood-Brain Barrier/metabolism , Central Nervous System/metabolism , Humans , Injections, Spinal/methods , Primates
5.
J Pharmacokinet Pharmacodyn ; 47(3): 255-266, 2020 06.
Article in English | MEDLINE | ID: mdl-32335844

ABSTRACT

A population pharmacokinetic/pharmacodynamic (popPK/PD) model for BIIB059 (anti-blood dendritic cell antigen 2 [anti-BDCA2]), a humanized immunoglobulin G1 monoclonal antibody currently under development for the treatment of SLE and CLE, is presented. BIIB059 binds BDCA2, a plasmacytoid dendritic cell (pDC)-specific receptor that inhibits the production of IFN-I and other inflammatory mediators when ligated. Phase 1 PK and PD data of healthy adult volunteers (HV, n = 87) and SLE subjects (n = 22) were utilized for the development of the popPK/PD model. The data included single and multiple dosing of intravenous and subcutaneous BIIB059. BDCA2 internalization (PD marker) was measured for all subjects by monitoring reduction of BDCA2 on pDC cell surface and used for development of the popPD model. A two-compartment popPK model with linear plus non-linear elimination was found to best describe BIIB059 PK. BDCA2 levels were best captured using an indirect response model with stimulation of the elimination of BDCA2. Clearance in SLE subjects was 25% higher compared to HV (6.87 vs 5.52 mL/h). Bodyweight was identified as only other covariate on clearance and central volume. The estimates of EC50 and Emax were 0.35 µg/mL and 8.92, respectively. No difference in EC50 and Emax was observed between SLE and HV. The popPK/PD model described the data accurately, as evaluated by pcVPCs and bootstrap. The presented popPK/PD model for BIIB059 provides valuable insight into the dynamics and dose-response relationship of BIIB059 for the treatment of SLE and CLE and was used to guide dose selection for the Phase 2 clinical study (NCT02847598).


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Lectins, C-Type/antagonists & inhibitors , Lupus Erythematosus, Cutaneous/drug therapy , Lupus Erythematosus, Systemic/drug therapy , Membrane Glycoproteins/antagonists & inhibitors , Models, Biological , Receptors, Immunologic/antagonists & inhibitors , Administration, Intravenous , Adult , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Area Under Curve , Biological Availability , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Half-Life , Humans , Injections, Subcutaneous , Lectins, C-Type/immunology , Lupus Erythematosus, Cutaneous/blood , Lupus Erythematosus, Cutaneous/immunology , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/immunology , Male , Membrane Glycoproteins/immunology , Metabolic Clearance Rate , Middle Aged , Receptors, Immunologic/immunology
6.
J Pharmacokinet Pharmacodyn ; 45(6): 817-827, 2018 12.
Article in English | MEDLINE | ID: mdl-30377889

ABSTRACT

BIIB059 is a novel humanized monoclonal antibody (mAb) that is currently under development for the treatment of Systemic Lupus Erythematosus and Cutaneous Lupus Erythematosus. BIIB059 is targeted against the blood dendritic cell antigen 2 (BDCA2), a receptor exclusively expressed on the surface of plasmacytoid dendritic cells (pDCs). Herein, we utilized pre-clinical pharmacokinetic (PK) and pharmacodynamic (PD) data to develop a non-human primate (NHP) model and to address whether the NHP model can be successfully scaled to predict the human PK/PD. In particular, PK data from 17 cynomolgus monkeys were utilized for PK model development, wherein BIIB059 was administered intravenously (1 and 10 mg/kg single-dosing and 5 mg/kg multiple-dosing) or subcutaneously (0.2 and 7.5 mg/kg single-dosing). Additionally, PD data (BDCA2 receptor density on pDCs) from 6 cynomolgus monkeys were used for the development of the PD model. The developed NHP two-compartment PK model, linked with an indirect response PD model, was subsequently scaled to humans by combining traditional allometric PK scaling with sensitivity-analysis-driven scaling of the PD. The scaled PK/PD model was then used to simulate the human PK/PD for different dose levels. When clinical data from the BIIB059 Phase I study became available, they were used to evaluate the predictability of the scaled PK/PD model and the model simulations were in agreement with the clinical data. Therefore, the presented approach is suggested to be employed in scaling pre-clinical mAb models to support the selection of safe first-in-human doses and, more broadly, the prediction of PK/PD in the clinic.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Lectins, C-Type/antagonists & inhibitors , Membrane Glycoproteins/antagonists & inhibitors , Models, Biological , Receptors, Immunologic/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Body Weight , Computer Simulation , Drug Evaluation, Preclinical , Humans , Immunoglobulin G , Lupus Erythematosus, Cutaneous/drug therapy , Lupus Erythematosus, Systemic/drug therapy , Macaca fascicularis
7.
J Pharmacokinet Pharmacodyn ; 44(3): 263-275, 2017 06.
Article in English | MEDLINE | ID: mdl-28251386

ABSTRACT

Natalizumab, a human immunoglobulin monoclonal antibody that targets α4ß1/α4ß7 integrin, is an effective therapy approved for the treatment of multiple sclerosis (MS). The objective of this analysis was to develop a population exposure-response model utilizing gadolinium-enhancing (Gd) lesion count data from four clinical studies and annualized relapse rate (ARR) data from three clinical studies. The natalizumab exposures were derived for the individuals using a population pharmacokinetic model. A log-linear exposure effect on Gd lesion count and ARR adequately characterized the relationship between exposure and disease response. In the case of the Gd lesion count model, a bimodal model that distributed subjects into two subpopulations based on low or high baseline Gd lesion count provided a superior goodness of fit. The mean (95% confidence interval) slopes from the exposure-Gd lesion count model and exposure-ARR model are -0.0903 (-0.100, -0.081) and -0.0222 (-0.026, -0.015) (mg/L)-1, respectively. From these slopes, it can be inferred that both Gd lesion count and ARR decrease with increasing exposure to natalizumab in MS subjects. Model-based simulations demonstrated that although reductions in Gd lesion count and ARR were observed with lower doses (75, 150, or 200 mg), only the dose of 300 mg every 4 weeks (q4w) was associated with an ARR ≤0.25 and was considered clinically effective. The results from the exposure-Gd lesion count and exposure-ARR models thus support the appropriateness of the approved natalizumab dose (300 mg q4w) in MS subjects.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/therapeutic use , Multiple Sclerosis/drug therapy , Natalizumab/pharmacokinetics , Natalizumab/therapeutic use , Adolescent , Adult , Aged , Clinical Trials as Topic , Female , Gadolinium/metabolism , Humans , Male , Middle Aged , Multiple Sclerosis/metabolism , Recurrence , Treatment Outcome , Young Adult
8.
Blood ; 123(3): 317-25, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24227821

ABSTRACT

This phase 3 pivotal study evaluated the safety, efficacy, and pharmacokinetics of a recombinant FVIII Fc fusion protein (rFVIIIFc) for prophylaxis, treatment of acute bleeding, and perioperative hemostatic control in 165 previously treated males aged ≥12 years with severe hemophilia A. The study had 3 treatment arms: arm 1, individualized prophylaxis (25-65 IU/kg every 3-5 days, n = 118); arm 2, weekly prophylaxis (65 IU/kg, n = 24); and arm 3, episodic treatment (10-50 IU/kg, n = 23). A subgroup compared recombinant FVIII (rFVIII) and rFVIIIFc pharmacokinetics. End points included annualized bleeding rate (ABR), inhibitor development, and adverse events. The terminal half-life of rFVIIIFc (19.0 hours) was extended 1.5-fold vs rFVIII (12.4 hours; P < .001). Median ABRs observed in arms 1, 2, and 3 were 1.6, 3.6, and 33.6, respectively. In arm 1, the median weekly dose was 77.9 IU/kg; approximately 30% of subjects achieved a 5-day dosing interval (last 3 months on study). Across arms, 87.3% of bleeding episodes resolved with 1 injection. Adverse events were consistent with those expected in this population; no subjects developed inhibitors. rFVIIIFc was well-tolerated, had a prolonged half-life compared with rFVIII, and resulted in low ABRs when dosed prophylactically 1 to 2 times per week.


Subject(s)
Factor VIII/therapeutic use , Hemophilia A/drug therapy , Immunoglobulin Fc Fragments/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Adolescent , Adult , Aged , Child , Drug Administration Schedule , Factor VIII/pharmacokinetics , Hemorrhage/drug therapy , Hemorrhage/prevention & control , Humans , Male , Middle Aged , Recombinant Fusion Proteins/pharmacokinetics , Time Factors , Treatment Outcome , Young Adult
9.
Br J Clin Pharmacol ; 82(5): 1333-1342, 2016 11.
Article in English | MEDLINE | ID: mdl-27333593

ABSTRACT

AIM: Daclizumab high yield process (HYP) is a humanized IgG1 monoclonal antibody that binds to the α-subunit of the interleukin-2 receptor and is being developed for treatment of multiple sclerosis (MS). This manuscript characterized the pharmacokinetic-pharmacodynamic (PK-PD) relationships of daclizumab HYP in subjects with MS. METHODS: Approximately 1400 subjects and 7000 PD measurements for each of three biomarkers [CD25 occupancy, CD56bright natural killer (NK) cell count, regulatory T cell (Treg) count] from four clinical trials were analyzed using non-linear mixed effects modelling. Evaluated regimens included 150 or 300 mg subcutaneous (s.c.) every 4 weeks. RESULTS: CD25 occupancy was characterized using a sigmoidal maximum response (Emax ) model. Upon daclizumab HYP treatment, CD25 saturation was rapid with complete saturation occurring after approximately 7 h and maintained when daclizumab HYP serum concentration was ≥5 mg l-1 . After the last 150 mg s.c. dose, unoccupied CD25 returned to baseline levels in approximately 24 weeks, with daclizumab HYP serum concentration approximately ≤1 mgl-1 1L. CD56bright NK cell expansion was characterized using an indirect response model. Following daclizumab HYP 150 mg s.c. every 4 weeks, expansion plateaus approximately at week 36, at which the average maximum expansion ratio is 5.2. After the last dose, CD56bright NK cells gradually declined to baseline levels within 24 weeks. Treg reduction was characterized by a sigmoidal Emax model. Average maximum reduction of 60% occurred approximately 4 days post 150 mg s.c. dose. After the last dose, Tregs were projected to return to baseline levels in approximately 20 weeks. CONCLUSIONS: Robust PK-PD models of CD25 occupancy, CD56bright NK cell expansion and Treg reduction by daclizumab HYP were developed to characterize its key pharmacodynamic effects in the target patient population.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/pharmacokinetics , CD56 Antigen/drug effects , Interleukin-2 Receptor alpha Subunit/drug effects , Killer Cells, Natural/drug effects , Multiple Sclerosis/blood , T-Lymphocytes, Regulatory/drug effects , Antibodies, Monoclonal, Humanized/blood , Clinical Trials as Topic , Daclizumab , Humans , Killer Cells, Natural/cytology , Lymphocyte Count , Nonlinear Dynamics , T-Lymphocytes, Regulatory/cytology
10.
Br J Clin Pharmacol ; 82(2): 380-8, 2016 08.
Article in English | MEDLINE | ID: mdl-27060836

ABSTRACT

AIM: Subcutaneous (s.c.) peginterferon beta-1a injected once every 2 weeks and s.c. interferon beta-1a injected three times per week (Rebif®) have demonstrated efficacy in relapsing-remitting multiple sclerosis, but direct comparisons of pharmacological activity and tolerability between the two products are lacking. COMPARE was an open label, crossover, pharmacokinetic (PK) study evaluating drug exposure and the safety and tolerability of s.c. peginterferon beta-1a and s.c. interferon beta-1a, over 2 weeks in healthy subjects. METHODS: Thirty healthy subjects received one dose of peginterferon beta-1a (125 µg s.c.) or six doses of interferon beta-1a (44 µg s.c.) over 2 weeks, followed by the alternate treatment after a 2 week washout period. Drug concentrations were measured using an enzyme-linked immunosorbent assay (ELISA) and PK parameters including cumulative area under the concentration-time curve (AUC0-336h ) over 2 weeks and maximum observed serum concentrations (Cmax ) were estimated using a non-compartmental analysis. RESULTS: The PK analysis population comprised 26 subjects for each treatment. Drug exposure (AUC0-336h ) was 60% higher with s.c. peginterferon than with s.c. interferon beta-1a (117.4 ng ml(-1) h, 95% confidence interval 95.6, 144.3 vs. 73.1 ng ml(-1) h, 95% confidence interval 61.2, 87.3, respectively; P < 0.0001). Injection-site reactions (ISRs) were the most common adverse events (AEs) observed with both treatments. Numerically lower frequencies and incidence rates of ISRs, headache, myalgia and chills were observed with s.c. peginterferon beta-1a. CONCLUSIONS: One dose of s.c. peginterferon delivered significantly greater drug exposure than s.c. interferon beta-1a three times a week over 2 weeks, and a lower frequency of AEs.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Interferon beta-1a/administration & dosage , Interferon-beta/administration & dosage , Polyethylene Glycols/administration & dosage , Adjuvants, Immunologic/adverse effects , Adjuvants, Immunologic/pharmacokinetics , Adolescent , Adult , Area Under Curve , Cross-Over Studies , Enzyme-Linked Immunosorbent Assay , Female , Humans , Injections, Subcutaneous , Interferon beta-1a/adverse effects , Interferon beta-1a/pharmacokinetics , Interferon-beta/adverse effects , Interferon-beta/pharmacokinetics , Male , Middle Aged , Polyethylene Glycols/adverse effects , Polyethylene Glycols/pharmacokinetics , Young Adult
11.
J Pharmacokinet Pharmacodyn ; 43(4): 371-83, 2016 08.
Article in English | MEDLINE | ID: mdl-27299457

ABSTRACT

The effect of subcutaneous (SC) peginterferon ß-1a exposure on reduction of gadolinium-enhanced (Gd+) lesion count over time was evaluated in patients with relapsing-remitting multiple sclerosis (RRMS) in a Phase 3 study (ADVANCE). Patients were randomized to receive SC injections of placebo (n = 500), 125 mcg every-2-weeks (n = 512), or 125 mcg every-4-weeks (n = 500) for 1 year, and then active treatment in the second year. Steady state 4-week AUC (AUCss) was derived for each individual based on sparse pharmacokinetic (PK) sample and a population PK model. Several longitudinal count models, including marginal, mixed effect, and mixture models, were compared to explore the relationship between AUCss and Gd+ lesion count (or T2 lesion count). A mixture model which divided subjects into two subpopulations by low and high baseline lesion activity was found to yield best goodness-of-fit for the data. In this model, the point estimate and 95 % CI for drug effect slope on log(λ) are -0.0256 (-0.0304, -0.0216) for Gd+ lesion and -0.0147 (-0.0170, -0.0124) for T2 lesion. This suggested that reduction of Gd+ lesion (or T2 lesion) count over time is significantly related to SC peginterferon ß-1a exposure, and that the increased reduction lesion count with the every-2-week regimen versus the every-4-week regimen was driven by the higher exposure achieved in that treatment arm (mean Gd+ lesion count 0.2 and 0.7 at Year 2, respectively). The every-2-week regimen produced an exposure range that was close to the plateau range of the exposure-response curve, supporting its selection as the regulatory approved dosage.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Interferon-beta/therapeutic use , Models, Biological , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/pathology , Polyethylene Glycols/therapeutic use , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacokinetics , Area Under Curve , Contrast Media/administration & dosage , Drug Administration Schedule , Gadolinium/administration & dosage , Half-Life , Humans , Image Enhancement , Injections, Subcutaneous , Interferon-beta/administration & dosage , Interferon-beta/pharmacokinetics , Magnetic Resonance Imaging , Multiple Sclerosis, Relapsing-Remitting/diagnostic imaging , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/pharmacokinetics , Recurrence , Treatment Outcome
12.
Br J Clin Pharmacol ; 79(3): 514-22, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25265472

ABSTRACT

AIMS: To evaluate the pharmacokinetics (PK) and pharmacodynamics (PD) of subcutaneous peginterferon beta-1a in patients with relapsing-remitting multiple sclerosis (RRMS) in the phase 3 ADVANCE study (n = 1512). METHODS: During year 1, patients were randomized (1:1:1) to placebo or peginterferon beta-1a 125 µg every 2 or 4 weeks. After year 1, patients randomized to placebo were re-randomized to 125 µg peginterferon beta-1a administered every 2 weeks or every 4 weeks for year 2. Patients randomized to peginterferon beta-1a in year 1 remained on the same dosing regimen in year 2. Intensive blood samples for PK and PD (neopterin elevation; a biomarker of pharmacological activity induced by interferon beta-1a) measurements were collected from 44 patients pre-dosing and at intervals over 240 h post-dosing at weeks 4 and 24. Sparse samples were collected from all patients after each dosing at weeks 4, 12, 24, 56 and 84. RESULTS: The PK profile of peginterferon beta-1a did not change over time or between dosing regimens. No accumulation was observed. Peak serum concentrations were reached 1-1.5 days post-dosing, with a mono-phasic decline and a median half-life of approximately 2-3 days. Dosing every 2 weeks provided approximately two-fold greater monthly cumulative area under the curve than every 4 weeks. Neopterin elevation was sustained for 10-14 days following each dose, indicating doubled cumulative duration of pharmacological activity for dosing every 2 weeks vs. every 4 weeks. CONCLUSIONS: These PK/PD profiles potentially explain the enhanced efficacy of dosing every 2 weeks in patients with RRMS.


Subject(s)
Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/pharmacokinetics , Interferon-beta/pharmacology , Interferon-beta/pharmacokinetics , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Polyethylene Glycols/pharmacology , Polyethylene Glycols/pharmacokinetics , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/therapeutic use , Adolescent , Adult , Aged , Antibodies, Neutralizing/blood , Area Under Curve , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Female , Humans , Injections, Subcutaneous , Interferon-beta/administration & dosage , Interferon-beta/therapeutic use , Male , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/blood , Multiple Sclerosis, Relapsing-Remitting/immunology , Neopterin/blood , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/therapeutic use , Treatment Outcome , Young Adult
13.
CPT Pharmacometrics Syst Pharmacol ; 12(2): 196-206, 2023 02.
Article in English | MEDLINE | ID: mdl-36471456

ABSTRACT

Phosphorylated neurofilament heavy subunit (pNfH) has been recently identified as a promising biomarker of disease onset and treatment efficacy in spinal muscular atrophy (SMA). This study introduces a quantitative systems pharmacology model representing the SMA pediatric scenario in the age range of 0-20 years with and without treatment with the antisense oligonucleotide nusinersen. Physiological changes typical of the pediatric age and the contribution of SMA and its treatment to the peripheral pNfH levels were included in the model by extending the equations of a previously developed mathematical model describing the neurofilament trafficking in healthy adults. All model parameters were estimated by fitting data from clinical trials that enrolled SMA patients treated with nusinersen. The data from the control group of the study was employed to build an in silico population of untreated subjects, and the parameters related to the treatment were estimated by fitting individual pNfH time series of SMA patients followed during the treatment. The final model reproduces well the pNfH levels in the presence of SMA in both the treated and untreated conditions. The results were validated by comparing model predictions with the data obtained from an additional cohort of SMA patients. The reported good predictive model performance makes it a valuable tool for investigating pNfH as a biomarker of disease progression and treatment response in SMA and for the in silico evaluation of novel treatment protocols.


Subject(s)
Muscular Atrophy, Spinal , Oligonucleotides, Antisense , Adult , Humans , Child , Infant, Newborn , Infant , Child, Preschool , Adolescent , Young Adult , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , Intermediate Filaments , Network Pharmacology , Muscular Atrophy, Spinal/drug therapy , Biomarkers
14.
J Clin Pharmacol ; 62(8): 1030-1046, 2022 08.
Article in English | MEDLINE | ID: mdl-35285968

ABSTRACT

Amyloid-related imaging abnormalities with edema (ARIA-E) have been reported in patients with early Alzheimer disease treated with aducanumab. ARIA-E incidence has been observed to be dependent on both dose and apolipoprotein E4 carrier status. A time-to-event (TTE) approach applying data from 2 phase 3 studies (studies 301 and 302) was used to describe the effect of aducanumab serum exposure on the instantaneous risk of 2 end points: the first incidence of ARIA-E and time to ARIA-E resolution. A total of 3251 subjects with 826 events supported the TTE model to characterize the first ARIA-E event. The TTE resolution model was supported by data from 768 of 826 subjects who had ARIA-E resolved. Relationships between drug concentrations and ARIA-E events were modeled with a hazard function dependent on time, aducanumab serum concentrations, attenuation of aducanumab exposure effects with time (ie, potential for tolerance to aducanumab exposure), study, and apolipoprotein E4 carrier status. The TTE model showed that ARIA-E incidence rates were higher during the first 200 days, followed by a reduction in rates. The change in event rate reflects the attenuation of drug effect, thereby providing support for the current proposed titration regimen. Time to ARIA-E resolution was characterized by a constant baseline hazard with a probability to resolution affected by baseline ARIA-E severity and aducanumab concentration. ARIA-E resolution was found to be driven primarily by baseline hazard and time and suggested that aducanumab concentration effect is a minor contributor to the time to resolution of ARIA-E.


Subject(s)
Alzheimer Disease , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Antibodies, Monoclonal, Humanized , Apolipoprotein E4/pharmacology , Apolipoprotein E4/therapeutic use , Brain/metabolism , Humans , Magnetic Resonance Imaging
15.
CPT Pharmacometrics Syst Pharmacol ; 11(3): 362-372, 2022 03.
Article in English | MEDLINE | ID: mdl-35029320

ABSTRACT

Alzheimer's disease (AD) is an irreversible, progressive brain disorder that impairs memory and cognitive function. Dysregulation of the amyloid-ß (Aß) pathway and amyloid plaque accumulation in the brain are hallmarks of AD. Aducanumab is a human, immunoglobulin gamma 1 monoclonal antibody targeting aggregated forms of Aß. In phase Ib and phase III studies, aducanumab reduced Aß plaques in a dose dependent manner, as measured by standard uptake value ratio of amyloid positron emission tomography imaging. The goal of this work was to develop a quantitative systems pharmacology model describing the production, aggregation, clearance, and transport of Aß as well as the mechanism of action for the drug to understand the relationship between aducanumab dosing regimens and changes of different Aß species, particularly plaques in the brain. The model was used to better understand the pharmacodynamic effects observed in the clinical trials of aducanumab and assist in the clinical development of future Aß therapies.


Subject(s)
Alzheimer Disease , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Antibodies, Monoclonal, Humanized , Brain/metabolism , Humans , Network Pharmacology , Plaque, Amyloid/drug therapy , Plaque, Amyloid/metabolism
16.
CPT Pharmacometrics Syst Pharmacol ; 11(1): 7-19, 2022 01.
Article in English | MEDLINE | ID: mdl-34697913

ABSTRACT

Aducanumab is a human immunoglobulin G1 anti-amyloid beta (Aß) antibody currently being evaluated for potential treatment of patients with early Alzheimer's disease. This paper describes the relationship between the population pharmacokinetics (PopPKs) and pharmacokinetics-pharmacodynamics (PKs-PDs) of aducanumab using data from phase I to III clinical studies, with standard uptake value ratio (SUVR) used as a PD marker. Across clinical studies, aducanumab was administered intravenously either as a single dose ranging from 0.3 to 60 mg/kg or as multiple doses of 1, 3, 6, or 10 mg/kg every 4 weeks. A titration regimen with maintenance doses of 3, 6, or 10 mg/kg was also evaluated. Aducanumab PK was characterized with a two-compartment model with first-order elimination. No nonlinearities in PKs were observed. The PopPK-PD model was developed using a sequential estimation approach. The time course of amyloid plaques, as expressed by composite SUVR measured using positron emission tomography, was described using an indirect response model with drug effect stimulating the elimination of SUVR. None of the identified covariates on PK and the PopPK-PD model were clinically relevant. The PopPK-PD model showed that magnitude, duration, and consistency of dosing are important factors determining the degree of Aß removal. The intrinsic pharmacology of aducanumab remained consistent across studies.


Subject(s)
Alzheimer Disease/drug therapy , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/therapeutic use , Plaque, Amyloid/drug therapy , Aged , Dose-Response Relationship, Drug , Female , Humans , Male , Models, Biological , Positron-Emission Tomography
17.
CPT Pharmacometrics Syst Pharmacol ; 11(4): 447-457, 2022 04.
Article in English | MEDLINE | ID: mdl-35146969

ABSTRACT

Neurofilaments (Nfs) are the major structural component of neurons. Their role as a potential biomarker of several neurodegenerative diseases has been investigated in past years with promising results. However, even under physiological conditions, little is known about the leaking of Nfs from the neuronal system and their detection in the cerebrospinal fluid (CSF) and blood. This study aimed at developing a mathematical model of Nf transport in healthy subjects in the 20-90 age range. The model was implemented as a set of ordinary differential equations describing the trafficking of Nfs from the nervous system to the periphery. Model parameters were calibrated on typical Nf levels obtained from the literature. An age-dependent function modeled on CSF data was also included and validated on data measured in serum. We computed a global sensitivity analysis of model rates and volumes to identify the most sensitive parameters affecting the model's steady state. Age, Nf synthesis, and degradation rates proved to be relevant for all model variables. Nf levels in the CSF and in blood were observed to be sensitive to the Nf leakage rates from neurons and to the blood clearance rate, and CSF levels were also sensitive to rates representing CSF turnover. An additional parameter perturbation analysis was also performed to investigate possible transient effects on the model variables not captured by the sensitivity analysis. The model provides useful insights into Nf transport and constitutes the basis for implementing quantitative system pharmacology extensions to investigate Nf trafficking in neurodegenerative diseases.


Subject(s)
Intermediate Filaments , Neurodegenerative Diseases , Biomarkers , Humans , Models, Theoretical , Neurofilament Proteins/cerebrospinal fluid
18.
J Pharmacol Exp Ther ; 338(3): 984-96, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21690216

ABSTRACT

Human interferon (IFN) ß has well established beneficial effects in treating relapsing forms of multiple sclerosis, but current first-line treatment requires frequent (from daily to weekly) parenteral administration. A 20-kDa polyethylene glycol (PEG)-conjugated IFN ß-1a (PEG-IFN ß-1a) is being developed to decrease the frequency of administration and improve patient convenience and compliance. We present pharmacokinetic (PK) and pharmacodynamic (PD) parameters, immunogenicity, and safety of PEG-IFN ß-1a in Rhesus monkeys in support of a phase 1 clinical trial. Two single-dose PK/PD studies and one 5-week repeat-dose toxicity study compliant with good laboratory practice were conducted. The PK of IFN ß-1a and PEG-IFN ß-1a were modeled with a two-compartment model, and the link between drug concentration and neopterin response (PD marker) was described with an indirect stimulatory model. PEG-IFN ß-1a showed greater exposure, longer half-life, lower clearance, and reduced volume of distribution than unmodified IFN ß-1a. Consistent with the pharmacology of type I IFNs, PEG-IFN ß-1a resulted in the elevation of neopterin concentration, a transient body temperature increase, and a reversible lymphocyte count decrease. As expected, neutralizing antibodies to PEG-IFN ß-1a formed in almost all monkeys after 5 weeks of treatment, which resulted in significantly reduced drug exposure and abrogation of neopterin induction. There were no drug-related adverse effects at doses up to 100 µg/kg (11 MIU/kg) given subcutaneously or intramuscularly once weekly for 5 weeks. The no-observed-adverse-effect level was determined to be 100 µg/kg (11 MIU/kg), the highest dose tested.


Subject(s)
Antineoplastic Agents/pharmacology , Interferons/pharmacology , Interferons/toxicity , Polyethylene Glycols/pharmacology , Polyethylene Glycols/toxicity , Animals , Antineoplastic Agents/chemistry , Area Under Curve , Body Temperature/drug effects , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Feedback, Physiological , Half-Life , Humans , Injections, Intramuscular , Injections, Subcutaneous , Interferon-beta , Interferons/immunology , Lymphocyte Count , Macaca mulatta , Models, Statistical , Neopterin/blood , No-Observed-Adverse-Effect Level
19.
Eur J Pharm Biopharm ; 158: 222-232, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33279603

ABSTRACT

To reduce cost and time for product development, an ideal strategy for the development of oral extended release (ER) product is to identify the desired formulation with minimum needsfor clinical evaluation. The aim of this work was to demonstrate the feasibility of adopting a "prediction-then-validation" strategy for the development of oral ER formulations. Instead of the traditional approach using multiple ER formulations for IVIVC development, an enteric-coated fast release formulation was successfully utilized for the development of a biopredictive tool to estimate the drug release from enteric coated polymeric ER formulations in the intestine. A TS1 (time scale factor between Tvitro and Tvivo equals to 1) system was designed and developed, based on which the in vivo pharmacokinetic (PK) performance of ER formulations in dog and in human were well predicted prior to in vivo evaluations. The model further passed a posteriori validation using the criteria for level A IVIVC and, as designed, provided a Tscale value of 1 for the IVIVC model.


Subject(s)
Delayed-Action Preparations/pharmacokinetics , Drug Carriers/chemistry , Models, Biological , Administration, Oral , Adult , Animals , Area Under Curve , Biological Availability , Cellulose/analogs & derivatives , Cellulose/chemistry , Cross-Over Studies , Delayed-Action Preparations/administration & dosage , Dogs , Drug Evaluation, Preclinical/methods , Drug Liberation , Feasibility Studies , Female , Healthy Volunteers , Humans , Intestinal Absorption , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Male , Middle Aged , Solubility , Tablets , Young Adult
20.
Neuromuscul Disord ; 31(4): 310-318, 2021 04.
Article in English | MEDLINE | ID: mdl-33781694

ABSTRACT

Nusinersen is an antisense oligonucleotide approved for the treatment of spinal muscular atrophy. The drug is given intrathecally at 12 mg, beginning with 3 loading doses at 2-week intervals, a fourth loading dose 30 days thereafter, and maintenance doses at 4-month intervals. This population pharmacokinetic model was developed to clarify how to maintain targeted nusinersen exposure after an unforeseen one-time delay or missed dose. Simulations demonstrated that the impact of a one-time delay in dosing or a missed dose on median cerebrospinal fluid exposures depended on duration of interruption and the regimen phase in which it occurred. Delays in loading doses delayed reaching the peak trough concentration by approximately the duration of the interruption. Resumption of the regimen as soon as possible resulted in achieving steady state trough concentration upon completion of the loading phase. A short delay (30-90 days) during the maintenance phase led to prolonged lower median cerebrospinal fluid concentration if all subsequent doses were shifted by the same 4-month interval. However, administration of the delayed dose, followed by the subsequent dose as originally scheduled, rapidly restored trough concentration. If a dose must be delayed, patients should return to the original dosing schedule as soon as possible.


Subject(s)
Muscular Atrophy, Spinal/drug therapy , Oligonucleotides/pharmacokinetics , Humans , Oligonucleotides/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL