Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Comput Biol ; 19(10): e1011507, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37792732

ABSTRACT

Mathematical modeling of unperturbed and perturbed tumor growth dynamics (TGD) in preclinical experiments provides an opportunity to establish translational frameworks. The most commonly used unperturbed tumor growth models (i.e. linear, exponential, Gompertz and Simeoni) describe a monotonic increase and although they capture the mean trend of the data reasonably well, systematic model misspecifications can be identified. This represents an opportunity to investigate possible underlying mechanisms controlling tumor growth dynamics through a mathematical framework. The overall goal of this work is to develop a data-driven semi-mechanistic model describing non-monotonic tumor growth in untreated mice. For this purpose, longitudinal tumor volume profiles from different tumor types and cell lines were pooled together and analyzed using the population approach. After characterizing the oscillatory patterns (oscillator half-periods between 8-11 days) and confirming that they were systematically observed across the different preclinical experiments available (p<10-9), a tumor growth model was built including the interplay between resources (i.e. oxygen or nutrients), angiogenesis and cancer cells. The new structure, in addition to improving the model diagnostic compared to the previously used tumor growth models (i.e. AIC reduction of 71.48 and absence of autocorrelation in the residuals (p>0.05)), allows the evaluation of the different oncologic treatments in a mechanistic way. Drug effects can potentially, be included in relevant processes taking place during tumor growth. In brief, the new model, in addition to describing non-monotonic tumor growth and the interaction between biological factors of the tumor microenvironment, can be used to explore different drug scenarios in monotherapy or combination during preclinical drug development.


Subject(s)
Models, Biological , Neoplasms , Animals , Mice , Tumor Microenvironment , Models, Theoretical , Cell Proliferation , Cell Line, Tumor
2.
Br J Clin Pharmacol ; 89(3): 1115-1126, 2023 03.
Article in English | MEDLINE | ID: mdl-36222177

ABSTRACT

AIMS: Pharmacokinetics of tacrolimus after sublingual administration is not characterized in paediatric liver transplant patients. Therefore, we aimed to develop a population pharmacokinetic model of sublingually administered tacrolimus in patients who cannot swallow the capsules due to their age, sedation status and/or mechanical ventilation during the first weeks post-transplantation. METHODS: Demographic, clinical and pharmacological variables, including tacrolimus whole blood concentrations obtained from therapeutic drug monitoring and data from dense-sampling pharmacokinetic profiles, were recorded in 26 paediatric patients with biliary atresia who underwent liver transplantation between 2016 and 2021. Population pharmacokinetic analysis was performed with NONMEM v7.4. RESULTS: Disposition of tacrolimus was best characterized by a 2-compartment model with clearance achieving half of the maximum elimination capacity (CLMAX  = 4.1 L/h) at 4.6 days post-transplantation (T50 ). Compared to sedated patients, nonsedated status showed an increased first-order absorption rate constant (1.1 vs. 0.1 h-1 ) and a 24% reduction in bioavailability (FNS ) at 14 days post-transplant. The model was able to explain the oral absorption pattern in nonsedated patients as the result of gut bioavailability (0.9) and hepatic extraction ratio, with the latter being responsible for first-pass effects. Estimates of interindividual variability remained moderate (25.9% for the gut bioavailability) to high (79.8% for the apparent volume of distribution of the central compartment, and 101% for T50 ). CONCLUSION: A population pharmacokinetic model of sublingually administered tacrolimus in paediatric patients was developed to characterize different absorption mechanisms. Once the model is externally validated, the effect of post-transplant time on clearance and the sedation status may be considered in routine dosing management.


Subject(s)
Liver Transplantation , Tacrolimus , Humans , Child , Infant , Child, Preschool , Tacrolimus/pharmacokinetics , Immunosuppressive Agents/pharmacokinetics , Models, Biological , Biological Availability
3.
Br J Clin Pharmacol ; 88(1): 166-177, 2022 01.
Article in English | MEDLINE | ID: mdl-34087010

ABSTRACT

AIMS: The aims of this work were to build a semi-mechanistic tumour growth inhibition (TGI) model for metastatic colorectal cancer (mCRC) patients receiving either cetuximab + chemotherapy or chemotherapy alone and to identify early predictors of overall survival (OS). METHODS: A total of 1716 patients from 4 mCRC clinical studies were included in the analysis. The TGI model was built with 8973 tumour size measurements where the probability of drop-out was also included and modelled as a time-to-event variable using parametric survival models, as it was the case in the OS analysis. The effects of patient- and tumour-related covariates on model parameters were explored. RESULTS: Chemotherapy and cetuximab effects were included in an additive form in the TGI model. Development of resistance was found to be faster for chemotherapy (drug effect halved at wk 8) compared to cetuximab (drug effect halved at wk 12). KRAS wild-type status and presenting a right-sided primary lesion were related to a 3.5-fold increase in cetuximab drug effect and a 4.7× larger cetuximab resistance, respectively. The early appearance of a new lesion (HR = 4.14), a large tumour size at baseline (HR = 1.62) and tumour heterogeneity (HR = 1.36) were the main predictors of OS. CONCLUSIONS: Semi-mechanistic TGI and OS models have been developed in a large population of mCRC patients receiving chemotherapy in combination or not with cetuximab. Tumour-related predictors, including a machine learning derived-index of tumour heterogeneity, were linked to changes in drug effect, resistance to treatment or OS, contributing to the understanding of the variability in clinical response.


Subject(s)
Colorectal Neoplasms , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cetuximab/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease-Free Survival , Humans , Mutation , Survival Analysis
4.
J Pharmacol Exp Ther ; 372(3): 299-307, 2020 03.
Article in English | MEDLINE | ID: mdl-31822515

ABSTRACT

Crohn's disease (CD) is a complex inflammatory bowel disease whose pathogenesis appears to involve several immunologic defects causing functional impairment of the gut. Its complexity and the reported loss of effectiveness over time of standard of care together with the increase in its worldwide incidence require the application of techniques aiming to find new therapeutic strategies. Currently, systems pharmacology modeling has been gaining importance as it integrates the available knowledge of the system into a single computational model. In this work, the following workflow for robust application of systems pharmacology modeling was followed: 1) scope definition; 2) species selection and circulating plasma levels based on a search in the literature; 3) representation of model topology and parametrization of the interactions, after literature data extraction and curation, and the implementation of ordinary differential equations in SimBiology (MATLAB version R2018b); and 4) model curation and evaluation by visual comparison of simulated interleukin (IL) concentrations with the reported levels in plasma, and sensitivity analysis performed to confirm model robustness and identify the most influential parameters. Finally, 5) exposure to two dose levels of recombinant human IL10 was evaluated by simulation and comparison with reported clinical study results. In summary, we present a quantitative systems pharmacology model for the main ILs involved in CD developed using a standardized methodology and supported by a comprehensive repository summarizing the most relevant literature in the field. However, it has to be taken into account that external validation is still pending as available clinical data were primarily used for model training. SIGNIFICANCE STATEMENT: Crohn's disease (CD) is a complex heterogeneous inflammatory bowel disorder. Systems pharmacology modeling offers a great opportunity for integration of the available knowledge on the disease using a computational framework. As a result of this work, a comprehensive repository along with a quantitative systems pharmacology model for the main interleukins involved in CD is provided. This model is useful for the in silico evaluation of biomarkers and potential therapeutic targets and can be adapted to address research gaps regarding CD.


Subject(s)
Crohn Disease/immunology , Interleukins/blood , Models, Biological , Biomarkers/blood , Computer Simulation , Crohn Disease/blood , Humans
5.
Mol Genet Metab ; 128(3): 367-375, 2019 11.
Article in English | MEDLINE | ID: mdl-30639045

ABSTRACT

INTRODUCTION: Acute intermittent porphyria (AIP) is characterized by hepatic over-production of the heme precursors when aminolevulinic acid (ALA)-synthase 1 is induced by endogenous or environmental factors. The aim of this study was to develop a semi-mechanistic computational model to characterize urine accumulation of heme precursors during acute attacks based on experimental pharmacodynamics data and support the development of new therapeutic strategies. METHODS: Male AIP mice received recurrent phenobarbital challenge starting on days 1, 9, 16 and 30. 24-h urine excretion of ALA, porphobilinogen (PBG) and porphyrins from challenges D1, D9 and D30 constituted the training data set to build the mechanistic model using the population approach. In a second study, porphyrin and porphyrin precursor excretion from challenge D16 were used as a validation data set. RESULTS: The computational model presented the following features: (i) urinary excretion of ALA, PBG and porphyrins was governed by unmeasured circulating heme precursor amounts, (ii) the circulating amounts of ALA and PBG were the precursors of circulating amounts of PBG and porphyrins, respectively, and (iii) the phenobarbital effect linearly increased the synthesis of circulating ALA and PBG levels. The model displayed good parameter precision (coefficient of variation below 32% in all parameters), and adequately described the experimental data. Finally, a theoretical hemin effect was implemented to illustrate the applicability of the model to dosage optimization in drug therapies. CONCLUSIONS: A semi-mechanistic disease model was successfully developed to describe the temporal evolution of urinary heme precursor excretion during recurrent biochemical-induced acute attacks in AIP mice. This model represents the first computational approach to explore and optimize current and new therapies.


Subject(s)
Computer Simulation , Disease Models, Animal , Phenobarbital/administration & dosage , Porphyria, Acute Intermittent/chemically induced , Aminolevulinic Acid/urine , Animals , Male , Mice , Mice, Inbred C57BL , Porphobilinogen/urine , Porphyria, Acute Intermittent/urine , Porphyrins/urine
6.
Clin Endocrinol (Oxf) ; 91(1): 33-40, 2019 07.
Article in English | MEDLINE | ID: mdl-30868607

ABSTRACT

CONTEXT: Optimization of hydrocortisone replacement therapy is important to prevent under- and over dosing. Hydrocortisone pharmacokinetics is complex as circulating cortisol is protein bound mainly to corticosteroid-binding globulin (CBG) that has a circadian rhythm. OBJECTIVE: A detailed analysis of the CBG circadian rhythm and its impact on cortisol exposure after hydrocortisone administration. DESIGN AND METHODS: CBG was measured over 24 hours in 14 healthy individuals and, employing a modelling and simulation approach using a semi-mechanistic hydrocortisone pharmacokinetic model, we evaluated the impact on cortisol exposure (area under concentration-time curve and maximum concentration of total cortisol) of hydrocortisone administration at different clock times and of the changing CBG concentrations. RESULTS: The circadian rhythm of CBG was well described with two cosine terms added to the baseline of CBG: baseline CBG was 21.8 µg/mL and interindividual variability 11.9%; the amplitude for the 24 and 12 hours cosine functions were relatively small (24 hours: 5.53%, 12 hours: 2.87%) and highest and lowest CBG were measured at 18:00 and 02:00, respectively. In simulations, the lowest cortisol exposure was observed after administration of hydrocortisone at 23:00-02:00, whereas the highest was observed at 15:00-18:00. The differences between the highest and lowest exposure were minor (≤12.2%), also regarding the free cortisol concentration and free fraction (≤11.7%). CONCLUSIONS: Corticosteroid-binding globulin has a circadian rhythm but the difference in cortisol exposure is ≤12.2% between times of highest and lowest CBG concentrations; therefore, hydrocortisone dose adjustment based on time of dosing to adjust for the CBG concentrations is unlikely to be of clinical benefit.


Subject(s)
Circadian Rhythm/drug effects , Hydrocortisone/pharmacology , Hydrocortisone/pharmacokinetics , Transcortin/metabolism , Adolescent , Adult , Circadian Rhythm/physiology , Female , Healthy Volunteers , Humans , Hydrocortisone/blood , Male , Middle Aged , Young Adult
7.
J Pharmacol Exp Ther ; 366(1): 96-104, 2018 07.
Article in English | MEDLINE | ID: mdl-29691287

ABSTRACT

Xenograft mice are largely used to evaluate the efficacy of oncological drugs during preclinical phases of drug discovery and development. Mathematical models provide a useful tool to quantitatively characterize tumor growth dynamics and also optimize upcoming experiments. To the best of our knowledge, this is the first report where unperturbed growth of a large set of tumor cell lines (n = 28) has been systematically analyzed using a previously proposed model of nonlinear mixed effects (NLME). Exponential growth was identified as the governing mechanism in the majority of the cell lines, with constant rate values ranging from 0.0204 to 0.203 day-1 No common patterns could be observed across tumor types, highlighting the importance of combining information from different cell lines when evaluating drug activity. Overall, typical model parameters were precisely estimated using designs in which tumor size measurements were taken every 2 days. Moreover, reducing the number of measurements to twice per week, or even once per week for cell lines with low growth rates, showed little impact on parameter precision. However, a sample size of at least 50 mice is needed to accurately characterize parameter variability (i.e., relative S.E. values below 50%). This work illustrates the feasibility of systematically applying NLME models to characterize tumor growth in drug discovery and development, and constitutes a valuable source of data to optimize experimental designs by providing an a priori sampling window and minimizing the number of samples required.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Xenograft Model Antitumor Assays , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Mice , Models, Statistical
8.
Pharm Res ; 32(4): 1493-504, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25355461

ABSTRACT

PURPOSE: Irosustat is the 'first-in-class' irreversible potent steroid sulphatase inhibitor with lack of oestrogenic activity. The objective of this work was to develop a population model characterizing simultaneously the pharmacokinetic profiles of irosustat in plasma and whole blood. METHODS: This clinical study was an open label, multicentre, phase I multiple cohort dose escalation trial conducted in 35 postmenopausal women with oestrogen-receptor positive breast cancer. Patients received 1, 5, 20, 40, or 80 mg oral doses. Irosustat was administered as a single oral dose to each patient followed by an observation period of 7 days. On day 8 each patient received once daily oral administration until day 34. Concentrations of irosustat in both blood and plasma were obtained and pharmacokinetic analyses were performed with NONMEM 7.2. RESULTS AND CONCLUSIONS: Irosustat showed non-linear disposition characteristics modelled as maximum binding capacity into the red blood cells. Plasma concentration corresponding to half of the maximum capacity was 32.79 ng/mL. The value of the blood to plasma concentration ratio in linear conditions was 419, indicating very high affinity for the red blood cells. Apparent plasma and blood clearances were estimated in 1199.52 and 3.90 L/day, respectively. Pharmacokinetics of irosustat showed low-moderate inter-subject variability, and neither the demographics (e.g., age, or weight) nor the phenotypes for CYP2C9, CYP2C19, and CYP3A5 enzymes showed statistically significant effects. Relative bioavailability was decreased as the administered dose was augmented. The model predicted a 47% decrease in relative bioavailability in the 40 mg with respect to the 1 mg dose.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacokinetics , Breast Neoplasms/metabolism , Erythrocytes/metabolism , Models, Biological , Postmenopause , Receptors, Estrogen/metabolism , Sulfonic Acids/pharmacokinetics , Administration, Oral , Aged , Aged, 80 and over , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Agents, Hormonal/blood , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/blood , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Female , Humans , Middle Aged , Nonlinear Dynamics , Sulfonic Acids/administration & dosage , Sulfonic Acids/blood , Sulfonic Acids/therapeutic use
9.
J Immunol ; 188(8): 3988-92, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22422884

ABSTRACT

IFN-α is widely used for the treatment of chronic viral hepatitis and malignancies. However, systemic IFN-α treatment causes severe neuropsychiatric complications in humans, including depression, anxiety, and cognitive impairments. We have previously reported that the fusion protein formed by IFN-α and apolipoprotein A-I (IA) circulates bound to high-density lipoproteins (HDLs) and exhibits liver targeting, increased half-life, enhanced immunostimulatory activity, and reduced cytotoxicity. As the transport of HDLs across the blood-brain barrier is a highly complex and regulated process, in this study, we examine the effects of IA on the brain. Determination of IFN-α in brain and serum after hydrodynamic administration of different doses of a plasmid encoding IFN-α or IA showed that IA penetrated into the brain by a saturable transport mechanism. Thus, at high serum levels of the transgenes, the induction of IFN-sensitive genes and the number of phospho-STAT1(+) cell nuclei in the brain were substantially higher with IFN-α than with IA. This was associated with attenuation of neurodepression in mice given IA, as manifested by shorter immobility time in the tail suspension test. However, when given low doses of rIFN-α or the same antiviral units of HDLs containing IA, the induction of IFN-stimulated genes in the brain was significantly greater with the latter. In conclusion, IA crosses the blood-brain barrier not by diffusion, as is the case of IFN-α, but by a facilitated saturable transport mechanism. Thus, linkage to apolipoprotein A-I may serve to modulate the effects of IFN-α on the CNS.


Subject(s)
Apolipoprotein A-I/metabolism , Blood-Brain Barrier/metabolism , Interferon-alpha/metabolism , Recombinant Fusion Proteins/pharmacokinetics , Animals , Apolipoprotein A-I/genetics , Depression/drug therapy , Depression/etiology , Female , Half-Life , Hindlimb Suspension , Humans , Injections, Intravenous , Interferon-alpha/genetics , Lipoproteins, HDL/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Transport , Recombinant Fusion Proteins/genetics
10.
Clin Pharmacol Ther ; 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39037559

ABSTRACT

Immuno-oncology (IO) is a growing strategy in cancer treatment. Oncolytic viruses (OVs) can selectively infect cancer cells and lead to direct and/or immune-dependent tumor lysis. This approach represents an opportunity to potentiate the efficacy of immune checkpoint inhibitors (ICI), such as pembrolizumab. Currently, there is a lack of comprehensive quantitative models for the aforementioned scenarios. In this work, we developed a mechanistic framework describing viral kinetics, viral dynamics, and tumor response after intratumoral (i.t.) or intravenous (i.v.) administration of V937 alone or in combination with pembrolizumab. The model accounts for tumor shrinkage, in both injected and non-injected lesions, induced by: viral-infected tumor cell death and activated CD8 cells. OV-infected tumor cells enhanced the expansion of CD8 cells, whereas pembrolizumab inhibits their exhaustion by competing with PD-L1 in their binding to PD-1. Circulating viral levels and treatment effects on tumor volume were adequately characterized in all the different scenarios. This mechanistic-based model has been developed by combining top-down and bottom-up approaches and provides individual estimates of viral and ICI responses. The robustness of the model is reflected by the description of the tumor size time profiles in a variety of clinical scenarios. Additionally, this platform allows us to investigate not only the contribution of processes related to the viral kinetics and dynamics on tumor response, but also the influence of its interaction with an ICI. Additionally, the model can be used to explore different scenarios aiming to optimize treatment combinations and support clinical development.

11.
J Pharmacol Exp Ther ; 346(3): 432-42, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23845890

ABSTRACT

The aims of this work were as follows: 1) to develop a semimechanistic pharmacodynamic model describing tumor shrinkage after administration of a previously developed antitumor vaccine (CyaA-E7) in combination with CpG (a TLR9 ligand) and/or cyclophosphamide (CTX), and 2) to assess the translational capability of the model to describe tumor effects of different immune-based treatments. Population approach with NONMEM version 7.2 was used to analyze the previously published data. These data were generated by injecting 5 × 10(5) tumor cells expressing human papillomavirus (HPV)-E7 proteins into C57BL/6 mice. Large and established tumors were treated with CpG and/or CTX administered alone or in combination with CyaA-E7. Applications of the model were assessed by comparing model-based simulations with preclinical and clinical outcomes obtained from literature. CpG effects were modeled: 1) as an amplification of the immune signal triggered by the vaccine and 2) by shortening the delayed response of the vaccine. CTX effects were included through a direct decrease of the tumor-induced inhibition of vaccine efficacy over time, along with a delayed induction of tumor cell death. A pharmacodynamic model, built based on plausible biologic mechanisms known for the coadjuvants, successfully characterized tumor response in all experimental scenarios. The model developed was satisfactory applied to reproduce clinical outcomes when CpG or CTX was used in combination with different vaccines. The results found after simulation exercise indicated that the contribution of the coadjuvants to the tumor response elicited by vaccines can be predicted for other immune-based treatments.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Cancer Vaccines/pharmacology , Neoplasms/immunology , Neoplasms/therapy , Algorithms , Animals , Cyclophosphamide/pharmacology , Cyclophosphamide/therapeutic use , Female , Humans , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Mice , Mice, Inbred C57BL , Models, Biological , Models, Statistical , Neoplasms/pathology , Papillomavirus E7 Proteins/biosynthesis
12.
Front Pharmacol ; 14: 1211452, 2023.
Article in English | MEDLINE | ID: mdl-37771727

ABSTRACT

Introduction: Oncolytic viruses (OVs) represent a novel therapeutic strategy in oncology due to their capability to selectively infect and replicate in cancer cells, triggering a direct and/or immune-induced tumor lysis. However, the mechanisms governing OV pharmacokinetics are still poorly understood. This work aims to develop a physiologically based pharmacokinetic model of the novel OV, V937, in non-tumor-bearing mice to get a quantitative understanding of its elimination and tissue uptake processes. Materials and methods: Model development was performed using data obtained from 60 mice. Viral levels were quantified from eight tissues after a single intravenous V937 dose. An external dataset was used for model validation. This test set included multiple-dose experiments with different routes of administration. V937 distribution in each organ was described using a physiological structure based on mouse-specific organ blood flows and volumes. Analyses were performed using the non-linear mixed-effects approach with NONMEM 7.4. Results: Viral levels showed a drop from 108 to 105 copies/µg RNA at day 1 in blood, reflected in a high estimate of total clearance (18.2 mL/h). A well-stirred model provided an adequate description for all organs except the muscle and heart, where a saturable uptake process improved data description. The highest numbers of viral copies were observed in the brain, lymph node, kidney, liver, lung, and spleen on the first day after injection. On the other hand, the maximum amount of viral copies in the heart, muscle, and pancreas occurred 3 days after administration. Conclusion: To the best of our knowledge, this is the first physiologically based pharmacokinetic model developed to characterize OV biodistribution, representing a relevant source of quantitative knowledge regarding the in vivo behavior of OVs. This model can be further expanded by adding a tumor compartment, where OVs could replicate.

13.
Clin Pharmacol Ther ; 114(3): 623-632, 2023 09.
Article in English | MEDLINE | ID: mdl-37170933

ABSTRACT

Oncolytic viruses (OVs) represent a potential therapeutic strategy in cancer treatment. However, there is currently a lack of comprehensive quantitative models characterizing clinical OV kinetics and distribution to the tumor. In this work, we present a mechanistic modeling framework for V937 OV, after intratumoral (i.t.) or intravascular (i.v.) administration in patients with cancer. A minimal physiologically-based pharmacokinetic model was built to characterize biodistribution of OVs in humans. Viral dynamics was incorporated at the i.t. cellular level and linked to tumor response, enabling the characterization of a direct OV killing triggered by the death of infected tumor cells and an indirect killing induced by the immune response. The model provided an adequate description of changes in V937 mRNA levels and tumor size obtained from phase I/II clinical trials after V937 administration. The model showed prominent role of viral clearance from systemic circulation and infectivity in addition to known tumor aggressiveness on clinical response. After i.v. administration, i.t. exposure of V937 was predicted to be several orders of magnitude lower compared with i.t. administration. These differences could be overcome if there is high virus infectivity and/or replication. Unfortunately, the latter process could not be identified at the current clinical setting. This work provides insights on selecting optimal OV considering replication rate and infectivity.


Subject(s)
Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Humans , Oncolytic Viruses/genetics , Tissue Distribution , Neoplasms/therapy , Immunity
15.
Br J Pharmacol ; 179(14): 3815-3830, 2022 07.
Article in English | MEDLINE | ID: mdl-35170015

ABSTRACT

BACKGROUND AND PURPOSE: Acute intermittent porphyria (AIP) is a rare disease caused by a genetic mutation in the hepatic activity of the porphobilinogen-deaminase. We aimed to develop a mechanistic model of the enzymatic restoration effects of a novel therapy based on the administration of different formulations of recombinant human-PBGD (rhPBGD) linked to the ApoAI lipoprotein. This fusion protein circulates in blood, incorporating into HDL and penetrating hepatocytes. EXPERIMENTAL APPROACH: Single i.v. dose of different formulations of rhPBGD linked to ApoAI were administered to AIP mice in which a porphyric attack was triggered by i.p. phenobarbital. Data consist on 24 h urine excreted amounts of heme precursors, 5-aminolevulinic acid (ALA), PBG and total porphyrins that were analysed using non-linear mixed-effects analysis. KEY RESULTS: The mechanistic model successfully characterized over time the amounts excreted in urine of the three heme precursors for different formulations of rhPBGD and unravelled several mechanisms in the heme pathway, such as the regulation in ALA synthesis by heme. Treatment with rhPBGD formulations restored PBGD activity, increasing up to 51 times the value of the rate of tPOR formation estimated from baseline. Model-based simulations showed that several formulation prototypes provided efficient protective effects when administered up to 1 week prior to the occurrence of the AIP attack. CONCLUSION AND IMPLICATIONS: The model developed had excellent performance over a range of doses and formulation type. This mechanistic model warrants use beyond ApoAI-conjugates and represents a useful tool towards more efficient drug treatments of other enzymopenias as well as for acute intermittent porphyria.


Subject(s)
Porphyria, Acute Intermittent , Aminolevulinic Acid/pharmacology , Aminolevulinic Acid/urine , Animals , Disease Models, Animal , Heme , Mice , Mice, Inbred C57BL , Porphyria, Acute Intermittent/drug therapy , Porphyria, Acute Intermittent/genetics , Porphyria, Acute Intermittent/metabolism , Recombinant Proteins
16.
Front Pharmacol ; 12: 705443, 2021.
Article in English | MEDLINE | ID: mdl-34366859

ABSTRACT

V937 is an investigational novel oncolytic non-genetically modified Kuykendall strain of Coxsackievirus A21 which is in clinical development for the treatment of advanced solid tumor malignancies. V937 infects and lyses tumor cells expressing the intercellular adhesion molecule I (ICAM-I) receptor. We integrated in vitro and in vivo data from six different preclinical studies to build a mechanistic model that allowed a quantitative analysis of the biological processes of V937 viral kinetics and dynamics, viral distribution to tumor, and anti-tumor response elicited by V937 in human xenograft models in immunodeficient mice following intratumoral and intravenous administration. Estimates of viral infection and replication which were calculated from in vitro experiments were successfully used to describe the tumor response in vivo under various experimental conditions. Despite the predicted high clearance rate of V937 in systemic circulation (t1/2 = 4.3 min), high viral replication was observed in immunodeficient mice which resulted in tumor shrinkage with both intratumoral and intravenous administration. The described framework represents a step towards the quantitative characterization of viral distribution, replication, and oncolytic effect of a novel oncolytic virus following intratumoral and intravenous administrations in the absence of an immune response. This model may further be expanded to integrate the role of the immune system on viral and tumor dynamics to support the clinical development of oncolytic viruses.

17.
Comput Struct Biotechnol J ; 19: 4997-5007, 2021.
Article in English | MEDLINE | ID: mdl-34589180

ABSTRACT

Hepatitis B liver infection is caused by hepatitis B virus (HBV) and represents a major global disease problem when it becomes chronic, as is the case for 80-90% of vertical or early life infections. However, in the vast majority (>95%) of adult exposures, the infected individuals are capable of mounting an effective immune response leading to infection resolution. A good understanding of HBV dynamics and the interaction between the virus and immune system during acute infection represents an essential step to characterize and understand the key biological processes involved in disease resolution, which may help to identify potential interventions to prevent chronic hepatitis B. In this work, a quantitative systems pharmacology model for acute hepatitis B characterizing viral dynamics and the main components of the innate, adaptive, and tolerant immune response has been successfully developed. To do so, information from multiple sources and across different organization levels has been integrated in a common mechanistic framework. The final model adequately describes the chronology and plausibility of an HBV-triggered immune response, as well as clinical data from acute patients reported in the literature. Given the holistic nature of the framework, the model can be used to illustrate the relevance of the different immune pathways and biological processes to ultimate response, observing the negligible contribution of the innate response and the key contribution of the cellular response on viral clearance. More specifically, moderate reductions of the proliferation of activated cytotoxic CD8+ lymphocytes or increased immunoregulatory effects can drive the system towards chronicity.

18.
Cancers (Basel) ; 13(20)2021 Oct 09.
Article in English | MEDLINE | ID: mdl-34680196

ABSTRACT

Immune checkpoint inhibitors, administered as single agents, have demonstrated clinical efficacy. However, when treating cold tumors, different combination strategies are needed. This work aims to develop a semi-mechanistic model describing the antitumor efficacy of immunotherapy combinations in cold tumors. Tumor size of mice treated with TC-1/A9 non-inflamed tumors and the drug effects of an antigen, a toll-like receptor-3 agonist (PIC), and an immune checkpoint inhibitor (anti-programmed cell death 1 antibody) were modeled using Monolix and following a middle-out strategy. Tumor growth was best characterized by an exponential model with an estimated initial tumor size of 19.5 mm3 and a doubling time of 3.6 days. In the treatment groups, contrary to the lack of response observed in monotherapy, combinations including the antigen were able to induce an antitumor response. The final model successfully captured the 23% increase in the probability of cure from bi-therapy to triple-therapy. Moreover, our work supports that CD8+ T lymphocytes and resistance mechanisms are strongly related to the clinical outcome. The activation of antigen-presenting cells might be needed to achieve an antitumor response in reduced immunogenic tumors when combined with other immunotherapies. These models can be used as a platform to evaluate different immuno-oncology combinations in preclinical and clinical scenarios.

19.
Pharm Res ; 27(8): 1487-97, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20387096

ABSTRACT

Since gene therapy started over 20 years ago, more than one-thousand clinical trials have been carried out. Nonviral vectors present interesting properties for their clinical application, but their efficiency in vivo is relatively low, and further improvements in these vectors are needed. Elucidating how nonviral vectors behave at the intracellular level is enlightening for vector improvement and optimization. Model-based approach is a powerful tool to understand and describe the different processes that gene transfer systems should overcome inside the body. Model-based approach allows for proposing and predicting the effect of parameter changes on the overall gene therapy response, as well as the known application of the pharmacokinetic/pharmacodynamic modelling in conventional therapies. The objective of this paper is to critically review the works in which the time-course of naked or formulated DNA have been quantitatively studied or modelled.


Subject(s)
Genetic Therapy , Models, Biological , Genetic Therapy/adverse effects , Genetic Therapy/trends , Genetic Vectors/adverse effects , Genetic Vectors/pharmacokinetics , Humans
20.
Clin Pharmacol Ther ; 107(3): 597-606, 2020 03.
Article in English | MEDLINE | ID: mdl-31562819

ABSTRACT

Over the past decade, the insulin-like growth factor (IGF)-signaling pathway has gained substantial interest as potential therapeutic target in oncology. Xentuzumab, a humanized IgG1 monoclonal antibody, binds to IGF-I and IGF-II thereby inhibiting the downstream signaling essential for survival and tumor growth. This pathway is further regulated by circulating IGF binding proteins (IGFBPs). In this work, a mechanistic model characterizing the dynamics and interactions of IGFs, IGFBPs, and Xentuzumab has been developed to guide dose selection. Therefore, in vitro and in vivo literature information was combined with temporal IGF-I, IGF-II, and IGFBP-3 total plasma concentrations from two phase I studies. Based on the established quantitative framework, the time-course of free IGFs as ultimate drug targets not measured in clinics was predicted. Finally, a dose of 1000 mg/week-predicted to reduce free IGF-I and free IGF-II at steady-state by at least 90% and 64%, respectively-was suggested for phase II.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Insulin-Like Growth Factor II/immunology , Insulin-Like Growth Factor I/immunology , Models, Biological , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Neutralizing/immunology , Clinical Trials, Phase I as Topic , Dose-Response Relationship, Drug , Humans , Insulin-Like Growth Factor Binding Protein 3/blood
SELECTION OF CITATIONS
SEARCH DETAIL