Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Bioconjug Chem ; 34(11): 2096-2111, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37916986

ABSTRACT

Antisense-oligonucleotides (ASOs) are a promising drug modality for the treatment of neurological disorders, but the currently established route of administration via intrathecal delivery is a major limitation to its broader clinical application. An attractive alternative is the conjugation of the ASO to an antibody that facilitates access to the central nervous system (CNS) after peripheral application and target engagement at the blood-brain barrier, followed by transcytosis. Here, we show that the diligent conjugate design of Brainshuttle-ASO conjugates is the key to generating promising delivery vehicles and thereby establishing design principles to create optimized molecules with drug-like properties. An innovative site-specific transglutaminase-based conjugation technology was chosen and optimized in a stepwise process to identify the best-suited conjugation site, tags, reaction conditions, and linker design. The overall conjugation performance was found to be specifically governed by the choice of buffer conditions and the structure of the linker. The combination of the peptide tags YRYRQ and RYESK was chosen, showing high conjugation fidelity. Elaborate conjugate analysis revealed that one leading differentiating factor was hydrophobicity. The increase of hydrophobicity by the ASO payload could be mitigated by the appropriate choice of conjugation site and the heavy chain position 297 proved to be the most optimal. Evaluating the properties of the linker suggested a short bicyclo[6.1.0]nonyne (BCN) unit as best suited with regards to conjugation performance and potency. Promising in vitro activity and in vivo pharmacokinetic behavior of optimized Brainshuttle-ASO conjugates, based on a microtubule-associated protein tau (MAPT) targeting oligonucleotide, suggest that such designs have the potential to serve as a blueprint for peripherally delivered ASO-based drugs for the CNS in the future.


Subject(s)
Antibodies , Oligonucleotides, Antisense , Oligonucleotides, Antisense/chemistry , Oligonucleotides , Peptides
2.
Drug Discov Today ; 27(6): 1604-1621, 2022 06.
Article in English | MEDLINE | ID: mdl-35304340

ABSTRACT

Many in vitro and in vivo models are used in pharmacological research to evaluate the role of targeted proteins in a disease. Understanding the translational relevance and limitation of these models for analyzing a drug's disposition, pharmacokinetic/pharmacodynamic (PK/PD) profile, mechanism, and efficacy, is essential when selecting the most appropriate model of the disease of interest and predicting clinically efficacious doses of the investigational drug. Selected animal models used in ophthalmology, infectious diseases, oncology, autoimmune diseases, and neuroscience are reviewed here. Each area has specific challenges around translatability and determination of an efficacious dose: new patient-specific dosing methods may help overcome these limitations.


Subject(s)
Drugs, Investigational , Medical Oncology , Animals , Models, Biological
4.
Hepatology ; 74(4): 1795-1808, 2021 10.
Article in English | MEDLINE | ID: mdl-34037271

ABSTRACT

BACKGROUND AND AIMS: RO7062931 is an N-acetylgalactosamine (GalNAc)-conjugated single-stranded locked nucleic acid oligonucleotide complementary to HBV RNA. GalNAc conjugation targets the liver through the asialoglycoprotein receptor (ASGPR). This two-part phase 1 study evaluated the safety, pharmacokinetics, and pharmacodynamics of RO7062931 in healthy volunteers and patients with chronic hepatitis B (CHB) who were virologically suppressed. APPROACH AND RESULTS: Part 1 was a single ascending dose study in healthy volunteers randomized to receive a single RO7062931 dose (0.1-4.0 mg/kg), or placebo. Part 2 was a multiple ascending dose study in patients with CHB randomized to receive RO7062931 at 0.5, 1.5, or 3.0 mg/kg or placebo every month for a total of 2 doses (Part 2a) or RO7062931 at 3.0 mg/kg every 2 weeks, 3.0 mg/kg every week (QW), or 4.0 mg/kg QW or placebo for a total of 3-5 doses (Part 2b). Sixty healthy volunteers and 59 patients received RO7062931 or placebo. The majority of adverse events (AEs) reported were mild in intensity. Common AEs included self-limiting injection site reactions and influenza-like illness. Supradose-proportional increases in RO7062931 plasma exposure and urinary excretion occurred at doses ≥3.0 mg/kg. In patients with CHB, RO7062931 resulted in dose-dependent and time-dependent reduction in HBsAg versus placebo. The greatest HBsAg declines from baseline were achieved with the 3.0 mg/kg QW dose regimen (mean nadir ~0.5 log10  IU/mL) independent of HBeAg status. CONCLUSIONS: RO7062931 is safe and well tolerated at doses up to 4.0 mg/kg QW. Supradose-proportional exposure at doses of 3.0-4.0 mg/kg was indicative of partial saturation of the ASGPR-mediated liver uptake system. Dose-dependent declines in HBsAg demonstrated target engagement with RO7062931.


Subject(s)
Acetylgalactosamine/therapeutic use , Hepatitis B, Chronic/drug therapy , Oligonucleotides, Antisense/therapeutic use , Oligonucleotides/therapeutic use , Acetylgalactosamine/analogs & derivatives , Adult , Asialoglycoprotein Receptor , Female , Healthy Volunteers , Hepatitis B Surface Antigens/blood , Hepatitis B virus/genetics , Hepatitis B, Chronic/blood , Humans , Male , Middle Aged , Oligonucleotides/genetics , Oligonucleotides, Antisense/genetics , RNA, Viral/genetics , Sustained Virologic Response
5.
Sci Rep ; 7(1): 18086, 2017 12 22.
Article in English | MEDLINE | ID: mdl-29273809

ABSTRACT

RG7787 is a re-engineered mesothelin-targeted immunotoxin with reduced immunogenicity composed of a humanized anti-mesothelin Fab fragment and a B-cell epitope silenced 24 kD fragment of Pseudomonas exotoxin A. High prevalence of mesothelin-positive cases and a large unmet medical need make ovarian cancer a promising indication for the clinical development of RG7787. However, ovarian cancer patients also frequently have elevated serum levels of the cancer antigen 125 (CA-125). In principle this could pose a problem, since the binding sites for CA-125 and RG7787 on mesothelin were reported to overlap. However, we show here that RG7787 can readily displace even excess amounts of CA-125 in different cellular assays. Moreover when tested in-vitro on a panel of 12 ovarian cancer cell lines, RG7787 had high cytotoxic activity on COV644, Caov-4, and SNU-119 cells and fully inhibited growth of EFO-21, KURAMOCHI, OVSAHO, and Caov-3 cells with potency values ranging from 1 to 86 pM. Finally, we evaluated the in-vivo efficacy of RG7787 in OvCa6668, a patient-derived ovarian cancer model with high levels of CA-125 expression. RG7787 had moderate monotherapy efficacy but in combination with standard chemotherapies (cisplatin, paclitaxel) achieved pronounced tumor regressions. In summary our data support clinical testing of RG7787 in ovarian cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Cell Proliferation/drug effects , Immunoconjugates/therapeutic use , Immunotoxins/therapeutic use , Ovarian Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Female , Humans , Immunoconjugates/pharmacology , Immunotoxins/pharmacology
6.
MAbs ; 9(8): 1360-1369, 2017.
Article in English | MEDLINE | ID: mdl-28876162

ABSTRACT

The abundant cell surface asialoglycoprotein receptor (ASGPR) is a highly selective receptor found on hepatocytes that potentially can be exploited as a selective shuttle for delivery. Various nucleic acid therapeutics that bind ASGPR are already in clinical development, but this receptor-mediated delivery mechanism can be saturated, which will likely result in reduced selectivity for the liver and therefore increase the likelihood for systemic adverse effects. Therefore, when aiming to utilize this mechanism, it is important to optimize both the administration protocol and the molecular properties. We here present a study using a novel ASGPR-targeted antibody to estimate ASGPR expression, turnover and internalization rates in vivo in mice. Using pharmacokinetic data (intravenous and subcutaneous dosing) and an in-silico target-mediated drug disposition (TMDD) model, we estimate an ASGPR expression level of 1.8 million molecules per hepatocyte. The half-life of the degradation of the receptor was found to be equal to 15 hours and the formed ligand-receptor complex is internalized with a half-life of 5 days. A biodistribution study was performed and confirmed the accuracy of the TMDD model predictions. The kinetics of the ASGPR shows that saturation of the shuttle at therapeutic concentrations is possible; however, simulation allows the dosing schedule to be optimized. The developed TMDD model can be used to support the development of therapies that use the ASGPR as a shuttle into hepatocytes.


Subject(s)
Asialoglycoprotein Receptor/metabolism , Drug Delivery Systems/methods , Hepatocytes/metabolism , Liver/metabolism , Pharmaceutical Preparations/administration & dosage , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Asialoglycoprotein Receptor/immunology , Endocytosis , HEK293 Cells , Humans , Kinetics , Mice , Molecular Targeted Therapy/methods , Tissue Distribution
7.
J Physiol ; 595(24): 7311-7330, 2017 12 15.
Article in English | MEDLINE | ID: mdl-28960303

ABSTRACT

KEY POINTS: For therapeutic antibodies, total tissue concentrations are frequently reported as a lump sum measure of the antibody in residual plasma, interstitial fluid and cells. In terms of correlating antibody exposure to a therapeutic effect, however, interstitial pharmacokinetics might be more relevant. In the present study, we collected total tissue and interstitial antibody biodistribution data in mice and assessed the composition of tissue samples aiming to correct total tissue measurements for plasma and cellular content. All data and parameters were integrated into a refined physiologically-based pharmacokinetic model for monoclonal antibodies to enable the tissue-specific description of antibody pharmacokinetics in the interstitial space. We found that antibody interstitial concentrations are highly tissue-specific and dependent on the underlying capillary structure but, in several tissues, they reach relatively high interstitial concentrations, contradicting the still-prevailing view that both the distribution to tissues and the interstitial concentrations for antibodies are generally low. ABSTRACT: For most therapeutic antibodies, the interstitium is the target space. Although experimental methods for measuring antibody pharmacokinetics (PK) in this space are not well established, thus making quantitative assessment difficult, the interstitial antibody concentration is assumed to be low. In the present study, we combined direct quantification of antibodies in the interstitial fluid with a physiologically-based PK (PBPK) modelling approach, with the aim of better describing the PK of monoclonal antibodies in the interstitial space of different tissues. We isolated interstitial fluid by tissue centrifugation and conducted an antibody biodistribution study in mice, measuring total tissue and interstitial concentrations in selected tissues. Residual plasma, interstitial volumes and lymph flows, which are important PBPK model parameters, were assessed in vivo. We could thereby refine the PBPK modelling of monoclonal antibodies, better interpret antibody biodistribution data and more accurately predict their PK in the different tissue spaces. Our results indicate that, in tissues with discontinuous capillaries (liver and spleen), interstitial concentrations are reflected by the plasma concentration. In tissues with continuous capillaries (e.g. skin and muscle), ∼50-60% of the plasma concentration is found in the interstitial space. In the brain and kidney, on the other hand, antibodies are restricted to the vascular space. Our data may significantly impact the interpretation of biodistribution data of monoclonal antibodies and might be important when relating measured concentrations to a therapeutic effect. By contrast to the view that the antibody distribution to the interstitial space is limited, using direct measurements and model-based data interpretation, we show that high antibody interstitial concentrations are reached in most tissues.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Extracellular Fluid/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Blood Vessels/metabolism , Female , Interleukin-17/immunology , Liver/metabolism , Male , Mice , Spleen/metabolism , Tissue Distribution
8.
Pharm Res ; 34(10): 2131-2141, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28681164

ABSTRACT

PURPOSE: To establish a continuous relationship between the size of various antibody fragments and their systemic clearance (CL) in mice. METHODS: Two different orthogonal approaches have been used to establish the relationship. First approach uses CL values estimated by non-compartmental analysis (NCA) to establish a correlation with protein size. The second approach simultaneously characterizes the PK data for all the proteins using a 2-compartment model to establish a relationship between protein size and pharmacokinetic (PK) parameters. RESULTS: Simple mathematical functions (e.g. sigmoidal, power law) were able to characterize the CL vs. protein size relationship generated using the investigated proteins. The relationship established in mouse was used to predict rat, rabbit, monkey, and human relationships using allometric scaling. The predicted relationships were found to capture the available spares data from each species reasonably well. CONCLUSIONS: The CL vs. protein size relationship is important for establishing a robust quantitative structure-PK relationship (QSPKR) for protein therapeutics. The relationship presented here can help in a priori predicting plasma exposure of therapeutic proteins, and together with our previously established relationship between plasma and tissue concentrations of proteins, it can predict the tissue exposure of non-binding proteins simply based on molecular weight/radius and dose.


Subject(s)
Antibodies, Monoclonal/pharmacology , Immunoglobulin Fragments/pharmacology , Models, Biological , Animals , Antibodies, Monoclonal/chemistry , Haplorhini , Humans , Immunoglobulin Fragments/chemistry , Kinetics , Mice , Molecular Structure , Molecular Weight , Rabbits , Rats , Small Molecule Libraries , Structure-Activity Relationship
9.
MAbs ; 9(6): 1007-1015, 2017.
Article in English | MEDLINE | ID: mdl-28613103

ABSTRACT

Monoclonal antibodies are an important therapeutic entity, and knowledge of antibody pharmacokinetics has steadily increased over the years. Despite this effort, little is known about the extent of IgG antibody degradation in different tissues of the body. While studies have been published identifying sites of degradation with the use of residualizing and non-residualizing radiolabels, quantitative tissue clearances have not yet been derived. Here, we show that in physiologically-based pharmacokinetic (PBPK) models we can combine mouse data of Indium-111 and Iodine-125 labeled antibodies with prior physiologic knowledge to determine tissue-specific intrinsic clearances. Unspecific total tissue clearance (mL/day) in the mouse was estimated to be: liver = 4.75; brain = 0.02; gut = 0.40; heart = 0.07; kidney = 0.97; lung = 0.20; muscle = 3.02; skin = 3.89; spleen = 0.45; rest of body = 2.16. The highest catabolic activity (per g tissue) was in spleen for an FcRn wild-type antibody, but shifts to the liver for an antibody with reduced FcRn affinity. In the model developed, this shift can be explained by the liver having a greater FcRn-mediated protection capacity than the spleen. The quantification of tissue intrinsic clearances and FcRn salvage capacity increases our understanding of quantitative processes that drive the therapeutic responses of antibodies. This knowledge is critical, for instance to estimate the non-specific cellular uptake and degradation of antibodies used for targeted delivery of payloads.

10.
Mol Oncol ; 10(8): 1317-29, 2016 10.
Article in English | MEDLINE | ID: mdl-27507537

ABSTRACT

Mesothelin overexpression in lung adenocarcinomas correlates with the presence of activating KRAS mutations and poor prognosis. Hence SS1P, a mesothelin-targeted immunotoxin, could offer valuable treatment options for these patients, but its use in solid tumor therapy is hampered by high immunogenicity and non-specific toxicity. To overcome both obstacles we developed RG7787, a de-immunized cytotoxic fusion protein comprising a humanized SS1 Fab fragment and a truncated, B-cell epitope silenced, 24 kD fragment of Pseudomonas exotoxin A (PE24). Reactivity of RG7787 with sera from immunotoxin-treated patients was >1000 fold reduced. In vitro RG7787 inhibited cell viability of lung cancer cell lines with picomolar potency. The pharmacokinetic properties of RG7787 in rodents were comparable to SS1P, yet it was tolerated up to 10 fold better without causing severe vascular leak syndrome or hepatotoxicity. A pharmacokinetic/pharmacodynamic model developed based on NCI-H596 xenograft studies showed that for RG7787 and SS1P, their in vitro and in vivo potencies closely correlate. At optimal doses of 2-3 mg/kg RG7787 is more efficacious than SS1P. Even large, well established tumors (600 mm(3)) underwent remission during three treatment cycles with RG7787. Also in two patient-derived lung cancer xenograft models, Lu7336 and Lu7187, RG7787 showed anti-tumor efficacy. In monotherapy two treatment cycles were moderately efficacious in the Lu7336 model but showed good anti-tumor activity in the KRAS mutant Lu7187 model (26% and 80% tumor growth inhibition, respectively). Combination of RG7787 with standard chemotherapies further enhanced efficacy in both models achieving near complete eradication of Lu7187 tumors.


Subject(s)
ADP Ribose Transferases/therapeutic use , Bacterial Toxins/therapeutic use , Exotoxins/therapeutic use , GPI-Linked Proteins/metabolism , Lung Neoplasms/drug therapy , Protein Engineering , Pseudomonas/metabolism , Recombinant Fusion Proteins/therapeutic use , Virulence Factors/therapeutic use , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Liver/drug effects , Liver/pathology , Lung Neoplasms/pathology , Mesothelin , Mice, SCID , Models, Biological , Rats , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/pharmacology , Xenograft Model Antitumor Assays , Pseudomonas aeruginosa Exotoxin A
11.
MAbs ; 8(1): 113-9, 2016.
Article in English | MEDLINE | ID: mdl-26496429

ABSTRACT

Biodistribution coefficients (BC) allow estimation of the tissue concentrations of proteins based on the plasma pharmacokinetics. We have previously established the BC values for monoclonal antibodies. Here, this concept is extended by development of a relationship between protein size and BC values. The relationship was built by deriving the BC values for various antibody fragments of known molecular weight from published biodistribution studies. We found that there exists a simple exponential relationship between molecular weight and BC values that allows the prediction of tissue distribution of proteins based on molecular weight alone. The relationship was validated by a priori predicting BC values of 4 antibody fragments that were not used in building the relationship. The relationship was also used to derive BC50 values for all the tissues, which is the molecular weight increase that would result in 50% reduction in tissue uptake of a protein. The BC50 values for most tissues were found to be ~35 kDa. An ability to estimate tissue distribution of antibody fragments based on the BC vs. molecular size relationship established here may allow better understanding of the biologics concentrations in tissues responsible for efficacy or toxicity. This relationship can also be applied for rational development of new biotherapeutic modalities with optimal biodistribution properties to target (or avoid) specific tissues.


Subject(s)
Immunoglobulin Fragments/pharmacology , Models, Biological , Animals , Humans , Molecular Weight , Tissue Distribution
12.
Integr Biol (Camb) ; 6(8): 736-42, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24934872

ABSTRACT

Cells sense information encoded in extracellular ligand concentrations and process it using intracellular signalling cascades. Using mathematical modelling and high-throughput imaging of individual cells, we studied how a transient extracellular growth factor signal is sensed by the epidermal growth factor receptor system, processed by downstream signalling, and transmitted to the nucleus. We found that transient epidermal growth factor signals are linearly translated into an activated epidermal growth factor receptor integrated over time. This allows us to generate a simplified model of receptor signaling where the receptor acts as a perfect sensor of extracellular information, while the nonlinear input-output relationship of EGF-EGFR triggered signalling is a consequence of the downstream MAPK cascade alone.


Subject(s)
ErbB Receptors/metabolism , MAP Kinase Signaling System , Active Transport, Cell Nucleus , Cell Membrane/metabolism , Cell Nucleus/metabolism , Endosomes/metabolism , HeLa Cells , Humans , Ligands , Models, Biological , Phosphorylation , Time Factors
13.
PLoS One ; 8(6): e67330, 2013.
Article in English | MEDLINE | ID: mdl-23840665

ABSTRACT

Capecitabine (CAP) is a 5-FU pro-drug approved for the treatment of several cancers and it is used in combination with gemcitabine (GEM) in the treatment of patients with pancreatic adenocarcinoma (PDAC). However, limited pre-clinical data of the effects of CAP in PDAC are available to support the use of the GEMCAP combination in clinic. Therefore, we investigated the pharmacokinetics and the efficacy of CAP as a single agent first and then in combination with GEM to assess the utility of the GEMCAP therapy in clinic. Using a model of spontaneous PDAC occurring in Kras(G12D); p53(R172H); Pdx1-Cre (KPC) mice and subcutaneous allografts of a KPC PDAC-derived cell line (K8484), we showed that CAP achieved tumour concentrations (∼25 µM) of 5-FU in both models, as a single agent, and induced survival similar to GEM in KPC mice, suggesting similar efficacy. In vitro studies performed in K8484 cells as well as in human pancreatic cell lines showed an additive effect of the GEMCAP combination however, it increased toxicity in vivo and no benefit of a tolerable GEMCAP combination was identified in the allograft model when compared to GEM alone. Our work provides pre-clinical evidence of 5-FU delivery to tumours and anti-tumour efficacy following oral CAP administration that was similar to effects of GEM. Nevertheless, the GEMCAP combination does not improve the therapeutic index compared to GEM alone. These data suggest that CAP could be considered as an alternative to GEM in future, rationally designed, combination treatment strategies for advanced pancreatic cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/toxicity , Capecitabine , Carcinoma, Pancreatic Ductal/enzymology , Cell Line, Tumor , Cytidine Deaminase/metabolism , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Disease Models, Animal , Drug Screening Assays, Antitumor , Fluorouracil/administration & dosage , Fluorouracil/analogs & derivatives , Mice , Mice, Transgenic , Neoplasm Transplantation , Pancreatic Neoplasms/enzymology , Prodrugs/administration & dosage , Tissue Distribution , Gemcitabine
14.
J Am Chem Soc ; 135(26): 9640-3, 2013 Jul 03.
Article in English | MEDLINE | ID: mdl-23782415

ABSTRACT

Synthetic lethality is a genetic concept in which cell death is induced by the combination of mutations in two sensitive genes, while mutation of either gene alone is not sufficient to affect cell survival. Synthetic lethality can also be achieved "chemically" by combination of drug-like molecules targeting distinct but cooperative pathways. Previously, we reported that the small molecule pyridostatin (PDS) stabilizes G-quadruplexes (G4s) in cells and elicits a DNA damage response by causing the formation of DNA double strand breaks (DSB). Cell death mediated by ligand-induced G4 stabilization can be potentiated in cells deficient in DNA damage repair genes. Here, we demonstrate that PDS acts synergistically both with NU7441, an inhibitor of the DNA-PK kinase crucial for nonhomologous end joining repair of DNA DSBs, and BRCA2-deficient cells that are genetically impaired in homologous recombination-mediated DSB repair. G4 targeting ligands have potential as cancer therapeutic agents, acting synergistically with inhibition or mutation of the DNA damage repair machinery.


Subject(s)
DNA, Neoplasm/genetics , G-Quadruplexes , Neoplasms/genetics , Aminoquinolines/chemistry , Aminoquinolines/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , BRCA2 Protein/deficiency , BRCA2 Protein/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chromones/chemistry , Chromones/pharmacology , DNA Breaks , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Morpholines/chemistry , Morpholines/pharmacology , Neoplasms/drug therapy , Neoplasms/pathology , Picolinic Acids/chemistry , Picolinic Acids/pharmacology , Structure-Activity Relationship
15.
J Pharmacokinet Pharmacodyn ; 39(2): 125-39, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22399130

ABSTRACT

Cell-level kinetic models for therapeutically relevant processes increasingly benefit the early stages of drug development. Later stages of the drug development processes, however, rely on pharmacokinetic compartment models while cell-level dynamics are typically neglected. We here present a systematic approach to integrate cell-level kinetic models and pharmacokinetic compartment models. Incorporating target dynamics into pharmacokinetic models is especially useful for the development of therapeutic antibodies because their effect and pharmacokinetics are inherently interdependent. The approach is illustrated by analysing the F(ab)-mediated inhibitory effect of therapeutic antibodies targeting the epidermal growth factor receptor. We build a multi-level model for anti-EGFR antibodies by combining a systems biology model with in vitro determined parameters and a pharmacokinetic model based on in vivo pharmacokinetic data. Using this model, we investigated in silico the impact of biochemical properties of anti-EGFR antibodies on their F(ab)-mediated inhibitory effect. The multi-level model suggests that the F(ab)-mediated inhibitory effect saturates with increasing drug-receptor affinity, thereby limiting the impact of increasing antibody affinity on improving the effect. This indicates that observed differences in the therapeutic effects of high affinity antibodies in the market and in clinical development may result mainly from Fc-mediated indirect mechanisms such as antibody-dependent cell cytotoxicity.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Cell Membrane/metabolism , Immunoglobulin Fab Fragments/physiology , Models, Biological , Animals , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Cell Membrane/drug effects , Forecasting , Humans , Macaca fascicularis , Signal Transduction/drug effects , Signal Transduction/physiology
16.
J Biomol Screen ; 14(8): 913-23, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19675314

ABSTRACT

The potential of enzyme inhibition of a drug is frequently quantified in terms of IC(50) values. Although this is a suitable quantity for reversible inhibitors, concerns arise when dealing with irreversible or mechanism-based inhibitors (MBIs). IC(50) values of MBIs are time dependent, causing serious problems when aiming at ranking different compounds with respect to their inhibitory potential. As a consequence, most studies and ranking schemes related to MBIs rely on the inhibition constant (K(I)) and the rate of enzyme inactivation (k(inact)) rather than on IC(50) values. In this article, the authors derive a novel relation between potentially time-dependent IC(50) values and K(I), k(inact) parameters for different types of inhibition. This allows for direct estimation of K(I) and k(inact) values from time-dependent IC(50) values, even without the need of additional preincubation experiments. The application of this approach is illustrated using a fluorimetric assay to access the drug-drug interaction potential associated with new chemical entities. The approach can easily be implemented using standard software tools (e.g., XLfit) and may also be suitable for applications where mechanism-based inhibition is a desired mode of action (e.g., at particular pharmacological drug targets).


Subject(s)
Drug Antagonism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacokinetics , Inhibitory Concentration 50 , Binding, Competitive , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP1A2 Inhibitors , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors , Humans , Models, Chemical , Models, Theoretical , Time Factors
17.
J Pharmacokinet Pharmacodyn ; 36(3): 239-60, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19554432

ABSTRACT

Receptor mediated endocytosis (RME) plays a major role in the disposition of therapeutic protein drugs in the body. It is suspected to be a major source of nonlinear pharmacokinetic behavior observed in clinical pharmacokinetic data. So far, mostly empirical or semi-mechanistic approaches have been used to represent RME. A thorough understanding of the impact of the properties of the drug and of the receptor system on the resulting nonlinear disposition is still missing, as is how to best represent RME in pharmacokinetic models. In this article, we present a detailed mechanistic model of RME that explicitly takes into account receptor binding and trafficking inside the cell and that is used to derive reduced models of RME which retain a mechanistic interpretation. We find that RME can be described by an extended Michaelis-Menten model that accounts for both the distribution and the elimination aspect of RME. If the amount of drug in the receptor system is negligible a standard Michaelis-Menten model is capable of describing the elimination by RME. Notably, a receptor system can efficiently eliminate drug from the extracellular space even if the total number of receptors is small. We find that drug elimination by RME can result in substantial nonlinear pharmacokinetics. The extent of nonlinearity is higher for drug/receptor systems with higher receptor availability at the membrane, or faster internalization and degradation of extracellular drug. Our approach is exemplified for the epidermal growth factor receptor system.


Subject(s)
Endocytosis/physiology , ErbB Receptors/physiology , Membrane Proteins/metabolism , Nonlinear Dynamics , Signal Transduction/physiology , ErbB Receptors/metabolism , Protein Transport/physiology
18.
J Biomol Screen ; 12(1): 92-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17130250

ABSTRACT

In drug discovery, the potential of cytochrome P450 inhibition of new chemical entities is frequently quantified in terms of IC50 values. In early drug discovery, a risk classification into low, medium, or high potential inhibitors is often sufficient for ranking and prioritizing of compounds. Although often 6 or more inhibitor concentrations are used to determine the IC50 value, the question arises whether it is possible to predict the risk class based on fewer inhibitor concentrations with comparable reliability. In this article, the authors propose a new integrated 2-point method with inhibitor concentrations chosen in accordance with the risk classification. They analyze its predictive power and the feasibility of not only classifying the compounds into different risk classes but also ranking those compounds that have been binned into the middle risk class. The proposed integrated 2-point method is thus highly suitable for automation. Altogether, it maintains the quality of the prediction while considerably reducing time and cost. The proposed method is applicable to other IC50 assays and risk classifications.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors , Drug Design , Enzyme Inhibitors/classification , Enzyme Inhibitors/pharmacology , Drug Interactions , Humans , Inhibitory Concentration 50 , Isoenzymes/antagonists & inhibitors , Substrate Specificity/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...