Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 10 de 10
1.
Drug Metab Dispos ; 50(6): 858-866, 2022 06.
Article En | MEDLINE | ID: mdl-35149542

Characterization of the pharmacokinetics and biodistribution of therapeutic proteins (TPs) is a hot topic within the pharmaceutical industry, particularly with an ever-increasing catalog of novel modality TPs. Here, we review the current practices, and provide a summary of extensive cross-company discussions as well as a survey completed by International Consortium for Innovation and Quality members on this theme. A wide variety of in vitro, in vivo and in silico techniques are currently used to assess pharmacokinetics and biodistribution of TPs, and we discuss the relevance of these from an industry perspective, focusing on pharmacokinetic/pharmacodynamic understanding at the preclinical stage of development, and translation to human. We consider that the 'traditional in vivo biodistribution study' is becoming insufficient as a standalone tool, and thorough characterization of the interaction of the TP with its target(s), target biology, and off-target interactions at a microscopic scale are key to understand the overall biodistribution on a full-body scale. Our summary of the current challenges and our recommendations to address these issues could provide insight into the implementation of best practices in this area of drug development, and continued cross-company collaboration will be of tremendous value. SIGNIFICANCE STATEMENT: The Innovation and Quality Consortium Translational and ADME Sciences Leadership Group working group for the absorption, distribution, metabolism, and excretion of therapeutic proteins evaluates the current practices and challenges in characterizing the pharmacokinetics and biodistribution of therapeutic proteins during drug development, and proposes recommendations to address these issues. Incorporating the in vitro, in vivo and in silico approaches discussed herein may provide a pragmatic framework to increase early understanding of pharmacokinetic/pharmacodynamic relationships, and aid translational modeling for first-in-human dose predictions.


Drug Industry , Pharmacokinetics , Drug Industry/methods , Humans , Pharmaceutical Preparations , Tissue Distribution
2.
J Pharmacol Toxicol Methods ; 103: 106872, 2020.
Article En | MEDLINE | ID: mdl-32387500

Undesired immune responses against protein therapeutics may adversely affect the pharmacokinetics, efficacy, and safety of the product. The presence of anti-drug-antibodies (ADA) has been the key determinant of immunogenic responses. Here we describe the use of a capillary electrophoresis platform for the identification of ADAs against several experimental camelid VHH biologics (Nanobodies®). Hereafter, we refer to this assay as WESADA. We modified the Wes platform by ProteinSimple to screen serum samples for ADA against covalently linked multi-modular Nanobodies and compared it to standard ADA methodologies. We were able to identify ADA positive samples and determine which individual VHH module in a multivalent Nanobody construct stimulated the predominant ADA response. WESADA requires denaturation of the experimental immobilized drug, which could affect recognition of the immunogenic epitope and alter ADA signal. To address this issue, we demonstrated that signal can be immunodepleted by pre-incubation of serum samples with native Nanobody. This capillary electrophoresis based approach allows for rapid analysis without the need for individually tailored assay optimization or reagent labeling, while consuming small amounts of sample and drug. It also allows for the simultaneous ADA analysis of multiple targets of different molecular size in the same experimental sample. WESADA is not intended to replace traditional ADA assay formats, but it facilitates the expedient immunogenic assessment of a large number of experimental drug candidates in the early developmental space.


Biological Products/immunology , Biological Products/therapeutic use , Drug Monitoring/methods , Electrophoresis, Capillary/methods , Animals , Antibodies, Monoclonal , Mice, Inbred BALB C , Mice, Inbred C57BL
3.
Mol Cell Biol ; 39(22)2019 11 15.
Article En | MEDLINE | ID: mdl-31501275

The MYC oncogene is upregulated in human cancers by translocation, amplification, and mutation of cellular pathways that regulate Myc. Myc/Max heterodimers bind to E box sequences in the promoter regions of genes and activate transcription. The MYC inhibitor Omomyc can reduce the ability of MYC to bind specific box sequences in promoters of MYC target genes by binding directly to E box sequences as demonstrated by chromatin immunoprecipitation (CHIP). Here, we demonstrate by both a proximity ligation assay (PLA) and double chromatin immunoprecipitation (ReCHIP) that Omomyc preferentially binds to Max, not Myc, to mediate inhibition of MYC-mediated transcription by replacing MYC/MAX heterodimers with Omomyc/MAX heterodimers. The formation of Myc/Max and Omomyc/Max heterodimers occurs cotranslationally; Myc, Max, and Omomyc can interact with ribosomes and Max RNA under conditions in which ribosomes are intact. Taken together, our data suggest that the mechanism of action of Omomyc is to bind DNA as either a homodimer or a heterodimer with Max that is formed cotranslationally, revealing a novel mechanism to inhibit the MYC oncogene. We find that in vivo, Omomyc distributes quickly to kidneys and liver and has a short effective half-life in plasma, which could limit its use in vivo.


Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Genes, myc , Peptide Fragments/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Line , Cell Line, Tumor , Chromatin Immunoprecipitation/methods , DNA/metabolism , DNA-Binding Proteins/metabolism , Female , HCT116 Cells , Humans , Mice , Mice, Inbred BALB C , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/pharmacology , Recombinant Proteins/pharmacology , Transcription, Genetic , Transcriptional Activation
4.
AAPS J ; 21(3): 39, 2019 03 13.
Article En | MEDLINE | ID: mdl-30868312

Assessment of the factors that regulate antibody exposure-response relationships in the relevant animal models is critical for the design of successful translational strategies from discovery to the clinic. Depending on the specific clinical indication, preclinical development paradigms may require that the efficacy or dosing-related attributes for the existing antibody be assessed in various species when cross-reactivity of the lead antibody to the intended species is justified. Additionally, with the success of monoclonal antibodies for management of various human conditions, a parallel interest in therapeutic use of these novel modalities in various veterinary species has followed. The protective role of neonatal Fc receptor (FcRn) in regulation of IgG homeostasis and clearance is now well recognized and the "nonspecific clearance" of antibodies through bone marrow-derived phagocytic and vascular endothelial cells (via lysosomal processes) is modulated by interactions with FcRn receptors. In this study, we have attempted to examine the PK properties of human IgG antibodies in dog and monkey. These studies establish a translational framework for evaluation of IgG antibody PK properties across species.


Antibodies, Monoclonal, Humanized/pharmacokinetics , Immunoglobulin G/pharmacology , Administration, Intravenous , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Dogs , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Female , Humans , Macaca fascicularis , Macaca mulatta , Mice , Models, Animal , Respiratory Syncytial Viruses/immunology , Species Specificity
5.
Drug Metab Dispos ; 44(5): 617-23, 2016 May.
Article En | MEDLINE | ID: mdl-26669328

An antibody-drug conjugate (ADC) is a unique therapeutic modality composed of a highly potent drug molecule conjugated to a monoclonal antibody. As the number of ADCs in various stages of nonclinical and clinical development has been increasing, pharmaceutical companies have been exploring diverse approaches to understanding the disposition of ADCs. To identify the key absorption, distribution, metabolism, and excretion (ADME) issues worth examining when developing an ADC and to find optimal scientifically based approaches to evaluate ADC ADME, the International Consortium for Innovation and Quality in Pharmaceutical Development launched an ADC ADME working group in early 2014. This white paper contains observations from the working group and provides an initial framework on issues and approaches to consider when evaluating the ADME of ADCs.


Antibodies, Monoclonal/metabolism , Immunoconjugates/metabolism , Pharmaceutical Preparations/metabolism , Animals , Drug Industry/methods , Humans
6.
J Pharmacol Exp Ther ; 356(3): 574-86, 2016 Mar.
Article En | MEDLINE | ID: mdl-26669426

Administration of biologics to enhance T-cell function is part of a rapidly growing field of cancer immunotherapy demonstrated by the unprecedented clinical success of several immunoregulatory receptor targeting antibodies. While these biologic agents confer significant anti-tumor activity through targeted immune response modulation, they can also elicit broad immune responses potentially including the production of anti-drug antibodies (ADAs). DTA-1, an agonist monoclonal antibody against GITR, is a highly effective anti-tumor treatment in preclinical models. We demonstrate that repeated dosing with murinized DTA-1 (mDTA-1) generates ADAs with corresponding reductions in drug exposure and engagement of GITR on circulating CD3(+) CD4(+) T cells, due to rapid hepatic drug uptake and catabolism. Mice implanted with tumors after induction of preexisting mDTA-1 ADA show no anti-tumor efficacy when given 3 mg/kg mDTA-1, an efficacious dose in naive mice. Nonetheless, increasing mDTA-1 treatment to 30 mg/kg in ADA-positive mice restores mDTA-1 exposure and GITR engagement on circulating CD3(+) CD4(+) T cells, thereby partially restoring anti-tumor efficacy. Formation of anti-mDTA-1 antibodies and changes in drug exposure and disposition does not occur in GITR(-/-) mice, consistent with a role for GITR agonism in humoral immunity. Finally, the administration of muDX400, a murinized monoclonal antibody against the checkpoint inhibitor PD-1, dosed alone or combined with mDTA-1 did not result in reduced muDX400 exposure, nor did it change the nature of the anti-mDTA-1 response. This indicates that anti-GITR immunogenicity may not necessarily impact the pharmacology of coadministered monoclonal antibodies, supporting combination immunomodulatory strategies.


Antibodies, Monoclonal/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Drug Delivery Systems/methods , Glucocorticoid-Induced TNFR-Related Protein/agonists , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tissue Distribution/drug effects , Tissue Distribution/physiology , Xenograft Model Antitumor Assays/methods
7.
Drug Metab Dispos ; 40(2): 360-73, 2012 Feb.
Article En | MEDLINE | ID: mdl-22083830

Interleukin 10 (IL-10) is a potent cytokine homodimer with multiple immunoregulatory functions. Here, we have characterized the effects of PEGylation and formation of human IL-10 (hIL-10)/humanized anti-human IL-10 (hαhIL-10) immune complexes in the pharmacokinetics, biodistribution, and biotransformation of IL-10 in mice. To assess the fate of native, PEGylated, and antibody-bound IL-10; we implemented an analytical set of fluorescence emission-linked assays. Plasma size exclusion chromatography analysis indicated that fluoro-labeled native and PEGylated murine IL-10 (PEG-mIL-10) are stable in the circulation. PEGylation of IL-10 resulted in a 21-fold increased exposure, 2.7-fold increase in half-life, and 20-fold reduction in clearance. Kidney is the major organ of disposition for both native and PEGylated mIL-10 with renal uptake directly related to systemic clearance. The fluorescence signal in the kidneys reached tissue/blood ratios up to 150 and 20 for native and PEG-mIL-10, respectively. hIL-10/hαhIL-10 immune complexes are detectable in the circulation without evidence of unbound or degraded hIL-10. The exposure of hIL-10 present in immune complexes versus that of hIL-10 alone increased from 0.53 to 11.28 µg · day/ml, with a half-life of 1.16 days and a 23-fold reduction in clearance. Unlike hIL-10 alone, antibody-bound hIL-10 was targeted mainly to the liver with minimal renal distribution. In addition, we found an 11-fold reduction (from 9.9 to 113 nM) in binding to the neonatal Fc receptor (FcRn) when the hαhIL10 antibody is conjugated to hIL-10. The potential changes in FcRn binding in vivo and increased liver uptake may explain the unique pharmacokinetic properties of hIL-10/hαhIL-10 immune complexes.


Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Antigen-Antibody Reactions , Interleukin-10/chemistry , Interleukin-10/pharmacokinetics , Polyethylene Glycols/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Antigen-Antibody Complex/analysis , Female , Half-Life , Histocompatibility Antigens Class I/metabolism , Humans , Injections, Intravenous , Injections, Subcutaneous , Interleukin-10/antagonists & inhibitors , Interleukin-10/metabolism , Kidney/cytology , Kidney/immunology , Kidney/metabolism , Liver/cytology , Liver/immunology , Liver/metabolism , Metabolic Clearance Rate , Mice , Mice, Inbred Strains , Receptors, Fc/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacokinetics , Tissue Distribution
8.
Bioconjug Chem ; 17(2): 410-8, 2006.
Article En | MEDLINE | ID: mdl-16536473

CC49 is a clinically validated antibody with specificity for TAG-72, a carbohydrate epitope that is overexpressed and exposed on the cell surface in a large fraction of solid malignancies. We constructed a single-chain fragment (scFv) based on CC49 and fused it to beta-lactamase (BLA). Following optimization of the scFv domain by combinatorial consensus mutagenesis (CCM) for increased expression and stability, we characterized the protein variant for binding, in vivo pharmacokinetics (PK), and antitumor efficacy. The fusion protein TAB2.5 possessed a similar binding specificity relative to the parent antibody CC49. TAB2.5 also showed prolonged retention (T(1/2) = 36.9 h) in tumor-bearing mice with tumor/plasma ratios of up to 1000. Preliminary evaluation of TAB2.5, in combination with a novel prodrug, GC-Mel, resulted in significant efficacy in a colorectal xenograft tumor model and supports the utility of the protein as an agent for tumor-selective prodrug activation.


Antibodies, Neoplasm , Cephalosporins/chemistry , Cephalosporins/metabolism , Cephalosporins/therapeutic use , Nitrogen Mustard Compounds/chemistry , Nitrogen Mustard Compounds/metabolism , Nitrogen Mustard Compounds/therapeutic use , Prodrugs , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use , beta-Lactamases , Animals , Antibiotics, Antineoplastic/therapeutic use , Antibodies, Neoplasm/chemistry , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/metabolism , Antibodies, Neoplasm/therapeutic use , Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/metabolism , Antineoplastic Agents, Alkylating/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Doxorubicin/therapeutic use , Drug Carriers/chemistry , Drug Carriers/metabolism , Drug Delivery Systems , Female , Humans , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/metabolism , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/metabolism , Irinotecan , Melphalan/chemistry , Melphalan/metabolism , Melphalan/therapeutic use , Mice , Mice, Nude , Molecular Structure , Neoplasm Transplantation , Prodrugs/chemistry , Prodrugs/metabolism , Prodrugs/therapeutic use , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , beta-Lactamases/chemistry , beta-Lactamases/genetics , beta-Lactamases/metabolism , beta-Lactamases/therapeutic use
9.
Drug Metab Dispos ; 32(11): 1230-8, 2004 Nov.
Article En | MEDLINE | ID: mdl-15282212

Apo2L/TRAIL [Apo2 ligand/tumor necrosis factor (TNF)-related apoptosis-inducing ligand], a member of the TNF cytokine superfamily, induces cell death by apoptosis in a number of human cancer cells and is a potential agent for cancer therapy. We have characterized the in vitro stability of Apo2L/TRAIL in human serum and the tissue distribution and metabolism of Apo2L/TRAIL in a xenograft model of human colon carcinoma (COLO205). Apo2L/TRAIL was stable after incubation in human serum, with no significant high molecular weight complexes or degradation products observed. After i.v. administration of 125I-Apo2L/TRAIL to mice, a small percentage of the radiolabeled drug was seen as high molecular weight complex or as low molecular weight degradation products in plasma. However, the most abundant radioactive species corresponded to the intact Apo2L/TRAIL monomer, indicative of the relative stability of this recombinant protein in blood. Distribution of 125I-Apo2L/TRAIL to organs and solid xenograft tumors was limited. Intact 125I-Apo2L/TRAIL was detectable in the solid tumor at all time points and was the only tissue in which radioactivity transiently increased over time. Kidney contained the highest levels of radioactivity. Radioactive signal reached a tissue-to-blood ratio of 18 in the kidney cortex region when 125I-Apo2L/TRAIL was given in the presence of excess unlabeled ligand. In contrast to blood, extensive 125I-Apo2L/TRAIL degradation was observed in the kidney and, to a lesser degree, in the solid tumor and other organs, including liver, spleen, and lung. Our studies demonstrated that Apo2L/TRAIL is stable in the circulation, localizes to human solid xenograft tumors, and is primarily eliminated through the kidney.


Colonic Neoplasms/metabolism , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/pharmacokinetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacokinetics , Xenograft Model Antitumor Assays/methods , Animals , Apoptosis Regulatory Proteins , Humans , Kidney/drug effects , Kidney/metabolism , Membrane Glycoproteins/pharmacology , Mice , Mice, Nude , Organ Specificity , TNF-Related Apoptosis-Inducing Ligand , Tissue Distribution , Tumor Necrosis Factor-alpha/pharmacology
10.
Oncogene ; 21(22): 3611-9, 2002 May 16.
Article En | MEDLINE | ID: mdl-12032863

Apo2L/TRAIL exhibits enhanced apoptotic activity in tumor xenograft models when used in combination with the topoisomerase 1 inhibitor CPT-11. To investigate the cellular mechanisms involved in this increased tumor-killing activity, a series of in vitro experiments were conducted using the human colon carcinoma cell line (HCT116). Apo2L/TRAIL induced a transient upregulation of DR5 mRNA, while CPT-11 increased both death and decoy receptor expression. Upregulation of decoy receptors by CPT-11 was partially inhibited by co-administration of Apo2L/TRAIL. CPT-11 treatment resulted in accumulation of cells at G(2)M-phase and correlated with a substantial increase in the protein levels of the cyclin-dependent kinase inhibitor p21. However, cells co-treated with CPT-11 and Apo2L/TRAIL, or pretreated with CPT-11 for up to 24 h followed by 2 h Apo2L/TRAIL, resulted in a caspase-dependent degradation of p21, reversal of G(2)-M phase arrest with a concomitant increase in apoptosis. The sequential treatment produced the greatest induction of DR5 and DR4, caspase-3-like cleavage/activation and p21 degradation, as well as increased apoptosis. These data indicate that the up-regulation of Apo2L/TRAIL ligand and its death receptors as well as cleavage of p21 protein in the Apo2L/TRAIL plus CPT-11 treatment contributes to the positive cooperation between these agents in enhancing tumor cell apoptosis.


Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/pharmacology , Cyclins/metabolism , Membrane Glycoproteins/pharmacology , Neoplasms/drug therapy , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis , Apoptosis Regulatory Proteins , Camptothecin/analogs & derivatives , Camptothecin/antagonists & inhibitors , Carcinoma/drug therapy , Carcinoma/metabolism , Carcinoma/pathology , Caspases/metabolism , Cell Cycle , Cells, Cultured , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p21 , Drug Synergism , Humans , Irinotecan , Kinetics , Neoplasms/metabolism , Neoplasms/pathology , RNA, Neoplasm/biosynthesis , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured
...