1
|
Schlosser P, Zhang J, Liu H, Surapaneni AL, Rhee EP, Arking DE, Yu B, Boerwinkle E, Welling PA, Chatterjee N, Susztak K, Coresh J, Grams ME. Transcriptome- and proteome-wide association studies nominate determinants of kidney function and damage. Genome Biol 2023; 24:150. [PMID: 37365616 PMCID: PMC10291807 DOI: 10.1186/s13059-023-02993-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 06/15/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND The pathophysiological causes of kidney disease are not fully understood. Here we show that the integration of genome-wide genetic, transcriptomic, and proteomic association studies can nominate causal determinants of kidney function and damage. RESULTS Through transcriptome-wide association studies (TWAS) in kidney cortex, kidney tubule, liver, and whole blood and proteome-wide association studies (PWAS) in plasma, we assess for effects of 12,893 genes and 1342 proteins on kidney filtration (glomerular filtration rate (GFR) estimated by creatinine; GFR estimated by cystatin C; and blood urea nitrogen) and kidney damage (albuminuria). We find 1561 associations distributed among 260 genomic regions that are supported as putatively causal. We then prioritize 153 of these genomic regions using additional colocalization analyses. Our genome-wide findings are supported by existing knowledge (animal models for MANBA, DACH1, SH3YL1, INHBB), exceed the underlying GWAS signals (28 region-trait combinations without significant GWAS hit), identify independent gene/protein-trait associations within the same genomic region (INHBC, SPRYD4), nominate tissues underlying the associations (tubule expression of NRBP1), and distinguish markers of kidney filtration from those with a role in creatinine and cystatin C metabolism. Furthermore, we follow up on members of the TGF-beta superfamily of proteins and find a prognostic value of INHBC for kidney disease progression even after adjustment for measured glomerular filtration rate (GFR). CONCLUSION In summary, this study combines multimodal, genome-wide association studies to generate a catalog of putatively causal target genes and proteins relevant to kidney function and damage which can guide follow-up studies in physiology, basic science, and clinical medicine.
Collapse
Affiliation(s)
- Pascal Schlosser
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| | - Jingning Zhang
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Hongbo Liu
- Department of Medicine and Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aditya L Surapaneni
- Welch Center for Prevention Epidemiology and Clinical Research, Johns Hopkins University, Baltimore, MD, USA
- Division of Precision Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Eugene P Rhee
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Dan E Arking
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bing Yu
- Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Eric Boerwinkle
- Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Paul A Welling
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nilanjan Chatterjee
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Katalin Susztak
- Department of Medicine and Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Morgan E Grams
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Division of Precision Medicine, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
2
|
Bensalem A, Ternant D. Pharmacokinetic Variability of Therapeutic Antibodies in Humans: A Comprehensive Review of Population Pharmacokinetic Modeling Publications. Clin Pharmacokinet 2020; 59:857-874. [DOI: 10.1007/s40262-020-00874-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
3
|
O'Donoghue JA, Danila DC, Pandit-Taskar N, Beylergil V, Cheal SM, Fleming SE, Fox JJ, Ruan S, Zanzonico PB, Ragupathi G, Lyashchenko SK, Williams SP, Scher HI, Fine BM, Humm JL, Larson SM, Morris MJ, Carrasquillo JA. Pharmacokinetics and Biodistribution of a [ 89Zr]Zr-DFO-MSTP2109A Anti-STEAP1 Antibody in Metastatic Castration-Resistant Prostate Cancer Patients. Mol Pharm 2019; 16:3083-3090. [PMID: 31117485 DOI: 10.1021/acs.molpharmaceut.9b00326] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A six-transmembrane epithelial antigen of prostate-1 (STEAP1) is a newly identified target in prostate cancer. The use of radio-labeled STEAP1-targeting antibodies with positron emission tomography (PET) may allow for detection of sites of metastatic prostate cancer and may refine patient selection for antigen-directed therapies. This was a prospective study in seven patients with metastatic castration-resistant prostate cancer who had at least one archival biopsy that was STEAP1-positive by immunohistochemistry. Patients received intravenous injections of ∼185 MBq and 10 mg of [89Zr]Zr-DFO-MSTP2109A, a humanized IgG1 monoclonal antibody directed against STEAP1. PET/CT images, blood samples, and whole-body counts were monitored longitudinally in six patients. Here, we report on safety, biodistribution, pharmacokinetics, dose estimates to normal tissues, and initial tumor targeting for this group of patients. There was no significant acute or subacute toxicity. Favorable biodistribution and enhanced lesion uptake (in both bone and soft tissue) were observed on imaging using a mass of 10 mg of DFO-MSTP2109A. The best lesion discrimination was seen at the latest imaging time, a median of 6 days postadministration. Pharmacokinetics showed a median serum T1/2 β of 198 h, volume of central compartment of 3.54 L (similar to plasma volume), and clearance of 19.7 mL/h. The median biologic T1/2 for whole-body retention was 469 h. The highest mean absorbed doses to normal organs (mGy/MBq) were 1.18, 1.11, 0.78, 0.73, and 0.71 for liver, heart wall, lung, kidney, and spleen, respectively. Excellent targeting of metastatic prostate sites in both bone and soft tissue was observed, with an optimal imaging time of 6 days postadministration. The liver and heart were the normal organs that experienced the highest absorbed doses. The pharmacokinetics were similar to other antibodies without major cross-reactivity with normal tissues. A more detailed analysis of lesion targeting in a larger patient population with correlation to immunohistology and standard imaging modalities has been reported.
Collapse
Affiliation(s)
| | - Daniel C Danila
- Department of Medicine , Joan and Sanford I. Weill College of Medicine of Cornell University , New York , New York 10065 , United States
| | - Neeta Pandit-Taskar
- Department of Radiology , Joan and Sanford I. Weill Cornell Medical Center , New York , New York 10065 , United States
| | | | | | | | | | | | | | | | | | - Simon P Williams
- Genentech , South San Francisco , California 94080 , United States
| | - Howard I Scher
- Department of Medicine , Joan and Sanford I. Weill College of Medicine of Cornell University , New York , New York 10065 , United States
| | - Bernard M Fine
- Genentech , South San Francisco , California 94080 , United States
| | | | - Steven M Larson
- Department of Radiology , Joan and Sanford I. Weill Cornell Medical Center , New York , New York 10065 , United States.,Center for Targeted Radioimmunotherapy and Diagnosis of the Ludwig Center for Cancer Immunotherapy , New York , New York 10065 , United States
| | - Michael J Morris
- Department of Medicine , Joan and Sanford I. Weill College of Medicine of Cornell University , New York , New York 10065 , United States
| | - Jorge A Carrasquillo
- Department of Radiology , Joan and Sanford I. Weill Cornell Medical Center , New York , New York 10065 , United States.,Center for Targeted Radioimmunotherapy and Diagnosis of the Ludwig Center for Cancer Immunotherapy , New York , New York 10065 , United States
| |
Collapse
|
4
|
Butt MA, Pye H, Haidry RJ, Oukrif D, Khan SUR, Puccio I, Gandy M, Reinert HW, Bloom E, Rashid M, Yahioglu G, Deonarain MP, Hamoudi R, Rodriguez-Justo M, Novelli MR, Lovat LB. Upregulation of mucin glycoprotein MUC1 in the progression to esophageal adenocarcinoma and therapeutic potential with a targeted photoactive antibody-drug conjugate. Oncotarget 2017; 8:25080-25096. [PMID: 28212575 PMCID: PMC5421911 DOI: 10.18632/oncotarget.15340] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 01/24/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Mucin glycoprotein 1 (MUC1) is a glycosylated transmembrane protein on epithelial cells. We investigate MUC1 as a therapeutic target in Barrett's epithelium (BE) and esophageal adenocarcinoma (EA) and provide proof of concept for a light based therapy targeting MUC1. RESULTS MUC1 was present in 21% and 30% of significantly enriched pathways comparing BE and EA to squamous epithelium respectively. MUC1 gene expression was x2.3 and x2.2 higher in BE (p=<0.001) and EA (p=0.03). MUC1 immunohistochemical expression increased during progression to EA and followed tumor invasion. HuHMFG1 based photosensitive antibody drug conjugates (ADC) showed cell internalization, MUC1 selective and light-dependent cytotoxicity (p=0.0006) and superior toxicity over photosensitizer alone (p=0.0022). METHODS Gene set enrichment analysis (GSEA) evaluated pathways during BE and EA development and quantified MUC1 gene expression. Immunohistochemistry and flow cytometry evaluated the anti-MUC1 antibody HuHMFG1 in esophageal cells of varying pathological grade. Confocal microscopy examined HuHMFG1 internalization and HuHMFG1 ADCs were created to deliver a MUC1 targeted phototoxic payload. CONCLUSIONS MUC1 is a promising target in EA. Molecular and light based targeting of MUC1 with a photosensitive ADC is effective in vitro and after development may enable treatment of locoregional tumors endoscopically.
Collapse
Affiliation(s)
- Mohammed Adil Butt
- Department for Tissue & Energy, University College London, London, UK
- Upper Gastrointestinal Service, University College London Hospitals NHS Foundation Trust, London, UK
| | - Hayley Pye
- Department for Tissue & Energy, University College London, London, UK
| | - Rehan J. Haidry
- Upper Gastrointestinal Service, University College London Hospitals NHS Foundation Trust, London, UK
| | - Dahmane Oukrif
- Department of Pathology, University College London, London, UK
| | | | - Ignazio Puccio
- Department for Tissue & Energy, University College London, London, UK
| | - Michael Gandy
- Department for Tissue & Energy, University College London, London, UK
| | - Halla W. Reinert
- Department for Tissue & Energy, University College London, London, UK
| | - Ellie Bloom
- Department for Tissue & Energy, University College London, London, UK
| | | | - Gokhan Yahioglu
- Antikor BioPharma, Stevenage Bioscience Catalyst, Hertfordshire, UK
- Department of Chemistry, Imperial College London, London, UK
| | - Mahendra P. Deonarain
- Department for Tissue & Energy, University College London, London, UK
- Antikor BioPharma, Stevenage Bioscience Catalyst, Hertfordshire, UK
- Department of Chemistry, Imperial College London, London, UK
| | - Rifat Hamoudi
- Department for Tissue & Energy, University College London, London, UK
| | | | | | - Laurence B. Lovat
- Department for Tissue & Energy, University College London, London, UK
- Upper Gastrointestinal Service, University College London Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
5
|
Pan WJ, Li H, Xiao JJ, Horner MJ, Lebrec HN, Butz EA, Kaliyaperumal A, Cheah TC, Ortiz RC, Prokop SP, Buntich SA, Boren BM, Wolford ST, Tsuji WH, Wienkers LC, Köck K. Modeling the pharmacokinetic-pharmacodynamic relationship of the monoclonal anti-macaque-IL-15 antibody Hu714MuXHu in cynomolgus monkeys. Pharmacol Res Perspect 2016; 3:e00199. [PMID: 27022472 PMCID: PMC4777250 DOI: 10.1002/prp2.199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 09/08/2015] [Indexed: 12/27/2022] Open
Abstract
Hu714MuXHu is a recombinant chimeric murine‐human monoclonal antibody directed against interleukin‐15 (IL‐15), a proinflammatory cytokine associated with memory CD8+ and natural killer (NK) T‐cell activation and implicated in the pathogenesis of inflammatory diseases. A pharmacokinetic‐pharmacodynamic (PK/PD) model was developed to describe the NK cell count reduction in cynomolgus monkeys after treatment with Hu714MuXHu. Cynomolgus monkeys were dosed with Hu714MuXHu in three studies: as a single dose at 0.1 or 1 mg·kg−1 i.v.; weekly for 5 weeks at 0, 30, 60, or 150 mg·kg−1 i.v. or 150 mg·kg−1 s.c.; weekly for 13 weeks at 0, 5, 30, or 150 mg·kg−1 s.c. Serum Hu714MuXHu concentration‐time data were analyzed using noncompartmental analysis and the PK/NK cell count relationship was assessed via simultaneous PK/PD modeling. Hu714MuXHu PK was approximately dose‐proportional between 0.1–150 mg·kg−1 for i.v. and 5–150 mg·kg−1 for s.c. administration with an elimination half‐life of 12.7–18 days. Hu714MuXHu administration resulted in rapid and marked reductions in NK cell counts after the first dose which recovered fully after the serum Hu714MuXHu concentrations approached 0.1 μg·mL−1 (assay limit of quantification). PK/PD modeled Hu714MuXHu effects on NK cells had an EC50 of 0.09 μg·mL−1. In summary, weekly i.v. or s.c. doses with Hu714MuXHu for up to 3 months in cynomolgus monkeys demonstrated linear PK and significant NK cell count reduction, which was described using PK/PD modeling. This approach may be used to guide investigative product dose selections for inflammatory diseases where NK cell count alterations are quantifiable.
Collapse
Affiliation(s)
- Wei J Pan
- Pharmacokinetics and Drug Metabolism Amgen Inc. Seattle Washington
| | - Hong Li
- Pharmacokinetics and Drug Metabolism Amgen Inc. Seattle Washington
| | - Jim J Xiao
- Pharmacokinetics and Drug Metabolism Amgen Inc. Thousand Oaks Washington
| | - Michelle J Horner
- Comparative Biology and Safety Sciences Amgen Inc. Thousand Oaks California
| | - Herve N Lebrec
- Comparative Biology and Safety Sciences Amgen Inc. Seattle Washington
| | - Eric A Butz
- Inflammation Discovery Research Amgen Inc. Seattle Washington
| | | | - Tsui C Cheah
- Pharmacokinetics and Drug Metabolism Amgen Inc. Thousand Oaks Washington
| | - Robert C Ortiz
- Pharmacokinetics and Drug Metabolism Amgen Inc. Thousand Oaks Washington
| | | | - Sabina A Buntich
- Comparative Biology and Safety Sciences Amgen Inc. Thousand Oaks California
| | - Babette M Boren
- Comparative Biology and Safety Sciences Amgen Inc. Seattle Washington
| | | | | | - Larry C Wienkers
- Pharmacokinetics and Drug Metabolism Amgen Inc. Seattle Washington
| | - Kathleen Köck
- Pharmacokinetics and Drug Metabolism Amgen Inc. Seattle Washington
| |
Collapse
|
6
|
Population pharmacokinetics of brodalumab in healthy adults and adults with psoriasis from single and multiple dose studies. J Clin Pharmacol 2014; 54:1230-8. [DOI: 10.1002/jcph.334] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 05/19/2014] [Indexed: 12/29/2022]
|
7
|
Matsushita T, Takada W, Igarashi K, Naruchi K, Miyoshi R, Garcia-Martin F, Amano M, Hinou H, Nishimura SI. A straightforward protocol for the preparation of high performance microarray displaying synthetic MUC1 glycopeptides. Biochim Biophys Acta Gen Subj 2013; 1840:1105-16. [PMID: 24246952 DOI: 10.1016/j.bbagen.2013.11.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 10/07/2013] [Accepted: 11/08/2013] [Indexed: 12/25/2022]
Abstract
BACKGROUND Human serum MUC1 peptide fragments bearing aberrant O-glycans are secreted from columnar epithelial cell surfaces and known as clinically important serum biomarkers for the epithelial carcinoma when a specific monoclonal antibody can probe disease-relevant epitopes. Despite the growing importance of MUC1 glycopeptides as biomarkers, the precise epitopes of most anti-MUC1 monoclonal antibodies remains unclear. METHODS A novel protocol for the fabrication of versatile microarray displaying peptide/glycopeptide library was investigated for the construction of highly sensitive and accurate epitope mapping assay of various anti-MUC1 antibodies. RESULTS Selective imine-coupling between aminooxy-functionalized methacrylic copolymer with phosphorylcholine unit and synthetic MUC1 glycopeptides-capped by a ketone linker at N-terminus provided a facile and seamless protocol for the preparation of glycopeptides microarray platform. It was demonstrated that anti-KL-6 monoclonal antibody shows an extremely specific and strong binding affinity toward MUC1 fragments carrying sialyl T antigen (Neu5Acα2,3Galβ1,3GalNAcα1→) at Pro-Asp-Thr-Arg motif when compared with other seven anti-MUC1 monoclonal antibodies such as VU-3D1, VU-12E1, VU-11E2, Ma552, VU-3C6, SM3, and DF3. The present microarray also uncovered the occurrence of IgG autoantibodies in healthy human sera that bind specifically with sialyl T antigen attached at five potential O-glycosylation sites of MUC1 tandem repeats. CONCLUSION We established a straightforward strategy toward the standardized microarray platform allowing highly sensitive and accurate epitope mapping analysis by reducing the background noise due to nonspecific protein adsorption. GENERAL SIGNIFICANCE The present approach would greatly accelerate the discovery research of new class autoantibodies as well as the development of therapeutic mAbs reacting specifically with disease-relevant epitopes.
Collapse
Affiliation(s)
- Takahiko Matsushita
- Field of Drug Discovery Research, Faculty of Advanced Life Science, Hokkaido University, N22, W11 Kita-ku, Sapporo 001-0021, Japan
| | | | | | - Kentaro Naruchi
- Medicinal Chemistry Pharmaceuticals, Co. Ltd., N22, W12, Kita-ku, Sapporo 001-0021, Japan
| | - Risho Miyoshi
- Medicinal Chemistry Pharmaceuticals, Co. Ltd., N22, W12, Kita-ku, Sapporo 001-0021, Japan
| | - Fayna Garcia-Martin
- Field of Drug Discovery Research, Faculty of Advanced Life Science, Hokkaido University, N22, W11 Kita-ku, Sapporo 001-0021, Japan
| | - Maho Amano
- Field of Drug Discovery Research, Faculty of Advanced Life Science, Hokkaido University, N22, W11 Kita-ku, Sapporo 001-0021, Japan; Medicinal Chemistry Pharmaceuticals, Co. Ltd., N22, W12, Kita-ku, Sapporo 001-0021, Japan
| | - Hiroshi Hinou
- Field of Drug Discovery Research, Faculty of Advanced Life Science, Hokkaido University, N22, W11 Kita-ku, Sapporo 001-0021, Japan; Medicinal Chemistry Pharmaceuticals, Co. Ltd., N22, W12, Kita-ku, Sapporo 001-0021, Japan
| | - Shin-Ichiro Nishimura
- Field of Drug Discovery Research, Faculty of Advanced Life Science, Hokkaido University, N22, W11 Kita-ku, Sapporo 001-0021, Japan; Medicinal Chemistry Pharmaceuticals, Co. Ltd., N22, W12, Kita-ku, Sapporo 001-0021, Japan.
| |
Collapse
|
8
|
Yang J, Shord S, Zhao H, Men Y, Rahman A. Are hepatic impairment studies necessary for therapeutic proteins? Clin Ther 2013; 35:1444-51. [PMID: 23891362 DOI: 10.1016/j.clinthera.2013.06.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 04/30/2013] [Accepted: 06/12/2013] [Indexed: 12/01/2022]
Abstract
OBJECTIVE This study assessed whether trials to investigate the effect of hepatic impairment on the pharmacokinetics of therapeutic proteins (TPs), which are conducted for small molecule drugs, are necessary. METHODS The product labeling for 91 TPs that have been approved by the US Food and Drug Administration were reviewed. A PubMed search was also conducted to identify completed studies that assessed the effect of hepatic impairment on the pharmacokinetics of TPs. Biologic License Applications were subsequently reviewed to gather further descriptions of the trials conducted in patients with hepatic impairment and data analyses. RESULTS No dedicated pharmacokinetics trials were conducted in patients with hepatic impairment for these approved TPs, but subgroup (n = 2 [2.2%]) or population (n = 5 [5.5%]) pharmacokinetic analyses were performed for 7 TPs. The pharmacokinetics of these proteins were not affected by the hepatic dysfunction, with the exception that the clearance of drotrecogin alfa seemed 25% higher in patients with hepatic impairment than in patients without hepatic impairment; however, no dose reduction was recommended. Thus, the effect of hepatic impairment on the pharmacokinetics of TPs is unclear based on the limited analyses completed to date. CONCLUSIONS A dedicated pharmacokinetics trial for TPs in patients with hepatic impairment is not necessary. Recognizing that the data are very limited, it would be important to continue collecting pharmacokinetic data of TP in patients with hepatic impairment and using population pharmacokinetic analyses to evaluate the effect of hepatic impairment on the pharmacokinetics of TP.
Collapse
Affiliation(s)
- Jun Yang
- Office of Clinical Pharmacology, Office of Translational Sciences, US Food and Drug Administration, Silver Spring, Maryland.
| | | | | | | | | |
Collapse
|
9
|
Chae JW, Baek IH, Lee BY, Cho SK, Kwon KI. Population PK/PD analysis of metformin using the signal transduction model. Br J Clin Pharmacol 2013; 74:815-23. [PMID: 22380769 DOI: 10.1111/j.1365-2125.2012.04260.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT Metformin, a biguanide glucose lowering agent, is commonly used to manage type 2 diabetes. The molecular mechanisms of metformin have not been fully identified, but turnover of biomarkers such as glucose and signalling pathways or translocation of glucose transporters are closely related to the glucose-lowering effects of metformin. The PK/PD of metformin have been investigated in healthy humans and patients with type 2 diabetes mellitus and modelling has been performed using an indirect response model. WHAT THIS STUDY ADDS The purpose of this investigation was to develop a population PK/PD model for metformin using a signal transduction model in healthy humans and predict the PK/PD profile in patients with type 2 diabetes. The aim was to compare a previous model (a biophase model) with the signal transduction model, and use a more appropriate model to follow the actions of metformin. Additionally, our developed model was appropriate to predict the time course of plasma metformin and fasting plasma glucose (FPG) concentrations in patients with type 2 diabetes. To our knowledge, this is the first published population PK/PD analysis using the signal transduction model for metformin. AIMS To develop a population pharmacokinetic (PK) and pharmacodynamic (PD) model for metformin (500 mg) using the signal transduction model in healthy humans and to predict the PK/PD profile in patients with type 2 diabetes. METHODS Following the oral administration of 500 mg metformin to healthy humans, plasma concentrations of metformin were measured using LC-MS/MS. A sequential modelling approach using NONMEM VI was used to facilitate data analysis. Monte Carlo simulation was performed to predict the antihyperglycaemic effect in patients with type 2 diabetes. RESULTS Forty-two healthy humans were included in the study. Population mean estimates (relative standard error, RSE) of apparent clearance, apparent volume of distribution and the absorption rate constant were 52.6 l h(-1) (4.18%), 113 l (56.6%) and 0.41 h(-1) , respectively. Covariate analyses revealed that creatinine clearance (CL(CR) ) significantly influenced metformin: CL/F= 52.6 × (CL(cr) /106.5)(0.782) . The signal transduction model was applied to describe the antihyperglycaemic effect of metformin. The population means for efficacy, potency, transit time and the Hill coefficient were estimated to be 19.8 (3.17%), 3.68 µg ml(-1) (3.89%), 0.5 h (2.89%) and 0.547 (9.05%), respectively. The developed model was used to predict the antihyperglycaemic effect in patients with type 2 diabetes. The predicted plasma glucose concentration value was similar to previous values. CONCLUSIONS The population signal transduction model was developed and evaluated for metformin use in healthy volunteers. Model evaluation by non-parametric bootstrap analysis suggested that the proposed model was robust and parameter values were estimated with good precision.
Collapse
Affiliation(s)
- Jung-woo Chae
- College of Pharmacy, Chungnam National University, Daejeon, Korea
| | | | | | | | | |
Collapse
|
10
|
Ibrahim NK, Yariz KO, Bondarenko I, Manikhas A, Semiglazov V, Alyasova A, Komisarenko V, Shparyk Y, Murray JL, Jones D, Senderovich S, Chau A, Erlandsson F, Acton G, Pegram M. Randomized phase II trial of letrozole plus anti-MUC1 antibody AS1402 in hormone receptor-positive locally advanced or metastatic breast cancer. Clin Cancer Res 2011; 17:6822-30. [PMID: 21878535 DOI: 10.1158/1078-0432.ccr-11-1151] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE AS1402 is a humanized immunoglobulin G1 antibody that targets the aberrantly glycosylated antigen MUC1, which is overexpressed in 90% of breast tumors and contributes to estrogen-mediated growth and survival of breast cancer cells in vitro by modulating estrogen receptor (ER) activity. Aromatase inhibitors have been reported to enhance antibody-dependent cell-mediated cytotoxicity elicited by antibodies in vitro. We compared the outcomes of patients with breast cancer treated with letrozole with or without AS1402. EXPERIMENTAL DESIGN The study population included 110 patients with locally advanced or metastatic hormone receptor-positive breast cancer randomized to receive 2.5 mg letrozole only once daily or with a weekly 9 mg/kg AS1402 infusion. The primary endpoint was overall response rate. Secondary endpoints included progression-free survival, time to progression, and safety. AS1402 exposure and influence of allotypes of FcγRIIIa, FcγRIIa, and MUC1 were evaluated. RESULTS The study was stopped early because of a trend toward worse response rates and a higher rate of early disease progression in the AS1402 + letrozole arm. Final analysis revealed no significant difference in efficacy between the study arms. Evaluated gene polymorphisms did not define patient subgroups with improved outcomes. Addition of AS1402 to letrozole was associated with manageable toxicity. CONCLUSIONS Because adding AS1402 to letrozole did not improve outcomes compared with letrozole only, blocking ER may be a better strategy for harnessing MUC1 modulation of the ER to a clinical advantage. FcγRIIIa, FcγRIIa, and MUC1 allotype did not predict outcome for patients treated with letrozole with or without AS1402.
Collapse
Affiliation(s)
- Nuhad K Ibrahim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Optimal sampling strategy development methodology using maximum a posteriori Bayesian estimation. Ther Drug Monit 2011; 33:133-46. [PMID: 21383653 DOI: 10.1097/ftd.0b013e31820f40f8] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Maximum a posteriori Bayesian (MAPB) pharmacokinetic parameter estimation is an accurate and flexible method of estimating individual pharmacokinetic parameters using individual blood concentrations and prior information. In the past decade, many studies have developed optimal sampling strategies to estimate pharmacokinetic parameters as accurately as possible using either multiple regression analysis or MAPB estimation. This has been done for many drugs, especially immunosuppressants and anticancer agents. Methods of development for optimal sampling strategies (OSS) are diverse and heterogeneous. This review provides a comprehensive overview of OSS development methodology using MAPB pharmacokinetic parameter estimation, determines the transferability of published OSSs, and compares sampling strategies determined by MAPB estimation and multiple regression analysis. OSS development has the following components: 1) prior distributions; 2) reference value determination; 3) optimal sampling time identification; and 4) validation of the OSS. Published OSSs often lack all data necessary for the OSS to be clinically transferable. MAPB estimation is similar to multiple regression analysis in terms of predictive performance but superior in flexibility.
Collapse
|
12
|
Roskos LK, Ren S, Robbie G. Application of Modeling and Simulation in the Development of Protein Drugs. CLINICAL TRIAL SIMULATIONS 2011. [DOI: 10.1007/978-1-4419-7415-0_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
13
|
Impact of in vivo alemtuzumab dose before reduced intensity conditioning and HLA-identical sibling stem cell transplantation: pharmacokinetics, GVHD, and immune reconstitution. Blood 2010; 116:3080-8. [PMID: 20587785 DOI: 10.1182/blood-2010-05-286856] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In vivo alemtuzumab reduces the risk of graft-versus-host disease (GVHD) and nonrelapse mortality after reduced intensity allogeneic transplantation. However, it also delays immune reconstitution, leading to frequent infections and potential loss of graft-versus-tumor responses. Here, we tested the feasibility of alemtuzumab dose deescalation in the context of fludarabine-melphalan conditioning and human leukocyte antigen (HLA)-identical sibling transplantation. Alemtuzumab was given 1-2 days before graft infusion, and dose reduced from 60 mg to 20 mg in 4 sequential cohorts (total n = 106). Pharmacokinetic studies were fitted to a linear, 2-compartment model in which dose reduction led to incomplete saturation of CD52 binding sites and greater antibody clearance. Increased elimination was particularly evident in the 20-mg group in patients who had CD52-expressing tumors at time of transplantation. The 20-mg dose was also associated with greater risk of severe GVHD (acute grade III-IV or chronic extensive) compared with > 20 mg (hazard ratio, 6.7; 95% CI, 2.5-18.3). In contrast, dose reduction to 30 mg on day -1 was associated with equivalent clinical outcomes to higher doses but better lymphocyte recovery at 12 months. In conclusion, alemtuzumab dose reduction to 30 mg is safe in the context of reduced intensity conditioning and HLA-identical sibling transplantation. This trial was registered at http://www.ncrn.org.uk as UKCRN study 1415.
Collapse
|