1
|
Gao Y, Xia Y, Chen Y, Zhou S, Fang Y, Yu J, Zhang L, Sun L. Key considerations based on pharmacokinetic/pharmacodynamic in the design of antibody-drug conjugates. Front Oncol 2025; 14:1459368. [PMID: 39850824 PMCID: PMC11754052 DOI: 10.3389/fonc.2024.1459368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 12/09/2024] [Indexed: 01/25/2025] Open
Abstract
Background Antibody-drug conjugate (ADC) is an anticancer drug that links toxins to specifically targeted antibodies via linkers, offering the advantages of high target specificity and high cytotoxicity. However, complexity of its structural composition poses a greater difficulty for drug design studies. Objectives Pharmacokinetic/pharmacodynamic (PK/PD) based consideration of ADCs has increasingly become a hot research topic for optimal drug design in recent years, providing possible ideas for obtaining ADCs with desirable properties. Methods From the assessment of the ADC action process based on PK/PD, we introduce the main research strategies of ADCs. In addition, we investigated the strategies to solve the prominent problems of ADC in the clinic in recent years, and summarized and evaluated the specific ways to optimize various problems of ADC based on the PK/PD model from two perspectives of optimizing the structure and properties of the drugs themselves. Through the selection of target antigen, the optimization of the linker, the optimization of novel small molecule toxins as payload, the optimization of ADC, overcoming the multi-drug resistance of ADC, improving the ADC tumor penetration of ADC, surface modification of ADC and surface bystander effect of ADC provide a more comprehensive and accurate framework for designing new ADCs. Results We've expounded comprehensively on applying pharmacokinetics or pharmacodynamics while designing ADC to obtain higher efficacy and fewer side effects. From the ADC's PK/PD property while coming into play in vivo and the PK/PD study strategy, to specific ADC optimization methods and recommendations based on PK/PD, it has been study-approved that the PK/PD properties exert a subtle role in the development of ADC, whether in preclinical trials or clinical promotion. Conclusion The study of PK/PD unfolds the detailed mechanism of ADC action, making it easier to control related parameters in the process of designing ADC, limited efficacy and inevitable off-target toxicity remain a challenging bottleneck.
Collapse
Affiliation(s)
- Yangyang Gao
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Yuwei Xia
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Yixin Chen
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Shiqi Zhou
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Yingying Fang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Jieru Yu
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Leyin Zhang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
- Department of Oncology, Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Chinese Medicine), Hangzhou, China
| | - Leitao Sun
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
2
|
Khera E, Dharmarajan L, Hainzl D, Engelhardt V, Vostiarova H, Davis J, Ebel N, Wuersch K, Romanet V, Sharaby S, Kearns JD. QSP modeling of a transiently inactivating antibody-drug conjugate highlights benefit of short antibody half life. J Pharmacokinet Pharmacodyn 2024; 52:7. [PMID: 39690276 DOI: 10.1007/s10928-024-09956-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/03/2024] [Indexed: 12/19/2024]
Abstract
Antibody drug conjugates (ADC) are a promising class of oncology therapeutics consisting of an antibody conjugated to a payload via a linker. DYP688 is a novel ADC comprising of a signaling protein inhibitor payload (FR900359) that undergoes unique on-antibody inactivation in plasma, resulting in complex pharmacology. To assess the impact of FR inactivation on DYP688 pharmacology and clinical developability, we performed translational modeling of preclinical PK and tumor growth inhibition (TGI) data, accompanied by mechanistic Krogh cylinder tumor modeling. Using a PK-TGI model, we identified a composite exposure-above-tumorostatic concentration (AUCTSC) metric as the PK-driver of efficacy. To underpin the mechanisms behind AUCTSC as the driver of efficacy, we performed quantitative systems pharmacology (QSP) modeling of DYP688 intratumoral pharmacokinetics and pharmacodynamics. Through exploratory simulations, we show that by deviating from canonical ADC design dogma, DYP688 has optimal FR900359 activity despite its transient inactivation. Finally, we performed the successful preclinical to clinical translation of DYP688 PK, including the payload inactivation kinetics, evidenced by good agreement of the predicted PK to the observed interim clinical PK. Overall, this work highlights early quantitative pharmacokinetics as a missing link in the ADC design-developability chasm.
Collapse
Affiliation(s)
- Eshita Khera
- PK Sciences, Translational Medicine, Novartis Biomedical Research, Cambridge, MA, USA
| | - Lekshmi Dharmarajan
- PK Sciences, Translational Medicine, Novartis Biomedical Research, Basel, Switzerland
| | - Dominik Hainzl
- PK Sciences, Translational Medicine, Novartis Biomedical Research, Cambridge, MA, USA
| | - Volker Engelhardt
- PK Sciences, Translational Medicine, Novartis Biomedical Research, Basel, Switzerland
| | - Helena Vostiarova
- PK Sciences, Translational Medicine, Novartis Biomedical Research, Basel, Switzerland
| | - John Davis
- PK Sciences, Translational Medicine, Novartis Biomedical Research, Cambridge, MA, USA
| | - Nicolas Ebel
- Oncology, Novartis Biomedical Research, Basel, Switzerland
| | - Kuno Wuersch
- Preclinical Safety, Novartis Biomedical Research, Basel, Switzerland
| | | | - Sherif Sharaby
- PK Sciences, Translational Medicine, Novartis Biomedical Research, East Hanover, NJ, USA
| | - Jeffrey D Kearns
- PK Sciences, Translational Medicine, Novartis Biomedical Research, Cambridge, MA, USA.
| |
Collapse
|
3
|
Stern S, Wange RL, Rogers H. An Evaluation of First-in-Human Studies for RNA Oligonucleotides. Nucleic Acid Ther 2024; 34:276-284. [PMID: 39311689 DOI: 10.1089/nat.2024.0036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024] Open
Abstract
Most oligonucleotide therapeutics use Watson-Crick-Franklin base-pairing hybridization to target RNA and mitigate disease-related protein production. Using targets that were previously inaccessible to small molecules and biologics, synthetic nucleotides have provided treatments for severely debilitating and life-threatening diseases. However, these therapeutics possess unique pharmacologies that require specific considerations for their distribution, clearance, and other clinical pharmacology characteristics. Namely, one hurdle in the drug development of these therapeutics remains the prediction of human dose that results in exposures comparable with or below those seen at no observed adverse effect level in animals. For first-in-human (FIH) clinical trials, this often involves allometric scaling based on body surface area (BSA) or body weight (BW). In this study, we reviewed the current literature and surveyed elements across 16 approved oligonucleotide therapeutic New Drug Applications approved by the U.S. Food and Drug Administration in the period from September 1998 to January 2024, and 89 Investigational New Drug (IND) programs with available FIH clinical trials conducted from January 2015 to January 2024, to understand dose selection in early-stage development of oligonucleotide therapeutics. The surveyed elements across these programs include study design, route of administration, dosing regimen, interspecies scaling approach, and the most sensitive species. Of 89 IND programs and 16 approved therapeutics, intravenous and subcutaneous were the most common route of administration, no observable adverse event levels were frequently derived from nonhuman primates, BSA and BW were adjusted for in similar frequencies, patients were predominantly enrolled in FIH trials, and the most common design was a single or multiple ascending dose trial.
Collapse
Affiliation(s)
- Sydney Stern
- Center for Drug Evaluation and Research, Office of Translational Science, Office of Clinical Pharmacology, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ronald L Wange
- Center for Drug Evaluation and Research, Office of the Medical Products and Tobacco (OMPT), Office of New Drug, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Hobart Rogers
- Center for Drug Evaluation and Research, Office of Translational Science, Office of Clinical Pharmacology, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
4
|
Fang P, You M, Cao Y, Feng Q, Shi L, Wang J, Sun X, Yu D, Zhou W, Yin L, Mei F, Zhu X, Cheng A, Tan X. Development and validation of bioanalytical assays for the quantification of 9MW2821, a nectin-4-targeting antibody-drug conjugate. J Pharm Biomed Anal 2024; 248:116318. [PMID: 38908237 DOI: 10.1016/j.jpba.2024.116318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/13/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
We designed and developed 9MW2821, an anti-Nectin-4 antibody-drug conjugate (ADC) with an enzymatically cleavable valine-citrulline linker and monomethyl auristatin E (MMAE) as the payload. Four bioanalytical assays for total antibodies, conjugated antibodies, conjugated payload, and free payload were then developed and validated for the comprehensive evaluation of the multiple drug forms of 9MW2821. Specific sandwich enzyme-linked immunosorbent assays were used to quantify total antibodies and conjugated antibody, showing good drug-to-antibody ratio (DAR) tolerance. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to determine free MMAE, and conjugated MMAE was quantified using a combination of ligand-binding assay (LBA) and LC-MS/MS. Based on these four assays, we studied the serum stability and monkey pharmacokinetic profiles of 9MW2821, and the in vivo DAR of 9MW2821 was calculated and dynamically monitored. In conclusion, we developed and validated series of bioanalytical assays to quantify multiple forms of 9MW2821, a new ADC, and used the assays to evaluate the serum stability and monkey pharmacokinetic characteristics. The results indicate good linker stability and suggest that the developed assays can be further used in clinical settings.
Collapse
Affiliation(s)
- Peng Fang
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Meng You
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Yuxia Cao
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Qingjun Feng
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Lei Shi
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Jin Wang
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Xiaowei Sun
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Dongan Yu
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Wei Zhou
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Long Yin
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Fei Mei
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Xiaohong Zhu
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Aidi Cheng
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China
| | - Xiaoding Tan
- Jiangsu Mabwell Health Pharmaceutical R&D Co. Ltd., Taizhou 225300, China.
| |
Collapse
|
5
|
Ayoun Alsoud R, Le Moan N, Holten-Andersen L, Knudsen T, Lennernäs H, Simonsson USH. Model-Based Interspecies Scaling for Predicting Human Pharmacokinetics of CB 4332, a Complement Factor I Protein. J Pharm Sci 2024; 113:2895-2903. [PMID: 38945365 DOI: 10.1016/j.xphs.2024.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/25/2024] [Accepted: 06/25/2024] [Indexed: 07/02/2024]
Abstract
Interspecies scaling of the pharmacokinetics (PK) of CB 4332, a 150 kDa recombinant complement factor I protein, was performed using traditional and model-based approaches to inform first-in-human dose selection. Plasma concentration versus time data from four preclinical PK studies of single intravenous and subcutaneous (SC) CB 4332 dosing in mice, rats and nonhuman primates (NHPs) were modeled simultaneously using naive pooling including allometric scaling. The human-equivalent dose was calculated using the preclinical no observed adverse effect level (NOAEL) as part of the dose-by-factor approach. Pharmacokinetic modeling of CB 4332 revealed species-specific differences in the elimination, which was accounted for by including an additional rat-specific clearance. Signs of anti-drug antibodies (ADA) formation in all rats and some NHPs were observed. Consequently, an additional ADA-induced clearance parameter was estimated including the time of onset. The traditional dose-by-factor approach calculated a maximum recommended starting SC dose of 0.9 mg/kg once weekly, which was predicted it to result in a trough steady-state concentration lower than the determined efficacy target range for CB 4332 in humans. Model simulations predicted the efficacy target range to be reached using 5 mg/kg once weekly SC dosing.
Collapse
Affiliation(s)
- Rami Ayoun Alsoud
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | | | | - Tom Knudsen
- Catalyst Biosciences, South San Francisco, California, USA
| | - Hans Lennernäs
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
6
|
Liao MZ, Leipold DD, Chen SC, Li Z, Kamath AV, Li C. Translational PK/PD framework for antibody-drug conjugates to inform drug discovery and development. Xenobiotica 2024; 54:543-551. [PMID: 38738473 DOI: 10.1080/00498254.2024.2351044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/30/2024] [Indexed: 05/14/2024]
Abstract
ADCs represent a transformative class of medicine that combines the specificity of monoclonal antibodies with the potency of highly cytotoxic agents through linkers, aiming to enhance the therapeutic index of cytotoxic drugs. Given the complex molecular structures of ADCs, combining the molecular characteristics of small-molecule drugs and those of large-molecule biotherapeutics, there are several unique considerations when designing nonclinical-to-clinical PK/PD translation strategies.This complexity also demands a thorough understanding of the ADC's components - antibody, linker, and payload - to the overall toxicological, PK/PD, and efficacy profile. ADC development is a multidisciplinary endeavour requiring a strategic integration of nonclinical safety, pharmacology, and PK/PD modelling to translate from bench to bedside successfully.The ADC development underscores the necessity for a robust scientific foundation, leveraging advanced analytical and modelling tools to predict human responses and optimise therapeutic outcomes.This review aims to provide an ADC translational PK/PD framework by discussing unique aspects of ADC nonclinical to clinical PK translation, starting dose determination, and leveraging PK/PD modelling for human efficacious dose prediction and potential safety mitigation.
Collapse
Affiliation(s)
| | | | | | - Zao Li
- Genentech Inc, South San Francisco, CA, USA
| | | | - Chunze Li
- Genentech Inc, South San Francisco, CA, USA
| |
Collapse
|
7
|
Filippini DM, Le Tourneau C. The potential roles of antibody-drug conjugates in head and neck squamous cell carcinoma. Curr Opin Oncol 2024; 36:147-154. [PMID: 38573203 DOI: 10.1097/cco.0000000000001022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
PURPOSE OF REVIEW To summarize the actual antibody-drug conjugates (ADCs) tested for patients with advanced head and neck squamous cell carcinoma (HNSCC), outlining the results of safety and efficacy through published clinical trials. RECENT FINDINGS ADCs combine the specificity of mAbs with the cytotoxic drug (known as payload) via a chemical linker and it is designed to selectively deliver the ultratoxic payload directly to the target cancer cells. To date, various ADCs have been investigated in multiple solid malignancies and others are in clinical development. In this study, we provide an overview of the structure and biology of ADC and we review recent clinical experience with the ADC in patients with advanced HNSCC, followed by a brief discussion of the evolvement of ADC conception, drug resistance and future perspectives. SUMMARY ADC strategy is emerging as a potential active treatment in previously treated patients with advanced HNSCC. However, the recent improvement in the bioengineering of ADC and a better comprehension of sequencing and association strategies could provide more benefit to HNSCC patients in need of innovative therapy.
Collapse
Affiliation(s)
- Daria Maria Filippini
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
- INSERM U900 Research Unit, Saint-Cloud
- Paris-Saclay University, Paris, France
| |
Collapse
|
8
|
Valsasina B, Orsini P, Caruso M, Albanese C, Ciavolella A, Cucchi U, Fraietta I, Melillo N, Fiorentini F, Rizzi S, Salsa M, Isacchi A, Gasparri F. Novel Thienoduocarmycin-Trastuzumab ADC Demonstrates Strong Antitumor Efficacy with Favorable Safety Profile in Preclinical Studies. Mol Cancer Ther 2023; 22:1465-1478. [PMID: 37722716 DOI: 10.1158/1535-7163.mct-23-0315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/19/2023] [Accepted: 09/08/2023] [Indexed: 09/20/2023]
Abstract
New antibodies-drug conjugate (ADC) payloads overcoming chemoresistance and killing also poorly proliferating tumors at well-tolerated doses are much desired. Duocarmycins are a well-known class of highly potent cytotoxic agents, with DNA minor groove-binding and alkylation properties, active also in chemoresistant tumors. Although different duocarmycin derivatives have been used during the years as payloads for ADC production, unfavorable physicochemical properties impaired the production of ADCs with optimal features. Optimization of the toxin to balance reactivity and stability features and best linker selection allowed us to develop the novel duocarmycin-like payload-linker NMS-P945 suitable for conjugation to mAbs with reproducible drug-antibody ratio (DAR) >3.5. When conjugated to trastuzumab, it generated an ADC with good internalization properties, ability to induce bystander effect and immunogenic cell death. Moreover, it showed strong target-driven activity in cells and cytotoxic activity superior to trastuzumab deruxtecan tested, in parallel, in cell lines with HER2 expression. High in vivo efficacy with cured mice at well-tolerated doses in HER2-driven models was also observed. A developed pharmacokinetic/pharmacodynamic (PK/PD) model based on efficacy in mice and cynomolgus monkey PK data, predicted tumor regression in patients upon administration of 2 doses of trastuzumab-NMS-P945-ADC at 0.5 mg/kg. Thus, considering the superior physicochemical features for ADC production and preclinical results obtained with the model trastuzumab ADC, including bystander effect, immunogenic cell death and activity in chemoresistant tumors, NMS-P945 represents a highly effective, innovative payload for the creation of novel, next-generation ADCs.
Collapse
Affiliation(s)
| | - Paolo Orsini
- Nerviano Medical Sciences srl, Nerviano, Milan, Italy
| | | | | | | | - Ulisse Cucchi
- Nerviano Medical Sciences srl, Nerviano, Milan, Italy
| | - Ivan Fraietta
- Nerviano Medical Sciences srl, Nerviano, Milan, Italy
| | | | | | - Simona Rizzi
- Nerviano Medical Sciences srl, Nerviano, Milan, Italy
| | - Matteo Salsa
- Nerviano Medical Sciences srl, Nerviano, Milan, Italy
| | | | | |
Collapse
|
9
|
Park S, Lee YW, Oh J, Kim SJ, Lee S, Lee H. Pharmacokinetic evaluation of radiolabeled intraocular anti-CLEC14a antibody in preclinical animal species and application in humans. Clin Transl Sci 2022; 15:2938-2946. [PMID: 36129122 PMCID: PMC9747121 DOI: 10.1111/cts.13412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/30/2022] [Accepted: 08/05/2022] [Indexed: 01/26/2023] Open
Abstract
Anti-angiogenic antibodies are widely used in the treatment of neovascular macular degeneration. Human antibody targeting C-type lectin domain family 14 member A (CLEC14a) is potential therapeutic agents owing to its antiangiogenic activity. In the present study, we aimed to predict the human intraocular pharmacokinetic (PK) properties of an anti-CLEC14a antibody. I-125 labeled aflibercept and anti-CLEC14a antibody were intravitreally injected into mice, rats, and rabbits. Single photon emission computed tomography/computed tomography imaging was performed, and the intraocular radioactivity concentration (%ID/ml) was obtained. The PK parameters in those three animal species were obtained by compartmental analysis. The PK parameters in humans were estimated by allometric scaling of the animal PK parameters with consideration of the hydrodynamic radius of the antibody. The mean half-life values of intraocular I-125-labeled aflibercept in mice, rats, and rabbits were 1.13 days, 1.25 days, and 4.91 days, respectively, by analysis with a one-compartment model. The predicted human half-life of intraocular aflibercept was 5.75 days based on vitreal volume by allometric scaling. The half-life values of intraocular I-125-labeled anti-CLEC14a in mice, rats and rabbits were 1.05 days, 1.84 days, and 6.37 days, respectively, by analysis with a one-compartment model. The predicted human half-life of intraocular anti-CLEC14a was 10.29 days based on vitreal volume. According to the hydrodynamic volume of the anti-CLEC14a, the predicted human half-life of intraocular anti-CLEC14a was 9.81 days. The PK characteristics of the intraocular anti-CLEC14a antibody were evaluated noninvasively in animals using I-125 labeling, and the intraocular PK characteristics in humans were predicted using these animal data. This methodology can be applied for the development of new antiangiogenic antibodies to treat macular degeneration.
Collapse
Affiliation(s)
- Sohyun Park
- Department of Nuclear MedicineNational Cancer CenterGoyang‐siGyeonggi‐doKorea,Division of Convergence TechnologyNational Cancer CenterGoyang‐siGyeonggi‐doKorea
| | - Youn Woo Lee
- Department of Nuclear MedicineSeoul National University Bundang HospitalSeongnam‐siGyeonggi‐doKorea
| | - Jaeseong Oh
- Department of Clinical Pharmacology and TherapeuticsSeoul National University College of Medicine and HospitalSeoulKorea
| | - Su Jin Kim
- Department of Nuclear MedicineSeoul National University Bundang HospitalSeongnam‐siGyeonggi‐doKorea
| | - Sukmook Lee
- Department of Biopharmaceutical ChemistryKookmin UniversitySeoulKorea
| | - Ho‐Young Lee
- Department of Nuclear MedicineSeoul National University Bundang HospitalSeongnam‐siGyeonggi‐doKorea,Office of eHealth Research and BusinessSeoul National University Bundang HospitalSeongnam‐siGyeonggi‐doKorea
| |
Collapse
|
10
|
Mahmood I. A Simple Method for the Prediction of Human Concentration-Time Profiles and Pharmacokinetics of Antibody-Drug Conjugates (ADC) from Rats or Monkeys. Antibodies (Basel) 2022; 11:antib11020042. [PMID: 35735361 PMCID: PMC9219807 DOI: 10.3390/antib11020042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/23/2022] [Accepted: 06/09/2022] [Indexed: 02/01/2023] Open
Abstract
Knowledge of human concentration-time profiles from animal data can be useful during early drug development. The objective of this study is to predict human concentration-time profiles of antibody-drug conjugates (ADCs) and subsequently predict pharmacokinetic parameters in humans from rats or monkeys. Eight methods with different exponents of volume of distribution (0.8-1) as well as exponents of clearance (0.85), along with the exponents of volume of distribution for 5 ADCs, were used to predict human concentration-time profiles. The PK parameters were also scaled to humans from monkeys or rats using fixed exponents and compared with the PK parameters predicted from predicted human concentration-time profiles. The results of the study indicated that the exponent 0.9 and the combination of exponents of 0.9 and 0.8 (two exponents, 0.8 and 0.9, were used) were the best method to predict human concentration-time profiles and, subsequently, human PK parameters. The predicted PK parameters from fixed exponents were comparable with the predicted PK parameters estimated from human concentration-time profiles. The proposed methods are applicable to rats or monkeys with the same degree of accuracy. Overall, the proposed methods are robust, accurate, and cost- and time-effective.
Collapse
Affiliation(s)
- Iftekhar Mahmood
- Mahmood Clinical Pharmacology Consultancy, LLC., Rockville, MD 20850, USA
| |
Collapse
|
11
|
Haraya K, Tsutsui H, Komori Y, Tachibana T. Recent Advances in Translational Pharmacokinetics and Pharmacodynamics Prediction of Therapeutic Antibodies Using Modeling and Simulation. Pharmaceuticals (Basel) 2022; 15:ph15050508. [PMID: 35631335 PMCID: PMC9145563 DOI: 10.3390/ph15050508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 02/05/2023] Open
Abstract
Therapeutic monoclonal antibodies (mAbs) have been a promising therapeutic approach for several diseases and a wide variety of mAbs are being evaluated in clinical trials. To accelerate clinical development and improve the probability of success, pharmacokinetics and pharmacodynamics (PKPD) in humans must be predicted before clinical trials can begin. Traditionally, empirical-approach-based PKPD prediction has been applied for a long time. Recently, modeling and simulation (M&S) methods have also become valuable for quantitatively predicting PKPD in humans. Although several models (e.g., the compartment model, Michaelis–Menten model, target-mediated drug disposition model, and physiologically based pharmacokinetic model) have been established and used to predict the PKPD of mAbs in humans, more complex mechanistic models, such as the quantitative systemics pharmacology model, have been recently developed. This review summarizes the recent advances and future direction of M&S-based approaches to the quantitative prediction of human PKPD for mAbs.
Collapse
Affiliation(s)
- Kenta Haraya
- Discovery Biologics Department, Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba 412-8513, Japan;
- Correspondence:
| | - Haruka Tsutsui
- Discovery Biologics Department, Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba 412-8513, Japan;
| | - Yasunori Komori
- Pharmaceutical Science Department, Translational Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba 412-8513, Japan; (Y.K.); (T.T.)
| | - Tatsuhiko Tachibana
- Pharmaceutical Science Department, Translational Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba 412-8513, Japan; (Y.K.); (T.T.)
| |
Collapse
|
12
|
Rodriguez-Fernandez K, Gras-Colomer E, Climente-Martí M, Mangas-Sanjuán V, Merino-Sanjuan M. Pharmacometric characterization of entero-hepatic circulation processes of orally administered formulations of amiodarone under complex binding kinetics. Eur J Pharm Sci 2022; 174:106198. [DOI: 10.1016/j.ejps.2022.106198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/19/2022] [Accepted: 04/28/2022] [Indexed: 11/03/2022]
|
13
|
Ball K, Bruin G, Escandon E, Funk C, Pereira JN, Yang TY, Yu H. Characterizing the pharmacokinetics and biodistribution of therapeutic proteins: an industry white paper. Drug Metab Dispos 2022; 50:858-866. [PMID: 35149542 DOI: 10.1124/dmd.121.000463] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 01/06/2022] [Indexed: 11/22/2022] Open
Abstract
Characterization of the pharmacokinetics (PK) 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 (IQ consortium) members on this theme. A wide variety of in vitro, in vivo and in silico techniques are currently used to assess PK and biodistribution of TPs, and we discuss the relevance of these from an industry perspective, focusing on PK/PD 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 at 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 & Quality Consortium (IQ) Translational and ADME Sciences Leadership Group (TALG) working group for the ADME of therapeutic proteins evaluates the current practices, recent advances, and challenges in characterizing the PK 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 PK/PD relationships, and aid translational modelling for first-in-human dose predictions.
Collapse
Affiliation(s)
| | - Gerard Bruin
- Novartis Institutes for Biomedical Research, Switzerland
| | | | - Christoph Funk
- Dept. of Drug Metabolism and Pharmacokinetics, F. Hoffmann-La Roche Ltd., Switzerland
| | | | | | - Hongbin Yu
- Boehringer Ingelheim Pharmaceuticals, Inc, United States
| |
Collapse
|
14
|
Clinical Pharmacology of Antibody-Drug Conjugates. Antibodies (Basel) 2021; 10:antib10020020. [PMID: 34063812 PMCID: PMC8161445 DOI: 10.3390/antib10020020] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/10/2021] [Accepted: 05/14/2021] [Indexed: 12/30/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are biopharmaceutical products where a monoclonal antibody is linked to a biologically active drug (a small molecule) forming a conjugate. Since the approval of first ADC (Gemtuzumab ozogamicin (trade name: Mylotarg)) for the treatment of CD33-positive acute myelogenous leukemia, several ADCs have been developed for the treatment of cancer. The goal of an ADC as a cancer agent is to release the cytotoxic drug to kill the tumor cells without harming the normal or healthy cells. With time, it is being realized that ADCS can also be used to manage or cure other diseases such as inflammatory diseases, atherosclerosis, and bacteremia and some research in this direction is ongoing. The focus of this review is on the clinical pharmacology aspects of ADC development. From the selection of an appropriate antibody to the finished product, the entire process of the development of an ADC is a difficult and challenging task. Clinical pharmacology is one of the most important tools of drug development since this tool helps in finding the optimum dose of a product, thus preserving the safety and efficacy of the product in a patient population. Unlike other small or large molecules where only one moiety and/or metabolite(s) is generally measured for the pharmacokinetic profiling, there are several moieties that need to be measured for characterizing the PK profiles of an ADC. Therefore, knowledge and understanding of clinical pharmacology of ADCs is vital for the selection of a safe and efficacious dose in a patient population.
Collapse
|
15
|
Nanavati C, McMullen G, Yu R, Geary RS, Henry SP, Wang Y. Interspecies Scaling of Human Clearance and Plasma Trough Exposure for Antisense Oligonucleotides: A Retrospective Analysis of GalNAc3-Conjugated and Unconjugated-Antisense Oligonucleotides. Nucleic Acid Ther 2021; 31:298-308. [PMID: 33891483 DOI: 10.1089/nat.2020.0911] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
It is well documented and generally accepted that human clearance (CL) of unconjugated single-strand antisense oligonucleotides (ASOs) can be directly predicted from monkeys by body weight (BW) on a mg/kg dose basis. However, the scaling for triantennary N-acetyl galactosamine (GalNAc3)-conjugated ASOs has not been fully established. In this study, we retrospectively analyzed pharmacokinetic data from 9 GalNAc3-conjugated and 12 unconjugated single-stranded ASOs (ten 2'-methoxyethyl and two 2', 4'-constrained ethyl ASOs) to identify an appropriate allometric scaling factor between the two species. In addition, we compared the trough plasma concentrations (Ctrough, a surrogate for tissue exposure) between monkeys and humans at comparable dose levels, aiming at predicting tissue distribution in humans from monkeys. Overall, the median plasma CL ratios (monkey CL/human CL) were 1.05 and 0.94 when CL was normalized by BW, as compared with 0.33 and 0.29 when CL was normalized by body surface area (BSA) for the 12 unconjugated and 9 GalNAc3-conjugated ASOs, respectively. Similarly, the median Ctrough ratios (Ctrough in monkeys/Ctrough in humans) were 0.96 and 1.71, respectively, when Ctrough was normalized by mg/kg dose as compared with 3.10 and 5.50 when Ctrough was normalized by mg/m2 dose for the same unconjugated and conjugated ASOs, respectively. Equivalent CL and dose-normalized plasma Ctrough between monkeys and humans suggest similar pharmacokinetic profiles and tissue distribution between the two species on a per kilogram BW basis. In conclusion, human CL and plasma Ctrough (a surrogate of tissue distribution) can be directly predicted (1:1 or within twofold) from monkeys by BW on a mg/kg dose basis but these parameters can be under- or over-predicted by BSA on a mg/m2 dose basis. These results provide evidence for single species scaling from monkeys to humans directly and, thus, they can facilitate early human dose prediction in ASO drug development.
Collapse
Affiliation(s)
| | - Gina McMullen
- Ionis Pharmaceuticals, Inc., Carlsbad, California, USA
| | - Rosie Yu
- Ionis Pharmaceuticals, Inc., Carlsbad, California, USA
| | | | - Scott P Henry
- Ionis Pharmaceuticals, Inc., Carlsbad, California, USA
| | - Yanfeng Wang
- Ionis Pharmaceuticals, Inc., Carlsbad, California, USA
| |
Collapse
|
16
|
Clinical pharmacology strategies in supporting drug development and approval of antibody-drug conjugates in oncology. Cancer Chemother Pharmacol 2021; 87:743-765. [PMID: 33792763 PMCID: PMC8110483 DOI: 10.1007/s00280-021-04250-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 02/18/2021] [Indexed: 11/12/2022]
Abstract
Antibody–drug conjugates (ADCs) are important molecular entities in the treatment of cancer. These conjugates combine the target specificity of monoclonal antibodies with the potent anti-cancer activity of small-molecule therapeutics. The complex structure of ADCs poses unique challenges to characterize the drug’s pharmacokinetics (PKs) and pharmacodynamics (PDs) since it requires a quantitative understanding of the PK and PD properties of multiple different molecular species (e.g., ADC conjugate, total antibody and unconjugated cytotoxic drug). As a result, clinical pharmacology strategy of an ADC is rather unique and dependent on the linker/cytotoxic drug technology, heterogeneity of the ADC, PK and safety/efficacy profile of the specific ADC in clinical development. In this review, we summarize the clinical pharmacology strategies in supporting development and approval of ADCs using the approved ADCs as specific examples to illustrate the customized approach to clinical pharmacology assessments in their clinical development.
Collapse
|
17
|
Mahmood I. Interspecies Scaling of Antibody-Drug Conjugates (ADC) for the Prediction of Human Clearance. Antibodies (Basel) 2021; 10:antib10010001. [PMID: 33430196 PMCID: PMC7839014 DOI: 10.3390/antib10010001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/18/2020] [Accepted: 01/05/2021] [Indexed: 12/15/2022] Open
Abstract
Allometric scaling is a useful tool for the extrapolation of pharmacokinetic parameters from animals to humans. The objective of this study was to predict human clearance of antibody-drug conjugates (ADC) allometrically from one to three animal species and compare the predicted human clearance with the observed human clearance. For three animal species allometric scaling, the "Rule of Exponents" (ROE) was used. The results of the study indicated that three-species allometric scaling in association with the ROE provides acceptable prediction (within 0.5-2-fold prediction error) of human clearance. The two-species allometric scaling resulted in substantial prediction error. One-species scaling using a fixed exponent of 1.0 provided acceptable prediction error (within 0.5-2-fold) by monkey, rat, and mouse, in which monkey and rat were comparable. Overall, the predicted human clearance values of ADCs from animal(s) was good. The allometric method proposed in this article can be used to predict human clearance from the animal data and subsequently to select the first-in-human dose of ADCs.
Collapse
Affiliation(s)
- Iftekhar Mahmood
- Mahmood Clinical Pharmacology Consultancy, LLC, Rockville, MD 20850, USA
| |
Collapse
|
18
|
Germovsek E, Cheng M, Giragossian C. Allometric scaling of therapeutic monoclonal antibodies in preclinical and clinical settings. MAbs 2021; 13:1964935. [PMID: 34530672 PMCID: PMC8463036 DOI: 10.1080/19420862.2021.1964935] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/19/2021] [Accepted: 08/03/2021] [Indexed: 02/06/2023] Open
Abstract
Constant technological advancement enabled the production of therapeutic monoclonal antibodies (mAbs) and will continue to contribute to their rapid expansion. Compared to small-molecule drugs, mAbs have favorable characteristics, but also more complex pharmacokinetics (PK), e.g., target-mediated nonlinear elimination and recycling by neonatal Fc-receptor. This review briefly discusses mAb biology, similarities and differences in PK processes across species and within human, and provides a detailed overview of allometric scaling approaches for translating mAb PK from preclinical species to human and extrapolating from adults to children. The approaches described here will remain vital in mAb drug development, although more data are needed, for example, from very young patients and mAbs with nonlinear PK, to allow for more confident conclusions and contribute to further growth of this field. Improving mAb PK predictions will facilitate better planning of (pediatric) clinical studies and enable progression toward the ultimate goal of expediting drug development.
Collapse
Affiliation(s)
- Eva Germovsek
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Ingelheim, Germany
| | - Ming Cheng
- Development Biologicals, Drug Metabolism And Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, US
| | - Craig Giragossian
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, US
| |
Collapse
|
19
|
Kim GS, Lee JH, Shin DY, Lee HS, Park H, Lee KW, Yang HM, Kim SJ, Park JB. Integrated whole liver histologic analysis of the allogeneic islet distribution and characteristics in a nonhuman primate model. Sci Rep 2020; 10:793. [PMID: 31964980 PMCID: PMC6972963 DOI: 10.1038/s41598-020-57701-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 01/06/2020] [Indexed: 12/21/2022] Open
Abstract
The most obvious method to observe transplanted islets in the liver is direct biopsy, but the distribution and location of the best biopsy site in the recipient's liver are poorly understood. Islets transplanted into the whole liver of five diabetic cynomolgus monkeys that underwent insulin-independent survival for an extended period of time after allo-islet transplantation were analyzed for characteristics and distribution tendency. The liver was divided into segments (S1-S8), and immunohistochemistry analysis was performed to estimate the diameter, beta cell area, and islet location. Islets were more distributed in S2 depending on tissue size; however, the number of islets per tissue size was high in S1 and S8. Statistical analysis revealed that the characteristics of islets in S1 and S8 were relatively similar to other segments despite various transplanted islet dosages and survival times. In conclusion, S1, which exhibited high islet density and reflected the overall characteristics of transplanted islets, can be considered to be a reasonable candidate for a liver biopsy site in this monkey model. The findings obtained from the five monkey livers with similar anatomical features to human liver can be used as a reference for monitoring transplanted islets after clinical islet transplantation.
Collapse
Affiliation(s)
- Geun Soo Kim
- Samsung Advanced Institute for Health Sciences & Technology, Graduate School, Department of Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea.,Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, Seoul, Republic of Korea.,Transplantation Research Center, Samsung Medical Center, Seoul, Republic of Korea
| | | | - Du Yeon Shin
- Samsung Advanced Institute for Health Sciences & Technology, Graduate School, Department of Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea.,Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, Seoul, Republic of Korea.,Transplantation Research Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Han Sin Lee
- Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, Seoul, Republic of Korea.,Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyojun Park
- Department of Medicine, Sungkyunkwan University School of Medicine, Gyeonggi, Republic of Korea.,GenNBio Inc, Seoul, Republic of Korea
| | - Kyo Won Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Department of Medicine, Sungkyunkwan University School of Medicine, Gyeonggi, Republic of Korea
| | - Heung-Mo Yang
- Department of Medicine, Sungkyunkwan University School of Medicine, Gyeonggi, Republic of Korea.,GenNBio Inc, Seoul, Republic of Korea
| | - Sung Joo Kim
- Department of Medicine, Sungkyunkwan University School of Medicine, Gyeonggi, Republic of Korea.,GenNBio Inc, Seoul, Republic of Korea
| | - Jae Berm Park
- Samsung Advanced Institute for Health Sciences & Technology, Graduate School, Department of Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea. .,Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, Seoul, Republic of Korea. .,Transplantation Research Center, Samsung Medical Center, Seoul, Republic of Korea. .,Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea. .,Department of Medicine, Sungkyunkwan University School of Medicine, Gyeonggi, Republic of Korea.
| |
Collapse
|
20
|
Deng R, Zhou C, Li D, Cai H, Sukumaran S, Carrasco-Triguero M, Saad O, Nazzal D, Lowe C, Ramanujan S, Kamath AV. Preclinical and translational pharmacokinetics of a novel THIOMAB™ antibody-antibiotic conjugate against Staphylococcus aureus. MAbs 2019; 11:1162-1174. [PMID: 31219754 DOI: 10.1080/19420862.2019.1627152] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
DSTA4637S, a novel THIOMAB™ antibody-antibiotic conjugate (TAC) against Staphylococcus aureus (S. aureus), is currently being investigated as a potential therapy for complicated S. aureus bloodstream infections. DSTA4637S is composed of a monoclonal THIOMABTM IgG1 recognizing S. aureus linked to a rifamycin-class antibiotic (dmDNA31) via a protease-cleavable linker. The pharmacokinetics (PK) of DSTA4637A (a liquid formulation of DSTA4637S) and its unconjugated antibody MSTA3852A were characterized in rats and monkeys. Systemic concentrations of three analytes, total antibody (TAb), antibody-conjugated dmDNA31 (ac-dmDNA31), and unconjugated dmDNA31, were measured to describe complex TAC PK in nonclinical studies. In rats and monkeys, following intravenous administration of a single dose of DSTA4637A, systemic concentration-time profiles of both TAb and ac-dmDNA31 were bi-exponential, characterized by a short distribution phase and a long elimination phase as expected for a monoclonal antibody-based therapeutic. Systemic exposures of both TAb and ac-dmDNA31 were dose proportional over the dose range tested, and ac-dmDNA31 cleared 2-3 times faster than TAb. Unconjugated dmDNA31 plasma concentrations were low (<4 ng/mL) in every study regardless of dose. In this report, an integrated semi-mechanistic PK model for two analytes (TAb and ac-dmDNA31) was successfully developed and was able to well describe the complicated DSTA4637A PK in mice, rats and monkeys. DSTA4637S human PK was predicted reasonably well using this model with allometric scaling of PK parameters from monkey data. This work provides insights into PK behaviors of DSTA4637A in preclinical species and informs clinical translatability of these observed results and further clinical development. Abbreviations: ADC: Antibody-drug conjugate; AUCinf: time curve extrapolated to infinity; ac-dmDNA31: antibody-conjugated dmDNA31; Cmax: maximum concentration observed; DAR: drug-to-antibody ratio; CL: clearance; CLD: distribution clearance; CL1: systemic clearance of all DAR species; kDC: deconjugation rate constant; PK: Pharmacokinetics; IV: Intravenous; IgG: Immunoglobulin G; mAb: monoclonal antibody; S. aureus: Staphylococcus aureus; TAC: THIOMABTM antibody-antibiotic conjugate; TDC: THIOMABTM antibody-drug conjugate; TAb: total antibody; t1/2, λz: terminal half-life; vc linker: valine-citrulline linker; Vss: volume of distribution at steady state; Vc: volume of distribution for the central compartment; Vp: the volume of distribution for the peripheral compartment.
Collapse
Affiliation(s)
- Rong Deng
- a Research and Early Development, Genentech Inc ., South San Francisco , CA , USA
| | - Chenguang Zhou
- a Research and Early Development, Genentech Inc ., South San Francisco , CA , USA
| | - Dongwei Li
- a Research and Early Development, Genentech Inc ., South San Francisco , CA , USA
| | - Hao Cai
- a Research and Early Development, Genentech Inc ., South San Francisco , CA , USA
| | - Siddharth Sukumaran
- a Research and Early Development, Genentech Inc ., South San Francisco , CA , USA
| | | | - Ola Saad
- a Research and Early Development, Genentech Inc ., South San Francisco , CA , USA
| | - Denise Nazzal
- a Research and Early Development, Genentech Inc ., South San Francisco , CA , USA
| | - Christopher Lowe
- a Research and Early Development, Genentech Inc ., South San Francisco , CA , USA
| | - Saroja Ramanujan
- a Research and Early Development, Genentech Inc ., South San Francisco , CA , USA
| | - Amrita V Kamath
- a Research and Early Development, Genentech Inc ., South San Francisco , CA , USA
| |
Collapse
|