1
|
Bohrmann L, Poulie CBM, Rodríguez-Rodríguez C, Karagiozov S, Saatchi K, Herth MM, Häfeli UO. Development of a 99mTc-labeled tetrazine for pretargeted SPECT imaging using an alendronic acid-based bone targeting model. PLoS One 2024; 19:e0300466. [PMID: 38626058 PMCID: PMC11020896 DOI: 10.1371/journal.pone.0300466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 02/27/2024] [Indexed: 04/18/2024] Open
Abstract
Pretargeting, which is the separation of target accumulation and the administration of a secondary imaging agent into two sequential steps, offers the potential to improve image contrast and reduce radiation burden for nuclear imaging. In recent years, the tetrazine ligation has emerged as a promising approach to facilitate covalent pretargeted imaging due to its unprecedented kinetics and bioorthogonality. Pretargeted bone imaging with TCO-modified alendronic acid (Aln-TCO) is an attractive model that allows the evaluation of tetrazines in healthy animals without the need for complex disease models or targeting regimens. Recent structure-activity relationship studies of tetrazines evaluated important parameters for the design of potent tetrazine-radiotracers for pretargeted imaging. However, limited information is available for 99mTc-labeled tetrazines. In this study, four tetrazines intended for labeling with fac-[99mTc(OH2)3 (CO)3]+ were synthesized and evaluated using an Aln-TCO mouse model. 3,6-bis(2-pyridyl)-1,2,4,5-Tz without additional linker showed higher pretargeted bone uptake and less background activity compared to the same scaffold with a PEG8 linker or 3-phenyl-1,2,4,5-Tz-based compounds. Additionally, improved bone/blood ratios were observed in pretargeted animals compared to animals receiving directly labeled Aln-TCO. The results of this study implicate 3,6-bis(2-pyridyl)-1,2,4,5-Tz as a promising scaffold for potential 99mTc-labeled tetrazines.
Collapse
Affiliation(s)
- Lennart Bohrmann
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken, Copenhagen, Denmark
| | - Christian B. M. Poulie
- Department of Drug Design and Pharmacology, Faculty of Health and Medicinal Sciences, University of Copenhagen, Universitetsparken, Copenhagen, Denmark
| | | | - Stoyan Karagiozov
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Katayoun Saatchi
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Matthias M. Herth
- Department of Drug Design and Pharmacology, Faculty of Health and Medicinal Sciences, University of Copenhagen, Universitetsparken, Copenhagen, Denmark
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej, Copenhagen, Denmark
| | - Urs O. Häfeli
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken, Copenhagen, Denmark
| |
Collapse
|
2
|
Photoaffinity labeling and bioorthogonal ligation: Two critical tools for designing "Fish Hooks" to scout for target proteins. Bioorg Med Chem 2022; 62:116721. [PMID: 35358862 DOI: 10.1016/j.bmc.2022.116721] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/14/2022] [Accepted: 03/17/2022] [Indexed: 11/21/2022]
Abstract
Small molecules remain an important category of therapeutic agents. Their binding to different proteins can lead to both desired and undesired biological effects. Identification of the proteins that a drug binds to has become an important step in drug development because it can lead to safer and more effective drugs. Parent bioactive molecules can be converted to appropriate probes that allow for visualization and identification of their target proteins. Typically, these probes are designed and synthesized utilizing some or all of five major tools; a photoactivatable group, a reporter tag, a linker, an affinity tag, and a bioorthogonal handle. This review covers two of the most challenging tools, photoactivation and bioorthogonal ligation. We provide a historical and theoretical background along with synthetic routes to prepare them. In addition, the review provides comparative analyses of the available tools that can assist decision making when designing such probes. A survey of most recent literature reports is included as well to identify recent trends in the field.
Collapse
|
3
|
Ye Z, Gastfriend BD, Umlauf BJ, Lynn DM, Shusta EV. Antibody-Targeted Liposomes for Enhanced Targeting of the Blood-Brain Barrier. Pharm Res 2022; 39:1523-1534. [PMID: 35169958 DOI: 10.1007/s11095-022-03186-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 02/01/2022] [Indexed: 11/28/2022]
Abstract
The blood-brain barrier (BBB) hinders therapeutic delivery to the central nervous system (CNS), thereby impeding the development of therapies for brain injury and disease. Receptor-mediated transcytosis (RMT) systems are a promising way to shuttle a targeted therapeutic into the brain. Here, we developed and evaluated an RMT antibody-targeted liposomal system. A previously identified antibody, scFv46.1, that binds to the human and murine BBB and can pass through the murine BBB by transcytosis after intravenous injection was used to decorate the surface of liposomes. Using an in vitro BBB model, we demonstrated the cellular uptake of scFv46.1-modified liposomes (46.1-Lipo). Next, the biodistribution and brain uptake capacity of 46.1-targeted liposomes were assessed after intravenous administration. Our results showed that 46.1-Lipo can lead to increased brain accumulation through targeting of the brain vasculature. Initial rate pharmacokinetic experiments and biodistribution analyses indicated that 46.1-Lipo loaded with pralidoxime exhibited a 10-fold increase in brain accumulation compared with a mock-targeted liposomal group, and this increased accumulation was brain-specific. These studies indicate the potential of this 46.1-Lipo system as a synthetic vehicle for the targeted transport of therapeutic molecules into the CNS.
Collapse
Affiliation(s)
- Zhou Ye
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA
| | - Benjamin D Gastfriend
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA
| | - Benjamin J Umlauf
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA.,Department of Neurosurgery, Dell Medical School and the Mulva Clinic for the Neurosciences, The University of Texas at Austin, Austin, TX, USA
| | - David M Lynn
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA.,Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA. .,Department of Neurological Surgery, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA.
| |
Collapse
|
4
|
Jemas A, Xie Y, Pigga JE, Caplan JL, am Ende CW, Fox JM. Catalytic Activation of Bioorthogonal Chemistry with Light (CABL) Enables Rapid, Spatiotemporally Controlled Labeling and No-Wash, Subcellular 3D-Patterning in Live Cells Using Long Wavelength Light. J Am Chem Soc 2022; 144:1647-1662. [PMID: 35072462 PMCID: PMC9364228 DOI: 10.1021/jacs.1c10390] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Described is the spatiotemporally controlled labeling and patterning of biomolecules in live cells through the catalytic activation of bioorthogonal chemistry with light, referred to as "CABL". Here, an unreactive dihydrotetrazine (DHTz) is photocatalytically oxidized in the intracellular environment by ambient O2 to produce a tetrazine that immediately reacts with a trans-cyclooctene (TCO) dienophile. 6-(2-Pyridyl)dihydrotetrazine-3-carboxamides were developed as stable, cell permeable DHTz reagents that upon oxidation produce the most reactive tetrazines ever used in live cells with Diels-Alder kinetics exceeding k2 of 106 M-1 s-1. CABL photocatalysts are based on fluorescein or silarhodamine dyes with activation at 470 or 660 nm. Strategies for limiting extracellular production of singlet oxygen are described that increase the cytocompatibility of photocatalysis. The HaloTag self-labeling platform was used to introduce DHTz tags to proteins localized in the nucleus, mitochondria, actin, or cytoplasm, and high-yielding subcellular activation and labeling with a TCO-fluorophore were demonstrated. CABL is light-dose dependent, and two-photon excitation promotes CABL at the suborganelle level to selectively pattern live cells under no-wash conditions. CABL was also applied to spatially resolved live-cell labeling of an endogenous protein target by using TIRF microscopy to selectively activate intracellular monoacylglycerol lipase tagged with DHTz-labeled small molecule covalent inhibitor. Beyond spatiotemporally controlled labeling, CABL also improves the efficiency of "ordinary" tetrazine ligations by rescuing the reactivity of commonly used 3-aryl-6-methyltetrazine reporters that become partially reduced to DHTzs inside cells. The spatiotemporal control and fast rates of photoactivation and labeling of CABL should enable a range of biomolecular labeling applications in living systems.
Collapse
Affiliation(s)
- Andrew Jemas
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, USA
| | - Yixin Xie
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, USA
| | - Jessica E. Pigga
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, USA
| | - Jeffrey L. Caplan
- Department of Plant and Soil Sciences and Delaware Biotechnology Institute, University of Delaware, Newark, DE 19716, USA
| | - Christopher W. am Ende
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Joseph M. Fox
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, USA
| |
Collapse
|
5
|
Spears RJ, McMahon C, Chudasama V. Cysteine protecting groups: applications in peptide and protein science. Chem Soc Rev 2021; 50:11098-11155. [PMID: 34605832 DOI: 10.1039/d1cs00271f] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Protecting group chemistry for the cysteine thiol group has enabled a vast array of peptide and protein chemistry over the last several decades. Increasingly sophisticated strategies for the protection, and subsequent deprotection, of cysteine have been developed, facilitating synthesis of complex disulfide-rich peptides, semisynthesis of proteins, and peptide/protein labelling in vitro and in vivo. In this review, we analyse and discuss the 60+ individual protecting groups reported for cysteine, highlighting their applications in peptide synthesis and protein science.
Collapse
Affiliation(s)
| | - Clíona McMahon
- Department of Chemistry, University College London, London, UK.
| | - Vijay Chudasama
- Department of Chemistry, University College London, London, UK.
| |
Collapse
|
6
|
Seki H, Walsh SJ, Bargh JD, Parker JS, Carroll J, Spring DR. Rapid and robust cysteine bioconjugation with vinylheteroarenes. Chem Sci 2021; 12:9060-9068. [PMID: 34276935 PMCID: PMC8261766 DOI: 10.1039/d1sc02722k] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 05/28/2021] [Indexed: 12/14/2022] Open
Abstract
Methods for residue-selective and stable modification of canonical amino acids enable the installation of distinct functionality which can aid in the interrogation of biological processes or the generation of new therapeutic modalities. Herein, we report an extensive investigation of reactivity and stability profiles for a series of vinylheteroarene motifs. Studies on small molecule and protein substrates identified an optimum vinylheteroarene scaffold for selective cysteine modification. Utilisation of this lead linker to modify a number of protein substrates with various functionalities, including the synthesis of a homogeneous, stable and biologically active antibody-drug conjugate (ADC) was then achieved. The reagent was also efficient in labelling proteome-wide cysteines in cell lysates. The efficiency and selectivity of these reagents as well as the stability of the products makes them suitable for the generation of biotherapeutics or studies in chemical biology.
Collapse
Affiliation(s)
- Hikaru Seki
- Department of Chemistry, University of Cambridge Lensfield Road Cambridge CB2 1EW UK
| | - Stephen J Walsh
- Department of Chemistry, University of Cambridge Lensfield Road Cambridge CB2 1EW UK
- Cancer Research UK Cambridge Institute, University of Cambridge Robinson Way Cambridge CB2 0RE UK
| | - Jonathan D Bargh
- Department of Chemistry, University of Cambridge Lensfield Road Cambridge CB2 1EW UK
| | - Jeremy S Parker
- Early Chemical Development, Pharmaceutical Sciences, R&D, AstraZeneca Macclesfield UK
| | - Jason Carroll
- Cancer Research UK Cambridge Institute, University of Cambridge Robinson Way Cambridge CB2 0RE UK
| | - David R Spring
- Department of Chemistry, University of Cambridge Lensfield Road Cambridge CB2 1EW UK
| |
Collapse
|
7
|
Arkenberg MR, Dimmitt NH, Johnson HC, Koehler KR, Lin CC. Dynamic Click Hydrogels for Xeno-Free Culture of Induced Pluripotent Stem Cells. ADVANCED BIOSYSTEMS 2020; 4:e2000129. [PMID: 32924337 PMCID: PMC7704730 DOI: 10.1002/adbi.202000129] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/05/2020] [Indexed: 12/25/2022]
Abstract
Xeno-free, chemically defined poly(ethylene glycol) (PEG)-based hydrogels are being increasingly used for in vitro culture and differentiation of human induced pluripotent stem cells (hiPSCs). These synthetic matrices provide tunable gelation and adaptable material properties crucial for guiding stem cell fate. Here, sequential norbornene-click chemistries are integrated to form synthetic, dynamically tunable PEG-peptide hydrogels for hiPSCs culture and differentiation. Specifically, hiPSCs are photoencapsulated in thiol-norbornene hydrogels crosslinked by multiarm PEG-norbornene (PEG-NB) and proteaselabile crosslinkers. These matrices are used to evaluate hiPSC growth under the influence of extracellular matrix properties. Tetrazine-norbornene (Tz-NB) click reaction is then employed to dynamically stiffen the cell-laden hydrogels. Fast reactive Tz and its stable derivative methyltetrazine (mTz) are tethered to multiarm PEG, yielding mono-functionalized PEG-Tz, PEG-mTz, and dualfunctionalized PEG-Tz/mTz that react with PEG-NB to form additional crosslinks in the cell-laden hydrogels. The versatility of Tz-NB stiffening is demonstrated with different Tz-modified macromers or by intermittent incubation of PEG-Tz for temporal stiffening. Finally, the Tz-NB-mediated dynamic stiffening is explored for 4D culture and definitive endoderm differentiation of hiPSCs. Overall, this dynamic hydrogel platform affords exquisite controls of hydrogel crosslinking for serving as a xeno-free and dynamic stem cell niche.
Collapse
Affiliation(s)
- Matthew R Arkenberg
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Nathan H Dimmitt
- Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Hunter C Johnson
- Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Karl R Koehler
- Departments of Otolaryngology and Plastic and Oral Surgery, F.M. Kirby Neurobiology Center, Boston Children's Hospital/Harvard Medical School, Boston, MA, 02115, USA
| | - Chien-Chi Lin
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| |
Collapse
|
8
|
Kamber DN, Nguyen SS, Liu F, Briggs JS, Shih HW, Row RD, Long ZG, Houk KN, Liang Y, Prescher JA. Isomeric triazines exhibit unique profiles of bioorthogonal reactivity. Chem Sci 2019; 10:9109-9114. [PMID: 31908754 PMCID: PMC6910137 DOI: 10.1039/c9sc01427f] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 08/11/2019] [Indexed: 11/29/2022] Open
Abstract
Expanding the scope of bioorthogonal reactivity requires access to new and mutually compatible reagents. We report here that 1,2,4-triazines can be tuned to exhibit unique reaction profiles with biocompatible strained alkenes and alkynes. Computational analyses were used to identify candidate orthogonal reactions, and the predictions were experimentally verified. Notably, 5-substituted triazines, unlike their 6-substituted counterparts, undergo rapid [4 + 2] cycloadditions with a sterically encumbered strained alkyne. This unique, sterically controlled reactivity was exploited for dual bioorthogonal labeling. Mutually orthogonal triazines and cycloaddition chemistries will enable new multi-component imaging applications.
Collapse
Affiliation(s)
- David N Kamber
- Department of Chemistry , University of California , Irvine , California 92697 , USA .
| | - Sean S Nguyen
- Department of Chemistry , University of California , Irvine , California 92697 , USA .
| | - Fang Liu
- Department of Chemistry and Biochemistry , University of California , Los Angeles , California 90095 , USA
- State Key Laboratory of Coordination Chemistry , Jiangsu Key Laboratory of Advanced Organic Materials , School of Chemistry and Chemical Engineering , Nanjing University , Nanjing 210023 , China .
| | - Jeffrey S Briggs
- Department of Chemistry , University of California , Irvine , California 92697 , USA .
| | - Hui-Wen Shih
- Department of Chemistry , University of California , Irvine , California 92697 , USA .
| | - R David Row
- Department of Chemistry , University of California , Irvine , California 92697 , USA .
| | - Zane G Long
- Department of Chemistry , University of California , Irvine , California 92697 , USA .
| | - K N Houk
- Department of Chemistry and Biochemistry , University of California , Los Angeles , California 90095 , USA
| | - Yong Liang
- State Key Laboratory of Coordination Chemistry , Jiangsu Key Laboratory of Advanced Organic Materials , School of Chemistry and Chemical Engineering , Nanjing University , Nanjing 210023 , China .
| | - Jennifer A Prescher
- Department of Chemistry , University of California , Irvine , California 92697 , USA .
- Department of Molecular Biology & Biochemistry , University of California , Irvine , California 92697 , USA
- Department of Pharmaceutical Sciences , University of California , Irvine , California 92697 , USA
| |
Collapse
|
9
|
Umlauf BJ, Clark PA, Lajoie JM, Georgieva JV, Bremner S, Herrin BR, Kuo JS, Shusta EV. Identification of variable lymphocyte receptors that can target therapeutics to pathologically exposed brain extracellular matrix. SCIENCE ADVANCES 2019; 5:eaau4245. [PMID: 31106264 PMCID: PMC6520025 DOI: 10.1126/sciadv.aau4245] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 04/02/2019] [Indexed: 05/21/2023]
Abstract
Diseases that lead to blood-brain barrier (BBB) disruption will pathologically expose normally inaccessible brain extracellular matrix (ECM) to circulating blood components. Therefore, we hypothesized that brain ECM-targeting moieties could specifically target the disrupted BBB and potentially deliver therapies. Variable lymphocyte receptors (VLRs) that preferentially associate with brain ECM were identified from an immune VLR library via yeast surface display biopanning coupled with a moderate throughput ECM screen. Brain ECM binding of VLR clones to murine and human brain tissue sections was confirmed. After systemic administration, P1C10, the lead brain ECM-targeting VLR candidate, specifically accumulated in brains with mannitol-disrupted BBB and at disrupted BBB regions in two different intracranial glioblastoma models. We also demonstrate P1C10's ability to deliver doxorubicin-loaded liposomes, leading to significantly improved survival in glioblastoma-bearing mice. Thus, VLRs can be used to selectively target pathologically exposed brain ECM and deliver drug payloads.
Collapse
Affiliation(s)
- Benjamin J. Umlauf
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Paul A. Clark
- Department of Neurological Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Jason M. Lajoie
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Julia V. Georgieva
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Samantha Bremner
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | | | - John S. Kuo
- Department of Neurological Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
- Mulva Clinic for the Neurosciences, The University of Texas at Austin, Austin, TX, USA
- Corresponding author. (E.V.S.); (J.S.K.)
| | - Eric V. Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Corresponding author. (E.V.S.); (J.S.K.)
| |
Collapse
|
10
|
Khera E, Thurber GM. Pharmacokinetic and Immunological Considerations for Expanding the Therapeutic Window of Next-Generation Antibody-Drug Conjugates. BioDrugs 2019; 32:465-480. [PMID: 30132210 DOI: 10.1007/s40259-018-0302-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Antibody-drug conjugate (ADC) development has evolved greatly over the last 3 decades, including the Food and Drug Administration (FDA) approval of several new drugs. However, translating ADCs from the design stage and preclinical promise to clinical success has been a major hurdle for the field, particularly for solid tumors. The challenge in clinical development can be attributed to the difficulty in connecting the design of these multifaceted agents with the impact on clinical efficacy, especially with the accelerated development of 'next-generation' ADCs containing a variety of innovative biophysical developments. Given their complex nature, there is an urgent need to integrate holistic ADC characterization approaches. This includes comprehensive in vivo assessment of systemic, intratumoral and cellular pharmacokinetics, pharmacodynamics, toxicodynamics, and interactions with the immune system, with the aim of optimizing the ADC therapeutic window. Pharmacokinetic/pharmacodynamic factors influencing the ADC therapeutic window include (1) selecting optimal target and ADC components for prolonged and stable plasma circulation to increase tumoral uptake with minimal non-specific systemic toxicity, (2) balancing homogeneous intratumoral distribution with efficient cellular uptake, and (3) translating improved ADC potency to better clinical efficacy. Balancing beneficial immunological effects such as Fc-mediated and payload-mediated immune cell activation against harmful immunogenic/toxic effects is also an emerging concern for ADCs. Here, we review practical considerations for tracking ADC efficacy and toxicity, as aided by high-resolution biomolecular and immunological tools, quantitative pharmacology, and mathematical models, all of which can elucidate the relative contributions of the multitude of interactions governing the ADC therapeutic window.
Collapse
Affiliation(s)
- Eshita Khera
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Rd, Ann Arbor, MI, 48109, USA
| | - Greg M Thurber
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Rd, Ann Arbor, MI, 48109, USA. .,Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|