1
|
Lee HJ, Chae BH, Ko DH, Lee SG, Yoon SR, Kim DS, Kim YS. Enhancing the cytotoxicity of immunotoxins by facilitating their dissociation from target receptors under the reducing conditions of the endocytic pathway. Int J Biol Macromol 2024; 278:134668. [PMID: 39137851 DOI: 10.1016/j.ijbiomac.2024.134668] [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: 06/18/2024] [Revised: 07/29/2024] [Accepted: 08/09/2024] [Indexed: 08/15/2024]
Abstract
Immunotoxins (ITs) are recombinant chimeric proteins that combine a protein toxin with a targeting moiety to facilitate the selective delivery of the toxin to cancer cells. Here, we present a novel strategy to enhance the cytosolic access of ITs by promoting their dissociation from target receptors under the reducing conditions of the endocytic pathway. We engineered monobodySS, a human fibronectin type III domain-based monobody with disulfide bond (SS)-containing paratopes, targeting receptors such as EGFR, EpCAM, Her2, and FAP. MonobodySS exhibited SS-dependent target receptor binding with a significant reduction in binding under reducing conditions. We then created monobodySS-based ITs carrying a 25 kDa fragment of Pseudomonas exotoxin A (PE25), termed monobodySS-PE25. These ITs showed dose-dependent cytotoxicity against target receptor-expressing cancer cells and a wider therapeutic window due to higher efficacy at lower doses compared to controls with SS reduction inhibited. ERSS/28-PE25, with a KD of 28 nM for EGFR, demonstrated superior tumor-killing potency compared to ER/21-PE25, which lacks an SS bond, at equivalent and lower doses. In vivo, ERSS/28-PE25 outperformed ER/21-PE25 in suppressing tumor growth in EGFR-overexpressing xenograft mouse models. This study presents a strategy for developing solid tumor-targeting ITs using SS-containing paratopes to enhance cytosolic delivery and antitumor efficacy.
Collapse
Affiliation(s)
- Hyun-Jin Lee
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Byeong-Ho Chae
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Deok-Han Ko
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Seul-Gi Lee
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Sang-Rok Yoon
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Dae-Seong Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Yong-Sung Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea; Advanced College of Bio-convergence Engineering, Ajou University, Suwon 16499, Republic of Korea.
| |
Collapse
|
2
|
Mohanan S, Guan X, Liang M, Karakoti A, Vinu A. Stimuli-Responsive Silica Silanol Conjugates: Strategic Nanoarchitectonics in Targeted Drug Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2301113. [PMID: 36967548 DOI: 10.1002/smll.202301113] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/28/2023] [Indexed: 06/18/2023]
Abstract
The design of novel drug delivery systems is exceptionally critical in disease treatments. Among the existing drug delivery systems, mesoporous silica nanoparticles (MSNs) have shown profuse promise owing to their structural stability, tunable morphologies/sizes, and ability to load different payload chemistry. Significantly, the presence of surface silanol groups enables functionalization with relevant drugs, imaging, and targeting agents, promoting their utility and popularity among researchers. Stimuli-responsive silanol conjugates have been developed as a novel, more effective way to conjugate, deliver, and release therapeutic drugs on demand and precisely to the selected location. Therefore, it is urgent to summarize the current understanding and the surface silanols' role in making MSN a versatile drug delivery platform. This review provides an analytical understanding of the surface silanols, chemistry, identification methods, and their property-performance correlation. The chemistry involved in converting surface silanols to a stimuli-responsive silica delivery system by endogenous/exogenous stimuli, including pH, redox potential, temperature, and hypoxia, is discussed in depth. Different chemistries for converting surface silanols to stimuli-responsive bonds are discussed in the context of drug delivery. The critical discussion is culminated by outlining the challenges in identifying silanols' role and overcoming the limitations in synthesizing stimuli-responsive mesoporous silica-based drug delivery systems.
Collapse
Affiliation(s)
- Shan Mohanan
- Global Innovative Centre for Advanced Nanomaterials, The School of Engineering, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, 2308, Australia
| | - Xinwei Guan
- Global Innovative Centre for Advanced Nanomaterials, The School of Engineering, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, 2308, Australia
| | - Mingtao Liang
- School of Biomedical Sciences and Pharmacy, College of Health Medicine and Wellbeing, The University of Newcastle, Callaghan, 2308, Australia
| | - Ajay Karakoti
- Global Innovative Centre for Advanced Nanomaterials, The School of Engineering, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, 2308, Australia
| | - Ajayan Vinu
- Global Innovative Centre for Advanced Nanomaterials, The School of Engineering, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, 2308, Australia
| |
Collapse
|
3
|
Slezak A, Chang K, Hossainy S, Mansurov A, Rowan SJ, Hubbell JA, Guler MO. Therapeutic synthetic and natural materials for immunoengineering. Chem Soc Rev 2024; 53:1789-1822. [PMID: 38170619 PMCID: PMC11557218 DOI: 10.1039/d3cs00805c] [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] [Indexed: 01/05/2024]
Abstract
Immunoengineering is a rapidly evolving field that has been driving innovations in manipulating immune system for new treatment tools and methods. The need for materials for immunoengineering applications has gained significant attention in recent years due to the growing demand for effective therapies that can target and regulate the immune system. Biologics and biomaterials are emerging as promising tools for controlling immune responses, and a wide variety of materials, including proteins, polymers, nanoparticles, and hydrogels, are being developed for this purpose. In this review article, we explore the different types of materials used in immunoengineering applications, their properties and design principles, and highlight the latest therapeutic materials advancements. Recent works in adjuvants, vaccines, immune tolerance, immunotherapy, and tissue models for immunoengineering studies are discussed.
Collapse
Affiliation(s)
- Anna Slezak
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
| | - Kevin Chang
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
| | - Samir Hossainy
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
| | - Aslan Mansurov
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
| | - Stuart J Rowan
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
- Department of Chemistry, The University of Chicago, Chicago, IL, 60637, USA
| | - Jeffrey A Hubbell
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
| | - Mustafa O Guler
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
4
|
Liu H, Lu HH, Alp Y, Wu R, Thayumanavan S. Structural Determinants of Stimuli-Responsiveness in Amphiphilic Macromolecular Nano-assemblies. Prog Polym Sci 2024; 148:101765. [PMID: 38476148 PMCID: PMC10927256 DOI: 10.1016/j.progpolymsci.2023.101765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Stimuli-responsive nano-assemblies from amphiphilic macromolecules could undergo controlled structural transformations and generate diverse macroscopic phenomenon under stimuli. Due to the controllable responsiveness, they have been applied for broad material and biomedical applications, such as biologics delivery, sensing, imaging, and catalysis. Understanding the mechanisms of the assembly-disassembly processes and structural determinants behind the responsive properties is fundamentally important for designing the next generation of nano-assemblies with programmable responsiveness. In this review, we focus on structural determinants of assemblies from amphiphilic macromolecules and their macromolecular level alterations under stimuli, such as the disruption of hydrophilic-lipophilic balance (HLB), depolymerization, decrosslinking, and changes of molecular packing in assemblies, which eventually lead to a series of macroscopic phenomenon for practical purposes. Applications of stimuli-responsive nano-assemblies in delivery, sensing and imaging were also summarized based on their structural features. We expect this review could provide readers an overview of the structural considerations in the design and applications of nanoassemblies and incentivize more explorations in stimuli-responsive soft matters.
Collapse
Affiliation(s)
- Hongxu Liu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065 P. R. China
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Hung-Hsun Lu
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Yasin Alp
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Ruiling Wu
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - S. Thayumanavan
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
5
|
Keller T, Trinks N, Brand J, Trippmacher S, Stahlhut P, Albrecht K, Papastavrou G, Koepsell H, Sauer M, Groll J. Design of Nanohydrogels for Targeted Intracellular Drug Transport to the Trans-Golgi Network. Adv Healthc Mater 2023; 12:e2201794. [PMID: 36739269 PMCID: PMC11469190 DOI: 10.1002/adhm.202201794] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 01/27/2023] [Indexed: 02/06/2023]
Abstract
Nanohydrogels combine advantages of hydrogels and nanoparticles. In particular, they represent promising drug delivery systems. Nanogel synthesis by oxidative condensation of polyglycidol prepolymers, that are modified with thiol groups, results in crosslinking by disulfide bonds. Hereby, biomolecules like the antidiabetic peptide RS1-reg, derived from the regulatory protein RS1 of the Na+ -D-glucose cotransporter SGLT1, can be covalently bound by cysteine residues to the nanogel in a hydrophilic, stabilizing environment. After oral uptake, the acid-stable nanogels protect their loading during gastric passage from proteolytic degradation. Under alkaline conditions in small intestine the nanohydrogels become mucoadhesive, pass the intestinal mucosa and are taken up into small intestinal enterocytes by endocytosis. Using Caco-2 cells as a model for small intestinal enterocytes, by confocal laser scanning microscopy and structured illumination microscopy, the colocalization of fluorescent-labeled RS1-reg with markers of endosomes, lysosomes, and trans-Golgi-network after uptake with polyglycidol-based nanogels formed by precipitation polymerization is demonstrated. This indicates that RS1-reg follows the endosomal pathway. In the following, the design of bespoken nanohydrogels for specific targeting of RS1-reg to its site of action at the trans-Golgi network is described that might also represent a way of targeted transport for other drugs to their targets at the Golgi apparatus.
Collapse
Affiliation(s)
- Thorsten Keller
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and BiofabricationUniversity of WürzburgPleicherwall 297070WürzburgGermany
| | - Nora Trinks
- Department of Biotechnology and BiophysicsUniversity of WürzburgAm Hubland97074WürzburgGermany
| | - Jessica Brand
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and BiofabricationUniversity of WürzburgPleicherwall 297070WürzburgGermany
| | - Steffen Trippmacher
- Physical Chemistry IIUniversity of BayreuthUniversitätsstr. 3095440BayreuthGermany
| | - Philipp Stahlhut
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and BiofabricationUniversity of WürzburgPleicherwall 297070WürzburgGermany
| | - Krystyna Albrecht
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and BiofabricationUniversity of WürzburgPleicherwall 297070WürzburgGermany
| | - Georg Papastavrou
- Physical Chemistry IIUniversity of BayreuthUniversitätsstr. 3095440BayreuthGermany
| | - Hermann Koepsell
- Institute of Anatomy and Cell BiologyUniversity of WürzburgKoellikerstraße 697070WürzburgGermany
| | - Markus Sauer
- Department of Biotechnology and BiophysicsUniversity of WürzburgAm Hubland97074WürzburgGermany
| | - Jürgen Groll
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and BiofabricationUniversity of WürzburgPleicherwall 297070WürzburgGermany
| |
Collapse
|
6
|
Peng Q, Xiong T, Ji F, Ren J, Jia L. Reduction-Activatable Fluorogenic Nanobody for Targeted and Low-Background Bioimaging. Anal Chem 2023; 95:2804-2811. [PMID: 36709506 DOI: 10.1021/acs.analchem.2c04132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Environment-sensitive fluorogenic antibodies enable target-specific bioimaging with reduced unspecific background signal and improved spatiotemporal resolution. However, current strategies for the construction of fluorogenic antibodies are hard to handle due to challenges that lie in the prior design of fluorogenic probes and subsequent antibody labeling. Here, we report a simple strategy to generate a fluorogenic nanobody, which we term D-body, by in situ incorporation of a reduction-responsive Nile blue foldamer which is self-quenched via a dimerization-caused quenching mechanism. The D-body can be efficiently internalized by cells with high epidermal growth factor receptor expression levels and is highly fluorogenic upon lysosomal activation, allowing wash-free cell imaging with exquisite specificity and fast in vivo imaging with a high tumor-to-background ratio. The modular D-body is readily available and easy to handle, offering a platform that is highly tunable for bioimaging applications.
Collapse
Affiliation(s)
- Qiang Peng
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116024, P. R. China
| | - Tao Xiong
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, P. R. China
| | - Fangling Ji
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116024, P. R. China
| | - Jun Ren
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116024, P. R. China
| | - Lingyun Jia
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116024, P. R. China
| |
Collapse
|
7
|
Sultana A, Zare M, Thomas V, Kumar TS, Ramakrishna S. Nano-based drug delivery systems: Conventional drug delivery routes, recent developments and future prospects. MEDICINE IN DRUG DISCOVERY 2022. [DOI: 10.1016/j.medidd.2022.100134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
8
|
Sousa de Almeida M, Rothen-Rutishauser B, Mayer M, Taskova M. Multi-Functionalized Heteroduplex Antisense Oligonucleotides for Targeted Intracellular Delivery and Gene Silencing in HeLa Cells. Biomedicines 2022; 10:biomedicines10092096. [PMID: 36140196 PMCID: PMC9495875 DOI: 10.3390/biomedicines10092096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/19/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Oligonucleotide therapeutics, antisense oligonucleotides (ASOs) and short interfering RNA (siRNA) are short synthetic nucleic acid molecules with a promising potential to treat a wide range of diseases. Despite considerable progress in the field, the development of safe and effective delivery systems that target organs and tissues other than the liver is challenging. While keeping possible off-target oligonucleotide interactions and toxicity related to chemical modifications in mind, innovative solutions for targeted intracellular delivery are highly needed. Herein, we report on the design, synthesis and testing of a novel multi-modified and multi-functionalized heteroduplex oligonucleotide (HDO) with respect to its intracellular delivery and its ability to silence genes in HeLa cells. Simultaneously, folic acid- and peptide- labeled HDO show proficient silencing of the green fluorescent protein (GFP) gene with an 84% reduction in the GFP fluorescence. In addition, the Bcl2 HDO achieved effective Bcl2 gene knockdown in the cells. The data show the proficiency of the multi-functionalization strategy and provide an example for advancing the design of safe and efficient forthcoming oligonucleotide therapeutics, such as HDO.
Collapse
Affiliation(s)
- Mauro Sousa de Almeida
- BioNanomaterials, Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Barbara Rothen-Rutishauser
- BioNanomaterials, Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Michael Mayer
- Biophysics, Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Maria Taskova
- Biophysics, Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
- Correspondence:
| |
Collapse
|
9
|
Valdez S, Robertson M, Qiang Z. Fluorescence Resonance Energy Transfer Measurements in Polymer Science: A Review. Macromol Rapid Commun 2022; 43:e2200421. [PMID: 35689335 DOI: 10.1002/marc.202200421] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/06/2022] [Indexed: 12/27/2022]
Abstract
Fluorescence resonance energy transfer (FRET) is a non-invasive characterization method for studying molecular structures and dynamics, providing high spatial resolution at nanometer scale. Over the past decades, FRET-based measurements are developed and widely implemented in synthetic polymer systems for understanding and detecting a variety of nanoscale phenomena, enabling significant advances in polymer science. In this review, the basic principles of fluorescence and FRET are briefly discussed. Several representative research areas are highlighted, where FRET spectroscopy and imaging can be employed to reveal polymer morphology and kinetics. These examples include understanding polymer micelle formation and stability, detecting guest molecule release from polymer host, characterizing supramolecular assembly, imaging composite interfaces, and determining polymer chain conformations and their diffusion kinetics. Finally, a perspective on the opportunities of FRET-based measurements is provided for further allowing their greater contributions in this exciting area.
Collapse
Affiliation(s)
- Sara Valdez
- School of Polymer Science and Engineering, University of Southern Mississippi, Hattiesburg, MS, 39406, USA
| | - Mark Robertson
- School of Polymer Science and Engineering, University of Southern Mississippi, Hattiesburg, MS, 39406, USA
| | - Zhe Qiang
- School of Polymer Science and Engineering, University of Southern Mississippi, Hattiesburg, MS, 39406, USA
| |
Collapse
|
10
|
Li X, Omonova Tuychi Qizi C, Mohamed Khamis A, Zhang C, Su Z. Nanotechnology for Enhanced Cytoplasmic and Organelle Delivery of Bioactive Molecules to Immune Cells. Pharm Res 2022; 39:1065-1083. [PMID: 35661086 DOI: 10.1007/s11095-022-03284-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 04/30/2022] [Indexed: 12/18/2022]
Abstract
Immune cells stand as a critical component of the immune system to maintain the internal environment homeostasis. The dysfunction of immune cells can result in various life-threatening diseases, including refractory infection, diabetes, cardiovascular disease, and cancer. Therefore, strategies to standardize or even enhance the function of immune cells are critical. Recently, nanotechnology has been highly researched and extensively applied for enhancing the cytoplasmic delivery of bioactive molecules to immune cells, providing efficient approaches to correct in vivo and in vitro dysfunction of immune cells. This review focuses on the technologies and challenges involved in improving endo-lysosomal escape, cytoplasmic release and organelle targeted delivery of different bioactive molecules in immune cells. Furthermore, it will elaborate on the broader vision of applying nanotechnology for treating immune cell-related diseases and constructing immune therapies and cytopharmaceuticals as potential treatments for diseases.
Collapse
Affiliation(s)
- Xiaoyu Li
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Charos Omonova Tuychi Qizi
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Amari Mohamed Khamis
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Can Zhang
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, 210009, China.
| | - Zhigui Su
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
11
|
Tian Y, Tirrell MV, LaBelle JL. Harnessing the Therapeutic Potential of Biomacromolecules through Intracellular Delivery of Nucleic Acids, Peptides, and Proteins. Adv Healthc Mater 2022; 11:e2102600. [PMID: 35285167 PMCID: PMC9232950 DOI: 10.1002/adhm.202102600] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 02/09/2022] [Indexed: 12/19/2022]
Abstract
Biomacromolecules have long been at the leading edge of academic and pharmaceutical drug development and clinical translation. With the clinical advances of new therapeutics, such as monoclonal antibodies and nucleic acids, the array of medical applications of biomacromolecules has broadened considerably. A major on-going effort is to expand therapeutic targets within intracellular locations. Owing to their large sizes, abundant charges, and hydrogen-bond donors and acceptors, advanced delivery technologies are required to deliver biomacromolecules effectively inside cells. In this review, strategies used for the intracellular delivery of three major forms of biomacromolecules: nucleic acids, proteins, and peptides, are highlighted. An emphasis is placed on synthetic delivery approaches and the major hurdles needed to be overcome for their ultimate clinical translation.
Collapse
Affiliation(s)
- Yu Tian
- Pritzker School of Molecular EngineeringThe University of Chicago5640 S Ellis AveChicagoIL60637USA
| | - Matthew V. Tirrell
- Pritzker School of Molecular EngineeringThe University of Chicago5640 S Ellis AveChicagoIL60637USA
| | - James L. LaBelle
- Department of Pediatrics, Section of Hematology/OncologyThe University of Chicago900 E 57th StChicagoIL60637USA
| |
Collapse
|
12
|
Napoleon JV, Zhang B, Luo Q, Srinivasarao M, Low PS. Design, Synthesis, and Targeted Delivery of an Immune Stimulant that Selectively Reactivates Exhausted CAR T Cells. Angew Chem Int Ed Engl 2022; 61:e202113341. [PMID: 35088497 DOI: 10.1002/anie.202113341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Indexed: 12/13/2022]
Abstract
Although chimeric antigen receptor (CAR) T cells have demonstrated significant promise in suppressing hematopoietic cancers, their applications in treating solid tumors have been limited by onset of CAR T cell exhaustion that accompanies continuous CAR T cell exposure to tumor antigen. To address this limitation, we have exploited the abilities of recently designed universal CARs to bind fluorescein and internalize a fluorescein-TLR7 agonist conjugate by CAR-mediated endocytosis. We demonstrate here that anti-fluorescein CAR-mediated uptake of a fluorescein-TLR7-3 conjugate can reactivate exhausted CAR T cells, leading to dramatic reduction in T cell exhaustion markers (PD-1+ Tim-3+ ) and shrinkage of otherwise resistant tumors without inducing systemic activation of the immune system. We conclude that CAR T cell exhaustion can be reversed by administration of a CAR-targeted TLR7 agonist, thereby enabling the CAR T cells to successfully treat solid tumors without incurring the systemic toxicity that commonly accompanies administration of nontargeted TLR7 agonists.
Collapse
Affiliation(s)
- John Victor Napoleon
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA
| | - Boning Zhang
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA
| | - Qian Luo
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA
| | - Madduri Srinivasarao
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA
| | - Philip S Low
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
13
|
Sheyi R, de la Torre BG, Albericio F. Linkers: An Assurance for Controlled Delivery of Antibody-Drug Conjugate. Pharmaceutics 2022; 14:pharmaceutics14020396. [PMID: 35214128 PMCID: PMC8874516 DOI: 10.3390/pharmaceutics14020396] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/27/2022] [Accepted: 02/04/2022] [Indexed: 12/15/2022] Open
Abstract
As one of the major therapeutic options for cancer treatment, chemotherapy has limited selectivity against cancer cells. Consequently, this therapeutic strategy offers a small therapeutic window with potentially high toxicity and thus limited efficacy of doses that can be tolerated by patients. Antibody-drug conjugates (ADCs) are an emerging class of anti-cancer therapeutic drugs that can deliver highly cytotoxic molecules directly to cancer cells. To date, twelve ADCs have received market approval, with several others in clinical stages. ADCs have become a powerful class of therapeutic agents in oncology and hematology. ADCs consist of recombinant monoclonal antibodies that are covalently bound to cytotoxic chemicals via synthetic linkers. The linker has a key role in ADC outcomes because its characteristics substantially impact the therapeutic index efficacy and pharmacokinetics of these drugs. Stable linkers and ADCs can maintain antibody concentration in blood circulation, and they do not release the cytotoxic drug before it reaches its target, thus resulting in minimum off-target effects. The linkers used in ADC development can be classified as cleavable and non-cleavable. The former, in turn, can be grouped into three types: hydrazone, disulfide, or peptide linkers. In this review, we highlight the various linkers used in ADC development and their design strategy, release mechanisms, and future perspectives.
Collapse
Affiliation(s)
- Rotimi Sheyi
- School of Chemistry and Physics, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Beatriz G. de la Torre
- Kwazulu-Natal Research Innovation and Sequencing Platform (KRISP), College of Health Sciences, University of KwaZulu-Natal, Durban 4001, South Africa
- Correspondence: (B.G.d.l.T.); (F.A.); Tel.: +27-614-047-528 (B.G.d.l.T.); +27-6140-09144 (F.A.)
| | - Fernando Albericio
- School of Chemistry and Physics, University of KwaZulu-Natal, Durban 4001, South Africa;
- Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
- Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Department of Organic Chemistry, University of Barcelona, 08028 Barcelona, Spain
- Correspondence: (B.G.d.l.T.); (F.A.); Tel.: +27-614-047-528 (B.G.d.l.T.); +27-6140-09144 (F.A.)
| |
Collapse
|
14
|
Van de Vyver T, De Smedt SC, Raemdonck K. Modulating intracellular pathways to improve non-viral delivery of RNA therapeutics. Adv Drug Deliv Rev 2022; 181:114041. [PMID: 34763002 DOI: 10.1016/j.addr.2021.114041] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/12/2021] [Accepted: 11/02/2021] [Indexed: 12/12/2022]
Abstract
RNA therapeutics (e.g. siRNA, oligonucleotides, mRNA, etc.) show great potential for the treatment of a myriad of diseases. However, to reach their site of action in the cytosol or nucleus of target cells, multiple intra- and extracellular barriers have to be surmounted. Several non-viral delivery systems, such as nanoparticles and conjugates, have been successfully developed to meet this requirement. Unfortunately, despite these clear advances, state-of-the-art delivery agents still suffer from relatively low intracellular delivery efficiencies. Notably, our current understanding of the intracellular delivery process is largely oversimplified. Gaining mechanistic insight into how RNA formulations are processed by cells will fuel rational design of the next generation of delivery carriers. In addition, identifying which intracellular pathways contribute to productive RNA delivery could provide opportunities to boost the delivery performance of existing nanoformulations. In this review, we discuss both established as well as emerging techniques that can be used to assess the impact of different intracellular barriers on RNA transfection performance. Next, we highlight how several modulators, including small molecules but also genetic perturbation technologies, can boost RNA delivery by intervening at differing stages of the intracellular delivery process, such as cellular uptake, intracellular trafficking, endosomal escape, autophagy and exocytosis.
Collapse
Affiliation(s)
- Thijs Van de Vyver
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Koen Raemdonck
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| |
Collapse
|
15
|
Napoleon JV, Zhang B, Luo Q, Srinivasarao M, Low PS. Design, Synthesis, and Targeted Delivery of an Immune Stimulant that Selectively Reactivates Exhausted CAR T Cells. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202113341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- John Victor Napoleon
- Purdue University Department of Chemistry Purdue University Institute for Drug Discovery720 Clinic Dr, 47907 West lafayette UNITED STATES
| | - Boning Zhang
- Purdue University Chemistry Purdue University Institute for Drug Discovery720 Clinic Dr, 47907 West Lafayette, UNITED STATES
| | - Qian Luo
- Purdue University Chemistry Purdue University Institute for Drug Discovery720 Clinic Dr, 47907 West lafayette UNITED STATES
| | - Madduri Srinivasarao
- Purdue University Chemistry Purdue University Institute for Drug Discovery720 Clinic Dr, 47907 West Lafayette UNITED STATES
| | - Philip S. Low
- Purdue University Department of Chemistry 720 clinic Dr 47907 West Lafayette UNITED STATES
| |
Collapse
|
16
|
Wylie B, Ong F, Belhoul-Fakir H, Priebatsch K, Bogdawa H, Stirnweiss A, Watt P, Cunningham P, Stone SR, Waithman J. Targeting Cross-Presentation as a Route to Improve the Efficiency of Peptide-Based Cancer Vaccines. Cancers (Basel) 2021; 13:6189. [PMID: 34944809 PMCID: PMC8699136 DOI: 10.3390/cancers13246189] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 12/06/2021] [Indexed: 11/17/2022] Open
Abstract
Cross-presenting dendritic cells (DC) offer an attractive target for vaccination due to their unique ability to process exogenous antigens for presentation on MHC class I molecules. Recent reports have established that these DC express unique surface receptors and play a critical role in the initiation of anti-tumor immunity, opening the way for the development of vaccination strategies specifically targeting these cells. This study investigated whether targeting cross-presenting DC by two complementary mechanisms could improve vaccine effectiveness, in both a viral setting and in a murine melanoma model. Our novel vaccine construct contained the XCL1 ligand, to target uptake to XCR1+ cross-presenting DC, and a cell penetrating peptide (CPP) with endosomal escape properties, to enhance antigen delivery into the cross-presentation pathway. Using a prime-boost regimen, we demonstrated robust expansion of antigen-specific T cells following vaccination with our CPP-linked peptide vaccine and protective immunity against HSV-1 skin infection, where vaccine epitopes were natively expressed by the virus. Additionally, our novel vaccination strategy slowed tumor outgrowth in a B16 murine melanoma model, compared to adjuvant only controls, suggesting antigen-specific anti-tumor immunity was generated following vaccination. These findings suggest that novel strategies to target the antigen cross-presentation pathway in DC may be beneficial for the generation of anti-tumor immunity.
Collapse
Affiliation(s)
- Ben Wylie
- Telethon Kids Institute, The University of Western Australia, Nedlands, WA 6009, Australia;
| | - Ferrer Ong
- PYC Therapeutics, Harry Perkins Institute, QEII Medical Centre, Nedlands, WA 6009, Australia; (F.O.); (A.S.); (P.C.)
| | - Hanane Belhoul-Fakir
- School of Public Health, Faculty of Health Sciences, Curtin University, Bentley, WA 6102, Australia;
| | | | | | - Anja Stirnweiss
- PYC Therapeutics, Harry Perkins Institute, QEII Medical Centre, Nedlands, WA 6009, Australia; (F.O.); (A.S.); (P.C.)
| | - Paul Watt
- Avicena, West Perth, WA 6005, Australia;
| | - Paula Cunningham
- PYC Therapeutics, Harry Perkins Institute, QEII Medical Centre, Nedlands, WA 6009, Australia; (F.O.); (A.S.); (P.C.)
| | - Shane R. Stone
- School of Agriculture and the Environment, University of Western Australia, Nedlands, WA 6009, Australia
| | - Jason Waithman
- Telethon Kids Institute, The University of Western Australia, Nedlands, WA 6009, Australia;
| |
Collapse
|
17
|
Kudruk S, Pottanam Chali S, Linard Matos AL, Bourque C, Dunker C, Gatsogiannis C, Ravoo BJ, Gerke V. Biodegradable and Dual-Responsive Polypeptide-Shelled Cyclodextrin-Containers for Intracellular Delivery of Membrane-Impermeable Cargo. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100694. [PMID: 34278745 PMCID: PMC8456233 DOI: 10.1002/advs.202100694] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/01/2021] [Indexed: 06/08/2023]
Abstract
The transport of membrane impermeable compounds into cells is a prerequisite for the efficient cellular delivery of hydrophilic and amphiphilic compounds and drugs. Transport into the cell's cytosolic compartment should ideally be controllable and it should involve biologically compatible and degradable vehicles. Addressing these challenges, nanocontainers based on cyclodextrin amphiphiles that are stabilized by a biodegradable peptide shell are developed and their potential to deliver fluorescently labeled cargo into human cells is analyzed. Host-guest mediated self-assembly of a thiol-containing short peptide or a cystamine-cross-linked polypeptide shell on cyclodextrin vesicles produce short peptide-shelled (SPSVss ) or polypeptide-shelled vesicles (PPSVss ), respectively, with redox-responsive and biodegradable features. Whereas SPSVss are permeable and less stable, PPSVss effectively encapsulate cargo and show a strictly regulated release of membrane impermeable cargo triggered by either reducing conditions or peptidase treatment. Live cell experiments reveal that the novel PPSVSS are readily internalized by primary human endothelial cells (human umbilical vein endothelial cells) and cervical cancer cells and that the reductive microenvironment of the cells' endosomes trigger release of the hydrophilic cargo into the cytosol. Thus, PPSVSS represent a highly efficient, biodegradable, and tunable system for overcoming the plasma membrane as a natural barrier for membrane-impermeable cargo.
Collapse
Affiliation(s)
- Sergej Kudruk
- Institute of Medical BiochemistryCenter for Molecular Biology of InflammationUniversity of MuensterVon‐Esmarch‐Str. 56Münster48149Germany
| | - Sharafudheen Pottanam Chali
- Center for Soft Nanoscience and Organic Chemistry InstituteUniversity of MuensterBusso Peus Straße 10Münster48149Germany
| | - Anna Livia Linard Matos
- Institute of Medical BiochemistryCenter for Molecular Biology of InflammationUniversity of MuensterVon‐Esmarch‐Str. 56Münster48149Germany
| | - Cole Bourque
- Center for Soft Nanoscience and Institute of Medical Physics and BiophysicsUniversity of MuensterBusso Peus Straße 10Münster48149Germany
- Max Planck Institute of Molecular PhysiologyOtto‐Hahn‐Straße 11Dortmund44227Germany
| | - Clara Dunker
- Institute of Medical BiochemistryCenter for Molecular Biology of InflammationUniversity of MuensterVon‐Esmarch‐Str. 56Münster48149Germany
| | - Christos Gatsogiannis
- Center for Soft Nanoscience and Institute of Medical Physics and BiophysicsUniversity of MuensterBusso Peus Straße 10Münster48149Germany
- Max Planck Institute of Molecular PhysiologyOtto‐Hahn‐Straße 11Dortmund44227Germany
| | - Bart Jan Ravoo
- Center for Soft Nanoscience and Organic Chemistry InstituteUniversity of MuensterBusso Peus Straße 10Münster48149Germany
| | - Volker Gerke
- Institute of Medical BiochemistryCenter for Molecular Biology of InflammationUniversity of MuensterVon‐Esmarch‐Str. 56Münster48149Germany
| |
Collapse
|
18
|
Mukherjee T, Kanvah S, Klymchenko AS, Collot M. Probing Variations of Reduction Activity at the Plasma Membrane Using a Targeted Ratiometric FRET Probe. ACS APPLIED MATERIALS & INTERFACES 2021; 13:40315-40324. [PMID: 34424677 DOI: 10.1021/acsami.1c11069] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Plasma membrane (PM) is the turntable of various reactions that regulate essential functionalities of cells. Among these reactions, the thiol disulfide exchange (TDE) reaction plays an important role in cellular processes. We herein designed a selective probe, called membrane reduction probe (MRP), that is able to report TDE activity at the PM. MRP is based on a green emitting BODIPY PM probe connected to rhodamine through a disulfide bond. MRP is fluorogenic as it is turned off in aqueous media due to aggregation-caused quenching, and once inserted in the PM, it displays a bright red signal due to an efficient fluorescence energy resonance transfer (FRET) between the BODIPY donor and the rhodamine acceptor. In the PM model, the MRP can undergo TDE reaction with external reductive agents as well as with thiolated lipids embedded in the bilayer. Upon TDE reaction, the FRET is turned off and a bright green signal appears allowing a ratiometric readout of this reaction. In cells, the MRP quickly labeled the PM and was able to probe variations of TDE activity using ratiometric imaging. With this tool in hand, we were able to monitor variations of TDE activity at the PM under stress conditions, and we showed that cancer cell lines presented a reduced TDE activity at the PM compared to noncancer cells.
Collapse
Affiliation(s)
- Tarushyam Mukherjee
- Discipline of Chemistry, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat 382355, India
| | - Sriram Kanvah
- Discipline of Chemistry, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat 382355, India
| | - Andrey S Klymchenko
- Laboratoire de Bioimagerie et Pathologies, UMR 7021, CNRS/Université de Strasbourg, 74 route du Rhin, 67401 Illkirch-Graffenstaden, France
| | - Mayeul Collot
- Laboratoire de Bioimagerie et Pathologies, UMR 7021, CNRS/Université de Strasbourg, 74 route du Rhin, 67401 Illkirch-Graffenstaden, France
| |
Collapse
|
19
|
Ghosal S, Walker JE, Alabi CA. Predictive Platforms of Bond Cleavage and Drug Release Kinetics for Macromolecule–Drug Conjugates. Annu Rev Chem Biomol Eng 2021; 12:241-261. [DOI: 10.1146/annurev-chembioeng-091720-030636] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Macromolecule–drug conjugates (MDCs) occupy a critical niche in modern pharmaceuticals that deals with the assembly and combination of a macromolecular carrier, a drug cargo, and a linker toward the creation of effective therapeutics. Macromolecular carriers such as synthetic biocompatible polymers and proteins are often exploited for their inherent ability to improve drug circulation, prevent off-target drug cytotoxicity, and widen the therapeutic index of drugs. One of the most significant challenges in MDC design involves tuning their drug release kinetics to achieve high spatiotemporal precision. This level of control requires a thorough qualitative and quantitative understanding of the bond cleavage event. In this review, we highlight specific research findings that emphasize the importance of establishing a precise structure–function relationship for MDCs that can be used to predict their bond cleavage and drug release kinetic parameters.
Collapse
Affiliation(s)
- Souvik Ghosal
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14850, USA
| | - Javon E. Walker
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14850, USA
| | - Christopher A. Alabi
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14850, USA
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14850, USA
| |
Collapse
|
20
|
Venkatesh C, Doorneweerd DD, Xia W, Putt KS, Low PS. Folate-targeted verrucarin A reduces the number of activated macrophages in a mouse model of acute peritonitis. Bioorg Med Chem Lett 2021; 42:128091. [PMID: 33964441 DOI: 10.1016/j.bmcl.2021.128091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/26/2021] [Accepted: 05/02/2021] [Indexed: 11/16/2022]
Abstract
Activated macrophages contribute prominently to the progression and maintenance of almost all inflammatory and autoimmune diseases. Although non-specific elimination of these phagocytes has been shown to treat animal models of inflammatory disease, the same therapies have been compromised by unacceptable toxicities, because they also kill quiescent macrophages in healthy tissues. In the studies below, we exploit upregulation of folate receptor beta (FRβ) on inflammatory (but not resting) macrophages to target a cytotoxic drug selectively to the inflammatory subset of macrophages. Because many of these activated macrophages are nondividing, we also employ verrucarin A as the cytotoxic payload, since it kills both mitotic and nonmitotic cells by blocking protein synthesis. By inserting a redox-sensitive self-immolative linker between the folate and verrucarin A, we further assure that release of unmodified verrucarin A is triggered primarily after internalization by an FRβ-positive cell. The resulting folate-verrucarin A conjugate is shown to kill FR-expressing cells in vitro in a manner that can be inhibited by competition with 100-fold excess folic acid. The folate-verrucarin A conjugate is also shown to successfully treat a murine model of inflammatory peritonitis by eliminating inflammatory macrophages without killing other cells in the same peritonitis fluid. Based on this high specificity for inflammatory macrophages, we conclude that folate-verrucarin A warrants continued exploration as a potential therapy for inflammatory and autoimmune diseases in humans.
Collapse
Affiliation(s)
- Chelvam Venkatesh
- Department of Chemistry, Indian Institute of Technology Indore, Indore, India; Department of Bioscience and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| | | | - Wei Xia
- Department of Chemistry, Purdue University, West Lafayette IN 47907, USA
| | - Karson S Putt
- Institute for Drug Discovery, Purdue University, West Lafayette IN 47907, USA
| | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette IN 47907, USA; Institute for Drug Discovery, Purdue University, West Lafayette IN 47907, USA.
| |
Collapse
|
21
|
Rana A, Bhatnagar S. Advancements in folate receptor targeting for anti-cancer therapy: A small molecule-drug conjugate approach. Bioorg Chem 2021; 112:104946. [PMID: 33989916 DOI: 10.1016/j.bioorg.2021.104946] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/17/2021] [Accepted: 04/22/2021] [Indexed: 10/21/2022]
Abstract
Targeted delivery combined with controlled release of drugs has a crucial role in future of personalized medicine. The majority of cancer drugs are intended to interfere with one or more cellular events. Anticancer agents can also be toxic to healthy cells, as healthy cells may also need to proliferate and avoid apoptosis. The focus of this review covers the principles, advantages, drawbacks and summarize criteria that must be met for design of small molecule-drug conjugates (SMDCs) to achieve the desired therapeutic potency with minimal toxicity. SMDCs are composed of a targeting ligand, a releasable bridge, a spacer, and a therapeutic payload. We summarize the criteria for the effective design that influences the selection of tumor specific receptor and optimum elements in the design of SMDCs. We also discuss the criteria for selecting the optimal therapeutic drug payload, spacer and linker. The linker chemistries and cleavage strategies are also discussed. Finally, we review the folate receptor targeting SMDCs that are in preclinical development and in clinical trials.
Collapse
Affiliation(s)
- Abhilash Rana
- Amity Institute of Biotechnology, Amity University, Sector125, Noida, Uttar Pradesh, India.
| | - Seema Bhatnagar
- Amity Institute of Biotechnology, Amity University, Sector125, Noida, Uttar Pradesh, India.
| |
Collapse
|
22
|
Dutta K, Das R, Medeiros J, Thayumanavan S. Disulfide Bridging Strategies in Viral and Nonviral Platforms for Nucleic Acid Delivery. Biochemistry 2021; 60:966-990. [PMID: 33428850 PMCID: PMC8753971 DOI: 10.1021/acs.biochem.0c00860] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Self-assembled nanostructures that are sensitive to environmental stimuli are promising nanomaterials for drug delivery. In this class, disulfide-containing redox-sensitive strategies have gained enormous attention because of their wide applicability and simplicity of nanoparticle design. In the context of nucleic acid delivery, numerous disulfide-based materials have been designed by relying on covalent or noncovalent interactions. In this review, we highlight major advances in the design of disulfide-containing materials for nucleic acid encapsulation, including covalent nucleic acid conjugates, viral vectors or virus-like particles, dendrimers, peptides, polymers, lipids, hydrogels, inorganic nanoparticles, and nucleic acid nanostructures. Our discussion will focus on the context of the design of materials and their impact on addressing the current shortcomings in the intracellular delivery of nucleic acids.
Collapse
Affiliation(s)
- Kingshuk Dutta
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Ritam Das
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
- The Center for Bioactive Delivery- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Jewel Medeiros
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
- The Center for Bioactive Delivery- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - S. Thayumanavan
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Massachusetts 01003, United States
- The Center for Bioactive Delivery- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
23
|
Discovery of De Novo Macrocyclic Peptides by Messenger RNA Display. Trends Pharmacol Sci 2021; 42:385-397. [PMID: 33771353 DOI: 10.1016/j.tips.2021.02.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/23/2021] [Accepted: 02/26/2021] [Indexed: 12/12/2022]
Abstract
Macrocyclic peptides are a promising class of compounds that can often engage challenging therapeutic targets. Display technologies, such as mRNA display, allow for the efficient discovery of macrocyclic peptides. This article reviews the current approaches for generating macrocyclic peptide libraries using mRNA display and highlights some recent examples of ribosomal incorporation of nonproteinogenic amino acids into macrocyclic peptides.
Collapse
|
24
|
Dutta K, Kanjilal P, Das R, Thayumanavan S. Synergistic Interplay of Covalent and Non-Covalent Interactions in Reactive Polymer Nanoassembly Facilitates Intracellular Delivery of Antibodies. Angew Chem Int Ed Engl 2021; 60:1821-1830. [PMID: 33034131 PMCID: PMC7855684 DOI: 10.1002/anie.202010412] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/23/2020] [Indexed: 01/29/2023]
Abstract
The primary impediments in developing large antibodies as drugs against intracellular targets involve their low transfection efficiency and suitable reversible encapsulation strategies for intracellular delivery with retention of biological activity. To address this, we outline an electrostatics-enhanced covalent self-assembly strategy to generate polymer-protein/antibody nanoassemblies. Through structure-activity studies, we down-select the best performing self-immolative pentafluorophenyl containing activated carbonate polymer for bioconjugation. With the help of an electrostatics-aided covalent self-assembly approach, we demonstrate efficient encapsulation of medium to large proteins (HRP, 44 kDa and β-gal, 465 kDa) and antibodies (ca. 150 kDa). The designed polymeric nanoassemblies are shown to successfully traffic functional antibodies (anti-NPC and anti-pAkt) to cytosol to elicit their bioactivity towards binding intracellular protein epitopes and inducing apoptosis.
Collapse
Affiliation(s)
| | | | - Ritam Das
- University of Massachusetts, Amherst, MA, 01003, USA
| | - Sankaran Thayumanavan
- Department of Chemistry, Molecular and Cellular Biology Program, and The Center for Bioactive Delivery-Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| |
Collapse
|
25
|
Zavoiura O, Brunner B, Casteels P, Zimmermann L, Ozog M, Boutton C, Helms MW, Wagenaar T, Adam V, Peterka J, Metz-Weidmann C, Deschaght P, Scheidler S, Jahn-Hofmann K. Nanobody-siRNA Conjugates for Targeted Delivery of siRNA to Cancer Cells. Mol Pharm 2021; 18:1048-1060. [PMID: 33444501 DOI: 10.1021/acs.molpharmaceut.0c01001] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Targeted extrahepatic delivery of siRNA remains a challenging task in the field of nucleic acid therapeutics. An ideal delivery tool must internalize siRNA exclusively into the cells of interest without affecting the silencing activity of siRNA. Here, we report the use of anti-EGFR Nanobodies (trademark of Ablynx N.V.) as tools for targeted siRNA delivery. A straightforward procedure for site-specific conjugation of siRNA to an engineered C-terminal cysteine residue on the Nanobody (trademark of Ablynx N.V.) is described. We show that siRNA-conjugated Nanobodies (Nb-siRNA) retain their binding to EGFR and enter EGFR-positive cells via receptor-mediated endocytosis. The activity of Nb-siRNAs was assessed by measuring the knockdown of a housekeeping gene (AHSA1) in EGFR-positive and EGFR-negative cells. We demonstrate that Nb-siRNAs are active in vitro and induce mRNA cleavage in the targeted cell line. In addition, we discuss the silencing activity of siRNA conjugated to fused Nbs with various combinations of EGFR-binding building blocks. Finally, we compare the performance of Nb-siRNA joined by four different linkers and discuss the advantages and limitations of using cleavable and noncleavable linkers in the context of Nanobody-mediated siRNA delivery.
Collapse
Affiliation(s)
- Oleksandr Zavoiura
- Sanofi, Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Bodo Brunner
- Sanofi, Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Peter Casteels
- Ablynx, a Sanofi Company, Technologiepark 21, 9052 Zwijnaarde, Belgium
| | - Luciana Zimmermann
- Sanofi, Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Matthias Ozog
- Sanofi, Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Carlo Boutton
- Ablynx, a Sanofi Company, Technologiepark 21, 9052 Zwijnaarde, Belgium
| | - Mike W Helms
- Sanofi, Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Timothy Wagenaar
- Sanofi, Research, 640 Memorial Drive, Cambridge, 02139 Massachusetts, United States
| | - Volker Adam
- Sanofi, Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Josefine Peterka
- Sanofi, TIDES platform, Industriepark Höchst, 65926, Frankfurt am Main, Germany
| | | | - Pieter Deschaght
- Ablynx, a Sanofi Company, Technologiepark 21, 9052 Zwijnaarde, Belgium
| | - Sabine Scheidler
- Sanofi, Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Kerstin Jahn-Hofmann
- Sanofi, Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| |
Collapse
|
26
|
Saw WS, Anasamy T, Foo YY, Kwa YC, Kue CS, Yeong CH, Kiew LV, Lee HB, Chung LY. Delivery of Nanoconstructs in Cancer Therapy: Challenges and Therapeutic Opportunities. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000206] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Wen Shang Saw
- Department of Pharmaceutical Chemistry Faculty of Pharmacy University of Malaya Kuala Lumpur 50603 Malaysia
| | - Theebaa Anasamy
- Department of Pharmaceutical Chemistry Faculty of Pharmacy University of Malaya Kuala Lumpur 50603 Malaysia
| | - Yiing Yee Foo
- Department of Pharmacology Faculty of Medicine University of Malaya Kuala Lumpur 50603 Malaysia
| | - Yee Chu Kwa
- Department of Pharmaceutical Chemistry Faculty of Pharmacy University of Malaya Kuala Lumpur 50603 Malaysia
| | - Chin Siang Kue
- Department of Diagnostic and Allied Health Sciences Faculty of Health and Life Sciences Management and Science University Shah Alam Selangor 40100 Malaysia
| | - Chai Hong Yeong
- School of Medicine Faculty of Health and Medical Sciences Taylor's University Subang Jaya Selangor 47500 Malaysia
| | - Lik Voon Kiew
- Department of Pharmacology Faculty of Medicine University of Malaya Kuala Lumpur 50603 Malaysia
| | - Hong Boon Lee
- Department of Pharmaceutical Chemistry Faculty of Pharmacy University of Malaya Kuala Lumpur 50603 Malaysia
- School of Biosciences Faculty of Health and Medical Sciences Taylor's University Subang Jaya Selangor 47500 Malaysia
| | - Lip Yong Chung
- Department of Pharmaceutical Chemistry Faculty of Pharmacy University of Malaya Kuala Lumpur 50603 Malaysia
| |
Collapse
|
27
|
Skubalova Z, Rex S, Sukupova M, Zahalka M, Skladal P, Pribyl J, Michalkova H, Weerasekera A, Adam V, Heger Z. Passive Diffusion vs Active pH-Dependent Encapsulation of Tyrosine Kinase Inhibitors Vandetanib and Lenvatinib into Folate-Targeted Ferritin Delivery System. Int J Nanomedicine 2021; 16:1-14. [PMID: 33442247 PMCID: PMC7797358 DOI: 10.2147/ijn.s275808] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/07/2020] [Indexed: 12/12/2022] Open
Abstract
Introduction The present study reports on examination of the effects of encapsulating the tyrosine kinase inhibitors (TKIs) vandetanib and lenvatinib into a biomacromolecular ferritin-based delivery system. Methods The encapsulation of TKIs was performed via two strategies: i) using an active reversible pH-dependent reassembly of ferritin´s quaternary structure and ii) passive loading of hydrophobic TKIs through the hydrophobic channels at the junctions of ferritin subunits. After encapsulation, ferritins were surface-functionalized with folic acid promoting active-targeting capabilities. Results The physico-chemical and nanomechanical analyses revealed that despite the comparable encapsulation efficiencies of both protocols, the active loading affects stability and rigidity of ferritins, plausibly due to their imperfect reassembly. Biological experiments with hormone-responsive breast cancer cells (T47-D and MCF-7) confirmed the cytotoxicity of encapsulated and folate-targeted TKIs to folate-receptor positive cancer cells, but only limited cytotoxic effects to healthy breast epithelium. Importantly, the long-term cytotoxic experiments revealed that compared to the pH-dependent encapsulation, the passively-loaded TKIs exert markedly higher anticancer activity, most likely due to undesired influence of harsh acidic environment used for the pH-dependent encapsulation on the TKIs’ structural and functional properties. Conclusion Since the passive loading does not require a reassembly step for which acids are needed, the presented investigation serves as a solid basis for future studies focused on encapsulation of small hydrophobic molecules.
Collapse
Affiliation(s)
- Zuzana Skubalova
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic.,Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Simona Rex
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic.,Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Martina Sukupova
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic
| | - Martin Zahalka
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic
| | - Petr Skladal
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Jan Pribyl
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Hana Michalkova
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic.,Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Akila Weerasekera
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic.,Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Zbynek Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic.,Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| |
Collapse
|
28
|
Kumari R, Sunil D. A mechanistic insight into benefits of aggregation induced emissive luminogens in cancer. J Drug Target 2021; 29:592-608. [PMID: 33399029 DOI: 10.1080/1061186x.2020.1868479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Exploration of advanced chemotheranostics that benefit from a combined in vivo strategy of cancer diagnosis and chemotherapy simultaneously is highly valued and will expose novel possibilities in modifying treatment and reduce side effects. In recent years, nanodrug delivery systems that incorporate aggregation-induced emissive luminogens (AIEgens) have been developed to track and monitor anticancer drug release, trace translocation processes and predict chemotherapeutic responses. There are several classes of AIEgen based chemotheranostics such us stimuli-responsive nanoprodrugs, pH-sensitive mesoporous silica nanocarriers, supramolecular polymer systems, drug encapsulated carriers, carrier-free nanodrugs, self-indicating drug delivery nanomachines and AIEgen-prodrug co-assembly. The present review conveys mechanistic insight into the benefits of AIEgens in the theranostic application by illustrating the recent breakthroughs in chemotheranostic nanomedicines that incorporate these unique fluorophores as signal reporters. The perspectives that can be further explored are also highlighted with the hope to instil more research interest in the advancement of AIE active cancer chemotheranostics for imaging and treatment in vivo.HIGHLIGHTSAggregation induced emissive materials (AIEgens) exhibit unique advantages over conventional luminogens for synergistic diagnosis and chemotherapy of cancer in vivo.The combination of AIE and nanotechnology offers an excellent platform to fabricate advanced chemotheranostics for cancer therapy.
Collapse
Affiliation(s)
- Rashmi Kumari
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, India
| | - Dhanya Sunil
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
29
|
Dutta K, Kanjilal P, Das R, Thayumanavan S. Synergistic Interplay of Covalent and Non‐Covalent Interactions in Reactive Polymer Nanoassembly Facilitates Intracellular Delivery of Antibodies. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202010412] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
| | | | - Ritam Das
- University of Massachusetts Amherst MA 01003 USA
| | - Sankaran Thayumanavan
- Department of Chemistry, Molecular and Cellular Biology Program, and The Center for Bioactive Delivery-Institute for Applied Life Sciences University of Massachusetts Amherst MA 01003 USA
| |
Collapse
|
30
|
Luong HX, Thanh TT, Tran TH. Antimicrobial peptides - Advances in development of therapeutic applications. Life Sci 2020; 260:118407. [PMID: 32931796 PMCID: PMC7486823 DOI: 10.1016/j.lfs.2020.118407] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/02/2020] [Accepted: 09/07/2020] [Indexed: 02/07/2023]
Abstract
The severe infection is becoming a significant health problem which threaten the lives of patients and the safety and economy of society. In the way of finding new strategy, antimicrobial peptides (AMPs) - an important part of host defense family, emerged with tremendous potential. Up to date, huge numbers of AMPs has been investigated from both natural and synthetic sources showing not only the ability to kill microbial pathogens but also propose other benefits such as wound healing, anti-tumor, immune modulation. In this review, we describe the involvements of AMPs in biological systems and discuss the opportunity in developing AMPs for clinical applications. In the detail, their properties in antibacterial activity is followed by their application in some infection diseases and cancer. The key discussions are the approaches to improve biological activities of AMPs either by modifying chemical structure or incorporating into delivery systems. The new applications and perspectives for the future of AMPs would open the new era of their development.
Collapse
Affiliation(s)
- Huy Xuan Luong
- Faculty of Pharmacy, PHENIKAA University, Yen Nghia, Ha Dong, Hanoi 12116, Viet Nam; PHENIKAA Institute for Advanced Study (PIAS), PHENIKAA University, Hanoi 12116, Viet Nam.
| | - Tung Truong Thanh
- Faculty of Pharmacy, PHENIKAA University, Yen Nghia, Ha Dong, Hanoi 12116, Viet Nam; PHENIKAA Institute for Advanced Study (PIAS), PHENIKAA University, Hanoi 12116, Viet Nam.
| | - Tuan Hiep Tran
- Faculty of Pharmacy, PHENIKAA University, Yen Nghia, Ha Dong, Hanoi 12116, Viet Nam; PHENIKAA Research and Technology Institute (PRATI), A&A Green Phoenix Group JSC, No.167 Hoang Ngan, Trung Hoa, Cau Giay, Hanoi 11313, Viet Nam.
| |
Collapse
|
31
|
Zhang B, Napoleon JV, Liu X, Luo Q, Srinivasarao M, Low PS. Sensitive manipulation of CAR T cell activity using a chimeric endocytosing receptor. J Immunother Cancer 2020; 8:jitc-2020-000756. [PMID: 33127654 PMCID: PMC7604868 DOI: 10.1136/jitc-2020-000756] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/18/2020] [Indexed: 01/22/2023] Open
Abstract
Background Most adoptive cell therapies (ACTs) suffer from an inability to control the therapeutic cell’s behavior following its transplantation into a patient. Thus, efforts to inhibit, activate, differentiate or terminate an ACT after patient reinfusion can be futile, because the required drug adversely affects other cells in the patient. Methods We describe here a two domain fusion receptor composed of a ligand-binding domain linked to a recycling domain that allows constitutive internalization and trafficking of the fusion receptor back to the cell surface. Because the ligand-binding domain is designed to bind a ligand not normally present in humans, any drug conjugated to this ligand will bind and endocytose selectively into the ACT. Results In two embodiments of our strategy, we fuse the chronically endocytosing domain of human folate receptor alpha to either a murine scFv that binds fluorescein or human FK506 binding protein that binds FK506, thereby creating a fusion receptor composed of largely human components. We then create the ligand-targeted drug by conjugating any desired drug to either fluorescein or FK506, thereby generating a ligand-drug conjugate with ~10-9 M affinity for its fusion receptor. Using these tools, we demonstrate that CAR T cell activities can be sensitively tuned down or turned off in vitro as well as tightly controlled following their reinfusion into tumor-bearing mice. Conclusions We suggest this ‘chimeric endocytosing receptor’ can be exploited to manipulate not only CAR T cells but other ACTs following their reinfusion into patients. With efforts to develop ACTs to treat diseases including diabetes, heart failure, osteoarthritis, cancer and sickle cell anemia accelerating, we argue an ability to manipulate ACT activities postinfusion will be important.
Collapse
Affiliation(s)
- Boning Zhang
- Chemistry, Purdue University System, West Lafayette, Indiana, USA
| | | | - Xin Liu
- Chemistry, Purdue University System, West Lafayette, Indiana, USA
| | - Qian Luo
- Chemistry, Purdue University System, West Lafayette, Indiana, USA
| | | | - Philip S Low
- Chemistry, Purdue University System, West Lafayette, Indiana, USA
| |
Collapse
|
32
|
Wu Y, Tam WS, Chau HF, Kaur S, Thor W, Aik WS, Chan WL, Zweckstetter M, Wong KL. Solid-phase fluorescent BODIPY-peptide synthesis via in situ dipyrrin construction. Chem Sci 2020; 11:11266-11273. [PMID: 34094367 PMCID: PMC8162834 DOI: 10.1039/d0sc04849f] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 09/23/2020] [Indexed: 12/28/2022] Open
Abstract
Traditional fluorescent peptide chemical syntheses hinge on the use of limited fluorescent/dye-taggable unnatural amino acids and entail multiple costly purifications. Here we describe a facile and efficient protocol for in situ construction of dipyrrins on the N-terminus with 20 natural and five unnatural amino acids and the lysine's side chain of selected peptides/peptide drugs through Fmoc-based solid-phase peptide synthesis. The new strategy enables the direct formation of boron-dipyrromethene (BODIPY)-peptide conjugates from simple aldehyde and pyrrole derivatives without pre-functionalization, and only requires a single-time chromatographic purification at the final stage. As a model study, synthesized EBNA1-targeting BODIPY1-Pep4 demonstrates intact selectivity in vitro, responsive fluorescence enhancement, and higher light cytotoxicity due to the photo-generation of cytotoxic singlet oxygen. This work offers a novel practical synthetic platform for fluorescent peptides for multifaceted biomedical applications.
Collapse
Affiliation(s)
- Yue Wu
- Department of Chemistry, Hong Kong Baptist University Kowloon Hong Kong SAR China
| | - Wing-Sze Tam
- Department of Chemistry, Hong Kong Baptist University Kowloon Hong Kong SAR China
| | - Ho-Fai Chau
- Department of Chemistry, Hong Kong Baptist University Kowloon Hong Kong SAR China
| | - Simranjeet Kaur
- Department of Chemistry, Hong Kong Baptist University Kowloon Hong Kong SAR China
| | - Waygen Thor
- Department of Chemistry, Hong Kong Baptist University Kowloon Hong Kong SAR China
| | - Wei Shen Aik
- Department of Chemistry, Hong Kong Baptist University Kowloon Hong Kong SAR China
| | - Wai-Lun Chan
- Department of Chemistry, Hong Kong Baptist University Kowloon Hong Kong SAR China
- Department for NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry Am Fassberg 11 37077 Göttingen Germany
- German Center for Neurodegenerative Diseases (DZNE) Von-Siebold-Str. 3a 37075 Göttingen Germany
| | - Markus Zweckstetter
- Department for NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry Am Fassberg 11 37077 Göttingen Germany
- German Center for Neurodegenerative Diseases (DZNE) Von-Siebold-Str. 3a 37075 Göttingen Germany
| | - Ka-Leung Wong
- Department of Chemistry, Hong Kong Baptist University Kowloon Hong Kong SAR China
| |
Collapse
|
33
|
Zhou Y, Mowlazadeh Haghighi S, Liu Z, Wang L, Hruby VJ, Cai M. Development of Ligand-Drug Conjugates Targeting Melanoma through the Overexpressed Melanocortin 1 Receptor. ACS Pharmacol Transl Sci 2020; 3:921-930. [PMID: 33073191 DOI: 10.1021/acsptsci.0c00072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Indexed: 12/31/2022]
Abstract
Melanoma is a lethal form of skin cancer. Despite recent breakthroughs of BRAF-V600E and PD-1 inhibitors showing remarkable clinical responses, melanoma can eventually survive these targeted therapies and become resistant. To solve the drug resistance issue, we designed and synthesized ligand-drug conjugates that couple cytotoxic drugs, which have a low cancer resistance issue, with the melanocortin 1 receptor (MC1R) agonist melanotan-II (MT-II), which provides specificity to MC1R-overexpressing melanoma. The drug-MT-II conjugates maintain strong binding interactions to MC1R and induce selective drug delivery to A375 melanoma cells through its MT-II moiety in vitro. Furthermore, using camptothecin as the cytotoxic drug, camptothecin-MT-II (compound 1) can effectively inhibit A375 melanoma cell growth with an IC50 of 16 nM. By providing selectivity to melanoma cells through its MT-II moiety, this approach of drug-MT-II conjugates enables us to have many more options for cytotoxic drug selection, which can be the key to solving the cancer resistant problem for melanoma.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Saghar Mowlazadeh Haghighi
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Zekun Liu
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Lingzhi Wang
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Victor J Hruby
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Minying Cai
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
34
|
Zhang F, Ayaub EA, Wang B, Puchulu‐Campanella E, Li Y, Hettiarachchi SU, Lindeman SD, Luo Q, Rout S, Srinivasarao M, Cox A, Tsoyi K, Nickerson‐Nutter C, Rosas IO, Low PS. Reprogramming of profibrotic macrophages for treatment of bleomycin-induced pulmonary fibrosis. EMBO Mol Med 2020; 12:e12034. [PMID: 32597014 PMCID: PMC7411553 DOI: 10.15252/emmm.202012034] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 05/30/2020] [Accepted: 06/03/2020] [Indexed: 12/19/2022] Open
Abstract
Fibrotic diseases cause organ failure that lead to ~45% of all deaths in the United States. Activated macrophages stimulate fibrosis by secreting cytokines that induce fibroblasts to synthesize collagen and extracellular matrix proteins. Although suppression of macrophage-derived cytokine production can halt progression of fibrosis, therapeutic agents that prevent release of these cytokines (e.g., TLR7 agonists) have proven too toxic to administer systemically. Based on the expression of folate receptor β solely on activated myeloid cells, we have created a folate-targeted TLR7 agonist (FA-TLR7-54) that selectively accumulates in profibrotic macrophages and suppresses fibrosis-inducing cytokine production. We demonstrate that FA-TLR7-54 reprograms M2-like fibrosis-inducing macrophages into fibrosis-suppressing macrophages, resulting in dramatic declines in profibrotic cytokine release, hydroxyproline biosynthesis, and collagen deposition, with concomitant increases in alveolar airspaces. Although nontargeted TLR7-54 is lethal at fibrosis-suppressing doses, FA-TLR7-54 halts fibrosis without evidence of toxicity. Taken together, FA-TLR7-54 is shown to constitute a novel and potent approach for treating fibrosis without causing dose-limiting systemic toxicities.
Collapse
Affiliation(s)
- Fenghua Zhang
- Department of Chemistry and Institute for Drug DiscoveryPurdue UniversityWest LafayetteINUSA
| | - Ehab A Ayaub
- Division of Pulmonary and Critical Care MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Bingbing Wang
- Department of Chemistry and Institute for Drug DiscoveryPurdue UniversityWest LafayetteINUSA
| | | | - Yen‐Hsing Li
- Department of Chemistry and Institute for Drug DiscoveryPurdue UniversityWest LafayetteINUSA
| | - Suraj U Hettiarachchi
- Department of Chemistry and Institute for Drug DiscoveryPurdue UniversityWest LafayetteINUSA
| | - Spencer D Lindeman
- Department of Chemistry and Institute for Drug DiscoveryPurdue UniversityWest LafayetteINUSA
| | - Qian Luo
- Department of Chemistry and Institute for Drug DiscoveryPurdue UniversityWest LafayetteINUSA
| | - Sasmita Rout
- Department of Chemistry and Institute for Drug DiscoveryPurdue UniversityWest LafayetteINUSA
| | - Madduri Srinivasarao
- Department of Chemistry and Institute for Drug DiscoveryPurdue UniversityWest LafayetteINUSA
| | - Abigail Cox
- Department of Comparative PathobiologyPurdue College of Veterinary MedicineWest LafayetteINUSA
| | - Konstantin Tsoyi
- Division of Pulmonary and Critical Care MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | | | - Ivan O Rosas
- Division of Pulmonary and Critical Care MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Philip S Low
- Department of Chemistry and Institute for Drug DiscoveryPurdue UniversityWest LafayetteINUSA
| |
Collapse
|
35
|
Bi X, Yin J, Zhang D, Zhang X, Balamkundu S, Lescar J, Dedon PC, Tam JP, Liu CF. Tagging Transferrin Receptor with a Disulfide FRET Probe To Gauge the Redox State in Endosomal Compartments. Anal Chem 2020; 92:12460-12466. [DOI: 10.1021/acs.analchem.0c02264] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Xiaobao Bi
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Juan Yin
- Program in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, 8 College Road, Singapore169857, Singapore
| | - Dingpeng Zhang
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Xiaohong Zhang
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Seetharamsing Balamkundu
- Singapore-MIT Alliance for Research and Technology Centre, 1 Create Way, #10-01 Create Tower, Singapore 138602, Singapore
| | - Julien Lescar
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Peter C. Dedon
- Singapore-MIT Alliance for Research and Technology Centre, 1 Create Way, #10-01 Create Tower, Singapore 138602, Singapore
| | - James P. Tam
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Chuan-Fa Liu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| |
Collapse
|
36
|
Tanaka H, Sakurai Y, Anindita J, Akita H. Development of lipid-like materials for RNA delivery based on intracellular environment-responsive membrane destabilization and spontaneous collapse. Adv Drug Deliv Rev 2020; 154-155:210-226. [PMID: 32650040 DOI: 10.1016/j.addr.2020.07.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/26/2020] [Accepted: 07/02/2020] [Indexed: 01/01/2023]
Abstract
Messenger RNA and small interfering RNA are attractive modalities for curing diseases by complementation or knock-down of proteins. For success of these RNAs, a drug delivery system (DDS) is required to control a pharmacokinetics, to enhance cellular uptake, to overcome biological membranes, and to release the cargo into the cytoplasm. Based on past research, developing nanoparticles that are neutrally charged have been the mainstream of their development. Also, the materials are further mounted with pH- and/or reducing environment-responsive units. In this review, we summarize progress made in the molecular design of these materials. We also focus on the importance of the hydrophobic scaffold for tissue/cell targeting, intracellular trafficking, and immune responses. As a practical example, the design concept of the SS-cleavable and pH-activated lipid-like material (ssPalm) and subsequent molecular modification tailored to the RNA-based medical application is discussed.
Collapse
|
37
|
Xie J, Gonzalez-Carter D, Tockary TA, Nakamura N, Xue Y, Nakakido M, Akiba H, Dirisala A, Liu X, Toh K, Yang T, Wang Z, Fukushima S, Li J, Quader S, Tsumoto K, Yokota T, Anraku Y, Kataoka K. Dual-Sensitive Nanomicelles Enhancing Systemic Delivery of Therapeutically Active Antibodies Specifically into the Brain. ACS NANO 2020; 14:6729-6742. [PMID: 32431145 DOI: 10.1021/acsnano.9b09991] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Delivering therapeutic antibodies into the brain across the blood-brain barrier at a therapeutic level is a promising while challenging approach in the treatment of neurological disorders. Here, we present a polymeric nanomicelle (PM) system capable of delivering therapeutically effective levels of 3D6 antibody fragments (3D6-Fab) into the brain parenchyma for inhibiting Aβ aggregation. PM assembly was achieved by charge-converting 3D6-Fab through pH-sensitive citraconylation to allow complexation with reductive-sensitive cationic polymers. Brain targeting was achieved by functionalizing the PM surface with glucose molecules to allow interaction with recycling glucose transporter (Glut)-1 proteins. Consequently, 41-fold enhanced 3D6-Fab accumulation in the brain was achieved by using the PM system compared to free 3D6-Fab. Furthermore, therapeutic benefits were obtained by successfully inhibiting Aβ1-42 aggregation in Alzheimer's disease mice systemically treated with 3D6-Fab-loaded glucosylated PM. Hence, this nanocarrier system represents a promising method for effectively delivering functional antibody agents into the brain and treating neurological diseases.
Collapse
Affiliation(s)
- Jinbing Xie
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
- Jiangsu Key Laboratory of Molecular Imaging and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing 210009, China
| | - Daniel Gonzalez-Carter
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Theofilus A Tockary
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Noriko Nakamura
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Yonger Xue
- Department of Pharmacy, Shanghai Jiaotong University, 800 Dongchun Road, Shanghai 200240, China
| | - Makoto Nakakido
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Hiroki Akiba
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Anjaneyulu Dirisala
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Xueying Liu
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Kazuko Toh
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Tao Yang
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Zengtao Wang
- Department of Pharmacy, Shanghai Jiaotong University, 800 Dongchun Road, Shanghai 200240, China
| | - Shigeto Fukushima
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Junjie Li
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Sabina Quader
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Kouhei Tsumoto
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Takanori Yokota
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Yasutaka Anraku
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
- Institute for Future Initiatives, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
38
|
Weng C, Fan N, Xu T, Chen H, Li Z, Li Y, Tan H, Fu Q, Ding M. FRET-based polymer materials for detection of cellular microenvironments. CHINESE CHEM LETT 2020. [DOI: 10.1016/j.cclet.2019.11.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
39
|
Numasawa K, Hanaoka K, Saito N, Yamaguchi Y, Ikeno T, Echizen H, Yasunaga M, Komatsu T, Ueno T, Miura M, Nagano T, Urano Y. A Fluorescent Probe for Rapid, High‐Contrast Visualization of Folate‐Receptor‐Expressing Tumors In Vivo. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201914826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Koji Numasawa
- Graduate School of Pharmaceutical SciencesThe University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| | - Kenjiro Hanaoka
- Graduate School of Pharmaceutical SciencesThe University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| | - Naoko Saito
- Graduate School of Pharmaceutical SciencesThe University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| | - Yoshifumi Yamaguchi
- Institute of Low Temperature ScienceHokkaido University Sapporo 060-0819 Japan
| | - Takayuki Ikeno
- Graduate School of Pharmaceutical SciencesThe University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| | - Honami Echizen
- Graduate School of Pharmaceutical SciencesThe University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| | - Masahiro Yasunaga
- Division of Developmental TherapeuticsExploratory Oncology Research & Clinical Trial CenterNational Cancer Center 6-5-1 Kashiwanoha, Kashiwa Chiba 277-8577 Japan
| | - Toru Komatsu
- Graduate School of Pharmaceutical SciencesThe University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| | - Tasuku Ueno
- Graduate School of Pharmaceutical SciencesThe University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| | - Masayuki Miura
- Graduate School of Pharmaceutical SciencesThe University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| | - Tetsuo Nagano
- Drug Discovery InitiativeThe University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| | - Yasuteru Urano
- Graduate School of Pharmaceutical SciencesThe University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
- Graduate School of MedicineThe University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
- CREST (Japan) Agency for Medical Research and Development (AMED) 1-7-1 Otemachi, Chiyoda-ku Tokyo 100-0004 Japan
| |
Collapse
|
40
|
Numasawa K, Hanaoka K, Saito N, Yamaguchi Y, Ikeno T, Echizen H, Yasunaga M, Komatsu T, Ueno T, Miura M, Nagano T, Urano Y. A Fluorescent Probe for Rapid, High‐Contrast Visualization of Folate‐Receptor‐Expressing Tumors In Vivo. Angew Chem Int Ed Engl 2020; 59:6015-6020. [DOI: 10.1002/anie.201914826] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/30/2019] [Indexed: 01/23/2023]
Affiliation(s)
- Koji Numasawa
- Graduate School of Pharmaceutical SciencesThe University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| | - Kenjiro Hanaoka
- Graduate School of Pharmaceutical SciencesThe University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| | - Naoko Saito
- Graduate School of Pharmaceutical SciencesThe University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| | - Yoshifumi Yamaguchi
- Institute of Low Temperature ScienceHokkaido University Sapporo 060-0819 Japan
| | - Takayuki Ikeno
- Graduate School of Pharmaceutical SciencesThe University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| | - Honami Echizen
- Graduate School of Pharmaceutical SciencesThe University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| | - Masahiro Yasunaga
- Division of Developmental TherapeuticsExploratory Oncology Research & Clinical Trial CenterNational Cancer Center 6-5-1 Kashiwanoha, Kashiwa Chiba 277-8577 Japan
| | - Toru Komatsu
- Graduate School of Pharmaceutical SciencesThe University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| | - Tasuku Ueno
- Graduate School of Pharmaceutical SciencesThe University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| | - Masayuki Miura
- Graduate School of Pharmaceutical SciencesThe University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| | - Tetsuo Nagano
- Drug Discovery InitiativeThe University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| | - Yasuteru Urano
- Graduate School of Pharmaceutical SciencesThe University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
- Graduate School of MedicineThe University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
- CREST (Japan) Agency for Medical Research and Development (AMED) 1-7-1 Otemachi, Chiyoda-ku Tokyo 100-0004 Japan
| |
Collapse
|
41
|
Yang Q, Deng Z, Wang D, He J, Zhang D, Tan Y, Peng T, Wang XQ, Tan W. Conjugating Aptamer and Mitomycin C with Reductant-Responsive Linker Leading to Synergistically Enhanced Anticancer Effect. J Am Chem Soc 2020; 142:2532-2540. [PMID: 31910340 DOI: 10.1021/jacs.9b12409] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mitomycin C (MMC) has been using for the treatment of a variety of digestive tract cancers. However, its nonspecific DNA-alkylating ability usually causes severe side effects, thus largely limiting its clinical applications. The utilization of an efficient active targeted drug delivery technique would address this issue. Accordingly, we report the design and development of aptamer-mitomycin C conjugates that use different cross-linking chemistry. The targeted delivery ability and cytotoxicity of these conjugates were carefully studied. It is worth noting that a linker-dependent cytotoxicity effect was observed for these conjugates. The use of a reductant-sensitive disulfide bond cross-linking strategy resulted in significantly enhanced cytotoxicity of MMC against the target cancer cell lines. Importantly, this cytotoxicity enhancement was suited to different types of aptamers, demonstrating the success of our design. Mechanistic studies of the enhanced cytotoxicity effect indicated that the target recognition, specific binding, and receptor-mediated internalization of aptamer were also critical for the observed effect.
Collapse
Affiliation(s)
- Qiuxia Yang
- Molecular Sciences and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering and College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine , Hunan University , Changsha 410082 , China
| | - Zhengyu Deng
- Molecular Sciences and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering and College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine , Hunan University , Changsha 410082 , China
| | - Dan Wang
- Molecular Sciences and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering and College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine , Hunan University , Changsha 410082 , China
| | - Jiaxuan He
- Molecular Sciences and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering and College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine , Hunan University , Changsha 410082 , China
| | - Dailiang Zhang
- Molecular Sciences and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering and College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine , Hunan University , Changsha 410082 , China
| | - Yan Tan
- Molecular Sciences and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering and College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine , Hunan University , Changsha 410082 , China
| | - Tianhuan Peng
- Molecular Sciences and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering and College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine , Hunan University , Changsha 410082 , China
| | - Xue-Qiang Wang
- Molecular Sciences and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering and College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine , Hunan University , Changsha 410082 , China
| | - Weihong Tan
- Molecular Sciences and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering and College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine , Hunan University , Changsha 410082 , China.,Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, College of Chemistry and Chemical Engineering , Shanghai Jiao Tong University , Shanghai 200127 , China.,Institute of Cancer and Basic Medicine (IBMC) , Chinese Academy of Sciences; The Cancer Hospital of the University of Chinese Academy of Sciences , Hangzhou , Zhejiang 310022 , China
| |
Collapse
|
42
|
Böttger R, Pauli G, Chao PH, AL Fayez N, Hohenwarter L, Li SD. Lipid-based nanoparticle technologies for liver targeting. Adv Drug Deliv Rev 2020; 154-155:79-101. [PMID: 32574575 DOI: 10.1016/j.addr.2020.06.017] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/26/2020] [Accepted: 06/16/2020] [Indexed: 12/18/2022]
Abstract
Liver diseases such as hepatitis, cirrhosis, and hepatocellular carcinoma are global health problems accounting for approximately 800 million cases and over 2 million deaths per year worldwide. Major drawbacks of standard pharmacological therapies are the inability to deliver a sufficient concentration of a therapeutic agent to the diseased liver, and nonspecific drug delivery leading to undesirable systemic side effects. Additionally, depending on the specific liver disease, drug delivery to a subset of liver cells is required. In recent years, lipid nanoparticles have been developed to passively and actively target drugs to the liver. The success of this approach has been highlighted by the FDA-approval of the first liver-targeting lipid nanoparticle, ONPATTRO, in 2018 and many other promising candidate technologies are expected to follow. This review summarizes recent developments of various lipid-based liver-targeting technologies, namely solid-lipid nanoparticles, liposomes, niosomes and micelles, and discusses the challenges and future perspectives in this field.
Collapse
|
43
|
Stasińska AR, Putaj P, Chmielewski MK. Disulfide bridge as a linker in nucleic acids' bioconjugation. Part II: A summary of practical applications. Bioorg Chem 2019; 95:103518. [PMID: 31911308 DOI: 10.1016/j.bioorg.2019.103518] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/11/2019] [Accepted: 12/16/2019] [Indexed: 12/15/2022]
Abstract
Disulfide conjugation invariably remains a key tool in research on nucleic acids. This versatile and cost-effective method plays a crucial role in structural studies of DNA and RNA as well as their interactions with other macromolecules in a variety of biological systems. In this article we review applications of disulfide-bridged conjugates of oligonucleotides with other (bio)molecules such as peptides, proteins etc. and present key findings obtained with their help.
Collapse
Affiliation(s)
- Anna R Stasińska
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, ul. Noskowskiego 12/14, 61-704 Poznań, Poland; FutureSynthesis sp. z o.o. ul. Rubież 46H, 61-612 Poznań, Poland
| | - Piotr Putaj
- FutureSynthesis sp. z o.o. ul. Rubież 46H, 61-612 Poznań, Poland
| | - Marcin K Chmielewski
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, ul. Noskowskiego 12/14, 61-704 Poznań, Poland; FutureSynthesis sp. z o.o. ul. Rubież 46H, 61-612 Poznań, Poland.
| |
Collapse
|
44
|
Scales SJ, Tsai SP, Zacharias N, Cruz-Chuh JD, Bullen G, Velasquez E, Chang J, Bruguera E, Kozak KR, Sadowsky J. Development of a Cysteine-Conjugatable Disulfide FRET Probe: Influence of Charge on Linker Cleavage and Payload Trafficking for an Anti-HER2 Antibody Conjugate. Bioconjug Chem 2019; 30:3046-3056. [PMID: 31726009 DOI: 10.1021/acs.bioconjchem.9b00678] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Disulfide-linked bioconjugates allow the delivery of pharmacologically active or other cargo to specific tissues in a redox-sensitive fashion. However, an understanding of the kinetics, subcellular distribution, and mechanism of disulfide cleavage in such bioconjugates is generally lacking. Here, we report a modular disulfide-linked TAMRA-BODIPY based FRET probe that can be readily synthesized, modified, and conjugated to a cysteine-containing biomolecule to enable real-time monitoring of disulfide cleavage during receptor-mediated endocytosis in cells. We demonstrate the utility of this probe to study disulfide reduction during HER2 receptor-mediated uptake of a Cys-engineered anti-HER2 THIOMAB antibody. We found that introduction of positive, but not negative, charges in the probe improved retention of the BODIPY catabolite. This permitted the observation of significant disulfide cleavage in endosomes or lysosomes on par with proteolytic cleavage of a similarly charged valine-citrulline peptide-based probe. In general, the FRET probe we describe should enable real-time cellular monitoring of disulfide cleavage in other targeted delivery systems for mechanistic or diagnostic applications. Furthermore, modifications to the released BODIPY moiety permit evaluation of physicochemical properties that govern lysosomal egress or retention, which may have implications for the development of next-generation antibody-drug conjugates.
Collapse
Affiliation(s)
- Suzie J Scales
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Siao Ping Tsai
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Neelie Zacharias
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Josefa Dela Cruz-Chuh
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Gordy Bullen
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Erick Velasquez
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Julie Chang
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Elise Bruguera
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Katherine R Kozak
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Jack Sadowsky
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| |
Collapse
|
45
|
Zhang R, Qin X, Kong F, Chen P, Pan G. Improving cellular uptake of therapeutic entities through interaction with components of cell membrane. Drug Deliv 2019; 26:328-342. [PMID: 30905189 PMCID: PMC6442206 DOI: 10.1080/10717544.2019.1582730] [Citation(s) in RCA: 183] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/08/2019] [Accepted: 02/11/2019] [Indexed: 12/24/2022] Open
Abstract
Efficient cellular delivery of biologically active molecules is one of the key factors that affect the discovery and development of novel drugs. The plasma membrane is the first barrier that prevents direct translocation of chemic entities, and thus obstructs their efficient intracellular delivery. Generally, hydrophilic small molecule drugs are poor permeability that reduce bioavailability and thus limit the clinic application. The cellular uptake of macromolecules and drug carriers is very inefficient without external assistance. Therefore, it is desirable to develop potent delivery systems for achieving effective intracellular delivery of chemic entities. Apart from of the types of delivery strategies, the composition of the cell membrane is critical for delivery efficiency due to the fact that cellular uptake is affected by the interaction between the chemical entity and the plasma membrane. In this review, we aimed to develop a profound understanding of the interactions between delivery systems and components of the plasma membrane. For the purpose, we attempt to present a broad overview of what delivery systems can be used to enhance the intracellular delivery of poorly permeable chemic entities, and how various delivery strategies are applied according to the components of plasma membrane.
Collapse
Affiliation(s)
- Renshuai Zhang
- Key Laboratory of Flexible Electronics & Institute of Advanced Materials, Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing, P.R. China
| | - Xiaofei Qin
- Key Laboratory of Flexible Electronics & Institute of Advanced Materials, Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing, P.R. China
| | - Fandong Kong
- Key Laboratory of Biology and Genetic Resources of Tropical Crops, Ministry of Agriculture, Institute of Tropical Bioscience and Biotechnology Chinese Academy of Tropical Agriculture Sciences, Haikou, P.R. China
| | - Pengwei Chen
- Key Laboratory of Biology and Genetic Resources of Tropical Crops, Ministry of Agriculture, Institute of Tropical Bioscience and Biotechnology Chinese Academy of Tropical Agriculture Sciences, Haikou, P.R. China
| | - Guojun Pan
- School of Life Sciences, Taishan Medical University, Tai’an, P.R. China
| |
Collapse
|
46
|
Varini K, Lécorché P, Sonnette R, Gassiot F, Broc B, Godard M, David M, Faucon A, Abouzid K, Ferracci G, Temsamani J, Khrestchatisky M, Jacquot G. Target engagement and intracellular delivery of mono- and bivalent LDL receptor-binding peptide-cargo conjugates: Implications for the rational design of new targeted drug therapies. J Control Release 2019; 314:141-161. [PMID: 31644939 DOI: 10.1016/j.jconrel.2019.10.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/14/2019] [Accepted: 10/17/2019] [Indexed: 10/25/2022]
Abstract
Targeted delivery to specific tissues and subcellular compartments is of paramount importance to optimize therapeutic or diagnostic interventions while minimizing side-effects. Using recently identified LDL receptor (LDLR) -targeting small synthetic peptide-vectors conjugated to model cargos of different nature and size, we investigated in LDLR-expressing cells the impact of vector-cargo molecular engineering and coupling valency, as well as the cellular exposure duration on their target engagement and intracellular trafficking and delivery profiles. All vector-cargo conjugates evaluated were found to be delivered to late compartments together with the natural ligand LDL, although to varying extents and with different kinetics. Partial recycling together with the LDLR was also consistently observed. Under continuous cellular exposure, the extent of intracellular vector-cargo delivery primarily relies on their endosomal unloading potential. In this condition, the highest intracellular delivery potential was observed with a monovalent conjugate displaying a rather high LDLR dissociation rate. On the contrary, under transient cellular exposure followed by chase, low dissociation-rate bivalent conjugates revealed a higher intracellular delivery potential than the monovalent conjugate. This was shown to rely on their ability to undergo multiple endocytosis-recycling rounds, with limited release in the ligand-free medium. The absence of reciprocal competition with the natural ligand LDL on their respective intracellular trafficking was also demonstrated, which is essential in terms of potential safety liabilities. These results demonstrate that not only molecular engineering of new therapeutic conjugates of interest, but also the cellular exposure mode used during in vitro evaluations are critical to anticipate and optimize their delivery potential.
Collapse
Affiliation(s)
- K Varini
- VECT-HORUS SAS, Marseille, France; Aix-Marseille Univ., CNRS, INP, Inst. Neurophysiopathol., Marseille, France
| | | | | | | | - B Broc
- VECT-HORUS SAS, Marseille, France
| | - M Godard
- VECT-HORUS SAS, Marseille, France
| | - M David
- VECT-HORUS SAS, Marseille, France
| | - A Faucon
- VECT-HORUS SAS, Marseille, France
| | | | - G Ferracci
- Aix-Marseille Univ., CNRS, INP, Inst. Neurophysiopathol., Marseille, France
| | | | - M Khrestchatisky
- Aix-Marseille Univ., CNRS, INP, Inst. Neurophysiopathol., Marseille, France
| | | |
Collapse
|
47
|
Stasińska AR, Putaj P, Chmielewski MK. Disulfide bridge as a linker in nucleic acids’ bioconjugation. Part I: An overview of synthetic strategies. Bioorg Chem 2019; 92:103223. [DOI: 10.1016/j.bioorg.2019.103223] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/26/2019] [Accepted: 08/26/2019] [Indexed: 12/23/2022]
|
48
|
Sefrin JP, Hillringhaus L, Mundigl O, Mann K, Ziegler-Landesberger D, Seul H, Tabares G, Knoblauch D, Leinenbach A, Friligou I, Dziadek S, Offringa R, Lifke V, Lifke A. Sensitization of Tumors for Attack by Virus-Specific CD8+ T-Cells Through Antibody-Mediated Delivery of Immunogenic T-Cell Epitopes. Front Immunol 2019; 10:1962. [PMID: 31555260 PMCID: PMC6712545 DOI: 10.3389/fimmu.2019.01962] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/05/2019] [Indexed: 01/22/2023] Open
Abstract
Anti-tumor immunity is limited by a number of factors including the lack of fully activated T-cells, insufficient antigenic stimulation and the immune-suppressive tumor microenvironment. We addressed these hurdles by developing a novel class of immunoconjugates, Antibody-Targeted Pathogen-derived Peptides (ATPPs), which were designed to efficiently deliver viral T-cell epitopes to tumors with the aim of redirecting virus-specific memory T-cells against the tumor. ATPPs were generated through covalent binding of mature MHC class I peptides to antibodies specific for cell surface-expressed tumor antigens that mediate immunoconjugate internalization. By means of a cleavable linker, the peptides are released in the endosomal compartment, from which they are loaded into MHC class I without the need for further processing. Pulsing of tumor cells with ATPPs was found to sensitize these for recognition by virus-specific CD8+ T-cells with much greater efficiency than exogenous loading with free peptides. Systemic injection of ATPPs into tumor-bearing mice enhanced the recruitment of virus-specific T-cells into the tumor and, when combined with immune checkpoint blockade, suppressed tumor growth. Our data thereby demonstrate the potential of ATPPs as a means of kick-starting the immune response against “cold” tumors and increasing the efficacy of checkpoint inhibitors.
Collapse
Affiliation(s)
- Julian P Sefrin
- Discovery Oncology, Roche Innovation Center Penzberg, Roche Pharma Research and Early Development, Penzberg, Germany
| | - Lars Hillringhaus
- Department of Early Development and Reagent Design, Roche Diagnostics GmbH, Penzberg, Germany
| | - Olaf Mundigl
- Large Molecule Research, Roche Innovation Center Penzberg, Roche Pharma Research and Early Development, Penzberg, Germany
| | - Karin Mann
- Discovery Oncology, Roche Innovation Center Penzberg, Roche Pharma Research and Early Development, Penzberg, Germany
| | - Doris Ziegler-Landesberger
- Large Molecule Research, Roche Innovation Center Penzberg, Roche Pharma Research and Early Development, Penzberg, Germany
| | - Heike Seul
- Large Molecule Research, Roche Innovation Center Penzberg, Roche Pharma Research and Early Development, Penzberg, Germany
| | - Gloria Tabares
- Department of Early Development and Reagent Design, Roche Diagnostics GmbH, Penzberg, Germany
| | - Dominic Knoblauch
- Department of Early Development and Reagent Design, Roche Diagnostics GmbH, Penzberg, Germany
| | - Andreas Leinenbach
- Department of Early Development and Reagent Design, Roche Diagnostics GmbH, Penzberg, Germany
| | - Irene Friligou
- Department of Early Development and Reagent Design, Roche Diagnostics GmbH, Penzberg, Germany
| | - Sebastian Dziadek
- Translational Medicine Oncology, Roche Innovation Center Basel, Roche Pharma Research and Early Development, Basel, Switzerland
| | - Rienk Offringa
- Department of General Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
| | - Valeria Lifke
- Personalized Healthcare Solution, Immunoassay Development and System Integration, Roche Diagnostics GmbH, Penzberg, Germany
| | - Alexander Lifke
- Pharma Biotech Penzberg, Roche Diagnostics GmbH, Penzberg, Germany
| |
Collapse
|
49
|
Geersing A, de Vries RH, Jansen G, Rots MG, Roelfes G. Folic acid conjugates of a bleomycin mimic for selective targeting of folate receptor positive cancer cells. Bioorg Med Chem Lett 2019; 29:1922-1927. [DOI: 10.1016/j.bmcl.2019.05.047] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/21/2019] [Accepted: 05/23/2019] [Indexed: 10/26/2022]
|
50
|
Knewtson K, Perera C, Hymel D, Gao Z, Lee MM, Peterson BR. Antibody-Drug Conjugate that Exhibits Synergistic Cytotoxicity with an Endosome-Disruptive Peptide. ACS OMEGA 2019; 4:12955-12968. [PMID: 31460422 PMCID: PMC6690568 DOI: 10.1021/acsomega.9b01585] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 07/17/2019] [Indexed: 05/18/2023]
Abstract
Antibody-drug conjugates are an important class of cancer therapeutics. These agents generally bind a specific cell surface receptor, undergo receptor-mediated endocytosis, and enter the endosomal-lysosomal system, where the environment in these organelles facilitates the release of a membrane-permeable cytotoxin. By using a membrane-impermeable cytotoxin, we describe here a method that allows the cytotoxicity of an antibody conjugate to be triggered by co-administration with an endosome-disruptive peptide that exhibits low toxicity. This approach was validated by conjugation of an anionic derivative of the tubulin-binding cytotoxin colchinol methyl ether to lysine residues of the HER2-targeting antibody trastuzumab (Herceptin) via a disulfide. When this antibody binds HER2 on SKBR3 breast cancer cells and undergoes endocytosis, the membrane-impermeable cytotoxin is released, but it becomes trapped in endosomes, resulting in relatively low cytotoxicity (IC50 > 1 μM). However, co-administration with an essentially nontoxic (IC50 > 10 μM) cholesterol-linked endosome-disruptive peptide promotes the release of this small molecule into the cytoplasm, conferring subnanomolar cytotoxic potency (IC50 = 0.11 ± 0.07 nM). Studies of a structurally related fluorophore conjugate revealed that the endosome-disruptive peptide does not substantially enhance cleavage of the disulfide (t 1/2 = 8 ± 2 h) within endosomes, suggesting that the mechanism of endosomal escape involves the efflux of some small molecules without facilitating substantial influx of reduced glutathione.
Collapse
Affiliation(s)
- Kelsey
E. Knewtson
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States
| | - Chamani Perera
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States
| | | | | | | | - Blake R. Peterson
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States
| |
Collapse
|