1
|
Zhang X, Zhuo J, Wang D, Zhu X. Supramolecular Polymers for Drug Delivery. Chemistry 2025; 31:e202404617. [PMID: 39961052 DOI: 10.1002/chem.202404617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Indexed: 03/21/2025]
Abstract
Supramolecular polymers are constructed through highly reversible and directionally specific non-covalent interactions between monomer units. This unique feature enables supramolecular polymers to undergo controlled structural reconfiguration and functional transformation in response to external stimuli, imparting them with high environmental responsiveness and self-healing properties. In particular, supramolecular polymers exhibit several specific advantages compared to conventional polymers, such as inherent degradability, the ease of preparation and the incorporation of functional units, and smart responsiveness to various biological stimuli. These characters make supramolecular polymers promising candidates for intelligent drug delivery systems in complex biological environments. In this review, we comprehensively summarize the latest developments and representative achievements of supramolecular polymers in drug delivery fields, focusing primarily on the design and synthesis, the properties and functionalities, and the practical applications of supramolecular polymers in small molecule drug delivery, gene therapy, and protein delivery. Finally, we highlight future research directions, focusing on multifunctionality, adaptability, and personalized therapy. We focus on recent studies that address key challenges in the field, providing rational polymer design, important properties, functionality, and understanding delivery strategies. These developments are expected to advance supramolecular polymers as new platforms of intelligent drug delivery systems, offering innovative solutions for the treatment of complex diseases.
Collapse
Affiliation(s)
- Xinyue Zhang
- School of Chemistry and Chemical Engineering, Zhangjiang Institute for Advanced Study, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Jiaxin Zhuo
- School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, P. R. China
| | - Dali Wang
- School of Chemistry and Chemical Engineering, Zhangjiang Institute for Advanced Study, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering, Zhangjiang Institute for Advanced Study, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| |
Collapse
|
2
|
Cornell H, Sose AT, Ilic S, Chinnabattigalla S, Lidman NE, Oldmixon CM, Yang X, Deshmukh SA, Morris AJ. Photoactivated Multivariate Metal-Organic Frameworks for On-Demand Drug Release: The Role of Host-Guest Interactions. J Am Chem Soc 2025; 147:7423-7432. [PMID: 39992360 PMCID: PMC11887053 DOI: 10.1021/jacs.4c15222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/25/2025]
Abstract
The development of smart drug delivery vehicles capable of controlled release upon application of an external stimulus is of paramount interest for the next generation of personalized medicine. Herein, we report a series of six multivariate (MTV) MOFs capable of visible light-activated drug delivery. The drug loading capacity and release rates were systematically tuned through variation of the linker ratio between 4,4'-azobenzene dicarboxylic acid (H2ABDA) and 4,4'-(diazene-1,2-diyl)bis(3,5-difluorobenzoic acid) (H2ABDA(3,5-F)). The drug loading capacity, dictated by host-guest interactions, was thoroughly explored via a combined experimental and computational approach using two model drug or drug-like molecules, 5-fluorouracil (5-FU) and Nile Red. Notably, the loading capacity for 5-FU follows a "Goldilocks" profile with a maximum loading at 33% H2ABDA(3,5-F) content. Computational results confirm the existence of a cooperative ligand environment that promotes strong, preferential binding at the tetrahedral/octahedral pore window formed between two H2ABDA and one H2ABDA(3,5-F). Thus, the MTV approach enhanced capacity over the native 100% H2ABDA(3,5-F) and 0% H2ABDA(3,5-F) MOFs. In addition to increased loading, the rate of cargo release upon green light excitation also increased as the percentage of H2ABDA(3,5-F) in the MOF was raised, reaching a maximum release rate of 0.9 ± 0.1% of total cargo per minute for the MOF containing 100% H2ABDA(3,5-F) MOF. The results highlight the promise of MTV MOF design for optimizing drug delivery vehicles with relevant payloads and patient-dictated dosing.
Collapse
Affiliation(s)
- Hannah
D. Cornell
- Department
of Chemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Abhishek T. Sose
- Department
of Chemical Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Stefan Ilic
- Department
of Chemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
| | | | - Naomei E. Lidman
- Department
of Chemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Colleen M. Oldmixon
- Department
of Chemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Xiaozhou Yang
- Department
of Chemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Sanket A. Deshmukh
- Department
of Chemical Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Amanda J. Morris
- Department
of Chemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
| |
Collapse
|
3
|
Pan Y, Fan Z, Yu S, Xia L, Li J. ROS-responsive supramolecular antimicrobial peptides-based nanoprodrugs for cervical cancer therapy. Colloids Surf B Biointerfaces 2025; 247:114411. [PMID: 39613501 DOI: 10.1016/j.colsurfb.2024.114411] [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: 10/17/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024]
Abstract
Although antimicrobial peptides (AMPs) as a promising natural drugs can efficiently inhibit cervical cancer, poor bioavailability, low tumor selectivity, and non-selective toxicity still hinder its further application in vivo. In order to effectively address these challenges, we have developed a reactive oxygen species (ROS)-responsive targeting nanoprodrug designed for selective therapy of cervical cancer. Such nanoprodrugs (CEC-OxbCD) are fabricated by the supramolecular self-assembly of the modified β-cyclodextrin (β-CD) and AMPs. Antimicrobial peptide, CecropinXJ (CEC), is a cationic antibacterial peptide isolated from 3rd instar larvae of Bombyx mori from Xinjiang, China. OxbCD is an oxidation-responsive β-cyclodextrin material. CEC-OxbCD were synthesized using the nanoprecipitation/self-assembly method. Subsequently, the particle size distribution, morphology, drug loading efficiency, and release behaviour of CEC-OxbCD were characterised. In vitro and in vivo anti-cancer activities were also evaluated. Nanoprodrugs can be effectively disassembled under stimuli of the tumor- endogenous ROS, resulting in a rapid and on-demand release of antimicrobial peptides (AMPs) with a release rate of 90 %. Furthermore, both in vitro and in vivo experimental results demonstrate that our nanoprodrugs exhibit remarkable therapeutic efficacy against cervical cancer. This work not only provides an effective and promising therapeutic strategy for cervical cancer, but also explores a novel application for AMPs.
Collapse
Affiliation(s)
- Yanzhu Pan
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Zhongxiong Fan
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Shaoqi Yu
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Lijie Xia
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China.
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China.
| |
Collapse
|
4
|
Ferrero R, Pantaleone S, Brunella V, Corno M, Jiménez-Osés G, Peccati F. Host-Guest Dynamic Behavior of Melatonin Encapsulated in β-Cyclodextrin Nanosponges. ACS OMEGA 2025; 10:4660-4669. [PMID: 39959078 PMCID: PMC11822501 DOI: 10.1021/acsomega.4c09367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/04/2025] [Accepted: 01/10/2025] [Indexed: 02/18/2025]
Abstract
Cyclodextrin-based nanosponges are cross-linked polymeric porous nanomaterials obtained by condensation of cyclodextrins with a polyfunctional reagent (cross-linker). Owing to their high surface area, they are attractive for encapsulation applications aimed at increasing the stability, solubility, and bioavailability of drugs. Due to the structural complexity of these emerging materials, computer modeling can provide atomistic-level insights into both the flexibility of nanosponges and their interactions with encapsulated drugs. In this contribution, we focus on nanosponges of β-cyclodextrin cross-linked with citric acid and provide full-atom models for linear and cyclic topologies. We use extensive molecular dynamics (MD) simulations to analyze the flexibility of these constructs and their interactions with encapsulated melatonin, a neurohormone involved in sleep-wake cycle regulation also used as an antioxidant and immunomodulator. We characterize the main interactions responsible for melatonin binding and show that it benefits from multivalence and crowding effects.
Collapse
Affiliation(s)
- Riccardo Ferrero
- Dipartimento
di Chimica and NIS Interdepartmental Centre, Università degli Studi di Torino, Via P. Giuria 7, 10125 Torino, Italy
| | - Stefano Pantaleone
- Dipartimento
di Chimica and NIS Interdepartmental Centre, Università degli Studi di Torino, Via P. Giuria 7, 10125 Torino, Italy
| | - Valentina Brunella
- Dipartimento
di Chimica and NIS Interdepartmental Centre, Università degli Studi di Torino, Via P. Giuria 7, 10125 Torino, Italy
| | - Marta Corno
- Dipartimento
di Chimica and NIS Interdepartmental Centre, Università degli Studi di Torino, Via P. Giuria 7, 10125 Torino, Italy
| | - Gonzalo Jiménez-Osés
- Center
for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research
and Technology Alliance (BRTA) Bizkaia Technology Park, Building 800, 48160 Derio, Spain
- Ikerbasque,
Basque Foundation for Science, 48013 Bilbao, Spain
| | - Francesca Peccati
- Center
for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research
and Technology Alliance (BRTA) Bizkaia Technology Park, Building 800, 48160 Derio, Spain
- Ikerbasque,
Basque Foundation for Science, 48013 Bilbao, Spain
| |
Collapse
|
5
|
Addonizio CJ, Braegelman AS, Schmidt CR, Ollier RC, Antwi AA, Su B, Farshad M, Whitmer JK, Webber MJ. Impact of Guest Orientation in Host-Guest Supramolecular Hydrogels. ACS Macro Lett 2025; 14:8-13. [PMID: 39670832 DOI: 10.1021/acsmacrolett.4c00531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
Host-guest interactions have been increasingly explored for use in the dynamic physical cross-linking of polymeric precursors to form hydrogel networks. However, the orientation of guest motifs is restricted upon macromolecule conjugation. The implications of such restriction on both the kinetics and thermodynamics of the resulting host-guest supramolecular cross-links are poorly understood. Herein, guest cross-linking motifs from controlled regioisomers are demonstrated to yield distinct material properties. Moreover, the underlying phenomena point to a further unexpected impact of modular guest topology on the molecular scale in both the affinity and dynamics of supramolecular complex formation.
Collapse
Affiliation(s)
- Christopher J Addonizio
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Adam S Braegelman
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
- Bioengineering Program, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Connor R Schmidt
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Rachel C Ollier
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Akwasi Ansah Antwi
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Bo Su
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Mohsen Farshad
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Jonathan K Whitmer
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Matthew J Webber
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| |
Collapse
|
6
|
Maity S, Deb VK, Mondal S, Chakraborty A, Pramanick K, Adhikari S. Leveraging supramolecular systems in biomedical breakthroughs. Biofactors 2025; 51:e70005. [PMID: 39902766 DOI: 10.1002/biof.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/21/2025] [Indexed: 02/06/2025]
Abstract
Supramolecular systems, intricate assemblies of molecular subunits organized through various intermolecular interactions, offer versatile platforms for diverse applications, including gene therapy, antimicrobial therapy, and cellular engineering. These systems are cost-effective and environmentally friendly, contributing to their attractiveness in biomaterial design. Furthermore, supramolecular biomaterials based on acyclic, macrocyclic compounds and lipid-based assembly offer potential applications in distinct types of biomedical approaches. In this context, they can transport several therapeutic agents very effectively to the target site. Supramolecular hydrogels exhibit potent antimicrobial activity by disrupting microbial membranes, offering promising solutions to combat drug-resistant pathogens. Additionally, supramolecular luminescent nanoparticles enable targeted cell imaging, facilitating disease diagnosis and treatment with high specificity and sensitivity. In cellular engineering, supramolecular assemblies of small molecules demonstrate biological activities, overcoming challenges in cancer treatment by inhibiting signaling pathways and inducing apoptosis in cancer cells. This review emphasizes the applications of supramolecular systems from gene therapy to cellular imaging, tissue engineering, and antimicrobial therapy, showcasing their potential to drive innovation and address pressing healthcare challenges.
Collapse
Affiliation(s)
- Shreya Maity
- Integrative Biology Research Unit, Department of Life Sciences, Presidency University, Kolkata, India
| | | | - Sayani Mondal
- Integrative Biology Research Unit, Department of Life Sciences, Presidency University, Kolkata, India
| | - Akansha Chakraborty
- Integrative Biology Research Unit, Department of Life Sciences, Presidency University, Kolkata, India
| | - Kousik Pramanick
- Integrative Biology Research Unit, Department of Life Sciences, Presidency University, Kolkata, India
| | - Suman Adhikari
- Department of Chemistry, Government Degree College, Dharmanagar, India
| |
Collapse
|
7
|
Hao Y, Shen X, Liu J, Cai Z, Wang X, Yang Z, Chen F, Dong B, Wang R, Du X, Qi Z, Ge Y. A Supramolecular Protein Assembly Intrinsically Rescues Memory Deficits in an Alzheimer's Disease Mouse Model. NANO LETTERS 2024; 24:15565-15574. [PMID: 39592140 PMCID: PMC11640758 DOI: 10.1021/acs.nanolett.4c03672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 11/28/2024]
Abstract
Supramolecular protein assemblies have been used as intelligent drug delivery systems that can encapsulate drugs and transport them to specific tissues or cells. However, the known methods for designing supramolecular protein assemblies for transportation across the blood-brain barrier (BBB) remain challenging and inefficient. Herein, we report that the supramolecular recombinant-protein-based strategy enables the biosynthesis and production of a supramolecular protein assembly that is intrinsically capable of crossing the BBB. The recombinant protein constituting the essential part of apolipoprotein A1 can self-assemble into a supramolecular protein assembly known as a nanodisc. The nanodisc could efficiently enter the brain of an Alzheimer's disease mouse model, recognize Aβ1-42, eliminate amyloid plaques, promote neurogenesis, and ameliorate cognitive impairment. This work opens a new field for supramolecular protein assemblies and offers a new avenue for designing versatile and intelligent supramolecular biomaterials.
Collapse
Affiliation(s)
- Yuchong Hao
- Sino-German
Joint Research Lab for Space Biomaterials and Translational Technology,
Synergetic Innovation Center of Biological Optoelectronics and Healthcare
Engineering, School of Life Sciences, Northwestern
Polytechnical University, Youyi West Road 127, Xi’an, Shaanxi 710072, China
| | - Xin Shen
- Sino-German
Joint Research Lab for Space Biomaterials and Translational Technology,
Synergetic Innovation Center of Biological Optoelectronics and Healthcare
Engineering, School of Life Sciences, Northwestern
Polytechnical University, Youyi West Road 127, Xi’an, Shaanxi 710072, China
| | - Jiantao Liu
- Guangdong
Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences
and Oceanography, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Zhongqi Cai
- Sino-German
Joint Research Lab for Space Biomaterials and Translational Technology,
Synergetic Innovation Center of Biological Optoelectronics and Healthcare
Engineering, School of Life Sciences, Northwestern
Polytechnical University, Youyi West Road 127, Xi’an, Shaanxi 710072, China
| | - Xinquan Wang
- Sino-German
Joint Research Lab for Space Biomaterials and Translational Technology,
Synergetic Innovation Center of Biological Optoelectronics and Healthcare
Engineering, School of Life Sciences, Northwestern
Polytechnical University, Youyi West Road 127, Xi’an, Shaanxi 710072, China
| | - Zerui Yang
- Sino-German
Joint Research Lab for Space Biomaterials and Translational Technology,
Synergetic Innovation Center of Biological Optoelectronics and Healthcare
Engineering, School of Life Sciences, Northwestern
Polytechnical University, Youyi West Road 127, Xi’an, Shaanxi 710072, China
| | - Fuqing Chen
- Sino-German
Joint Research Lab for Space Biomaterials and Translational Technology,
Synergetic Innovation Center of Biological Optoelectronics and Healthcare
Engineering, School of Life Sciences, Northwestern
Polytechnical University, Youyi West Road 127, Xi’an, Shaanxi 710072, China
| | - Baorui Dong
- Sino-German
Joint Research Lab for Space Biomaterials and Translational Technology,
Synergetic Innovation Center of Biological Optoelectronics and Healthcare
Engineering, School of Life Sciences, Northwestern
Polytechnical University, Youyi West Road 127, Xi’an, Shaanxi 710072, China
| | - Ruibing Wang
- State
Key Laboratory of Quality Research in Chinese Medicine, Institute
of Chinese Medical Sciences & MoE Frontiers Science Center for
Precision Oncology, University of Macau, Taipa, Macau SAR 999078, China
| | - Xiubo Du
- Guangdong
Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences
and Oceanography, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Zhenhui Qi
- Sino-German
Joint Research Lab for Space Biomaterials and Translational Technology,
Synergetic Innovation Center of Biological Optoelectronics and Healthcare
Engineering, School of Life Sciences, Northwestern
Polytechnical University, Youyi West Road 127, Xi’an, Shaanxi 710072, China
| | - Yan Ge
- Sino-German
Joint Research Lab for Space Biomaterials and Translational Technology,
Synergetic Innovation Center of Biological Optoelectronics and Healthcare
Engineering, School of Life Sciences, Northwestern
Polytechnical University, Youyi West Road 127, Xi’an, Shaanxi 710072, China
| |
Collapse
|
8
|
Li S, Li P, Tian Y, Zeng R, Zhang Q, Pi C. A mini review of supramolecular antagonists based on macrocyclic host compounds. Bioorg Chem 2024; 153:107974. [PMID: 39571303 DOI: 10.1016/j.bioorg.2024.107974] [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: 07/28/2024] [Revised: 11/11/2024] [Accepted: 11/15/2024] [Indexed: 12/12/2024]
Abstract
In the interdisciplinary domains of medicine and chemistry, addressing the issue of residual drugs (toxicants) that fail to fully exert therapeutic effects while potentially inducing toxic side effects has become increasingly critical. Researchers are actively seeking innovative solutions to this multifaceted challenge. Conventional small-molecule antagonists, commonly used in clinical settings, typically depend on "drug-receptor interactions" yet pose substantial developmental challenges. Recent advancements in the investigation of macrocyclic host compounds present a promising alternative. By leveraging the principles of host-guest chemistry, these macrocyclic hosts form stable inclusion complexes with residual drugs (toxicants), thereby decreasing their free concentration in the bloodstream and effectively mitigating associated toxic side effects. Consequently, macrocyclic host compounds represent a novel class of supramolecular antagonists (SAs). This article reviews recent progress in the application of macrocyclic host molecules-such as cyclodextrin, calix[n]arene, pillar[n]arene, and cucurbit[n]uril-as SA and examines current issues and future development prospects within the field.
Collapse
Affiliation(s)
- Shanshan Li
- College of Pharmacy, Key Laboratory of Research and Application of Ethnic Medicine Processing and Preparation on the Qinghai-Tibet Plateau, Southwest Minzu University, Chengdu 610041, China
| | - Pengcheng Li
- College of Pharmacy, Key Laboratory of Research and Application of Ethnic Medicine Processing and Preparation on the Qinghai-Tibet Plateau, Southwest Minzu University, Chengdu 610041, China
| | - Yuhan Tian
- College of Pharmacy, Key Laboratory of Research and Application of Ethnic Medicine Processing and Preparation on the Qinghai-Tibet Plateau, Southwest Minzu University, Chengdu 610041, China
| | - Rui Zeng
- College of Pharmacy, Key Laboratory of Research and Application of Ethnic Medicine Processing and Preparation on the Qinghai-Tibet Plateau, Southwest Minzu University, Chengdu 610041, China
| | - Qixiong Zhang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Department of Pharmacy, Sichuan Provincial People's Hospital East Sichuan Hospital & Dazhou First People's Hospital, Dazhou, 635000, China.
| | - Chuan Pi
- Department of Pharmacy, Sichuan Provincial People's Hospital East Sichuan Hospital & Dazhou First People's Hospital, Dazhou, 635000, China
| |
Collapse
|
9
|
Yu S, Webber MJ. Engineering disease analyte response in peptide self-assembly. J Mater Chem B 2024; 12:10757-10769. [PMID: 39382032 DOI: 10.1039/d4tb01860e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
A need to enhance the precision and specificity of therapeutic nanocarriers inspires the development of advanced nanomaterials capable of sensing and responding to disease-related cues. Self-assembled peptides offer a promising nanocarrier platform with versatile use to create precisely defined nanoscale materials. Disease-relevant cues can range from large biomolecules, such as enzymes, to ubiquitous small molecules with varying concentrations in healthy versus diseased states. Notably, pH changes (i.e., H+ concentration), redox species (e.g., H2O2), and glucose levels are significant spatial and/or temporal indicators of therapeutic need. Self-assembled peptides respond to these cues by altering their solubility, modulating electrostatic interactions, or facilitating chemical transformations through dynamic or labile bonds. This review explores the design and construction of therapeutic nanocarriers using self-assembled peptides, focusing on how peptide sequence engineering along with the inclusion of non-peptidic components can link the assembly state of these nanocarriers to the presence of disease-relevant small molecules.
Collapse
Affiliation(s)
- Sihan Yu
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Matthew J Webber
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
10
|
Dietl MC, Maag M, Ber S, Rominger F, Rudolph M, Caligiuri I, Andele PK, Mkhalid IAI, Rizzolio F, Nogara PA, Orian L, Scattolin T, Hashmi ASK. Comparative study of the antiproliferative activity of heterometallic carbene gold(i)-platinum(ii) and gold(i)-palladium(ii) complexes in cancer cell lines. Chem Sci 2024:d4sc04585h. [PMID: 39246355 PMCID: PMC11376197 DOI: 10.1039/d4sc04585h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 07/26/2024] [Indexed: 09/10/2024] Open
Abstract
The stepwise, one-pot synthesis of heterobimetallic carbene gold(i) platinum(ii) complexes from readily available starting materials is presented. The protecting group free methodology is based on the graduated nucleophilicities of aliphatic and aromatic amines as linkers between both metal centers. This enables the selective, sequential installation of the metal fragments. In addition, the obtained complexes were tested as potential anticancer agents and directly compared to their gold(i) palladium(ii) counterparts.
Collapse
Affiliation(s)
- Martin C Dietl
- Organisch-Chemisches Institut, Heidelberg University Im Neuenheimer Feld 270 69120 Heidelberg Germany
| | - Melina Maag
- Organisch-Chemisches Institut, Heidelberg University Im Neuenheimer Feld 270 69120 Heidelberg Germany
| | - Sophia Ber
- Organisch-Chemisches Institut, Heidelberg University Im Neuenheimer Feld 270 69120 Heidelberg Germany
| | - Frank Rominger
- Organisch-Chemisches Institut, Heidelberg University Im Neuenheimer Feld 270 69120 Heidelberg Germany
| | - Matthias Rudolph
- Organisch-Chemisches Institut, Heidelberg University Im Neuenheimer Feld 270 69120 Heidelberg Germany
| | - Isabella Caligiuri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS via Franco Gallini 2 33081 Aviano Italy
| | - Pacome K Andele
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS via Franco Gallini 2 33081 Aviano Italy
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari Campus Scientifico Via Torino 155 30174 Venezia-Mestre Italy
| | - Ibraheem A I Mkhalid
- Chemistry Department, Faculty of Science, King Abdulaziz University Jeddah 21589 Saudi Arabia
| | - Flavio Rizzolio
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS via Franco Gallini 2 33081 Aviano Italy
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari Campus Scientifico Via Torino 155 30174 Venezia-Mestre Italy
| | - Pablo A Nogara
- Instituto Federal de Educação, Ciência e Tecnologia Sul-rio-grandense (IFSul) Av. Leonel de Moura Brizola, 2501 96418-400 Bagé RS Brazil
| | - Laura Orian
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova via Marzolo 1 35131 Padova Italy
| | - Thomas Scattolin
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova via Marzolo 1 35131 Padova Italy
| | - A Stephen K Hashmi
- Organisch-Chemisches Institut, Heidelberg University Im Neuenheimer Feld 270 69120 Heidelberg Germany
- Chemistry Department, Faculty of Science, King Abdulaziz University Jeddah 21589 Saudi Arabia
| |
Collapse
|
11
|
Li WB, Xu LL, Wang SL, Wang YY, Pan YC, Shi LQ, Guo DS. Co-Assembled Nanoparticles toward Multi-Target Combinational Therapy of Alzheimer's Disease by Making Full Use of Molecular Recognition and Self-Assembly. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2401918. [PMID: 38662940 DOI: 10.1002/adma.202401918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/10/2024] [Indexed: 05/07/2024]
Abstract
The complex pathologies in Alzheimer's disease (AD) severely limit the effectiveness of single-target pharmic interventions, thus necessitating multi-pronged therapeutic strategies. While flexibility is essentially demanded in constructing such multi-target systems, for achieving optimal synergies and also accommodating the inherent heterogeneity within AD. Utilizing the dynamic reversibility of supramolecular strategy for conferring sufficient tunability in component substitution and proportion adjustment, amphiphilic calixarenes are poised to be a privileged molecular tool for facilely achieving function integration. Herein, taking β-amyloid (Aβ) fibrillation and oxidative stress as model combination pattern, a supramolecular multifunctional integration is proposed by co-assembling guanidinium-modified calixarene with ascorbyl palmitate and loading dipotassium phytate within calixarene cavity. Serial pivotal events can be simultaneously addressed by this versatile system, including 1) inhibition of Aβ production and aggregation, 2) disintegration of Aβ fibrils, 3) acceleration of Aβ metabolic clearance, and 4) regulation of oxidative stress, which is verified to significantly ameliorate the cognitive impairment of 5×FAD mice, with reduced Aβ plaque content, neuroinflammation, and neuronal apoptosis. Confronted with the extremely intricate clinical realities of AD, the strategy presented here exhibits ample adaptability for necessary alterations on combinations, thereby may immensely expedite the advancement of AD combinational therapy through providing an exceptionally convenient platform.
Collapse
Affiliation(s)
- Wen-Bo Li
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, 300071, China
- State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin, 300071, China
| | - Lin-Lin Xu
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Si-Lei Wang
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Ying-Yue Wang
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, 300071, China
- State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin, 300071, China
| | - Yu-Chen Pan
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, 300071, China
- State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin, 300071, China
| | - Lin-Qi Shi
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin, 300090, China
| | - Dong-Sheng Guo
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, 300071, China
- State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin, 300071, China
- Xinjiang Key Laboratory of Novel Functional Materials Chemistry, College of Chemistry and Environmental Sciences, Kashi University, Kashi, 844000, China
| |
Collapse
|
12
|
Chen FY, Li CZ, Han H, Geng WC, Zhang SX, Jiang ZT, Zhao QY, Cai K, Guo DS. Expanding the Hydrophobic Cavity Surface of Azocalix[4]arene to Enable Biotin/Avidin Affinity with Controlled Release. Angew Chem Int Ed Engl 2024; 63:e202402139. [PMID: 38563765 DOI: 10.1002/anie.202402139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/04/2024]
Abstract
The development of artificial receptors that combine ultrahigh-affinity binding and controllable release for active guests holds significant importance in biomedical applications. On one hand, a complex with an exceedingly high binding affinity can resist unwanted dissociation induced by dilution effect and complex interferents within physiological environments. On the other hand, stimulus-responsive release of the guest is essential for precisely activating its function. In this context, we expanded hydrophobic cavity surface of a hypoxia-responsive azocalix[4]arene, affording Naph-SAC4A. This modification significantly enhanced its aqueous binding affinity to 1013 M-1, akin to the naturally occurring strongest recognition pair, biotin/(strept-)avidin. Consequently, Naph-SAC4A emerges as the first artificial receptor to simultaneously integrate ultrahigh recognition affinity and actively controllable release. The markedly enhanced affinity not only improved Naph-SAC4A's sensitivity in detecting rocuronium bromide in serum, but also refined the precision of hypoxia-responsive doxorubicin delivery at the cellular level, demonstrating its immense potential for diverse practical applications.
Collapse
Affiliation(s)
- Fang-Yuan Chen
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, 300071, Tianjin, China
| | - Cheng-Zhi Li
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, 300071, Tianjin, China
| | - Han Han
- Department of Chemistry, The University of Hong Kong, 999077, Hong Kong SAR, China
| | - Wen-Chao Geng
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, 300071, Tianjin, China
| | - Shu-Xin Zhang
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, 300071, Tianjin, China
| | - Ze-Tao Jiang
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, 300071, Tianjin, China
| | - Qing-Yu Zhao
- College of Chemistry, Nankai University, 300071, Tianjin, China
| | - Kang Cai
- College of Chemistry, Nankai University, 300071, Tianjin, China
| | - Dong-Sheng Guo
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, 300071, Tianjin, China
| |
Collapse
|
13
|
Geng WC, Jiang ZT, Chen SL, Guo DS. Supramolecular interaction in the action of drug delivery systems. Chem Sci 2024; 15:7811-7823. [PMID: 38817563 PMCID: PMC11134347 DOI: 10.1039/d3sc04585d] [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: 10/04/2023] [Accepted: 04/27/2024] [Indexed: 06/01/2024] Open
Abstract
Complex diseases and diverse clinical needs necessitate drug delivery systems (DDSs), yet the current performance of DDSs is far from ideal. Supramolecular interactions play a pivotal role in various aspects of drug delivery, encompassing biocompatibility, drug loading, stability, crossing biological barriers, targeting, and controlled release. Nevertheless, despite having some understanding of the role of supramolecular interactions in drug delivery, their incorporation is frequently overlooked in the design and development of DDSs. This perspective provides a brief analysis of the involved supramolecular interactions in the action of drug delivery, with a primary emphasis on the DDSs employed in the clinic, mainly liposomes and polymers, and recognized phenomena in research, such as the protein corona. The supramolecular interactions implicated in various aspects of drug delivery systems, including biocompatibility, drug loading, stability, spatiotemporal distribution, and controlled release, were individually analyzed and discussed. This perspective aims to trigger a comprehensive and systematic consideration of supramolecular interactions in the further development of DDSs. Supramolecular interactions embody the true essence of the interplay between the majority of DDSs and biological systems.
Collapse
Affiliation(s)
- Wen-Chao Geng
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University Tianjin 300071 China
| | - Ze-Tao Jiang
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University Tianjin 300071 China
| | - Shi-Lin Chen
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University Tianjin 300071 China
| | - Dong-Sheng Guo
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University Tianjin 300071 China
| |
Collapse
|
14
|
Yao SY, Ying AK, Jiang ZT, Cheng YQ, Geng WC, Hu XY, Cai K, Guo DS. Single Molecular Nanomedicines Based on Macrocyclic Carrier-Drug Conjugates for Concentration-Independent Encapsulation and Precise Activation of Drugs. J Am Chem Soc 2024; 146:14203-14212. [PMID: 38733560 DOI: 10.1021/jacs.4c03238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2024]
Abstract
Nanomedicines often rely on noncovalent self-assembly and encapsulation for drug loading and delivery. However, challenges such as reproducibility issues due to the multicomponent nature, off-target activation caused by premature drug release, and complex pharmacokinetics arising from assembly dissociation have hindered their clinical translation. In this study, we introduce an innovative design concept termed single molecular nanomedicine (SMNM) based on macrocyclic carrier-drug conjugates. Through the covalent linkage of two chemotherapy drugs to a hypoxia-cleavable macrocyclic carrier, azocalix[4]arene, we obtained two self-included complexes to serve as SMNMs. The intramolecular inclusion feature of the SMNMs has not only demonstrated comprehensive shielding and protection for the drugs but also effectively prevented off-target drug leakage, thereby significantly reducing their side effects and enhancing their antitumor therapeutic efficacy. Additionally, the attributes of being a single component and molecularly dispersed confer advantages such as ease of preparation and good reproducibility for SMNMs, which is desirable for clinical applications.
Collapse
Affiliation(s)
- Shun-Yu Yao
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - An-Kang Ying
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Ze-Tao Jiang
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Yuan-Qiu Cheng
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Wen-Chao Geng
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Xin-Yue Hu
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Kang Cai
- College of Chemistry, Nankai University, Tianjin 300071, China
| | - Dong-Sheng Guo
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
- Xinjiang Key Laboratory of Novel Functional Materials Chemistry, College of Chemistry and Environmental Sciences, Kashi University, Kashi 844000, China
| |
Collapse
|
15
|
Su B, Chi T, Chen W, Xian S, Liu D, Addonizio CJ, Xiang Y, Webber MJ. Using a biocatalyzed reaction cycle for transient and pH-dependent host-guest supramolecular hydrogels. J Mater Chem B 2024; 12:4666-4672. [PMID: 38647183 PMCID: PMC11095629 DOI: 10.1039/d4tb00545g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
The formation of transient structures plays important roles in biological processes, capturing temporary states of matter through influx of energy or biological reaction networks catalyzed by enzymes. These natural transient structures inspire efforts to mimic this elegant mechanism of structural control in synthetic analogues. Specifically, though traditional supramolecular materials are designed on the basis of equilibrium formation, recent efforts have explored out-of-equilibrium control of these materials using both direct and indirect mechanisms; the preponderance of such works has been in the area of low molecular weight gelators. Here, a transient supramolecular hydrogel is realized through cucurbit[7]uril host-guest physical crosslinking under indirect control from a biocatalyzed network that regulates and oscillates pH. The duration of transient hydrogel formation, and resulting mechanical properties, are tunable according to the dose of enzyme, substrate, or pH stimulus. This tunability enables control over emergent functions, such as the programmable burst release of encapsulated model macromolecular payloads.
Collapse
Affiliation(s)
- Bo Su
- Department of Chemcial & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana, USA.
| | - Teng Chi
- Department of Chemcial & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana, USA.
| | - Weike Chen
- Department of Chemcial & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana, USA.
| | - Sijie Xian
- Department of Chemcial & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana, USA.
| | - Dongping Liu
- Department of Chemcial & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana, USA.
| | - Christopher J Addonizio
- Department of Chemcial & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana, USA.
| | - Yuanhui Xiang
- Department of Chemcial & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana, USA.
| | - Matthew J Webber
- Department of Chemcial & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana, USA.
| |
Collapse
|
16
|
Yan M, Wu S, Wang Y, Liang M, Wang M, Hu W, Yu G, Mao Z, Huang F, Zhou J. Recent Progress of Supramolecular Chemotherapy Based on Host-Guest Interactions. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2304249. [PMID: 37478832 DOI: 10.1002/adma.202304249] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/10/2023] [Indexed: 07/23/2023]
Abstract
Chemotherapy is widely recognized as an effective approach for treating cancer due to its ability to eliminate cancer cells using chemotherapeutic drugs. However, traditional chemotherapy suffers from various drawbacks, including limited solubility and stability of drugs, severe side effects, low bioavailability, drug resistance, and challenges in tracking treatment efficacy. These limitations greatly hinder its widespread clinical application. In contrast, supramolecular chemotherapy, which relies on host-guest interactions, presents a promising alternative by offering highly efficient and minimally toxic anticancer drug delivery. In this review, an overview of recent advancements in supramolecular chemotherapy based on host-guest interactions is provided. The significant role it plays in guiding cancer therapy is emphasized. Drawing on a wealth of cutting-edge research, herein, a timely and valuable resource for individuals interested in the field of supramolecular chemotherapy or cancer therapy, is presented. Furthermore, this review contributes to the progression of the field of supramolecular chemotherapy toward clinical application.
Collapse
Affiliation(s)
- Miaomiao Yan
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, P. R. China
| | - Sha Wu
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, P. R. China
| | - Yuhao Wang
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, P. R. China
| | - Minghao Liang
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, P. R. China
| | - Mengbin Wang
- Stoddart Institute of Molecular Science, Department of Chemistry, Zhejiang University, Hangzhou, 310058, P. R. China
- Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311215, P. R. China
| | - Wenting Hu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P. R. China
| | - Guocan Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, P. R. China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Feihe Huang
- Stoddart Institute of Molecular Science, Department of Chemistry, Zhejiang University, Hangzhou, 310058, P. R. China
- Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311215, P. R. China
| | - Jiong Zhou
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, P. R. China
- Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou, 510632, P. R. China
| |
Collapse
|
17
|
Gong N, Han X, Xue L, Billingsley MM, Huang X, El-Mayta R, Qin J, Sheppard NC, June CH, Mitchell MJ. Small-molecule-mediated control of the anti-tumour activity and off-tumour toxicity of a supramolecular bispecific T cell engager. Nat Biomed Eng 2024; 8:513-528. [PMID: 38378820 DOI: 10.1038/s41551-023-01147-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 10/24/2023] [Indexed: 02/22/2024]
Abstract
The broader clinical use of bispecific T cell engagers for inducing anti-tumour toxicity is hindered by their on-target off-tumour toxicity and the associated neurotoxicity and cytokine-release syndrome. Here we show that the off-tumour toxicity of a supramolecular bispecific T cell engager binding to the T cell co-receptor CD3 and to the human epidermal growth factor receptor 2 on breast tumour cells can be halted by disengaging the T cells from the tumour cells via the infusion of the small-molecule drug amantadine, which disassembles the supramolecular aggregate. In mice bearing human epidermal growth factor receptor 2-expressing tumours and with a human immune system, high intravenous doses of such a 'switchable T cell nanoengager' elicited strong tumour-specific adaptive immune responses that prevented tumour relapse, while the infusion of amantadine restricted off-tumour toxicity, cytokine-release syndrome and neurotoxicity. Supramolecular chemistry may be further leveraged to control the anti-tumour activity and off-tumour toxicity of bispecific antibodies.
Collapse
Affiliation(s)
- Ningqiang Gong
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Xuexiang Han
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Lulu Xue
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Xisha Huang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Rakan El-Mayta
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Jingya Qin
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Neil C Sheppard
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for RNA Innovation, University of Pennsylvania, Philadelphia, PA, USA.
- Center for Precision Engineering for Health, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
18
|
Chen MM, Tang X, Li JJ, Chen FY, Jiang ZT, Fu R, Li HB, Hu XY, Geng WC, Guo DS. Active targeting tumor therapy using host-guest drug delivery system based on biotin functionalized azocalix[4]arene. J Control Release 2024; 368:691-702. [PMID: 38492860 DOI: 10.1016/j.jconrel.2024.03.017] [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: 11/24/2023] [Revised: 02/25/2024] [Accepted: 03/12/2024] [Indexed: 03/18/2024]
Abstract
Host-guest drug delivery systems (HGDDSs) provided a facile method for incorporating biomedical functions, including efficient drug-loading, passive targeting, and controlled drug release. However, developing HGDDSs with active targeting is hindered by the difficult functionalization of popular macrocycles. Herein, we report an active targeting HGDDS based on biotin-modified sulfonated azocalix[4]arene (Biotin-SAC4A) to efficiently deliver drug into cancer cells for improving anti-tumor effect. Biotin-SAC4A was synthesized by amide condensation and azo coupling. Biotin-SAC4A demonstrated hypoxia responsive targeting and active targeting through azo and biotin groups, respectively. DOX@Biotin-SAC4A, which was prepared by loading doxorubicin (DOX) in Biotin-SAC4A, was evaluated for tumor targeting and therapy in vitro and in vivo. DOX@Biotin-SAC4A formulation effectively killed cancer cells in vitro and more efficiently delivered DOX to the lesion than the similar formulation without active targeting. Therefore, DOX@Biotin-SAC4A significantly improved the in vivo anti-tumor effect of free DOX. The facilely prepared Biotin-SAC4A offers strong DOX complexation, active targeting, and hypoxia-triggered release, providing a favorable host for effective breast cancer chemotherapy in HGDDSs. Moreover, Biotin-SAC4A also has potential to deliver agents for other therapeutic modalities and diseases.
Collapse
Affiliation(s)
- Meng-Meng Chen
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Collaborative Innovation Center of Chemical Science and Engineering, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, PR China
| | - Xingchen Tang
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Collaborative Innovation Center of Chemical Science and Engineering, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, PR China
| | - Juan-Juan Li
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Collaborative Innovation Center of Chemical Science and Engineering, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, PR China
| | - Fang-Yuan Chen
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Collaborative Innovation Center of Chemical Science and Engineering, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, PR China
| | - Ze-Tao Jiang
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Collaborative Innovation Center of Chemical Science and Engineering, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, PR China
| | - Rong Fu
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Collaborative Innovation Center of Chemical Science and Engineering, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, PR China
| | - Hua-Bin Li
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Collaborative Innovation Center of Chemical Science and Engineering, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, PR China
| | - Xin-Yue Hu
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Collaborative Innovation Center of Chemical Science and Engineering, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, PR China
| | - Wen-Chao Geng
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Collaborative Innovation Center of Chemical Science and Engineering, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, PR China.
| | - Dong-Sheng Guo
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Collaborative Innovation Center of Chemical Science and Engineering, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, PR China.
| |
Collapse
|
19
|
Alešković M, Šekutor M. Overcoming barriers with non-covalent interactions: supramolecular recognition of adamantyl cucurbit[ n]uril assemblies for medical applications. RSC Med Chem 2024; 15:433-471. [PMID: 38389878 PMCID: PMC10880950 DOI: 10.1039/d3md00596h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 11/30/2023] [Indexed: 02/24/2024] Open
Abstract
Adamantane, a staple in medicinal chemistry, recently became a cornerstone of a supramolecular host-guest drug delivery system, ADA/CB[n]. Owing to a good fit between the adamantane cage and the host cavity of the cucurbit[n]uril macrocycle, formed strong inclusion complexes find applications in drug delivery and controlled drug release. Note that the cucurbit[n]uril host is not solely a delivery vehicle of the ADA/CB[n] system but rather influences the bioactivity and bioavailability of drug molecules and can tune drug properties. Namely, as host-guest interactions are capable of changing the intrinsic properties of the guest molecule, inclusion complexes can become more soluble, bioavailable and more resistant to metabolic conditions compared to individual non-complexed molecules. Such synergistic effects have implications for practical bioapplicability of this complex system and provide a new viewpoint to therapy, beyond the traditional single drug molecule approach. By achieving a balance between guest encapsulation and release, the ADA/CB[n] system has also found use beyond just drug delivery, in fields like bioanalytics, sensing assays, bioimaging, etc. Thus, chemosensing in physiological conditions, indicator displacement assays, in vivo diagnostics and hybrid nanostructures are just some recent examples of the ADA/CB[n] applicability, be it for displacements purposes or as cargo vehicles.
Collapse
Affiliation(s)
- Marija Alešković
- Department of Organic Chemistry and Biochemistry, Ruđer Bošković Institute Bijenička 54 10 000 Zagreb Croatia
| | - Marina Šekutor
- Department of Organic Chemistry and Biochemistry, Ruđer Bošković Institute Bijenička 54 10 000 Zagreb Croatia
| |
Collapse
|
20
|
Attia MF, Ogunnaike EA, Pitz M, Elbaz NM, Panda DK, Alexander-Bryant A, Saha S, Whitehead DC, Kabanov A. Enhancing drug delivery with supramolecular amphiphilic macrocycle nanoparticles: selective targeting of CDK4/6 inhibitor palbociclib to melanoma. Biomater Sci 2024; 12:725-737. [PMID: 38099834 PMCID: PMC10872447 DOI: 10.1039/d3bm01888a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2023]
Abstract
Drug delivery systems based on amphiphilic supramolecular macrocycles have garnered increased attention over the past two decades due to their ability to successfully formulate nanoparticles. Macrocyclic (MC) materials can self-assemble at lower concentrations without the need for surfactants and polymers, but surfactants are required to form and stabilize nanoparticles at higher concentrations. Using MCs to deliver both hydrophilic and hydrophobic guest molecules is advantageous. We developed two novel types of amphiphilic macrocycle nanoparticles (MC NPs) capable of delivering either Nile Red (NR) (a hydrophobic model) or Rhodamine B (RhB) (a hydrophilic model) fluorescent dyes. We extensively characterized the materials using various techniques to determine size, morphology, stability, hemolysis, fluorescence, loading efficiency (LE), and loading capacity (LC). We then loaded the CDK4/6 inhibitor Palbociclib (Palb) into both MC NPs using a solvent diffusion method. This yielded Palb-MC NPs in the size range of 65-90 nm. They exhibited high stability over time and in fetal bovine serum with negligible toxicity against erythrocytes. Cytotoxicity was minimal when tested against RAW macrophages, human fibroblast HDFn, and adipose stromal cells (ASCs) at higher concentrations of MC NPs. Cell viability studies were conducted with different concentrations of MC NPs, Palb-MC NPs, and free Palb against RAW macrophages, human U-87 GBM, and human M14 melanoma cell lines in vitro. Flow cytometry experiments revealed that blank MC NPs and Palb-MC NPs were selectively targeted to melanoma cells, resulting in cell death compared to the other two cell lines. Future work will focus on studying the biological effect of MC NPs including their binding affinity with molecules/receptors expressed on the M14 and other melanoma cell surfaces by molecular docking simulations. Subsequently, we will evaluate the MCs as a component of combination therapy in a murine melanoma model.
Collapse
Affiliation(s)
- Mohamed F Attia
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA.
| | - Edikan A Ogunnaike
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA.
| | - Megan Pitz
- Department of Bioengineering, Clemson University, Clemson, SC, 29634, USA
| | - Nancy M Elbaz
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA.
| | - Dillip K Panda
- Department of Chemistry, Clemson University, Clemson, SC, 29634, USA.
| | | | - Sourav Saha
- Department of Chemistry, Clemson University, Clemson, SC, 29634, USA.
| | | | - Alexander Kabanov
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA.
- Laboratory of Chemical Design of Bionanomaterials, Faculty of Chemistry, M.V. Lomonosov Moscow State University, Moscow, 119992, Russia
| |
Collapse
|
21
|
Dutton KG, Jones TJ, Emge TJ, Lipke MC. Cage Match: Comparing the Anion Binding Ability of Isostructural Versus Isofunctional Pairs of Metal-Organic Nanocages. Chemistry 2024; 30:e202303013. [PMID: 37907394 DOI: 10.1002/chem.202303013] [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] [Received: 09/15/2023] [Revised: 10/25/2023] [Accepted: 10/31/2023] [Indexed: 11/02/2023]
Abstract
Affinities of six anions (mesylate, acetate, trifluoroacetate, p-toluenecarboxylate, p-toluenesulfonate, and perfluorooctanoate) for three related Pt2+ -linked porphyrin nanocages were measured to probe the influence of different noncovalent recognition motifs (e. g., hydrogen bonding, electrostatics, π bonding) on anion binding. Two new hosts of M6 L3 12+ (1b) and M4 L2 8+ (2) composition (M=(en)Pt2+ , L=(3-py)4 porphyrin) were prepared in a one-pot synthesis and allowed comparison of hosts that differ in structure while maintaining similar N-H hydrogen-bond donor ability. Comparisons of isostructural hosts that differ in hydrogen-bonding ability were made between 1b and a related M6 L3 12+ nanoprism (1a, M=(tmeda)Pt2+ ) that lacks N-H groups. Considerable variation in association constants (K1 =1.6×103 M-1 to 1.3×108 M-1 ) and binding mode (exo vs. endo) were found for different host-guest combinations. Strongest binding was seen between p-toluenecarboxylate and 1b, but surprisingly, association of this guest with 1a was only slightly weaker despite the absence of NH⋅⋅⋅O interactions. The high affinity between p-toluenecarboxylate and 1a could be turned off by protonation, and this behavior was used to toggle between the binding of this guest and the environmental pollutant perfluorooctanoate, which otherwise has a lower affinity for the host.
Collapse
Affiliation(s)
- Kaitlyn G Dutton
- Department of Chemistry and Chemical Biology, Rutgers University - New Brunswick, 123 Bevier Road Piscataway, NJ, 08854, USA
| | - Taro J Jones
- Department of Chemistry and Chemical Biology, Rutgers University - New Brunswick, 123 Bevier Road Piscataway, NJ, 08854, USA
| | - Thomas J Emge
- Department of Chemistry and Chemical Biology, Rutgers University - New Brunswick, 123 Bevier Road Piscataway, NJ, 08854, USA
| | - Mark C Lipke
- Department of Chemistry and Chemical Biology, Rutgers University - New Brunswick, 123 Bevier Road Piscataway, NJ, 08854, USA
| |
Collapse
|
22
|
Hidalgo-Alvarez V, Madl CM. Leveraging Biomaterial Platforms to Study Aging-Related Neural and Muscular Degeneration. Biomolecules 2024; 14:69. [PMID: 38254669 PMCID: PMC10813704 DOI: 10.3390/biom14010069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 12/28/2023] [Accepted: 12/30/2023] [Indexed: 01/24/2024] Open
Abstract
Aging is a complex multifactorial process that results in tissue function impairment across the whole organism. One of the common consequences of this process is the loss of muscle mass and the associated decline in muscle function, known as sarcopenia. Aging also presents with an increased risk of developing other pathological conditions such as neurodegeneration. Muscular and neuronal degeneration cause mobility issues and cognitive impairment, hence having a major impact on the quality of life of the older population. The development of novel therapies that can ameliorate the effects of aging is currently hindered by our limited knowledge of the underlying mechanisms and the use of models that fail to recapitulate the structure and composition of the cell microenvironment. The emergence of bioengineering techniques based on the use of biomimetic materials and biofabrication methods has opened the possibility of generating 3D models of muscular and nervous tissues that better mimic the native extracellular matrix. These platforms are particularly advantageous for drug testing and mechanistic studies. In this review, we discuss the developments made in the creation of 3D models of aging-related neuronal and muscular degeneration and we provide a perspective on the future directions for the field.
Collapse
Affiliation(s)
| | - Christopher M. Madl
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA;
| |
Collapse
|
23
|
Pinto AF, Nunes JS, Severino Martins JE, Leal AC, Silva CCVC, da Silva AJFS, da Cruz Olímpio DS, da Silva ETN, Campos TA, Lima Leite AC. Thiazole, Isatin and Phthalimide Derivatives Tested in vivo against Cancer Models: A Literature Review of the Last Six Years. Curr Med Chem 2024; 31:2991-3032. [PMID: 37170994 DOI: 10.2174/0929867330666230426154055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 02/06/2023] [Accepted: 02/16/2023] [Indexed: 05/13/2023]
Abstract
BACKGROUND Cancer is a disease characterized by the abnormal multiplication of cells and is the second leading cause of death in the world. The search for new effective and safe anticancer compounds is ongoing due to factors such as low selectivity, high toxicity, and multidrug resistance. Thus, heterocyclic compounds derived from isatin, thiazole and phthalimide that have achieved promising in vitro anticancer activity have been tested in vivo and in clinical trials. OBJECTIVE This review focused on the compilation of promising data from thiazole, isatin, and phthalimide derivatives, reported in the literature between 2015 and 2022, with in vivo anticancer activity and clinical trials. METHODS A bibliographic search was carried out in the PUBMED, MEDLINE, ELSEVIER, and CAPES PERIODIC databases, selecting relevant works for each pharmacophoric group with in vivo antitumor activity in the last 6 years. RESULTS In our study, 68 articles that fit the scope were selected and critically analyzed. These articles were organized considering the type of antitumor activity and their year of publication. Some compounds reported here demonstrated potent antitumor activity against several tumor types. CONCLUSION This review allowed us to highlight works that reported promising structures for the treatment of various cancer types and also demonstrated that the privileged structures thiazole, isatin and phthalimide are important in the design of new syntheses and molecular optimization of compounds with antitumor activity.
Collapse
Affiliation(s)
- Aline Ferreira Pinto
- Laboratory of Planning in Medicinal Chemistry, Department of Pharmaceutical Sciences, Center for Health Sciences, Federal University of Pernambuco, 50740-520, Recife, PE, Brazil
| | - Janine Siqueira Nunes
- Laboratory of Planning in Medicinal Chemistry, Department of Pharmaceutical Sciences, Center for Health Sciences, Federal University of Pernambuco, 50740-520, Recife, PE, Brazil
| | - José Eduardo Severino Martins
- Regulatory Affairs Advisory, Empresa Brasileira de Hemoderivados e Biotecnologia (HEMOBRAS), CEP 51021-410, Recife, PE, Brazil
| | - Amanda Calazans Leal
- Laboratory of Planning in Medicinal Chemistry, Department of Pharmaceutical Sciences, Center for Health Sciences, Federal University of Pernambuco, 50740-520, Recife, PE, Brazil
| | - Carla Cauanny Vieira Costa Silva
- Laboratory of Planning in Medicinal Chemistry, Department of Pharmaceutical Sciences, Center for Health Sciences, Federal University of Pernambuco, 50740-520, Recife, PE, Brazil
| | - Anderson José Firmino Santos da Silva
- Laboratory of Planning in Medicinal Chemistry, Department of Pharmaceutical Sciences, Center for Health Sciences, Federal University of Pernambuco, 50740-520, Recife, PE, Brazil
| | - Daiane Santiago da Cruz Olímpio
- Laboratory of Planning in Medicinal Chemistry, Department of Pharmaceutical Sciences, Center for Health Sciences, Federal University of Pernambuco, 50740-520, Recife, PE, Brazil
| | - Elineide Tayse Noberto da Silva
- Laboratory of Planning in Medicinal Chemistry, Department of Pharmaceutical Sciences, Center for Health Sciences, Federal University of Pernambuco, 50740-520, Recife, PE, Brazil
| | - Thiers Araújo Campos
- Laboratory of Planning in Medicinal Chemistry, Department of Pharmaceutical Sciences, Center for Health Sciences, Federal University of Pernambuco, 50740-520, Recife, PE, Brazil
| | - Ana Cristina Lima Leite
- Laboratory of Planning in Medicinal Chemistry, Department of Pharmaceutical Sciences, Center for Health Sciences, Federal University of Pernambuco, 50740-520, Recife, PE, Brazil
| |
Collapse
|
24
|
Li S, Li JJ, Zhao YY, Chen MM, Su SS, Yao SY, Wang ZH, Hu XY, Geng WC, Wang W, Wang KR, Guo DS. Supramolecular Integration of Multifunctional Nanomaterial by Mannose-Decorated Azocalixarene with Ginsenoside Rb1 for Synergistic Therapy of Rheumatoid Arthritis. ACS NANO 2023; 17:25468-25482. [PMID: 38096153 DOI: 10.1021/acsnano.3c09140] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
The complexity and progressive nature of diseases require the exploitation of multifunctional materials. However, introducing a function inevitably increases the complexity of materials, which complicates preparation and decreases reproducibility. Herein, we report a supramolecular integration of multifunctional nanomaterials based on mannose-modified azocalix[4]arene (ManAC4A) and ginsenoside Rb1 (Rb1), which showed advances of simplicity and reproducibility. ManAC4A possesses reactive oxygen species (ROS) scavenging capacity and hypoxia-responsiveness, together with macrophage-targeting and induction functionality. Collectively, the Rb1@ManAC4A assembly simply prepared by two components is integrated with multifunction, including triple targeting (ELVIS targeting, macrophage-targeting, and hypoxia-targeted release) and triple therapy (ROS scavenging, macrophage polarization, and the anti-inflammatory effect of Rb1). The spontaneous assembly and recognition of ManAC4A, with its precise structure and molecular weight, facilitated the simple and straightforward preparation of Rb1@ManAC4A, leading to excellent batch consistency. Progress in simplicity and reproducibility, as directed by this research, will catalyze the clinical translation of multifunctional materials.
Collapse
Affiliation(s)
- Shihui Li
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin 300071, China
- State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, China
| | - Juan-Juan Li
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin 300071, China
- State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, China
| | - Ying-Ying Zhao
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding 071002, China
| | - Meng-Meng Chen
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin 300071, China
- State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, China
| | - Shan-Shan Su
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding 071002, China
| | - Shun-Yu Yao
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin 300071, China
- State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, China
| | - Ze-Han Wang
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin 300071, China
- State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, China
| | - Xin-Yue Hu
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin 300071, China
- State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, China
| | - Wen-Chao Geng
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin 300071, China
- State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, China
| | - Wei Wang
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin 300071, China
| | - Ke-Rang Wang
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding 071002, China
| | - Dong-Sheng Guo
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin 300071, China
- State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, China
| |
Collapse
|
25
|
Attia MF, Ogunnaike EA, Pitz M, Elbaz NM, Panda DK, Alexander-Bryant A, Saha S, Whitehead DC, Kabanov A. Enhancing Drug Delivery with Supramolecular Amphiphilic Macrocycle Nanoparticles: Selective Targeting of CDK4/6 Inhibitor Palbociclib to Melanoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.21.567974. [PMID: 38045274 PMCID: PMC10690174 DOI: 10.1101/2023.11.21.567974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Drug delivery systems based on amphiphilic supramolecular macrocycles have garnered increased attention over the past two decades due to their ability to successfully formulate nanoparticles. Macrocyclic (MC) materials can self-assemble at lower concentrations without the need for surfactants and polymers, but surfactants are required to form and stabilize nanoparticles at higher concentrations. Using MCs to deliver both hydrophilic and hydrophobic guest molecules is advantageous. We developed two novel types of amphiphilic macrocycle nanoparticles (MC NPs) capable of delivering either Nile Red (NR) (a hydrophobic model) or Rhodamine B (RhB) (a hydrophilic model) fluorescent dyes. We extensively characterized the materials using various techniques to determine size, morphology, stability, hemolysis, fluorescence, loading efficiency (LE), and loading capacity (LC). We then loaded the CDK4/6 inhibitor Palbociclib (Palb) into both MC NPs using a solvent diffusion method. This yielded Palb-MC NPs in the size range of 65-90 nm. They exhibited high stability over time and in fetal bovine serum with negligible toxicity against erythrocytes. Cytotoxicity was minimal when tested against RAW macrophages, human fibroblast HDFn , and adipose stromal cells (ASCs) at higher concentrations of MC NPs. Cell viability studies were conducted with different concentrations of MC NPs, Palb-MC NPs, and free Palb against RAW macrophages, human U-87 GBM, and human M14 melanoma cell lines in vitro. Flow cytometry experiments revealed that blank MC NPs and Palb-MC NPs were selectively targeted to melanoma cells, resulting in cell death compared to the other two cell lines. Future work will focus on studying the biological effect of MC NPs including their binding affinity with molecules/receptors expressed on the M14 and other melanoma cell surface by molecular docking simulations. Subsequently, we will evaluate the MCs as a component of combination therapy in a murine melanoma model. Graphical abstract
Collapse
|
26
|
Grimm LM, Setiadi J, Tkachenko B, Schreiner PR, Gilson MK, Biedermann F. The temperature-dependence of host-guest binding thermodynamics: experimental and simulation studies. Chem Sci 2023; 14:11818-11829. [PMID: 37920355 PMCID: PMC10619620 DOI: 10.1039/d3sc01975f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 09/24/2023] [Indexed: 11/04/2023] Open
Abstract
The thermodynamic parameters of host-guest binding can be used to describe, understand, and predict molecular recognition events in aqueous systems. However, interpreting binding thermodynamics remains challenging, even for these relatively simple molecules, as they are determined by both direct and solvent-mediated host-guest interactions. In this contribution, we focus on the contributions of water to binding by studying binding thermodynamics, both experimentally and computationally, for a series of nearly rigid, electrically neutral host-guest systems and report the temperature-dependent thermodynamic binding contributions ΔGb(T), ΔHb(T), ΔSb(T), and ΔCp,b. Combining isothermal titration calorimetry (ITC) measurements with molecular dynamics (MD) simulations, we provide insight into the binding forces at play for the macrocyclic hosts cucurbit[n]uril (CBn, n = 7-8) and β-cyclodextrin (β-CD) with a range of guest molecules. We find consistently negative changes in heat capacity on binding (ΔCp,b) for all systems studied herein - as well as for literature host-guest systems - indicating increased enthalpic driving forces for binding at higher temperatures. We ascribe these trends to solvation effects, as the solvent properties of water deteriorate as temperature rises. Unlike the entropic and enthalpic contributions to binding, with their differing signs and magnitudes for the classical and non-classical hydrophobic effect, heat capacity changes appear to be a unifying and more general feature of host-guest complex formation in water. This work has implications for understanding protein-ligand interactions and other complex systems in aqueous environments.
Collapse
Affiliation(s)
- Laura M Grimm
- Institute of Nanotechnology (INT), Karlsruhe Institute of Technology (KIT) Hermann-von-Helmholtz Platz 1 76344 Eggenstein-Leopoldshafen Germany
| | - Jeffry Setiadi
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego 9255 Pharmacy Lane La Jolla CA 92093 USA
| | - Boryslav Tkachenko
- Institute of Organic Chemistry, Justus Liebig University Giessen Heinrich-Buff-Ring 17 35392 Giessen Germany
| | - Peter R Schreiner
- Institute of Organic Chemistry, Justus Liebig University Giessen Heinrich-Buff-Ring 17 35392 Giessen Germany
| | - Michael K Gilson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego 9255 Pharmacy Lane La Jolla CA 92093 USA
| | - Frank Biedermann
- Institute of Nanotechnology (INT), Karlsruhe Institute of Technology (KIT) Hermann-von-Helmholtz Platz 1 76344 Eggenstein-Leopoldshafen Germany
| |
Collapse
|
27
|
Tiwari P, Yadav K, Shukla RP, Gautam S, Marwaha D, Sharma M, Mishra PR. Surface modification strategies in translocating nano-vesicles across different barriers and the role of bio-vesicles in improving anticancer therapy. J Control Release 2023; 363:290-348. [PMID: 37714434 DOI: 10.1016/j.jconrel.2023.09.016] [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: 04/17/2023] [Revised: 09/03/2023] [Accepted: 09/06/2023] [Indexed: 09/17/2023]
Abstract
Nanovesicles and bio-vesicles (BVs) have emerged as promising tools to achieve targeted cancer therapy due to their ability to overcome many of the key challenges currently being faced with conventional chemotherapy. These challenges include the diverse and often complex pathophysiology involving the progression of cancer, as well as the various biological barriers that circumvent therapeutic molecules reaching their target site in optimum concentration. The scientific evidence suggests that surface-functionalized nanovesicles and BVs camouflaged nano-carriers (NCs) both can bypass the established biological barriers and facilitate fourth-generation targeting for the improved regimen of treatment. In this review, we intend to emphasize the role of surface-functionalized nanovesicles and BVs camouflaged NCs through various approaches that lead to an improved internalization to achieve improved and targeted oncotherapy. We have explored various strategies that have been employed to surface-functionalize and biologically modify these vesicles, including the use of biomolecule functionalized target ligands such as peptides, antibodies, and aptamers, as well as the targeting of specific receptors on cancer cells. Further, the utility of BVs, which are made from the membranes of cells such as mesenchymal stem cells (MSCs), white blood cells (WBCs), red blood cells (RBCs), platelets (PLTs) as well as cancer cells also been investigated. Lastly, we have discussed the translational challenges and limitations that these NCs can encounter and still need to be overcome in order to fully realize the potential of nanovesicles and BVs for targeted cancer therapy. The fundamental challenges that currently prevent successful cancer therapy and the necessity of novel delivery systems are in the offing.
Collapse
Affiliation(s)
- Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Krishna Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Madhu Sharma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, U.P., India.
| |
Collapse
|
28
|
Ma WJ, Chen HY, Huang YL, Chen JM, Lu TB. Complexation of Fluorofenidone by Cucurbit[7]uril and β-Cyclodextrin: Keto-Enol Tautomerization to Enhance the Solubility. Mol Pharm 2023; 20:4517-4527. [PMID: 37526016 DOI: 10.1021/acs.molpharmaceut.3c00213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
This study is designed to compare drug encapsulation by cucurbit[7]uril and β-cyclodextrin, using fluorofenidone as a model drug. Single-crystal X-ray diffraction analysis was employed to successfully determine the crystal structures of fluorofenidone·H+@cucurbit[7]uril Form, fluorofenidone@cucurbit[7]uril Form, and fluorofenidone@β-cyclodextrin Form. Keto-enol tautomerization of fluorofenidone mediated by cucurbit[7]uril in acid solution is confirmed by crystal structures, pH titration, and nuclear magnetic resonance experiments. However, β-cyclodextrin cannot cause the keto-enol tautomerization of fluorofenidone under similar conditions. The phase solubility study demonstrates that cucurbit[7]uril has a much higher solubilization capacity for fluorofenidone than β-cyclodextrin in 0.1 M HCl since the Kc values of fluorofenidone with cucurbit[7]uril and β-cyclodextrin were 1223.97 ± 452.68 and 78.49 ± 10.56 M-1, respectively. Excellent solubility can be attributed to the keto-enol tautomerization of fluorofenidone under the conditions of cucurbit[7]uril in acid solution. The enol form of fluorofenidone is encapsulated by cucurbit[7]uril by hydrogen bonding interaction and hydrophobic interaction to increase binding affinity. Rat pharmacokinetic studies demonstrate that the area under the plasma concentration-time curve from time 0 to 7 h value of fluorofenidone@cucurbit[7]uril complex is 1.70-fold greater than that of free fluorofenidone, and the mean residence time from time 0 to 7 h is slightly prolonged from 1.29 to 1.76 h (P < 0.01) after oral administration. However, no significant difference is found between fluorofenidone and fluorofenidone@β-cyclodextrin complex. This work indicates that the induction of keto-enol tautomerization of drugs using macrocyclic molecules has the potential to be an effective method to improve their solubility and bioavailability, providing valuable insights for the application of macrocyclic molecules in the biomedical field.
Collapse
Affiliation(s)
- Wen-Juan Ma
- Department of Pharmacy, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Hua-Yu Chen
- Department of Dermatology, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Yong-Liang Huang
- Department of Medicinal Chemistry, Shantou University Medical College, Shantou, Guangdong 515041, P. R. China
| | - Jia-Mei Chen
- Department of Pharmacy, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- School of Chemistry and Chemical Engineering, Tianjin University of Technology, Tianjin 300384, China
| | - Tong-Bu Lu
- Institute for New Energy Materials and Low Carbon Technologies, School of Materials Science and Engineering, Tianjin University of Technology, Tianjin 300384, China
| |
Collapse
|
29
|
Andrianov AK. Noncovalent PEGylation of protein and peptide therapeutics. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1897. [PMID: 37138514 DOI: 10.1002/wnan.1897] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/15/2023] [Accepted: 04/20/2023] [Indexed: 05/05/2023]
Abstract
Clinical applications of protein therapeutics-an advanced generation of drugs characterized by high biological specificity-are rapidly expanding. However, their development is often impeded by unfavorable pharmacokinetic profiles and largely relies on the use of drug delivery systems to prolong their in vivo half-life and suppress undesirable immunogenicity. Although a commercially established PEGylation technology based on protein conjugation with poly(ethylene glycol) (PEG)-protective steric shield resolves some of the challenges, the search for alternatives continues. Noncovalent PEGylation, which mainly relies on multivalent (cooperative) interactions and high affinity (host-guest) complexes formed between protein and PEG offers a number of potential advantages. Among them are dynamic or reversible protection of the protein with minimal loss of biological activity, drastically lower manufacturing costs, "mix-and-match" formulations approaches, and expanded scope of PEGylation targets. While a great number of innovative chemical approaches have been proposed in recent years, the ability to effectively control the stability of noncovalently assembled protein-PEG complexes under physiological conditions presents a serious challenge for the commercial development of the technology. In an attempt to identify critical factors affecting pharmacological behavior of noncovalently linked complexes, this Review follows a hierarchical analysis of various experimental techniques and resulting supramolecular architectures. The importance of in vivo administration routes, degradation patterns of PEGylating agents, and a multitude of potential exchange reactions with constituents of physiological compartments are highlighted. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Alexander K Andrianov
- Institute of Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland, USA
| |
Collapse
|
30
|
Tong F, Zhou Y, Xu Y, Chen Y, Yudintceva N, Shevtsov M, Gao H. Supramolecular nanomedicines based on host-guest interactions of cyclodextrins. EXPLORATION (BEIJING, CHINA) 2023; 3:20210111. [PMID: 37933241 PMCID: PMC10624390 DOI: 10.1002/exp.20210111] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 02/09/2023] [Indexed: 11/08/2023]
Abstract
In the biomedical and pharmaceutical fields, cyclodextrin (CD) is undoubtedly one of the most frequently used macrocyclic compounds as the host molecule because it has good biocompatibility and can increase the solubility, bioavailability, and stability of hydrophobic drug guests. In this review, we generalized the unique properties of CDs, CD-related supramolecular nanocarriers, supramolecular controlled release systems, and targeting systems based on CDs, and introduced the paradigms of these nanomedicines. In addition, we also discussed the prospects and challenges of CD-based supramolecular nanomedicines to facilitate the development and clinical translation of these nanomedicines.
Collapse
Affiliation(s)
- Fan Tong
- Key Laboratory of Drug Targeting and Drug Delivery SystemsWest China School of PharmacySichuan UniversityChengduChina
| | - Yang Zhou
- Key Laboratory of Drug Targeting and Drug Delivery SystemsWest China School of PharmacySichuan UniversityChengduChina
| | - Yanyan Xu
- Key Laboratory of Drug Targeting and Drug Delivery SystemsWest China School of PharmacySichuan UniversityChengduChina
| | - Yuxiu Chen
- Key Laboratory of Drug Targeting and Drug Delivery SystemsWest China School of PharmacySichuan UniversityChengduChina
| | - Natalia Yudintceva
- Institute of Cytology of the Russian Academy of Sciences (RAS)St. PetersburgRussia
| | - Maxim Shevtsov
- Institute of Cytology of the Russian Academy of Sciences (RAS)St. PetersburgRussia
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery SystemsWest China School of PharmacySichuan UniversityChengduChina
| |
Collapse
|
31
|
Alrabiah H, Ali EA, Alsalahi RA, Attwa MW, Mostafa GAE. Fabrication and Applications of Potentiometric Membrane Sensors Based on γ-Cyclodextrin and Calixarene as Ionophores for the Determination of a Histamine H1-Receptor Antagonist: Fexofenadine. Polymers (Basel) 2023; 15:2808. [PMID: 37447454 DOI: 10.3390/polym15132808] [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] [Received: 05/05/2023] [Revised: 06/18/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Supramolecular fexofenadine sensors have been constructed. Although noncovalent intermolecular and intramolecular interactions, which are far weaker than covalent contacts, are the main focus of supramolecular chemistry, they can be used to create sensors with an exceptional affinity for a target analyte. The objective of the current research study is to adapt two PVC membrane sensors into an electrochemical approach for the dosage form determination of histamine H1-receptor antagonists: fexofenadine. The general performance characteristics of two new modified potentiometric membrane sensors responsive to fexofenadine hydrochloride were established. The technique was based on the employment of γ-cyclodextrin (CD) (sensor 1), 4-tert-butylcalix[8]arene (calixarene) (sensor 2) as an ionophore, potassium tetrakis (4-chlorophenyl) borate (KTpClPB) as an ion additive, and (o-NPOE) as a plasticizer for sensors 1 and 2. The sensors showed fast responses over a wide fexofenadine concentration range (1 × 10-2 to 4.5 (4.7) × 10-6 M), with detection limits of 1.3 × 10-6 M and 1.4 × 10-6 M for sensors 1 and 2, respectively, in the pH range of 2-8. The tested sensors exhibit the fexofenadine near-Nernstian cationic response at 56 and 58 mV/decade for sensors 1 and 2, respectively. The sensors exhibit good stability, fast response times, accuracy, precision, and longer life for fexofenadine. Throughout the day and between days, the sensors exhibit good recovery and low relative standard deviations. Fexofenadine in its pure, dose form has been identified with success using the modified sensors. The sensors were employed as end-point indications for the titration of fexofenadine with NaTPB.
Collapse
Affiliation(s)
- Haitham Alrabiah
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Essam A Ali
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Rashad A Alsalahi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Mohamed W Attwa
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Gamal A E Mostafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| |
Collapse
|
32
|
Wu J, Sun X, Li X, Li X, Feng W, Yuan L. Multi-Responsive Molecular Encapsulation and Release Based on Hydrogen-Bonded Azo-Macrocycle. Molecules 2023; 28:molecules28114437. [PMID: 37298912 DOI: 10.3390/molecules28114437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/27/2023] [Accepted: 05/27/2023] [Indexed: 06/12/2023] Open
Abstract
Research on stimuli-responsive host-guest systems is at the cutting edge of supramolecular chemistry, owing to their numerous potential applications such as catalysis, molecular machines, and drug delivery. Herein, we present a multi-responsive host-guest system comprising azo-macrocycle 1 and 4,4'-bipyridinium salt G1 for pH-, photo-, and cation- responsiveness. Previously, we reported a novel hydrogen-bonded azo-macrocycle 1. The size of this host can be controlled through light-induced E↔Z photo-isomerization of the constituent azo-benzenes. The host is found in this work to be capable of forming stable complexes with bipyridinium/pyridinium salts, and implementing guest capture and release with G1 under light in a controlled manner. The binding and release of the guest in the complexes can also be easily controlled reversibly by using acid and base. Moreover, the cation competition-induced dissociation of the complex 1a2⊃G1 is achieved. These findings are expected to be useful in regulating encapsulation for sophisticated supramolecular systems.
Collapse
Affiliation(s)
- Jinyang Wu
- College of Chemistry, Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, China
| | - Xuan Sun
- College of Chemistry, Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, China
| | - Xianghui Li
- College of Chemistry, Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, China
| | - Xiaowei Li
- College of Chemistry, Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, China
| | - Wen Feng
- College of Chemistry, Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, China
| | - Lihua Yuan
- College of Chemistry, Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, China
| |
Collapse
|
33
|
Shukla S, Sagar B, Sood AK, Gaur A, Batra S, Gulati S. Supramolecular Chemotherapy with Cucurbit[ n]urils as Encapsulating Hosts. ACS APPLIED BIO MATERIALS 2023. [PMID: 37224296 DOI: 10.1021/acsabm.3c00244] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
The cucurbit[n]urils (CB[n]) belong to the field of relatively young supramolecules which act as containers for a large variety of guests and are being explored extensively for their numerous biomedical applications. This includes drug formulation and delivery, controlled drug release, photodynamic therapy, sensing for bioanalytical purposes, etc. These supramolecular host-guest systems have distinctive recognition properties and have successfully been shown to enhance the in vitro and in vivo utility of various chemotherapeutic agents. The CB[n]s are tailored to optimize their application in payload delivery and diagnostics and in lowering the toxicity of existing drugs. This review has listed the recent studies on working mechanisms and host-guest complexation of the biologically vital molecules with CB[n] and highlighted their implementation in anticancer therapeutics. Various modifications in CB-drug inclusion compounds like CB supramolecular nanoarchitectures along with application in photodynamic therapy, which has shown potential as targeted drug delivery vehicles in cancer chemotherapy, have also been discussed.
Collapse
|
34
|
Wu X, Yang J, Xing J, Lyu Y, Zou R, Wang X, Yao J, Zhang D, Qi D, Shao G, Wu A, Li J. Using host-guest interactions at the interface of quantum dots to load drug molecules for biocompatible, safe, and effective chemo-photodynamic therapy against cancer. J Mater Chem B 2023. [PMID: 37161740 DOI: 10.1039/d3tb00592e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Combining photodynamic therapy (PDT) and chemotherapy (CHT) by loading an anti-cancer drug and a photosensitizer (PS) into the same delivery nanosystem has been proposed as an effective approach to achieve synergistic effects for a safe cancer treatment. However, exploring an ideal delivery nanosystem has been challenging, because the noncovalent interactions must be maintained between the multiple components to produce a stable yet responsive nanostructure that takes into account the encapsulation of drug molecules. We addressed this issue by engineering the interfacial interaction between Ag2S quantum dots (QDs) using a pillararene derivative to direct the co-self-assembly of the entire system. The high surface area-to-volume ratio of the Ag2S QDs provided ample hydrophobic space to accommodate the anti-drug molecule doxrubicine. Moreover, Ag2S QDs served as PSs triggered by 808 nm near-infrared (NIR) light and also as carriers for high-efficiency delivery of drug molecules to the tumor site. Drug release experiments showed smart drug release under the acidic microenvironments (pH 5.5) in tumor cells. Additionally, the Ag2S QDs demonstrated outstanding PDT ability under NIR light, as confirmed by extracellular and intracellular reactive oxygen species generation. Significant treatment efficacy of the chemo-photodynamic synergistic therapy for cancer using the co-delivery system was demonstrated via in vitro and in vivo studies. These findings suggest that our system offers intelligent control of CHT and PDT, which will provide a promising strategy for constructing hybrid systems with synergistic effects for advanced applications in biomedicine, catalysis, and optoelectronics.
Collapse
Affiliation(s)
- Xiaoxia Wu
- MediCity Research Laboratory, University of Turku, Tykistökatu 6, FI-20520 Turku, Finland.
- Department of Interventional Radiology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences (CAS), Hangzhou 310022, China
| | - Jinghui Yang
- MediCity Research Laboratory, University of Turku, Tykistökatu 6, FI-20520 Turku, Finland.
- Department of Chemistry, University of Turku, Vatselankatu 2, FI-20014 Turku, Finland
| | - Jie Xing
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo 315201, China.
| | - Yonglei Lyu
- MediCity Research Laboratory, University of Turku, Tykistökatu 6, FI-20520 Turku, Finland.
- Department of Chemistry, University of Turku, Vatselankatu 2, FI-20014 Turku, Finland
| | - Ruifen Zou
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo 315201, China.
| | - Xin Wang
- MediCity Research Laboratory, University of Turku, Tykistökatu 6, FI-20520 Turku, Finland.
- Department of Chemistry, University of Turku, Vatselankatu 2, FI-20014 Turku, Finland
| | - Junlie Yao
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo 315201, China.
| | - Dinghu Zhang
- Department of Interventional Radiology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences (CAS), Hangzhou 310022, China
| | - Dawei Qi
- MediCity Research Laboratory, University of Turku, Tykistökatu 6, FI-20520 Turku, Finland.
| | - Guoliang Shao
- Department of Interventional Radiology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences (CAS), Hangzhou 310022, China
| | - Aiguo Wu
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo 315201, China.
| | - Jianwei Li
- MediCity Research Laboratory, University of Turku, Tykistökatu 6, FI-20520 Turku, Finland.
- Hainan Provincial Key Laboratory of Fine Chem, School of Chemical Engineering and Technology, Hainan University, Haikou 570228, China
| |
Collapse
|
35
|
Li JJ, Rong RX, Yang Y, Hu ZY, Hu B, Zhao YY, Li HB, Hu XY, Wang KR, Guo DS. Triple targeting host-guest drug delivery system based on lactose-modified azocalix[4]arene for tumor ablation. MATERIALS HORIZONS 2023; 10:1689-1696. [PMID: 36825769 DOI: 10.1039/d3mh00018d] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Host-guest drug delivery systems (HGDDSs) have been studied in an effort to modify the characteristics of therapeutic agents through noncovalent interactions, reduce toxic side effects and improve therapeutic effects. However, it is still an important task to continuously improve the targeting ability of HGDDSs, which is conducive to the development of precision medicine. Herein, we utilize the lactose-modified azocalix[4]arene (LacAC4A) as a triple targeting drug carrier customized for antitumor purposes. LacAC4A integrates three targeting features, passive targeting through the enhancing permeability and retention effect, active targeting by the interactions of lactose and the asialoglycoprotein receptors on the surface of tumor cells, and stimuli-responsive targeting via the reduction of the azo group under a hypoxia microenvironment. After loading doxorubicin (DOX) in LacAC4A, the supramolecular nanoformulation DOX@LacAC4A clearly showed the effective suppression of tumor growth through in vivo experiments. LacAC4A can achieve effective targeting, rapid release, and improve drug bioavailability. This design principle will provide a new material for drug delivery systems.
Collapse
Affiliation(s)
- Juan-Juan Li
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China.
| | - Rui-Xue Rong
- Department of Medical Microbiology and Immunology, School of Basic Medical Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Key Laboratory of Chemical Biology of Hebei Province, Medical Comprehensive Experimental Center, Hebei University, Baoding 071002, China
| | - Yan Yang
- Department of Medical Microbiology and Immunology, School of Basic Medical Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Key Laboratory of Chemical Biology of Hebei Province, Medical Comprehensive Experimental Center, Hebei University, Baoding 071002, China
| | - Zong-Ying Hu
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China.
| | - Bing Hu
- College of Chemistry and Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding 071002, China.
| | - Ying-Ying Zhao
- College of Chemistry and Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding 071002, China.
| | - Hua-Bin Li
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China.
| | - Xin-Yue Hu
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China.
| | - Ke-Rang Wang
- College of Chemistry and Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding 071002, China.
| | - Dong-Sheng Guo
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China.
| |
Collapse
|
36
|
Liu Y, Wu Y, Luo Z, Li M. Designing supramolecular self-assembly nanomaterials as stimuli-responsive drug delivery platforms for cancer therapy. iScience 2023; 26:106279. [PMID: 36936787 PMCID: PMC10014307 DOI: 10.1016/j.isci.2023.106279] [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] [Indexed: 03/03/2023] Open
Abstract
Stimuli-responsive nanomaterials have attracted substantial interest in cancer therapy, as they hold promise to deliver anticancer agents to tumor sites in a precise and on-demand manner. Interestingly, supramolecular chemistry is a burgeoning discipline that entails the reversible bonding between components at the molecular and nanoscale levels, and the recent advances in this area offer the possibility to design nanotherapeutics with improved controllability and functionality for cancer therapy. Herein, we provide a comprehensive summary of typical non-covalent interaction modes, which primarily include hydrophobic interaction, hydrogel bonding, host-guest interaction, π-π stacking, and electrostatic interaction. Special emphasis is placed on the implications of these interaction modes to design novel stimuli-responsive drug delivery principles and concepts, aiming to enhance the spatial, temporal, and dosage precision of drug delivery to cancer cells. Finally, future perspectives are discussed to highlight current challenges and future opportunities in self-assembly-based stimuli-responsive drug delivery nanotechnologies for cancer therapy.
Collapse
Affiliation(s)
- Yingqi Liu
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Yunyun Wu
- Chongqing Municipal Center for Disease Control and Prevention, Chongqing 400042, China
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| |
Collapse
|
37
|
Mariottini D, Idili A, Ercolani G, Ricci F. Thermo-Programmed Synthetic DNA-Based Receptors. ACS NANO 2023; 17:1998-2006. [PMID: 36689298 PMCID: PMC9933611 DOI: 10.1021/acsnano.2c07039] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 01/18/2023] [Indexed: 06/17/2023]
Abstract
Herein, we present a generalizable and versatile strategy to engineer synthetic DNA ligand-binding devices that can be programmed to load and release a specific ligand at a defined temperature. We do so by re-engineering two model DNA-based receptors: a triplex-forming bivalent DNA-based receptor that recognizes a specific DNA sequence and an ATP-binding aptamer. The temperature at which these receptors load/release their ligands can be finely modulated by controlling the entropy associated with the linker connecting the two ligand-binding domains. The availability of a set of receptors with tunable and reversible temperature dependence allows achieving complex load/release behavior such as sustained ligand release over a wide temperature range. Similar programmable thermo-responsive synthetic ligand-binding devices can be of utility in applications such as drug delivery and production of smart materials.
Collapse
Affiliation(s)
- Davide Mariottini
- Chemistry
Department, University of Rome, Tor Vergata, Via della Ricerca Scientifica, 00133, Rome, Italy
| | - Andrea Idili
- Chemistry
Department, University of Rome, Tor Vergata, Via della Ricerca Scientifica, 00133, Rome, Italy
| | - Gianfranco Ercolani
- Chemistry
Department, University of Rome, Tor Vergata, Via della Ricerca Scientifica, 00133, Rome, Italy
| | - Francesco Ricci
- Chemistry
Department, University of Rome, Tor Vergata, Via della Ricerca Scientifica, 00133, Rome, Italy
| |
Collapse
|
38
|
Yan M, Zhou J. Pillararene-Based Supramolecular Polymers for Cancer Therapy. Molecules 2023; 28:molecules28031470. [PMID: 36771136 PMCID: PMC9919256 DOI: 10.3390/molecules28031470] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Supramolecular polymers have attracted considerable interest due to their intriguing features and functions. The dynamic reversibility of noncovalent interactions endows supramolecular polymers with tunable physicochemical properties, self-healing, and externally stimulated responses. Among them, pillararene-based supramolecular polymers show great potential for biomedical applications due to their fascinating host-guest interactions and easy modification. Herein, we summarize the state of the art of pillararene-based supramolecular polymers for cancer therapy and illustrate its developmental trend and future perspective.
Collapse
|
39
|
Xiao T, Elmes R, Yao Y. Editorial: Host-guest chemistry of macrocycles- Volume II. Front Chem 2023; 11:1162019. [PMID: 36895319 PMCID: PMC9990904 DOI: 10.3389/fchem.2023.1162019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/23/2023] Open
Affiliation(s)
- Tangxin Xiao
- School of Petrochemical Engineering, Changzhou University, Changzhou, China
| | - Robert Elmes
- Department of Chemistry, Maynooth University, National University of Ireland, Maynooth, Ireland.,Synthesis and Solid-State Pharmaceutical Centre (SSPC), Maynooth University, National University of Ireland, Maynooth, Ireland
| | - Yong Yao
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, China
| |
Collapse
|
40
|
Gates BD, Vyletel JB, Zou L, Webber MJ. Multivalent Cucurbituril Dendrons for Cell Membrane Engineering with Supramolecular Receptors. Bioconjug Chem 2022; 33:2262-2268. [PMID: 35802933 PMCID: PMC11144120 DOI: 10.1021/acs.bioconjchem.2c00242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The affinity possible from certain supramolecular motifs rivals that for some of the best-recognized interactions in biology. Cucurbit[7]uril (CB[7]) macrocycles, for example, are capable of achieving affinities in their binding to certain guests that rival that of biotin-avidin. Supramolecular host-guest recognition between CB[7] and certain guests has been demonstrated to spatially localize guest-linked agents to desired sites in vivo, offering opportunities to better exploit this affinity axis for applications in biomedicine. Herein, architectures of CB[7] are prepared from a polyamidoamine (PAMAM) dendrimer scaffold, installing a PEG-linked cholesterol anchor on the opposite end of the dendron to facilitate cell membrane integration. Cells are then modified with this dendritic CB[7] construct in vitro, demonstrating the ability to deliver a model guest-linked agent to the cell membrane. This approach to realize synthetic supramolecular "membrane receptors" may be leveraged in the future for in situ imaging or modulation of cell-based therapies or to facilitate a synthetic supramolecular recognition axis on the cell membrane.
Collapse
Affiliation(s)
- Brant D. Gates
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, IN 46556 USA
| | - Jackson B. Vyletel
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, IN 46556 USA
| | - Lei Zou
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, IN 46556 USA
| | - Matthew J. Webber
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, IN 46556 USA
| |
Collapse
|
41
|
Xian S, VandenBerg MA, Xiang Y, Yu S, Webber MJ. Glucose-Responsive Injectable Thermogels via Dynamic-Covalent Cross-Linking of Pluronic Micelles. ACS Biomater Sci Eng 2022; 8:4873-4885. [PMID: 36317822 DOI: 10.1021/acsbiomaterials.2c00979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Sijie Xian
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Michael A. VandenBerg
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Yuanhui Xiang
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Sihan Yu
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Matthew J. Webber
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| |
Collapse
|
42
|
Maksoud FJ, Velázquez de la Paz MF, Hann AJ, Thanarak J, Reilly GC, Claeyssens F, Green NH, Zhang YS. Porous biomaterials for tissue engineering: a review. J Mater Chem B 2022; 10:8111-8165. [PMID: 36205119 DOI: 10.1039/d1tb02628c] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The field of biomaterials has grown rapidly over the past decades. Within this field, porous biomaterials have played a remarkable role in: (i) enabling the manufacture of complex three-dimensional structures; (ii) recreating mechanical properties close to those of the host tissues; (iii) facilitating interconnected structures for the transport of macromolecules and cells; and (iv) behaving as biocompatible inserts, tailored to either interact or not with the host body. This review outlines a brief history of the development of biomaterials, before discussing current materials proposed for use as porous biomaterials and exploring the state-of-the-art in their manufacture. The wide clinical applications of these materials are extensively discussed, drawing on specific examples of how the porous features of such biomaterials impact their behaviours, as well as the advantages and challenges faced, for each class of the materials.
Collapse
Affiliation(s)
- Fouad Junior Maksoud
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA.
| | - María Fernanda Velázquez de la Paz
- Department of Materials Science and Engineering, Kroto Research Building, North Campus, Broad Lane, University of Sheffield, Sheffield, S3 7HQ, UK.
| | - Alice J Hann
- Department of Materials Science and Engineering, Kroto Research Building, North Campus, Broad Lane, University of Sheffield, Sheffield, S3 7HQ, UK.
| | - Jeerawan Thanarak
- Department of Materials Science and Engineering, Kroto Research Building, North Campus, Broad Lane, University of Sheffield, Sheffield, S3 7HQ, UK.
| | - Gwendolen C Reilly
- Department of Materials Science and Engineering, Kroto Research Building, North Campus, Broad Lane, University of Sheffield, Sheffield, S3 7HQ, UK. .,INSIGNEO Institute for in silico Medicine, University of Sheffield, S3 7HQ, UK
| | - Frederik Claeyssens
- Department of Materials Science and Engineering, Kroto Research Building, North Campus, Broad Lane, University of Sheffield, Sheffield, S3 7HQ, UK. .,INSIGNEO Institute for in silico Medicine, University of Sheffield, S3 7HQ, UK
| | - Nicola H Green
- Department of Materials Science and Engineering, Kroto Research Building, North Campus, Broad Lane, University of Sheffield, Sheffield, S3 7HQ, UK. .,INSIGNEO Institute for in silico Medicine, University of Sheffield, S3 7HQ, UK
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA.
| |
Collapse
|
43
|
GOH SQ, CHAN QH, ADNAN R, RAHIM NY. Preparation and characterisation of ethambutol with β-cyclodextrin: a comprehensive molecular insight into host-guest interaction. Turk J Chem 2022; 46:1946-1955. [PMID: 37621338 PMCID: PMC10446918 DOI: 10.55730/1300-0527.3493] [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: 05/25/2022] [Revised: 12/19/2022] [Accepted: 08/10/2022] [Indexed: 12/24/2022] Open
Abstract
The increase in new cases of drug resistance to first-line drugs such as ethambutol (ETB) makes it necessary to develop improvements for antituberculosis drugs. A new method for improving the bioavailability of active pharmaceutical ingredient (API) was investigated for preventing drug resistance and side-effects of antituberculosis drugs. In this study, an antituberculosis drug delivery system using β-cyclodextrin (β-CD) as the supramolecular carrier of ETB was prepared using the kneading method. The inclusion behaviour of β-CD/ETB inclusion complex in solid and liquid state was investigated. The inclusion complex was characterized using Fourier transform infrared spectroscopy, thermogravimetric analysis, nuclear magnetic resonance (NMR) spectroscopy, and UV-visible spectroscopy. The 1H and nuclear Overhauser effect spectroscopy NMR results indicated the hydrophobic interaction between β-CD and ETB. Meanwhile, the Benesi-Hildebrand equation was used to calculate the formation constant (K) of β-CD/ETB complex in natural condition, pH4, and pH9, which were 105.43, 107.06, and 119.11, respectively. The stoichiometry ratio of β-CD/ETB complex was proven to be 1:1.
Collapse
Affiliation(s)
- Soen Qeng GOH
- School of Chemical Sciences, Universiti Sains Malaysia, Penang,
Malaysia
| | - Qiao Hui CHAN
- School of Chemical Sciences, Universiti Sains Malaysia, Penang,
Malaysia
| | - Rohana ADNAN
- School of Chemical Sciences, Universiti Sains Malaysia, Penang,
Malaysia
| | - Nurul Yani RAHIM
- School of Chemical Sciences, Universiti Sains Malaysia, Penang,
Malaysia
| |
Collapse
|
44
|
Li S, Ma R, Hu XY, Li HB, Geng WC, Kong X, Zhang C, Guo DS. Drug in Drug: A Host-Guest Formulation of Azocalixarene with Hydroxychloroquine for Synergistic Anti-Inflammation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2203765. [PMID: 35680644 DOI: 10.1002/adma.202203765] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/09/2022] [Indexed: 06/15/2023]
Abstract
Macrocyclic delivery and therapeutics are two significant topics in supramolecular biomedicine. The functional integration of these topics would open new avenues for treating diseases synergistically. However, these two individual topics have only been occasionally merged, probably because of the lack of functionalized design of macrocyclic host and the lack of efficient recognition between host and guest drugs. Herein, a "drug-in-drug" strategy is proposed, in which an active drug is encapsulated by a macrocycle possessing therapeutic activity to form a multifunctional supramolecular active pharmaceutical ingredient. As a proof-of-concept, a complex of hydroxychloroquine (HCQ) with sulfonated azocalix[4]arene (HCQ@SAC4A) is prepared to treat rheumatoid arthritis (RA) in a combined fashion. SAC4A is a therapeutic agent that exhibits scavenging capacity for reactive oxygen species and exerts an anti-inflammatory effect. It is also a hypoxia-responsive carrier that can deliver HCQ directly to the inflammatory articular cavity. Consequently, HCQ@SAC4A achieves the synergistic anti-inflammatory effect on both inflamed RAW 264.7 cells and RA rats. This effect is attributed to the temporal and spatial consistency of the two active ingredients of the complex. As a new paradigm for combinational therapy, the drug-in-drug strategy advances in easy preparation, mix-and-match combination, and precise ratiometric control.
Collapse
Affiliation(s)
- Shihui Li
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| | - Rong Ma
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| | - Xin-Yue Hu
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| | - Hua-Bin Li
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| | - Wen-Chao Geng
- Key Laboratory of Systems Microbial Biotechnology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
| | - Xianglei Kong
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| | - Chao Zhang
- Orthopedics Department, The First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300380, China
| | - Dong-Sheng Guo
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| |
Collapse
|
45
|
Akmal M, Wahyuningrum D, Ivansyah A. Theoretical Insight and Molecular Recognition of Oxatub[4]arene-based Organic Macrocycle as a Supramolecular Host for Antipsychotic Drug Risperidone. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.120195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
46
|
Braegelman AS, Ollier RC, Su B, Addonizio CJ, Zou L, Cole SL, Webber MJ. Macromolecular Solute Transport in Supramolecular Hydrogels Spanning Dynamic to Quasi-Static States. ACS APPLIED BIO MATERIALS 2022; 5:10.1021/acsabm.2c00165. [PMID: 35623099 PMCID: PMC10019485 DOI: 10.1021/acsabm.2c00165] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Hydrogels prepared from supramolecular cross-linking motifs are appealing for use as biomaterials and drug delivery technologies. The inclusion of macromolecules (e.g., protein therapeutics) in these materials is relevant to many of their intended uses. However, the impact of dynamic network cross-linking on macromolecule diffusion must be better understood. Here, hydrogel networks with identical topology but disparate cross-link dynamics are explored. These materials are prepared from cross-linking with host-guest complexes of the cucurbit[7]uril (CB[7]) macrocycle and two guests of different affinity. Rheology confirms differences in bulk material dynamics arising from differences in cross-link thermodynamics. Fluorescence recovery after photobleaching (FRAP) provides insight into macromolecule diffusion as a function of probe molecular weight and hydrogel network dynamics. Together, both rheology and FRAP enable the estimation of the mean network mesh size, which is then related to the solute hydrodynamic diameters to further understand macromolecule diffusion. Interestingly, the thermodynamics of host-guest cross-linking are correlated with a marked deviation from classical diffusion behavior for higher molecular weight probes, yielding solute aggregation in high-affinity networks. These studies offer insights into fundamental macromolecular transport phenomena as they relate to the association dynamics of supramolecular networks. Translation of these materials from in vitro to in vivo is also assessed by bulk release of an encapsulated macromolecule. Contradictory in vitro to in vivo results with inverse relationships in release between the two hydrogels underscores the caution demanded when translating supramolecular biomaterials into application.
Collapse
Affiliation(s)
- Adam S. Braegelman
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, IN 46556 USA
- University of Notre Dame, Bioengineering PhD Program, Notre Dame, IN 46556 USA
| | - Rachel C. Ollier
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, IN 46556 USA
| | - Bo Su
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, IN 46556 USA
| | - Christopher J. Addonizio
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, IN 46556 USA
| | - Lei Zou
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, IN 46556 USA
| | - Sara L. Cole
- University of Notre Dame, Integrated Imaging Facility, Notre Dame, IN 46556 USA
| | - Matthew J. Webber
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, IN 46556 USA
| |
Collapse
|
47
|
Carbajo‐Gordillo AI, López‐Fernández J, Benito JM, Blanco JLJ, Santana‐Armas ML, Marcelo G, Giorgio CD, Przybylski C, Mellet CO, Ilarduya CT, Mendicuti F, Fernández JMG. Enhanced Gene Delivery Triggered by Dual pH/Redox Responsive Host‐Guest Dimerization of Cyclooligosaccharide Star Polycations. Macromol Rapid Commun 2022; 43:e2200145. [DOI: 10.1002/marc.202200145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/23/2022] [Indexed: 11/10/2022]
Affiliation(s)
| | - José López‐Fernández
- Instituto de Investigaciones Químicas (IIQ) C/ Américo Vespucio 49 Sevilla 41092 Spain
| | - Juan M. Benito
- Instituto de Investigaciones Químicas (IIQ) C/ Américo Vespucio 49 Sevilla 41092 Spain
| | - José L. Jiménez Blanco
- Department of Organic Chemistry Faculty of Chemistry University of Seville C/ Profesor García González 1 Seville 41012 Spain
| | - María L. Santana‐Armas
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition University of Navarra Pamplona 31080 Spain
| | - Gema Marcelo
- Department of Analytical Chemistry Physical Chemistry and Chemical Engineering Faculty of Chemistry University of Alcalá Alcalá de Henares Madrid Spain
| | - Christophe Di Giorgio
- Institut de Chimie Nice UMR 7272 Université Côte d'Azur 28, Avenue de Valrose Nice F‐06108 France
| | - Cédric Przybylski
- Institut Parisien de Chimie Moléculaire (IPCM) CNRS Sorbonne Université Paris France
| | - Carmen Ortiz Mellet
- Department of Organic Chemistry Faculty of Chemistry University of Seville C/ Profesor García González 1 Seville 41012 Spain
| | - Conchita Tros Ilarduya
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition University of Navarra Pamplona 31080 Spain
| | - Francisco Mendicuti
- Department of Analytical Chemistry Physical Chemistry and Chemical Engineering Faculty of Chemistry University of Alcalá Alcalá de Henares Madrid Spain
| | | |
Collapse
|
48
|
Omar J, Ponsford D, Dreiss CA, Lee TC, Loh XJ. Supramolecular Hydrogels: Design Strategies and Contemporary Biomedical Applications. Chem Asian J 2022; 17:e202200081. [PMID: 35304978 DOI: 10.1002/asia.202200081] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/08/2022] [Indexed: 12/19/2022]
Abstract
Self-assembly of supramolecular hydrogels is driven by dynamic, non-covalent interactions between molecules. Considerable research effort has been exerted to fabricate and optimise supramolecular hydrogels that display shear-thinning, self-healing, and reversibility, in order to develop materials for biomedical applications. This review provides a detailed overview of the chemistry behind the dynamic physicochemical interactions that sustain hydrogel formation (hydrogen bonding, hydrophobic interactions, ionic interactions, metal-ligand coordination, and host-guest interactions). Novel design strategies and methodologies to create supramolecular hydrogels are highlighted, which offer promise for a wide range of applications, specifically drug delivery, wound healing, tissue engineering and 3D bioprinting. To conclude, future prospects are briefly discussed, and consideration given to the steps required to ultimately bring these biomaterials into clinical settings.
Collapse
Affiliation(s)
- Jasmin Omar
- Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, SE1 9NH, London, UK.,Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Daniel Ponsford
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), Singapore.,Department of Chemistry, University College London, London, WC1H 0AJ, UK.,Institute for Materials Discovery, University College London, London, WC1E 7JE, UK
| | - Cécile A Dreiss
- Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, SE1 9NH, London, UK
| | - Tung-Chun Lee
- Department of Chemistry, University College London, London, WC1H 0AJ, UK.,Institute for Materials Discovery, University College London, London, WC1E 7JE, UK
| | - Xian Jun Loh
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), Singapore.,Department of Materials Science and Engineering, National University of Singapore, Singapore
| |
Collapse
|
49
|
Zhang TX, Hou X, Kong Y, Yang F, Yue YX, Shah MR, Li HB, Huang F, Liu J, Guo DS. A hypoxia-responsive supramolecular formulation for imaging-guided photothermal therapy. Theranostics 2022; 12:396-409. [PMID: 34987652 PMCID: PMC8690909 DOI: 10.7150/thno.67036] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/27/2021] [Indexed: 12/13/2022] Open
Abstract
Photothermal agents (PTAs) based on organic small-molecule dyes emerge as promising theranostic strategy in imaging and photothermal therapy (PTT). However, hydrophobicity, photodegradation, and low signal-to-noise ratio impede their transformation from bench to bedside. In this study, a novel supramolecular PTT formulation by a stimuli-responsive macrocyclic host is prepared to overcome these obstacles of organic small-molecule PTAs. Methods: Sulfonated azocalix[4]arene (SAC4A) was synthesized as a hypoxia-responsive macrocyclic host. Taking IR780 as an example, the supramolecular nanoformulation IR780@SAC4A was constructed by grinding method, and its solubility, photostability, and photothermal conversion were evaluated. The hypoxia tumor-selective imaging and supramolecular PTT of IR780@SAC4A were further evaluated in vitro and in vivo. Results: IR780@SAC4A is capable of enhancing the solubility, photostability, and photothermal conversion of IR780 significantly, which achieve this supramolecular formulation with good imaging-guided PTT efficacy in vitro and in vivo. Conclusions: This study demonstrates that the supramolecular PTT strategy is a promising cancer theranostic method. Moreover, this supramolecular approach is applicative to construct kinds of supramolecular PTAs, opening a general avenue for extending smart PTT formulations.
Collapse
|
50
|
Molecular Recognition by Pillar[5]arenes: Evidence for Simultaneous Electrostatic and Hydrophobic Interactions. Pharmaceutics 2021; 14:pharmaceutics14010060. [PMID: 35056956 PMCID: PMC8777861 DOI: 10.3390/pharmaceutics14010060] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/17/2021] [Accepted: 12/22/2021] [Indexed: 11/21/2022] Open
Abstract
The formation of inclusion complexes between alkylsulfonate guests and a cationic pillar[5]arene receptor in water was investigated by NMR and ITC techniques. The results show the formation of host-guest complexes stabilized by electrostatic interactions and hydrophobic effects with binding constants of up to 107 M−1 for the guest with higher hydrophobic character. Structurally, the alkyl chain of the guest is included in the hydrophobic aromatic cavity of the macrocycle while the sulfonate groups are held in the multicationic portal by ionic interactions.
Collapse
|