1
|
Slaughter KV, Donders EN, Jones MS, Sabbah SG, Elliott MJ, Shoichet BK, Cescon DW, Shoichet MS. Ionizable Drugs Enable Intracellular Delivery of Co-Formulated siRNA. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403701. [PMID: 39148215 DOI: 10.1002/adma.202403701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/02/2024] [Indexed: 08/17/2024]
Abstract
Targeting complementary pathways in diseases such as cancer can be achieved with co-delivery of small interfering ribonucleic acid (siRNA) and small molecule drugs; however, current formulation strategies are typically limited to one, but not both. Here, ionizable small molecule drugs and siRNA are co-formulated in drug-rich nanoparticles. Ionizable analogs of the selective estrogen receptor degrader fulvestrant self-assemble into colloidal drug aggregates and cause endosomal disruption, allowing co-delivery of siRNA against a non-druggable target. siRNA is encapsulated in lipid-stabilized, drug-rich colloidal nanoparticles where the ionizable lipid used in conventional lipid nanoparticles is replaced with an ionizable fulvestrant analog. The selection of an appropriate phospholipid and formulation buffer enables endocytosis and potent reporter gene knockdown in cancer cells. Importantly, siRNA targeting cyclin E1 is effectively delivered to drug-resistant breast cancer cells, demonstrating the utility of this approach. This strategy opens the possibility of using ionizable drugs to co-deliver RNA and ultimately improve therapeutic outcomes.
Collapse
Affiliation(s)
- Kai V Slaughter
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, M5S 3G9, Canada
- Donnelly Centre, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
| | - Eric N Donders
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, M5S 3G9, Canada
- Donnelly Centre, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON, M5S 3E5, Canada
| | - Michael S Jones
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, M5S 3G9, Canada
- Donnelly Centre, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
| | - Sami G Sabbah
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, M5S 3G9, Canada
- Donnelly Centre, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
| | - Mitchell J Elliott
- Princess Margaret Cancer Centre, University Health Network, 610 University Avenue, Toronto, ON, M5G 2C1, Canada
| | - Brian K Shoichet
- Department of Pharmaceutical Chemistry, University of California San Francisco, 1700 Fourth Street, Mail Box 2550, San Francisco, CA, 94143, USA
| | - David W Cescon
- Princess Margaret Cancer Centre, University Health Network, 610 University Avenue, Toronto, ON, M5G 2C1, Canada
| | - Molly S Shoichet
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, M5S 3G9, Canada
- Donnelly Centre, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON, M5S 3E5, Canada
| |
Collapse
|
2
|
Nesabi A, Kalayan J, Al-Rawashdeh S, Ghattas MA, Bryce RA. Molecular dynamics simulations as a guide for modulating small molecule aggregation. J Comput Aided Mol Des 2024; 38:11. [PMID: 38470532 PMCID: PMC10933209 DOI: 10.1007/s10822-024-00557-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024]
Abstract
Small colloidally aggregating molecules (SCAMs) can be problematic for biological assays in drug discovery campaigns. However, the self-associating properties of SCAMs have potential applications in drug delivery and analytical biochemistry. Consequently, the ability to predict the aggregation propensity of a small organic molecule is of considerable interest. Chemoinformatics-based filters such as ChemAGG and Aggregator Advisor offer rapid assessment but are limited by the assay quality and structural diversity of their training set data. Complementary to these tools, we explore here the ability of molecular dynamics (MD) simulations as a physics-based method capable of predicting the aggregation propensity of diverse chemical structures. For a set of 32 molecules, using simulations of 100 ns in explicit solvent, we find a success rate of 97% (one molecule misclassified) as opposed to 75% by Aggregator Advisor and 72% by ChemAGG. These short timescale MD simulations are representative of longer microsecond trajectories and yield an informative spectrum of aggregation propensities across the set of solutes, capturing the dynamic behaviour of weakly aggregating compounds. Implicit solvent simulations using the generalized Born model were less successful in predicting aggregation propensity. MD simulations were also performed to explore structure-aggregation relationships for selected molecules, identifying chemical modifications that reversed the predicted behaviour of a given aggregator/non-aggregator compound. While lower throughput than rapid cheminformatics-based SCAM filters, MD-based prediction of aggregation has potential to be deployed on the scale of focused subsets of moderate size, and, depending on the target application, provide guidance on removing or optimizing a compound's aggregation propensity.
Collapse
Affiliation(s)
- Azam Nesabi
- Division of Pharmacy and Optometry, School of Health Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Jas Kalayan
- Daresbury Laboratory, Science and Technologies Facilities Council (STFC), Keckwick Lane, Daresbury, Warrington, WA4 4AD, UK
| | - Sara Al-Rawashdeh
- Division of Pharmacy and Optometry, School of Health Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | | | - Richard A Bryce
- Division of Pharmacy and Optometry, School of Health Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Oxford Road, Manchester, M13 9PL, UK.
| |
Collapse
|
3
|
Dasram MH, Naidoo P, Walker RB, Khamanga SM. Targeting the Endocannabinoid System Present in the Glioblastoma Tumour Microenvironment as a Potential Anti-Cancer Strategy. Int J Mol Sci 2024; 25:1371. [PMID: 38338649 PMCID: PMC10855826 DOI: 10.3390/ijms25031371] [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: 09/16/2023] [Revised: 01/08/2024] [Accepted: 01/17/2024] [Indexed: 02/12/2024] Open
Abstract
The highly aggressive and invasive glioblastoma (GBM) tumour is the most malignant lesion among adult-type diffuse gliomas, representing the most common primary brain tumour in the neuro-oncology practice of adults. With a poor overall prognosis and strong resistance to treatment, this nervous system tumour requires new innovative treatment. GBM is a polymorphic tumour consisting of an array of stromal cells and various malignant cells contributing to tumour initiation, progression, and treatment response. Cannabinoids possess anti-cancer potencies against glioma cell lines and in animal models. To improve existing treatment, cannabinoids as functionalised ligands on nanocarriers were investigated as potential anti-cancer agents. The GBM tumour microenvironment is a multifaceted system consisting of resident or recruited immune cells, extracellular matrix components, tissue-resident cells, and soluble factors. The immune microenvironment accounts for a substantial volume of GBM tumours. The barriers to the treatment of glioblastoma with cannabinoids, such as crossing the blood-brain barrier and psychoactive and off-target side effects, can be alleviated with the use of nanocarrier drug delivery systems and functionalised ligands for improved specificity and targeting of pharmacological receptors and anti-cancer signalling pathways. This review has shown the presence of endocannabinoid receptors in the tumour microenvironment, which can be used as a potential unique target for specific drug delivery. Existing cannabinoid agents, studied previously, show anti-cancer potencies via signalling pathways associated with the hallmarks of cancer. The results of the review can be used to provide guidance in the design of future drug therapy for glioblastoma tumours.
Collapse
Affiliation(s)
| | | | | | - Sandile M. Khamanga
- Division of Pharmaceutics, Faculty of Pharmacy, Rhodes University, Makhanda 6139, South Africa (R.B.W.)
| |
Collapse
|
4
|
Shireen Z, Curk T, Brandl C, B Babu S. Rigidity-Induced Controlled Aggregation of Binary Colloids. ACS OMEGA 2023; 8:37225-37232. [PMID: 37841185 PMCID: PMC10568703 DOI: 10.1021/acsomega.3c04909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023]
Abstract
Here, we report the proof-of-concept for controlled aggregation in a binary colloidal system. The binary systems are studied by varying bond flexibility of only one species, while the other species' bonds remain fully flexible. By establishing the underlying relation between gelation and bond rigidity, we demonstrate how the interplay among bond flexibility, critical concentration, and packing volume fraction influenced the aggregation kinetics. Our result shows that rigidity in bonds increases the critical concentration for gels to be formed in the binary mixture. Furthermore, the average number of bonded neighbor analyses reveal the influence of bond rigidity both above and below critical concentrations and show that variation in bond flexibility in only one species alters the kinetics of aggregation of both species. This finding improves our understanding of colloidal aggregation in soft and biological systems.
Collapse
Affiliation(s)
- Zakiya Shireen
- Department
of Mechanical Engineering, Faculty of Engineering and Information
Technology, University of Melbourne, 3010 Parkville, Victoria Australia
| | - Tine Curk
- Department
of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Christian Brandl
- Department
of Mechanical Engineering, Faculty of Engineering and Information
Technology, University of Melbourne, 3010 Parkville, Victoria Australia
| | - Sujin B Babu
- Out
of Equilibrium Group, Department of Physics, Indian Institute of Technology Delhi, 110016 New Delhi, India
| |
Collapse
|
5
|
Donders EN, Slaughter KV, Dank C, Ganesh AN, Shoichet BK, Lautens M, Shoichet MS. Synthetic Ionizable Colloidal Drug Aggregates Enable Endosomal Disruption. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300311. [PMID: 36905240 PMCID: PMC10161099 DOI: 10.1002/advs.202300311] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Indexed: 05/06/2023]
Abstract
Colloidal drug aggregates enable the design of drug-rich nanoparticles; however, the efficacy of stabilized colloidal drug aggregates is limited by entrapment in the endo-lysosomal pathway. Although ionizable drugs are used to elicit lysosomal escape, this approach is hindered by toxicity associated with phospholipidosis. It is hypothesized that tuning the pKa of the drug would enable endosomal disruption while avoiding phospholipidosis and minimizing toxicity. To test this idea, 12 analogs of the nonionizable colloidal drug fulvestrant are synthesized with ionizable groups to enable pH-dependent endosomal disruption while maintaining bioactivity. Lipid-stabilized fulvestrant analog colloids are endocytosed by cancer cells, and the pKa of these ionizable colloids influenced the mechanism of endosomal and lysosomal disruption. Four fulvestrant analogs-those with pKa values between 5.1 and 5.7-disrupted endo-lysosomes without measurable phospholipidosis. Thus, by manipulating the pKa of colloid-forming drugs, a tunable and generalizable strategy for endosomal disruption is established.
Collapse
Affiliation(s)
- Eric N. Donders
- Department of Chemical Engineering & Applied ChemistryUniversity of Toronto200 College StreetTorontoONM5S 3E5Canada
- Institute of Biomedical EngineeringUniversity of Toronto164 College StreetTorontoONM5S 3G9Canada
- Donnelly CentreUniversity of Toronto160 College StreetTorontoONM5S3E1Canada
| | - Kai V. Slaughter
- Institute of Biomedical EngineeringUniversity of Toronto164 College StreetTorontoONM5S 3G9Canada
- Donnelly CentreUniversity of Toronto160 College StreetTorontoONM5S3E1Canada
| | - Christian Dank
- Department of ChemistryUniversity of Toronto80 St. George StreetTorontoONM5S 3H6Canada
| | - Ahil N. Ganesh
- Department of Chemical Engineering & Applied ChemistryUniversity of Toronto200 College StreetTorontoONM5S 3E5Canada
- Institute of Biomedical EngineeringUniversity of Toronto164 College StreetTorontoONM5S 3G9Canada
- Donnelly CentreUniversity of Toronto160 College StreetTorontoONM5S3E1Canada
| | - Brian K. Shoichet
- Department of Pharmaceutical ChemistryUniversity of California San Francisco1700 Fourth Street, Mail Box 2550San FranciscoCA94143USA
| | - Mark Lautens
- Department of ChemistryUniversity of Toronto80 St. George StreetTorontoONM5S 3H6Canada
| | - Molly S. Shoichet
- Department of Chemical Engineering & Applied ChemistryUniversity of Toronto200 College StreetTorontoONM5S 3E5Canada
- Institute of Biomedical EngineeringUniversity of Toronto164 College StreetTorontoONM5S 3G9Canada
- Donnelly CentreUniversity of Toronto160 College StreetTorontoONM5S3E1Canada
| |
Collapse
|
6
|
Sheridan R, Spelman K. Polyphenolic promiscuity, inflammation-coupled selectivity: Whether PAINs filters mask an antiviral asset. Front Pharmacol 2022; 13:909945. [PMID: 36339544 PMCID: PMC9634583 DOI: 10.3389/fphar.2022.909945] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 10/03/2022] [Indexed: 11/26/2023] Open
Abstract
The Covid-19 pandemic has elicited much laboratory and clinical research attention on vaccines, mAbs, and certain small-molecule antivirals against SARS-CoV-2 infection. By contrast, there has been comparatively little attention on plant-derived compounds, especially those that are understood to be safely ingested at common doses and are frequently consumed in the diet in herbs, spices, fruits and vegetables. Examining plant secondary metabolites, we review recent elucidations into the pharmacological activity of flavonoids and other polyphenolic compounds and also survey their putative frequent-hitter behavior. Polyphenols, like many drugs, are glucuronidated post-ingestion. In an inflammatory milieu such as infection, a reversion back to the active aglycone by the release of β-glucuronidase from neutrophils and macrophages allows cellular entry of the aglycone. In the context of viral infection, virions and intracellular virus particles may be exposed to promiscuous binding by the polyphenol aglycones resulting in viral inhibition. As the mechanism's scope would apply to the diverse range of virus species that elicit inflammation in infected hosts, we highlight pre-clinical studies of polyphenol aglycones, such as luteolin, isoginkgetin, quercetin, quercetagetin, baicalein, curcumin, fisetin and hesperetin that reduce virion replication spanning multiple distinct virus genera. It is hoped that greater awareness of the potential spatial selectivity of polyphenolic activation to sites of pathogenic infection will spur renewed research and clinical attention for natural products antiviral assaying and trialing over a wide array of infectious viral diseases.
Collapse
Affiliation(s)
| | - Kevin Spelman
- Massachusetts College of Pharmacy and Health Sciences, Boston, MA, United States
- Health Education and Research, Driggs, ID, United States
| |
Collapse
|
7
|
Taheri-Ledari R, Asl FR, Saeidirad M, Kashtiaray A, Maleki A. Convenient synthesis of dipeptide structures in solution phase assisted by a thioaza functionalized magnetic nanocatalyst. Sci Rep 2022; 12:4719. [PMID: 35304475 PMCID: PMC8933478 DOI: 10.1038/s41598-022-07303-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 02/16/2022] [Indexed: 12/14/2022] Open
Abstract
In this study, a heterogeneous nanocatalyst is presented that is capable to efficiently catalyze the synthetic reactions of amide bond formation between the amino acids. This nanocatalyst which is named Fe3O4@SiO2/TABHA (TABHA stands for thio-aza-bicyclo-hepten amine), was composed of several layers that increased the surface area to be functionalized with 2-aminothiazole rings via Diels-Alder approach. Firstly, various analytic methods such as Fourier-transform infrared (FTIR) and energy-dispersive X-ray (EDX) spectroscopic methods, thermogravimetric analysis (TGA), electron microscopy (EM), and UV-vis diffuse reflectance spectroscopy (UV-DRS) have been used to characterize the desired structure of the Fe3O4@SiO2/TABHA catalyst. Afterward, the application of the presented catalytic system has been studied in the peptide bond formation reactions. Due to the existence of a magnetic core in the structure of the nanocatalyst, the nanoparticles (NPs) could be easily separated from the reaction medium by an external magnet. This special feature has been corroborated by the obtained results from vibrating-sample magnetometer (VSM) analysis that showed 24 emu g-1 magnetic saturation for the catalytic system. Amazingly, a small amount of Fe3O4@SiO2/TABHA particles (0.2 g) has resulted in ca. 90% efficiency in catalyzing the peptide bond formation at ambient temperature, over 4 h. Also, this nanocatalyst has demonstrated an acceptable recycling ability, where ca. 76% catalytic performance has been observed after four recycles. Due to high convenience in the preparation, application, and recyclization processes, and also because of lower cost than the traditional coupling reagents (like TBTU), the presented catalytic system is recommended for the industrial utilization.
Collapse
Affiliation(s)
- Reza Taheri-Ledari
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran
| | - Fereshteh Rasouli Asl
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran
| | - Mahdi Saeidirad
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran
| | - Amir Kashtiaray
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran
| | - Ali Maleki
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran.
| |
Collapse
|
8
|
Farshbaf M, Valizadeh H, Panahi Y, Fatahi Y, Chen M, Zarebkohan A, Gao H. The impact of protein corona on the biological behavior of targeting nanomedicines. Int J Pharm 2022; 614:121458. [PMID: 35017025 DOI: 10.1016/j.ijpharm.2022.121458] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/27/2021] [Accepted: 01/05/2022] [Indexed: 12/17/2022]
Abstract
For successful translation of targeting nanomedicines from bench to bedside, it is vital to address their most common drawbacks namely rapid clearance and off-target accumulation. These complications evidently originate from a phenomenon called "protein corona (PC) formation" around the surface of targeting nanoparticles (NPs) which happens once they encounter the bloodstream and interact with plasma proteins with high collision frequency. This phenomenon endows the targeting nanomedicines with a different biological behavior followed by an unexpected fate, which is usually very different from what we commonly observe in vitro. In addition to the inherent physiochemical properties of NPs, the targeting ligands could also remarkably dictate the amount and type of adsorbed PC. As very limited studies have focused their attention on this particular factor, the present review is tasked to discuss the best simulated environment and latest characterization techniques applied to PC analysis. The effect of PC on the biological behavior of targeting NPs engineered with different targeting moieties is further discussed. Ultimately, the recent progresses in manipulation of nano-bio interfaces to achieve the most favorite therapeutic outcome are highlighted.
Collapse
Affiliation(s)
- Masoud Farshbaf
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Valizadeh
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yunes Panahi
- Pharmacotherapy Department, Faculty of Pharmacy, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Yousef Fatahi
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Amir Zarebkohan
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Sichuan 610041, China.
| |
Collapse
|
9
|
LaPlante SR, Roux V, Shahout F, LaPlante G, Woo S, Denk MM, Larda ST, Ayotte Y. Probing the free-state solution behavior of drugs and their tendencies to self-aggregate into nano-entities. Nat Protoc 2021; 16:5250-5273. [PMID: 34707256 DOI: 10.1038/s41596-021-00612-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 08/09/2021] [Indexed: 02/08/2023]
Abstract
The free-state solution behaviors of drugs profoundly affect their properties. Therefore, it is critical to properly evaluate a drug's unique multiphase equilibrium when in an aqueous enviroment, which can comprise lone molecules, self-associating aggregate states and solid phases. To date, the full range of nano-entities that drugs can adopt has been a largely unexplored phenomenon. This protocol describes how to monitor the solution behavior of drugs, revealing the nano-entities formed as a result of self-associations. The procedure begins with a simple NMR 1H assay, and depending on the observations, subsequent NMR dilution, NMR T2-CPMG (spin-spin relaxation Carr-Purcell-Meiboom-Gill) and NMR detergent assays are used to distinguish between the existence of fast-tumbling lone drug molecules, small drug aggregates and slow-tumbling colloids. Three orthogonal techniques (dynamic light scattering, transmission electron microscopy and confocal laser scanning microscopy) are also described that can be used to further characterize any large colloids. The protocol can take a non-specialist between minutes to a few hours; thus, libraries of compounds can be evaluated within days.
Collapse
Affiliation(s)
- Steven R LaPlante
- Université du Québec, INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Quebec, Canada.
- NMX Research and Solutions, Inc., Laval, Quebec, Canada.
| | - Valérie Roux
- Université du Québec, INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Quebec, Canada
| | - Fatma Shahout
- Université du Québec, INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Quebec, Canada
| | | | - Simon Woo
- Université du Québec, INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Quebec, Canada
| | - Maria M Denk
- NMX Research and Solutions, Inc., Laval, Quebec, Canada
| | - Sacha T Larda
- NMX Research and Solutions, Inc., Laval, Quebec, Canada
| | - Yann Ayotte
- Université du Québec, INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Quebec, Canada
| |
Collapse
|
10
|
Fan M, Li J. A Novel Combinational Nanodrug Delivery System Induces Synergistic Inhibition of Lung Adenocarcinoma Cells In vitro. LETT DRUG DES DISCOV 2021. [DOI: 10.2174/1570180817999200719152426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
The combination of two or more therapeutic drugs is an attractive approach
to improve the treatment of experimental tumors. Leveraging nanocarriers for combinational drug
delivery can allow control over drug biological fate and promote co-localization in the same area of
the body. However, there are certain concerns regarding the biodegradability and potential longterm
toxicity arising from these synthetic nanoscale carriers.
Objective:
Our aim was to develop a combinational nanodrug delivery system formed by selfassembling
of amphiphilic drug molecules.minimizing potential toxicities associated with using
additional synthetic nanocarriers.
Methods:
A novel prodrug chlorambucil gemcitabine conjugate was synthesized, this prodrug was
used for the encapsulation of an additional hydrophobic anticancer drug paclitaxel, taking the form
of combinational nanodrugs. Particle size and zeta potential were evaluated, cytotoxicity assay and
apoptosis/cell cycle analysis were also performed to validate the anticancer efficacy of the combinational
nanodrugs.
Results:
The combinational nanodrugs were acquired by means of nanoprecipitation. In A549 lung
adenocarcinoma cell line, cellular assays revealed that co-delivery of low dosage paclitaxel with
chlorambucil gemcitabine conjugate can act synergistically to inhibit cell growth and induce accumulation
of cells in the G2/M phase with a concomitant decrease in G0/G1 compartment.
Conclusion:
Chlorambucil gemcitabine conjugate and paclitaxel can co-assemble into composite
nanoparticles by a nanoprecipitation process and the resulting combinational nanodrugs showed a
synergistic anticancer effect. This synthetic nanocarrier-free approach might broaden the nanodrug
concept and have potential in cancer therapy.
Collapse
Affiliation(s)
- Mingliang Fan
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Jiping Li
- Department of Otolaryngology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200032, China
| |
Collapse
|
11
|
Daso RE, Banerjee IA. Self-Assembled Peptide-Based Biocomposites for Near-Infrared Light Triggered Drug Release to Tumor Cells. Biotechnol J 2020; 15:e2000128. [PMID: 32845561 DOI: 10.1002/biot.202000128] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/21/2020] [Indexed: 11/11/2022]
Abstract
Peptide-based nanomaterials are increasingly gaining popularity due to their specificity, biocompatibility, and biodegradability. In this work, a new multi-layered peptide-based biocomposite for targeting MCF-7 breast cancer cells is developed. The amphipathic Fluorenylmethyloxycarbonyl (Fmoc)-Leu-Ser peptide is synthesized, which is conjugated to a tumor-targeting peptide sequence Gly-Cys-Gly-Asn-Ser to form Fmoc-L-S-G-C-G-N-S (FLS) assemblies. To the FLS assemblies, gold nanorods are then attached to develop drug delivery vehicles (DDVs). The DDVs are entrapped with the anti-cancer drug fulvestrant. Entrapment efficiency is found to be 50.6%. Release studies indicate that irradiating the gold nanorod bound DDVs at NIR wavelength (785 nm) increases drug release by fourfold compared to assemblies that are not irradiated. These results also show higher cytotoxicity and lower cell invasion due to photo-triggered drug release. Furthermore, distinct actin cytoskeletal changes are observed. Such novel peptide-based gold nanorod bound DDVs demonstrate potential in dual targeting of MCF-7 breast cancer cells.
Collapse
Affiliation(s)
- Rachel E Daso
- Department of Chemistry, Fordham University, 441 E. Fordham Road, Bronx, NY, 10458, USA
| | - Ipsita A Banerjee
- Department of Chemistry, Fordham University, 441 E. Fordham Road, Bronx, NY, 10458, USA
| |
Collapse
|
12
|
Ghattas MA, Al Rawashdeh S, Atatreh N, Bryce RA. How Do Small Molecule Aggregates Inhibit Enzyme Activity? A Molecular Dynamics Study. J Chem Inf Model 2020; 60:3901-3909. [PMID: 32628846 DOI: 10.1021/acs.jcim.0c00540] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Small molecule compounds which form colloidal aggregates in solution are problematic in early drug discovery; adsorption of the target protein by these aggregates can lead to false positives in inhibition assays. In this work, we probe the molecular basis of this inhibitory mechanism using molecular dynamics simulations. Specifically, we examine in aqueous solution the adsorption of the enzymes β-lactamase and PTP1B onto aggregates of the drug miconazole. In accordance with experiment, molecular dynamics simulations observe formation of miconazole aggregates as well as subsequent association of these aggregates with β-lactamase and PTP1B. When complexed with aggregate, the proteins do not exhibit significant alteration in protein tertiary structure or dynamics on the microsecond time scale of the simulations, but they do indicate persistent occlusion of the protein active site by miconazole molecules. MD simulations further suggest this occlusion can occur via surficial interactions of protein with miconazole but also potentially by envelopment of the protein by miconazole. The heterogeneous polarity of the miconazole aggregate surface seems to underpin its activity as an invasive and nonspecific inhibitory agent. A deeper understanding of these protein/aggregate systems has implications not only for drug design but also for their exploitation as tools in drug delivery and analytical biochemistry.
Collapse
Affiliation(s)
| | - Sara Al Rawashdeh
- Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Oxford Road, Manchester M13 9PL, United Kingdom
| | - Noor Atatreh
- College of Pharmacy, Al Ain University, Abu Dhabi, United Arab Emirates
| | - Richard A Bryce
- Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Oxford Road, Manchester M13 9PL, United Kingdom
| |
Collapse
|
13
|
Abstract
Small-molecule aggregates are a leading cause of artifacts in early drug discovery, but little is known about their interactions with proteins, nor why some proteins are more susceptible to inhibition than others. A possible reason for this apparent selectivity is that aggregation-based inhibition, as a stoichiometric process, is sensitive to protein concentration, which varies across assays. Alternatively, local protein unfolding by aggregates may lead to selectivity since stability varies among proteins. To deconvolute these effects, we used differentially stable point mutants of a single protein, TEM-1 β-lactamase. Broadly, destabilized mutants had higher affinities for and were more potently inhibited by aggregates versus more stable variants. The addition of the irreversible inhibitor moxalactam destabilized several mutants, and these typically bound tighter to a colloidal particle, while the only mutant it stabilized bound weaker. These results suggest that less-stable enzymes are more easily sequestered and inhibited by colloidal aggregates.
Collapse
Affiliation(s)
- Hayarpi Torosyan
- Department of Pharmaceutical Chemistry , University of California, San Francisco , 1700 Fourth Street , San Francisco , California 94143-2550 , United States
| | - Brian K Shoichet
- Department of Pharmaceutical Chemistry , University of California, San Francisco , 1700 Fourth Street , San Francisco , California 94143-2550 , United States
| |
Collapse
|
14
|
Ayotte Y, Marando VM, Vaillancourt L, Bouchard P, Heffron G, Coote PW, Larda ST, LaPlante SR. Exposing Small-Molecule Nanoentities by a Nuclear Magnetic Resonance Relaxation Assay. J Med Chem 2019; 62:7885-7896. [PMID: 31422659 DOI: 10.1021/acs.jmedchem.9b00653] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Small molecules can self-assemble in aqueous solution into a wide range of nanoentity types and sizes (dimers, n-mers, micelles, colloids, etc.), each having their own unique properties. This has important consequences in the context of drug discovery including issues related to nonspecific binding, off-target effects, and false positives and negatives. Here, we demonstrate the use of the spin-spin relaxation Carr-Purcell-Meiboom-Gill NMR experiment, which is sensitive to molecular tumbling rates and can expose larger aggregate species that have slower rotational correlations. The strategy easily distinguishes lone-tumbling molecules versus nanoentities of various sizes. The technique is highly sensitive to chemical exchange between single-molecule and aggregate states and can therefore be used as a reporter when direct measurement of aggregates is not possible by NMR. Interestingly, we found differences in solution behavior for compounds within structurally related series, demonstrating structure-nanoentity relationships. This practical experiment is a valuable tool to support drug discovery efforts.
Collapse
Affiliation(s)
- Yann Ayotte
- INRS-Centre Armand-Frappier Santé Biotechnologie , 531 Boulevard des Prairies , Laval , Québec H7V 1B7 , Canada
| | - Victoria M Marando
- NMX Research and Solutions, Inc. , 500 Boulevard Cartier Ouest , Laval , Québec , H7V 5B7 , Canada
| | - Louis Vaillancourt
- NMX Research and Solutions, Inc. , 500 Boulevard Cartier Ouest , Laval , Québec , H7V 5B7 , Canada
| | - Patricia Bouchard
- NMX Research and Solutions, Inc. , 500 Boulevard Cartier Ouest , Laval , Québec , H7V 5B7 , Canada
| | - Gregory Heffron
- Harvard Medical School , 240 Longwood Avenue , Boston , Massachusetts 02115 , United States
| | - Paul W Coote
- NMX Research and Solutions, Inc. , 500 Boulevard Cartier Ouest , Laval , Québec , H7V 5B7 , Canada.,Harvard Medical School , 240 Longwood Avenue , Boston , Massachusetts 02115 , United States
| | - Sacha T Larda
- NMX Research and Solutions, Inc. , 500 Boulevard Cartier Ouest , Laval , Québec , H7V 5B7 , Canada
| | - Steven R LaPlante
- INRS-Centre Armand-Frappier Santé Biotechnologie , 531 Boulevard des Prairies , Laval , Québec H7V 1B7 , Canada.,NMX Research and Solutions, Inc. , 500 Boulevard Cartier Ouest , Laval , Québec , H7V 5B7 , Canada.,Harvard Medical School , 240 Longwood Avenue , Boston , Massachusetts 02115 , United States
| |
Collapse
|
15
|
Donders EN, Ganesh AN, Torosyan H, Lak P, Shoichet BK, Shoichet MS. Triggered Release Enhances the Cytotoxicity of Stable Colloidal Drug Aggregates. ACS Chem Biol 2019; 14:1507-1514. [PMID: 31243955 DOI: 10.1021/acschembio.9b00247] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chemotherapeutics that self-assemble into colloids have limited efficacy above their critical aggregation concentration due to their inability to penetrate intact plasma membranes. Even when colloid uptake is promoted, issues with colloid escape from the endolysosomal pathway persist. By stabilizing acid-responsive lapatinib colloids through coaggregation with fulvestrant, and inclusion of transferrin, we demonstrate colloid internalization by cancer cells, where subsequent lapatinib ionization leads to endosomal leakage and increased cytotoxicity. These results demonstrate a strategy for triggered drug release from stable colloidal aggregates.
Collapse
Affiliation(s)
- Eric N. Donders
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
| | - Ahil N. Ganesh
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
| | - Hayarpi Torosyan
- Department of Pharmaceutical Chemistry, University of California, San Francisco, 1700 Fourth Street, Mail Box 2550, San Francisco, California 94143, United States
| | - Parnian Lak
- Department of Pharmaceutical Chemistry, University of California, San Francisco, 1700 Fourth Street, Mail Box 2550, San Francisco, California 94143, United States
| | - Brian K. Shoichet
- Department of Pharmaceutical Chemistry, University of California, San Francisco, 1700 Fourth Street, Mail Box 2550, San Francisco, California 94143, United States
| | - Molly S. Shoichet
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| |
Collapse
|
16
|
Xu L, Chu Z, Wang H, Cai L, Tu Z, Liu H, Zhu C, Shi H, Pan D, Pan J, Fei X. Electrostatically Assembled Multilayered Films of Biopolymer Enhanced Nanocapsules for on-Demand Drug Release. ACS APPLIED BIO MATERIALS 2019; 2:3429-3438. [DOI: 10.1021/acsabm.9b00381] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Li Xu
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Zihan Chu
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Hailong Wang
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Lawrence Cai
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Zhigang Tu
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Hanqing Liu
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Chunyin Zhu
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Haifeng Shi
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Donghui Pan
- Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
| | - Jia Pan
- Novo Nordisk Research Center−Indianapolis, Inc., Indianapolis, Indiana 46241, United States
| | - Xiang Fei
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| |
Collapse
|
17
|
Maleki A, Taheri-Ledari R, Rahimi J, Soroushnejad M, Hajizadeh Z. Facile Peptide Bond Formation: Effective Interplay between Isothiazolone Rings and Silanol Groups at Silver/Iron Oxide Nanocomposite Surfaces. ACS OMEGA 2019; 4:10629-10639. [PMID: 31460161 PMCID: PMC6649058 DOI: 10.1021/acsomega.9b00986] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 05/17/2019] [Indexed: 05/08/2023]
Abstract
Proportional to considerable progress in protein-drug conjugations, attention to the efficient peptide coupling reagents is being increased. Hence, in this study, a versatile heterogeneous nanoscale reagent is presented for chemical, biological, and medical purposes. A combination of silver and silica-coated iron oxide nanoparticles (Ag/Fe3O4) has been well functionalized with isothiazolone rings via a silver-modified Heck mechanism. An appropriate condition is provided for peptide bond formation through the surface interplay between silanol groups and the loaded isothiazolone rings. A logical mechanism including a series of successive covalent bonds onto the surface of Ag/Fe3O4 nanocomposites is suggested for this catalyzed peptide bond formation. Accurate comparisons have been made to obtain the optimum value of the nanocatalyst and suitable conditions. As an additional application, the biological activity of the desired product has also been investigated through antibacterial assay tests. The results showed that our desired product could also be used as an effective heterogeneous nanoscale antibacterial agent for different purposes. In this regard, all of the essential structural and practical analyses have been carried out and precisely interpreted.
Collapse
Affiliation(s)
- Ali Maleki
- E-mail: . Tel: +98 21 73228313. Fax: +98 21 73021584
| | | | | | | | | |
Collapse
|
18
|
Ganesh AN, Aman A, Logie J, Barthel BL, Cogan P, Al-awar R, Koch TH, Shoichet BK, Shoichet MS. Colloidal Drug Aggregate Stability in High Serum Conditions and Pharmacokinetic Consequence. ACS Chem Biol 2019; 14:751-757. [PMID: 30840432 PMCID: PMC6474797 DOI: 10.1021/acschembio.9b00032] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Colloidal drug aggregates have been a nuisance in drug screening, yet, because they inherently comprise drug-rich particles, they may be useful in vivo if issues of stability can be addressed. As the first step toward answering this question, we optimized colloidal drug aggregate formulations using a fluorescence-based assay to study fulvestrant colloidal formation and stability in high (90%) serum conditions in vitro. We show, for the first time, that the critical aggregation concentration of fulvestrant depends on media composition and increases with serum concentration. Excipients, such as polysorbate 80, stabilize fulvestrant colloids in 90% serum in vitro for over 48 h. Using fulvestrant and an investigational pro-drug, pentyloxycarbonyl-( p-aminobenzyl) doxazolidinylcarbamate (PPD), as proof-of-concept colloidal formulations, we demonstrate that the in vivo plasma half-life for stabilized colloids is greater than their respective monomeric forms. These studies demonstrate the potential of turning the nuisance of colloidal drug aggregation into an opportunity for drug-rich formulations.
Collapse
Affiliation(s)
- Ahil N. Ganesh
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
| | - Ahmed Aman
- Drug Discovery Program, Ontario Institute for Cancer Research, 661 University Avenue, Suite 510, Toronto, Ontario M5G 0A3, Canada
| | - Jennifer Logie
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
| | - Ben L. Barthel
- Drug Discovery Program, Ontario Institute for Cancer Research, 661 University Avenue, Suite 510, Toronto, Ontario M5G 0A3, Canada
| | - Peter Cogan
- School of Pharmacy, Regis University, 3333 Regis Boulevard, Denver, Colorado 80221-1099, United States
| | - Rima Al-awar
- Department of Chemistry and Biochemistry, University of Colorado, Boulder, Colorado 80309-0215, United States
| | - Tad H. Koch
- Drug Discovery Program, Ontario Institute for Cancer Research, 661 University Avenue, Suite 510, Toronto, Ontario M5G 0A3, Canada
| | - Brian K. Shoichet
- Department of Pharmaceutical Chemistry and Quantitative Biology Institute, University of California, San Francisco, 1700 Fourth Street, Mail Box 2550, San Francisco, California 94143, United States
| | - Molly S. Shoichet
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| |
Collapse
|
19
|
Reker D, Bernardes GJL, Rodrigues T. Computational advances in combating colloidal aggregation in drug discovery. Nat Chem 2019; 11:402-418. [PMID: 30988417 DOI: 10.1038/s41557-019-0234-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 02/21/2019] [Indexed: 02/07/2023]
Abstract
Small molecule effectors are essential for drug discovery. Specific molecular recognition, reversible binding and dose-dependency are usually key requirements to ensure utility of a novel chemical entity. However, artefactual frequent-hitter and assay interference compounds may divert lead optimization and screening programmes towards attrition-prone chemical matter. Colloidal aggregates are the prime source of false positive readouts, either through protein sequestration or protein-scaffold mimicry. Nevertheless, assessment of colloidal aggregation remains somewhat overlooked and under-appreciated. In this Review, we discuss the impact of aggregation in drug discovery by analysing select examples from the literature and publicly-available datasets. We also examine and comment on technologies used to experimentally identify these potentially problematic entities. We focus on evidence-based computational filters and machine learning algorithms that may be swiftly deployed to flag chemical matter and mitigate the impact of aggregates in discovery programmes. We highlight the tools that can be used to scrutinize libraries, and identify and eliminate these problematic compounds.
Collapse
Affiliation(s)
- Daniel Reker
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA. .,MIT-IBM Watson AI Lab, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Gonçalo J L Bernardes
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, UK.,Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Tiago Rodrigues
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
20
|
Ganesh AN, Donders EN, Shoichet BK, Shoichet MS. Colloidal aggregation: from screening nuisance to formulation nuance. NANO TODAY 2018; 19:188-200. [PMID: 30250495 PMCID: PMC6150470 DOI: 10.1016/j.nantod.2018.02.011] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
It is well known that small molecule colloidal aggregation is a leading cause of false positives in early drug discovery. Colloid-formers are diverse and well represented among corporate and academic screening decks, and even among approved drugs. Less appreciated is how colloid formation by drug-like compounds fits into the wider understanding of colloid physical chemistry. Here we introduce the impact that colloidal aggregation has had on early drug discovery, and then turn to the physical and thermodynamic driving forces for small molecule colloidal aggregation, including the particulate nature of the colloids, their critical aggregation concentration-governed formation, their mechanism of protein adsorption and subsequent inhibition, and their sensitivity to detergent. We describe methods that have been used extensively to both identify aggregate-formers and to study and control their physical chemistry. While colloidal aggregation is widely recognized as a problem in early drug discovery, we highlight the opportunities for exploiting this phenomenon in biological milieus and for drug formulation.
Collapse
Affiliation(s)
- Ahil N. Ganesh
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, ON,Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, ON, Canada
| | - Eric N. Donders
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, ON,Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, ON, Canada
| | - Brian K. Shoichet
- Department of Pharmaceutical Chemistry, University of California – San Francisco, CA, USA
| | - Molly S. Shoichet
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, ON,Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, ON, Canada
- Department of Chemistry, University of Toronto, ON, Canada
- To whom correspondence should be addressed: Molly S. Shoichet, University of Toronto, 160 College Street, Room 514, Toronto, ON, Canada M5S 3E1,
| |
Collapse
|
21
|
Drug self-assembly for synthesis of highly-loaded antimicrobial drug-silica particles. Sci Rep 2018; 8:895. [PMID: 29343729 PMCID: PMC5772632 DOI: 10.1038/s41598-018-19166-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 12/17/2017] [Indexed: 12/16/2022] Open
Abstract
Antimicrobial drug release from biomaterials for orthopedic repair and dental restorations can prevent biofilm growth and caries formation. Carriers for drug incorporation would benefit from long-term drug storage, controlled release, and structural stability. Mesoporous silica, synthesized through a co-assembly of silica and surfactant template, is an ideal drug encapsulation scaffold that maintains structural integrity upon release. However, conventional loading of drug within meso-silica pores via concentration-gradient diffusion limits the overall payload, concentration uniformity, and drug release control. Herein we demonstrate the co-assembly of an antimicrobial drug (octenidine dihydrochloride, OCT), and silica, to form highly-loaded (35% wt.) OCT-silica nanocomposite spheres of 500 nm diameter. Drug release significantly outlasted conventional OCT-loaded mesoporous silica, closely fit Higuchi models of diffusive release, and was visualized via electron microscopy. Extension of this concept to the broad collection of self-assembling drugs grants biomedical community a powerful tool for synthesizing drug-loaded inorganic nanomaterials from the bottom-up.
Collapse
|
22
|
Ghattas MA, Bryce RA, Al Rawashdah S, Atatreh N, Zalloum WA. Comparative Molecular Dynamics Simulation of Aggregating and Non-Aggregating Inhibitor Solutions: Understanding the Molecular Basis of Promiscuity. ChemMedChem 2017; 13:500-506. [PMID: 29058775 DOI: 10.1002/cmdc.201700654] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Indexed: 11/08/2022]
Abstract
The presence of false positives in enzyme inhibition assays is a common problem in early drug discovery, especially for compounds that form colloid aggregates in solution. The molecular basis of these aggregates could not be thoroughly explored because of their transient stability. In this study we conducted comparative molecular dynamics (MD) simulations of miconazole, a strong aggregator, and fluconazole, a known non-aggregator. Interestingly, miconazole displays full aggregation within only 50 ns, whilst fluconazole shows no aggregation over the 500 ns simulation. The simulations indicate that the center of the aggregate is densely packed by the hydrophobic groups of miconazole, whereas polar and nonpolar groups comprise the surface to form a micelle-like colloid. The amphiphilic moment and planar nature of the miconazole structure appear to promote its aggregating behavior. The simulations also predict rapid aggregate formation for a second known promiscuous inhibitor, nicardipine. Thus, MD appears to be a useful tool to characterize aggregate-prone inhibitors at molecular-level detail and has the potential to provide useful information for drug discovery and formulation design.
Collapse
Affiliation(s)
- Mohammad A Ghattas
- College of Pharmacy, Al Ain University of Science and Technology, Al Ain, 64141, UAE
| | - Richard A Bryce
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Oxford Road, Manchester, M13 9PL, UK
| | - Sara Al Rawashdah
- College of Pharmacy, Al Ain University of Science and Technology, Al Ain, 64141, UAE
| | - Noor Atatreh
- College of Pharmacy, Al Ain University of Science and Technology, Al Ain, 64141, UAE
| | - Waleed A Zalloum
- Faculty of Health Sciences, American University of Madaba, Amman, Jordan
| |
Collapse
|