1
|
Hefnawy A, Abdelhamid AS, Abdelaziz MM, Elzoghby AO, Khalil IA. Recent advances in nano-based drug delivery systems for treatment of liver cancer. J Pharm Sci 2024; 113:3145-3172. [PMID: 39151795 DOI: 10.1016/j.xphs.2024.08.012] [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: 04/01/2024] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Liver cancer is one of the aggressive primary tumors as evident by high rate of incidence and mortality. Conventional treatments (e.g. chemotherapy) suffer from various drawbacks including wide drug distribution, low localized drug concentration, and severe off-site toxicity. Therefore, they cannot satisfy the mounting need for safe and efficient cancer therapeutics, and alternative novel strategies are needed. Nano-based drug delivery systems (NDDSs) are among these novel approaches that can improve the overall therapeutic outcomes. NDDSs are designed to encapsulate drug molecules and target them specifically to liver cancer. Thus, NDDSs can selectively deliver therapeutic agents to the tumor cells and avoid distribution to off-target sites which should improve the safety profile of the active agents. Nonetheless, NDDSs should be well designed, in terms of the preparing materials, nanocarriers structure, and the targeting strategy, in order to accomplish these objectives. This review discusses the latest advances of NDDSs for cancer therapy with emphasis on the aforementioned essential design components. The review also entails the challenges associated with the clinical translation of NDDSs, and the future perspectives towards next-generation NDDSs.
Collapse
Affiliation(s)
- Amr Hefnawy
- Smyth Lab, College of Pharmacy, University of Texas at Austin, TX 78712, USA.
| | - Ahmed S Abdelhamid
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt.
| | - Moustafa M Abdelaziz
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS 66047, USA.
| | - Ahmed O Elzoghby
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt; Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt; Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Islam A Khalil
- Department of Pharmaceutics, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City 12582, Giza, Egypt.
| |
Collapse
|
2
|
Nasaj M, Chehelgerdi M, Asghari B, Ahmadieh-Yazdi A, Asgari M, Kabiri-Samani S, Sharifi E, Arabestani M. Factors influencing the antimicrobial mechanism of chitosan action and its derivatives: A review. Int J Biol Macromol 2024; 277:134321. [PMID: 39084423 DOI: 10.1016/j.ijbiomac.2024.134321] [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: 12/02/2023] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 08/02/2024]
Abstract
Chitosan, a versatile amino polysaccharide biopolymer derived from chitin, exhibits broad-spectrum antimicrobial activity against various pathogenic microorganisms, including gram-negative and gram-positive bacteria, as well as fungi. Due to its ubiquitous use in medications, food, cosmetics, chemicals, and crops, it is an effective antibacterial agent. However, the antimicrobial performance of chitosan is influenced by multiple factors, which have been extensively investigated and reported in the literature. The goal of this review paper is to present a thorough grasp of the mechanisms of action and determining variables of chitosan and its derivatives' antibacterial activity. The article begins by providing a brief background on chitosan and its antimicrobial properties, followed by the importance of understanding the mechanism of action and factors influencing its activity".
Collapse
Affiliation(s)
- Mona Nasaj
- Department of Microbiology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, IR, Iran
| | - Mohammad Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran; Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Babak Asghari
- Department of Microbiology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, IR, Iran
| | - Amirhossein Ahmadieh-Yazdi
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Masoumeh Asgari
- Department of Nutritional Science, School of Medicine, Hamadan university of Medical Science, Hamadan, Iran
| | - Saber Kabiri-Samani
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Esmaeel Sharifi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran; Cancer Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Mohammadreza Arabestani
- Department of Microbiology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, IR, Iran; Infectious Disease Research Centre, Hamadan University of Medical Sciences, Hamadan, IR, Iran.
| |
Collapse
|
3
|
Ijaz M, Hasan I, Chaudhry TH, Huang R, Zhang L, Hu Z, Tan Q, Guo B. Bacterial derivatives mediated drug delivery in cancer therapy: a new generation strategy. J Nanobiotechnology 2024; 22:510. [PMID: 39182109 PMCID: PMC11344338 DOI: 10.1186/s12951-024-02786-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024] Open
Abstract
Cancer is measured as a major threat to human life and is a leading cause of death. Millions of cancer patients die every year, although a burgeoning number of researchers have been making tremendous efforts to develop cancer medicine to fight against cancer. Owing to the complexity and heterogeneity of cancer, lack of ability to treat deep tumor tissues, and high toxicity to the normal cells, it complicates the therapy of cancer. However, bacterial derivative-mediated drug delivery has raised the interest of researchers in overcoming the restrictions of conventional cancer chemotherapy. In this review, we show various examples of tumor-targeting bacteria and bacterial derivatives for the delivery of anticancer drugs. This review also describes the advantages and limitations of delivering anticancer treatment drugs under regulated conditions employing these tumor-targeting bacteria and their membrane vesicles. This study highlights the substantial potential for clinical translation of bacterial-based drug carriers, improve their ability to work with other treatment modalities, and provide a more powerful, dependable, and distinctive tumor therapy.
Collapse
Affiliation(s)
- Muhammad Ijaz
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Ikram Hasan
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, 518060, Guangdong, China
| | - Tamoor Hamid Chaudhry
- Antimicrobial Resistance (AMR) Containment & Infection Prevention & Control (IPC) Program, National Institute of Health, Chak Shahzad, Islamabad, Pakistan
| | - Rui Huang
- Department of Blood Transfusion, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330000, China
| | - Lan Zhang
- Department of Blood Transfusion, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330000, China
| | - Ziwei Hu
- Institute of Otolaryngology Head and Neck Surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510282, China.
| | - Qingqin Tan
- Department of Blood Transfusion, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330000, China.
| | - Bing Guo
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China.
| |
Collapse
|
4
|
Degerstedt O, O'Callaghan P, Clavero AL, Gråsjö J, Eriksson O, Sjögren E, Hansson P, Heindryckx F, Kreuger J, Lennernäs H. Quantitative imaging of doxorubicin diffusion and cellular uptake in biomimetic gels with human liver tumor cells. Drug Deliv Transl Res 2024; 14:970-983. [PMID: 37824040 DOI: 10.1007/s13346-023-01445-1] [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] [Accepted: 09/29/2023] [Indexed: 10/13/2023]
Abstract
Novel tumor-on-a-chip approaches are increasingly used to investigate tumor progression and potential treatment options. To improve the effect of any cancer treatment it is important to have an in depth understanding of drug diffusion, penetration through the tumor extracellular matrix and cellular uptake. In this study, we have developed a miniaturized chip where drug diffusion and cellular uptake in different hydrogel environments can be quantified at high resolution using live imaging. Diffusion of doxorubicin was reduced in a biomimetic hydrogel mimicking tissue properties of cirrhotic liver and early stage hepatocellular carcinoma (373 ± 108 µm2/s) as compared to an agarose gel (501 ± 77 µm2/s, p = 0.019). The diffusion was further lowered to 256 ± 30 µm2/s (p = 0.028) by preparing the biomimetic gel in cell media instead of phosphate buffered saline. The addition of liver tumor cells (Huh7 or HepG2) to the gel, at two different densities, did not significantly influence drug diffusion. Clinically relevant and quantifiable doxorubicin concentration gradients (1-20 µM) were established in the chip within one hour. Intracellular increases in doxorubicin fluorescence correlated with decreasing fluorescence of the DNA-binding stain Hoechst 33342 and based on the quantified intracellular uptake of doxorubicin an apparent cell permeability (9.00 ± 0.74 × 10-4 µm/s for HepG2) was determined. Finally, the data derived from the in vitro model were applied to a spatio-temporal tissue concentration model to evaluate the potential clinical impact of a cirrhotic extracellular matrix on doxorubicin diffusion and tumor cell uptake.
Collapse
Affiliation(s)
- Oliver Degerstedt
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Paul O'Callaghan
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Ada Lerma Clavero
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Johan Gråsjö
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Olle Eriksson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Erik Sjögren
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Per Hansson
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Femke Heindryckx
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Johan Kreuger
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Hans Lennernäs
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
5
|
Madkhali OA, Moni SS, Sultan MH, Bakkari MA, Almoshari Y, Shaheen ES, Alshammari A. Design and characterization of Lactotransferrin peptide-loaded dextran-docosahexaenoic acid nanoparticles: an immune modulator for hepatic damage. Sci Rep 2023; 13:13537. [PMID: 37598258 PMCID: PMC10439908 DOI: 10.1038/s41598-023-40674-9] [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: 04/04/2023] [Accepted: 08/16/2023] [Indexed: 08/21/2023] Open
Abstract
The primary objective of this research was to create injectable delivery formulations using Lactotransferrin (LTF) peptide-loaded dextran nanoparticles coated with docosahexaenoic acid. These nanoparticles, designated as LLDDNP, underwent a lyophilization process. The study encompassed a comprehensive investigation, including physicochemical characterization, in vivo assessment of biomarkers, and an examination of immune response through cytokine modulation. The zeta potential of LLDDNP was - 24.5 ± 12 mV, while their average particle size was 334.9 z.d.nm. The particles exhibited a conductivity of 2.10 mS/cm, while their mobility in the injectable dosage form was measured at - 3.65 µm cm/Vs. The scanning electron microscopy investigation, the lyophilization processes resulted in discrete particles forming particle aggregations. However, transmission electron microscopy analysis revealed that LLDDNP is spherical and smooth. The thermogram showed that about 95% of LLDDNP's weight was lost at 270 °C, indicating that the particles are extremely thermal stable. The XRD analysis of LLDDNP exhibited clear and distinctive peaks at 2θ angles, specifically at 9.6°, 20.3°, 21.1°, 22°, 24.6°, 25.2°, 36°, and 44.08°, providing compelling evidence of the crystalline nature of the particles. According to proton NMR studies, the proton dimension fingerprint region of LLDDNP ranges from 1.00 to 1.03 ppm. The in vitro release of LTF from LLDDNP was found to follow zero-order kinetics, with a commendable R2 value of 0.942, indicating a consistent and predictable release pattern over time. The in vivo investigation revealed a significant impact of hepatotoxicity on the elevation of various cytokines, including IL-1β, IL-6, IL-8R, TNF-α, IL-2, IL-4, IL-10, and IFN-γ. Additionally, the presence of hepatotoxicity led to an increase in apoptosis markers, namely caspase 3 and caspase 9, as well as elevated levels of liver biomarkers such as CRP, ALP, ALT, and AST. In contrast, the treatment with LLDDNP modulated the levels of all biomarkers, including cytokines level in the treatment group extremely high significant at p < 0.001.
Collapse
Affiliation(s)
- Osama A Madkhali
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Sivakumar S Moni
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia.
| | - Muhammad H Sultan
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Mohammed Ali Bakkari
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Yosif Almoshari
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | | | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
6
|
Singh N, Shi S, Goel S. Ultrasmall silica nanoparticles in translational biomedical research: Overview and outlook. Adv Drug Deliv Rev 2023; 192:114638. [PMID: 36462644 PMCID: PMC9812918 DOI: 10.1016/j.addr.2022.114638] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/06/2022] [Accepted: 11/23/2022] [Indexed: 12/05/2022]
Abstract
The exemplary progress of silica nanotechnology has attracted extensive attention across a range of biomedical applications such as diagnostics and imaging, drug delivery, and therapy of cancer and other diseases. Ultrasmall silica nanoparticles (USNs) have emerged as a particularly promising class demonstrating unique properties that are especially suitable for and have shown great promise in translational and clinical biomedical research. In this review, we discuss synthetic strategies that allow precise engineering of USNs with excellent control over size and surface chemistry, functionalization, and pharmacokinetic and toxicological profiles. We summarize the current state-of-the-art in the biomedical applications of USNs with a particular focus on select clinical studies. Finally, we illustrate long-standing challenges in the translation of inorganic nanotechnology, particularly in the context of ultrasmall nanomedicines, and provide our perspectives on potential solutions and future opportunities in accelerating the translation and widespread adoption of USN technology in biomedical research.
Collapse
Affiliation(s)
- Neetu Singh
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112
| | - Sixiang Shi
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112,Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84112,Correspondence to ;
| | - Shreya Goel
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112,Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112,Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84112,Correspondence to ;
| |
Collapse
|
7
|
Interaction between Nanoparticles, Membranes and Proteins: A Surface Plasmon Resonance Study. Int J Mol Sci 2022; 24:ijms24010591. [PMID: 36614033 PMCID: PMC9820549 DOI: 10.3390/ijms24010591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/20/2022] [Accepted: 09/23/2022] [Indexed: 12/31/2022] Open
Abstract
Regardless of the promising use of nanoparticles (NPs) in biomedical applications, several toxic effects have increased the concerns about the safety of these nanomaterials. Although the pathways for NPs toxicity are diverse and dependent upon many parameters such as the nature of the nanoparticle and the biochemical environment, numerous studies have provided evidence that direct contact between NPs and biomolecules or cell membranes leads to cell inactivation or damage and may be a primary mechanism for cytotoxicity. In such a context, this work focused on developing a fast and accurate method to characterize the interaction between NPs, proteins and lipidic membranes by surface plasmon resonance imaging (SPRi) technique. The interaction of gold NPs with mimetic membranes was evaluated by monitoring the variation of reflectivity after several consecutive gold NPs injections on the lipidic membranes prepared on the SPRi biochip. The interaction on the membranes with varied lipidic composition was compared regarding the total surface concentration density of gold NPs adsorbed on them. Then, the interaction of gold and silver NPs with blood proteins was analyzed regarding their kinetic profile of the association/dissociation and dissociation constants (koff). The surface concentration density on the membrane composed of 1-palmitoyl-2-oleoyl-glycero-3-phosphocholine and cholesterol (POPC/cholesterol) was 2.5 times higher than the value found after the injections of gold NPs on POPC only or with dimethyldioctadecylammonium (POPC/DDAB). Regarding the proteins, gold NPs showed preferential binding to fibrinogen resulting in a value of the variation of reflectivity that was 8 times higher than the value found for the other proteins. Differently, silver NPs showed similar interaction on all the tested proteins but with a variation of reflectivity on immunoglobulin G (IgG) 2 times higher than the value found for the other tested proteins.
Collapse
|
8
|
Hu H, Quintana J, Weissleder R, Parangi S, Miller M. Deciphering albumin-directed drug delivery by imaging. Adv Drug Deliv Rev 2022; 185:114237. [PMID: 35364124 PMCID: PMC9117484 DOI: 10.1016/j.addr.2022.114237] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/10/2022] [Accepted: 03/23/2022] [Indexed: 01/03/2023]
Abstract
Albumin is the most abundant plasma protein, exhibits extended circulating half-life, and its properties have long been exploited for diagnostics and therapies. Many drugs intrinsically bind albumin or have been designed to do so, yet questions remain about true rate limiting factors that govern albumin-based transport and their pharmacological impacts, particularly in advanced solid cancers. Imaging techniques have been central to quantifying - at a molecular and single-cell level - the impact of mechanisms such as phagocytic immune cell signaling, FcRn-mediated recycling, oncogene-driven macropinocytosis, and albumin-drug interactions on spatial albumin deposition and related pharmacology. Macroscopic imaging of albumin-binding probes quantifies vessel structure, permeability, and supports efficiently targeted molecular imaging. Albumin-based imaging in patients and animal disease models thus offers a strategy to understand mechanisms, guide drug development and personalize treatments.
Collapse
Affiliation(s)
- Huiyu Hu
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States; Department of Surgery, Massachusetts General Hospital and Harvard Medical School, United States; Department of General Surgery, Xiangya Hospital, Central South University, China
| | - Jeremy Quintana
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States; Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States; Department of Systems Biology, Harvard Medical School, United States
| | - Sareh Parangi
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, United States
| | - Miles Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States; Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States.
| |
Collapse
|
9
|
Drug diffusion in biomimetic hydrogels: importance for drug transport and delivery in non-vascular tumor tissue. Eur J Pharm Sci 2022; 172:106150. [DOI: 10.1016/j.ejps.2022.106150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/25/2022] [Accepted: 02/21/2022] [Indexed: 11/22/2022]
|
10
|
Gupta R, Chen Y, Xie H. In vitro dissolution considerations associated with nano drug delivery systems. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1732. [PMID: 34132050 PMCID: PMC8526385 DOI: 10.1002/wnan.1732] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 12/17/2022]
Abstract
Nano drug delivery systems (NDDS) offer promising solution for the translation of future nanomedicines. As bioavailability and therapeutic outcomes can be improved by altering the drug release from these NDDS, it becomes essential to thoroughly understand their drug release kinetics. Moreover, U.S. Food and Drug Administration requires critical evaluation of potential safety, efficacy, and public health impacts of nanomaterials. Spiraling up market share of NDDS has also stimulated the pharmaceutical industry to develop their cost-effective generic versions after the expiry of patent and associated exclusivity. However, unlike the conventional dosage forms, the in vivo disposition of NDDS is highly intricate and different from their in vitro behavior. Significant challenges exist in the establishment of in vitro-in vivo correlation (IVIVC) due to incomplete understanding of nanoparticles' in vivo biofate and its impact on in vitro experimental protocols. A rational design of dissolution may serve as quality and quantity control tool and help develop a meaningful IVIVC for favorable economic implications. Clinically relevant drug product specifications (critical quality attributes) can be identified by establishing a link between in vitro performance and in vivo exposure. In vitro dissolution may also play a pivotal role to understand the dissolution-mediated clearance and safety of NDDS. Prevalent in vitro dissolution methods for NDDS and their limitations are discussed in this review, among which USP 4 is gaining more interest recently. Researchers are working diligently to develop biorelevant in vitro release assays to ensure optimal therapeutic performance of generic versions of these NDDS. This article focuses on these studies and presents important considerations for the future development of clinically relevant in vitro release methods. This article is categorized under: Toxicology and Regulatory Issues in Nanomedicine > Regulatory and Policy Issues in Nanomedicine.
Collapse
Affiliation(s)
- Ritu Gupta
- Department of Pharmaceutical Science, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA 77004
| | - Yuan Chen
- Department of Pharmaceutical Science, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA 77004
| | - Huan Xie
- Department of Pharmaceutical Science, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA 77004
| |
Collapse
|
11
|
Yang M, Wu E, Tang W, Qian J, Zhan C. Interplay between nanomedicine and protein corona. J Mater Chem B 2021; 9:6713-6727. [PMID: 34328485 DOI: 10.1039/d1tb01063h] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nanomedicine is recognized as a promising agent for diverse biomedical applications; however, its safety and efficiency in clinical practice remains to be enhanced. A priority issue is the protein corona (PC), which imparts unique biological identities to prototype and determines the actual biological functions in biological fluids. Decades of work has already illuminated abundant considerations that influence the composition of the protein corona. Thereinto, the physical assets of nanomedicines (e.g., size and shape, surface properties, nanomaterials) and the biological environment collectively play fundamental roles in shaping the PC, including the types and quantities of plasma proteins. The properties of nanomedicines are dependent on certain factors. This review aims to explore the applications of nanomedicines by regulating their interplay with PC.
Collapse
Affiliation(s)
- Min Yang
- Department of Pharmacology, School of Basic Medical Sciences & Center of Medical Research and Innovation, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P. R. China.
| | - Ercan Wu
- MOE Key Laboratory of Smart Drug Delivery, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Wenjing Tang
- MOE Key Laboratory of Smart Drug Delivery, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Jun Qian
- MOE Key Laboratory of Smart Drug Delivery, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Changyou Zhan
- Department of Pharmacology, School of Basic Medical Sciences & Center of Medical Research and Innovation, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P. R. China. and MOE Key Laboratory of Smart Drug Delivery, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| |
Collapse
|
12
|
Guan J, Chen W, Yang M, Wu E, Qian J, Zhan C. Regulation of in vivo delivery of nanomedicines by herbal medicines. Adv Drug Deliv Rev 2021; 174:210-228. [PMID: 33887404 DOI: 10.1016/j.addr.2021.04.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 04/06/2021] [Accepted: 04/16/2021] [Indexed: 12/15/2022]
Abstract
Nanomedicines are of increasing scrutiny due to their improved efficacy and/or mitigated side effects. They can be integrated with many other therapeutics to further boost the clinical benefits. Among those, herbal medicines are arousing great interest to be combined with nanomedicines to exert synergistic effects in multifaceted mechanisms. The in vivo performance of nanomedicines which determines the therapeutic efficacy and safety is believed to be heavily influenced by the physio-pathological characters of the body. Activation of multiple immune factors, e.g., complement system, phagocytic cells, lymphocytes, and among many others, can affect the fate of nanomedicines in blood circulation, biodistribution, interaction with single cells and intracellular transport. Immunomodulatory effects and metabolic regulation by herbal medicines have been widely witnessed during the past decades, which alter the physio-pathological conditions and dramatically affect in vivo delivery of nanomedicines. In this review, we summarize recent progress of understanding on the in vivo delivery process of nanomedicines and analyze the major affecting factors that regulate the interaction of nanomedicines with organisms. We discuss the immunomodulatory roles and metabolic regulation by herbal medicines and their effects on in vivo delivery process of nanomedicines, as well as the prospective clinical benefits from the combination of nanomedicines and herbal medicines.
Collapse
|
13
|
Wang J, Yeung BZ, Wientjes MG, Cui M, Peer CJ, Lu Z, Figg WD, Woo S, Au JLS. A Quantitative Pharmacology Model of Exosome-Mediated Drug Efflux and Perturbation-Induced Synergy. Pharmaceutics 2021; 13:pharmaceutics13070997. [PMID: 34209265 PMCID: PMC8308960 DOI: 10.3390/pharmaceutics13070997] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/21/2021] [Accepted: 06/24/2021] [Indexed: 01/15/2023] Open
Abstract
Exosomes, naturally occurring vesicles secreted by cells, are undergoing development as drug carriers. We used experimental and computational studies to investigate the kinetics of intracellular exosome processing and exosome-mediated drug efflux and the effects of exosome inhibition. The experiments used four human-breast or ovarian cancer cells, a cytotoxic drug paclitaxel (PTX), two exosome inhibitors (omeprazole (OME), which inhibits exosome release, and GW4869 (GW), which inhibits synthesis of sphingolipid ceramide required for exosome formation), LC-MS/MS analysis of PTX levels in exosomes, and confocal microscopic study of endocytic transport (monitored using fluorescent nanoparticles and endocytic organelle markers). In all four cells, exosome production was enhanced by PTX but diminished by OME or GW (p < 0.05); the PTX enhancement was completely reversed by OME or GW. Co-treatment with OME or GW simultaneously reduced PTX amount in exosomes and increased PTX amount and cytotoxicity in exosome-donor cells (corresponding to >2-fold synergy as indicated by curve shift and uncertainty envelope analyses). This synergy is consistent with the previous reports that OME co-administration significantly enhances the taxane activity in tumor-bearing mice and in patients with triple negative metastatic breast cancer. The experimental results were used to develop a quantitative pharmacology model; model simulations revealed the different effects of the two exosome inhibitors on intracellular PTX processing and subcellular distribution.
Collapse
Affiliation(s)
- Jin Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (J.W.); (B.Z.Y.); (M.C.)
- Institute of Quantitative Systems Pharmacology, Carlsbad, CA 92008, USA;
| | - Bertrand Z. Yeung
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (J.W.); (B.Z.Y.); (M.C.)
- Optimum Therapeutics LLC, Carlsbad, CA 92008, USA;
| | - M. Guillaume Wientjes
- Institute of Quantitative Systems Pharmacology, Carlsbad, CA 92008, USA;
- Optimum Therapeutics LLC, Carlsbad, CA 92008, USA;
| | - Minjian Cui
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (J.W.); (B.Z.Y.); (M.C.)
- Optimum Therapeutics LLC, Carlsbad, CA 92008, USA;
| | - Cody J. Peer
- Clinical Pharmacology Program, National Cancer Institute, NIH, Bethesda, MD 20892, USA; (C.J.P.); (W.D.F.)
| | - Ze Lu
- Optimum Therapeutics LLC, Carlsbad, CA 92008, USA;
| | - William D. Figg
- Clinical Pharmacology Program, National Cancer Institute, NIH, Bethesda, MD 20892, USA; (C.J.P.); (W.D.F.)
| | - Sukyung Woo
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (J.W.); (B.Z.Y.); (M.C.)
- Correspondence: (S.W.); (J.L.-S.A.)
| | - Jessie L.-S. Au
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (J.W.); (B.Z.Y.); (M.C.)
- Institute of Quantitative Systems Pharmacology, Carlsbad, CA 92008, USA;
- Optimum Therapeutics LLC, Carlsbad, CA 92008, USA;
- College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence: (S.W.); (J.L.-S.A.)
| |
Collapse
|
14
|
Wigner P, Zielinski K, Labieniec-Watala M, Marczak A, Szwed M. Doxorubicin-transferrin conjugate alters mitochondrial homeostasis and energy metabolism in human breast cancer cells. Sci Rep 2021; 11:4544. [PMID: 33633284 PMCID: PMC7907108 DOI: 10.1038/s41598-021-84146-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 02/09/2021] [Indexed: 12/18/2022] Open
Abstract
Doxorubicin (DOX) is considered one of the most powerful chemotherapeutic agents but its clinical use has several limitations, including cardiomyopathy and cellular resistance to the drug. By using transferrin (Tf) as a drug carrier, however, the adverse effects of doxorubicin as well as drug resistance can be reduced. The main objective of this study was to determine the exact nature and extent to which mitochondrial function is influenced by DOX-Tf conjugate treatment, specifically in human breast adenocarcinoma cells. We assessed the potential of DOX-Tf conjugate as a drug delivery system, monitoring its cytotoxicity using the MTT assay and ATP measurements. Moreover, we measured the alterations of mitochondrial function and oxidative stress markers. The effect of DOX-Tf was the most pronounced in MDA-MB-231, triple-negative breast cancer cells, whereas non-cancer endothelial HUVEC-ST cells were more resistant to DOX-Tf conjugate than to free DOX treatment. A different sensitivity of two investigate breast cancer cell lines corresponded to the functionality of their cellular antioxidant systems and expression of estrogen receptors. Our data also revealed that conjugate treatment mediated free radical generation and altered the mitochondrial bioenergetics in breast cancer cells.
Collapse
Affiliation(s)
- Paulina Wigner
- Department of Medical Biophysics, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Krzysztof Zielinski
- Department of Medical Biophysics, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Magdalena Labieniec-Watala
- Department of Medical Biophysics, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Agnieszka Marczak
- Department of Medical Biophysics, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Marzena Szwed
- Department of Medical Biophysics, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland.
| |
Collapse
|
15
|
Disturbance of cellular homeostasis as a molecular risk evaluation of human endothelial cells exposed to nanoparticles. Sci Rep 2021; 11:3849. [PMID: 33589697 PMCID: PMC7884700 DOI: 10.1038/s41598-021-83291-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 02/01/2021] [Indexed: 12/27/2022] Open
Abstract
Even though application of nanoparticles in medicine seems to provide unique solutions for drug delivery and diagnosis diseases, understanding interactions between nanoscale materials and biological systems is imperative. Therefore, this study determined the effect of different types of nanoparticles (NPs) on human endothelial cells and examined the types of toxicity responses they can induce. Four different types of NPs were tested (PLA/MMT/TRASTUZUMAB, PLA/EDTMP, PLGA/MDP, and Pluronic F127 MICELLES), representing three putative areas of application: anticancer therapy, scintigraphy, and cosmetology. The experiments were performed on immortalized human umbilical vein endothelial cells (HUVEC-STs). Light contrast phase microscopy as well as cell viability assays showed that only Pluronic F127 MICELLES decreased the number of HUVEC-STs in contrast to PLA/MMT/TRASTUZUMAB, PLA/EDTMP, and PLGA/MDP NPs, which altered cell morphology, but not their confluency. The tested NPs induced not only DNA strand-breaks and alkali-labile sites, but also internucleosomal DNA fragmentation, visualized as a DNA ladder pattern typical of apoptosis. Moreover, generation of free radicals and subsequent mitochondrial membrane potential collapse showed the significance of free radical production during interactions between NPs and endothelial cells. High concentrations of NPs had different degrees of toxicity in human endothelial cells and affected cell proliferation, redox homeostasis, and triggered mitochondrial dysfunction.
Collapse
|
16
|
Wang H, Ding T, Guan J, Liu X, Wang J, Jin P, Hou S, Lu W, Qian J, Wang W, Zhan C. Interrogation of Folic Acid-Functionalized Nanomedicines: The Regulatory Roles of Plasma Proteins Reexamined. ACS NANO 2020; 14:14779-14789. [PMID: 33084315 DOI: 10.1021/acsnano.0c02821] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Folic acid (FA) has been extensively exploited to facilitate targeted delivery of nanomedicines by recognizing the folate receptor-α (FR-α) overexpressed in many human cancers. Unfortunately, none have been approved for clinical use yet. Here we reveal that FA functionalization induces heavy natural IgM absorption on the liposomal surface, depriving FA of receptor recognition and accelerating complement activation in vivo. FA functionalization does not enhance distribution of liposomes in FR-α-overexpressed tumors in comparison to plain liposomes (without FA), but leads to aggravated capture of liposomes by macrophages in the tumor, liver, and spleen. In addition, FA-functionalized polymeric nanoparticles are also vulnerable to natural IgM absorption. This work highlights the pivotal roles of natural IgM in regulating in vivo delivery of FA-functionalized nanomedicines. Due to the prevalent association of immune disorders and varying levels of immunoglobulins with cancer patients, extraordinary cautiousness is urged for clinical translation of FA-enabled targeted delivery systems.
Collapse
Affiliation(s)
- Huan Wang
- Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, People's Republic of China
| | - Tianhao Ding
- Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, People's Republic of China
| | - Juan Guan
- Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, People's Republic of China
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, People's Republic of China
| | - Xia Liu
- Center of Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, People's Republic of China
| | - Jing Wang
- Shanghai Institute of Immunology, Shanghai Jiaotong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Pengpeng Jin
- Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, People's Republic of China
- Center of Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, People's Republic of China
| | - Shuangxing Hou
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, People's Republic of China
| | - Weiyue Lu
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, People's Republic of China
| | - Jun Qian
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, People's Republic of China
| | - Weiping Wang
- Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Changyou Zhan
- Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, People's Republic of China
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, People's Republic of China
- Center of Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, People's Republic of China
| |
Collapse
|
17
|
Cottura N, Howarth A, Rajoli RKR, Siccardi M. The Current Landscape of Novel Formulations and the Role of Mathematical Modeling in Their Development. J Clin Pharmacol 2020; 60 Suppl 1:S77-S97. [PMID: 33205431 DOI: 10.1002/jcph.1715] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/25/2020] [Indexed: 12/15/2022]
Abstract
Drug delivery is an integral part of the drug development process, influencing safety and efficacy of active pharmaceutical ingredients. The application of nanotechnology has enabled the discovery of novel formulations for numerous therapeutic purposes across multiple disease areas. However, evaluation of novel formulations in clinical scenarios is slow and hampered due to various ethical and logistical barriers. Computational models have the ability to integrate existing domain knowledge and mathematical correlations, to rationalize the feasibility of using novel formulations for safely enhancing drug delivery, identifying suitable candidates, and reducing the burden on preclinical and clinical studies. In this review, types of novel formulations and their application through several routes of administration and the use of modeling approaches that can find application in different stages of the novel formulation development process are discussed.
Collapse
Affiliation(s)
- Nicolas Cottura
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Alice Howarth
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Rajith K R Rajoli
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Marco Siccardi
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| |
Collapse
|
18
|
Chen Y, Du M, Yu J, Rao L, Chen X, Chen Z. Nanobiohybrids: A Synergistic Integration of Bacteria and Nanomaterials in Cancer Therapy. BIO INTEGRATION 2020. [DOI: 10.15212/bioi-2020-0008] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Abstract Cancer is a common cause of mortality in the world. For cancer treatment modalities such as chemotherapy, photothermal therapy and immunotherapy, the concentration of therapeutic agents in tumor tissue is the key factor which determines therapeutic efficiency. In
view of this, developing targeted drug delivery systems are of great significance in selectively delivering drugs to tumor regions. Various types of nanomaterials have been widely used as drug carriers. However, the low tumor-targeting ability of nanomaterials limits their clinical application.
It is difficult for nanomaterials to penetrate the tumor tissue through passive diffusion due to the elevated tumoral interstitial fluid pressure. As a biological carrier, bacteria can specifically colonize and proliferate inside tumors and inhibit tumor growth, making it an ideal candidate
as delivery vehicles. In addition, synthetic biology techniques have been applied to enable bacteria to controllably express various functional proteins and achieve targeted delivery of therapeutic agents. Nanobiohybrids constructed by the combination of bacteria and nanomaterials have an
abundance of advantages, including tumor targeting ability, genetic modifiability, programmed product synthesis, and multimodal therapy. Nowadays, many different types of bacteria-based nanobiohybrids have been used in multiple targeted tumor therapies. In this review, firstly we summarized
the development of nanomaterial-mediated cancer therapy. The mechanism and advantages of the bacteria in tumor therapy are described. Especially, we will focus on introducing different therapeutic strategies of nanobiohybrid systems which combine bacteria with nanomaterials in cancer therapy.
It is demonstrated that the bacteria-based nanobiohybrids have the potential to provide a targeted and effective approach for cancer treatment.
Collapse
Affiliation(s)
- Yuhao Chen
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Meng Du
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Jinsui Yu
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Lang Rao
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Zhiyi Chen
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| |
Collapse
|
19
|
Kolitsi LI, Yiantsios SG. Transport of nanoparticles in magnetic targeting: Comparison of magnetic, diffusive and convective forces and fluxes in the microvasculature, through vascular pores and across the interstitium. Microvasc Res 2020; 130:104007. [PMID: 32305349 DOI: 10.1016/j.mvr.2020.104007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/13/2020] [Accepted: 04/13/2020] [Indexed: 02/06/2023]
Abstract
Magnetic nanoparticle targeting in tumor areas is examined by an integrated consideration of the transport steps from the microcirculation to the vascular walls, through their pores and into the interstitium. Brownian, flow- and magnetically induced forces and fluxes are compared on the basis of order-of-magnitude estimates and numerical simulations. The main resistance to nanoparticle transport is found to be within the interstitium, since fluxes there are much smaller than the extravasation fluxes, and the latter are much smaller than the convective-diffusive ones within the microvasculature. For typical nanoparticle sizes, magnetic properties and strengths of magnetic fields as in MRI equipment, magnetic targeting is rather unlikely to play a significant role in directing nanoparticles towards vascular walls or through vascular pores. However, magnetic drift can have an effect within the interstitium and a tangible overall outcome, despite the fact that typical magnetic forces are smaller than Brownian ones or interstitial flow convective forces. The reason behind such an effect has to do with the much larger length scales involved in interstitial transport. Magnetic drift creates a front of large nanoparticle concentrations, flooding the inadequately perfused and poorly accessible tumor area. On the basis of time-scale estimates, it is suggested that sequential cycles of magnetic nanoparticle dosage may help in more efficient access of cell layers ever closer to the tumor center. The present results may assist in the quest for optimal parameters and conditions, given the conflicting requirements for particles small enough to evade hydrodynamic and steric hindrances in vascular pores and the interstitium, yet large enough to bear a substantial magnetic load.
Collapse
Affiliation(s)
- Lydia I Kolitsi
- Department of Chemical Engineering, Aristotle University of Thessaloniki, Univ. Box 453, GR 541 24, Thessaloniki, Greece
| | - Stergios G Yiantsios
- Department of Chemical Engineering, Aristotle University of Thessaloniki, Univ. Box 453, GR 541 24, Thessaloniki, Greece.
| |
Collapse
|
20
|
Ng TS, Garlin MA, Weissleder R, Miller MA. Improving nanotherapy delivery and action through image-guided systems pharmacology. Theranostics 2020; 10:968-997. [PMID: 31938046 PMCID: PMC6956809 DOI: 10.7150/thno.37215] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 08/04/2019] [Indexed: 12/12/2022] Open
Abstract
Despite recent advances in the translation of therapeutic nanoparticles (TNPs) into the clinic, the field continues to face challenges in predictably and selectively delivering nanomaterials for the treatment of solid cancers. The concept of enhanced permeability and retention (EPR) has been coined as a convenient but simplistic descriptor of high TNP accumulation in some tumors. However, in practice EPR represents a number of physiological variables rather than a single one (including dysfunctional vasculature, compromised lymphatics and recruited host cells, among other aspects of the tumor microenvironment) — each of which can be highly heterogenous within a given tumor, patient and across patients. Therefore, a clear need exists to dissect the specific biophysical factors underlying the EPR effect, to formulate better TNP designs, and to identify patients with high-EPR tumors who are likely to respond to TNP. The overall pharmacology of TNP is governed by an interconnected set of spatially defined and dynamic processes that benefit from a systems-level quantitative approach, and insights into the physiology have profited from the marriage between in vivo imaging and quantitative systems pharmacology (QSP) methodologies. In this article, we review recent developments pertinent to image-guided systems pharmacology of nanomedicines in oncology. We first discuss recent developments of quantitative imaging technologies that enable analysis of nanomaterial pharmacology at multiple spatiotemporal scales, and then examine reports that have adopted these imaging technologies to guide QSP approaches. In particular, we focus on studies that have integrated multi-scale imaging with computational modeling to derive insights about the EPR effect, as well as studies that have used modeling to guide the manipulation of the EPR effect and other aspects of the tumor microenvironment for improving TNP action. We anticipate that the synergistic combination of imaging with systems-level computational methods for effective clinical translation of TNPs will only grow in relevance as technologies increase in resolution, multiplexing capability, and in the ability to examine heterogeneous behaviors at the single-cell level.
Collapse
|
21
|
Nguyen L, Li M, Woo S, You Y. Development of Prodrugs for PDT-Based Combination Therapy Using a Singlet-Oxygen-Sensitive Linker and Quantitative Systems Pharmacology. J Clin Med 2019; 8:jcm8122198. [PMID: 31847080 PMCID: PMC6947033 DOI: 10.3390/jcm8122198] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/05/2019] [Accepted: 12/06/2019] [Indexed: 12/20/2022] Open
Abstract
Photodynamic therapy (PDT) has become an effective treatment for certain types of solid tumors. The combination of PDT with other therapies has been extensively investigated in recent years to improve its effectiveness and expand its applications. This focused review summarizes the development of a prodrug system in which anticancer drugs are activated locally at tumor sites during PDT treatment. The development of a singlet-oxygen-sensitive linker that can be conveniently conjugated to various drugs and efficiently cleaved to release intact drugs is recapitulated. The initial design of prodrugs, preliminary efficacy evaluation, pharmacokinetics study, and optimization using quantitative systems pharmacology is discussed. Current treatment optimization in animal models using physiologically based a pharmacokinetic (PBPK) modeling approach is also explored.
Collapse
Affiliation(s)
- Luong Nguyen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (L.N.); (M.L.); (S.W.)
| | - Mengjie Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (L.N.); (M.L.); (S.W.)
| | - Sukyung Woo
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (L.N.); (M.L.); (S.W.)
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, USA
| | - Youngjae You
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (L.N.); (M.L.); (S.W.)
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, USA
- Correspondence: ; Tel.: +1-716-645-4843
| |
Collapse
|
22
|
Liang Y, Xie M, Li J, Liu L, Cao Y. Influence of 3-Hydroxyflavone on Colloidal Stability and Internationalization of Ag Nanomaterials Into THP-1 Macrophages. Dose Response 2019; 17:1559325819865713. [PMID: 31384242 PMCID: PMC6657132 DOI: 10.1177/1559325819865713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/10/2019] [Accepted: 07/02/2019] [Indexed: 11/19/2022] Open
Abstract
Polyphenols as typical food components can influence the colloidal properties and internalization of nanomaterials (NMs) into mammalian cells. Recently, we found that 3-hydroxyflavone (H3) promoted intracellular Zn ions in ZnO nanoparticle (NP) exposed Caco-2 and HepG2 cells. However, it is unclear if H3 could affect the internalization of metal-based NMs with different morphologies. This study investigated the influence of H3 on colloidal aspects of Ag NPs and Ag nanoflakes (NFs) as well as the internalization of Ag NMs into THP-1 macrophages. 3-Hydroxyflavone at 50 μM promoted the solubility and altered hydrodynamic size, polydispersity index, and ζ potential of Ag NPs and Ag NFs, which indicated that H3 could affect the colloidal stability of Ag NMs. Only H3 but not Ag NMs significantly decreased mitochondrial activities of THP-1 macrophages. The internalization of Ag NMs was markedly increased due to the presence of H3. 3-Hydroxyflavone also exhibited antioxidative properties as it reduced intracellular reactive oxygen species and promoted the activities of ABC transporters as it reduced retention of Calcein in Ag NM-exposed THP-1 macrophages. We concluded that H3 promoted the internalization of Ag NMs into macrophages probably by altering the colloidal stability of Ag NMs and consequently NM-macrophage interactions.
Collapse
Affiliation(s)
- Yongqi Liang
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha, People’s Republic of China
- Key Laboratory of Environment-Friendly Chemistry and Applications of Ministry Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan, People’s Republic of China
| | - Min Xie
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha, People’s Republic of China
- Key Laboratory of Environment-Friendly Chemistry and Applications of Ministry Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan, People’s Republic of China
| | - Juan Li
- Key Laboratory of Environment-Friendly Chemistry and Applications of Ministry Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan, People’s Republic of China
| | - Liangliang Liu
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha, People’s Republic of China
| | - Yi Cao
- Key Laboratory of Environment-Friendly Chemistry and Applications of Ministry Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan, People’s Republic of China
| |
Collapse
|
23
|
Au JLS, Lu Z, Abbiati RA, Wientjes MG. Systemic Bioequivalence Is Unlikely to Equal Target Site Bioequivalence for Nanotechnology Oncologic Products. AAPS J 2019; 21:24. [PMID: 30710324 PMCID: PMC6432930 DOI: 10.1208/s12248-019-0296-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 01/09/2019] [Indexed: 11/30/2022] Open
Abstract
Approval of generic drugs by the US Food and Drug Administration (FDA) requires the product to be pharmaceutically equivalent to the reference listed drug (RLD) and demonstrate bioequivalence (BE) in effectiveness when administered to patients under the conditions in the RLD product labeling. Effectiveness is determined by drug exposure at the target sites. However, since such measurement is usually unavailable, systemic exposure is assumed to equal target site exposure and systemic BE to equal target site BE. This assumption, while it often applies to small molecule drug products that are readily dissolved in biological fluids and systemically absorbed, is unlikely to apply to nanotechnology products (NP) that exist as heterogeneous systems and are subjected to dimension- and material-dependent changes. This commentary provides an overview of the intersecting and spatial-dependent processes and variables governing the delivery and residence of oncologic NP in solid tumors. In order to provide a quantitative perspective of the collective effects of these processes, we used quantitative systems pharmacology (QSP) multi-scale modeling to capture the physicochemical and biological events on several scales (whole-body, organ/suborgan, cell/subcellular, spatial locations, time). QSP is an emerging field that entails using modeling and computation to facilitate drug development; an analogous approach (i.e., model-informed drug development) is advocated by to FDA. The QSP model-based simulations illustrated that small changes in NP attributes (e.g., size variations during manufacturing, interactions with proteins in biological milieu) could lead to disproportionately large differences in target site exposure, rending systemic BE unlikely to equal target site BE.
Collapse
Affiliation(s)
- Jessie L-S Au
- Institute of Quantitative Systems Pharmacology, 1815 Aston Avenue, suite 107, Carlsbad, California, 92008, USA.
- Optimum Therapeutics LLC, Carlsbad, California, 92008, USA.
- Department of Pharmaceutical Sciences, University of Oklahoma, Oklahoma City, Oklahoma, 73117, USA.
- College of Pharmacy, Taipei Medical University, Taipei, Taiwan, Republic of China.
| | - Ze Lu
- Institute of Quantitative Systems Pharmacology, 1815 Aston Avenue, suite 107, Carlsbad, California, 92008, USA
- Optimum Therapeutics LLC, Carlsbad, California, 92008, USA
| | - Roberto A Abbiati
- Institute of Quantitative Systems Pharmacology, 1815 Aston Avenue, suite 107, Carlsbad, California, 92008, USA
- Department of Pharmaceutical Sciences, University of Oklahoma, Oklahoma City, Oklahoma, 73117, USA
| | - M Guillaume Wientjes
- Institute of Quantitative Systems Pharmacology, 1815 Aston Avenue, suite 107, Carlsbad, California, 92008, USA
- Optimum Therapeutics LLC, Carlsbad, California, 92008, USA
| |
Collapse
|