1
|
Einolghasi FB, Zeinali B, Vafai K, Mojra A. Development of a prediction model for hyperthermia-enhanced drug delivery using thermosensitive nanoparticles. J Therm Biol 2025; 129:104124. [PMID: 40300402 DOI: 10.1016/j.jtherbio.2025.104124] [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: 01/21/2025] [Revised: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 05/01/2025]
Abstract
Therapies targeting solid tumors are often hindered by drug resistance mechanisms, which presenting significant challenges. This study investigates the synergistic effect of hyperthermia and chemotherapy by utilizing thermosensitive nanoparticles (NPs) to enhance drug uptake by cancer cells. The thermoresponsive nature of NPs enables precise drug release under hyperthermic conditions, making them ideal for localized treatment. To achieve this, we developed a multi-component numerical model of focused ultrasound-enhanced doxorubicin delivery using tumor-targeting nanoparticles. The model incorporates critical factors such as blood coagulation and non-Fourier heat transfer, and it was validated against experimental results, thereby enhancing the reliability and accuracy of our findings. Ultimately, a prediction model was developed to estimate heat-driven irreversible cell damage in relation to heating power and NP size. The results demonstrated a remarkable 13-fold increase in drug penetration and a 4.6-fold enhancement in the fraction of killed cells with hyperthermia. These findings underscore the importance of accurately controlling the temperature field and heating power for optimal hyperthermia-assisted chemotherapy outcomes.
Collapse
Affiliation(s)
| | - Behnam Zeinali
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Kambiz Vafai
- Department of Mechanical Engineering, University of California, Riverside, CA, 92521, USA.
| | - Afsaneh Mojra
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran.
| |
Collapse
|
2
|
Koehler JK, Schmager S, Schnur J, Gedda L, Edwards K, Heerklotz H, Massing U. Novel thermosensitive small multilamellar lipid nanoparticles with promising release characteristics made by dual centrifugation. Eur J Pharm Sci 2025; 206:106999. [PMID: 39730031 DOI: 10.1016/j.ejps.2024.106999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024]
Abstract
Thermosensitive liposomes (TSLs) have great potential for the selective delivery of cytostatic drugs to the tumor site with greatly reduced side effects. Here we report the discovery and characterization of new thermosensitive small multilamellar lipid nanoparticles (tSMLPs) with unusually high temperature selectivity. Furthermore, the temperature-dependent release of the fluorescent marker calcein from tSMLPs is enhanced by human serum albumin. tSMLPs can easily be prepared through dual centrifugation (DC) at very high lipid concentrations using dipalmitoyl and distearoyl phosphatidylcholine (DPPC, DSPC) and the phospholipid dipalmitoyl-sn-glycero-phosphatidyldiglycerol (DPPG2). The new particles have a hydrodynamic diameter of about 175 nm and a narrow size distribution (PDI 0.02). tSMLPs consist of multiple lipid membranes, which become increasingly closer packed towards the particle center, and have no visible aqueous core. The particles are highly stable due to strong hydrogen bond-based membrane interactions mediated by DPPG2. tSMLPs can be used as carriers for water-soluble drugs (EE 25 %) entrapped within the interlamellar spaces. Based on biophysical (DSC, DLS and ITC) and morphological (cryo-EM) studies, a hypothesis is presented to explain the structural basis underlying the high temperature selectivity, as well as the unusual morphology of the new thermosensitive lipid nanoparticles.
Collapse
Affiliation(s)
- Jonas K Koehler
- Institute of Pharmaceutical Sciences, University of Freiburg, 79104 Freiburg im Breisgau, Germany.
| | - Stefanie Schmager
- Institute of Pharmaceutical Sciences, University of Freiburg, 79104 Freiburg im Breisgau, Germany
| | - Johannes Schnur
- Institute of Pharmaceutical Sciences, University of Freiburg, 79104 Freiburg im Breisgau, Germany
| | - Lars Gedda
- Department of Chemistry-Ångström, Uppsala University, 75237 Uppsala, Sweden
| | - Katarina Edwards
- Department of Chemistry-Ångström, Uppsala University, 75237 Uppsala, Sweden
| | - Heiko Heerklotz
- Institute of Pharmaceutical Sciences, University of Freiburg, 79104 Freiburg im Breisgau, Germany; Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada; Signaling Research Centers BIOSS and CIBBS, University of Freiburg, 79104 Freiburg im Breisgau, Germany
| | - Ulrich Massing
- Institute of Pharmaceutical Sciences, University of Freiburg, 79104 Freiburg im Breisgau, Germany; Andreas Hettich GmbH, 78532 Tuttlingen, Germany.
| |
Collapse
|
3
|
Gleue L, Graefen B, Voigt M, Schupp J, Schneider D, Fichter M, Kuske M, Mailaender V, Tuettenberg A, Helm M. Dual Centrifugation-Based Screening for pH-Responsive Liposomes. ChemMedChem 2025; 20:e202400648. [PMID: 39328087 DOI: 10.1002/cmdc.202400648] [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: 08/19/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024]
Abstract
In liposomal drug delivery development, the delicate balance of membrane stability is a major challenge to prevent leakage (during shelf-life and blood circulation), and to ensure efficient payload release at the therapeutic destination. Our composite screening approach uses the processing by dual centrifugation technique to speed up the identification of de novo formulations of intermediate membrane stability. By screening binary lipid combinations at systemically varied ratios we highlight liposomal formulations of intermediate stability, what we termed "the edge of stability", requiring moderate stimuli for destabilization. Supplementation with a pH-sensitive cholesterol derivative (to obtain acid labile liposomes) and renewed assessment with cargo load led to the discovery of three formulations with sufficient shelf-life stability, acceptable cargo retention and efficient pH-responsive cargo release in vitro. The "lead candidates" exhibited promising in cellulo uptake with increased intracellular cargo release and revealed in vivo performance advantages compared to a control liposome. Our approach filters lipid compositions on "the edge of stability" that were introduced with a pH-sensitive cholesterol derivate leading pH-responsive liposomes, out of a multidimensional parameter space. Their discovery by rational approaches would have been highly unlikely, thus highlighting the potential of our screening approach.
Collapse
Affiliation(s)
- Lukas Gleue
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudingerweg 5, 55128, Mainz, Germany
| | - Barbara Graefen
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Matthias Voigt
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudingerweg 5, 55128, Mainz, Germany
- BioNTech SE, An der Goldgrube 12, 55131, Mainz, Germany
| | - Jonathan Schupp
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
- Frankfurt Cancer Institute, Paul-Ehrlich-Straße 42-44, 60596, Frankfurt, Germany
- Goethe University Frankfurt, Institute of Neurology (Edinger Institute), Heinrich-Hoffmann-Straße 7, 60528, Frankfurt, Germany
| | - Dirk Schneider
- Department of Chemistry-Biochemistry, Johannes Gutenberg University, Hanns-Dieter-Hüsch-Weg 17, 55128, Mainz, Germany
| | - Michael Fichter
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
- Paul-Klein-Center for Immunintervention, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Michael Kuske
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
- Paul-Klein-Center for Immunintervention, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
- Institute for translational oncology Mainz (TRON-Mainz), Freiligrathstraße 12, 55131, Mainz, Germany
| | - Volker Mailaender
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
- Paul-Klein-Center for Immunintervention, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Andrea Tuettenberg
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Mark Helm
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudingerweg 5, 55128, Mainz, Germany
| |
Collapse
|
4
|
Abtahi MS, Fotouhi A, Rezaei N, Akalin H, Ozkul Y, Hossein-Khannazer N, Vosough M. Nano-based drug delivery systems in hepatocellular carcinoma. J Drug Target 2024; 32:977-995. [PMID: 38847573 DOI: 10.1080/1061186x.2024.2365937] [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: 03/12/2024] [Revised: 05/22/2024] [Accepted: 06/02/2024] [Indexed: 06/19/2024]
Abstract
The high recurrence rate of hepatocellular carcinoma (HCC) and poor prognosis after medical treatment reflects the necessity to improve the current chemotherapy protocols, particularly drug delivery methods. Development of targeted and efficient drug delivery systems (DDSs), in all active, passive and stimuli-responsive forms for selective delivery of therapeutic drugs to the tumour site has been extended to improve efficacy and reduce the severe side effects. Recent advances in nanotechnology offer promising breakthroughs in the diagnosis, treatment and monitoring of cancer cells. In this review, the specific design of DDSs based on the different nano-particles and their surface engineering is discussed. In addition, the innovative clinical studies in which nano-based DDS was used in the treatment of HCC were highlighted.
Collapse
Affiliation(s)
- Maryam Sadat Abtahi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Alireza Fotouhi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Niloufar Rezaei
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hilal Akalin
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Yusuf Ozkul
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Nikoo Hossein-Khannazer
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
5
|
Alrbyawi H. Stimuli-Responsive Liposomes of 5-Fluorouracil: Progressive Steps for Safe and Effective Treatment of Colorectal Cancer. Pharmaceutics 2024; 16:966. [PMID: 39065663 PMCID: PMC11280302 DOI: 10.3390/pharmaceutics16070966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
5-Fluorouracil (5-FU) has become one of the most widely employed antimetabolite chemotherapeutic agents in recent decades to treat various types of cancer. It is considered the standard first-line treatment for patients with metastatic colorectal cancer. Unfortunately, traditional chemotherapy with 5-FU presents many limitations, such as a short half-life, a low bioavailability, and a high cytotoxicity, affecting both tumor tissue and healthy tissue. In order to overcome the drawbacks of 5-FU and enhance its therapeutic effectiveness against colorectal cancer, many studies have focused on designing new delivery systems to successfully deliver 5-FU to tumor sites. Liposomes have gained attention as a well-accepted nanocarrier for several chemotherapeutic agents. These amphipathic spherical vesicles consist of one or more phospholipid bilayers, showing promise for the drug delivery of both hydrophobic and hydrophilic components in addition to distinctive properties, such as biodegradability, biocompatibility, a low toxicity, and non-immunogenicity. Recent progress in liposomes has mainly focused on chemical and structural modifications to specifically target and activate therapeutic actions against cancer within the proximity of tumors. This review provides a comprehensive overview of both internal-stimuli-responsive liposomes, such as those activated by enzymes or pH, and external-stimuli-responsive liposomes, such as those activated by the application of a magnetic field, light, or temperature variations, for the site-specific delivery of 5-FU in colorectal cancer therapy, along with the future perspectives of these smart-delivery liposomes in colorectal cancer. In addition, this review critically highlights recent innovations in the literature on various types of stimuli-responsive liposomal formulations designed to be applied either exogenously or endogenously and that have great potential in delivering 5-FU to colorectal cancer sites.
Collapse
Affiliation(s)
- Hamad Alrbyawi
- Department of Pharmaceutics and Pharmaceutical Industries, College of Pharmacy, Taibah University, Madinah 41477, Saudi Arabia
| |
Collapse
|
6
|
Lammers T. Nanomedicine Tumor Targeting. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312169. [PMID: 38361435 DOI: 10.1002/adma.202312169] [Citation(s) in RCA: 51] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/24/2024] [Indexed: 02/17/2024]
Abstract
Nanomedicines are extensively explored for cancer therapy. By delivering drug molecules more efficiently to pathological sites and by attenuating their accumulation in healthy organs and tissues, nanomedicine formulations aim to improve the balance between drug efficacy and toxicity. More than 20 cancer nanomedicines are approved for clinical use, and hundreds of formulations are in (pre)clinical development. Over the years, several key pitfalls have been identified as bottlenecks in nanomedicine tumor targeting and translation. These go beyond materials- and production-related issues, and particularly also encompass biological barriers and pathophysiological heterogeneity. In this manuscript, the author describes the most important principles, progress, and products in nanomedicine tumor targeting, delineates key current problems and challenges, and discusses the most promising future prospects to create clinical impact.
Collapse
Affiliation(s)
- Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Center for Biohyhrid Medical Systems, University Hospital RWTH Aachen, Forckenbeckstrasse 55, 52074, Aachen, Germany
| |
Collapse
|
7
|
Shomope I, Percival KM, Abdel Jabbar NM, Husseini GA. Predicting Calcein Release from Ultrasound-Targeted Liposomes: A Comparative Analysis of Random Forest and Support Vector Machine. Technol Cancer Res Treat 2024; 23:15330338241296725. [PMID: 39539114 PMCID: PMC11561990 DOI: 10.1177/15330338241296725] [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: 07/03/2024] [Revised: 09/29/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVE This study presents a comparative analysis of RF and SVM for predicting calcein release from ultrasound-triggered, targeted liposomes under varied low-frequency ultrasound (LFUS) power densities (6.2, 9, and 10 mW/cm2). METHODS Liposomes loaded with calcein and targeted with seven different moieties (cRGD, estrone, folate, Herceptin, hyaluronic acid, lactobionic acid, and transferrin) were synthesized using the thin-film hydration method. The liposomes were characterized using Dynamic Light Scattering and Bicinchoninic Acid assays. Extensive data collection and preprocessing were performed. RF and SVM models were trained and evaluated using mean absolute error (MAE), mean squared error (MSE), coefficient of determination (R²), and the a20 index as performance metrics. RESULTS RF consistently outperformed SVM, achieving R2 scores above 0.96 across all power densities, particularly excelling at higher power densities and indicating a strong correlation with the actual data. CONCLUSION RF outperforms SVM in drug release prediction, though both show strengths and apply based on specific prediction needs.
Collapse
Affiliation(s)
- Ibrahim Shomope
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah 26666, United Arab Emirates
| | - Kelly M. Percival
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah 26666, United Arab Emirates
| | - Nabil M. Abdel Jabbar
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah 26666, United Arab Emirates
| | - Ghaleb A. Husseini
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah 26666, United Arab Emirates
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah PO Box 26666, United Arab Emirates
| |
Collapse
|
8
|
Rysin A, Lokerse WJM, Paal M, Habler K, Wedmann B, Hossann M, Winter G, Lindner LH. Heat-Triggered Release of Dexamethasone from Thermosensitive Liposomes Using Prodrugs or Excipients. J Pharm Sci 2023; 112:1947-1956. [PMID: 37030437 DOI: 10.1016/j.xphs.2023.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/02/2023] [Accepted: 04/02/2023] [Indexed: 04/10/2023]
Abstract
Dexamethasone (DXM) is a potent glucocorticoid with an anti-inflammatory and anti-angiogenic activity which is widely clinically used. Systemic side effects limit the long-term use of DXM in patients requiring formulations which deliver and selectively release the drug to the diseased tissues. This in vitro study compares the suitability of DXM and commonly used prodrugs dexamethasone-21-phosphate (DXMP) and dexamethasone-21-palmitate (DP) as well as DXM complexed by 2-hydroxypropyl-γ-cyclodextrin (HP-γ-CD) for the use in thermosensitive liposomes (TSL). DXM showed a poor retention and a low final drug:lipid ratio in a 1,2-dipalmitoyl-sn‑glycero-3-phosphodiglycerol-based TSL (DPPG2-TSL) and a low-temperature sensitive liposome (LTSL). In contrast to DXM, DXMP and DP were stably retained at 37 °C in TSL in serum and could be encapsulated with high drug:lipid ratios in DPPG2-TSL and LTSL. DXMP showed a rapid release at mild hyperthermia (HT) from both TSL in serum, whereas DP remained incorporated in the TSL bilayer. According to release experiments with carboxyfluorescein (CF), HP-γ-CD and 2-hydroxypropyl-β-cyclodextrin (HP-β-CD) are suitable vehicles for the loading of DXM into DPPG2-TSL and LTSL. Complexation of DXM with HP-γ-CD increased the aqueous solubility of the drug leading to approx. ten times higher DXM:lipid ratio in DPPG2-TSL and LTSL in comparison to un-complexed DXM. Both DXM and HP-γ-CD showed increased release at HT in comparison to 37 °C in serum. In conclusion, DXMP and DXM complexed by HP-γ-CD represent promising candidates for TSL delivery.
Collapse
Affiliation(s)
- Alexander Rysin
- Department of Medicine III, University Hospital, LMU Munich, Germany; Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, LMU Munich, Germany.
| | | | - Michael Paal
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Germany
| | - Katharina Habler
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Germany
| | | | | | - Gerhard Winter
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, LMU Munich, Germany
| | - Lars H Lindner
- Department of Medicine III, University Hospital, LMU Munich, Germany
| |
Collapse
|
9
|
Recent Preclinical and Clinical Progress in Liposomal Doxorubicin. Pharmaceutics 2023; 15:pharmaceutics15030893. [PMID: 36986754 PMCID: PMC10054554 DOI: 10.3390/pharmaceutics15030893] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/12/2023] Open
Abstract
Doxorubicin (DOX) is a potent anti-cancer agent that has garnered great interest in research due to its high efficacy despite dose-limiting toxicities. Several strategies have been exploited to enhance the efficacy and safety profile of DOX. Liposomes are the most established approach. Despite the improvement in safety properties of liposomal encapsulated DOX (in Doxil and Myocet), the efficacy is not superior to conventional DOX. Functionalized (targeted) liposomes present a more effective system to deliver DOX to the tumor. Moreover, encapsulation of DOX in pH-sensitive liposomes (PSLs) or thermo-sensitive liposomes (TSLs) combined with local heating has improved DOX accumulation in the tumor. Lyso-thermosensitive liposomal DOX (LTLD), MM-302, and C225-immunoliposomal(IL)-DOX have reached clinical trials. Further functionalized PEGylated liposomal DOX (PLD), TSLs, and PSLs have been developed and evaluated in preclinical models. Most of these formulations improved the anti-tumor activity compared to the currently available liposomal DOX. However, the fast clearance, the optimization of ligand density, stability, and release rate need more investigations. Therefore, we reviewed the latest approaches applied to deliver DOX more efficiently to the tumor, preserving the benefits obtained from FDA-approved liposomes.
Collapse
|
10
|
Magnetic Thermosensitive Liposomes Loaded with Doxorubicin. Methods Mol Biol 2023; 2622:103-119. [PMID: 36781754 DOI: 10.1007/978-1-0716-2954-3_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Liposome-mediated anticancer drug delivery has the advantage of limiting the massive cytotoxicity of chemotherapeutic agents. Doxorubicin (DOX) PEG-liposomal does however have a slow-release rate that hinders its therapeutic efficacy. In this study, an integrated therapeutic system based on magnetic thermosensitive liposomes was designed. The chelated gadolinium acquired magnetic properties in the liposomes. The hyperthermia induced by ultra-high-field magnetic resonance imaging (UHF-MRI) enhances the chemotherapeutic effects of DOX. The DOX release from liposomes was facilitated over a narrow range of temperatures owing to the phase transition temperature of the liposomes. The magnetic properties of the liposomes were evident by the elevation of contrast after the exposure to UHF-MRI. Moreover, triple-negative breast cancer (TNBC) cells showed a significant decrease in cellular viability reaching less than 40% viability after 1 h of exposure to UHF-MRI. The liposomes demonstrated a physiological coagulation time and a minimal hemolytic potential in hemocompatibility studies; therefore, they were considered safe for physiological application. As a result, magnetic-thermosensitive liposomal guidance of local delivery of DOX could increase the therapeutic index, thereby reducing the amount of the drug required for systemic administration and the chance of affecting the adjacent tissues.
Collapse
|
11
|
Haemmerich D, Ramajayam KK, Newton DA. Review of the Delivery Kinetics of Thermosensitive Liposomes. Cancers (Basel) 2023; 15:cancers15020398. [PMID: 36672347 PMCID: PMC9856714 DOI: 10.3390/cancers15020398] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/10/2023] Open
Abstract
Thermosensitive liposomes (TSL) are triggered nanoparticles that release the encapsulated drug in response to hyperthermia. Combined with localized hyperthermia, TSL enabled loco-regional drug delivery to tumors with reduced systemic toxicities. More recent TSL formulations are based on intravascular triggered release, where drug release occurs within the microvasculature. Thus, this delivery strategy does not require enhanced permeability and retention (EPR). Compared to traditional nanoparticle drug delivery systems based on EPR with passive or active tumor targeting (typically <5%ID/g tumor), TSL can achieve superior tumor drug uptake (>10%ID/g tumor). Numerous TSL formulations have been combined with various drugs and hyperthermia devices in preclinical and clinical studies over the last four decades. Here, we review how the properties of TSL dictate delivery and discuss the advantages of rapid drug release from TSL. We show the benefits of selecting a drug with rapid extraction by tissue, and with quick cellular uptake. Furthermore, the optimal characteristics of hyperthermia devices are reviewed, and impact of tumor biology and cancer cell characteristics are discussed. Thus, this review provides guidelines on how to improve drug delivery with TSL by optimizing the combination of TSL, drug, and hyperthermia method. Many of the concepts discussed are applicable to a variety of other triggered drug delivery systems.
Collapse
Affiliation(s)
- Dieter Haemmerich
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Bioengineering, Clemson University, Clemson, SC 29634, USA
- Correspondence:
| | - Krishna K. Ramajayam
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Danforth A. Newton
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
12
|
Abbasi H, Kouchak M, Mirveis Z, Hajipour F, Khodarahmi M, Rahbar N, Handali S. What We Need to Know about Liposomes as Drug Nanocarriers: An Updated Review. Adv Pharm Bull 2023; 13:7-23. [PMID: 36721822 PMCID: PMC9871273 DOI: 10.34172/apb.2023.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 01/13/2022] [Accepted: 03/31/2022] [Indexed: 02/03/2023] Open
Abstract
Liposomes have been attracted considerable attention as phospholipid spherical vesicles, over the past 40 years. These lipid vesicles are valued in biomedical application due to their ability to carry both hydrophobic and hydrophilic agents, high biocompatibility and biodegradability. Various methods have been used for the synthesis of liposomes, so far and numerous modifications have been performed to introduce liposomes with different characteristics like surface charge, size, number of their layers, and length of circulation in biological fluids. This article provides an overview of the significant advances in synthesis of liposomes via active or passive drug loading methods, as well as describes some strategies developed to fabricate their targeted formulations to overcome limitations of the "first-generation" liposomes.
Collapse
Affiliation(s)
- Hanieh Abbasi
- Department of Medicinal Chemistry, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Kouchak
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Pharmaceutics, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zohreh Mirveis
- Department of Medicinal Chemistry, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fatemeh Hajipour
- Department of Medicinal Chemistry, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohsen Khodarahmi
- Department of Medicinal Chemistry, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nadereh Rahbar
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Corresponding Authors: Nadereh Rahbar and Somayeh Handali, and
| | - Somayeh Handali
- Medical Biomaterials Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran.,Corresponding Authors: Nadereh Rahbar and Somayeh Handali, and
| |
Collapse
|
13
|
Image-guided drug delivery in nanosystem-based cancer therapies. Adv Drug Deliv Rev 2023; 192:114621. [PMID: 36402247 DOI: 10.1016/j.addr.2022.114621] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/18/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022]
Abstract
The past decades have shown significant advancements in the development of solid tumor treatment. For instance, implementation of nanosystems for drug delivery has led to a reduction in side effects and improved delivery to the tumor region. However, clinical translation has faced challenges, as tumor drug levels are still considered to be inadequate. Interdisciplinary research has resulted in the development of more advanced drug delivery systems. These are coined "smart" due to the ability to be followed and actively manipulated in order to have better control over local drug release. Therefore, image-guided drug delivery can be a powerful strategy to improve drug activity at the target site. Being able to visualize the inflow of the administered smart nanosystem within the tumor gives the potential to determine the right moment to apply the facilitator to initiate drug release. Here we provide an overview of available nanosystems, imaging moieties, and imaging techniques. We discuss preclinical application of these smart drug delivery systems, the strength of image-guided drug delivery, and the future of personalized treatment.
Collapse
|
14
|
Dastidar DG, Ghosh D, Das A. Recent developments in nanocarriers for cancer chemotherapy. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
15
|
Bahutair WN, Abuwatfa WH, Husseini GA. Ultrasound Triggering of Liposomal Nanodrugs for Cancer Therapy: A Review. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12173051. [PMID: 36080088 PMCID: PMC9458162 DOI: 10.3390/nano12173051] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 08/27/2022] [Accepted: 08/28/2022] [Indexed: 05/11/2023]
Abstract
Efficient conventional chemotherapy is limited by its nonspecific nature, which causes severe systemic toxicity that can lead to patient discomfort and low therapeutic efficacy. The emergence of smart drug delivery systems (SDDSs) utilizing nanoparticles as drug nanocarriers has shown great potential in enhancing the targetability of anticancer agents and limiting their side effects. Liposomes are among the most investigated nanoplatforms due to their promising capabilities of encapsulating hydrophilic, lipophilic, and amphiphilic drugs, biocompatibility, physicochemical and biophysical properties. Liposomal nanodrug systems have demonstrated the ability to alter drugs' biodistribution by sufficiently delivering the entrapped chemotherapeutics at the targeted diseased sites, sparing normal cells from undesired cytotoxic effects. Combining liposomal treatments with ultrasound, as an external drug release triggering modality, has been proven effective in spatially and temporally controlling and stimulating drug release. Therefore, this paper reviews recent literature pertaining to the therapeutic synergy of triggering nanodrugs from liposomes using ultrasound. It also highlights the effects of multiple physical and chemical factors on liposomes' sonosensetivity, several ultrasound-induced drug release mechanisms, and the efficacy of ultrasound-responsive liposomal systems in cancer therapy. Overall, liposomal nanodrug systems triggered by ultrasound are promising cancer therapy platforms that can potentially alleviate the detriments of conventional cancer treatments.
Collapse
Affiliation(s)
- Wafa N. Bahutair
- Department of Chemical Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box. 26666, United Arab Emirates
| | - Waad H. Abuwatfa
- Department of Chemical Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box. 26666, United Arab Emirates
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box. 26666, United Arab Emirates
| | - Ghaleb A. Husseini
- Department of Chemical Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box. 26666, United Arab Emirates
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box. 26666, United Arab Emirates
- Correspondence:
| |
Collapse
|
16
|
Huang H, Shao L, Chen Y, Han W, Zhou Y, Liu T, Gu J, Zhu H. Sequential Dual Delivery System Based on siCOX-2-Loaded Gold Nanostar and Thermal-Sensitive Liposomes Overcome Hypoxia-Mediated Multidrug Resistance in Tumors. Mol Pharm 2022; 19:2390-2405. [PMID: 35639669 DOI: 10.1021/acs.molpharmaceut.2c00164] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Reversing hypoxia-mediated multidrug resistance (MDR) presents a unique challenge in clinical chemotherapy. Here, a sequential dual delivery system composited with Cyclooxygenase-2 siRNA (siCOX-2) in poly-d-arginine (9R)/2-deoxyglucose (DG)-loaded gold nanostar (GNS) (siCOX-2@RDG) and paclitaxel (PTX)-loaded thermosensitive liposome (PTSL) was proposed to conquer the hypoxia-mediated MDR in tumors. As a result, the prepared siCOX-2@RDG exhibited a starlike morphology with a uniform particle size of 194.36 ± 1.44 nm and a ζ-potential of -11.83 ± 2.01 mV. In vitro, PTSL displayed expected thermal-responsive release properties. As expected, siCOX-2@RDG displayed exceptional DG-mediated hypoxia-targeting capability both in vitro and in vivo and downregulated the expression of COX-2 successfully. Meanwhile, GNS-triggered hyperthermia elevated the cellular uptake of PTSL in PTX-resistant HepG2(HepG2/PTX) cells in vitro and enhanced the permeability of tumor tissues, thus elevating the valid retention of PTX into solid tumors. Finally, we demonstrated that the sequential dual systems composed of siCOX-2@RDG and PTSL could reverse hypoxia-mediated MDR and exhibit excellent synergistic antitumor effects both in vitro and in vivo, prolonging the survival of tumor-bearing mice. The devised sequential dual systems, composed of two independent nanosystems, have a promising potential to overcome hypoxia-mediated MDR in clinical practice.
Collapse
Affiliation(s)
- Haiqin Huang
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Lanlan Shao
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Yan Chen
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Weili Han
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Yao Zhou
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Tianqing Liu
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia
| | - Jinhua Gu
- Department of Clinical Pharmacy, The Affiliated Maternal and Child Health Hospital of Nantong University/Nantong Children's Hospital, Nantong 226001, China
| | - Hongyan Zhu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| |
Collapse
|
17
|
Alrbyawi H, Poudel I, Annaji M, Arnold RD, Tiwari AK, Babu RJ. Recent Advancements of Stimuli-Responsive Targeted Liposomal Formulations for Cancer Drug Delivery. Pharm Nanotechnol 2022; 10:3-23. [PMID: 35156590 DOI: 10.2174/2211738510666220214102626] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/09/2022] [Accepted: 01/18/2022] [Indexed: 11/22/2022]
Abstract
Liposomes have gained attention as a well-accepted nanocarrier for several chemotherapeutic drugs and are considered a drug delivery system of choice for a wide range of products. These amphipathic spherical vesicles primarily consist of one or more phospholipid bilayers, showing promise for drug delivery of both hydrophilic and hydrophobic components in addition to unique properties such as biocompatibility, biodegradability, low toxicity, and non-immunogenicity. Recent advances in liposomes are mainly centered on chemical and structural modification with the multifunctional approach to target the cancer cells activating the offensive mechanisms within the proximity of the tumors. Stimuli-responsive liposomes are a precisive approach to deliver and release chemotherapeutic drugs in the tumor site in a controlled fashion, thus reducing damage to normal tissues and preventing the side effects of the conventional chemotherapy regimen. The unique characteristics in the tumor microenvironment facilitate applying an endogenous stimulus (pH, redox potential, or enzymatic activity) to trigger the release of the drug, or external stimulus (heat or light) could be applied to tailor the drug release from liposomes. This review focuses on newer developments in stimuli-sensitive liposomal drug delivery systems designed to apply either exogenous (temperature, light, and magnetic field) or endogenous (pH changes, enzymatic triggers, or redox potential) approaches.
Collapse
Affiliation(s)
- Hamad Alrbyawi
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA
- Pharmaceutics and Pharmaceutical Technology Department, College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - Ishwor Poudel
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA
| | - Manjusha Annaji
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA
| | - Robert D Arnold
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, The University of Toledo, Toledo, Ohio, 43614, USA
| | - R Jayachandra Babu
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA
| |
Collapse
|
18
|
Sebeke L, Gómez JDC, Heijman E, Rademann P, Maul AC, Ekdawi S, Vlachakis S, Toker D, Mink BL, Schubert-Quecke C, Yeo SY, Schmidt P, Lucas C, Brodesser S, Hossann M, Lindner LH, Grüll H. Hyperthermia-induced doxorubicin delivery from thermosensitive liposomes via MR-HIFU in a pig model. J Control Release 2022; 343:798-812. [DOI: 10.1016/j.jconrel.2022.02.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 01/27/2022] [Accepted: 02/02/2022] [Indexed: 12/17/2022]
|
19
|
Milan A, Mioc A, Prodea A, Mioc M, Buzatu R, Ghiulai R, Racoviceanu R, Caruntu F, Şoica C. The Optimized Delivery of Triterpenes by Liposomal Nanoformulations: Overcoming the Challenges. Int J Mol Sci 2022; 23:1140. [PMID: 35163063 PMCID: PMC8835305 DOI: 10.3390/ijms23031140] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/13/2022] [Accepted: 01/18/2022] [Indexed: 02/06/2023] Open
Abstract
The last decade has witnessed a sustained increase in the research development of modern-day chemo-therapeutics, especially for those used for high mortality rate pathologies. However, the therapeutic landscape is continuously changing as a result of the currently existing toxic side effects induced by a substantial range of drug classes. One growing research direction driven to mitigate such inconveniences has converged towards the study of natural molecules for their promising therapeutic potential. Triterpenes are one such class of compounds, intensively investigated for their therapeutic versatility. Although the pharmacological effects reported for several representatives of this class has come as a well-deserved encouragement, the pharmacokinetic profile of these molecules has turned out to be an unwelcomed disappointment. Nevertheless, the light at the end of the tunnel arrived with the development of nanotechnology, more specifically, the use of liposomes as drug delivery systems. Liposomes are easily synthesizable phospholipid-based vesicles, with highly tunable surfaces, that have the ability to transport both hydrophilic and lipophilic structures ensuring superior drug bioavailability at the action site as well as an increased selectivity. This study aims to report the results related to the development of different types of liposomes, used as targeted vectors for the delivery of various triterpenes of high pharmacological interest.
Collapse
Affiliation(s)
- Andreea Milan
- Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy, 2 E. Murgu Sq., 300041 Timişoara, Romania; (A.M.); (A.M.); (A.P.); (R.G.); (R.R.); (C.Ş.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq., No. 2, 300041 Timişoara, Romania
| | - Alexandra Mioc
- Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy, 2 E. Murgu Sq., 300041 Timişoara, Romania; (A.M.); (A.M.); (A.P.); (R.G.); (R.R.); (C.Ş.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq., No. 2, 300041 Timişoara, Romania
| | - Alexandra Prodea
- Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy, 2 E. Murgu Sq., 300041 Timişoara, Romania; (A.M.); (A.M.); (A.P.); (R.G.); (R.R.); (C.Ş.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq., No. 2, 300041 Timişoara, Romania
| | - Marius Mioc
- Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy, 2 E. Murgu Sq., 300041 Timişoara, Romania; (A.M.); (A.M.); (A.P.); (R.G.); (R.R.); (C.Ş.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq., No. 2, 300041 Timişoara, Romania
| | - Roxana Buzatu
- Faculty of Dental Medicine, “Victor Babeş” University of Medicine and Pharmacy Timişoara, 2 Eftimie Murgu Street, 300041 Timişoara, Romania
| | - Roxana Ghiulai
- Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy, 2 E. Murgu Sq., 300041 Timişoara, Romania; (A.M.); (A.M.); (A.P.); (R.G.); (R.R.); (C.Ş.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq., No. 2, 300041 Timişoara, Romania
| | - Roxana Racoviceanu
- Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy, 2 E. Murgu Sq., 300041 Timişoara, Romania; (A.M.); (A.M.); (A.P.); (R.G.); (R.R.); (C.Ş.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq., No. 2, 300041 Timişoara, Romania
| | - Florina Caruntu
- Faculty of Medicine, “Victor Babeş” University of Medicine and Pharmacy Timişoara, 2 Eftimie Murgu Street, 300041 Timişoara, Romania;
| | - Codruţa Şoica
- Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy, 2 E. Murgu Sq., 300041 Timişoara, Romania; (A.M.); (A.M.); (A.P.); (R.G.); (R.R.); (C.Ş.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq., No. 2, 300041 Timişoara, Romania
| |
Collapse
|
20
|
Han M, Beon J, Lee JY, Oh SS. Systematic Combination of Oligonucleotides and Synthetic Polymers for Advanced Therapeutic Applications. Macromol Res 2021; 29:665-680. [PMID: 34754286 PMCID: PMC8568687 DOI: 10.1007/s13233-021-9093-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/22/2021] [Accepted: 09/25/2021] [Indexed: 11/16/2022]
Abstract
The potential of oligonucleotides is exceptional in therapeutics because of their high safety, potency, and specificity compared to conventional therapeutic agents. However, many obstacles, such as low in vivo stability and poor cellular uptake, have hampered their clinical success. Use of polymeric carriers can be an effective approach for overcoming the biological barriers and thereby maximizing the therapeutic efficacy of the oligonucleotides due to the availability of highly tunable synthesis and functional modification of various polymers. As loaded in the polymeric carriers, the therapeutic oligonucleotides, such as antisense oligonucleotides, small interfering RNAs, microRNAs, and even messenger RNAs, become nuclease-resistant by bypassing renal filtration and can be efficiently internalized into disease cells. In this review, we introduced a variety of systematic combinations between the therapeutic oligonucleotides and the synthetic polymers, including the uses of highly functionalized polymers responding to a wide range of endogenous and exogenous stimuli for spatiotemporal control of oligonucleotide release. We also presented intriguing characteristics of oligonucleotides suitable for targeted therapy and immunotherapy, which can be fully supported by versatile polymeric carriers.
Collapse
Affiliation(s)
- Moohyun Han
- Department of Materials Science and Engineering, Pohang University of Science Technology (POSTECH), Pohang, Gyeongbuk, 37673 Korea
| | - Jiyun Beon
- Department of Materials Science and Engineering, Pohang University of Science Technology (POSTECH), Pohang, Gyeongbuk, 37673 Korea
| | - Ju Young Lee
- Research Center for Bio-based Chemistry, Korea Research Institute of Chemical Technology (KRICT), Ulsan, 44429 Korea
| | - Seung Soo Oh
- Department of Materials Science and Engineering, Pohang University of Science Technology (POSTECH), Pohang, Gyeongbuk, 37673 Korea
| |
Collapse
|
21
|
Preclinical Studies in Small Animals for Advanced Drug Delivery Using Hyperthermia and Intravital Microscopy. Cancers (Basel) 2021; 13:cancers13205146. [PMID: 34680296 PMCID: PMC8534089 DOI: 10.3390/cancers13205146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/05/2021] [Accepted: 10/05/2021] [Indexed: 01/15/2023] Open
Abstract
This paper presents three devices suitable for the preclinical application of hyperthermia via the simultaneous high-resolution imaging of intratumoral events. (Pre)clinical studies have confirmed that the tumor micro-environment is sensitive to the application of local mild hyperthermia. Therefore, heating is a promising adjuvant to aid the efficacy of radiotherapy or chemotherapy. More so, the application of mild hyperthermia is a useful stimulus for triggered drug release from heat-sensitive nanocarriers. The response of thermosensitive nanoparticles to hyperthermia and ensuing intratumoral kinetics are considerably complex in both space and time. To obtain better insight into intratumoral processes, longitudinal imaging (preferable in high spatial and temporal resolution) is highly informative. Our devices are based on (i) an external electric heating adaptor for the dorsal skinfold model, (ii) targeted radiofrequency application, and (iii) a microwave antenna for heating of internal tumors. These models, while of some technical complexity, significantly add to the understanding of effects of mild hyperthermia warranting implementation in research on hyperthermia.
Collapse
|
22
|
Chen Y, Guo M, Qu D, Liu Y, Guo J, Chen Y. Furin-responsive triterpenine-based liposomal complex enhances anticervical cancer therapy through size modulation. Drug Deliv 2021; 27:1608-1624. [PMID: 33179521 PMCID: PMC7676817 DOI: 10.1080/10717544.2020.1827086] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The accumulation and penetration of antitumor drugs in tumor tissues are directly related to their antitumor effects. The particle size of the nanodrug delivery system is one of the most important factors for the accumulation and penetration of antitumor drugs within tumor tissues. Generally, nanodelivery systems of intermediate size (100–120 nm) are capable of efficient accumulation owing to prolonged circulation and enhanced permeability and retention (EPR) effect; however, smaller ones (20–40 nm) are effective for deep penetration within tumor tissue. Currently a conventional drug delivery system cannot possess two types of optimal sizes, simultaneously. To solve this and to enhance cervical cancer treatment, a furin-responsive triterpenine-based liposomal complex (PEGcleavable Tf-CTM/L), with Tf-CTM (transferrin-modified tripterine-loaded coix seed oil microemulsion) in core, coated with a thermo-sensitive lipid and a kind of PEG shell modified with a furin-cleavable peptide was developed to improve tumor-specific accumulation and penetration. Herein, PEGcleavable Tf-CTM/L was capable of efficient accumulation because of EPR effect. The PEG shells could timely detach under stimulation of overexpressed furin protein to solve the problem of the steric hindrance dilemma. The small-sized Tf-CTM released under stimulation of tumor microthermal environment in cervical cancer, which was efficient with regards to deep penetration at tumor sites. Notably, compared to the use of triterpenine alone, PEGcleavable Tf-CTM/L promoted anticervical efficacy and displayed diminished systemic toxicity by efficient accumulation and deep penetration of antitumor drugs within tumor tissues. Our study provides a new strategy, and holds promising potential for anticervical cancer treatment.
Collapse
Affiliation(s)
- Yunyan Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing, China.,School of Pharmacy,Wannan Medical College, Wuhu, China
| | - Mengfei Guo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing, China
| | - Ding Qu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing, China
| | - Yuping Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing, China
| | - Jian Guo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing, China
| | - Yan Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing, China
| |
Collapse
|
23
|
Drug transport kinetics of intravascular triggered drug delivery systems. Commun Biol 2021; 4:920. [PMID: 34321602 PMCID: PMC8319190 DOI: 10.1038/s42003-021-02428-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 07/05/2021] [Indexed: 01/17/2023] Open
Abstract
Intravascular triggered drug delivery systems (IV-DDS) for local drug delivery include various stimuli-responsive nanoparticles that release the associated agent in response to internal (e.g., pH, enzymes) or external stimuli (e.g., temperature, light, ultrasound, electromagnetic fields, X-rays). We developed a computational model to simulate IV-DDS drug delivery, for which we quantified all model parameters in vivo in rodent tumors. The model was validated via quantitative intravital microscopy studies with unencapsulated fluorescent dye, and with two formulations of temperature-sensitive liposomes (slow, and fast release) encapsulating a fluorescent dye as example IV-DDS. Tumor intra- and extravascular dye concentration dynamics were extracted from the intravital microscopy data by quantitative image processing, and were compared to computer model results. Via this computer model we explain IV-DDS delivery kinetics and identify parameters of IV-DDS, of drug, and of target tissue for optimal delivery. Two parameter ratios were identified that exclusively dictate how much drug can be delivered with IV-DDS, indicating the importance of IV-DDS with fast drug release (~sec) and choice of a drug with rapid tissue uptake (i.e., high first-pass extraction fraction). The computational model thus enables engineering of improved future IV-DDS based on tissue parameters that can be quantified by imaging. Ten Hagen et al. developed a computational model to simulate intravascular triggered drug delivery systems (IV-DDS), which they validated using quantitative intravital microscopy in mice. Their model potentially enables the engineering of more efficacious IV-DDS based on parameters that can be quantified by imaging.
Collapse
|
24
|
Evaluation of release and pharmacokinetics of hexadecylphosphocholine (miltefosine) in phosphatidyldiglycerol-based thermosensitive liposomes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2021; 1863:183698. [PMID: 34283999 DOI: 10.1016/j.bbamem.2021.183698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 07/12/2021] [Accepted: 07/12/2021] [Indexed: 12/18/2022]
Abstract
Hexadecylphosphocholine (HePC, Miltefosine) is a drug from the class of alkylphosphocholines with an antineoplastic and antiprotozoal activity. We previously reported that HePC uptake from thermosensitive liposomes (TSL) containing 1,2-dipalmitoyl-sn-glycero-3-phosphodiglycerol (DPPG2) into cancer cells is accelerated at mild hyperthermia (HT) resulting in increased cytotoxicity. In this study, we compared HePC release of different TSL formulations in serum. HePC showed rapid but incomplete release below the transition temperature (Tm) of investigated TSL formulations in serum. Short heating (5 min) to 42 °C increased HePC release from DPPG2-TSL (Tm = 41 °C) by a factor of two in comparison to body temperature (37 °C). Bovine serum albumin (BSA) induced HePC release from DPPG2-TSL comparable to serum. Furthermore, multilamellar vesicles (MLV) were capable to extract HePC from DPPG2-TSL in a concentration- and temperature-dependent manner. Repetitive exposure of DPPG2-TSL to MLV at 37 °C led to a fast initial release of HePC which slowed down after subsequent extraction cycles finally reaching approx. 50% HePC release. A pharmacokinetic study in rats revealed a biphasic pattern with an immediate clearance of approx. 50% HePC whereas the remaining 50% HePC showed a prolonged circulation time. We speculate that HePC located in the external leaflet of DPPG2-TSL is rapidly released upon contact with suitable biological acceptors. As demonstrated by MLV transfer experiments, asymmetric incorporation of HePC into the internal leaflet of DPPG2-TSL might improve HePC retention in presence of complex biological media and still give rise to HT-induced HePC release.
Collapse
|
25
|
Petrini M, Lokerse WJM, Mach A, Hossann M, Merkel OM, Lindner LH. Effects of Surface Charge, PEGylation and Functionalization with Dipalmitoylphosphatidyldiglycerol on Liposome-Cell Interactions and Local Drug Delivery to Solid Tumors via Thermosensitive Liposomes. Int J Nanomedicine 2021; 16:4045-4061. [PMID: 34163158 PMCID: PMC8214027 DOI: 10.2147/ijn.s305106] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/24/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose Previous studies demonstrated the possibility of targeting tumor-angiogenic endothelial cells with positively charged nanocarriers, such as cationic liposomes. We investigated the active targeting potential of positively charged nanoparticles in combination with the heat-induced drug release function of thermosensitive liposomes (TSL). This novel dual-targeted approach via cationic TSL (CTSL) was thoroughly explored using either a novel synthetic phospholipid 1,2-dipalmitoyl-sn-glycero-3-phosphodiglycerol (DPPG2) or a conventional polyethylene glycol (PEG) surface modification. Anionic particles containing either DPPG2 or PEG were also included in the study to highlight difference in tumor enrichment driven by surface charge. With this study, we aim to provide a deep insight into the main differences between DPPG2- and PEG-functionalized liposomes, focusing on the delivery of a well-known cytotoxic drug (doxorubicin; DOX) in combination with local hyperthermia (HT, 41–43°C). Materials and Methods DPPG2- and PEG-based cationic TSLs (PG2-CTSL/PEG-CTSL) were thoroughly analyzed for size, surface charge, and heat-triggered DOX release. Cancer cell targeting and DOX delivery was evaluated by FACS, fluorescence imaging, and HPLC. In vivo particle behavior was analyzed by assessing DOX biodistribution with local HT application in tumor-bearing animals. Results The absence of PEG in PG2-CTSL promoted more efficient liposome–cell interactions, resulting in a higher DOX delivery and cancer cell toxicity compared with PEG-CTSL. By exploiting the dual-targeting function of CTSLs, we were able to selectively trigger DOX release in the intracellular compartment by HT. When tested in vivo, local HT promoted an increase in intratumoral DOX levels for all (C)TSLs tested, with DOX enrichment factors ranging from 3 to 14-fold depending on the type of formulation. Conclusion Cationic particles showed lower hemocompatibility than their anionic counterparts, which was partially mitigated when PEG was grafted on the liposome surface. DPPG2-based anionic TSL showed optimal local drug delivery compared to all other formulations tested, demonstrating the potential advantages of using DPPG2 lipid in designing liposomes for tumor-targeted applications.
Collapse
Affiliation(s)
- Matteo Petrini
- Department of Internal Medicine III, University Hospital, Ludwig Maximilian University, Munich, Germany.,Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig Maximilian University, Munich, Germany
| | - Wouter J M Lokerse
- Department of Internal Medicine III, University Hospital, Ludwig Maximilian University, Munich, Germany
| | - Agnieszka Mach
- Department of Internal Medicine III, University Hospital, Ludwig Maximilian University, Munich, Germany
| | | | - Olivia M Merkel
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig Maximilian University, Munich, Germany
| | - Lars H Lindner
- Department of Internal Medicine III, University Hospital, Ludwig Maximilian University, Munich, Germany
| |
Collapse
|
26
|
Mechanistic investigation of thermosensitive liposome immunogenicity and understanding the drivers for circulation half-life: A polyethylene glycol versus 1,2-dipalmitoyl-sn-glycero-3-phosphodiglycerol study. J Control Release 2021; 333:1-15. [DOI: 10.1016/j.jconrel.2021.03.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 03/10/2021] [Indexed: 12/18/2022]
|
27
|
Lu T, Haemmerich D, Liu H, Seynhaeve AL, van Rhoon GC, Houtsmuller AB, ten Hagen TL. Externally triggered smart drug delivery system encapsulating idarubicin shows superior kinetics and enhances tumoral drug uptake and response. Am J Cancer Res 2021; 11:5700-5712. [PMID: 33897876 PMCID: PMC8058728 DOI: 10.7150/thno.55163] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/09/2021] [Indexed: 12/20/2022] Open
Abstract
Rationale: Increasing the bioavailable drug level in a tumor is the key to enhance efficacy of chemotherapy. Thermosensitive smart drug delivery systems (SDDS) in combination with local hyperthermia facilitate high local drug levels, thus improving uptake in the tumor. However, inability to rapidly and efficiently absorb the locally released drug results in reduced efficacy, as well as undesired redistribution of the drug away from the tumor to the system. Methods: Based on this paradigm we propose a novel approach in which we replaced doxorubicin (DXR), one of the classic drugs for nanocarrier-based delivery, with idarubicin (IDA), a hydrophobic anthracycline used solely in the free form for treatment hematologic cancers. We established a series of in vitro and in vivo experiments to in depth study the kinetics of SDDS-based delivery, drug release, intratumor biodistribution and subsequent cell uptake. Results: We demonstrate that IDA is taken up over 10 times more rapidly by cancer cells than DXR in vitro. Similar trend is observed in in vivo online imaging and less drug redistribution is shown for IDA, together resulting in 4-times higher whole tumor drug uptake for IDA vs. DXR. Together his yielded an improved intratumoral drug distribution for IDA-SDDS, translating into superior tumor response compared to DXR-SDDS treatment at the same dose. Thus, IDA - a drug that is not used for treatment of solid cancers - shows superior therapeutic index and better outcome when administered in externally triggered SDDS. Conclusions: We show that a shift in selection of chemotherapeutics is urgently needed, away from the classic drugs towards selection based on properties of a chemotherapeutic in context of the nanoparticle and delivery mode, to maximize the therapeutic efficacy.
Collapse
|
28
|
Hossann M, Hirschberger J, Schmidt R, Baumgartner C, Zimmermann K, Baer S, Ratzlaff C, Peller M, Troedson K, Limmer S, Brühschwein A, Dörfelt R, Kreutzmann N, Wess G, Knösel T, Schagon O, Fischer J, Grüll H, Willerding L, Schmidt M, Meyer-Lindenberg A, Issels RD, Schwaiger M, Eggermont AM, ten Hagen TL, Lindner LH. A Heat‐Activated Drug‐Delivery Platform Based on Phosphatidyl‐(oligo)‐glycerol Nanocarrier for Effective Cancer Treatment. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202000089] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Martin Hossann
- Department of Medicine III & Sarcoma Center (SarKUM) University Hospital LMU Munich Marchioninistraße 15 81377 Munich Germany
- Thermosome GmbH 82152 Planegg/Martinsried Germany
| | | | - Rebecca Schmidt
- Department of Medicine III & Sarcoma Center (SarKUM) University Hospital LMU Munich Marchioninistraße 15 81377 Munich Germany
| | - Christine Baumgartner
- Department of Nuclear Medicine Klinikum Rechts der Isar Ismaninger Straße 22 81675 Munich Germany
| | - Katja Zimmermann
- Clinic of Small Animal Medicine LMU Munich Veterinärstr. 13 80539 Munich Germany
| | - Silke Baer
- Clinic of Small Animal Medicine LMU Munich Veterinärstr. 13 80539 Munich Germany
| | - Christina Ratzlaff
- Clinic of Small Animal Medicine LMU Munich Veterinärstr. 13 80539 Munich Germany
| | - Michael Peller
- Department of Radiology University Hospital LMU Munich Marchioninistr. 15 81377 Munich Germany
| | - Karin Troedson
- Clinic of Small Animal Medicine LMU Munich Veterinärstr. 13 80539 Munich Germany
| | - Simone Limmer
- Department of Medicine III & Sarcoma Center (SarKUM) University Hospital LMU Munich Marchioninistraße 15 81377 Munich Germany
| | - Andreas Brühschwein
- Clinic of Small Animal Surgery and Reproduction LMU Munich Veterinärstr. 13 80539 Munich Germany
| | - Rene Dörfelt
- Clinic of Small Animal Medicine LMU Munich Veterinärstr. 13 80539 Munich Germany
| | - Nina Kreutzmann
- Clinic of Small Animal Medicine LMU Munich Veterinärstr. 13 80539 Munich Germany
| | - Gerhard Wess
- Clinic of Small Animal Medicine LMU Munich Veterinärstr. 13 80539 Munich Germany
| | - Thomas Knösel
- Institute of Pathology LMU Munich Thalkirchner Str. 36 80337 Munich Germany
| | - Olaf Schagon
- Phospholipid Research Group Max Planck Institute for Biophysical Chemistry Am Faßberg 11 37073 Göttingen Germany
| | - Johannes Fischer
- Department of Nuclear Medicine Klinikum Rechts der Isar Ismaninger Straße 22 81675 Munich Germany
| | - Holger Grüll
- University of Cologne Faculty of Medicine University Hospital of Cologne Institute of Diagnostic and Interventional Radiology Kerpener Str. 62 50937 Cologne Germany
| | - Linus Willerding
- Department of Medicine III & Sarcoma Center (SarKUM) University Hospital LMU Munich Marchioninistraße 15 81377 Munich Germany
| | - Michael Schmidt
- Munich Cancer Registry Institute for Medical Information Processing, Biometry, and Epidemiology University of Munich Marchioninistr. 15 81377 Munich Germany
| | - Andrea Meyer-Lindenberg
- Clinic of Small Animal Surgery and Reproduction LMU Munich Veterinärstr. 13 80539 Munich Germany
| | - Rolf D. Issels
- Department of Medicine III & Sarcoma Center (SarKUM) University Hospital LMU Munich Marchioninistraße 15 81377 Munich Germany
| | - Markus Schwaiger
- Department of Nuclear Medicine Klinikum Rechts der Isar Ismaninger Straße 22 81675 Munich Germany
| | - Alexander M. Eggermont
- Princess Máxima Center for Pediatric Oncology University Medical Center Utrecht Heidelberglaan 25 3584 CS Utrecht The Netherlands
| | - Timo L. ten Hagen
- Department of Pathology Laboratory Experimental Oncology and Nanomedicine Innovation Center Erasmus (NICE) Erasmus MC 3015 CE Rotterdam The Netherlands
| | - Lars H. Lindner
- Department of Medicine III & Sarcoma Center (SarKUM) University Hospital LMU Munich Marchioninistraße 15 81377 Munich Germany
| |
Collapse
|
29
|
Franco MS, Gomes ER, Roque MC, Oliveira MC. Triggered Drug Release From Liposomes: Exploiting the Outer and Inner Tumor Environment. Front Oncol 2021; 11:623760. [PMID: 33796461 PMCID: PMC8008067 DOI: 10.3389/fonc.2021.623760] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/02/2021] [Indexed: 12/12/2022] Open
Abstract
Since more than 40 years liposomes have being extensively studied for their potential as carriers of anticancer drugs. The basic principle behind their use for cancer treatment consists on the idea that they can take advantage of the leaky vasculature and poor lymphatic drainage present at the tumor tissue, passively accumulating in this region. Aiming to further improve their efficacy, different strategies have been employed such as PEGlation, which enables longer circulation times, or the attachment of ligands to liposomal surface for active targeting of cancer cells. A great challenge for drug delivery to cancer treatment now, is the possibility to trigger release from nanosystems at the tumor site, providing efficacious levels of drug in the tumor. Different strategies have been proposed to exploit the outer and inner tumor environment for triggering drug release from liposomes and are the focus of this review.
Collapse
Affiliation(s)
- Marina Santiago Franco
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Eliza Rocha Gomes
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marjorie Coimbra Roque
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mônica Cristina Oliveira
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
30
|
Moholkar DN, Sadalage PS, Havaldar DV, Pawar KD. Engineering the liposomal formulations from natural peanut phospholipids for pH and temperature sensitive release of folic acid, levodopa and camptothecin. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 123:111979. [PMID: 33812607 DOI: 10.1016/j.msec.2021.111979] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 01/12/2021] [Accepted: 02/13/2021] [Indexed: 11/29/2022]
Abstract
The present study demonstrates the extraction and identification of phospholipids (PLs) from peanut seed for formulation of liposomes for pH and thermo-sensitive delivery and release of folic acid (FA), levodopa (DOPA) and, camptothecin (CPT). The TLC, FTIR and GC-MS based characterization of extracted peanut PLs showed phosphatidylethanolamine, cardiolipin and phosphatidic acid as major PLs and palmitic acid and oleic acid as major fatty acids. Liposomes (LSMs) of size 1-2 μm formulated by optimized thin-film hydration method were found to entrap FA, DOPA and CPT with 58, 61.4 and 52.12% efficiency, respectively with good stability. The effect of external stimuli like pH and temperature on the release pattern of FA, DOPA and CPT indicated that FA was optimally released at pH 10 and 57 °C, DOPA at pH 2 and 37 °C, while CPT was best released at pH 6 and 47 °C. When tested for the in vitro activity, DOPA released by DOPA@LSMs showed lower toxicity to 3T3 than to SH-SY5Y cells. Similarly, CPT released by CPT@LSMs showed remarkable anticancer activity against MCF-7 cells with an IC50 value of 17.99 μg/mL. Thus peanut PLs can be efficiently used for liposomal formulations for pH and thermo-sensitive release of drugs.
Collapse
Affiliation(s)
- Disha N Moholkar
- School of Nanoscience and Biotechnology, Shivaji University, Kolhapur, Maharashtra, India
| | | | - Darshana V Havaldar
- School of Nanoscience and Biotechnology, Shivaji University, Kolhapur, Maharashtra, India
| | - Kiran D Pawar
- School of Nanoscience and Biotechnology, Shivaji University, Kolhapur, Maharashtra, India.
| |
Collapse
|
31
|
Rahim MA, Jan N, Khan S, Shah H, Madni A, Khan A, Jabar A, Khan S, Elhissi A, Hussain Z, Aziz HC, Sohail M, Khan M, Thu HE. Recent Advancements in Stimuli Responsive Drug Delivery Platforms for Active and Passive Cancer Targeting. Cancers (Basel) 2021; 13:670. [PMID: 33562376 PMCID: PMC7914759 DOI: 10.3390/cancers13040670] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 12/13/2022] Open
Abstract
The tumor-specific targeting of chemotherapeutic agents for specific necrosis of cancer cells without affecting the normal cells poses a great challenge for researchers and scientists. Though extensive research has been carried out to investigate chemotherapy-based targeted drug delivery, the identification of the most promising strategy capable of bypassing non-specific cytotoxicity is still a major concern. Recent advancements in the arena of onco-targeted therapies have enabled safe and effective tumor-specific localization through stimuli-responsive drug delivery systems. Owing to their promising characteristic features, stimuli-responsive drug delivery platforms have revolutionized the chemotherapy-based treatments with added benefits of enhanced bioavailability and selective cytotoxicity of cancer cells compared to the conventional modalities. The insensitivity of stimuli-responsive drug delivery platforms when exposed to normal cells prevents the release of cytotoxic drugs into the normal cells and therefore alleviates the off-target events associated with chemotherapy. Contrastingly, they showed amplified sensitivity and triggered release of chemotherapeutic payload when internalized into the tumor microenvironment causing maximum cytotoxic responses and the induction of cancer cell necrosis. This review focuses on the physical stimuli-responsive drug delivery systems and chemical stimuli-responsive drug delivery systems for triggered cancer chemotherapy through active and/or passive targeting. Moreover, the review also provided a brief insight into the molecular dynamic simulations associated with stimuli-based tumor targeting.
Collapse
Affiliation(s)
- Muhammad Abdur Rahim
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Punjab, Pakistan; (M.A.R.); (N.J.); (S.K.); (H.S.); (A.K.)
| | - Nasrullah Jan
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Punjab, Pakistan; (M.A.R.); (N.J.); (S.K.); (H.S.); (A.K.)
| | - Safiullah Khan
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Punjab, Pakistan; (M.A.R.); (N.J.); (S.K.); (H.S.); (A.K.)
| | - Hassan Shah
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Punjab, Pakistan; (M.A.R.); (N.J.); (S.K.); (H.S.); (A.K.)
| | - Asadullah Madni
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Punjab, Pakistan; (M.A.R.); (N.J.); (S.K.); (H.S.); (A.K.)
| | - Arshad Khan
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Punjab, Pakistan; (M.A.R.); (N.J.); (S.K.); (H.S.); (A.K.)
| | - Abdul Jabar
- College of Pharmacy, University of Sargodha, Sargodha 40100, Punjab, Pakistan;
| | - Shahzeb Khan
- Department of Pharmacy, University of Malakand, Chakdara, Dir Lower 18800, Khyber Pakhtunkhwa, Pakistan;
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Westville 3631, Durban 4000, South Africa
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Abdelbary Elhissi
- College of Pharmacy, QU Health and Office of VP for Research and Graduate Studies, Qatar University, P.O. Box 2713, Doha, Qatar;
| | - Zahid Hussain
- Department of Pharmaceutics & Pharmaceutical Technology, College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates;
- Research Institute for Medical and Health Sciences (SIMHR), University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Heather C Aziz
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA;
| | - Muhammad Sohail
- Department of Pharmacy, COMSATS University Abbottabad Campus, Abbottabad 45550, Khyber Pakhtunkhwa, Pakistan;
| | - Mirazam Khan
- Department of Pharmacy, University of Malakand, Chakdara, Dir Lower 18800, Khyber Pakhtunkhwa, Pakistan;
| | - Hnin Ei Thu
- Research and Innovation Department, Lincolon University College, Petaling Jaya 47301, Selangor, Malaysia;
- Innoscience Research Institute, Skypark, Subang Jaya 47650, Selangor, Malaysia
| |
Collapse
|
32
|
Moosavian SA, Bianconi V, Pirro M, Sahebkar A. Challenges and pitfalls in the development of liposomal delivery systems for cancer therapy. Semin Cancer Biol 2021; 69:337-348. [PMID: 31585213 DOI: 10.1016/j.semcancer.2019.09.025] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/03/2019] [Accepted: 09/28/2019] [Indexed: 12/21/2022]
Abstract
Despite considerable advances in the application of liposomal drug delivery systems in cancer treatment, the clinical application of liposomal formulations has been limited by many factors. It seems that there is a wide gap between results of experimental studies and clinical application of liposomes. In this review, we discuss barriers which limit the translation of liposomal delivery systems in cancer therapy. The main focus of this review will be on differences between preclinical and clinical studies and potential approaches to overcome the main pitfalls in the clinical application of liposomal delivery systems.
Collapse
Affiliation(s)
- Seyedeh Alia Moosavian
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vanessa Bianconi
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Italy
| | - Matteo Pirro
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Italy
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
33
|
Majumder J, Minko T. Targeted Nanotherapeutics for Respiratory Diseases: Cancer, Fibrosis, and Coronavirus. ADVANCED THERAPEUTICS 2021; 4:2000203. [PMID: 33173809 PMCID: PMC7646027 DOI: 10.1002/adtp.202000203] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/27/2020] [Indexed: 12/13/2022]
Abstract
Systemic delivery of therapeutics for treatment of lung diseases has several limitations including poor organ distribution of delivered payload with relatively low accumulation of active substances in the lungs and severe adverse side effects. In contrast, nanocarrier based therapeutics provide a broad range of opportunities due to their ability to encapsulate substances with different aqueous solubility, transport distinct types of cargo, target therapeutics specifically to the deceased organ, cell, or cellular organelle limiting adverse side effects and increasing the efficacy of therapy. Moreover, many nanotherapeutics can be delivered by inhalation locally to the lungs avoiding systemic circulation. In addition, nanoscale based delivery systems can be multifunctional, simultaneously carrying out several tasks including diagnostics, treatment and suppression of cellular resistance to the treatment. Nanoscale delivery systems improve the clinical efficacy of conventional therapeutics allowing new approaches for the treatment of respiratory diseases which are difficult to treat or possess intrinsic or acquired resistance to treatment. The present review summarizes recent advances in the development of nanocarrier based therapeutics for local and targeted delivery of drugs, nucleic acids and imaging agents for diagnostics and treatment of various diseases such as cancer, cystic fibrosis, and coronavirus.
Collapse
Affiliation(s)
- Joydeb Majumder
- Department of PharmaceuticsErnest Mario School of Pharmacy, RutgersThe State University of New JerseyPiscatawayNJ08854USA
| | - Tamara Minko
- Department of PharmaceuticsErnest Mario School of Pharmacy, RutgersThe State University of New JerseyPiscatawayNJ08854USA
| |
Collapse
|
34
|
van Valenberg FJP, Brummelhuis ISG, Lindner LH, Kuhnle F, Wedmann B, Schweizer P, Hossann M, Witjes JA, Oosterwijk E. DPPG 2-Based Thermosensitive Liposomes with Encapsulated Doxorubicin Combined with Hyperthermia Lead to Higher Doxorubicin Concentrations in the Bladder Compared to Conventional Application in Pigs: A Rationale for the Treatment of Muscle-Invasive Bladder Cancer. Int J Nanomedicine 2021; 16:75-88. [PMID: 33447028 PMCID: PMC7802347 DOI: 10.2147/ijn.s280034] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/10/2020] [Indexed: 12/27/2022] Open
Abstract
Purpose Current treatment options for muscle-invasive bladder cancer (MIBC) are associated with substantial morbidity. Local release of doxorubicin (DOX) from phosphatidyldiglycerol-based thermosensitive liposomes (DPPG2-TSL-DOX) potentiated by hyperthermia (HT) in the bladder wall may result in bladder sparing without toxicity of systemic chemotherapy. We investigated whether this approach, compared to conventional DOX application, increases DOX concentrations in the bladder wall while limiting DOX in essential organs. Materials and Methods Twenty-one pigs were anaesthetized, and a urinary catheter equipped with a radiofrequency-emitting antenna for HT (60 minutes) was placed. Experimental groups consisted of iv low or full dose (20 or 60 mg/m2) DPPG2-TSL-DOX with/without HT, iv low dose (20 mg/m2) free DOX with HT, and full dose (50 mg/50 mL) intravesical DOX with/without HT. After the procedure, animals were immediately sacrificed. HPLC was used to measure DOX levels in the bladder, essential organs and serum, and fluorescence microscopy to evaluate DOX distribution in the bladder wall. Results Iv DPPG2-TSL-DOX with HT resulted in a significantly higher bladder wall DOX concentration which was more homogeneous distributed, than iv and intravesical free DOX administration with HT. Specifically in the detrusor, DPPG2-TSL-DOX with HT led to a >7- and 44-fold higher DOX concentration, compared to iv free DOX with HT and intravesical DOX, respectively. Organ DOX concentrations were significantly lower in heart and kidneys, and similar in liver, spleen and lungs, following iv DPPG2-TSL-DOX with HT, compared to iv free DOX. Intravesical DOX led to the lowest organ DOX concentrations. Conclusion Iv DPPG2-TSL-DOX combined with HT achieved higher DOX concentrations in the bladder wall including the detrusor, compared to conventional iv and intravesical DOX application. In combination with lower DOX accumulation in heart and kidneys, compared to iv free chemotherapy, DPPG2-TSL-DOX with HT has great potential to attain a role as a bladder-sparing treatment for MIBC.
Collapse
Affiliation(s)
| | - Iris S G Brummelhuis
- Department of Urology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Lars H Lindner
- Department of Medicine III, University Hospital LMU Munich, Munich, Germany
| | - Felix Kuhnle
- Department of Medicine III, University Hospital LMU Munich, Munich, Germany
| | - Barbara Wedmann
- Department of Medicine III, University Hospital LMU Munich, Munich, Germany
| | | | | | - J Alfred Witjes
- Department of Urology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Egbert Oosterwijk
- Department of Urology, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
35
|
Amin M, Huang W, Seynhaeve ALB, ten Hagen TLM. Hyperthermia and Temperature-Sensitive Nanomaterials for Spatiotemporal Drug Delivery to Solid Tumors. Pharmaceutics 2020; 12:E1007. [PMID: 33105816 PMCID: PMC7690578 DOI: 10.3390/pharmaceutics12111007] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/18/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023] Open
Abstract
Nanotechnology has great capability in formulation, reduction of side effects, and enhancing pharmacokinetics of chemotherapeutics by designing stable or long circulating nano-carriers. However, effective drug delivery at the cellular level by means of such carriers is still unsatisfactory. One promising approach is using spatiotemporal drug release by means of nanoparticles with the capacity for content release triggered by internal or external stimuli. Among different stimuli, interests for application of external heat, hyperthermia, is growing. Advanced technology, ease of application and most importantly high level of control over applied heat, and as a result triggered release, and the adjuvant effect of hyperthermia in enhancing therapeutic response of chemotherapeutics, i.e., thermochemotherapy, make hyperthermia a great stimulus for triggered drug release. Therefore, a variety of temperature sensitive nano-carriers, lipid or/and polymeric based, have been fabricated and studied. Importantly, in order to achieve an efficient therapeutic outcome, and taking the advantages of thermochemotherapy into consideration, release characteristics from nano-carriers should fit with applicable clinical thermal setting. Here we introduce and discuss the application of the three most studied temperature sensitive nanoparticles with emphasis on release behavior and its importance regarding applicability and therapeutic potentials.
Collapse
Affiliation(s)
- Mohamadreza Amin
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands; (M.A.); (W.H.); (A.L.B.S.)
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| | - Wenqiu Huang
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands; (M.A.); (W.H.); (A.L.B.S.)
| | - Ann L. B. Seynhaeve
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands; (M.A.); (W.H.); (A.L.B.S.)
| | - Timo L. M. ten Hagen
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands; (M.A.); (W.H.); (A.L.B.S.)
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| |
Collapse
|
36
|
Gleue L, Schupp J, Zimmer N, Becker E, Frey H, Tuettenberg A, Helm M. Stability of Alkyl Chain-Mediated Lipid Anchoring in Liposomal Membranes. Cells 2020; 9:E2213. [PMID: 33003620 PMCID: PMC7599733 DOI: 10.3390/cells9102213] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/21/2020] [Accepted: 09/24/2020] [Indexed: 12/31/2022] Open
Abstract
Lipid exchange among biological membranes, lipoprotein particles, micelles, and liposomes is an important yet underrated phenomenon with repercussions throughout the life sciences. The premature loss of lipid molecules from liposomal formulations severely impacts therapeutic applications of the latter and thus limits the type of lipids and lipid conjugates available for fine-tuning liposomal properties. While cholesterol derivatives, with their irregular lipophilic surface shape, are known to readily undergo lipid exchange and interconvert, e.g., with serum, the situation is unclear for lipids with regular, linear-shaped alkyl chains. This study compares the propensity of fluorescence-labeled lipid conjugates of systematically varied lengths to migrate from liposomal particles consisting mainly of egg phosphatidyl choline 3 (EPC3) and cholesterol into biomembranes. We show that dialkyl glyceryl lipids with chains of 18-20 methylene units are inherently stable in liposomal membranes. In contrast, C16 lipids show some lipid exchange, albeit significantly less than comparable cholesterol conjugates. Remarkably, the C18 chain length, which confers noticeable anchor stability, corresponds to the typical chain length in biological membranes.
Collapse
Affiliation(s)
- Lukas Gleue
- Institute of Pharmaceutical and Biomedical Science, Johannes Gutenberg-University Mainz, 55128 Mainz, Germany;
| | - Jonathan Schupp
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (J.S.); (N.Z.)
| | - Niklas Zimmer
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (J.S.); (N.Z.)
| | - Eyleen Becker
- Department of Chemistry, Johannes Gutenberg-University Mainz, 55128 Mainz, Germany; (E.B.); (H.F.)
| | - Holger Frey
- Department of Chemistry, Johannes Gutenberg-University Mainz, 55128 Mainz, Germany; (E.B.); (H.F.)
| | - Andrea Tuettenberg
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (J.S.); (N.Z.)
| | - Mark Helm
- Institute of Pharmaceutical and Biomedical Science, Johannes Gutenberg-University Mainz, 55128 Mainz, Germany;
| |
Collapse
|
37
|
Skóra B, Szychowski KA, Gmiński J. A concise review of metallic nanoparticles encapsulation methods and their potential use in anticancer therapy and medicine. Eur J Pharm Biopharm 2020; 154:153-165. [PMID: 32681962 DOI: 10.1016/j.ejpb.2020.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/29/2020] [Accepted: 07/02/2020] [Indexed: 02/08/2023]
Abstract
Interest in the use of metallic nanoparticles (NPs) in medicine is constantly increasing. The key challenge to the introduction of NPs into anticancer treatment is to limit the contact of their surface with healthy cells and to enable specific targeting of certain tissues, for example, cancerous cells. These aspects have raised a question whether the recent methods of drug delivery allow restricting the contact of NPs with healthy and/or nontarget cells. NPs can be restricted by encapsulation, which involves entrapping them into organic layers. This review is the first to present the different approaches for the encapsulation of metallic NPs, using liposomes, dendrimers, and proteins. The types and methods of entrapping are shown in an accessible way, enriched with graphics, and the pros and cons of these methods are disputable. Furthermore, the potential uses of NP complexes in medicine are described.
Collapse
Affiliation(s)
- Bartosz Skóra
- Department of Lifestyle Disorders and Regenerative Medicine, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland.
| | - Konrad A Szychowski
- Department of Lifestyle Disorders and Regenerative Medicine, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland
| | - Jan Gmiński
- Department of Lifestyle Disorders and Regenerative Medicine, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland
| |
Collapse
|
38
|
Lu T, Ten Hagen TLM. A novel kinetic model to describe the ultra-fast triggered release of thermosensitive liposomal drug delivery systems. J Control Release 2020; 324:669-678. [PMID: 32512013 DOI: 10.1016/j.jconrel.2020.05.047] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 05/21/2020] [Accepted: 05/31/2020] [Indexed: 12/20/2022]
Abstract
Thermosensitive liposomes, as one of the stimuli-responsive drug delivery systems, receive growing attention, due to their ability to generate rapid and massive drug release in the heated area, and marginal release of contents in non-heated parts of the body. This typical triggered release behavior cannot be fitted adequately by most of the current mathematical kinetic models. The aim of this study was to establish the proper kinetic equation to describe the rapid release of drugs from trigger-sensitive drug delivery systems. We summarized all commonly used kinetic models mentioned in the literature and fitted the release data with these models, finding that only the Korsmeyer-Peppas and the Weibull models show acceptable fitting results. To better describe the release from thermosensitive liposomes with a size below 100 nm, we took Laplace pressure as a release-driving force and proposed a new equation that demonstrates improved fitting in liposomes ranging down to a size of 70 nm. Our new kinetic model shows desirable fitting, not only at the optimal temperature but also of releases within the whole release-temperature range, providing a useful kinetic model to describe release profiles of smaller nano-sized stimuli-responsive drug delivery systems.
Collapse
Affiliation(s)
- Tao Lu
- Laboratory Experimental Oncology, Department of Pathology, Erasmus MC, 3015GD Rotterdam, the Netherlands.
| | - Timo L M Ten Hagen
- Laboratory Experimental Oncology, Department of Pathology, Erasmus MC, 3015GD Rotterdam, the Netherlands.
| |
Collapse
|
39
|
Saha S, Banskota S, Roberts S, Kirmani N, Chilkoti A. Engineering the Architecture of Elastin-Like Polypeptides: From Unimers to Hierarchical Self-Assembly. ADVANCED THERAPEUTICS 2020; 3:1900164. [PMID: 34307837 PMCID: PMC8297442 DOI: 10.1002/adtp.201900164] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Indexed: 12/12/2022]
Abstract
Well-defined tunable nanostructures formed through the hierarchical self-assembly of peptide building blocks have drawn significant attention due to their potential applications in biomedical science. Artificial protein polymers derived from elastin-like polypeptides (ELPs), which are based on the repeating sequence of tropoelastin (the water-soluble precursor to elastin), provide a promising platform for creating nanostructures due to their biocompatibility, ease of synthesis, and customizable architecture. By designing the sequence and composition of ELPs at the gene level, their physicochemical properties can be controlled to a degree that is unmatched by synthetic polymers. A variety of ELP-based nanostructures are designed, inspired by the self-assembly of elastin and other proteins in biological systems. The choice of building blocks determines not only the physical properties of the nanostructures, but also their self-assembly into architectures ranging from spherical micelles to elongated nanofibers. This review focuses on the molecular determinants of ELP and ELP-hybrid self-assembly and formation of spherical, rod-like, worm-like, fibrillar, and vesicle architectures. A brief discussion of the potential biomedical applications of these supramolecular assemblies is also included.
Collapse
Affiliation(s)
- Soumen Saha
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Samagya Banskota
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Stefan Roberts
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Nadia Kirmani
- Department of Biology, Trinity College of Arts and Sciences, Duke University, Durham, NC 27708, USA
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| |
Collapse
|
40
|
Seynhaeve A, Amin M, Haemmerich D, van Rhoon G, ten Hagen T. Hyperthermia and smart drug delivery systems for solid tumor therapy. Adv Drug Deliv Rev 2020; 163-164:125-144. [PMID: 32092379 DOI: 10.1016/j.addr.2020.02.004] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/13/2020] [Accepted: 02/19/2020] [Indexed: 12/31/2022]
Abstract
Chemotherapy is a cornerstone of cancer therapy. Irrespective of the administered drug, it is crucial that adequate drug amounts reach all cancer cells. To achieve this, drugs first need to be absorbed, then enter the blood circulation, diffuse into the tumor interstitial space and finally reach the tumor cells. Next to chemoresistance, one of the most important factors for effective chemotherapy is adequate tumor drug uptake and penetration. Unfortunately, most chemotherapeutic agents do not have favorable properties. These compounds are cleared rapidly, distribute throughout all tissues in the body, with only low tumor drug uptake that is heterogeneously distributed within the tumor. Moreover, the typical microenvironment of solid cancers provides additional hurdles for drug delivery, such as heterogeneous vascular density and perfusion, high interstitial fluid pressure, and abundant stroma. The hope was that nanotechnology will solve most, if not all, of these drug delivery barriers. However, in spite of advances and decades of nanoparticle development, results are unsatisfactory. One promising recent development are nanoparticles which can be steered, and release content triggered by internal or external signals. Here we discuss these so-called smart drug delivery systems in cancer therapy with emphasis on mild hyperthermia as a trigger signal for drug delivery.
Collapse
|
41
|
Motamarry A, Negussie AH, Rossmann C, Small J, Wolfe AM, Wood BJ, Haemmerich D. Real-time fluorescence imaging for visualization and drug uptake prediction during drug delivery by thermosensitive liposomes. Int J Hyperthermia 2020; 36:817-826. [PMID: 31451077 DOI: 10.1080/02656736.2019.1642521] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Objective: Thermosensitive liposomal doxorubicin (TSL-Dox) is a promising stimuli-responsive nanoparticle drug delivery system that rapidly releases the contained drug in response to hyperthermia (HT) (>40 °C). Combined with localized heating, TSL-Dox allows highly localized delivery. The goals of this study were to demonstrate that real-time fluorescence imaging can visualize drug uptake during delivery, and can predict tumor drug uptake. Methods: Nude mice carrying subcutaneous tumors (Lewis lung carcinoma) were anesthetized and injected with TSL-Dox (5 mg/kg dose). Localized HT was induced by heating tumors for 15, 30 or 60 min via a custom-designed HT probe placed superficially at the tumor location. In vivo fluorescence imaging (excitation 523 nm, emission 610 nm) was performed before, during, and for 5 min following HT. After imaging, tumors were extracted, drug uptake was quantified by high-performance liquid chromatography, and correlated with in vivo fluorescence. Plasma samples were obtained before and after HT to measure TSL-Dox pharmacokinetics. Results: Local drug uptake could be visualized in real-time during HT. Compared to unheated control tumors, fluorescence of heated tumors increased by 4.6-fold (15 min HT), 9.3-fold (30 min HT), and 13.2-fold (60 min HT). HT duration predicted tumor drug uptake (p = .02), with tumor drug concentrations of 4.2 ± 1.3 µg/g (no HT), 7.1 ± 5.9 µg/g (15 min HT), 14.1 ± 6.7 µg/g (30 min HT) and 21.4 ± 12.6 µg/g (60 min HT). There was good correlation (R2 = 0.67) between fluorescence of the tumor region and tumor drug uptake. Conclusions: Real-time in vivo fluorescence imaging can visualize drug uptake during delivery, and can predict tumor drug uptake.
Collapse
Affiliation(s)
- Anjan Motamarry
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina , Charleston , SC , USA.,Department of Pediatrics, Medical University of South Carolina , Charleston , SC , USA
| | - Ayele H Negussie
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health , Bethesda , MD , USA
| | - Christian Rossmann
- Department of Pediatrics, Medical University of South Carolina , Charleston , SC , USA
| | - James Small
- Department of Public Health Sciences, Medical University of South Carolina , Charleston , SC , USA
| | - A Marissa Wolfe
- Department of Comparative Medicine, Medical University of South Carolina , Charleston , SC , USA
| | - Bradford J Wood
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health , Bethesda , MD , USA
| | - Dieter Haemmerich
- Department of Pediatrics, Medical University of South Carolina , Charleston , SC , USA.,Department of Bioengineering, Clemson University , Clemson , SC , USA
| |
Collapse
|
42
|
Chen Z, Tu Y, Zhang D, Liu C, Zhou Y, Li X, Wu X, Liu R. A thermosensitive nanoplatform for photoacoustic imaging and NIR light triggered chemo-photothermal therapy. Biomater Sci 2020; 8:4299-4307. [DOI: 10.1039/d0bm00810a] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A thermosensitive nanoplatform CDTSL achieves NIR light controlled drug release and can be applied for photoacoustic imaging and chemo-photothermal therapy.
Collapse
Affiliation(s)
- Zikang Chen
- Guangdong Provincial Key Laboratory of Medical Image Processing
- School of Biomedical Engineering
- Southern Medical University
- Guangzhou
- P.R. China
| | - Yinuo Tu
- Department of Thoracic Surgery
- Huiqiao Medical Center
- Nanfang Hospital
- Southern Medical University
- Guangzhou
| | - Di Zhang
- Guangdong Provincial Key Laboratory of Medical Image Processing
- School of Biomedical Engineering
- Southern Medical University
- Guangzhou
- P.R. China
| | - Chuang Liu
- Department of Thoracic Surgery
- Huiqiao Medical Center
- Nanfang Hospital
- Southern Medical University
- Guangzhou
| | - Yuping Zhou
- Guangdong Provincial Key Laboratory of Medical Image Processing
- School of Biomedical Engineering
- Southern Medical University
- Guangzhou
- P.R. China
| | - Xiang Li
- Department of Thoracic Surgery
- Huiqiao Medical Center
- Nanfang Hospital
- Southern Medical University
- Guangzhou
| | - Xu Wu
- Department of Thoracic Surgery
- Huiqiao Medical Center
- Nanfang Hospital
- Southern Medical University
- Guangzhou
| | - Ruiyuan Liu
- Guangdong Provincial Key Laboratory of Medical Image Processing
- School of Biomedical Engineering
- Southern Medical University
- Guangzhou
- P.R. China
| |
Collapse
|
43
|
Combined Modality Therapy Based on Hybrid Gold Nanostars Coated with Temperature Sensitive Liposomes to Overcome Paclitaxel-Resistance in Hepatic Carcinoma. Pharmaceutics 2019; 11:pharmaceutics11120683. [PMID: 31847496 PMCID: PMC6969923 DOI: 10.3390/pharmaceutics11120683] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/07/2019] [Accepted: 12/13/2019] [Indexed: 12/11/2022] Open
Abstract
In this study, we prepared gold nanostar (GNS) composite nanoparticles containing siRNA of cyclooxygenase-2(siCOX-2) that were modified by tumor targeting ligand 2-deoxyglucose (DG) and transmembrane peptide 9-poly-D-arginine (9R) to form siCOX-2(9R/DG-GNS). Paclitaxel loaded temperature sensitive liposomes (PTX-TSL) were surface-modified to produce PTX-TSL-siCOX-2(9R/DG-GNS) displaying homogeneous star-shaped structures of suitable size (293.93 nm ± 3.21) and zeta potentials (2.47 mV ± 0.22). PTX-TSL-siCOX-2(9R/DG-GNS) had a high thermal conversion efficiency under 808 nm laser radiation and a superior transfection efficiency, which may be related to the targeting effects of DG and increased heat induced membrane permeability. COX-2 expression in HepG2/PTX cells was significantly suppressed by PTX-TSL-siCOX-2(9R/DG-GNS) in high temperatures. The co-delivery system inhibited drug-resistant cell growth rates by ≥77% and increased the cell apoptosis rate about 47% at elevated temperatures. PTX-TSL and siCOX-2 loaded gold nanostar particles, therefore, show promise for overcoming tumor resistance.
Collapse
|
44
|
Abri Aghdam M, Bagheri R, Mosafer J, Baradaran B, Hashemzaei M, Baghbanzadeh A, de la Guardia M, Mokhtarzadeh A. Recent advances on thermosensitive and pH-sensitive liposomes employed in controlled release. J Control Release 2019; 315:1-22. [DOI: 10.1016/j.jconrel.2019.09.018] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 12/12/2022]
|
45
|
Abstract
Liposomes have been employed as cancer therapy clinically since the 1990s, with the primary benefit of reduced toxicity but no appreciable efficacy improvement. Thermosensitive liposomes (TSLs) are specifically formulated such that they release the encapsulated drug when exposed to hyperthermic temperatures in the fever range (~40-42°C) and have been investigated as cancer therapy for several decades, with first clinical trials initiated in the last decade. Combined with localized hyperthermia, TSLs allow precise drug delivery to a targeted region. Typically, the targeted tissue is exposed to localized hyperthermia facilitated by an image-guided hyperthermia device. Thus, TSLs enable image-guided drug delivery where drug is delivered to a tissue region identified by medical imaging. Recent TSL formulations are based on the more recent paradigm of intravascular triggered release, where drug is released rapidly (within seconds) while TSLs pass through the vasculature of the heated tissue region. The drug released within the blood then extravasates and is taken up by cancer cells. These TSLs enable up to 20-30 times higher tumor drug uptake compared to infusion of unencapsulated drug, and the dose locally delivered to the heated region can be modulated based on heating duration. This chapter reviews various TSL formulations, the different anticancer agents that have been encapsulated, as well as targeted cancer types. Further, the various hyperthermia devices that have been used for image-guided hyperthermia are reviewed, focusing on those that have been employed in human patients.
Collapse
Affiliation(s)
- Dieter Haemmerich
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States.
| | - Anjan Motamarry
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States; Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
46
|
Dunne M, Epp-Ducharme B, Sofias AM, Regenold M, Dubins DN, Allen C. Heat-activated drug delivery increases tumor accumulation of synergistic chemotherapies. J Control Release 2019; 308:197-208. [PMID: 31195059 DOI: 10.1016/j.jconrel.2019.06.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/31/2019] [Accepted: 06/09/2019] [Indexed: 12/20/2022]
Abstract
Doxorubicin is a clinically important anthracycline chemotherapeutic agent that is used to treat many cancers. Nanomedicine formulations including Doxil® and ThermoDox® have been developed to mitigate doxorubicin cardiotoxicity. Doxil is used clinically to treat ovarian cancer, AIDS-related Kaposi's sarcoma, and multiple myeloma, but there is evidence that therapeutic efficacy is hampered by lack of drug release. ThermoDox is a lipid-based heat-activated formulation of doxorubicin that relies on externally applied energy to increase tissue temperatures and efficiently trigger drug release, thereby affording therapeutic advantages compared to Doxil. However, elevating tissue temperatures is a complex treatment process requiring significant time, cost, and expertise compared to standard intravenous chemotherapy. This work endeavors to develop a companion therapeutic to ThermoDox that also relies on heat-triggered release in order to increase the therapeutic index of doxorubicin. To this end, a thermosensitive liposome formulation of the heat shock protein 90 inhibitor alvespimycin has been developed and characterized. This research demonstrates that both doxorubicin and alvespimycin are potent anti-cancer agents and that heat amplifies their cytotoxic effects. Furthermore, the two drugs are proven to act synergistically when cancer cells are treated with the drugs in combination. The formulation of alvespimycin was rationally designed to exhibit similar pharmacokinetics and drug release kinetics compared to ThermoDox, enabling the two drugs to be delivered to heated tumors at similar efficiencies resulting in control of a particular synergistic ratio of drugs. In vivo measurements demonstrated effective heat-mediated triggering of doxorubicin and alvespimycin release from thermosensitive liposomes within tumor vasculature. This treatment strategy resulted in a ~10-fold increase in drug concentration within tumors compared to free drug administered without tumor heating.
Collapse
Affiliation(s)
- Michael Dunne
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | | | - Alexandros Marios Sofias
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada; Utrecht Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Utrecht University, Utrecht, The Netherlands; Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Maximilian Regenold
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - David N Dubins
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Christine Allen
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
47
|
Zhan W, Gedroyc W, Xu XY. Towards a multiphysics modelling framework for thermosensitive liposomal drug delivery to solid tumour combined with focused ultrasound hyperthermia. BIOPHYSICS REPORTS 2019. [DOI: 10.1007/s41048-019-0083-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
48
|
Nigatu AS, Ashar H, Sethuraman SN, Wardlow R, Maples D, Malayer J, Ranjan A. Elastin-like polypeptide incorporated thermally sensitive liposome improve antibiotic therapy against musculoskeletal bacterial pathogens. Int J Hyperthermia 2019; 34:201-208. [PMID: 29278945 DOI: 10.1080/02656736.2017.1420249] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Musculoskeletal infections caused by bacteria such as Staphylococcus aureus and Pseudomonas aeruginosa in children and adults can lead to adverse outcomes including a need for extensive surgical debridement and limb amputation. To enable targeted antimicrobial release in infected tissues, the objective of this study was to design and investigate novel elastin-like polypeptide (ELP)-based thermally sensitive liposomes in vitro. ELP biopolymers can change their phase behaviour at higher temperatures. We hypothesised that ELP-TSL will improve therapeutic efficacy by releasing antimicrobial payloads locally at higher temperatures (≥39 °C). ELP-TSL library were formulated by varying cholesterol and phospholipid composition by the thin film and extrusion method. A broad-spectrum antimicrobial (Ciprofloxacin or Cipro) was encapsulated inside the liposomes by the ammonium sulphate gradient method. Cipro release from ELP-TSLs was assessed in physiological buffers containing ∼25% serum by fluorescence spectroscopy, and efficacy against Staphylococcus aureus and Pseudomonas aeruginosa was assessed by disc diffusion and planktonic assay. Active loading of Cipro achieved an encapsulation efficiency of 40-70% in the ELP-TSL depending upon composition. ELP-TSL Cipro release was near complete at ≥39 °C; however, the release rates could be delayed by cholesterol. Triggered release of Cipro from ELP-TSL at ∼42 °C induced significant killing of S. aureus and P. aeruginosa compared to 37 °C. Our in vitro data suggest that ELP-TSL may potentially improve bacterial wound therapy in patients.
Collapse
Affiliation(s)
- Adane S Nigatu
- a Center for Veterinary Health Sciences , Oklahoma State University , Stillwater , OK , USA
| | - Harshini Ashar
- a Center for Veterinary Health Sciences , Oklahoma State University , Stillwater , OK , USA
| | | | - Rachel Wardlow
- a Center for Veterinary Health Sciences , Oklahoma State University , Stillwater , OK , USA
| | - Danny Maples
- a Center for Veterinary Health Sciences , Oklahoma State University , Stillwater , OK , USA
| | - Jerry Malayer
- a Center for Veterinary Health Sciences , Oklahoma State University , Stillwater , OK , USA
| | - Ashish Ranjan
- a Center for Veterinary Health Sciences , Oklahoma State University , Stillwater , OK , USA
| |
Collapse
|
49
|
Nardecchia S, Sánchez-Moreno P, Vicente JD, Marchal JA, Boulaiz H. Clinical Trials of Thermosensitive Nanomaterials: An Overview. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E191. [PMID: 30717386 PMCID: PMC6409767 DOI: 10.3390/nano9020191] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/15/2019] [Accepted: 01/30/2019] [Indexed: 01/18/2023]
Abstract
Currently, we are facing increasing demand to develop efficient systems for the detection and treatment of diseases that can realistically improve distinct aspects of healthcare in our society. Sensitive nanomaterials that respond to environmental stimuli can play an important role in this task. In this manuscript, we review the clinical trials carried out to date on thermosensitive nanomaterials, including all those clinical trials in hybrid nanomaterials that respond to other stimuli (e.g., magnetic, infrared radiation, and ultrasound). Specifically, we discuss their use in diagnosis and treatment of different diseases. At present, none of the existing trials focused on diagnosis take advantage of the thermosensitive characteristics of these nanoparticles. Indeed, almost all clinical trials consulted explore the use of Ferumoxytol as a current imaging test enhancer. However, the thermal property is being further exploited in the field of disease treatment, especially for the delivery of antitumor drugs. In this regard, ThermoDox®, based on lysolipid thermally sensitive liposome technology to encapsulate doxorubicin (DOX), is the flagship drug. In this review, we have evidenced the discrepancy existing between the number of published papers in thermosensitive nanomaterials and their clinical use, which could be due to the relative novelty of this area of research; more time is needed to validate it through clinical trials. We have no doubt that in the coming years there will be an explosion of clinical trials related to thermosensitive nanomaterials that will surely help to improve current treatments and, above all, will impact on patients' quality of life and life expectancy.
Collapse
Affiliation(s)
- Stefania Nardecchia
- Department of Applied Physics, Faculty of Sciences, University of Granada, C/Fuentenueva s/n, 18071 Granada, Spain.
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, 18016 Granada, Spain.
| | - Paola Sánchez-Moreno
- Nanobiointeractions & Nanodiagnostics, Istituto Italiano di Tecnologia, Via Morego, 30, 16163 Genova, Italy.
| | - Juan de Vicente
- Department of Applied Physics, Faculty of Sciences, University of Granada, C/Fuentenueva s/n, 18071 Granada, Spain.
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, 18016 Granada, Spain.
| | - Juan A Marchal
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, 18016 Granada, Spain.
- Department of Human Anatomy and Embryology, University of Granada, 18016 Granada, Spain.
- Biopathology and Medicine Regenerative Institute (IBIMER), University of Granada, 18016 Granada, Spain.
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-Universidad de Granada, 18016 Granada, Spain.
| | - Houria Boulaiz
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, 18016 Granada, Spain.
- Department of Human Anatomy and Embryology, University of Granada, 18016 Granada, Spain.
- Biopathology and Medicine Regenerative Institute (IBIMER), University of Granada, 18016 Granada, Spain.
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-Universidad de Granada, 18016 Granada, Spain.
| |
Collapse
|
50
|
Zhou L, Qiu T, Lv F, Liu L, Ying J, Wang S. Self-Assembled Nanomedicines for Anticancer and Antibacterial Applications. Adv Healthc Mater 2018; 7:e1800670. [PMID: 30080319 DOI: 10.1002/adhm.201800670] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/03/2018] [Indexed: 01/28/2023]
Abstract
Self-assembly strategies have been widely applied in the nanomedicine field, which provide a convenient approach for building various structures for delivery carriers. When cooperating with biomolecules, self-assembly systems have significant influence on the cell activity and life process and could be used for regulating nanodrug activity. In this review, self-assembled nanomedicines are introduced, including materials, encapsulation, and releasing strategies, where self-assembly strategies are involved. Furthermore, as a promising and emerging area for nanomedicine, in situ self-assembly of anticancer drugs and supramolecular antibiotic switches is also discussed about how to regulate drug activity. Selective pericellular assembly can block mass transformation of cancer cells inducing cell apoptosis, and the intracellular assembly can either cause cell death or effectively avoid drug elimination from cytosol of cancer cells because of the assembly-induced retention (AIR) effect. Host-guest interactions of drug and competitive molecules offer reversible regulations of antibiotic activity, which can reduce drug-resistance and inhibit the generation of drug-resistant bacteria. Finally, the challenges and development trend in the field are discussed.
Collapse
Affiliation(s)
- Lingyun Zhou
- Beijing National Laboratory for Molecular Sciences; Key Laboratory of Organic Solids; Institute of Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
- College of Chemistry; University of Chinese Academy of Sciences; Beijing 100049 P. R. China
| | - Tian Qiu
- Department of Pathology; National Cancer Center/National Clinical Research Center for; Cancer/Cancer Hospital; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing 100021 P. R. China
| | - Fengting Lv
- Beijing National Laboratory for Molecular Sciences; Key Laboratory of Organic Solids; Institute of Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
| | - Libing Liu
- Beijing National Laboratory for Molecular Sciences; Key Laboratory of Organic Solids; Institute of Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
| | - Jianming Ying
- Department of Pathology; National Cancer Center/National Clinical Research Center for; Cancer/Cancer Hospital; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing 100021 P. R. China
| | - Shu Wang
- Beijing National Laboratory for Molecular Sciences; Key Laboratory of Organic Solids; Institute of Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
- College of Chemistry; University of Chinese Academy of Sciences; Beijing 100049 P. R. China
| |
Collapse
|