1
|
Ubanako P, Mirza S, Ruff P, Penny C. Exosome-mediated delivery of siRNA molecules in cancer therapy: triumphs and challenges. Front Mol Biosci 2024; 11:1447953. [PMID: 39355533 PMCID: PMC11442288 DOI: 10.3389/fmolb.2024.1447953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/03/2024] [Indexed: 10/03/2024] Open
Abstract
The discovery of novel and innovative therapeutic strategies for cancer treatment and management remains a major global challenge. Exosomes are endogenous nanoscale extracellular vesicles that have garnered increasing attention as innovative vehicles for advanced drug delivery and targeted therapy. The attractive physicochemical and biological properties of exosomes, including increased permeability, biocompatibility, extended half-life in circulation, reduced toxicity and immunogenicity, and multiple functionalization strategies, have made them preferred drug delivery vehicles in cancer and other diseases. Small interfering RNAs (siRNAs) are remarkably able to target any known gene: an attribute harnessed to knock down cancer-associated genes as a viable strategy in cancer management. Extensive research on exosome-mediated delivery of siRNAs for targeting diverse types of cancer has yielded promising results for anticancer therapy, with some formulations progressing through clinical trials. This review catalogs recent advances in exosome-mediated siRNA delivery in several types of cancer, including the manifold benefits and minimal drawbacks of such innovative delivery systems. Additionally, we have highlighted the potential of plant-derived exosomes as innovative drug delivery systems for cancer treatment, offering numerous advantages such as biocompatibility, scalability, and reduced toxicity compared to traditional methods. These exosomes, with their unique characteristics and potential for effective siRNA delivery, represent a significant advancement in nanomedicine and cancer therapeutics. Further exploration of their manufacturing processes and biological mechanisms could significantly advance natural medicine and enhance the efficacy of exosome-based therapies.
Collapse
Affiliation(s)
- Philemon Ubanako
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sheefa Mirza
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Paul Ruff
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Clement Penny
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
2
|
Kalra J, Baker J, Sun X, Kyle A, Minchinton A, Bally MB. Accumulation of liposomes in metastatic tumor sites is not necessary for anti-cancer drug efficacy. J Transl Med 2024; 22:621. [PMID: 38961395 PMCID: PMC11223361 DOI: 10.1186/s12967-024-05428-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/21/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND The tumor microenvironment is profoundly heterogeneous particularly when comparing sites of metastases. Establishing the extent of this heterogeneity may provide guidance on how best to design lipid-based drug delivery systems to treat metastatic disease. Building on our previous research, the current study employs a murine model of metastatic cancer to explore the distribution of ~ 100 nm liposomes. METHODS Female NCr nude mice were inoculated with a fluorescently labeled, Her2/neu-positive, trastuzumab-resistant breast cancer cell line, JIMT-1mkate, either in the mammary fat pad to create an orthotopic tumor (OT), or via intracardiac injection (IC) to establish tumors throughout the body. Animals were dosed with fluorescent and radio-labeled liposomes. In vivo and ex vivo fluorescent imaging was used to track liposome distribution over a period of 48 h. Liposome distribution in orthotopic tumors was compared to sites of tumor growth that arose following IC injection. RESULTS A significant amount of inter-vessel heterogeneity for DiR distribution was observed, with most tumor blood vessels showing little to no presence of the DiR-labelled liposomes. Further, there was limited extravascular distribution of DiR liposomes in the perivascular regions around DiR-positive vessels. While all OT tumors contained at least some DiR-positive vessels, many metastases had very little or none. Despite the apparent limited distribution of liposomes within metastases, two liposomal drug formulations, Irinophore C and Doxil, showed similar efficacy for both the OT and IC JIMT-1mkate models. CONCLUSION These findings suggest that liposomal formulations achieve therapeutic benefits through mechanisms that extend beyond the enhanced permeability and retention effect.
Collapse
Affiliation(s)
- Jessica Kalra
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC, Canada.
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada.
| | - Jennifer Baker
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - XuXin Sun
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Alastair Kyle
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Andrew Minchinton
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Marcel B Bally
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
- NanoMedicine Innovation Network, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
3
|
Gaballa SA, Shimizu T, Ando H, Takata H, Emam SE, Ramadan E, Naguib YW, Mady FM, Khaled KA, Ishida T. Treatment-induced and Pre-existing Anti-peg Antibodies: Prevalence, Clinical Implications, and Future Perspectives. J Pharm Sci 2024; 113:555-578. [PMID: 37931786 DOI: 10.1016/j.xphs.2023.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 11/08/2023]
Abstract
Polyethylene glycol (PEG) is a versatile polymer that is used in numerous pharmaceutical applications like the food industry, a wide range of disinfectants, cosmetics, and many commonly used household products. PEGylation is the term used to describe the covalent attachment of PEG molecules to nanocarriers, proteins and peptides, and it is used to prolong the circulation half-life of the PEGylated products. Consequently, PEGylation improves the efficacy of PEGylated therapeutics. However, after four decades of research and more than two decades of clinical applications, an unappealing side of PEGylation has emerged. PEG immunogenicity and antigenicity are remarkable challenges that confound the widespread clinical application of PEGylated therapeutics - even those under clinical trials - as anti-PEG antibodies (Abs) are commonly reported following the systemic administration of PEGylated therapeutics. Furthermore, pre-existing anti-PEG Abs have also been reported in healthy individuals who have never been treated with PEGylated therapeutics. The circulating anti-PEG Abs, both treatment-induced and pre-existing, selectively bind to PEG molecules of the administered PEGylated therapeutics inducing activation of the complement system, which results in remarkable clinical implications with varying severity. These include increased blood clearance of the administered PEGylated therapeutics through what is known as the accelerated blood clearance (ABC) phenomenon and initiation of serious adverse effects through complement activation-related pseudoallergic reactions (CARPA). Therefore, the US FDA industry guidelines have recommended the screening of anti-PEG Abs, in addition to Abs against PEGylated proteins, in the clinical trials of PEGylated protein therapeutics. In addition, strategies revoking the immunogenic response against PEGylated therapeutics without compromising their therapeutic efficacy are important for the further development of advanced PEGylated therapeutics and drug-delivery systems.
Collapse
Affiliation(s)
- Sherif A Gaballa
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Taro Shimizu
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Hidenori Ando
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan; Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Haruka Takata
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan; Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Sherif E Emam
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig, 44519 Egypt
| | - Eslam Ramadan
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Youssef W Naguib
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Fatma M Mady
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Khaled A Khaled
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan; Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University; 1-78-1 Sho-machi, Tokushima 770-8505, Japan.
| |
Collapse
|
4
|
Mutalik C, Nivedita, Sneka C, Krisnawati DI, Yougbaré S, Hsu CC, Kuo TR. Zebrafish Insights into Nanomaterial Toxicity: A Focused Exploration on Metallic, Metal Oxide, Semiconductor, and Mixed-Metal Nanoparticles. Int J Mol Sci 2024; 25:1926. [PMID: 38339204 PMCID: PMC10856345 DOI: 10.3390/ijms25031926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/27/2024] [Accepted: 02/02/2024] [Indexed: 02/12/2024] Open
Abstract
Nanomaterials are widely used in various fields, and ongoing research is focused on developing safe and sustainable nanomaterials. Using zebrafish as a model organism for studying the potentially toxic effects of nanomaterials highlights the importance of developing safe and sustainable nanomaterials. Studies conducted on nanomaterials and their toxicity and potential risks to human and environmental health are vital in biomedical sciences. In the present review, we discuss the potential toxicity of nanomaterials (inorganic and organic) and exposure risks based on size, shape, and concentration. The review further explores various types of nanomaterials and their impacts on zebrafish at different levels, indicating that exposure to nanomaterials can lead to developmental defects, changes in gene expressions, and various toxicities. The review also covers the importance of considering natural organic matter and chorion membranes in standardized nanotoxicity testing. While some nanomaterials are biologically compatible, metal and semiconductor nanomaterials that enter the water environment can increase toxicity to aquatic creatures and can potentially accumulate in the human body. Further investigations are necessary to assess the safety of nanomaterials and their impacts on the environment and human health.
Collapse
Affiliation(s)
- Chinmaya Mutalik
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan;
| | - Nivedita
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan; (N.); (C.S.)
| | - Chandrasekaran Sneka
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan; (N.); (C.S.)
| | - Dyah Ika Krisnawati
- Department of Nursing, Faculty of Nursing and Midwifery, Universitas Nahdlatul Ulama Surabaya, Surabaya 60237, East Java, Indonesia;
| | - Sibidou Yougbaré
- Institut de Recherche en Sciences de La Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), Nanoro BP 218, 11, Burkina Faso;
| | - Chuan-Chih Hsu
- Division of Cardiovascular Surgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 11031, Taiwan
| | - Tsung-Rong Kuo
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan;
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan; (N.); (C.S.)
- Stanford Byers Center for Biodesign, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
5
|
Gaballa SA, Shimizu T, Takata H, Ando H, Ibrahim M, Emam SE, Amorim Matsuo NC, Kim Y, Naguib YW, Mady FM, Khaled KA, Ishida T. Impact of Anti-PEG IgM Induced via the Topical Application of a Cosmetic Product Containing PEG Derivatives on the Antitumor Effects of PEGylated Liposomal Antitumor Drug Formulations in Mice. Mol Pharm 2024; 21:622-632. [PMID: 38273445 DOI: 10.1021/acs.molpharmaceut.3c00774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
Poly(ethylene glycol) (PEG) is used in many common products, such as cosmetics. PEG, however, is also used to covalently conjugate drug molecules, proteins, or nanocarriers, which is termed PEGylation, to serve as a shield against the natural immune system of the human body. Repeated administration of some PEGylated products, however, is known to induce anti-PEG antibodies. In addition, preexisting anti-PEG antibodies are now being detected in healthy individuals who have never received PEGylated therapeutics. Both treatment-induced and preexisting anti-PEG antibodies alter the pharmacokinetic properties, which can result in a subsequent reduction in the therapeutic efficacy of administered PEGylated therapeutics through the so-called accelerated blood clearance (ABC) phenomenon. Moreover, these anti-PEG antibodies are widely reported to be related to severe hypersensitivity reactions following the administration of PEGylated therapeutics, including COVID-19 vaccines. We recently reported that the topical application of a cosmetic product containing PEG derivatives induced anti-PEG immunoglobulin M (IgM) in a mouse model. Our finding indicates that the PEG derivatives in cosmetic products could be a major cause of the preexistence of anti-PEG antibodies in healthy individuals. In this study, therefore, the pharmacokinetics and therapeutic effects of Doxil (doxorubicin hydrochloride-loaded PEGylated liposomes) and oxaliplatin-loaded PEGylated liposomes (Liposomal l-OHP) were studied in mice. The anti-PEG IgM antibodies induced by the topical application of cosmetic products obviously accelerated the blood clearance of both PEGylated liposomal formulations. Moreover, in C26 tumor-bearing mice, the tumor growth suppressive effects of both Doxil and Liposomal l-OHP were significantly attenuated in the presence of anti-PEG IgM antibodies induced by the topical application of cosmetic products. These results confirm that the topical application of a cosmetic product containing PEG derivatives could produce preexisting anti-PEG antibodies that then affect the therapeutic efficacy of subsequent doses of PEGylated therapeutics.
Collapse
Affiliation(s)
- Sherif A Gaballa
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Taro Shimizu
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Haruka Takata
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
- Institute of Innovative Drug Delivery System, Graduate School of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Hidenori Ando
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
- Institute of Innovative Drug Delivery System, Graduate School of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Mohamed Ibrahim
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Sherif E Emam
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Nana Cristina Amorim Matsuo
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Yuri Kim
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | - Youssef W Naguib
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Fatma M Mady
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Khaled A Khaled
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
- Institute of Innovative Drug Delivery System, Graduate School of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| |
Collapse
|
6
|
Maphutha J, Twilley D, Lall N. The Role of the PTEN Tumor Suppressor Gene and Its Anti-Angiogenic Activity in Melanoma and Other Cancers. Molecules 2024; 29:721. [PMID: 38338464 PMCID: PMC10856229 DOI: 10.3390/molecules29030721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/29/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
Human malignant melanoma and other solid cancers are largely driven by the inactivation of tumor suppressor genes and angiogenesis. Conventional treatments for cancer (surgery, radiation therapy, and chemotherapy) are employed as first-line treatments for solid cancers but are often ineffective as monotherapies due to resistance and toxicity. Thus, targeted therapies, such as bevacizumab, which targets vascular endothelial growth factor, have been approved by the US Food and Drug Administration (FDA) as angiogenesis inhibitors. The downregulation of the tumor suppressor, phosphatase tensin homolog (PTEN), occurs in 30-40% of human malignant melanomas, thereby elucidating the importance of the upregulation of PTEN activity. Phosphatase tensin homolog (PTEN) is modulated at the transcriptional, translational, and post-translational levels and regulates key signaling pathways such as the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) and mitogen-activated protein kinase (MAPK) pathways, which also drive angiogenesis. This review discusses the inhibition of angiogenesis through the upregulation of PTEN and the inhibition of hypoxia-inducible factor 1 alpha (HIF-1-α) in human malignant melanoma, as no targeted therapies have been approved by the FDA for the inhibition of angiogenesis in human malignant melanoma. The emergence of nanocarrier formulations to enhance the pharmacokinetic profile of phytochemicals that upregulate PTEN activity and improve the upregulation of PTEN has also been discussed.
Collapse
Affiliation(s)
- Jacqueline Maphutha
- Department of Plant and Soil Sciences, University of Pretoria, Pretoria 0002, South Africa
| | - Danielle Twilley
- Department of Plant and Soil Sciences, University of Pretoria, Pretoria 0002, South Africa
| | - Namrita Lall
- Department of Plant and Soil Sciences, University of Pretoria, Pretoria 0002, South Africa
- School of Natural Resources, University of Missouri, Columbia, MO 65211, USA
- College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, India
| |
Collapse
|
7
|
Chen J, Hu S, Sun M, Shi J, Zhang H, Yu H, Yang Z. Recent advances and clinical translation of liposomal delivery systems in cancer therapy. Eur J Pharm Sci 2024; 193:106688. [PMID: 38171420 DOI: 10.1016/j.ejps.2023.106688] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 11/23/2023] [Accepted: 12/31/2023] [Indexed: 01/05/2024]
Abstract
The limitations of conventional cancer treatment are driving the emergence and development of nanomedicines. Research in liposomal nanomedicine for cancer therapy is rapidly increasing, opening up new horizons for cancer treatment. Liposomal nanomedicine, which focuses on targeted drug delivery to improve the therapeutic effect of cancer while reducing damage to normal tissues and cells, has great potential in the field of cancer therapy. This review aims to clarify the advantages of liposomal delivery systems in cancer therapy. We describe the recent understanding of spatiotemporal fate of liposomes in the organism after different routes of drug administration. Meanwhile, various types of liposome-based drug delivery systems that exert their respective advantages in cancer therapy while reducing side effects were discussed. Moreover, the combination of liposomal agents with other therapies (such as photodynamic therapy and photothermal therapy) has demonstrated enhanced tumor-targeting efficiency and therapeutic efficacy. Finally, the opportunities and challenges faced by the field of liposome nanoformulations for entering the clinical treatment of cancer are highlighted.
Collapse
Affiliation(s)
- Jiayi Chen
- School of Life Sciences, Jilin University, Changchun, China
| | - Siyuan Hu
- School of Life Sciences, Jilin University, Changchun, China
| | - Man Sun
- School of Life Sciences, Jilin University, Changchun, China
| | - Jianan Shi
- School of Life Sciences, Jilin University, Changchun, China
| | - Huan Zhang
- School of Life Sciences, Jilin University, Changchun, China
| | - Hongmei Yu
- China-Japan Union Hospital, Jilin University, Changchun, China.
| | - Zhaogang Yang
- School of Life Sciences, Jilin University, Changchun, China.
| |
Collapse
|
8
|
Fernandes S, Cassani M, Cavalieri F, Forte G, Caruso F. Emerging Strategies for Immunotherapy of Solid Tumors Using Lipid-Based Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305769. [PMID: 38054651 PMCID: PMC10885677 DOI: 10.1002/advs.202305769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/09/2023] [Indexed: 12/07/2023]
Abstract
The application of lipid-based nanoparticles for COVID-19 vaccines and transthyretin-mediated amyloidosis treatment have highlighted their potential for translation to cancer therapy. However, their use in delivering drugs to solid tumors is limited by ineffective targeting, heterogeneous organ distribution, systemic inflammatory responses, and insufficient drug accumulation at the tumor. Instead, the use of lipid-based nanoparticles to remotely activate immune system responses is an emerging effective strategy. Despite this approach showing potential for treating hematological cancers, its application to treat solid tumors is hampered by the selection of eligible targets, tumor heterogeneity, and ineffective penetration of activated T cells within the tumor. Notwithstanding, the use of lipid-based nanoparticles for immunotherapy is projected to revolutionize cancer therapy, with the ultimate goal of rendering cancer a chronic disease. However, the translational success is likely to depend on the use of predictive tumor models in preclinical studies, simulating the complexity of the tumor microenvironment (e.g., the fibrotic extracellular matrix that impairs therapeutic outcomes) and stimulating tumor progression. This review compiles recent advances in the field of antitumor lipid-based nanoparticles and highlights emerging therapeutic approaches (e.g., mechanotherapy) to modulate tumor stiffness and improve T cell infiltration, and the use of organoids to better guide therapeutic outcomes.
Collapse
Affiliation(s)
- Soraia Fernandes
- Center for Translational Medicine (CTM)International Clinical Research Centre (ICRC)St. Anne HospitalBrno656 91Czech Republic
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Marco Cassani
- Center for Translational Medicine (CTM)International Clinical Research Centre (ICRC)St. Anne HospitalBrno656 91Czech Republic
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Francesca Cavalieri
- School of ScienceRMIT UniversityMelbourneVictoria3000Australia
- Dipartimento di Scienze e Tecnologie ChimicheUniversita di Roma “Tor Vergata”Via della Ricerca Scientifica 1Rome00133Italy
| | - Giancarlo Forte
- Center for Translational Medicine (CTM)International Clinical Research Centre (ICRC)St. Anne HospitalBrno656 91Czech Republic
- School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonSE5 9NUUK
| | - Frank Caruso
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| |
Collapse
|
9
|
Cassani M, Fernandes S, Oliver‐De La Cruz J, Durikova H, Vrbsky J, Patočka M, Hegrova V, Klimovic S, Pribyl J, Debellis D, Skladal P, Cavalieri F, Caruso F, Forte G. YAP Signaling Regulates the Cellular Uptake and Therapeutic Effect of Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302965. [PMID: 37946710 PMCID: PMC10787066 DOI: 10.1002/advs.202302965] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 09/27/2023] [Indexed: 11/12/2023]
Abstract
Interactions between living cells and nanoparticles are extensively studied to enhance the delivery of therapeutics. Nanoparticles size, shape, stiffness, and surface charge are regarded as the main features able to control the fate of cell-nanoparticle interactions. However, the clinical translation of nanotherapies has so far been limited, and there is a need to better understand the biology of cell-nanoparticle interactions. This study investigates the role of cellular mechanosensitive components in cell-nanoparticle interactions. It is demonstrated that the genetic and pharmacologic inhibition of yes-associated protein (YAP), a key component of cancer cell mechanosensing apparatus and Hippo pathway effector, improves nanoparticle internalization in triple-negative breast cancer cells regardless of nanoparticle properties or substrate characteristics. This process occurs through YAP-dependent regulation of endocytic pathways, cell mechanics, and membrane organization. Hence, the study proposes targeting YAP may sensitize triple-negative breast cancer cells to chemotherapy and increase the selectivity of nanotherapy.
Collapse
Affiliation(s)
- Marco Cassani
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Soraia Fernandes
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Jorge Oliver‐De La Cruz
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- Institute for Bioengineering of Catalonia (IBEC)The Barcelona Institute for Science and Technology (BIST)BarcelonaSpain
| | - Helena Durikova
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
| | - Jan Vrbsky
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
| | - Marek Patočka
- NenoVisionPurkynova 649/127Brno61200Czech Republic
- Faculty of Mechanical EngineeringBrno University of TechnologyTechnicka 2896/2Brno61669Czech Republic
| | | | - Simon Klimovic
- Department of Bioanalytical InstrumentationCEITEC Masaryk UniversityBrno60200Czech Republic
| | - Jan Pribyl
- Department of Bioanalytical InstrumentationCEITEC Masaryk UniversityBrno60200Czech Republic
| | - Doriana Debellis
- Electron Microscopy FacilityFondazione Istituto Italiano Di TecnologiaVia Morego 30Genoa16163Italy
| | - Petr Skladal
- Department of Bioanalytical InstrumentationCEITEC Masaryk UniversityBrno60200Czech Republic
| | - Francesca Cavalieri
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
- School of ScienceRMIT UniversityMelbourne3000VictoriaAustralia
- Dipartimento di Scienze e Tecnologie ChimicheUniversità di Roma “Tor Vergata”Via Della Ricerca ScientificaRome00133Italy
| | - Frank Caruso
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Giancarlo Forte
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonWC2R 2LSUK
| |
Collapse
|
10
|
Komedchikova EN, Kolesnikova OA, Syuy AV, Volkov VS, Deyev SM, Nikitin MP, Shipunova VO. Targosomes: Anti-HER2 PLGA nanocarriers for bioimaging, chemotherapy and local photothermal treatment of tumors and remote metastases. J Control Release 2024; 365:317-330. [PMID: 37996056 DOI: 10.1016/j.jconrel.2023.11.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/14/2023] [Accepted: 11/18/2023] [Indexed: 11/25/2023]
Abstract
Developing combined cancer therapy strategies is of utmost importance as it can enhance treatment efficacy, overcome drug resistance, and ultimately improve patient outcomes by targeting multiple pathways and mechanisms involved in cancer growth and progression. Specifically, the potential of developing a combination chemo&photothermal therapy using targeted polymer nanoparticles as nanocarriers offers a promising approach for synergistic cancer treatment by combining the benefits of both therapies, such as targeted drug delivery and localized hyperthermia. Here, we report the first targeted anti-HER2 PLGA nanocarriers, called targosomes, that simultaneously possess photothermal, chemotherapeutic and diagnostic properties using only molecular payloads. Biocompatible poly(lactic-co-glycolic acid), PLGA, nanoparticles were loaded with photosensitizer phthalocyanine, diagnostic dye Nile Blue, and chemotherapeutic drug irinotecan, which was chosen as a result of screening a panel of theragnostic nanoparticles. The targeted delivery to cell surface oncomarker HER2 was ensured by nanoparticle modification with the anti-HER2 monoclonal antibody, trastuzumab, using the one-pot synthesis method without chemical conjugation. The irradiation tests revealed prominent photothermal properties of nanoparticles, namely heating by 35 °C in 10 min. Nanoparticles exhibited a 7-fold increase in binding and nearly an 18-fold increase in cytotoxicity for HER2-overexpressing cells compared to cells lacking HER2 expression. This enhancement of cytotoxicity was further amplified by >20-fold under NIR light irradiation. In vivo studies proved the efficacy of nanoparticles for bioimaging of primary tumor and metastasis sites and demonstrated 93% tumor growth inhibition, making these nanoparticles excellent candidates for translation into theragnostic applications.
Collapse
Affiliation(s)
- E N Komedchikova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - O A Kolesnikova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - A V Syuy
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - V S Volkov
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - S M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - M P Nikitin
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia; Nanobiomedicine Division, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - V O Shipunova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; Nanobiomedicine Division, Sirius University of Science and Technology, 354340 Sochi, Russia.
| |
Collapse
|
11
|
Tantray J, Patel A, Prajapati BG, Kosey S, Bhattacharya S. The Use of Lipid-based Nanocarriers to Improve Ovarian Cancer Treatment: An Overview of Recent Developments. Curr Pharm Biotechnol 2024; 25:2200-2217. [PMID: 38357950 DOI: 10.2174/0113892010279572240126052844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/08/2024] [Accepted: 01/16/2024] [Indexed: 02/16/2024]
Abstract
Ovarian cancer poses a formidable health challenge for women globally, necessitating innovative therapeutic approaches. This review provides a succinct summary of the current research status on lipid-based nanocarriers in the context of ovarian cancer treatment. Lipid-based nanocarriers, including liposomes, solid lipid nanoparticles (SLNs), and nanostructured lipid carriers (NLCs), offer a promising solution for delivering anticancer drugs with enhanced therapeutic effectiveness and reduced adverse effects. Their versatility in transporting both hydrophobic and hydrophilic medications makes them well-suited for a diverse range of anticancer drugs. Active targeting techniques like ligand-conjugation and surface modifications have been used to reduce off-target effects and achieve tumour-specific medication delivery. The study explores formulation techniques and adjustments meant to enhance drug stability and encapsulation in these nanocarriers. Encouraging results from clinical trials and preclinical investigations underscore the promise of lipid-based nanocarriers in ovarian cancer treatment, providing optimism for improved patient outcomes. Notwithstanding these advancements, challenges related to clearance, long-term stability, and scalable manufacturing persist. Successfully translating lipidbased nanocarriers into clinical practice requires addressing these hurdles. To sum up, lipidbased nanocarriers are a viable strategy to improve the effectiveness of therapy for ovarian cancer. With their more focused medication administration and lower systemic toxicity, they may completely change the way ovarian cancer is treated and increase patient survival rates. Lipidbased nanocarriers need to be further researched and developed to become a therapeutically viable treatment for ovarian cancer.
Collapse
Affiliation(s)
- Junaid Tantray
- Department of Pharmacology, NIMS Institute of Pharmacy, NIMS University, Rajasthan, India
| | - Akhilesh Patel
- Department of Pharmacology, NIMS Institute of Pharmacy, NIMS University, Rajasthan, India
| | - Bhupendra G Prajapati
- Department of Pharmaceutics and Pharmaceutical Technology, Shree S.K. Patel College of Pharmaceutical Education & Research, Ganpat University, Gujarat, India
| | - Sourabh Kosey
- Department of Pharmacy Practice, ISF College of Pharmacy, Punjab, India
| | - Sankha Bhattacharya
- School of Pharmacy & Technology, Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| |
Collapse
|
12
|
Adekiya TA, Owoseni O. Emerging frontiers in nanomedicine targeted therapy for prostate cancer. Cancer Treat Res Commun 2023; 37:100778. [PMID: 37992539 DOI: 10.1016/j.ctarc.2023.100778] [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: 08/01/2023] [Revised: 10/23/2023] [Accepted: 11/13/2023] [Indexed: 11/24/2023]
Abstract
Prostate cancer is a prevalent cancer in men, often treated with chemotherapy. However, it tumor cells are clinically grows slowly and is heterogeneous, leading to treatment resistance and recurrence. Nanomedicines, through targeted delivery using nanocarriers, can enhance drug accumulation at the tumor site, sustain drug release, and counteract drug resistance. In addition, combination therapy using nanomedicines can target multiple cancer pathways, improving effectiveness and addressing tumor heterogeneity. The application of nanomedicine in prostate cancer treatment would be an important strategy in controlling tumor dynamic process as well as improve survival. Thus, this review highlights therapeutic nanoparticles as a solution for prostate cancer chemotherapy, exploring targeting strategies and approaches to combat drug resistance.
Collapse
Affiliation(s)
- Tayo Alex Adekiya
- Department of Pharmaceutical Sciences, Howard University, Washington, DC 20059, United States.
| | - Oluwanifemi Owoseni
- Department of Pharmaceutical Sciences, Howard University, Washington, DC 20059, United States
| |
Collapse
|
13
|
Yuan M, Chen T, Jin L, Zhang P, Xie L, Zhou S, Fan L, Wang L, Zhang C, Tang N, Guo L, Xie C, Duo Y, Li L, Shi L. A carrier-free supramolecular nano-twin-drug for overcoming irinotecan-resistance and enhancing efficacy against colorectal cancer. J Nanobiotechnology 2023; 21:393. [PMID: 37898773 PMCID: PMC10612220 DOI: 10.1186/s12951-023-02157-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/11/2023] [Indexed: 10/30/2023] Open
Abstract
Irinotecan (Ir) is commonly employed as a first-line chemotherapeutic treatment for colorectal cancer (CRC). However, tremendous impediments remain to be addressed to surmount drug resistance and ameliorate adverse events. Poly-ADP-Ribose Polymerase (PARP) participates in the maintenance of genome stability and the repair of DNA damage, thus playing a critical role in chemotherapy resistance. In this work, we introduce a novel curative strategy that utilizes nanoparticles (NPs) prepared by dynamic supramolecular co-assembly of Ir and a PARP inhibitor (PARPi) niraparib (Nir) through π-π stacking and hydrogen bond interactions. The Ir and Nir self-assembled Nano-Twin-Drug of (Nir-Ir NPs) could enhance the therapeutic effect on CRC by synergistically inhibiting the DNA damage repair pathway and activating the tumor cell apoptosis process without obvious toxicity. In addition, the Nir-Ir NPs could effectively reverse irinotecan-resistance by inhibiting the expression of multiple resistance protein-1 (MRP-1). Overall, our study underscores the distinctive advantages and potential of Nir-Ir NPs as a complementary strategy to chemotherapy by simultaneously overcoming the Ir resistance and improving the anti-tumor efficacy against CRC.
Collapse
Affiliation(s)
- Miaomiao Yuan
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of pharmacology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong, Sun Yat-sen University, Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Shenzhen, China
| | - Tong Chen
- Department of pharmacology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong, Sun Yat-sen University, Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Shenzhen, China
| | - Lu Jin
- School of Pharmaceutical Sciences, Sun Yat-sen University, 510006, Guangzhou, China
| | - Peng Zhang
- Department of Pharmacy, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, 47 Youyi Road, Shenzhen, 518001, China.
| | - Luoyijun Xie
- Department of pharmacology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong, Sun Yat-sen University, Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Shenzhen, China
| | - Shuyi Zhou
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lianfeng Fan
- Department of pharmacology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong, Sun Yat-sen University, Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Shenzhen, China
| | - Li Wang
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Cai Zhang
- Department of pharmacology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong, Sun Yat-sen University, Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Shenzhen, China
| | - Ning Tang
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - LiHao Guo
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chengmei Xie
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yanhong Duo
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Ling Li
- Department of pharmacology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong, Sun Yat-sen University, Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Shenzhen, China.
| | - Leilei Shi
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
14
|
Chattopadhyay S, Liao YP, Wang X, Nel AE. Use of Stromal Intervention and Exogenous Neoantigen Vaccination to Boost Pancreatic Cancer Chemo-Immunotherapy by Nanocarriers. Bioengineering (Basel) 2023; 10:1205. [PMID: 37892935 PMCID: PMC10604647 DOI: 10.3390/bioengineering10101205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Despite the formidable treatment challenges of pancreatic ductal adenocarcinoma (PDAC), considerable progress has been made in improving drug delivery via pioneering nanocarriers. These innovations are geared towards overcoming the obstacles presented by dysplastic stroma and fostering anti-PDAC immune reactions. We are currently conducting research aimed at enhancing chemotherapy to stimulate anti-tumor immunity by inducing immunogenic cell death (ICD). This is accomplished using lipid bilayer-coated nanocarriers, which enable the attainment of synergistic results. Noteworthy examples include liposomes and lipid-coated mesoporous silica nanoparticles known as "silicasomes". These nanocarriers facilitate remote chemotherapy loading, as well as the seamless integration of immunomodulators into the lipid bilayer. In this communication, we elucidate innovative ways for further improving chemo-immunotherapy. The first is the development of a liposome platform engineered by the remote loading of irinotecan while incorporating a pro-resolving lipoxin in the lipid bilayer. This carrier interfered in stromal collagen deposition, as well as boosting the irinotecan-induced ICD response. The second approach was to synthesize polymer nanoparticles for the delivery of mutated KRAS peptides in conjunction with a TLR7/8 agonist. The dual delivery vaccine particle boosted the generation of antigen-specific cytotoxic T-cells that are recruited to lymphoid structures at the cancer site, with a view to strengthening the endogenous vaccination response achieved by chemo-immunotherapy.
Collapse
Affiliation(s)
- Saborni Chattopadhyay
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Yu-Pei Liao
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Xiang Wang
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - André E. Nel
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
15
|
Song S, Sun D, Wang H, Wang J, Yan H, Zhao X, Fawcett JP, Xu X, Cai D, Gu J. Full-profile pharmacokinetics, anticancer activity and toxicity of an extended release trivalent PEGylated irinotecan prodrug. Acta Pharm Sin B 2023; 13:3444-3453. [PMID: 37655324 PMCID: PMC10466002 DOI: 10.1016/j.apsb.2023.01.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/22/2022] [Accepted: 12/03/2022] [Indexed: 01/12/2023] Open
Abstract
Irinotecan is an anticancer topoisomerase I inhibitor that acts as a prodrug of the active metabolite, SN-38. Unfortunately, the limited utility of irinotecan is attributed to its pH-dependent stability, short half-life and dose-limiting toxicity. To address this problem, a novel trivalent PEGylated prodrug (PEG-[Irinotecan]3) has been synthesized and its full-profile pharmacokinetics, antitumor activity and toxicity compared with those of irinotecan. The results show that after intravenous administration to rats, PEG-[Irinotecan]3 undergoes stepwise loss of irinotecan to form PEG-[Irinotecan]3‒x (x = 1,2) and PEG-[linker] during which time the released irinotecan undergoes conversion to SN-38. As compared with conventional irinotecan, PEG-[Irinotecan]3 displays extended release of irinotecan and efficient formation of SN-38 with significantly improved AUC and half-life. In a colorectal cancer-bearing model in nude mice, the tumor concentrations of irinotecan and SN-38 produced by PEG-[Irinotecan]3 were respectively 86.2 and 2293 times higher at 48 h than produced by irinotecan. In summary, PEG-[Irinotecan]3 displays superior pharmacokinetic characteristics and antitumor activity with lower toxicity than irinotecan. This supports the view that PEG-[Irinotecan]3 is a superior anticancer drug to irinotecan and it has entered the phase II trial stage.
Collapse
Affiliation(s)
- Shiwen Song
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Dong Sun
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Hong Wang
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun 130012, China
- Center for Food and Drug Inspection of NMPA, Changchun 130012, China
| | | | - Huijing Yan
- JenKem Technology Co., Ltd., Tianjin 300450, China
| | - Xuan Zhao
- JenKem Technology Co., Ltd., Tianjin 300450, China
| | - John Paul Fawcett
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xin Xu
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Deqi Cai
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun 130012, China
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Jingkai Gu
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun 130012, China
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
- Beijing Institute of Drug Metabolism, Beijing 102209, China
| |
Collapse
|
16
|
Moholkar DN, Kandimalla R, Gupta RC, Aqil F. Advances in lipid-based carriers for cancer therapeutics: Liposomes, exosomes and hybrid exosomes. Cancer Lett 2023; 565:216220. [PMID: 37209944 PMCID: PMC10325927 DOI: 10.1016/j.canlet.2023.216220] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/18/2023] [Accepted: 05/06/2023] [Indexed: 05/22/2023]
Abstract
Cancer has recently surpassed heart disease as the leading cause of deaths worldwide for the age group 45-65 and has been the primary focus for biomedical researchers. Presently, the drugs involved in the first-line cancer therapy are raising concerns due to high toxicity and lack of selectivity to cancer cells. There has been a significant increase in research with innovative nano formulations to entrap the therapeutic payload to enhance efficacy and eliminate or minimize toxic effects. Lipid-based carriers stand out due to their unique structural properties and biocompatible nature. The two main leaders of lipid-based drug carriers: long known liposomes and comparatively new exosomes have been well-researched. The similarity between the two lipid-based carriers is the vesicular structure with the core's capability to carry the payload. While liposomes utilize chemically derived and altered phospholipid components, the exosomes are naturally occurring vesicles with inherent lipids, proteins, and nucleic acids. More recently, researchers have focused on developing hybrid exosomes by fusing liposomes and exosomes. Combining these two types of vesicles may offer some advantages such as high drug loading, targeted cellular uptake, biocompatibility, controlled release, stability in harsh conditions and low immunogenicity.
Collapse
Affiliation(s)
- Disha N Moholkar
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA
| | - Raghuram Kandimalla
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA; Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA
| | - Ramesh C Gupta
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA; Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA.
| | - Farrukh Aqil
- Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA; Department of Medicine, University of Louisville, Louisville, KY, 40202, USA.
| |
Collapse
|
17
|
López-Goerne T, Padilla-Godínez FJ. Catalytic Nanomedicine as a Therapeutic Approach to Brain Tumors: Main Hypotheses for Mechanisms of Action. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:nano13091541. [PMID: 37177086 PMCID: PMC10180296 DOI: 10.3390/nano13091541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 04/19/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive primary malignant tumor of the brain. Although there are currently a wide variety of therapeutic approaches focused on tumor elimination, such as radiotherapy, chemotherapy, and tumor field therapy, among others, the main approach involves surgery to remove the GBM. However, since tumor growth occurs in normal brain tissue, complete removal is impossible, and patients end up requiring additional treatments after surgery. In this line, Catalytic Nanomedicine has achieved important advances in developing bionanocatalysts, brain-tissue-biocompatible catalytic nanostructures capable of destabilizing the genetic material of malignant cells, causing their apoptosis. Previous work has demonstrated the efficacy of bionanocatalysts and their selectivity for cancer cells without affecting surrounding healthy tissue cells. The present review provides a detailed description of these nanoparticles and their potential mechanisms of action as antineoplastic agents, covering the most recent research and hypotheses from their incorporation into the tumor bed, internalization via endocytosis, specific chemotaxis by mitochondrial and nuclear genetic material, and activation of programmed cell death. In addition, a case report of a patient with GBM treated with the bionanocatalysts following tumor removal surgery is described. Finally, the gaps in knowledge that must be bridged before the clinical translation of these compounds with such a promising future are detailed.
Collapse
Affiliation(s)
- Tessy López-Goerne
- Nanotechnology and Nanomedicine Laboratory, Department of Health Care, Metropolitan Autonomous University-Xochimilco, Mexico City 04960, Mexico
| | - Francisco J Padilla-Godínez
- Nanotechnology and Nanomedicine Laboratory, Department of Health Care, Metropolitan Autonomous University-Xochimilco, Mexico City 04960, Mexico
| |
Collapse
|
18
|
Olajubutu O, Ogundipe OD, Adebayo A, Adesina SK. Drug Delivery Strategies for the Treatment of Pancreatic Cancer. Pharmaceutics 2023; 15:pharmaceutics15051318. [PMID: 37242560 DOI: 10.3390/pharmaceutics15051318] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/15/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
Pancreatic cancer is fast becoming a global menace and it is projected to be the second leading cause of cancer-related death by 2030. Pancreatic adenocarcinomas, which develop in the pancreas' exocrine region, are the predominant type of pancreatic cancer, representing about 95% of total pancreatic tumors. The malignancy progresses asymptomatically, making early diagnosis difficult. It is characterized by excessive production of fibrotic stroma known as desmoplasia, which aids tumor growth and metastatic spread by remodeling the extracellular matrix and releasing tumor growth factors. For decades, immense efforts have been harnessed toward developing more effective drug delivery systems for pancreatic cancer treatment leveraging nanotechnology, immunotherapy, drug conjugates, and combinations of these approaches. However, despite the reported preclinical success of these approaches, no substantial progress has been made clinically and the prognosis for pancreatic cancer is worsening. This review provides insights into challenges associated with the delivery of therapeutics for pancreatic cancer treatment and discusses drug delivery strategies to minimize adverse effects associated with current chemotherapy options and to improve the efficiency of drug treatment.
Collapse
Affiliation(s)
| | - Omotola D Ogundipe
- Department of Pharmaceutical Sciences, Howard University, Washington, DC 20059, USA
| | - Amusa Adebayo
- Department of Pharmaceutical Sciences, Howard University, Washington, DC 20059, USA
| | - Simeon K Adesina
- Department of Pharmaceutical Sciences, Howard University, Washington, DC 20059, USA
| |
Collapse
|
19
|
Zhang J, Ding H, Zhang F, Xu Y, Liang W, Huang L. New trends in diagnosing and treating ovarian cancer using nanotechnology. Front Bioeng Biotechnol 2023; 11:1160985. [PMID: 37082219 PMCID: PMC10110946 DOI: 10.3389/fbioe.2023.1160985] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/22/2023] [Indexed: 04/07/2023] Open
Abstract
Ovarian cancer stands as the fifth most prevalent cancer among women, causing more mortalities than any other disease of the female reproductive system. There are numerous histological subtypes of ovarian cancer, each of which has distinct clinical characteristics, risk factors, cell origins, molecular compositions, and therapeutic options. Typically, it is identified at a late stage, and there is no efficient screening method. Standard therapies for newly diagnosed cancer are cytoreductive surgery and platinum-based chemotherapy. The difficulties of traditional therapeutic procedures encourage researchers to search for other approaches, such as nanotechnology. Due to the unique characteristics of matter at the nanoscale, nanomedicine has emerged as a potent tool for creating novel drug carriers that are more effective and have fewer adverse effects than traditional treatments. Nanocarriers including liposomes, dendrimers, polymer nanoparticles, and polymer micelles have unique properties in surface chemistry, morphology, and mechanism of action that can distinguish between malignant and normal cells, paving the way for targeted drug delivery. In contrast to their non-functionalized counterparts, the development of functionalized nano-formulations with specific ligands permits selective targeting of ovarian cancers and ultimately increases the therapeutic potential. This review focuses on the application of various nanomaterials to the treatment and diagnosis of ovarian cancer, their advantages over conventional treatment methods, and the effective role of controlled drug delivery systems in the therapy of ovarian cancer.
Collapse
Affiliation(s)
- Juan Zhang
- Department of Gynecology, Shaoxing Maternity and Child Healthcare Hospital, Shaoxing, China
- Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Haigang Ding
- Department of Gynecology, Shaoxing Maternity and Child Healthcare Hospital, Shaoxing, China
- Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Feng Zhang
- Department of Gynecology, Shaoxing Maternity and Child Healthcare Hospital, Shaoxing, China
- Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Yan Xu
- Intensive Care Unit, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Wenqing Liang
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
- *Correspondence: Liping Huang, ; Wenqing Liang,
| | - Liping Huang
- Department of Medical Oncology, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
- *Correspondence: Liping Huang, ; Wenqing Liang,
| |
Collapse
|
20
|
Yusuf A, Almotairy ARZ, Henidi H, Alshehri OY, Aldughaim MS. Nanoparticles as Drug Delivery Systems: A Review of the Implication of Nanoparticles' Physicochemical Properties on Responses in Biological Systems. Polymers (Basel) 2023; 15:polym15071596. [PMID: 37050210 PMCID: PMC10096782 DOI: 10.3390/polym15071596] [Citation(s) in RCA: 143] [Impact Index Per Article: 71.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/11/2023] [Accepted: 03/14/2023] [Indexed: 04/14/2023] Open
Abstract
In the last four decades, nanotechnology has gained momentum with no sign of slowing down. The application of inventions or products from nanotechnology has revolutionised all aspects of everyday life ranging from medical applications to its impact on the food industry. Nanoparticles have made it possible to significantly extend the shelf lives of food product, improve intracellular delivery of hydrophobic drugs and improve the efficacy of specific therapeutics such as anticancer agents. As a consequence, nanotechnology has not only impacted the global standard of living but has also impacted the global economy. In this review, the characteristics of nanoparticles that confers them with suitable and potentially toxic biological effects, as well as their applications in different biological fields and nanoparticle-based drugs and delivery systems in biomedicine including nano-based drugs currently approved by the U.S. Food and Drug Administration (FDA) are discussed. The possible consequence of continuous exposure to nanoparticles due to the increased use of nanotechnology and possible solution is also highlighted.
Collapse
Affiliation(s)
- Azeez Yusuf
- Irish Centre for Genetic Lung Disease, Department of Medicine, RCSI University of Medicine and Health Sciences, Beaumont Hospital, D02 YN77 Dublin, Ireland
| | | | - Hanan Henidi
- Research Department, Health Sciences Research Center, Princess Nourah bint Abdulrahman University, Riyadh 84428, Saudi Arabia
| | - Ohoud Y Alshehri
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11564, Saudi Arabia
| | - Mohammed S Aldughaim
- Research Center, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh 11451, Saudi Arabia
| |
Collapse
|
21
|
Current Update on Nanotechnology-Based Approaches in Ovarian Cancer Therapy. Reprod Sci 2023; 30:335-349. [PMID: 35585292 DOI: 10.1007/s43032-022-00968-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 05/06/2022] [Indexed: 10/18/2022]
Abstract
Ovarian cancer is one of the leading causes of cancer-related deaths among women. The drawbacks of conventional therapeutic strategies encourage researchers to look for alternative strategies, including nanotechnology. Nanotechnology is one of the upcoming domains of science that is rechanneled towards targeted cancer therapy and diagnosis. Nanocarriers such as dendrimers, liposomes, polymer micelles, and polymer nanoparticles present distinct surface characteristics in morphology, surface chemistry, and mode of action that help differentiate normal and malignant cells, which paves the way for target-specific drug delivery. Similarly, nanoparticles have been strategically utilized as efficacious vehicles to deliver drugs that alter the epigenetic modifications in epigenetic therapy. Some studies suggest that the use of specialized target-modified nanoparticles in siRNA-based nanotherapy prevents internalization and improves the antitumor activity of siRNA by ensuring unrestrained entry of siRNA into the tumor vasculature and efficient intracellular delivery of siRNA. Moreover, research findings highlight the significance of utilizing nanoparticles as depots for photosensitive drugs in photodynamic therapy. The applicability of nanoparticles is further extended to medical imaging. They serve as contrast agents in combination with conventional imaging modalities such as MRI, CT, and fluorescence-based imaging to produce vivid and enhanced images of tumors. Therefore, this review aims to explore and delve deeper into the advent of various nanotechnology-based therapeutic and imaging techniques that provide non-invasive and effective means to tackle ovarian cancers.
Collapse
|
22
|
Ding R, Zhao Z, He J, Tao Y, Zhang H, Yuan R, Sun K, Shi Y. Preparation, Drug Distribution, and In Vivo Evaluation of the Safety of Protein Corona Liposomes for Liraglutide Delivery. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:nano13030540. [PMID: 36770503 PMCID: PMC9920406 DOI: 10.3390/nano13030540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/17/2023] [Accepted: 01/17/2023] [Indexed: 05/13/2023]
Abstract
The development of oral drug delivery systems is challenging, and issues related to the mucus layer and low intestinal epithelial permeability have not yet been surmounted. The purpose of this study was to develop a promising formulation that is more adapted to in vivo absorption and to facilitate the administration of oral liraglutide. Cationic liposomes (CLs) linked to AT-1002 were prepared using a double-emulsion method, and BSA was adsorbed on the surface of the AT-CLs, resulting in protein corona cationic liposomes with AT-1002 (Pc-AT-CLs). The preparation method was determined by investigating various process parameters. The particle size, potential, and encapsulation efficiency (EE%) of the Pc-AT-CLs were 202.9 ± 12.4 nm, 1.76 ± 4.87 mV, and 84.63 ± 5.05%, respectively. The transmission electron microscopy (TEM) imaging revealed a nearly spherical structure of the Pc-AT-CLs, with a recognizable coating. The circular dichroism experiments confirmed that the complex preparation process did not affect the secondary structure of liraglutide. With the addition of BSA and AT-1002, the mucosal accumulation of the Pc-AT-CLs was nearly two times lower than that of the AT-CLs, and the degree of enteric metaplasia was 1.35 times higher than that of the PcCLs. The duration of the intestinal absorption of the Pc-AT-CLs was longer, offering remarkable biological safety.
Collapse
Affiliation(s)
- Ruihuan Ding
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 261400, China
| | - Zhenyu Zhao
- School of Life Science, Yantai University, Yantai 261400, China
| | - Jibiao He
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 261400, China
| | - Yuping Tao
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 261400, China
| | - Houqian Zhang
- School of Life Science, Yantai University, Yantai 261400, China
| | - Ranran Yuan
- School of Life Science, Yantai University, Yantai 261400, China
| | - Kaoxiang Sun
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 261400, China
- Correspondence: (K.S.); (Y.S.)
| | - Yanan Shi
- School of Life Science, Yantai University, Yantai 261400, China
- Correspondence: (K.S.); (Y.S.)
| |
Collapse
|
23
|
Biological Use of Nanostructured Silica-Based Materials Functionalized with Metallodrugs: The Spanish Perspective. Int J Mol Sci 2023; 24:ijms24032332. [PMID: 36768659 PMCID: PMC9917151 DOI: 10.3390/ijms24032332] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/11/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Since the pioneering work of Vallet-Regí's group on the design and synthesis of mesoporous silica-based materials with therapeutic applications, during the last 15 years, the potential use of mesoporous silica nanostructured materials as drug delivery vehicles has been extensively explored. The versatility of these materials allows the design of a wide variety of platforms that can incorporate numerous agents of interest (fluorophores, proteins, drugs, etc.) in a single scaffold. However, the use of these systems loaded with metallodrugs as cytotoxic agents against different diseases and with distinct therapeutic targets has been studied to a much lesser extent. This review will focus on the work carried out in this field, highlighting both the pioneering and recent contributions of Spanish groups that have synthesized a wide variety of systems based on titanium, tin, ruthenium, copper and silver complexes supported onto nanostructured silica. In addition, this article will also discuss the importance of the structural features of the systems for evaluating and modulating their therapeutic properties. Finally, the most interesting results obtained in the study of the potential therapeutic application of these metallodrug-functionalized silica-based materials against cancer and bacteria will be described, paying special attention to preclinical trials in vivo.
Collapse
|
24
|
Das CGA, Kumar VG, Dhas TS, Karthick V, Kumar CMV. Nanomaterials in anticancer applications and their mechanism of action - A review. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 47:102613. [PMID: 36252911 DOI: 10.1016/j.nano.2022.102613] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 10/01/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022]
Abstract
The current challenges in cancer treatment using conventional therapies have made the emergence of nanotechnology with more advancements. The exponential growth of nanoscience has drawn to develop nanomaterials (NMs) with therapeutic activities. NMs have enormous potential in cancer treatment by altering the drug toxicity profile. Nanoparticles (NPs) with enhanced surface characteristics can diffuse more easily inside tumor cells, thus delivering an optimal concentration of drugs at tumor site while reducing the toxicity. Cancer cells can be targeted with greater affinity by utilizing NMs with tumor specific constituents. Furthermore, it bypasses the bottlenecks of indiscriminate biodistribution of the antitumor agent and high administration dosage. Here, we focus on the recent advances on the use of various nanomaterials for cancer treatment, including targeting cancer cell surfaces, tumor microenvironment (TME), organelles, and their mechanism of action. The paradigm shift in cancer management is achieved through the implementation of anticancer drug delivery using nano routes.
Collapse
Affiliation(s)
- C G Anjali Das
- Centre for Ocean Research, Col. Dr. Jeppiaar Research Park, Sathyabama Institute of Science and Technology, Jeppiaar Nagar, Rajiv Gandhi Salai, Chennai 600119, India; Earth Science and Technology Cell (Marine Biotechnological Studies), Sathyabama Institute of Science and Technology, Rajiv Gandhi Salai, Chennai 600119, India.
| | - V Ganesh Kumar
- Centre for Ocean Research, Col. Dr. Jeppiaar Research Park, Sathyabama Institute of Science and Technology, Jeppiaar Nagar, Rajiv Gandhi Salai, Chennai 600119, India; Earth Science and Technology Cell (Marine Biotechnological Studies), Sathyabama Institute of Science and Technology, Rajiv Gandhi Salai, Chennai 600119, India.
| | - T Stalin Dhas
- Centre for Ocean Research, Col. Dr. Jeppiaar Research Park, Sathyabama Institute of Science and Technology, Jeppiaar Nagar, Rajiv Gandhi Salai, Chennai 600119, India; Earth Science and Technology Cell (Marine Biotechnological Studies), Sathyabama Institute of Science and Technology, Rajiv Gandhi Salai, Chennai 600119, India.
| | - V Karthick
- Centre for Ocean Research, Col. Dr. Jeppiaar Research Park, Sathyabama Institute of Science and Technology, Jeppiaar Nagar, Rajiv Gandhi Salai, Chennai 600119, India; Earth Science and Technology Cell (Marine Biotechnological Studies), Sathyabama Institute of Science and Technology, Rajiv Gandhi Salai, Chennai 600119, India.
| | - C M Vineeth Kumar
- Centre for Ocean Research, Col. Dr. Jeppiaar Research Park, Sathyabama Institute of Science and Technology, Jeppiaar Nagar, Rajiv Gandhi Salai, Chennai 600119, India; Earth Science and Technology Cell (Marine Biotechnological Studies), Sathyabama Institute of Science and Technology, Rajiv Gandhi Salai, Chennai 600119, India.
| |
Collapse
|
25
|
Mercatali L, Vanni S, Miserocchi G, Liverani C, Spadazzi C, Cocchi C, Calabrese C, Gurrieri L, Fausti V, Riva N, Genovese D, Lucarelli E, Focarete ML, Ibrahim T, Calabrò L, De Vita A. The emerging role of cancer nanotechnology in the panorama of sarcoma. Front Bioeng Biotechnol 2022; 10:953555. [PMID: 36324885 PMCID: PMC9618700 DOI: 10.3389/fbioe.2022.953555] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/28/2022] [Indexed: 11/21/2022] Open
Abstract
In the field of nanomedicine a multitude of nanovectors have been developed for cancer application. In this regard, a less exploited target is represented by connective tissue. Sarcoma lesions encompass a wide range of rare entities of mesenchymal origin affecting connective tissues. The extraordinary diversity and rarity of these mesenchymal tumors is reflected in their classification, grading and management which are still challenging. Although they include more than 70 histologic subtypes, the first line-treatment for advanced and metastatic sarcoma has remained unchanged in the last fifty years, excluding specific histotypes in which targeted therapy has emerged. The role of chemotherapy has not been completely elucidated and the outcomes are still very limited. At the beginning of the century, nano-sized particles clinically approved for other solid lesions were tested in these neoplasms but the results were anecdotal and the clinical benefit was not substantial. Recently, a new nanosystem formulation NBTXR3 for the treatment of sarcoma has landed in a phase 2-3 trial. The preliminary results are encouraging and could open new avenues for research in nanotechnology. This review provides an update on the recent advancements in the field of nanomedicine for sarcoma. In this regard, preclinical evidence especially focusing on the development of smart materials and drug delivery systems will be summarized. Moreover, the sarcoma patient management exploiting nanotechnology products will be summed up. Finally, an overlook on future perspectives will be provided.
Collapse
Affiliation(s)
- Laura Mercatali
- Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Silvia Vanni
- Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Giacomo Miserocchi
- Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Chiara Liverani
- Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Chiara Spadazzi
- Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Claudia Cocchi
- Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Chiara Calabrese
- Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Lorena Gurrieri
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Valentina Fausti
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Nada Riva
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Damiano Genovese
- Department of Chemistry “Giacomo Ciamician”, University of Bologna, Bologna, Italy
| | - Enrico Lucarelli
- Osteoncologia, Sarcomi dell’osso e dei tessuti molli, e Terapie Innovative, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | | | - Toni Ibrahim
- Osteoncologia, Sarcomi dell’osso e dei tessuti molli, e Terapie Innovative, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Luana Calabrò
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Alessandro De Vita
- Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- *Correspondence: Alessandro De Vita,
| |
Collapse
|
26
|
Amin M, Lammers T, Ten Hagen TLM. Temperature-sensitive polymers to promote heat-triggered drug release from liposomes: Towards bypassing EPR. Adv Drug Deliv Rev 2022; 189:114503. [PMID: 35998827 DOI: 10.1016/j.addr.2022.114503] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/14/2022] [Accepted: 08/17/2022] [Indexed: 01/24/2023]
Abstract
Heat-triggered drug release from temperature-sensitive nanocarriers upon the application of mild hyperthermia is a promising approach to achieve site-specific delivery of drugs. The combination of mild hyperthermia (41-42 °C) and temperature-sensitive liposomes (TSL) that undergo lipid phase-transition and drug release has been studied extensively and has shown promising therapeutic outcome in a variety of animal tumor models as well as initial indications of success in humans. Sensitization of liposomes to mild hyperthermia by means of exploiting the thermal behavior of temperature-sensitive polymers (TSP) provides novel opportunities. Recently, TSP-modified liposomes (TSPL) have shown potential for enhancing tumor-directed drug delivery, either by triggered drug release or by triggered cell interactions in response to heat. In this review, we describe different classes of TSPL, and analyze and discuss the mechanisms and kinetics of content release from TSPL in response to local heating. In addition, the impact of lipid composition, polymer and copolymer characteristics, serum components and PEGylation on the mechanism of content release and TSPL performance is addressed. This is done from the perspective of rationally designing TSPL, with the overall goal of conceiving efficient strategies to increase the efficacy of TSPL plus hyperthermia to improve the outcome of targeted anticancer therapy.
Collapse
Affiliation(s)
- Mohamadreza Amin
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands; Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Center for Biohybrid Medical Systems, Aachen, Germany.
| | - Timo L M Ten Hagen
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands; Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
27
|
Li ZH, Ma YJ, Jia ZH, Weng YY, Zhang P, Zhu SJ, Wang F. Meta-analysis of gemcitabine plus nab-paclitaxel combined with targeted agents in the treatment of metastatic pancreatic cancer. World J Clin Cases 2022; 10:9703-9713. [PMID: 36186177 PMCID: PMC9516936 DOI: 10.12998/wjcc.v10.i27.9703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 05/17/2022] [Accepted: 08/15/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Gemcitabine plus nab-paclitaxel (GA) is a commonly used first-line treatment regimen for metastatic pancreatic cancer, and many studies will add a novel targeted agent to this regimen for improving patient survival rate. However, the clinical effectiveness of GA is the most controversial issue.
AIM To compare the efficacy and safety of GA regimen with a targeted agent and GA regimen.
METHODS Up to 1 December 2021, the eligible randomized controlled trials (RCTs) relating to GA and GA with a targeted agent were searched on PubMed, EMBASE and Cochrane Library for eligible data. We screened out appropriate studies for overall survival (OS), progression-free survival (PFS), objective response rate (ORR), and toxicity, which had been pooled and finally analyzed by using Stata version 15.1. In addition, we use Reference Citation Analysis (https://www.referencecitationanalysis.com/) to collect the latest related literature to improve the latest cutting-edge research results.
RESULTS Seven RCTs involving 1544 patients (848 men and 696 women) were included. There were no significant differences between GA with a targeted agent and GA in PFS [hazard ratio (HR): 1.18 95% confidence interval (CI): 0.91-1.53], OS (HR: 1.12 95%CI: 0.99-1.27), and ORR (HR: 0.96 95%CI: 0.71-1.29). There was no notable difference in the two groups in grade 3/4 toxicity (fatigue, anemia, vomiting and neutropenia), whereas the incidence of grade 3/4 diarrhea considerably increased in GA with a targeted drug.
CONCLUSION Adding a novel targeted agent to the GA regimen did not improve survival rate of patients with metastatic pancreatic cancer.
Collapse
Affiliation(s)
- Zhong-Hui Li
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Yin-Jie Ma
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Zong-Hang Jia
- Department of Oncology, Shandong University of Traditional Chinese Medicine, Jinan 250022, Shandong Province, China
| | - Yue-Yan Weng
- Department of Acupuncture and Moxibustion, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Ping Zhang
- Department of Pathology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Shi-Jie Zhu
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Fang Wang
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| |
Collapse
|
28
|
Ghosh S, He X, Huang WC, Lovell JF. Immune checkpoint blockade enhances chemophototherapy in a syngeneic pancreatic tumor model. APL Bioeng 2022; 6:036105. [PMID: 36164594 PMCID: PMC9509203 DOI: 10.1063/5.0099811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/29/2022] [Indexed: 11/14/2022] Open
Abstract
Pancreatic cancer (PaCa) suffers from poor treatment options for locally advanced cases. Chemophototherapy (CPT) is an emerging anti-tumor modality, and porphyrin–phospholipid liposomes have been shown to be versatile drug carriers for CPT in preclinical rodent models. Here we show that in the syngeneic subcutaneous KPC PaCa tumor model, exhausted CD8+ T cells are localized in the tumor, and that CPT is enhanced in combination with immune checkpoint blockade (ICB). Addition of ICB using anti-programmed cell death 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) antibodies resulted in ablation of medium-sized, established KPC tumors (∼200 mm3) without recurrence for over 100 days. Mice rejected subsequent tumor re-challenge. Flow cytometry and tumor slice analysis following injection of a fluorescently labeled anti-PD-1 antibody showed that CPT improved antibody delivery to the tumor microenvironment. Treatment of large established tumors (∼400 mm3) using with CPT and ICB induced appreciable tumor regression and delay in regrowth. Taken together, these data demonstrate the utility of combining CPT with immunotherapies.
Collapse
Affiliation(s)
- Sanjana Ghosh
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York 14260, USA
| | - Xuedan He
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York 14260, USA
| | - Wei-Chiao Huang
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York 14260, USA
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York 14260, USA
| |
Collapse
|
29
|
Development of Irinotecan Liposome Armed with Dual-Target Anti-Epidermal Growth Factor Receptor and Anti-Fibroblast Activation Protein-Specific Antibody for Pancreatic Cancer Treatment. Pharmaceutics 2022; 14:pharmaceutics14061202. [PMID: 35745775 PMCID: PMC9227843 DOI: 10.3390/pharmaceutics14061202] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/27/2022] [Accepted: 06/03/2022] [Indexed: 01/27/2023] Open
Abstract
Pancreatic cancer is one of the most common causes of death in Taiwan. Previous studies have shown that more than 90% of pancreatic cancer cells presented epidermal growth factor receptor (EGFR) cell marker, and this marker is thought to be important as it is related to activation of cancer cell proliferation, angiogenesis, and cancer progression. Moreover, tumor-associated fibroblasts were involved in tumor proliferation and progression. In this study, we fabricated an anti-EGFR and anti-fibroblast activation protein bispecific antibody-targeted liposomal irinotecan (BS−LipoIRI), which could specifically bind to pancreatic cancer cells and tumor-associated fibroblasts. The drug encapsulation efficiency of BS−LipoIRI was 80.95%, and the drug loading was 8.41%. We proved that both pancreatic cancer cells and fibroblasts could be targeted by BS−LipoIRI, which showed better cellular uptake efficacy compared to LipoIRI. Furthermore, an in vivo mouse tumor test indicated that BS−LipoIRI could inhibit pancreatic cancer growth up to 46.2% compared to phosphate-buffered saline control, suggesting that BS−LipoIRI could be useful in clinical cancer treatment.
Collapse
|
30
|
Zhao YQ, Li LJ, Zhou EF, Wang JY, Wang Y, Guo LM, Zhang XX. Lipid-Based Nanocarrier Systems for Drug Delivery: Advances and Applications. PHARMACEUTICAL FRONTS 2022. [DOI: 10.1055/s-0042-1751036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
Lipid-based nanocarriers have been extensively investigated for drug delivery due to their advantages including biodegradability, biocompatibility, nontoxicity, and nonimmunogenicity. However, the shortcomings of traditional lipid-based nanocarriers such as insufficient targeting, capture by the reticuloendothelial system, and fast elimination limit the efficiency of drug delivery and therapeutic efficacy. Therefore, a series of multifunctional lipid-based nanocarriers have been developed to enhance the accumulation of drugs in the lesion site, aiming for improved diagnosis and treatment of various diseases. In this review, we summarized the advances and applications of lipid-based nanocarriers from traditional to novel functional lipid preparations, including liposomes, stimuli-responsive lipid-based nanocarriers, ionizable lipid nanoparticles, lipid hybrid nanocarriers, as well as biomembrane-camouflaged nanoparticles, and further discussed the challenges and prospects of this system. This exploration may give a complete idea viewing the lipid-based nanocarriers as a promising choice for drug delivery system, and fuel the advancement of pharmaceutical products by materials innovation and nanotechnology.
Collapse
Affiliation(s)
- Yan-Qi Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Li-Jun Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Er-Fen Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Jiang-Yue Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
- School of Pharmacy, East China University of Science and Technology, Shanghai, People's Republic of China
| | - Ying Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Lin-Miao Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Xin-Xin Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
| |
Collapse
|
31
|
Evaluating nanobiomaterial-induced DNA strand breaks using the alkaline comet assay. Drug Deliv Transl Res 2022; 12:2243-2258. [PMID: 35612707 PMCID: PMC9360128 DOI: 10.1007/s13346-022-01178-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2022] [Indexed: 11/12/2022]
Abstract
Due to their unique chemical and physical properties, nanobiomaterials (NBMs) are extensively studied for applications in medicine and drug delivery. Despite these exciting properties, their small sizes also make them susceptible to toxicity. Whilst nanomaterial immunotoxicity and cytotoxicity are studied in great depth, there is still limited data on their potential genotoxicity or ability to cause DNA damage. In the past years, new medical device regulations, which came into place in 2020, were developed, which require the assessment of long-term NBM exposure; therefore, in recent years, increased attention is being paid to genotoxicity screening of these materials. In this article, and through an interlaboratory comparison (ILC) study conducted within the Horizon 2020 REFINE project, we assess five different NBM formulations, each with different uses, namely, a bio-persistent gold nanoparticle (AuNP), an IR-780 dye-loaded liposome which is used in deep tissue imaging (LipImage™815), an unloaded PACA polymeric nanoparticle used as a drug delivery system (PACA), and two loaded PACA NBMs, i.e. the cabazitaxel drug-loaded PACA (CBZ-PACA) and the NR668 dye-loaded PACA (NR668 PACA) for their potential to cause DNA strand breaks using the alkaline comet assay and discuss the current state of genotoxicity testing for nanomaterials. We have found through our interlaboratory comparison that the alkaline comet assay can be suitably applied to the pre-clinical assessment of NBMs, as a reproducible and repeatable methodology for assessing NBM-induced DNA damage.
Collapse
|
32
|
Nsairat H, Khater D, Sayed U, Odeh F, Al Bawab A, Alshaer W. Liposomes: structure, composition, types, and clinical applications. Heliyon 2022; 8:e09394. [PMID: 35600452 PMCID: PMC9118483 DOI: 10.1016/j.heliyon.2022.e09394] [Citation(s) in RCA: 308] [Impact Index Per Article: 102.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/19/2022] [Accepted: 05/06/2022] [Indexed: 12/18/2022] Open
Abstract
Liposomes are now considered the most commonly used nanocarriers for various potentially active hydrophobic and hydrophilic molecules due to their high biocompatibility, biodegradability, and low immunogenicity. Liposomes also proved to enhance drug solubility and controlled distribution, as well as their capacity for surface modifications for targeted, prolonged, and sustained release. Based on the composition, liposomes can be considered to have evolved from conventional, long-circulating, targeted, and immune-liposomes to stimuli-responsive and actively targeted liposomes. Many liposomal-based drug delivery systems are currently clinically approved to treat several diseases, such as cancer, fungal and viral infections; more liposomes have reached advanced phases in clinical trials. This review describes liposomes structure, composition, preparation methods, and clinical applications.
Collapse
Affiliation(s)
- Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Dima Khater
- Department of Chemistry, Faculty of Arts and Science, Applied Science Private University, Amman, Jordan
| | - Usama Sayed
- Department of Biology, The University of Jordan, Amman, 11942, Jordan
| | - Fadwa Odeh
- Department of Chemistry, The University of Jordan, Amman, 11942, Jordan
| | - Abeer Al Bawab
- Department of Chemistry, The University of Jordan, Amman, 11942, Jordan.,Hamdi Mango Center for Scientific Research, The University of Jordan, Amman, 11942, Jordan
| | - Walhan Alshaer
- Cell Therapy Center, The University of Jordan, Amman, 11942, Jordan
| |
Collapse
|
33
|
Irinotecan-loaded ROS-responsive liposomes containing thioether phosphatidylcholine for improving anticancer activity. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
34
|
Ercin E, Kecel-Gunduz S, Gok B, Aydin T, Budama-Kilinc Y, Kartal M. Laurus nobilis L. Essential Oil-Loaded PLGA as a Nanoformulation Candidate for Cancer Treatment. Molecules 2022; 27:1899. [PMID: 35335262 PMCID: PMC8951774 DOI: 10.3390/molecules27061899] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/10/2022] [Accepted: 03/10/2022] [Indexed: 12/18/2022] Open
Abstract
The aim of this study was to obtain essential oil (LNEO) from the Laurus nobilis L. plant, and to prepare LNEO-loaded poly lactic-co-glycolic acid (PLGA) nanoparticles (NPs) as an approach in cancer treatment. The components of the obtained LNEO were analyzed using GC-MS. The LNEO-NPs were synthesized by the single-emulsion method. The LNEO-NPs were characterized using UV-Vis spectrometry, Dynamic Light Scattering (DLS), Scanning Electron Microscopy (SEM), and a DNA binding assay, which was performed via the UV-Vis titration method. According to the results, the LNEO-NPs had a 211.4 ± 4.031 nm average particle size, 0.068 ± 0.016 PdI, and -7.87 ± 1.15 mV zeta potential. The encapsulation efficiency and loading capacity were calculated as 59.25% and 25.65%, respectively, and the in vitro drug release study showed an LNEO release of 93.97 ± 3.78% over the 72 h period. Moreover, the LNEO was intercalatively bound to CT-DNA. In addition, the mechanism of action of LNEO on a dual PI3K/mTOR inhibitor was predicted, and its antiproliferative activity and mechanism were determined using molecular docking analysis. It was concluded that LNEO-loaded PLGA NPs may be used for cancer treatment as a novel phytotherapeutic agent-based controlled-release system.
Collapse
Affiliation(s)
- Esin Ercin
- Department of Pharmacognosy and Natural Product Chemistry, Institute of Health Sciences, Bezmialem Vakıf University, Istanbul 34093, Turkey; (E.E.); (T.A.)
| | - Serda Kecel-Gunduz
- Department of Physics, Faculty of Science, Istanbul University, Istanbul 34134, Turkey;
| | - Bahar Gok
- Department of Bioengineering, Graduate School of Natural and Applied Science, Yildiz Technical University, Istanbul 34220, Turkey;
| | - Tugba Aydin
- Department of Pharmacognosy and Natural Product Chemistry, Institute of Health Sciences, Bezmialem Vakıf University, Istanbul 34093, Turkey; (E.E.); (T.A.)
- Department of Pharmacognosy, Faculty of Pharmacy, Istinye University, Istanbul 34010, Turkey
| | - Yasemin Budama-Kilinc
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yildiz Technical University, Istanbul 34220, Turkey
| | - Murat Kartal
- Department of Pharmacognosy, Faculty of Pharmacy, Bezmialem Vakif University, Istanbul 34093, Turkey;
| |
Collapse
|
35
|
Álvarez-Benedicto E, Farbiak L, Márquez Ramírez M, Wang X, Johnson LT, Mian O, Guerrero ED, Siegwart DJ. Optimization of phospholipid chemistry for improved lipid nanoparticle (LNP) delivery of messenger RNA (mRNA). Biomater Sci 2022; 10:549-559. [PMID: 34904974 PMCID: PMC9113778 DOI: 10.1039/d1bm01454d] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Lipid nanoparticles (LNPs) have been established as an essential platform for nucleic acid delivery. Efforts have led to the development of vaccines that protect against SARS-CoV-2 infection using LNPs to deliver messenger RNA (mRNA) coding for the viral spike protein. Out of the four essential components that comprise LNPs, phospholipids represent an underappreciated opportunity for fundamental and translational study. We investigated this avenue by systematically modulating the identity of the phospholipid in LNPs with the goal of identifying specific moieties that directly enhance or hinder delivery efficacy. Results indicate that phospholipid chemistry can enhance mRNA delivery by increasing membrane fusion and enhancing endosomal escape. Phospholipids containing phosphoethanolamine (PE) head groups likely increase endosomal escape due to their fusogenic properties. Additionally, it was found that zwitterionic phospholipids mainly aided liver delivery, whereas negatively charged phospholipids changed the tropism of the LNPs from liver to spleen. These results demonstrate that the choice of phospholipid plays a role intracellularly by enhancing endosomal escape, while also driving organ tropism in vivo. These findings were then applied to Selective Organ Targeting (SORT) LNPs to manipulate and control spleen-specific delivery. Overall, selection of the phospholipid in LNPs provides an important handle to design and optimize LNPs for improved mRNA delivery and more effective therapeutics.
Collapse
Affiliation(s)
- Ester Álvarez-Benedicto
- Department of Biochemistry, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| | - Lukas Farbiak
- Department of Biochemistry, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| | - Martha Márquez Ramírez
- Department of Biochemistry, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| | - Xu Wang
- Department of Biochemistry, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| | - Lindsay T Johnson
- Department of Biochemistry, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| | - Osamah Mian
- Department of Biochemistry, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| | - Erick D Guerrero
- Department of Biochemistry, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| | - Daniel J Siegwart
- Department of Biochemistry, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| |
Collapse
|
36
|
Journey to the Market: The Evolution of Biodegradable Drug Delivery Systems. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12020935] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Biodegradable polymers have been used as carriers in drug delivery systems for more than four decades. Early work used crude natural materials for particle fabrication, whereas more recent work has utilized synthetic polymers. Applications include the macroscale, the microscale, and the nanoscale. Since pioneering work in the 1960’s, an array of products that use biodegradable polymers to encapsulate the desired drug payload have been approved for human use by international regulatory agencies. The commercial success of these products has led to further research in the field aimed at bringing forward new formulation types for improved delivery of various small molecule and biologic drugs. Here, we review recent advances in the development of these materials and we provide insight on their drug delivery application. We also address payload encapsulation and drug release mechanisms from biodegradable formulations and their application in approved therapeutic products.
Collapse
|
37
|
Turchin I, Bano S, Kirillin M, Orlova A, Perekatova V, Plekhanov V, Sergeeva E, Kurakina D, Khilov A, Kurnikov A, Subochev P, Shirmanova M, Komarova A, Yuzhakova D, Gavrina A, Mallidi S, Hasan T. Combined Fluorescence and Optoacoustic Imaging for Monitoring Treatments against CT26 Tumors with Photoactivatable Liposomes. Cancers (Basel) 2021; 14:197. [PMID: 35008362 PMCID: PMC8750546 DOI: 10.3390/cancers14010197] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/22/2021] [Accepted: 12/29/2021] [Indexed: 12/12/2022] Open
Abstract
The newly developed multimodal imaging system combining raster-scan optoacoustic (OA) microscopy and fluorescence (FL) wide-field imaging was used for characterizing the tumor vascular structure with 38/50 μm axial/transverse resolution and assessment of photosensitizer fluorescence kinetics during treatment with novel theranostic agents. A multifunctional photoactivatable multi-inhibitor liposomal (PMILs) nano platform was engineered here, containing a clinically approved photosensitizer, Benzoporphyrin derivative (BPD) in the bilayer, and topoisomerase I inhibitor, Irinotecan (IRI) in its inner core, for a synergetic therapeutic impact. The optimized PMIL was anionic, with the hydrodynamic diameter of 131.6 ± 2.1 nm and polydispersity index (PDI) of 0.05 ± 0.01, and the zeta potential between -14.9 ± 1.04 to -16.9 ± 0.92 mV. In the in vivo studies on BALB/c mice with CT26 tumors were performed to evaluate PMILs' therapeutic efficacy. PMILs demonstrated the best inhibitory effect of 97% on tumor growth compared to the treatment with BPD-PC containing liposomes (PALs), 81%, or IRI containing liposomes (L-[IRI]) alone, 50%. This confirms the release of IRI within the tumor cells upon PMILs triggering by NIR light, which is additionally illustrated by FL monitoring demonstrating enhancement of drug accumulation in tumor initiated by PDT in 24 h after the treatment. OA monitoring revealed the largest alterations of the tumor vascular structure in the PMILs treated mice as compared to BPD-PC or IRI treated mice. The results were further corroborated with histological data that also showed a 5-fold higher percentage of hemorrhages in PMIL treated mice compared to the control groups. Overall, these results suggest that multifunctional PMILs simultaneously delivering PDT and chemotherapy agents along with OA and FL multi-modal imaging offers an efficient and personalized image-guided platform to improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Ilya Turchin
- Institute of Applied Physics RAS, 46 Ulyanov St., 603950 Nizhny Novgorod, Russia; (M.K.); (A.O.); (V.P.); (V.P.); (E.S.); (D.K.); (A.K.); (A.K.); (P.S.)
| | - Shazia Bano
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; (S.B.); (S.M.); (T.H.)
| | - Mikhail Kirillin
- Institute of Applied Physics RAS, 46 Ulyanov St., 603950 Nizhny Novgorod, Russia; (M.K.); (A.O.); (V.P.); (V.P.); (E.S.); (D.K.); (A.K.); (A.K.); (P.S.)
| | - Anna Orlova
- Institute of Applied Physics RAS, 46 Ulyanov St., 603950 Nizhny Novgorod, Russia; (M.K.); (A.O.); (V.P.); (V.P.); (E.S.); (D.K.); (A.K.); (A.K.); (P.S.)
| | - Valeriya Perekatova
- Institute of Applied Physics RAS, 46 Ulyanov St., 603950 Nizhny Novgorod, Russia; (M.K.); (A.O.); (V.P.); (V.P.); (E.S.); (D.K.); (A.K.); (A.K.); (P.S.)
| | - Vladimir Plekhanov
- Institute of Applied Physics RAS, 46 Ulyanov St., 603950 Nizhny Novgorod, Russia; (M.K.); (A.O.); (V.P.); (V.P.); (E.S.); (D.K.); (A.K.); (A.K.); (P.S.)
| | - Ekaterina Sergeeva
- Institute of Applied Physics RAS, 46 Ulyanov St., 603950 Nizhny Novgorod, Russia; (M.K.); (A.O.); (V.P.); (V.P.); (E.S.); (D.K.); (A.K.); (A.K.); (P.S.)
| | - Daria Kurakina
- Institute of Applied Physics RAS, 46 Ulyanov St., 603950 Nizhny Novgorod, Russia; (M.K.); (A.O.); (V.P.); (V.P.); (E.S.); (D.K.); (A.K.); (A.K.); (P.S.)
| | - Aleksandr Khilov
- Institute of Applied Physics RAS, 46 Ulyanov St., 603950 Nizhny Novgorod, Russia; (M.K.); (A.O.); (V.P.); (V.P.); (E.S.); (D.K.); (A.K.); (A.K.); (P.S.)
| | - Alexey Kurnikov
- Institute of Applied Physics RAS, 46 Ulyanov St., 603950 Nizhny Novgorod, Russia; (M.K.); (A.O.); (V.P.); (V.P.); (E.S.); (D.K.); (A.K.); (A.K.); (P.S.)
| | - Pavel Subochev
- Institute of Applied Physics RAS, 46 Ulyanov St., 603950 Nizhny Novgorod, Russia; (M.K.); (A.O.); (V.P.); (V.P.); (E.S.); (D.K.); (A.K.); (A.K.); (P.S.)
| | - Marina Shirmanova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (M.S.); (A.K.); (D.Y.); (A.G.)
| | - Anastasiya Komarova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (M.S.); (A.K.); (D.Y.); (A.G.)
| | - Diana Yuzhakova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (M.S.); (A.K.); (D.Y.); (A.G.)
| | - Alena Gavrina
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (M.S.); (A.K.); (D.Y.); (A.G.)
| | - Srivalleesha Mallidi
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; (S.B.); (S.M.); (T.H.)
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; (S.B.); (S.M.); (T.H.)
- Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
38
|
Ghosh S, Lovell JF. Two Laser Treatments Can Improve Tumor Ablation Efficiency of Chemophototherapy. Pharmaceutics 2021; 13:pharmaceutics13122183. [PMID: 34959464 PMCID: PMC8704214 DOI: 10.3390/pharmaceutics13122183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 11/16/2022] Open
Abstract
Chemophototherapy is an emerging tumor ablation modality that can improve local delivery of chemotherapeutic agents. Long circulating doxorubicin (Dox) in porphyrin-phospholipid (PoP) liposomes (LC-Dox-PoP) has previously been developed as an effective chemophototherapy agent. In the present study, we observed that in mice, LC-Dox-PoP showed enhanced accumulation in human pancreatic tumor xenografts even with suboptimal light doses, as assessed by fluorometric analysis of tissue homogenates and microscopic imaging of Dox and PoP in tumor slices. A second laser treatment, at a time point in which tumors had greater drug accumulation as a result of the first laser treatment, induced potent tumor ablation. Efficacy studies were carried out in two human pancreatic cancer subcutaneous mouse tumor models; MIA PaCa-2 or low-passage patient derived pancreatic cancer xenografts. A single treatment of 3 mg/kg LC-Dox-PoP and an initial 150 J/cm2 laser treatment 1 h after drug administration, followed by second laser treatment of 50 J/cm2 8 h after drug administration, was more effective than a single laser treatment of 200 J/cm2 at either of those time points. Thus, this study presents proof-of-principle and rationale for using two discrete laser treatments to enhance the efficacy of chemophototherapy.
Collapse
|
39
|
Kumar N, Fazal S, Miyako E, Matsumura K, Rajan R. Avengers against cancer: A new era of nano-biomaterial-based therapeutics. MATERIALS TODAY 2021; 51:317-349. [DOI: 10.1016/j.mattod.2021.09.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
40
|
The Evolution and Future of Targeted Cancer Therapy: From Nanoparticles, Oncolytic Viruses, and Oncolytic Bacteria to the Treatment of Solid Tumors. NANOMATERIALS 2021; 11:nano11113018. [PMID: 34835785 PMCID: PMC8623458 DOI: 10.3390/nano11113018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 02/07/2023]
Abstract
While many classes of chemotherapeutic agents exist to treat solid tumors, few can generate a lasting response without substantial off-target toxicity despite significant scientific advancements and investments. In this review, the paths of development for nanoparticles, oncolytic viruses, and oncolytic bacteria over the last 20 years of research towards clinical translation and acceptance as novel cancer therapeutics are compared. Novel nanoparticle, oncolytic virus, and oncolytic bacteria therapies all start with a common goal of accomplishing therapeutic drug activity or delivery to a specific site while avoiding off-target effects, with overlapping methodology between all three modalities. Indeed, the degree of overlap is substantial enough that breakthroughs in one therapeutic could have considerable implications on the progression of the other two. Each oncotherapeutic modality has accomplished clinical translation, successfully overcoming the potential pitfalls promising therapeutics face. However, once studies enter clinical trials, the data all but disappears, leaving pre-clinical researchers largely in the dark. Overall, the creativity, flexibility, and innovation of these modalities for solid tumor treatments are greatly encouraging, and usher in a new age of pharmaceutical development.
Collapse
|
41
|
Ali N, Srivastava N. Recent Advancements for the Management of Pancreatic Cancer: Current Insights. CURRENT CANCER THERAPY REVIEWS 2021. [DOI: 10.2174/1573394717666210625153256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
One of the most fatal forms of cancer includes cancer of the pancreas And the most
rapid malignancy is observed in PDAC (pancreatic ductal adenocarcinoma). The high lethality rate
is generally due to very late diagnosis and resistance to traditional chemotherapeutic agents. Desmoplastic
stromal barrier results in resistance to immunotherapy. Other reasons for the high lethality
rate include the absence of effective treatment and standard screening tests. Hence, there is a
need for effective novel carrier systems. “A formulation, method, or device that allows the desired
therapeutic substance to reach its site of action in such a manner that nontarget cells experience
minimum effect is referred to as a drug delivery system”. The delivery system is responsible for introducing
the active component into the body. They are also liable for boosting the efficacy and desirable
targeted action on the tumorous tissues. Several studies, researches, and developments have
yielded various advanced drug delivery systems, which include liposomes, nanoparticles, carbon
nanotubules, renovoCath, etc. These systems control rate and location of the release. They are designed
while taking into consideration characteristic properties of the tumor and tumor stroma. These
delivery systems overcome the barriers in drug deliverance in pancreatic cancer. Alongside providing
palliative benefits, these delivery systems also aim to correct the underlying reason for the
defect. The following review article aims and focuses to bring out a brief idea about systems, methods,
and technologies for futuristic drug deliverance in pancreatic cancer therapy.
Collapse
Affiliation(s)
- Naureen Ali
- Amity Institute of Pharmacy, Amity University, Uttar Pradesh, Lucknow Campus, Lucknow,India
| | - Nimisha Srivastava
- Amity Institute of Pharmacy, Amity University, Uttar Pradesh, Lucknow Campus, Lucknow,India
| |
Collapse
|
42
|
Wang T, He W, Du Y, Wang J, Li X. Redox-sensitive irinotecan liposomes with active ultra-high loading and enhanced intracellular drug release. Colloids Surf B Biointerfaces 2021; 206:111967. [PMID: 34256270 DOI: 10.1016/j.colsurfb.2021.111967] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/01/2021] [Accepted: 07/04/2021] [Indexed: 01/21/2023]
Abstract
In this report, a novel irinotecan (IR) encapsulated redox-responsive liposome was developed. The redox-responsive liposomes were prepared based on disulfide phosphatidylcholine (SS-PC), DSPC, DSPE-PEG2000 and cholesterol by ethanol injection method. IR was actively loaded by triethylammonium sucrose octasulfate (TEA8-SOS) gradient method to generate IR/SS-PC liposomes (IR/SS-LP). The particle size of IR/SS-PC was characterized by using dynamic light scattering (DLS) and transmission electron microscopy (TEM). It was found that IR/SS-LP with 30 % content of SS-PC (IR/SS30-LP) had an average size of 125.5 ± 5.8 nm with a negative zeta potential of -19.5 ± 0.1. The encapsulation efficiency (EE) was further determined to be 98.1 ± 0.8 % and drug loading (DL) was 31.8 ± 0.1 %. The redox-responsiveness of IR/SS-LP was investigated by observing the change of particle size and morphology as well as the release behavior of IR triggered by glutathione (GSH). The data indicated GSH breaks the disulfide bonds in SS-PC and leads to the controlled release of IR. At 1 mM GSH, 60.2 % irinotecan was released from IR/SS30-LP within 24 h. Finally, the effects of IR/SS-LP in cell and animal experiments were evaluated in detail. The results showed that IR/SS30-LP had superior pharmacokinetic and antitumor efficacy compared to free irinotecan and traditional irinotecan liposome (ONIVYDE®-like). Taken together, IR/SS30-LP displayed redox-responsive release of IR, ultra-high loading and enhanced anti-tumor activity, which has great potential for clinical application as a new generation of IR liposomal formulation.
Collapse
Affiliation(s)
- Tao Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, PR China
| | - Wei He
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, PR China
| | - Yawei Du
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, PR China
| | - Ji Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, PR China
| | - Xinsong Li
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, PR China.
| |
Collapse
|
43
|
Dimitriou P, Li J, Tornillo G, McCloy T, Barrow D. Droplet Microfluidics for Tumor Drug-Related Studies and Programmable Artificial Cells. GLOBAL CHALLENGES (HOBOKEN, NJ) 2021; 5:2000123. [PMID: 34267927 PMCID: PMC8272004 DOI: 10.1002/gch2.202000123] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/19/2021] [Indexed: 05/11/2023]
Abstract
Anticancer drug development is a crucial step toward cancer treatment, that requires realistic predictions of malignant tissue development and sophisticated drug delivery. Tumors often acquire drug resistance and drug efficacy, hence cannot be accurately predicted in 2D tumor cell cultures. On the other hand, 3D cultures, including multicellular tumor spheroids (MCTSs), mimic the in vivo cellular arrangement and provide robust platforms for drug testing when grown in hydrogels with characteristics similar to the living body. Microparticles and liposomes are considered smart drug delivery vehicles, are able to target cancerous tissue, and can release entrapped drugs on demand. Microfluidics serve as a high-throughput tool for reproducible, flexible, and automated production of droplet-based microscale constructs, tailored to the desired final application. In this review, it is described how natural hydrogels in combination with droplet microfluidics can generate MCTSs, and the use of microfluidics to produce tumor targeting microparticles and liposomes. One of the highlights of the review documents the use of the bottom-up construction methodologies of synthetic biology for the formation of artificial cellular assemblies, which may additionally incorporate both target cancer cells and prospective drug candidates, as an integrated "droplet incubator" drug assay platform.
Collapse
Affiliation(s)
- Pantelitsa Dimitriou
- Applied Microfluidic LaboratorySchool of EngineeringCardiff UniversityCardiffCF24 3AAUK
| | - Jin Li
- Applied Microfluidic LaboratorySchool of EngineeringCardiff UniversityCardiffCF24 3AAUK
| | - Giusy Tornillo
- Hadyn Ellis BuildingCardiff UniversityMaindy RoadCardiffCF24 4HQUK
| | - Thomas McCloy
- Applied Microfluidic LaboratorySchool of EngineeringCardiff UniversityCardiffCF24 3AAUK
| | - David Barrow
- Applied Microfluidic LaboratorySchool of EngineeringCardiff UniversityCardiffCF24 3AAUK
| |
Collapse
|
44
|
Khaiwa N, Maarouf NR, Darwish MH, Alhamad DWM, Sebastian A, Hamad M, Omar HA, Orive G, Al-Tel TH. Camptothecin's journey from discovery to WHO Essential Medicine: Fifty years of promise. Eur J Med Chem 2021; 223:113639. [PMID: 34175539 DOI: 10.1016/j.ejmech.2021.113639] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/24/2021] [Accepted: 06/06/2021] [Indexed: 12/16/2022]
Abstract
Nature represents a rich source of compounds used for the treatment of many diseases. Camptothecin (CPT), isolated from the bark of Camptotheca acuminata, is a cytotoxic alkaloid that attenuates cancer cell replication by inhibiting DNA topoisomerase 1. Despite its promising and wide spectrum antiproliferative activity, its use is limited due to low solubility, instability, acquired tumour cell resistance, and remarkable toxicity. This has led to the development of numerous CPT analogues with improved pharmacodynamic and pharmacokinetic profiles. Three natural product-inspired drugs, namely, topotecan, irinotecan, and belotecan, are clinically approved and prescribed drugs for the treatment of several types of cancer, whereas other derivatives are in clinical trials. In this review, which covers literature from 2015 to 2020, we aim to provide a comprehensive overview and describe efforts that led to the development of a variety of CPT analogues. These efforts have led to the discovery of potent, first-in-class chemotherapeutic agents inspired by CPT. In addition, the mechanism of action, SAR studies, and recent advances of novel CPT drug delivery systems and antibody drug conjugates are discussed.
Collapse
Affiliation(s)
- Noura Khaiwa
- College of Pharmacy, University of Sharjah, 27272, Sharjah, United Arab Emirates
| | - Noor R Maarouf
- College of Pharmacy, University of Sharjah, 27272, Sharjah, United Arab Emirates
| | - Mhd H Darwish
- College of Pharmacy, University of Sharjah, 27272, Sharjah, United Arab Emirates
| | - Dima W M Alhamad
- Sharjah Institute for Medical Research, 27272, Sharjah, United Arab Emirates
| | - Anusha Sebastian
- Sharjah Institute for Medical Research, 27272, Sharjah, United Arab Emirates
| | - Mohamad Hamad
- Sharjah Institute for Medical Research, 27272, Sharjah, United Arab Emirates; College of Health Sciences, 27272, Sharjah, United Arab Emirates
| | - Hany A Omar
- College of Pharmacy, University of Sharjah, 27272, Sharjah, United Arab Emirates; Sharjah Institute for Medical Research, 27272, Sharjah, United Arab Emirates
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain
| | - Taleb H Al-Tel
- College of Pharmacy, University of Sharjah, 27272, Sharjah, United Arab Emirates; Sharjah Institute for Medical Research, 27272, Sharjah, United Arab Emirates.
| |
Collapse
|
45
|
Liu S, Khan AR, Yang X, Dong B, Ji J, Zhai G. The reversal of chemotherapy-induced multidrug resistance by nanomedicine for cancer therapy. J Control Release 2021; 335:1-20. [PMID: 33991600 DOI: 10.1016/j.jconrel.2021.05.012] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 05/08/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022]
Abstract
Multidrug resistance (MDR) of cancer is a persistent problem in chemotherapy. Scientists have considered the overexpressed efflux transporters responsible for MDR and chemotherapy failure. MDR extremely limits the therapeutic effect of chemotherapy in cancer treatment. Many strategies have been applied to solve this problem. Multifunctional nanoparticles may be one of the most promising approaches to reverse MDR of tumor. These nanoparticles can keep stability in the blood circulation and selectively accumulated in the tumor microenvironment (TME) either by passive or active targeting. The stimuli-sensitive or organelle-targeting nanoparticles can release the drug at the targeted-site without exposure to normal tissues. In order to better understand reversal of MDR, three main strategies are concluded in this review. First strategy is the synergistic effect of chemotherapeutic drugs and ABC transporter inhibitors. Through directly inhibiting overexpressed ABC transporters, chemotherapeutic drugs can enter into resistant cells without being efflux. Second strategy is based on nanoparticles circumventing over-expressed efflux transporters and directly targeting resistance-related organelles. Third approach is the combination of multiple therapy modes overcoming cancer resistance. At last, numerous researches demonstrated cancer stem-like cells (CSCs) had a deep relation with drug resistance. Here, we discuss two different drug delivery approaches of nanomedicine based on CSC therapy.
Collapse
Affiliation(s)
- Shangui Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, PR China
| | - Abdur Rauf Khan
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, PR China
| | - Xiaoye Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, PR China
| | - Bo Dong
- Department of cardiovascular medicine, Shandong Provincial Hospital, Jinan 250021, PR China
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, PR China
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, PR China.
| |
Collapse
|
46
|
Lin WW, Cheng YA, Li CC, Ho KW, Chen HJ, Chen IJU, Huang BC, Liu HJ, Lu YC, Cheng CM, Huang MY, Lai HW, Cheng TL. Enhancement of tumor tropism of mPEGylated nanoparticles by anti-mPEG bispecific antibody for ovarian cancer therapy. Sci Rep 2021; 11:7598. [PMID: 33828191 PMCID: PMC8027450 DOI: 10.1038/s41598-021-87271-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 03/22/2021] [Indexed: 12/16/2022] Open
Abstract
Ovarian cancer is highly metastatic, with a high frequency of relapse, and is the most fatal gynecologic malignancy in women worldwide. It is important to elevate the drug susceptibility and cytotoxicity of ovarian cancer cells, thereby eliminating resident cancer cells for more effective therapeutic efficacy. Here, we developed a bispecific antibody (BsAb; mPEG × HER2) that can easily provide HER2+ tumor tropism to mPEGylated liposomal doxorubicin (PLD) and further increase the drug accumulation in cancer cells via receptor-mediated endocytosis, and improve the cytotoxicity and therapeutic efficacy of HER2+ ovarian tumors. The mPEG × HER2 can simultaneously bind to mPEG molecules on the surface of PLD and HER2 antigen on the surface of ovarian cancer cells. Simply mixing the mPEG × HER2 with PLD was able to confer HER2 specificity of PLD to HER2+ ovarian cancer cells and efficiently trigger endocytosis and enhance cytotoxicity by 5.4-fold as compared to non-targeted PLD. mPEG × HER2-modified PLD was able to significantly increase the targeting and accumulation of HER2+ ovarian tumor by 220% as compared with non-targeted PLD. It could also significantly improve the anti-tumor activity of PLD (P < 0.05) with minimal obvious toxicity in a tumor-bearing mouse model. We believe that the mPEG × HER2 can significantly improve the therapeutic efficacy, potentially reduce the relapse freqency and thereby achieve good prognosis in ovarian cancer patients.
Collapse
Affiliation(s)
- Wen-Wei Lin
- Department of Laboratory Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Laboratory Medicine, Post Baccalaureat Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-An Cheng
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, 100 Shih-Chuan 1st Road, Kaohsiung, 80708, Taiwan
| | - Chia-Ching Li
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, 100 Shih-Chuan 1st Road, Kaohsiung, 80708, Taiwan
| | - Kai-Wen Ho
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Huei-Jen Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - I-J U Chen
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, 100 Shih-Chuan 1st Road, Kaohsiung, 80708, Taiwan
| | - Bo-Cheng Huang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Hui-Ju Liu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yun-Chi Lu
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, 100 Shih-Chuan 1st Road, Kaohsiung, 80708, Taiwan
| | - Chiu-Min Cheng
- Department of Aquaculture, National Kaohsiung University of Science and Technology, Kaohsiung, Taiwan
| | - Ming-Yii Huang
- Department of Radiation Oncology, Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Hung-Wen Lai
- Endoscopic and Oncoplastic Breast Surgery Center, Comprehensive Breast Cancer Center, Changhua Christian Hospital, 135 Nanxiao Street, Changhua, 500, Taiwan. .,Division of General Surgery, Changhua Christian Hospital, Changhua, Taiwan. .,Comprehensive Breast Cancer Center, Changhua Christian Hospital, Changhua, Taiwan. .,Minimal Invasive Surgery Research Center, Changhua Christian Hospital, Changhua, Taiwan.
| | - Tian-Lu Cheng
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan. .,Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan. .,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan. .,Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, 100 Shih-Chuan 1st Road, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
47
|
Janjua TI, Rewatkar P, Ahmed-Cox A, Saeed I, Mansfeld FM, Kulshreshtha R, Kumeria T, Ziegler DS, Kavallaris M, Mazzieri R, Popat A. Frontiers in the treatment of glioblastoma: Past, present and emerging. Adv Drug Deliv Rev 2021; 171:108-138. [PMID: 33486006 DOI: 10.1016/j.addr.2021.01.012] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/13/2020] [Accepted: 01/09/2021] [Indexed: 12/13/2022]
Abstract
Glioblastoma (GBM) is one of the most aggressive cancers of the brain. Despite extensive research over the last several decades, the survival rates for GBM have not improved and prognosis remains poor. To date, only a few therapies are approved for the treatment of GBM with the main reasons being: 1) significant tumour heterogeneity which promotes the selection of resistant subpopulations 2) GBM induced immunosuppression and 3) fortified location of the tumour in the brain which hinders the delivery of therapeutics. Existing therapies for GBM such as radiotherapy, surgery and chemotherapy have been unable to reach the clinical efficacy necessary to prolong patient survival more than a few months. This comprehensive review evaluates the current and emerging therapies including those in clinical trials that may potentially improve both targeted delivery of therapeutics directly to the tumour site and the development of agents that may specifically target GBM. Particular focus has also been given to emerging delivery technologies such as focused ultrasound, cellular delivery systems nanomedicines and immunotherapy. Finally, we discuss the importance of developing novel materials for improved delivery efficacy of nanoparticles and therapeutics to reduce the suffering of GBM patients.
Collapse
|
48
|
Hartman SJ, Bagby SM, Yacob BW, Simmons DM, MacBeth M, Lieu CH, Davis SL, Leal AD, Tentler JJ, Diamond JR, Eckhardt SG, Messersmith WA, Pitts TM. WEE1 Inhibition in Combination With Targeted Agents and Standard Chemotherapy in Preclinical Models of Pancreatic Ductal Adenocarcinoma. Front Oncol 2021; 11:642328. [PMID: 33869031 PMCID: PMC8044903 DOI: 10.3389/fonc.2021.642328] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/09/2021] [Indexed: 12/15/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal cancer with high incidences of p53 mutations. AZD1775 (adavosertib, previously MK-1775) is a small molecule WEE1 inhibitor that abrogates the G2M checkpoint and can potentially synergize with DNA damaging therapies commonly used in PDAC treatment. The purpose of this study was to identify combination partners for AZD1775, including standard chemotherapy or targeted agents, in PDAC preclinical models. Low powered preliminary screens demonstrated that two of the four PDX models responded better to the combinations of AZD1775 with irinotecan or capecitabine than to either single agent. Following the screens, two full powered PDAC PDX models of differing p53 status were tested with the combinations of AZD1775 and irinotecan or capecitabine. The combinations of AZD1775 and SN38 or 5-FU were also tested on PDAC cell lines. Cellular proliferation was measured using an IncuCyte Live Cell Imager and apoptosis was measured using a Caspase-Glo 3/7 assay. Flow cytometry was conducted to measure alterations in cell cycle distribution. Western blot analysis was used to determine the effects of the drug combinations on downstream effectors. In PDX models with mutated p53 status, there was significant tumor growth inhibition from the combination of AZD1775 with irinotecan or capecitabine (P ≤ 0.03), while PDX models with wild type p53 did not show anti-tumor synergy from the same combinations (P ≥ 0.08). The combination of AZD1775 with SN38 or 5-FU significantly decreased proliferation in all PDAC cell lines, and enhanced apoptosis in multiple cell lines. Cell cycle distribution was disrupted from the combination of AZD1775 with SN38 or 5-FU which was recorded as G2M arrest and decreased G1 phase. AZD1775 inhibited phospho-CDC2 and increased the expression of γH2AX that was either maintained or enhanced after combination with SN38 or 5-FU. The combination of AZD1775 with irinotecan/SN38 or capecitabine/5-FU showed anti-tumor effects in vivo and in vitro in PDAC models. These results support further investigation for these combination strategies to enhance outcomes for PDAC patients.
Collapse
Affiliation(s)
- Sarah J Hartman
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Stacey M Bagby
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Betelehem W Yacob
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Dennis M Simmons
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Morgan MacBeth
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Christopher H Lieu
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - S Lindsey Davis
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Alexis D Leal
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - John J Tentler
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jennifer R Diamond
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - S Gail Eckhardt
- Department of Oncology, Dell Medical School, The University of Texas Austin, Austin, TX, United States
| | - Wells A Messersmith
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Todd M Pitts
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
49
|
Zhang Z, Song J, Xie C, Pan J, Lu W, Liu M. Pancreatic Cancer: Recent Progress of Drugs in Clinical Trials. AAPS JOURNAL 2021; 23:29. [PMID: 33580411 DOI: 10.1208/s12248-021-00556-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 01/06/2021] [Indexed: 12/18/2022]
Abstract
Pancreatic cancer is a highly malignant tumor and one of the primary causes of cancer-related death. Because pancreatic cancer is difficult to diagnose in the early course of the disease, most patients present with advanced lesions at the time of diagnosis, and only 20% of patients are eligible for surgery. Consequently, drug treatment has become extremely important. At present, the main treatment regimens for pancreatic cancer are gemcitabine and the FORFIRINOX and MPACT regimens. However, none of these regimens substantially improves the prognosis of patients with pancreatic cancer. Extensive efforts have been dedicated to the study of pancreatic cancer in recent years. With the development and clinical application of biological targeted drugs, the biological targeted treatment of tumors has been widely accepted. Therefore, this article used relevant clinical trial data to summarize the research progress of traditional chemotherapy drugs and biological targeted drugs for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Zhiyi Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, People's Republic of China
| | - Jie Song
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, People's Republic of China
| | - Cao Xie
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, People's Republic of China
| | - Jun Pan
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, People's Republic of China
| | - Weiyue Lu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, People's Republic of China
| | - Min Liu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, People's Republic of China.
| |
Collapse
|
50
|
Nanomedicines accessible in the market for clinical interventions. J Control Release 2021; 330:372-397. [DOI: 10.1016/j.jconrel.2020.12.034] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 12/17/2020] [Accepted: 12/17/2020] [Indexed: 02/06/2023]
|