1
|
Azimizonuzi H, Ghayourvahdat A, Ahmed MH, Kareem RA, Zrzor AJ, Mansoor AS, Athab ZH, Kalavi S. A state-of-the-art review of the recent advances of theranostic liposome hybrid nanoparticles in cancer treatment and diagnosis. Cancer Cell Int 2025; 25:26. [PMID: 39871316 PMCID: PMC11773959 DOI: 10.1186/s12935-024-03610-z] [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: 08/19/2024] [Accepted: 12/10/2024] [Indexed: 01/29/2025] Open
Abstract
Theranostics is a way of treating illness that blends medicine with testing. Specific characteristics should be present in the best theranostic agents for cancer: (1) the drugs should be safe and non-toxic; (2) they should be able to treat cancer selectively; and (3) they should be able to build up only in the cancerous tissue. Liposomes (LPs) are one of the most efficient drug delivery methods based on nanotechnology. Stealth LPs and commercial LPs have recently had an impact on cancer treatment. Using the valuable information from each imaging technique, along with the multimodality imaging functionality of liposomal therapeutic agents, makes them very appealing for personalized monitoring of how well therapeutic drugs are working against cancer in vivo and for predicting how well therapies will work. On the other hand, their use as nanoparticle delivery systems is currently in the research and development phase. Nanoscale delivery system innovation has made LP-nanoparticle hybrid structures very useful for combining therapeutic and imaging methods. LP-hybrid nanoparticles are better at killing cancer cells than their LP counterparts, making them excellent options for in vivo and in vitro drug delivery applications. Hybrid liposomes (HLs) could be used in the future as theranostic carriers to find and treat cancer targets. This would combine the best features of synthetic and biological drug delivery systems. Overarchingly, this article provided a comprehensive overview of the many LP types used in cancer detection, therapy, and theranostic analysis. An evaluation of the pros and cons of the many HLs types used in cancer detection and treatment has also been conducted. The study also included recent and significant research on HLs for cancer theranostic applications. We conclude by outlining the potential benefits and drawbacks of this theranostic approach to the concurrent detection and treatment of different malignancies, as well as its prospects.
Collapse
Affiliation(s)
- Hannaneh Azimizonuzi
- Inventor Member of International Federation of Inventors Associations, Geneva, Switzerland
| | - Arman Ghayourvahdat
- Inventor Member of International Federation of Inventors Associations, Geneva, Switzerland
| | | | | | - Athmar Jaber Zrzor
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Shaylan Kalavi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Islamic Azad University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Al-Baidhani SAS, Pouresmaeil V, Homayouni Tabrizi M. Synthesis of liposomal nanoparticles to load 4-farnesyloxycoumarin and investigating its anti-cancer and anti-metastatic effects. J Liposome Res 2024:1-10. [PMID: 39550704 DOI: 10.1080/08982104.2024.2428168] [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: 06/25/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 11/18/2024]
Abstract
The aim of this study was to load 4-farnesyloxycoumarin (4-FLC) in nanoliposomes (4-FLC-LNPs) and evaluate its anti-cancer and anti-metastatic effects. 4-FLC-LNPs were synthesized using a combination of lecithin-cholesterol-polyethylene glycol. The physicochemical properties were evaluated using DLS, FTIR, and microscopy methods. The toxicity against breast cancer (MCF-7), prostate cancer (PS3), pancreatic cancer (PANC), gastric cancer (AGS), and normal cell lines (HUVEC) was evaluated using the MTT assay. Fluorescent staining and flow cytometry were used to assess the occurrence of apoptosis. Molecular analysis methods were used to study the apoptosis and metastasis effects of these nanoliposomes. The antioxidant power of 4-FLC-LNPs was measured using the ABTS and DPPH free radicals methods. 4-FLC-LNPs exhibit a spherical morphology, with an average size of 57.43 nm, a polydispersity index of 0.29, and a zeta potential of -31.4 mV. They demonstrate an encapsulation efficiency of 82.4% for 4-FLC. The IC50 value of 4-FLC-LNPs against the breast cancer cell line was reported as the most sensitive, at approximately 60 μg/mL. ABTS and DPPH results were reported at approximately 30 µg/mL. The inductive effects of nanoliposomes on the apoptosis process were confirmed by an increase in the number of apoptotic cells, as well as the arrest of cells in various phases of cell growth. The increased expression of BAX and decreased expression of Bcl-2, MMP-2, and MMP-9 confirmed the pro-apoptotic and anti-metastatic effects of 4-FLC-LNPs. These finding validate the therapeutic potential of 4-FLC-LNPs, which may be utilized in preclinical studies.
Collapse
Affiliation(s)
| | - Vahid Pouresmaeil
- Department of Biochemistry, Faculty of Medicine, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran
| | | |
Collapse
|
3
|
Saladino GM, Chao PH, Brodin B, Li SD, Hertz HM. Liposome biodistribution mapping with in vivo X-ray fluorescence imaging. NANOSCALE 2024; 16:17404-17411. [PMID: 39212620 DOI: 10.1039/d4nr02793k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Lipid-based nanoparticles are organic nanostructures constituted of phospholipids and cholesterol, displaying high in vivo biocompatibility. They have been demonstrated as effective nanocarriers for drug delivery and targeting. Mapping liposome distribution is crucial as it enables a precise understanding of delivery kinetics, tissue targeting efficiency, and potential off-target effects. Recently, ruthenium-encapsulated liposomes have shown potential for targeted drug delivery, photodynamic therapy, and optical fluorescence imaging. In the present work, we design Ru(bpy)3-encapsulated liposomes (Ru-Lipo) empowering optical and X-ray fluorescence (XRF) properties for dual mode imaging and demonstrate the passivation role of liposomes over the free Ru(bpy)3 compound. We employ whole-body XRF imaging to map the in vivo biodistribution of Ru-Lipo in mice, enabling tumor detection and longitudinal studies with elemental specificity and resolution down to the sub-millimeter scale. Quantitative XRF computed tomography on extracted organs permits targeting efficiency evaluations. These findings highlight the promising role of XRF imaging in pharmacokinetic studies and theranostic applications for the rapid optimization of drug delivery and assessment of targeting efficiency.
Collapse
Affiliation(s)
- Giovanni Marco Saladino
- Department of Applied Physics, Bio-Opto-Nano Physics, KTH Royal Institute of Technology, SE 10691, Stockholm, Sweden.
- Faculty of Pharmaceutical Sciences, University of British Columbia, V6T 1Z3, Vancouver, British Columbia, Canada
| | - Po-Han Chao
- Faculty of Pharmaceutical Sciences, University of British Columbia, V6T 1Z3, Vancouver, British Columbia, Canada
| | - Bertha Brodin
- Department of Applied Physics, Bio-Opto-Nano Physics, KTH Royal Institute of Technology, SE 10691, Stockholm, Sweden.
| | - Shyh-Dar Li
- Faculty of Pharmaceutical Sciences, University of British Columbia, V6T 1Z3, Vancouver, British Columbia, Canada
| | - Hans Martin Hertz
- Department of Applied Physics, Bio-Opto-Nano Physics, KTH Royal Institute of Technology, SE 10691, Stockholm, Sweden.
| |
Collapse
|
4
|
Paramshetti S, Angolkar M, Talath S, Osmani RAM, Spandana A, Al Fatease A, Hani U, Ramesh KVRNS, Singh E. Unravelling the in vivo dynamics of liposomes: Insights into biodistribution and cellular membrane interactions. Life Sci 2024; 346:122616. [PMID: 38599316 DOI: 10.1016/j.lfs.2024.122616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/21/2024] [Accepted: 04/05/2024] [Indexed: 04/12/2024]
Abstract
Liposomes, as a colloidal drug delivery system dating back to the 1960s, remain a focal point of extensive research and stand as a highly efficient drug delivery method. The amalgamation of technological and biological advancements has propelled their evolution, elevating them to their current status. The key attributes of biodegradability and biocompatibility have been instrumental in driving substantial progress in liposome development. Demonstrating a remarkable ability to surmount barriers in drug absorption, enhance stability, and achieve targeted distribution within the body, liposomes have become pivotal in pharmaceutical research. In this comprehensive review, we delve into the intricate details of liposomal drug delivery systems, focusing specifically on their pharmacokinetics and cell membrane interactions via fusion, lipid exchange, endocytosis etc. Emphasizing the nuanced impact of various liposomal characteristics, we explore factors such as lipid composition, particle size, surface modifications, charge, dosage, and administration routes. By dissecting the multifaceted interactions between liposomes and biological barriers, including the reticuloendothelial system (RES), opsonization, enhanced permeability and retention (EPR) effect, ATP-binding cassette (ABC) phenomenon, and Complement Activation-Related Pseudoallergy (CARPA) effect, we provide a deeper understanding of liposomal behaviour in vivo. Furthermore, this review addresses the intricate challenges associated with translating liposomal technology into practical applications, offering insights into overcoming these hurdles. Additionally, we provide a comprehensive analysis of the clinical adoption and patent landscape of liposomes across diverse biomedical domains, shedding light on their potential implications for future research and therapeutic developments.
Collapse
Affiliation(s)
- Sharanya Paramshetti
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, Karnataka, India.
| | - Mohit Angolkar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, Karnataka, India.
| | - Sirajunisa Talath
- Department of Pharmaceutical Chemistry, RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates.
| | - Riyaz Ali M Osmani
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, Karnataka, India.
| | - Asha Spandana
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, Karnataka, India.
| | - Adel Al Fatease
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia.
| | - Umme Hani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia.
| | - K V R N S Ramesh
- Department of Pharmaceutics, RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates.
| | - Ekta Singh
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States.
| |
Collapse
|
5
|
Kimura T, Okada K, Morohashi Y, Kato Y, Mori M, Kato H, Matsumoto T, Shimoyama S. Quantification of Unencapsulated Drug in Target Tissues Demonstrates Pharmacological Properties and Therapeutic Effects of Liposomal Topotecan (FF-10850). Pharm Res 2024; 41:795-806. [PMID: 38536615 PMCID: PMC11024016 DOI: 10.1007/s11095-023-03652-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 12/27/2023] [Indexed: 04/18/2024]
Abstract
PURPOSE Quantifying unencapsulated drug concentrations in tissues is crucial for understanding the mechanisms underlying the efficacy and safety of liposomal drugs; however, the methodology for this has not been fully established. Herein, we aimed to investigate the enhanced therapeutic potential of a pegylated liposomal formulation of topotecan (FF-10850) by analyzing the concentrations of the unencapsulated drug in target tissues, to guide the improvement of its dosing regimen. METHODS We developed a method for measuring unencapsulated topotecan concentrations in tumor and bone marrow interstitial fluid (BM-ISF) and applied this method to pharmacokinetic assessments. The ratios of the area under the concentration-time curves (AUCs) between tumor and BM-ISF were calculated for total and unencapsulated topotecan. DNA damage and antitumor effects of FF-10850 or non-liposomal topotecan (TPT) were evaluated in an ES-2 mice xenograft model. RESULTS FF-10850 exhibited a much larger AUC ratio between tumor and BM-ISF for unencapsulated topotecan (2.96), but not for total topotecan (0.752), than TPT (0.833). FF-10850 promoted milder DNA damage in the bone marrow than TPT; however, FF-10850 and TPT elicited comparable DNA damage in the tumor. These findings highlight the greater tumor exposure to unencapsulated topotecan and lower bone marrow exposure to FF-10850 than TPT. The dosing regimen was successfully improved based on the kinetics of unencapsulated topotecan and DNA damage. CONCLUSIONS Tissue pharmacokinetics of unencapsulated topotecan elucidated the favorable pharmacological properties of FF-10850. Evaluation of tissue exposure to an unencapsulated drug with appropriate pharmacodynamic markers can be valuable in optimizing liposomal drugs and dosing regimens.
Collapse
Affiliation(s)
- Toshifumi Kimura
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-Machi, Ashigarakami-Gun, Kanagawa, 258-8577, Japan.
| | - Ken Okada
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-Machi, Ashigarakami-Gun, Kanagawa, 258-8577, Japan
| | - Yasushi Morohashi
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-Machi, Ashigarakami-Gun, Kanagawa, 258-8577, Japan
| | - Yukio Kato
- Faculty of Pharmacy, Kanazawa University, Kakuma-Machi, Kanazawa, Ishikawa, 920-1192, Japan
| | - Mikinaga Mori
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-Machi, Ashigarakami-Gun, Kanagawa, 258-8577, Japan
| | - Hiroshi Kato
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-Machi, Ashigarakami-Gun, Kanagawa, 258-8577, Japan
| | - Takeshi Matsumoto
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-Machi, Ashigarakami-Gun, Kanagawa, 258-8577, Japan
| | - Susumu Shimoyama
- FUJIFILM Pharmaceuticals U.S.A., Inc, One Broadway, Cambridge, MA, 02142, USA
| |
Collapse
|
6
|
Mathis BJ, Kato H, Matsuishi Y, Hiramatsu Y. Endogenous and exogenous protection from surgically induced reactive oxygen and nitrogen species. Surg Today 2024; 54:1-13. [PMID: 36348164 DOI: 10.1007/s00595-022-02612-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 10/11/2022] [Indexed: 11/09/2022]
Abstract
Surgical intervention creates reactive oxygen species through diverse molecular mechanisms, including direct stimulation of immune-mediated inflammation necessary for wound healing. However, dysregulation of redox homeostasis in surgical patients overwhelms the endogenous defense system, slowing the healing process and damaging organs. We broadly surveyed reactive oxygen species that result from surgical interventions and the endogenous and/or exogenous antioxidants that control them. This study assimilates current reports on surgical sources of reactive oxygen and nitrogen species along with literature reports on the effects of endogenous and exogenous antioxidants in human, animal, and clinical settings. Although exogenous antioxidants are generally beneficial, endogenous antioxidant systems account for over 80% of total activity, varying based on patient age, sex, and health or co-morbidity status, especially in smokers, the diabetic, and the obese. Supplementation of exogenous compounds for support in surgical patients is thus theoretically beneficial, but a lack of persuasive clinical evidence has left this potential patient support strategy without clear guidelines. A more thorough understanding of the mechanisms of exogenous antioxidants in patients with compromised health statuses and pharmacokinetic profiling may increase the utility of such support in both the operating and recovery rooms.
Collapse
Affiliation(s)
- Bryan J Mathis
- International Medical Center, University of Tsukuba Affiliated Hospital, 2-1-1 Amakubo, Tsukuba, 305-8576, Ibaraki, Japan.
| | - Hideyuki Kato
- Department of Cardiovascular Surgery, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yujiro Matsuishi
- Department of Neuroscience Nursing, St. Luke's International University, Tokyo, Japan
| | - Yuji Hiramatsu
- Department of Cardiovascular Surgery, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
7
|
André AS, Dias JNR, Aguiar SI, Leonardo A, Nogueira S, Amaral JD, Fernandes C, Gano L, Correia JDG, Cavaco M, Neves V, Correia J, Castanho M, Rodrigues CMP, Gaspar MM, Tavares L, Aires-da-Silva F. Panobinostat-loaded folate targeted liposomes as a promising drug delivery system for treatment of canine B-cell lymphoma. Front Vet Sci 2023; 10:1236136. [PMID: 37711439 PMCID: PMC10498770 DOI: 10.3389/fvets.2023.1236136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/07/2023] [Indexed: 09/16/2023] Open
Abstract
Introduction Cancer is a major public health problem with over 19 million cases reported in 2020. Similarly to humans, dogs are also largely affected by cancer, with non-Hodgkin's lymphoma (NHL) among the most common cancers in both species. Comparative medicine has the potential to accelerate the development of new therapeutic options in oncology by leveraging commonalities between diseases affecting both humans and animals. Within this context, in the present study, we investigated the potential of panobinostat (Pan)-loaded folate-targeted PEGylated liposomes (FA-PEG-Pan-Lip) for the treatment of canine B-cell lymphoma, while contributing to new perspectives in comparative oncology. Methods and results Two formulations were developed, namely: PEG-Pan-Lip and FA-PEG-Pan-Lip. Firstly, folate receptor expression in the CLBL-1 canine B-cell lymphoma cell line was assessed. After confirming receptor expression, both Pan-loaded formulations (PEG-Pan-Lip, FA-PEG-Pan-Lip) demonstrated dose-dependent inhibitory effects on CLBL-1 cell proliferation. The FA-PEG-Pan-Lip formulation (IC50 = 10.9 ± 0.03 nM) showed higher cytotoxicity than the non-targeted PEG-Pan-Lip formulation (IC50 = 12.9 ± 0.03 nM) and the free panobinostat (Pan) compound (IC50 = 18.32±0.03 nM). Moreover, mechanistically, both Pan-containing formulations induced acetylation of H3 histone and apoptosis. Flow cytometry and immunofluorescence analysis of intracellular uptake of rhodamine-labeled liposome formulations in CLBL-1 cells confirmed cellular internalization of PEG-Lip and FA-PEG-Lip formulations and higher uptake profile for the latter. Biodistribution studies of both radiolabeled formulations in CD1 and SCID mice revealed a rapid clearance from the major organs and a 1.6-fold enhancement of tumor uptake at 24 h for 111In-FA-PEG-Pan-Lip (2.2 ± 0.1 %ID/g of tumor) compared to 111In-PEG-Pan-Lip formulation (1.2±0.2 %ID/g of tumor). Discussion In summary, our results provide new data validating Pan-loaded folate liposomes as a promising targeted drug delivery system for the treatment of canine B-cell lymphoma and open innovative perspectives for comparative oncology.
Collapse
Affiliation(s)
- Ana S. André
- Faculty of Veterinary Medicine, CIISA-Centre for Interdisciplinary Research in Animal Health, University of Lisbon, Avenida da Universidade Técnica, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Joana N. R. Dias
- Faculty of Veterinary Medicine, CIISA-Centre for Interdisciplinary Research in Animal Health, University of Lisbon, Avenida da Universidade Técnica, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Sandra I. Aguiar
- Faculty of Veterinary Medicine, CIISA-Centre for Interdisciplinary Research in Animal Health, University of Lisbon, Avenida da Universidade Técnica, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Ana Leonardo
- Faculty of Veterinary Medicine, CIISA-Centre for Interdisciplinary Research in Animal Health, University of Lisbon, Avenida da Universidade Técnica, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Sara Nogueira
- Faculty of Veterinary Medicine, CIISA-Centre for Interdisciplinary Research in Animal Health, University of Lisbon, Avenida da Universidade Técnica, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Joana D. Amaral
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Célia Fernandes
- Departamento de Engenharia e Ciências Nucleares, Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Bobadela, Portugal
| | - Lurdes Gano
- Departamento de Engenharia e Ciências Nucleares, Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Bobadela, Portugal
| | - João D. G. Correia
- Departamento de Engenharia e Ciências Nucleares, Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Bobadela, Portugal
| | - Marco Cavaco
- Faculdade de Medicina, Instituto de Medicina Molecular-João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| | - Vera Neves
- Faculdade de Medicina, Instituto de Medicina Molecular-João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| | - Jorge Correia
- Faculty of Veterinary Medicine, CIISA-Centre for Interdisciplinary Research in Animal Health, University of Lisbon, Avenida da Universidade Técnica, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Miguel Castanho
- Faculdade de Medicina, Instituto de Medicina Molecular-João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| | - Cecília M. P. Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Maria Manuela Gaspar
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Luís Tavares
- Faculty of Veterinary Medicine, CIISA-Centre for Interdisciplinary Research in Animal Health, University of Lisbon, Avenida da Universidade Técnica, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Frederico Aires-da-Silva
- Faculty of Veterinary Medicine, CIISA-Centre for Interdisciplinary Research in Animal Health, University of Lisbon, Avenida da Universidade Técnica, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| |
Collapse
|
8
|
Bhatia A, Upadhyay AK, Sharma S. miRNAs are now starring in "No Time to Die: Overcoming the chemoresistance in cancer". IUBMB Life 2023; 75:238-256. [PMID: 35678612 DOI: 10.1002/iub.2652] [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: 03/15/2022] [Accepted: 05/04/2022] [Indexed: 12/24/2022]
Abstract
Cancer is a leading cause of death globally, with about 19.3 million new cases reported each year. Current therapies for cancer management include-chemotherapy, radiotherapy, and surgery. However, they are loaded with side effects and tend to cause toxicity in the patient's body posttreatment, ultimately hindering the response towards the treatment building up resistance. This is where noncoding RNAs such as miRNAs help provide us with a helping hand for taming the chemoresistance and providing potential holistic cancer management. MicroRNAs are promising targets for anticancer therapy as they perform critical regulatory roles in various signaling cascades related to cell proliferation, apoptosis, migration, and invasion. Combining miRNAs and anticancer drugs and devising a combination therapy has managed cancer well in various independent studies. This review aims to provide insights into how miRNAs play a mechanistic role in cancer development and progression and regulate drug resistance in various types of cancers. Furthermore, next-generation novel therapies using miRNAs in combination with anticancer treatments in multiple cancers have been put forth and how they improve the efficacy of the treatments. Exemplary studies currently in the preclinical and clinical models have been summarized. Ultimately, we briefly talk through the challenges that come forward with it and minimize them.
Collapse
Affiliation(s)
- Anmol Bhatia
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala, India
| | - Atul Kumar Upadhyay
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala, India
| | - Siddharth Sharma
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala, India
| |
Collapse
|
9
|
Sarkar M, Wang Y, Ekpenyong O, Liang D, Xie H. Pharmacokinetic behaviors of soft nanoparticulate formulations of chemotherapeutics. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1846. [PMID: 35979879 PMCID: PMC9938089 DOI: 10.1002/wnan.1846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/17/2022] [Accepted: 07/12/2022] [Indexed: 11/10/2022]
Abstract
Chemotherapeutic treatment with conventional drug formulations pose numerous challenges, such as poor solubility, high cytotoxicity and serious off-target side effects, low bioavailability, and ultimately subtherapeutic tumoral concentration leading to poor therapeutic outcomes. In the field of Nanomedicine, advances in nanotechnology have been applied with great success to design and develop novel nanoparticle-based formulations for the treatment of various types of cancer. The approval of the first nanomedicine, Doxil® (liposomal doxorubicin) in 1995, paved the path for further development for various types of novel delivery platforms. Several different types of nanoparticles, especially organic (soft) nanoparticles (liposomes, polymeric micelles, and albumin-bound nanoparticles), have been developed and approved for several anticancer drugs. Nanoparticulate drug delivery platform have facilitated to overcome of these challenges and offered key advantages of improved bioavailability, higher intra-tumoral concentration of the drug, reduced toxicity, and improved efficacy. This review introduces various commonly used nanoparticulate systems in biomedical research and their pharmacokinetic (PK) attributes, then focuses on the various physicochemical and physiological factors affecting the in vivo disposition of chemotherapeutic agents encapsulated in nanoparticles in recent years. Further, it provides a review of the current landscape of soft nanoparticulate formulations for the two most widely investigated anticancer drugs, paclitaxel, and doxorubicin, that are either approved or under investigation. Formulation details, PK profiles, and therapeutic outcomes of these novel strategies have been discussed individually and in comparison, to traditional formulations. This article is categorized under: Nanotechnology Approaches to Biology > Cells at the Nanoscale Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Mahua Sarkar
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, Texas, USA
| | - Yang Wang
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, Texas, USA
| | | | - Dong Liang
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, Texas, USA
| | - Huan Xie
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, Texas, USA
| |
Collapse
|
10
|
Gomes IP, Silva JDO, Cassali GD, De Barros ALB, Leite EA. Cisplatin-Loaded Thermosensitive Liposomes Functionalized with Hyaluronic Acid: Cytotoxicity and In Vivo Acute Toxicity Evaluation. Pharmaceutics 2023; 15:pharmaceutics15020583. [PMID: 36839905 PMCID: PMC9961010 DOI: 10.3390/pharmaceutics15020583] [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: 12/29/2022] [Revised: 01/31/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Cisplatin (CDDP) is a potent antitumor drug used in first-line chemotherapy against several solid tumors, including breast cancer. However, toxicities and drug resistance limit its clinical application. Thermosensitive liposome (TSL) functionalized with hyaluronic acid (HA) containing cisplatin (TSL-CDDP-HA) was developed by our research group aiming to promote the release of CDDP in the tumor region under hyperthermia conditions, as well as to decrease toxicity. Thus, this study aimed to evaluate this new formulation (HA-coated TSL-CDDP) concerning in vitro behavior and in vivo toxicity compared to non-coated TSL-CDDP and free CDDP. Cytotoxicity assays and nuclear morphology were carried out against triple-negative breast cancer cells (MDA-MB-231), while an in vivo toxicity study was performed using healthy Swiss mice. The results showed an increase (around 3-fold) in cytotoxicity of the cationic formulation (non-coated TSL-CDDP) compared to free CDDP. On the other hand, TSL-CDDP treatment induced the appearance of 2.5-fold more senescent cells with alteration of nuclear morphology than the free drug after hyperthermia condition. Furthermore, the association of liposomal formulations treatment with hyperthermia increased the percentage of apoptotic cells compared to those without heating. The percentage of apoptotic cells was 1.7-fold higher for TSL-CDDP-HA than for TSL-CDDP. For the in vivo toxicity data, the TSL-CDDP treatment was also toxic to healthy cells, inducing nephrotoxicity with a significant increase in urea levels compared to the saline control group (73.1 ± 2.4 vs. 49.2 ± 2.8 mg/mL). On the other hand, the HA-coated TSL-CDDP eliminated the damages related to the use of CDDP since the animals did not show changes in hematological and biochemical examinations and histological analyses. Thus, data suggest that this new formulation is a potential candidate for the intravenous therapy of solid tumors.
Collapse
Affiliation(s)
- Isabela Pereira Gomes
- Department of Pharmaceutical Products, Faculty of Pharmacy, Federal University of Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil
| | - Juliana de Oliveira Silva
- Department of Pharmaceutical Products, Faculty of Pharmacy, Federal University of Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil
| | - Geovanni Dantas Cassali
- Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil
| | - André Luís Branco De Barros
- Department of Clinical and Toxicological Analyses, Faculty of Pharmacy, Federal University of Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil
| | - Elaine Amaral Leite
- Department of Pharmaceutical Products, Faculty of Pharmacy, Federal University of Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil
- Correspondence: or ; Tel.: +55-3134096944; Fax: +55-3134096935
| |
Collapse
|
11
|
Bandeira LC, Pinto L, Carneiro CM. Pharmacometrics: The Already-Present Future of Precision Pharmacology. Ther Innov Regul Sci 2023; 57:57-69. [PMID: 35984633 DOI: 10.1007/s43441-022-00439-4] [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: 02/14/2022] [Accepted: 07/20/2022] [Indexed: 02/01/2023]
Abstract
The use of mathematical modeling to represent, analyze, make predictions or providing information on data obtained in drug research and development has made pharmacometrics an area of great prominence and importance. The main purpose of pharmacometrics is to provide information relevant to the search for efficacy and safety improvements in pharmacotherapy. Regulatory agencies have adopted pharmacometrics analysis to justify their regulatory decisions, making those decisions more efficient. Demand for specialists trained in the field is therefore growing. In this review, we describe the meaning, history, and development of pharmacometrics, analyzing the challenges faced in the training of professionals. Examples of applications in current use, perspectives for the future, and the importance of pharmacometrics for the development and growth of precision pharmacology are also presented.
Collapse
Affiliation(s)
- Lorena Cera Bandeira
- Laboratory of Immunopathology, Nucleus of Biological Sciences Research, Federal University of Ouro Preto, Ouro Preto, Minas Gerais, Brazil.
| | - Leonardo Pinto
- Laboratory of Immunopathology, Nucleus of Biological Sciences Research, Federal University of Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Cláudia Martins Carneiro
- Laboratory of Immunopathology, Nucleus of Biological Sciences Research, Federal University of Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| |
Collapse
|
12
|
Mukherjee A, Bisht B, Dutta S, Paul MK. Current advances in the use of exosomes, liposomes, and bioengineered hybrid nanovesicles in cancer detection and therapy. Acta Pharmacol Sin 2022; 43:2759-2776. [PMID: 35379933 PMCID: PMC9622806 DOI: 10.1038/s41401-022-00902-w] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 03/15/2022] [Indexed: 12/17/2022] Open
Abstract
Three major approaches of cancer therapy can be enunciated as delivery of biotherapeutics, tumor image analysis, and immunotherapy. Liposomes, artificial fat bubbles, are long known for their capacity to encapsulate a diverse range of bioactive molecules and release the payload in a sustained, stimuli-responsive manner. They have already been widely explored as a delivery vehicle for therapeutic drugs as well as imaging agents. They are also extensively being used in cancer immunotherapy. On the other hand, exosomes are naturally occurring nanosized extracellular vesicles that serve an important role in cell-cell communication. Importantly, the exosomes also have proven their capability to carry an array of active pharmaceuticals and diagnostic molecules to the tumor cells. Exosomes, being enriched with tumor antigens, have numerous immunomodulatory effects. Much to our intrigue, in recent times, efforts have been directed toward developing smart, bioengineered, exosome-liposome hybrid nanovesicles, which are augmented by the benefits of both vesicular systems. This review attempts to summarize the contemporary developments in the use of exosome and liposome toward cancer diagnosis, therapy, as a vehicle for drug delivery, diagnostic carrier for tumor imaging, and cancer immunotherapy. We shall also briefly reflect upon the recent advancements of the exosome-liposome hybrids in cancer therapy. Finally, we put forward future directions for the use of exosome/liposome and/or hybrid nanocarriers for accurate diagnosis and personalized therapies for cancers.
Collapse
Affiliation(s)
| | - Bharti Bisht
- Division of Thoracic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Suman Dutta
- International Institute of Innovation and Technology, New Town, Kolkata, 700156, India
| | - Manash K Paul
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
13
|
Lu CH, Hsiao JK. Diagnostic and therapeutic roles of iron oxide nanoparticles in biomedicine. Tzu Chi Med J 2022; 35:11-17. [PMID: 36866343 PMCID: PMC9972926 DOI: 10.4103/tcmj.tcmj_65_22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/03/2022] [Accepted: 06/08/2022] [Indexed: 11/04/2022] Open
Abstract
Nanotechnology changed our understanding of physics and chemics and influenced the biomedical field. Iron oxide nanoparticles (IONs) are one of the first emerging biomedical applications of nanotechnology. The IONs are composed of iron oxide core exhibiting magnetism and coated with biocompatible molecules. The small size, strong magnetism, and biocompatibility of IONs facilitate the application of IONs in the medical imaging field. We listed several clinical available IONs including Resovist (Bayer Schering Pharma, Berlin, Germany) and Feridex intravenous (I.V.)/Endorem as magnetic resonance (MR) contrast agents for liver tumor detection. We also illustrated GastroMARK as a gastrointestinal contrast agent for MR imaging. Recently, IONs named Feraheme for treating iron-deficiency anemia have been approved by the Food and Drug Administration. Moreover, tumor ablation by IONs named NanoTherm has also been discussed. In addition to the clinical application, several potential biomedical applications of IONs including cancer-targeting capability by conjugating IONs with cancer-specific ligands, cell trafficking tools, or tumor ablation agents have also been discussed. With the growing awareness of nanotechnology, further application of IONs is still on the horizon that would shed light on biomedicine.
Collapse
Affiliation(s)
- Chia-Hung Lu
- Department of Medical Imaging, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Jong-Kai Hsiao
- Department of Medical Imaging, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan,School of Medicine, Tzu Chi University, Hualien, Taiwan,Address for correspondence: Dr. Jong-Kia Hsiao, Department of Medical Imaging, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, 289, Jianguo Road, Xindian District, New Taipei, Taiwan. E-mail:
| |
Collapse
|
14
|
Givinostat-Liposomes: Anti-Tumor Effect on 2D and 3D Glioblastoma Models and Pharmacokinetics. Cancers (Basel) 2022; 14:cancers14122978. [PMID: 35740641 PMCID: PMC9220922 DOI: 10.3390/cancers14122978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/10/2022] [Accepted: 06/14/2022] [Indexed: 11/16/2022] Open
Abstract
Glioblastoma is the most common and aggressive brain tumor, associated with poor prognosis and survival, representing a challenging medical issue for neurooncologists. Dysregulation of histone-modifying enzymes (HDACs) is commonly identified in many tumors and has been linked to cancer proliferation, changes in metabolism, and drug resistance. These findings led to the development of HDAC inhibitors, which are limited by their narrow therapeutic index. In this work, we provide the proof of concept for a delivery system that can improve the in vivo half-life and increase the brain delivery of Givinostat, a pan-HDAC inhibitor. Here, 150-nm-sized liposomes composed of cholesterol and sphingomyelin with or without surface decoration with mApoE peptide, inhibited human glioblastoma cell growth in 2D and 3D models by inducing a time- and dose-dependent reduction in cell viability, reduction in the receptors involved in cholesterol metabolism (from -25% to -75% of protein levels), and reduction in HDAC activity (-25% within 30 min). In addition, liposome-Givinostat formulations showed a 2.5-fold increase in the drug half-life in the bloodstream and a 6-fold increase in the amount of drug entering the brain in healthy mice, without any signs of overt toxicity. These features make liposomes loaded with Givinostat valuable as potential candidates for glioblastoma therapy.
Collapse
|
15
|
Sukocheva OA, Liu J, Neganova ME, Beeraka NM, Aleksandrova YR, Manogaran P, Grigorevskikh EM, Chubarev VN, Fan R. Perspectives of using microRNA-loaded nanocarriers for epigenetic reprogramming of drug resistant colorectal cancers. Semin Cancer Biol 2022; 86:358-375. [PMID: 35623562 DOI: 10.1016/j.semcancer.2022.05.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 02/07/2023]
Abstract
Epigenetic regulation by microRNAs (miRs) demonstrated a promising therapeutic potential of these molecules to regulate genetic activity in different cancers, including colorectal cancers (CRCs). The RNA-based therapy does not change genetic codes in tumor cells but can silence oncogenes and/or reactivate inhibited tumor suppressor genes. In many cancers, specific miRs were shown to promote or stop tumor progression. Among confirmed and powerful epigenetic regulators of colon carcinogenesis and development of resistance are onco-miRs, which include let-7, miR-21, miR-22, miR-23a, miR-27a, miR-34, miR-92, miR-96, miR-125b, miR-135b, miR-182, miR-200c, miR-203, miR-221, miR-421, miR-451, and others. Moreover, various tumor-suppressor miRs (miR-15b-5b, miR-18a, miR-20b, miR-22, miR-96, miR-139-5p, miR-145, miR-149, miR-197, miR-199b, miR-203, miR-214, miR-218, miR-320, miR-375-3p, miR-409-3p, miR-450b-5p, miR-494, miR-577, miR-874, and others) were found silenced in drug-resistant CRCs. Re-expression of tumor suppressor miR is complicated by the chemical nature of miRs that are not long-lasting compounds and require protection from the enzymatic degradation. Several recent studies explored application of miRs using nanocarrier complexes. This study critically describes the most successfully tested nanoparticle complexes used for intracellular delivery of nuclear acids and miRs, including micelles, liposomes, inorganic and polymeric NPs, dendrimers, and aptamers. Nanocarriers shield incorporated miRs and improve the agent stability in circulation. Attachment of antibodies and/or specific peptide or ligands facilitates cell-targeted miR delivery. Addressing in vivo challenges, a broad spectrum of non-toxic materials has been tested and indicated reliable advantages of lipid-based (lipoplexes) and polymer-based liposomes. Recent cutting-edge developments indicated that lipid-based complexes with multiple cargo, including several miRs, are the most effective approach to eradicate drug-resistant tumors. Focusing on CRC-specific miRs, this review provides a guidance and insights towards the most promising direction to achieve dramatic reduction in tumor growth and metastasis using miR-nanocarrier complexes.
Collapse
Affiliation(s)
- Olga A Sukocheva
- Cancer Center and Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshedong Str., Zhengzhou, 450052, China; The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute, Griffith University, Queensland, Australia; Discipline of Health Sciences, College of Nursing and Health Sciences, Flinders University, Bedford Park, South Australia, 5042, Australia.
| | - Junqi Liu
- Cancer Center and Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshedong Str., Zhengzhou, 450052, China
| | - Margarita E Neganova
- Institute of Physiologically Active Compounds of the Russian Academy of Sciences, 1, Severnii pr., Chernogolovka, 142432, Russia
| | - Narasimha M Beeraka
- Discipline of Health Sciences, College of Nursing and Health Sciences, Flinders University, Bedford Park, South Australia, 5042, Australia; Department of Human Anatomy, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Street, Moscow, 119991, Russia; Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Academy of Higher Education and Research (JSS AHER), JSS Medical College, Mysuru, Karnataka, India
| | - Yulia R Aleksandrova
- Institute of Physiologically Active Compounds of the Russian Academy of Sciences, 1, Severnii pr., Chernogolovka, 142432, Russia
| | - Prasath Manogaran
- Translational Research Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
| | - Ekaterina M Grigorevskikh
- Department of Human Anatomy, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Street, Moscow, 119991, Russia
| | - Vladimir N Chubarev
- Department of Human Anatomy, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Street, Moscow, 119991, Russia
| | - Ruitai Fan
- Cancer Center and Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshedong Str., Zhengzhou, 450052, China.
| |
Collapse
|
16
|
Sheikholeslami B, Lam NW, Dua K, Haghi M. Exploring the impact of physicochemical properties of liposomal formulations on their in vivo fate. Life Sci 2022; 300:120574. [DOI: 10.1016/j.lfs.2022.120574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/12/2022] [Accepted: 04/18/2022] [Indexed: 12/16/2022]
|
17
|
Tadini MC, Fernandes FS, Ozelin SD, de Melo MRS, Mansur AL, de Toledo TB, de Albuquerque NCP, Tavares DC, Marquele-Oliveira F, de Oliveira ARM. Pharmacokinetic study of AmB-NP-GR: a new granule form with amphotericin B to treat leishmaniasis and fungal infections. Eur J Pharm Sci 2022; 173:106173. [PMID: 35331860 DOI: 10.1016/j.ejps.2022.106173] [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: 09/09/2021] [Revised: 03/18/2022] [Accepted: 03/20/2022] [Indexed: 11/03/2022]
Abstract
Amphotericin B (AmB) has been the gold standard to treat systemic fungal infections. The use of AmB is restricted to hospitals because it poses several risks, mainly risks related to its high nephrotoxicity. Given the importance of this drug in medicine, new therapeutics and AmB formulations with nanotechnological improvements are required and could bring many benefits to patients. A new drug formulation with gastro-resistant coated granules has been proposed. The lipid-based system containing AmB was produced and used as raw material in the granulation/coated process. The new developed formulation (AmB-NP-GR) was characterized by optical microscopy, granulometry, and atomic force microscopy (AFM) after disintegration test. AmB-NP-GR showed granular shape, with most granules measured between 250 and 500 µm (37 ± 7% w/w). The AFM images indicated that the granule formulation should disintegrate in the intestine, to release the lipid-based carriers, making them available for absorption and allowing them to reach the blood circulation. The developed formulation was administered to rats in a single dose of 4.0 or 8.0 mg kg-1 and the pharmacokinetics was studied. The samples were analyzed by liquid chromatography coupled to mass spectrometry. Before the pharmacokinetic studies were conducted, the bioanalytical method was validated according to the EMA guideline and all evaluated parameters agreed with this guideline. The pharmacokinetic results showed that Cmax was similar for both doses and that tmax was reached at 4-12 hours for dose of 4.0 mg kg-1 and 4 hours for dose of 8.0 mg kg-1. The half-life related to the dose of 8.0 mg kg-1 increased significantly compared to the dose of 4.0 mg kg-1 (an increase of more than 3 times). In addition, the mean residence time related to the dose of 8.0 mg kg-1 was 4 times higher than for the lower dose. The clearance value showed to be higher for the lower dose. Together, these results provide important conclusions for experimental design of other in vivo safety and efficacy studies of AmB-NP-GR.
Collapse
Affiliation(s)
- Maraine Catarina Tadini
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901, Ribeirão Preto, SP, Brazil; Eleve Science Pesquisa e Desenvolvimento, Supera Parque Av. Dra. Nadir Aguiar, n. 1805, 14056-680, Ribeirão Preto, São Paulo, Brazil
| | | | - Saulo Duarte Ozelin
- University of Franca, Av. Dr. Armando Salles Oliveira, n. 201, 14404-600, Franca, SP, Brazil
| | | | - Ana Luiza Mansur
- Eleve Science Pesquisa e Desenvolvimento, Supera Parque Av. Dra. Nadir Aguiar, n. 1805, 14056-680, Ribeirão Preto, São Paulo, Brazil
| | - Thaís Bueno de Toledo
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901, Ribeirão Preto, SP, Brazil; Eleve Science Pesquisa e Desenvolvimento, Supera Parque Av. Dra. Nadir Aguiar, n. 1805, 14056-680, Ribeirão Preto, São Paulo, Brazil
| | | | - Denise Crispim Tavares
- University of Franca, Av. Dr. Armando Salles Oliveira, n. 201, 14404-600, Franca, SP, Brazil
| | - Franciane Marquele-Oliveira
- Eleve Science Pesquisa e Desenvolvimento, Supera Parque Av. Dra. Nadir Aguiar, n. 1805, 14056-680, Ribeirão Preto, São Paulo, Brazil.
| | - Anderson Rodrigo Moraes de Oliveira
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, 14040-901, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
18
|
Đorđević S, Gonzalez MM, Conejos-Sánchez I, Carreira B, Pozzi S, Acúrcio RC, Satchi-Fainaro R, Florindo HF, Vicent MJ. Current hurdles to the translation of nanomedicines from bench to the clinic. Drug Deliv Transl Res 2022; 12:500-525. [PMID: 34302274 PMCID: PMC8300981 DOI: 10.1007/s13346-021-01024-2] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2021] [Indexed: 02/07/2023]
Abstract
The field of nanomedicine has significantly influenced research areas such as drug delivery, diagnostics, theranostics, and regenerative medicine; however, the further development of this field will face significant challenges at the regulatory level if related guidance remains unclear and unconsolidated. This review describes those features and pathways crucial to the clinical translation of nanomedicine and highlights considerations for early-stage product development. These include identifying those critical quality attributes of the drug product essential for activity and safety, appropriate analytical methods (physical, chemical, biological) for characterization, important process parameters, and adequate pre-clinical models. Additional concerns include the evaluation of batch-to-batch consistency and considerations regarding scaling up that will ensure a successful reproducible manufacturing process. Furthermore, we advise close collaboration with regulatory agencies from the early stages of development to assure an aligned position to accelerate the development of future nanomedicines.
Collapse
Affiliation(s)
- Snežana Đorđević
- Polymer Therapeutics Laboratory, Prince Felipe Research Center (CIPF), Eduardo Primo Yúfera 3, 46012, Valencia, Av, Spain
| | - María Medel Gonzalez
- Polymer Therapeutics Laboratory, Prince Felipe Research Center (CIPF), Eduardo Primo Yúfera 3, 46012, Valencia, Av, Spain
| | - Inmaculada Conejos-Sánchez
- Polymer Therapeutics Laboratory, Prince Felipe Research Center (CIPF), Eduardo Primo Yúfera 3, 46012, Valencia, Av, Spain
| | - Barbara Carreira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisboa, Portugal
| | - Sabina Pozzi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Rita C Acúrcio
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisboa, Portugal
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel.
- Sagol School of Neuroscience, Tel Aviv University, 69978, Tel Aviv, Israel.
| | - Helena F Florindo
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisboa, Portugal.
| | - María J Vicent
- Polymer Therapeutics Laboratory, Prince Felipe Research Center (CIPF), Eduardo Primo Yúfera 3, 46012, Valencia, Av, Spain.
| |
Collapse
|
19
|
Alanazi SA, Harisa GI, Badran MM, Alanazi FK, Elzayat E, Alomrani AH, Al Meanazel OT, Al Meanazel AT. Crosstalk of low density lipoprotein and liposome as a paradigm for targeting of 5-fluorouracil into hepatic cells: cytotoxicity and liver deposition. Bioengineered 2021; 12:914-926. [PMID: 33678142 PMCID: PMC8806320 DOI: 10.1080/21655979.2021.1896202] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 02/19/2021] [Indexed: 11/20/2022] Open
Abstract
This study aimed to utilize cholesterol conjugation of 5-fluorouracil (5-FUC) and liposomal formulas to enhance the partitioning of 5-FU into low density lipoprotein (LDL) to target hepatocellular carcinoma (HCC). Thus, 5-FU and 5-FUCwere loaded into liposomes. Later, the direct loading and transfer of 5-FU, and 5-FUC from liposomes into LDL were attained. The preparations were characterized in terms of particle size, zeta potential, morphology, entrapment efficiency, and cytotoxicity using the HepG2 cell line. Moreover, the drug deposition into the LDL and liver tissues was investigated. The present results revealed that liposomal preparations have a nanosize range (155 - 194 nm), negative zeta potential (- 0.82 to - 16 mV), entrapment efficiency of 69% for 5-FU, and 66% for 5-FUC. Moreover, LDL particles have a nanosize range (28-49 nm), negative zeta potential (- 17 to -27 mV), and the entrapment efficiency is 11% for 5-FU and 85% for 5-FUC. Furthermore, 5-FUC loaded liposomes displayed a sustained release profile (57%) at 24 h compared to fast release (92%) of 5-FU loaded liposomes. 5-FUC and liposomal formulas enhanced the transfer of 5-FUC into LDL compared to 5-FU. 5-FUC loaded liposomes and LDL have greater cytotoxicity against HepG2 cell lines compared to 5-FU and 5-FUC solutions. Moreover, the deposition of 5-FUC in LDL (26.87ng/mg) and liver tissues (534 ng/gm tissue) was significantly increased 5-FUC liposomes compared to 5-FU (11.7 ng/g tissue) liposomal formulation. In conclusion, 5-FUC is a promising strategy for hepatic targeting of 5-FU through LDL-mediated gateway.
Collapse
Affiliation(s)
- Saleh A. Alanazi
- Kayyali Chair for Pharmaceutical Industries, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Pharmaceutical Care Services, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Gamaleldin I. Harisa
- Kayyali Chair for Pharmaceutical Industries, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Biochemistry, College of Pharmacy, Al-Azhar University, Nasr City, Cairo, Egypt
| | - Mohamed M. Badran
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmaceutics, College of Pharmacy, Al-Azhar University, Nasr City Cairo, Egypt
| | - Fars K. Alanazi
- Kayyali Chair for Pharmaceutical Industries, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ehab Elzayat
- Kayyali Chair for Pharmaceutical Industries, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah H. Alomrani
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Nanobiotechnology Unit, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Osaid T. Al Meanazel
- Kayyali Chair for Pharmaceutical Industries, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Michael Sayegh Faculty of Pharmacy, Aqaba University of Technology, Aqaba, Jordan
| | - Ahmed T. Al Meanazel
- Prince Naif for the Health Research Center, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
20
|
Moghassemi S, Dadashzadeh A, Azevedo RB, Feron O, Amorim CA. Photodynamic cancer therapy using liposomes as an advanced vesicular photosensitizer delivery system. J Control Release 2021; 339:75-90. [PMID: 34562540 DOI: 10.1016/j.jconrel.2021.09.024] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 09/16/2021] [Accepted: 09/18/2021] [Indexed: 12/26/2022]
Abstract
The multidisciplinary field of photodynamic therapy (PDT) is a combination of photochemistry and photophysics sciences, which has shown tremendous potential for cancer therapy application. PDT employs a photosensitizing agent (PS) and light to form cytotoxic reactive oxygen species and subsequently oxidize light-exposed tissue. Despite numerous advantages of PDT and enormous progress in this field, common PSs are still far from ideal treatment because of their poor permeability, non-specific phototoxicity, side effects, hydrophobicity, weak bioavailability, and tendency to self-aggregation. To circumvent these limitations, PS can be encapsulated in liposomes, an advanced drug delivery system that has demonstrated the ability to enhance drug permeability into biological membranes and loading both hydrophobic and lipophilic agents. Moreover, liposomes can also be coated by targeting agents to improve delivery efficiency. The present review aims to summarize the principles of PDT, various PS generations, PS-loaded nanoparticles, liposomes, and their impact on PDT, then discuss recent photodynamic cancer therapy strategies using liposomes as PS-loaded vectors, and highlight future possibilities and perspectives.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Ricardo Bentes Azevedo
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasília, Brasília, DF, Brazil
| | - Olivier Feron
- Pôle de Pharmacologie et thérapeutique, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Christiani A Amorim
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
21
|
Ewert KK, Scodeller P, Simón-Gracia L, Steffes VM, Wonder EA, Teesalu T, Safinya CR. Cationic Liposomes as Vectors for Nucleic Acid and Hydrophobic Drug Therapeutics. Pharmaceutics 2021; 13:1365. [PMID: 34575441 PMCID: PMC8465808 DOI: 10.3390/pharmaceutics13091365] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 08/09/2021] [Accepted: 08/21/2021] [Indexed: 12/15/2022] Open
Abstract
Cationic liposomes (CLs) are effective carriers of a variety of therapeutics. Their applications as vectors of nucleic acids (NAs), from long DNA and mRNA to short interfering RNA (siRNA), have been pursued for decades to realize the promise of gene therapy, with approvals of the siRNA therapeutic patisiran and two mRNA vaccines against COVID-19 as recent milestones. The long-term goal of developing optimized CL-based NA carriers for a broad range of medical applications requires a comprehensive understanding of the structure of these vectors and their interactions with cell membranes and components that lead to the release and activity of the NAs within the cell. Structure-activity relationships of lipids for CL-based NA and drug delivery must take into account that these lipids act not individually but as components of an assembly of many molecules. This review summarizes our current understanding of how the choice of the constituting lipids governs the structure of their CL-NA self-assemblies, which constitute distinct liquid crystalline phases, and the relation of these structures to their efficacy for delivery. In addition, we review progress toward CL-NA nanoparticles for targeted NA delivery in vivo and close with an outlook on CL-based carriers of hydrophobic drugs, which may eventually lead to combination therapies with NAs and drugs for cancer and other diseases.
Collapse
Affiliation(s)
- Kai K. Ewert
- Materials, Physics, and Molecular, Cellular, and Developmental Biology Departments, and Biomolecular Science and Engineering Program, University of California at Santa Barbara, Santa Barbara, CA 93106, USA; (V.M.S.); (E.A.W.)
| | - Pablo Scodeller
- Laboratory of Precision- and Nanomedicine, Institute of Biomedicine and Translational Medicine, Centre of Excellence for Translational Medicine, University of Tartu, Ravila 14b, 50411 Tartu, Estonia; (P.S.); (L.S.-G.)
| | - Lorena Simón-Gracia
- Laboratory of Precision- and Nanomedicine, Institute of Biomedicine and Translational Medicine, Centre of Excellence for Translational Medicine, University of Tartu, Ravila 14b, 50411 Tartu, Estonia; (P.S.); (L.S.-G.)
| | - Victoria M. Steffes
- Materials, Physics, and Molecular, Cellular, and Developmental Biology Departments, and Biomolecular Science and Engineering Program, University of California at Santa Barbara, Santa Barbara, CA 93106, USA; (V.M.S.); (E.A.W.)
| | - Emily A. Wonder
- Materials, Physics, and Molecular, Cellular, and Developmental Biology Departments, and Biomolecular Science and Engineering Program, University of California at Santa Barbara, Santa Barbara, CA 93106, USA; (V.M.S.); (E.A.W.)
| | - Tambet Teesalu
- Laboratory of Precision- and Nanomedicine, Institute of Biomedicine and Translational Medicine, Centre of Excellence for Translational Medicine, University of Tartu, Ravila 14b, 50411 Tartu, Estonia; (P.S.); (L.S.-G.)
- Center for Nanomedicine and Department of Cell, Molecular and Developmental Biology, University of California at Santa Barbara, Santa Barbara, CA 93106, USA
| | - Cyrus R. Safinya
- Materials, Physics, and Molecular, Cellular, and Developmental Biology Departments, and Biomolecular Science and Engineering Program, University of California at Santa Barbara, Santa Barbara, CA 93106, USA; (V.M.S.); (E.A.W.)
| |
Collapse
|
22
|
Li J, Zhou S, Yu J, Cai W, Yang Y, Kuang X, Liu H, He Z, Wang Y. Low dose shikonin and anthracyclines coloaded liposomes induce robust immunogenetic cell death for synergistic chemo-immunotherapy. J Control Release 2021; 335:306-319. [PMID: 34081995 DOI: 10.1016/j.jconrel.2021.05.040] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/20/2021] [Accepted: 05/29/2021] [Indexed: 12/14/2022]
Abstract
Chemo-immunotherapy based on immunogenic cell death (ICD) is a promising strategy for cancer therapy. However, the effective ICD requires a high dosage of ICD stimulus, which could be associated to a dose-dependent toxicity. Therefore, in this study, a liposome remote-loaded with shikonin (a potent ICD stimulus) was developed, with the ability to effectively induce ICD at high dosage in vivo. However, a hepatotoxic effect was observed. To circumvent this problem, shikonin was combined with the anthracycline mitoxantrone or doxorubicin to develop co-loaded liposomes inducing a synergistic ICD effect and cytotoxicity to tumor cells. Cytotoxicity and uptake experiment in vitro were performed to analyze the optimal synergistic ratio of shikonin and anthracyclines based on a "formulated strategy". Interestingly, copper mediated co-loaded liposomes resulted in a pH and GSH dual-responsive release property. More importantly, pharmacokinetics and tumor biodistribution studies revealed an outstanding capacity of ratiometric delivery of dual drugs. Thus, the dual-loaded liposome enhanced the antitumor effect by the stimulation of a robust immune response at lower doses of the drugs with a higher safety compared to single-loaded liposomes. Summarized, the current work provided a reference for a rational design and development of liposomal co-delivery system of drugs and ICD-induced chemo-immunotherapy, and established a potential clinical application of shikonin-based drug combinations as a new chemo-immunotherapeutic strategy for cancer treatment.
Collapse
Affiliation(s)
- Jinbo Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, Shenyang 110016, China
| | - Shuang Zhou
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, Shenyang 110016, China
| | - Jiang Yu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, Shenyang 110016, China
| | - Wenxu Cai
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, Shenyang 110016, China
| | - Yinxian Yang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, Shenyang 110016, China
| | - Xiao Kuang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, Shenyang 110016, China
| | - Hongzhuo Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, Shenyang 110016, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, Shenyang 110016, China
| | - Yongjun Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
23
|
Rommasi F, Esfandiari N. Liposomal Nanomedicine: Applications for Drug Delivery in Cancer Therapy. NANOSCALE RESEARCH LETTERS 2021; 16:95. [PMID: 34032937 PMCID: PMC8149564 DOI: 10.1186/s11671-021-03553-8] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/17/2021] [Indexed: 05/23/2023]
Abstract
The increasing prevalence of cancer, a disease in which rapid and uncontrollable cell growth causes complication and tissue dysfunction, is one of the serious and tense concerns of scientists and physicians. Nowadays, cancer diagnosis and especially its effective treatment have been considered as one of the biggest challenges in health and medicine in the last century. Despite significant advances in drug discovery and delivery, their many adverse effects and inadequate specificity and sensitivity, which usually cause damage to healthy tissues and organs, have been great barriers in using them. Limitation in the duration and amount of these therapeutic agents' administration is also challenging. On the other hand, the incidence of tumor cells that are resistant to typical methods of cancer treatment, such as chemotherapy and radiotherapy, highlights the intense need for innovation, improvement, and development in antitumor drug properties. Liposomes have been suggested as a suitable candidate for drug delivery and cancer treatment in nanomedicine due to their ability to store drugs with different physical and chemical characteristics. Moreover, the high flexibility and potential of liposome structure for chemical modification by conjugating various polymers, ligands, and molecules is a significant pro for liposomes not only to enhance their pharmacological merits but also to improve the effectiveness of anticancer drugs. Liposomes can increase the sensitivity, specificity, and durability of these anti-malignant cell agents in the body and provide remarkable benefits to be applied in nanomedicines. We reviewed the discovery and development of liposomes focusing on their clinical applications to treat diverse sorts of cancers and diseases. How the properties of liposomal drugs can be improved and their opportunity and challenges for cancer therapy were also considered and discussed.
Collapse
Affiliation(s)
- Foad Rommasi
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Neda Esfandiari
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
24
|
Matsumoto T, Komori T, Yoshino Y, Ioroi T, Kitahashi T, Kitahara H, Ono K, Higuchi T, Sakabe M, Kori H, Kano M, Hori R, Kato Y, Hagiwara S. A Liposomal Gemcitabine, FF-10832, Improves Plasma Stability, Tumor Targeting, and Antitumor Efficacy of Gemcitabine in Pancreatic Cancer Xenograft Models. Pharm Res 2021; 38:1093-1106. [PMID: 33961188 PMCID: PMC8217058 DOI: 10.1007/s11095-021-03045-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 04/15/2021] [Indexed: 01/02/2023]
Abstract
Purpose The clinical application of gemcitabine (GEM) is limited by its pharmacokinetic properties. The aim of this study was to characterize the stability in circulating plasma, tumor targeting, and payload release of liposome-encapsulated GEM, FF-10832. Methods Antitumor activity was assessed in xenograft mouse models of human pancreatic cancer. The pharmacokinetics of GEM and its active metabolite dFdCTP were also evaluated. Results In mice with Capan-1 tumors, the dose-normalized areas under the curve (AUCs) after FF-10832 administration in plasma and tumor were 672 and 1047 times higher, respectively, than after using unencapsulated GEM. The tumor-to-bone marrow AUC ratio of dFdCTP was approximately eight times higher after FF-10832 administration than after GEM administration. These results indicated that liposomal encapsulation produced long-term stability in circulating plasma and tumor-selective targeting of GEM. In mice with Capan-1, SUIT-2, and BxPC-3 tumors, FF-10832 had better antitumor activity and tolerability than GEM. Internalization of FF-10832 in tumor-associated macrophages (TAMs) was revealed by flow cytometry and confocal laser scanning microscopy, and GEM was efficiently released from isolated macrophages of mice treated with FF-10832. These results suggest that TAMs are one of the potential reservoirs of GEM in tumors. Conclusion This study found that FF-10832 had favorable pharmacokinetic properties. The liposomal formulation was more effective and tolerable than unencapsulated GEM in mouse xenograft tumor models. Hence, FF-10832 is a promising candidate for the treatment of pancreatic cancer. Supplementary Information The online version contains supplementary material available at 10.1007/s11095-021-03045-5.
Collapse
Affiliation(s)
- Takeshi Matsumoto
- Bioscience and Engineering laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa, 258-8577, Japan.
| | - Takashi Komori
- Bioscience and Engineering laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa, 258-8577, Japan
| | - Yuta Yoshino
- Bioscience and Engineering laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa, 258-8577, Japan
| | - Tadaaki Ioroi
- Bioscience and Engineering laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa, 258-8577, Japan
| | - Tsukasa Kitahashi
- Bioscience and Engineering laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa, 258-8577, Japan
| | - Hiromu Kitahara
- Bioscience and Engineering laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa, 258-8577, Japan
| | - Kohei Ono
- Bioscience and Engineering laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa, 258-8577, Japan
| | - Tamami Higuchi
- Bioscience and Engineering laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa, 258-8577, Japan
| | - Masayo Sakabe
- Bioscience and Engineering laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa, 258-8577, Japan
| | - Hiroshi Kori
- Bioscience and Engineering laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa, 258-8577, Japan
| | - Masahiro Kano
- Bioscience and Engineering laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa, 258-8577, Japan
| | - Ritsuko Hori
- Analysis Technology Center, FUJIFILM Corporation, Nakanuma 210, Minamiashigara, Kanagawa, 250-0193, Japan
| | - Yukio Kato
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, 920-1192, Japan
| | - Shinji Hagiwara
- Bioscience and Engineering laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa, 258-8577, Japan
| |
Collapse
|
25
|
Zhou S, Li J, Yu J, Yang L, Kuang X, Wang Z, Wang Y, Liu H, Lin G, He Z, Liu D, Wang Y. A facile and universal method to achieve liposomal remote loading of non-ionizable drugs with outstanding safety profiles and therapeutic effect. Acta Pharm Sin B 2021; 11:258-270. [PMID: 33532191 PMCID: PMC7838024 DOI: 10.1016/j.apsb.2020.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 10/28/2022] Open
Abstract
Liposomes have made remarkable achievements as drug delivery vehicles in the clinic. Liposomal products mostly benefited from remote drug loading techniques that succeeded in amphipathic and/or ionizable drugs, but seemed impracticable for nonionizable and poorly water-soluble therapeutic agents, thereby impeding extensive promising drugs to hitchhike liposomal vehicles for disease therapy. In this study, a series of weak acid drug derivatives were designed by a simplistic one step synthesis, which could be remotely loaded into liposomes by pH gradient method. Cabazitaxel (CTX) weak acid derivatives were selected to evaluate regarding its safety profiles, pharmacodynamics, and pharmacokinetics. CTX weak acid derivative liposomes were superior to Jevtana® in terms of safety profiles, including systemic toxicity, hematological toxicity, and potential central nerve toxicity. Specifically, it was demonstrated that liposomes had capacity to weaken potential toxicity of CTX on cortex and hippocampus neurons. Significant advantages of CTX weak acid derivative-loaded liposomes were achieved in prostate cancer and metastatic cancer therapy resulting from higher safety and elevated tolerated doses.
Collapse
Key Words
- AUC0‒t, area under the curve
- CR, creatinine
- CTX, cabazitaxel
- Cabazitaxel
- Cancer
- Chol, cholesterol
- DA, trans-2-butene-1,4-dicarboxylic acid
- DA-CTX, cabazitaxel trans-2-butene-1,4-dicarboxylic acid derivate
- DSPC, 1,2-dioctadecanoyl-sn-glycero-3-phophocholine
- DSPE-PEG2000, 2-distearoyl-snglycero-3-phosphoethanolamine-N-[methyl(polyethylene glycol)-2000
- EE, encapsulation efficiency
- EPR, enhanced permeability and retention
- GA, glutaric anhydride
- GA-CTX, cabazitaxel glutaric acid derivate
- Lung metastasis
- MED, minimum effective dose
- MPS, mononuclear phagocyte system
- MTD, maximum tolerated dose
- Non-ionizable drugs
- PCa, prostate cancer
- PSA, prostate-specific antigen
- Remote loading liposome
- SA, succinic anhydride
- SA-CTX, cabazitaxel succinic acid derivate
- Safety
- TI, therapeutic index
- Tolerated doses
- Weak acid derivatives
- lipo DA-CTX, DA-CTX liposome
- lipo GA-CTX, GA-CTX liposome
- lipo SA-CTX, SA-CTX liposome
- mCRPCa, metastatic castration-resistant prostate cancer
Collapse
Affiliation(s)
- Shuang Zhou
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jinbo Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jiang Yu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Liyuan Yang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiao Kuang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhenjie Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yingli Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hongzhuo Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Guimei Lin
- School of Pharmaceutical Science, Shandong University, Jinan 250012, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Dan Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yongjun Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
26
|
Harnessing the nano-bio interface: Application of membrane coating to long acting silica particles. Eur J Pharm Biopharm 2020; 158:382-389. [PMID: 33309845 DOI: 10.1016/j.ejpb.2020.12.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/10/2020] [Accepted: 12/02/2020] [Indexed: 01/15/2023]
Abstract
Interaction of conventional drug delivery systems such as polymeric or lipid based nano- and microparticles with the in vivo milieu has garnered significant interest, primarily to orchestrate immune escape and/or improve targeting. Surface modification with targeting ligands has been heavily relied upon for the mentioned purpose in the recent years. However, the surface modified particles can also activate the immune system. Large-scale manufacturing can also be a challenge, as surface modification needs to be reproducible. Furthermore, in vivo, the targeting is dependent on the receptor expression density and number of target sites, which adds to the pharmacokinetic variability of the constructs. An evolving paradigm to overcome complications of surface functionalization is the incorporation of bio-inspired topographies into these conventional delivery systems to enable them to better interact with biological systems. Biomimetic delivery systems combine the unique surface composition of cells or cell membranes, and versatility of synthetic nanoparticles. In this review, we focus on one such delivery system, silica particles, and explore their interaction with different biological membranes.
Collapse
|
27
|
Weber F, Rahnfeld L, Luciani P. Analytical profiling and stability evaluation of liposomal drug delivery systems: A rapid UHPLC-CAD-based approach for phospholipids in research and quality control. Talanta 2020; 220:121320. [DOI: 10.1016/j.talanta.2020.121320] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 06/19/2020] [Accepted: 06/22/2020] [Indexed: 01/26/2023]
|
28
|
Chen W, Boras B, Sung T, Hu W, Spilker ME, D'Argenio DZ. Predicting Chemotherapy-Induced Neutropenia and Granulocyte Colony-Stimulating Factor Response Using Model-Based In Vitro to Clinical Translation. AAPS J 2020; 22:143. [PMID: 33156437 PMCID: PMC7764847 DOI: 10.1208/s12248-020-00529-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 10/20/2020] [Indexed: 11/30/2022] Open
Abstract
The ability to predict the incidence of chemotherapy-induced neutropenia in early drug development can inform risk monitoring and mitigation strategies, as well as decisions on advancing compounds to clinical trials. In this report, a physiological model of granulopoiesis that incorporates the drug's mechanism of action on cell cycle proliferation of bone marrow progenitor cells was extended to include the action of the cytotoxic agents paclitaxel, carboplatin, doxorubicin, and gemcitabine. In vitro bone marrow studies were conducted with each compound, and results were used to determine the model's drug effect parameters. Population simulations were performed to predict the absolute neutrophil count (ANC) and incidence of neutropenia for each compound, which were compared to results reported in the literature. In addition, using the single agent in vitro study results, the model was able to predict ANC time course in response to paclitaxel plus carboplatin in combination, which compared favorably to the results reported in a phase 1 clinical trial of 46 patients (r2 = 0.70). Model simulations were used to compare the relative risk (RR) of neutropenia in patients with high baseline ANCs for five chemotherapeutic regimens: doxorubicin (RR = 0.59), paclitaxel plus carboplatin combination (RR = 0.079), carboplatin (RR = 0.047), paclitaxel (RR = 0.031), and gemcitabine (RR = 0.013). Finally, the model was applied to quantify the reduced incidence of neutropenia with coadministration of pegfilgrastim or filgrastim, for both paclitaxel and the combination of paclitaxel plus carboplatin. The model provides a framework for predicting clinical neutropenia using in vitro bone marrow studies of anticancer agents that may guide drug development decisions.
Collapse
Affiliation(s)
- Wenbo Chen
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, 90089, USA
| | - Britton Boras
- Pfizer Worldwide Research, Development and Medicine, San Diego, California, USA
| | - Tae Sung
- Pfizer Worldwide Research, Development and Medicine, San Diego, California, USA
| | - Wenyue Hu
- Pfizer Worldwide Research, Development and Medicine, San Diego, California, USA
| | - Mary E Spilker
- Pfizer Worldwide Research, Development and Medicine, San Diego, California, USA
| | - David Z D'Argenio
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, 90089, USA.
| |
Collapse
|
29
|
De Silva P, Saad MA, Thomsen HC, Bano S, Ashraf S, Hasan T. Photodynamic therapy, priming and optical imaging: Potential co-conspirators in treatment design and optimization - a Thomas Dougherty Award for Excellence in PDT paper. J PORPHYR PHTHALOCYA 2020; 24:1320-1360. [PMID: 37425217 PMCID: PMC10327884 DOI: 10.1142/s1088424620300098] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Photodynamic therapy is a photochemistry-based approach, approved for the treatment of several malignant and non-malignant pathologies. It relies on the use of a non-toxic, light activatable chemical, photosensitizer, which preferentially accumulates in tissues/cells and, upon irradiation with the appropriate wavelength of light, confers cytotoxicity by generation of reactive molecular species. The preferential accumulation however is not universal and, depending on the anatomical site, the ratio of tumor to normal tissue may be reversed in favor of normal tissue. Under such circumstances, control of the volume of light illumination provides a second handle of selectivity. Singlet oxygen is the putative favorite reactive molecular species although other entities such as nitric oxide have been credibly implicated. Typically, most photosensitizers in current clinical use have a finite quantum yield of fluorescence which is exploited for surgery guidance and can also be incorporated for monitoring and treatment design. In addition, the photodynamic process alters the cellular, stromal, and/or vascular microenvironment transiently in a process termed photodynamic priming, making it more receptive to subsequent additional therapies including chemo- and immunotherapy. Thus, photodynamic priming may be considered as an enabling technology for the more commonly used frontline treatments. Recently, there has been an increase in the exploitation of the theranostic potential of photodynamic therapy in different preclinical and clinical settings with the use of new photosensitizer formulations and combinatorial therapeutic options. The emergence of nanomedicine has further added to the repertoire of photodynamic therapy's potential and the convergence and co-evolution of these two exciting tools is expected to push the barriers of smart therapies, where such optical approaches might have a special niche. This review provides a perspective on current status of photodynamic therapy in anti-cancer and anti-microbial therapies and it suggests how evolving technologies combined with photochemically-initiated molecular processes may be exploited to become co-conspirators in optimization of treatment outcomes. We also project, at least for the short term, the direction that this modality may be taking in the near future.
Collapse
Affiliation(s)
- Pushpamali De Silva
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Mohammad A. Saad
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Hanna C. Thomsen
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Shazia Bano
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Shoaib Ashraf
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
30
|
Zhao Q, Sun XY, Wu B, Shang Y, Huang X, Dong H, Liu H, Chen W, Gui R, Li J. Construction of biomimetic silver nanoparticles in the treatment of lymphoma. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 119:111648. [PMID: 33321684 DOI: 10.1016/j.msec.2020.111648] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 10/10/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023]
Abstract
Lymphoma is a well-known malignant tumor in the human body. Although many anticancer drugs have been developed to improve the survival rate of patients, about 40% of patients continue to be recurrent or refractory, a key issue needing remedy. Therefore, it is necessary to identify alternative treatments to reduce the disease's mortality. To this effect, a new type of anti-lymphoma nanocomplex FA@RBCm-AgNPs was prepared using AgNPs as the core of nanoparticles along with the targeting molecule folic acid inserted erythrocyte membrane as the shell. The biomimetic properties of red blood cell membrane (RBCm) endow F-RAN with good biocompatibility as well as the ability to evade clearance of the reticuloendothelial system. In addition, F-RAN was modified with folic acid to actively and selectively identify tumor cells. In vivo and in vitro experiments demonstrate that F-RAN can inhibit lymphoma cells and induce apoptosis of stem cells while promoting apoptosis of lymphoma with no obvious side effects. Hence, F-RAN may serve as a new treatment for lymphoma.
Collapse
Affiliation(s)
- Qiangqiang Zhao
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha 410013, PR China; Department of Hematology, The Qinghai Provincial People's Hospital, Xining 810007, PR China
| | - Xiao Ying Sun
- Nursing School, Soochow University, Suzhou 215000, PR China; Department of Emergency, The Qinghai Provincial People's Hospital, Xining 810007, PR China
| | - Bin Wu
- Department of Transfusion Medicine, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China
| | - Yinghui Shang
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha 410013, PR China
| | - Xueyuan Huang
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha 410013, PR China
| | - Hang Dong
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha 410013, PR China
| | - Haiting Liu
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha 410013, PR China
| | - Wansong Chen
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, PR China
| | - Rong Gui
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha 410013, PR China.
| | - Jian Li
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha 410013, PR China.
| |
Collapse
|
31
|
Yu J, Mu Q, Perazzolo S, Griffin JI, Zhu L, McConnachie LA, Shen DD, Ho RJ. Novel Long-Acting Drug Combination Nanoparticles Composed of Gemcitabine and Paclitaxel Enhance Localization of Both Drugs in Metastatic Breast Cancer Nodules. Pharm Res 2020; 37:197. [PMID: 32968837 PMCID: PMC8686529 DOI: 10.1007/s11095-020-02888-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/21/2020] [Indexed: 12/25/2022]
Abstract
PURPOSE To develop drug-combination nanoparticles (DcNPs) composed of hydrophilic gemcitabine (G) and hydrophobic paclitaxel (T) and deliver both drugs to metastatic cancer cells. METHODS GT DcNPs were evaluated based on particle size and drug association efficiency (AE%). The effect of DcNP on GT plasma time-course and tissue distribution was characterized in mice and a pharmacokinetic model was developed. A GT distribution study into cancer nodules (derived from 4 T1 cells) was performed. RESULTS An optimized GT DcNP composition (d = 59.2 nm ±9.2 nm) was found to be suitable for IV formulation. Plasma exposure of G and T were enhanced 61-fold and 3.8-fold when given in DcNP form compared to the conventional formulation, respectively. Mechanism based pharmacokinetic modeling and simulation show that both G and T remain highly associated to DcNPs in vivo (G: 98%, T:75%). GT DcNPs have minimal distribution to healthy organs with selective distribution and retention in tumor burdened tissue. Tumor bearing lungs had a 5-fold higher tissue-to-plasma ratio of gemcitabine in GT DcNPs compared to healthy lungs. CONCLUSIONS DcNPs can deliver hydrophilic G and hydrophobic T together to cancer nodules and produce long acting exposure, likely due to stable GT association to DcNPs in vivo.
Collapse
Affiliation(s)
- Jesse Yu
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - Qingxin Mu
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - Simone Perazzolo
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - James I Griffin
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - Linxi Zhu
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - Lisa A McConnachie
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - Danny D Shen
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - Rodney Jy Ho
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA.
- Departments of Bioengineering, University of Washington, Seattle, Washington, 98195, USA.
| |
Collapse
|
32
|
Cutting-edge advances in therapy for the posterior segment of the eye: Solid lipid nanoparticles and nanostructured lipid carriers. Int J Pharm 2020; 589:119831. [PMID: 32877729 DOI: 10.1016/j.ijpharm.2020.119831] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/23/2020] [Accepted: 08/26/2020] [Indexed: 12/11/2022]
Abstract
Posterior segment eye diseases affect more than 300 million patients worldwide resulting in severe visual impairment. The treatments available are invasive, costly, present irregular effectiveness, and cause serious adverse effects. These drawbacks significantly reduce patient compliance. In the last decade, solid lipid nanoparticle (SLN) and nanostructured lipid carrier (NLC) have shown potential as innovative carriers for lipophilic drug substances to overcome hurdles in treating the eye posterior segment. This review shows the advantages of these formulations, focusing on their compatibility with ocular tissues, which increases the internalization of the drug substances. Additionally, SLN and NLC can reduce the clearance by the eye's protective mechanisms due to adhesive properties related to nanometric size. Therefore, these preparations may allow the treating of several ophthalmic diseases by topical administration, increasing the interval between doses. This feature can decrease adverse effects and enhance efficacy, ultimately improving patient compliance. Thus, this critical review presents the performance of the in vitro, ex vivo, and in vivo assays that support the potential of SLN and NLC to treat diseases of the posterior segment of the eye. These nanoparticles have shown to be promising alternative towards a major shift in developing ophthalmic products.
Collapse
|
33
|
Forterre A, Komuro H, Aminova S, Harada M. A Comprehensive Review of Cancer MicroRNA Therapeutic Delivery Strategies. Cancers (Basel) 2020; 12:E1852. [PMID: 32660045 PMCID: PMC7408939 DOI: 10.3390/cancers12071852] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
In the field of molecular oncology, microRNAs (miRNAs) and their role in regulating physiological processes and cancer pathogenesis have been a revolutionary discovery over the last decade. It is now considered that miRNA dysregulation influences critical molecular pathways involved in tumor progression, invasion, angiogenesis and metastasis in a wide range of cancer types. Hence, altering miRNA levels in cancer cells has promising potential as a therapeutic intervention, which is discussed in many other articles in this Special Issue. Some of the most significant hurdles in therapeutic miRNA usage are the stability and the delivery system. In this review, we cover a comprehensive update on the challenges and strategies for the development of therapeutic miRNA delivery systems that includes virus-based delivery, non-viral delivery (artificial lipid-based vesicles, polymer-based or chemical structures), and recently emerged extracellular vesicle (EV)-based delivery systems.
Collapse
Affiliation(s)
- Alexis Forterre
- UMR DIATHEC, EA 7294, Centre Européen d’Etude du Diabète, 67200 Strasbourg, France;
| | - Hiroaki Komuro
- Department of Cardiovascular Physiology, Tokyo Medical and Dental University, Tokyo 113-8510, Japan;
| | - Shakhlo Aminova
- Lyman Briggs College, Michigan State University, East Lansing, MI 48825, USA;
- Institute for Quantitative Health Sciences and Engineering (IQ), Michigan State University, East Lansing, MI 48824, USA
| | - Masako Harada
- Institute for Quantitative Health Sciences and Engineering (IQ), Michigan State University, East Lansing, MI 48824, USA
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
34
|
Zou H, Banerjee P, Leung SSY, Yan X. Application of Pharmacokinetic-Pharmacodynamic Modeling in Drug Delivery: Development and Challenges. Front Pharmacol 2020; 11:997. [PMID: 32719604 PMCID: PMC7348046 DOI: 10.3389/fphar.2020.00997] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 06/19/2020] [Indexed: 12/19/2022] Open
Abstract
With the advancement of technology, drug delivery systems and molecules with more complex architecture are developed. As a result, the drug absorption and disposition processes after administration of these drug delivery systems and engineered molecules become exceedingly complex. As the pharmacokinetic and pharmacodynamic (PK-PD) modeling allows for the separation of the drug-, carrier- and pharmacological system-specific parameters, it has been widely used to improve understanding of the in vivo behavior of these complex delivery systems and help their development. In this review, we summarized the basic PK-PD modeling theory in drug delivery and demonstrated how it had been applied to help the development of new delivery systems and modified large molecules. The linkage between PK and PD was highlighted. In particular, we exemplified the application of PK-PD modeling in the development of extended-release formulations, liposomal drugs, modified proteins, and antibody-drug conjugates. Furthermore, the model-based simulation using primary PD models for direct and indirect PD responses was conducted to explain the assertion of hypothetical minimal effective concentration or threshold in the exposure-response relationship of many drugs and its misconception. The limitations and challenges of the mechanism-based PK-PD model were also discussed.
Collapse
Affiliation(s)
- Huixi Zou
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Parikshit Banerjee
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Sharon Shui Yee Leung
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Xiaoyu Yan
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
35
|
Price LSL, Stern ST, Deal AM, Kabanov AV, Zamboni WC. A reanalysis of nanoparticle tumor delivery using classical pharmacokinetic metrics. SCIENCE ADVANCES 2020; 6:eaay9249. [PMID: 32832614 PMCID: PMC7439617 DOI: 10.1126/sciadv.aay9249] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 05/29/2020] [Indexed: 05/15/2023]
Abstract
Nanoparticle (NP) delivery to solid tumors has recently been questioned. To better understand the magnitude of NP tumor delivery, we reanalyzed published murine NP tumor pharmacokinetic (PK) data used in the Wilhelm et al. study. Studies included in their analysis reporting matched tumor and blood concentration versus time data were evaluated using classical PK endpoints and compared to the unestablished percent injected dose (%ID) in tumor metric from the Wilhelm et al. study. The %ID in tumor was poorly correlated with standard PK metrics that describe NP tumor delivery (AUCtumor/AUCblood ratio) and only moderately associated with maximal tumor concentration. The relative tumor delivery of NPs was ~100-fold greater as assessed by the standard AUCtumor/AUCblood ratio than by %ID in tumor. These results strongly suggest that PK metrics and calculations can influence the interpretation of NP tumor delivery and stress the need to properly validate novel PK metrics against traditional approaches.
Collapse
Affiliation(s)
- Lauren S. L. Price
- Carolina Center of Cancer Nanotechnology Excellence (C-CCNE), University of North Carolina, Chapel Hill, NC, USA
- Translational Oncology and Nanoparticle Drug Development (TOND2I) Lab, University of North Carolina, Chapel Hill, NC, USA
| | - Stephan T. Stern
- Nanotechnology Characterization Lab (NCL), Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Allison M. Deal
- UNC Lineberger Biostatistics Shared Resource, Chapel Hill, NC, USA
| | - Alexander V. Kabanov
- Carolina Center of Cancer Nanotechnology Excellence (C-CCNE), University of North Carolina, Chapel Hill, NC, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
- Center for Nanotechnology in Drug Delivery, UNC Eshelman School of Pharmacy, Chapel Hill, NC, USA
- Carolina Institute for Nanomedicine, University of North Carolina, Chapel Hill, NC, USA
| | - William C. Zamboni
- Carolina Center of Cancer Nanotechnology Excellence (C-CCNE), University of North Carolina, Chapel Hill, NC, USA
- Translational Oncology and Nanoparticle Drug Development (TOND2I) Lab, University of North Carolina, Chapel Hill, NC, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
- Center for Nanotechnology in Drug Delivery, UNC Eshelman School of Pharmacy, Chapel Hill, NC, USA
- Carolina Institute for Nanomedicine, University of North Carolina, Chapel Hill, NC, USA
- Corresponding author.
| |
Collapse
|
36
|
Adiwidjaja J, Boddy AV, McLachlan AJ. Physiologically-Based Pharmacokinetic Predictions of the Effect of Curcumin on Metabolism of Imatinib and Bosutinib: In Vitro and In Vivo Disconnect. Pharm Res 2020; 37:128. [PMID: 32529309 DOI: 10.1007/s11095-020-02834-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 04/26/2020] [Indexed: 12/14/2022]
Abstract
PURPOSE This study aimed to investigate the potential pharmacokinetic interactions between curcumin, imatinib and bosutinib, combining In Vitro and in silico methods. METHODS In Vitro metabolism of imatinib and bosutinib were investigated in pooled human liver microsomes and recombinant CYP3A4 enzyme in the presence and absence of curcumin and curcumin glucuronide using an LC-MS/MS assay for N-desmethyl metabolites. A physiologically-based pharmacokinetic (PBPK) model for curcumin formulated as solid lipid nanoparticles (SLN) was constructed using In Vitro glucuronidation kinetics and published clinical pharmacokinetic data. The potential effects of curcumin coadministration on systemic exposures of imatinib and bosutinib were predicted in silico using PBPK simulations. RESULTS Curcumin demonstrated potent reversible inhibition of cytochrome P450 (CYP)3A4-mediated N-demethylation of imatinib and bosutinib and CYP2C8-mediated metabolism of imatinib with inhibitory constants (ki,u) of ≤1.5 μmol. L-1. A confirmatory In Vitro study with paclitaxel, the 6α-hydroxylation of which is exclusively mediated by CYP2C8, was consistent with a potent inhibition of this enzyme by curcumin. Curcumin glucuronide also inhibited both CYP enzymes In Vitro, albeit to a lesser extent than that of curcumin. PBPK model simulations predicted that at recommended dosing regimens of SLN curcumin, coadministration would result in an increase in systemic exposures of imatinib and bosutinib of up to only 10%. CONCLUSION A PBPK model for curcumin in a SLN formulation was successfully developed. Although curcumin possesses a strong In Vitro inhibitory activity towards CYP3A4 and CYP2C8 enzymes, its interactions with imatinib and bosutinib were unlikely to be of clinical importance due to curcumin's poor bioavailability.
Collapse
Affiliation(s)
- Jeffry Adiwidjaja
- Sydney Pharmacy School, The University of Sydney, Sydney, NSW, 2006, Australia.
| | - Alan V Boddy
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, 5001, Australia
- University of South Australia Cancer Research Institute, University of South Australia, Adelaide, SA, 5000, Australia
| | - Andrew J McLachlan
- Sydney Pharmacy School, The University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
37
|
Chude-Okonkwo UAK, Maharaj BT, Vasilakos AV, Malekian R. Information-Theoretic Model and Analysis of Molecular Signaling in Targeted Drug Delivery. IEEE Trans Nanobioscience 2020; 19:270-284. [PMID: 31985433 DOI: 10.1109/tnb.2020.2968567] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Targeted drug delivery (TDD) modality promises a smart localization of appropriate dose of therapeutic drugs to the targeted part of the body at reduced system toxicity. To achieve the desired goals of TDD, accurate analysis of the system is important. Recent advances in molecular communication (MC) present prospects to analyzing the TDD process using engineering concepts and tools. Specifically, the MC platform supports the abstraction of TDD process as a communication engineering problem in which the injection and transportation of drug particles in the human body and the delivery to a specific tissue or organ can be analyzed using communication engineering tools. In this paper we stand on the MC platform to present the information-theoretic model and analysis of the TDD systems. We present a modular structure of the TDD system and the probabilistic models of the MC-abstracted modules in an intuitive manner. Simulated results of information-theoretic measures such as the mutual information are employed to analyze the performance of the TDD system. Results indicate that uncertainties in drug injection/release systems, nanoparticles propagation channel and nanoreceiver systems influence the mutual information of the system, which is relative to the system's bioequivalence measure.
Collapse
|
38
|
Abstract
Invasive fungal diseases caused by Candida albicans, Cryptococcus neoformans, and Aspergillus fumigatus have mortality rates ranging from 10 to 95%. Individual patient costs may exceed $100,000 in the United States. All antifungals in current use have serious limitations due to host toxicity and/or insufficient fungal cell killing that results in recurrent infections. Few new antifungal drugs have been introduced in the last 2 decades. Hence, there is a critical need for improved antifungal therapeutics. By targeting antifungal-loaded liposomes to α-mannans in the extracellular matrices secreted by these fungi, we dramatically reduced the effective dose of drug. Dectin-2-coated liposomes loaded with amphotericin B bound 50- to 150-fold more strongly to C. albicans, C. neoformans, and A. fumigatus than untargeted liposomes and killed these fungi more than an order of magnitude more efficiently. Targeting drug-loaded liposomes specifically to fungal cells has the potential to greatly enhance the efficacy of most antifungal drugs. Candida albicans, Cryptococcus neoformans, and Aspergillus fumigatus cause life-threatening candidiasis, cryptococcosis, and aspergillosis, resulting in several hundred thousand deaths annually. The patients at the greatest risk of developing these life-threatening invasive fungal infections have weakened immune systems. The vulnerable population is increasing due to rising numbers of immunocompromised individuals as a result of HIV infection or immunosuppressed individuals receiving anticancer therapies and/or stem cell or organ transplants. While patients are treated with antifungals such as amphotericin B, all antifungals have serious limitations due to lack of sufficient fungicidal effect and/or host toxicity. Even with treatment, 1-year survival rates are low. We explored methods of increasing drug effectiveness by designing fungicide-loaded liposomes specifically targeted to fungal cells. Most pathogenic fungi are encased in cell walls and exopolysaccharide matrices rich in mannans. Dectin-2 is a mammalian innate immune membrane receptor that binds as a dimer to mannans and signals fungal infection. We coated amphotericin-loaded liposomes with monomers of Dectin-2’s mannan-binding domain, sDectin-2. sDectin monomers were free to float in the lipid membrane and form dimers that bind mannan substrates. sDectin-2-coated liposomes bound orders of magnitude more efficiently to the extracellular matrices of several developmental stages of C. albicans, C. neoformans, and A. fumigatus than untargeted control liposomes. Dectin-2-coated amphotericin B-loaded liposomes reduced the growth and viability of all three species more than an order of magnitude more efficiently than untargeted control liposomes and dramatically decreased the effective dose. Future efforts focus on examining pan-antifungal targeted liposomal drugs in animal models of fungal diseases. IMPORTANCE Invasive fungal diseases caused by Candida albicans, Cryptococcus neoformans, and Aspergillus fumigatus have mortality rates ranging from 10 to 95%. Individual patient costs may exceed $100,000 in the United States. All antifungals in current use have serious limitations due to host toxicity and/or insufficient fungal cell killing that results in recurrent infections. Few new antifungal drugs have been introduced in the last 2 decades. Hence, there is a critical need for improved antifungal therapeutics. By targeting antifungal-loaded liposomes to α-mannans in the extracellular matrices secreted by these fungi, we dramatically reduced the effective dose of drug. Dectin-2-coated liposomes loaded with amphotericin B bound 50- to 150-fold more strongly to C. albicans, C. neoformans, and A. fumigatus than untargeted liposomes and killed these fungi more than an order of magnitude more efficiently. Targeting drug-loaded liposomes specifically to fungal cells has the potential to greatly enhance the efficacy of most antifungal drugs.
Collapse
|
39
|
Hu Y, Hammarlund-Udenaes M, Fridén M. Understanding the Influence of Nanocarrier-Mediated Brain Delivery on Therapeutic Performance Through Pharmacokinetic-Pharmacodynamic Modeling. J Pharm Sci 2019; 108:3425-3433. [PMID: 31163187 DOI: 10.1016/j.xphs.2019.05.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 12/16/2022]
Abstract
This study aimed at evaluating how encapsulation in a regular nanocarrier (NC) (providing extended circulation time) or in a brain-targeting NC (providing prolonged circulation time and increased brain uptake) may influence the therapeutic index compared with the unformulated drug and to explore the key parameters affecting therapeutic performance using a model-based approach. Pharmacokinetic (PK) models were built with chosen PK parameters. For a scenario where central effect depends on area under the unbound brain concentration curve and peripheral toxicity relates to peak unbound plasma concentration, dose-effect and drug-side effect curves were constructed, and the therapeutic index was evaluated. Regular NC improved the therapeutic index compared with the unformulated drug due to reduced peripheral toxicity, while brain-targeting NC enhanced the therapeutic index by lowering peripheral toxicity and increasing central effect. Decreasing drug release rate or systemic clearance of NC with drug still encapsulated could increase the therapeutic index. Also, a drug with shorter half-life would therapeutically benefit more from a NC encapsulation. This work provides insights into how a NC for brain delivery should be optimized to maximize the therapeutic performance and is helpful to predict if and to what extent a drug with certain PK properties would obtain therapeutic benefit from nanoencapsulation.
Collapse
Affiliation(s)
- Yang Hu
- Translational PKPD Research Group, Department of Pharmaceutical Biosciences, Associate Member of SciLife Lab, Uppsala University, Uppsala, Sweden
| | - Margareta Hammarlund-Udenaes
- Translational PKPD Research Group, Department of Pharmaceutical Biosciences, Associate Member of SciLife Lab, Uppsala University, Uppsala, Sweden.
| | - Markus Fridén
- Translational PKPD Research Group, Department of Pharmaceutical Biosciences, Associate Member of SciLife Lab, Uppsala University, Uppsala, Sweden; Department of Drug Metabolism and Pharmacokinetics, Early Respiratory, Inflammation and Autoimmunity, R&D Biopharmaceuticals, AstraZeneca R&D, Gothenburg, Sweden
| |
Collapse
|
40
|
Chadha J, Hussein S, Zhan Y, Shulman J, Brody J, Ratner L, Steinberg A. Liposomal Vincristine as a Bridge Therapy Prior to CAR-T Therapy in Relapsed and Refractory Diffuse Large B-Cell Lymphoma? Int J Hematol Oncol Stem Cell Res 2019; 13:102-107. [PMID: 31372204 PMCID: PMC6660481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
We report a case of a 76-year-old male with a history of relapsed and refractory diffuse large B-cell lymphoma (DLBCL).Our patient was initially treated with front line chemotherapy along with central nervous system (CNS) prophylaxis with complete response. He subsequently relapsed, was sensitive to second-line chemotherapy, and underwent autologous stem cell transplantation achieving a complete remission. Only a few months after transplant, the patient suffered his second relapse and was deemed a candidate for Chimeric Antigen Receptor T-Cell Therapy (CAR-T). Given his aggressive disease, combined with the time needed to generate CAR-T cells, a multidisciplinary team recommended to treat our patient with liposomal vincristine in combination with rituximab as a bridge therapy. Durable responses have been seen using liposomal vincristine based on results from a recent phase II trial in heavily pretreated patients with DLBCL1. This therapy was effective in stabilizing and reducing active disease in our patient. This case looks to illustrate the use of liposomal vincristine in combination with immunotherapy in a novel setting bridging highly selected patients with active and refractory lymphoma prior to CAR-T. Moreover, we expanded an additional therapeutic point, highlighting the importance of optimal disease control prior to CAR-T cell harvesting, as recent literature has shown that residual malignant cells in the pheresis product may be inadvertently be transfected with the CAR gene, resulting in resistance and further relapse2.
Collapse
Affiliation(s)
- Juskaran Chadha
- Department of Hematology & Medical Oncology, Lenox Hill Hospital Northwell Health, New York, NY, USA
| | | | - Yougen Zhan
- Tisch Cancer Institute, Mount Sinai Hospital, New York, NY, USA
| | - Jonah Shulman
- Tisch Cancer Institute, Mount Sinai Hospital, New York, NY, USA
| | - Joshua Brody
- Tisch Cancer Institute, Mount Sinai Hospital, New York, NY, USA
| | - Lynn Ratner
- Department of Hematology & Medical Oncology, Lenox Hill Hospital Northwell Health, New York, NY, USA
| | - Amir Steinberg
- Tisch Cancer Institute, Mount Sinai Hospital, New York, NY, USA
| |
Collapse
|
41
|
He H, Yuan D, Wu Y, Cao Y. Pharmacokinetics and Pharmacodynamics Modeling and Simulation Systems to Support the Development and Regulation of Liposomal Drugs. Pharmaceutics 2019; 11:E110. [PMID: 30866479 PMCID: PMC6471205 DOI: 10.3390/pharmaceutics11030110] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/25/2019] [Accepted: 02/28/2019] [Indexed: 12/27/2022] Open
Abstract
Liposomal formulations have been developed to improve the therapeutic index of encapsulated drugs by altering the balance of on- and off-targeted distribution. The improved therapeutic efficacy of liposomal drugs is primarily attributed to enhanced distribution at the sites of action. The targeted distribution of liposomal drugs depends not only on the physicochemical properties of the liposomes, but also on multiple components of the biological system. Pharmacokinetic⁻pharmacodynamic (PK⁻PD) modeling has recently emerged as a useful tool with which to assess the impact of formulation- and system-specific factors on the targeted disposition and therapeutic efficacy of liposomal drugs. The use of PK⁻PD modeling to facilitate the development and regulatory reviews of generic versions of liposomal drugs recently drew the attention of the U.S. Food and Drug Administration. The present review summarizes the physiological factors that affect the targeted delivery of liposomal drugs, challenges that influence the development and regulation of liposomal drugs, and the application of PK⁻PD modeling and simulation systems to address these challenges.
Collapse
Affiliation(s)
- Hua He
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China.
- Division of Pharmacotherapy and Experimental Therapeutics, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Dongfen Yuan
- Division of Pharmacotherapy and Experimental Therapeutics, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Yun Wu
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, 332 Bonner Hall, Buffalo, NY 14260, USA.
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
42
|
Bhowmik S, Bhowmick S, Maiti K, Chakra A, Shahi P, Jain D, Rajamannar T. Two multicenter Phase I randomized trials to compare the bioequivalence and safety of a generic doxorubicin hydrochloride liposome injection with Doxil ® or Caelyx ® in advanced ovarian cancer. Cancer Chemother Pharmacol 2018; 82:521-532. [PMID: 29995186 DOI: 10.1007/s00280-018-3643-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 07/04/2018] [Indexed: 10/28/2022]
Abstract
PURPOSE To compare the pharmacokinetic bioequivalence and safety of a generic pegylated liposomal doxorubicin formulation (SPIL DXR hydrochloride liposome injection) with that of the reference products, Caelyx or Doxil. METHODS Two open-label, two-way reference crossover studies were conducted in patients with ovarian cancer. Cmax, AUC0 - t, and AUC0-∞, Vd, and Cl for total, free, and encapsulated DXR were evaluated in 18 blood samples taken pre-dose (t = 0), at increasing time intervals over the following 14 days. A washout period of 28 days was observed before crossing over. RESULTS Studies 1 and 2 were completed by 24/29 and 41/60 patients, respectively. Pharmacokinetic data from 24 patients from each study established bioequivalence for free DXR in study 2, and for total and encapsulated DXR in both studies. Data from 29 and 54 patients, respectively, were included in the safety evaluation. Of these, 37 patients experienced 81 post-dose adverse events (40 related to the test product and 41 related to the reference product). In study 1, four patients were withdrawn owing to adverse events. Eleven patients experienced serious adverse events and one death occurred in study 2. CONCLUSIONS Bioequivalence between the test and the reference products was established for total and encapsulated DXR in both studies, and for free DXR in the study with the larger sample size (study 2). There were no significant differences between the safety profiles of the generic formulation and the reference products. No correlation was found between drug level and adverse events. TRIAL REGISTRATION Study 1 was registered retrospectively; registration number is NCT03055143, dated February 15, 2017. Study 2 registration number is NCT00862355, dated March 13, 2009.
Collapse
Affiliation(s)
- Shravanti Bhowmik
- Sun Pharmaceutical Industries Limited (SPIL), 17 B Mahal Industrial Estate, Mahakali Caves Road, Andheri (East), Mumbai, Maharashtra, 400093, India.
| | - Subhas Bhowmick
- Sun Pharmaceutical Industries Limited (SPIL), 17 B Mahal Industrial Estate, Mahakali Caves Road, Andheri (East), Mumbai, Maharashtra, 400093, India
| | - Kuntal Maiti
- Sun Pharmaceutical Industries Limited (SPIL), 17 B Mahal Industrial Estate, Mahakali Caves Road, Andheri (East), Mumbai, Maharashtra, 400093, India
| | - Amaresh Chakra
- Sun Pharmaceutical Industries Limited (SPIL), 17 B Mahal Industrial Estate, Mahakali Caves Road, Andheri (East), Mumbai, Maharashtra, 400093, India
| | - Pradeep Shahi
- Sun Pharmaceutical Industries Limited (SPIL), 17 B Mahal Industrial Estate, Mahakali Caves Road, Andheri (East), Mumbai, Maharashtra, 400093, India
| | - Deepak Jain
- Sun Pharmaceutical Industries Limited (SPIL), 17 B Mahal Industrial Estate, Mahakali Caves Road, Andheri (East), Mumbai, Maharashtra, 400093, India
| | - Thennati Rajamannar
- Sun Pharmaceutical Industries Limited (SPIL), 17 B Mahal Industrial Estate, Mahakali Caves Road, Andheri (East), Mumbai, Maharashtra, 400093, India
| |
Collapse
|
43
|
Rodallec A, Benzekry S, Lacarelle B, Ciccolini J, Fanciullino R. Pharmacokinetics variability: Why nanoparticles are not just magic-bullets in oncology. Crit Rev Oncol Hematol 2018; 129:1-12. [PMID: 30097227 DOI: 10.1016/j.critrevonc.2018.06.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/16/2018] [Accepted: 06/13/2018] [Indexed: 12/11/2022] Open
Abstract
Developing nanoparticles to improve the specificity of anticancer agents towards tumor tissue and to better control drug delivery is a rising strategy in oncology. An increasing number of forms (e.g., conjugated nanoparticles, liposomes, immunoliposomes…) are now available on the shelves and numerous other scaffolds (e.g., dendrimeres, nanospheres, squalenes …) are currently at various stages of development. However, as of today most nanoparticles made available remain lipidic carriers. Pharmacokinetic variability is a major, yet largely underestimated issue with liposomal nanoparticles. A wide variety of causes (e.g., tumor type and disease staging, comorbidities, patient's immune system) can explain this variability, which can in return negatively impact pharmacodynamic endpoints such as poor efficacy or severe toxicities. This review aims to cover the main causes for erratic pharmacokinetics observed with most nanoparticles, especially liposomes used in oncology. Should the main causes of such variability be identified, specific studies in non-clinical or clinical development stages could be undertaken using dedicated models (i.e., mechanistic or semi-mechanistic mathematical models such as PBPK approaches) to better describe nanoparticles pharmacokinetics and decipher PK/PD relationships. In addition, identifying relevant biomarkers or parameters likely to impact nanoparticles pharmacokinetics would allow for either the modification of their characteristics to reduce the influence of the expected variability during development phases or the development of biomarker-based adaptive dosing strategies to maintain an optimal efficacy/toxicity balance. Broadly, we call for the development of comprehensive distribution studies and state-of-the-art modeling support to better understand and anticipate nanoparticle pharmacokinetics in oncology.
Collapse
Affiliation(s)
- Anne Rodallec
- SMARTc unit, Center for Research on Cancer of Marseille (CRCM): UMR Inserm 1068, CNRS UMR 7258, Aix Marseille Université, Marseille, France
| | | | - Bruno Lacarelle
- SMARTc unit, Center for Research on Cancer of Marseille (CRCM): UMR Inserm 1068, CNRS UMR 7258, Aix Marseille Université, Marseille, France
| | - Joseph Ciccolini
- SMARTc unit, Center for Research on Cancer of Marseille (CRCM): UMR Inserm 1068, CNRS UMR 7258, Aix Marseille Université, Marseille, France
| | - Raphaelle Fanciullino
- SMARTc unit, Center for Research on Cancer of Marseille (CRCM): UMR Inserm 1068, CNRS UMR 7258, Aix Marseille Université, Marseille, France.
| |
Collapse
|
44
|
Rodallec A, Fanciullino R, Lacarelle B, Ciccolini J. Seek and destroy: improving PK/PD profiles of anticancer agents with nanoparticles. Expert Rev Clin Pharmacol 2018; 11:599-610. [PMID: 29768060 DOI: 10.1080/17512433.2018.1477586] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The Pharmacokinetics/pharmacodynamics (PK/PD) relationships with cytotoxics are usually based on a steepening concentration-effect relationship; the greater the drug amount, the greater the effect. The Maximum Tolerated Dose paradigm, finding the balance between efficacy, while keeping toxicities at their manageable level, has been the rule of thumb for the last 50-years. Developing nanodrugs is an appealing strategy to help broaden this therapeutic window. The fact that efficacy and toxicity with cytotoxics are intricately linked is primarily due to the complete lack of specificity toward the tumor tissue during their distribution phase. Because nanoparticles are expected to better target tumor tissue while sparing healthy cells, accumulating large amounts of cytotoxics in tumors could be achieved in a safer way. Areas covered: This review aims at presenting how nanodrugs present unique features leading to reconsidering PK/PD relationships of anticancer agents. Expert commentary: The constant interplay between carrier PK, interactions with cancer cells, payload release, payload PK, target expression and target engagement, makes picturing the exact PK/PD relationships of nanodrugs particularly challenging. However, those improved PK/PD relationships now make the once contradictory higher efficacy and lower toxicities requirement an achievable goal in cancer patients.
Collapse
Affiliation(s)
- Anne Rodallec
- a SMARTc Unit, Pharmacokinetics Laboratory, Inserm UMR U1068 Centre de Recherche en Cancérologie de Marseille , Aix-Marseille Universite , Marseille , France
| | - Raphaelle Fanciullino
- a SMARTc Unit, Pharmacokinetics Laboratory, Inserm UMR U1068 Centre de Recherche en Cancérologie de Marseille , Aix-Marseille Universite , Marseille , France
| | - Bruno Lacarelle
- a SMARTc Unit, Pharmacokinetics Laboratory, Inserm UMR U1068 Centre de Recherche en Cancérologie de Marseille , Aix-Marseille Universite , Marseille , France
| | - Joseph Ciccolini
- a SMARTc Unit, Pharmacokinetics Laboratory, Inserm UMR U1068 Centre de Recherche en Cancérologie de Marseille , Aix-Marseille Universite , Marseille , France
| |
Collapse
|
45
|
Sandzhieva AV, Sybachin AV, Zaborova OV, Yaroslavov AA. Competitive Reactions in Three-Component System Cationic Colloid–Anionic Liposome–Protein. POLYMER SCIENCE SERIES B 2018. [DOI: 10.1134/s1560090418030090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
46
|
Merino M, Zalba S, Garrido MJ. Immunoliposomes in clinical oncology: State of the art and future perspectives. J Control Release 2018; 275:162-176. [DOI: 10.1016/j.jconrel.2018.02.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/09/2018] [Accepted: 02/10/2018] [Indexed: 02/02/2023]
|
47
|
Cholesterol modulates the liposome membrane fluidity and permeability for a hydrophilic molecule. Food Chem Toxicol 2018; 113:40-48. [PMID: 29337230 DOI: 10.1016/j.fct.2018.01.017] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 01/27/2023]
Abstract
The effect of cholesterol (CHOL) content on the permeability and fluidity of dipalmitoylphosphatidylcholine (DPPC) liposome membrane was investigated. Liposomes encapsulating sulforhodamine B (SRB), a fluorescent dye, were prepared by reverse phase evaporation technique (REV) at various DPPC:CHOL molar ratios (from 100:0 to 100:100). The release kinetics of SRB was studied during 48 h in buffer (pH 7.4) containing NaCl at 37 °C. The DPPC:CHOL formulations were also characterized for their size, polydispersity index and morphology. Increasing CHOL concentration induced an increase in the mean liposomes size accompanying with a shape transition from irregular to nanosized, regular and spherical vesicles. The release kinetics of SRB showed a biphasic pattern; the release data was then analyzed using different mathematical models. On the overall, the SRB release was governed by a non-Fickian diffusion during the first period (0-10 h) while it followed a Fickian diffusion between 10 and 48 h. Changes in DPPC liposome membrane fluidity of various batches (CHOL% 0, 10, 20, 30 and 100) were monitored by using 5- and 16 doxyl stearic acids (DSA) as spin labels. CHOL induced a decrease in the bilayer fluidity. Concisely, CHOL represents a critical component in modulating the release of hydrophilic molecules from lipid vesicles.
Collapse
|
48
|
Steffes VM, Murali MM, Park Y, Fletcher BJ, Ewert KK, Safinya CR. Distinct solubility and cytotoxicity regimes of paclitaxel-loaded cationic liposomes at low and high drug content revealed by kinetic phase behavior and cancer cell viability studies. Biomaterials 2017; 145:242-255. [PMID: 28889081 PMCID: PMC5610109 DOI: 10.1016/j.biomaterials.2017.08.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 07/19/2017] [Accepted: 08/14/2017] [Indexed: 01/20/2023]
Abstract
Lipid-based particles are used worldwide in clinical trials as carriers of hydrophobic paclitaxel (PTXL) for cancer chemotherapy, albeit with little improvement over the standard-of-care. Improving efficacy requires an understanding of intramembrane interactions between PTXL and lipids to enhance PTXL solubilization and suppress PTXL phase separation into crystals. We studied the solubility of PTXL in cationic liposomes (CLs) composed of positively charged 2,3-dioleyloxypropyltrimethylammonium chloride (DOTAP) and neutral 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine (DOPC) as a function of PTXL membrane content and its relation to efficacy. Time-dependent kinetic phase diagrams were generated from observations of PTXL crystal formation by differential-interference-contrast microscopy. Furthermore, a new synchrotron small-angle x-ray scattering in situ methodology applied to DOTAP/DOPC/PTXL membranes condensed with DNA enabled us to detect the incorporation and time-dependent depletion of PTXL from membranes by measurements of variations in the membrane interlayer and DNA interaxial spacings. Our results revealed three regimes with distinct time scales for PTXL membrane solubility: hours for >3 mol% PTXL (low), days for ≈ 3 mol% PTXL (moderate), and ≥20 days for < 3 mol% PTXL (long-term). Cell viability experiments on human cancer cell lines using CLPTXL nanoparticles (NPs) in the distinct CLPTXL solubility regimes reveal an unexpected dependence of efficacy on PTXL content in NPs. Remarkably, formulations with lower PTXL content and thus higher stability show higher efficacy than those formulated at the membrane solubility limit of ≈3 mol% PTXL (which has been the focus of most previous physicochemical studies and clinical trials of PTXL-loaded CLs). Furthermore, an additional high-efficacy regime is seen on occasion for liposome compositions with PTXL ≥9 mol% applied to cells at short time scales (hours) after formation. At longer time scales (days), CLPTXL NPs with ≥3 mol% PTXL lose efficacy while formulations with 1-2 mol% PTXL maintain high efficacy. Our findings underscore the importance of understanding the relationship of the kinetic phase behavior and physicochemical properties of CLPTXL NPs to efficacy.
Collapse
Affiliation(s)
- Victoria M Steffes
- Chemistry and Biochemistry Department, University of California, Santa Barbara, CA 93106, USA; Materials Department, University of California, Santa Barbara, CA 93106, USA
| | - Meena M Murali
- Materials Department, University of California, Santa Barbara, CA 93106, USA
| | - Yoonsang Park
- Materials Department, University of California, Santa Barbara, CA 93106, USA
| | - Bretton J Fletcher
- Materials Department, University of California, Santa Barbara, CA 93106, USA
| | - Kai K Ewert
- Materials Department, University of California, Santa Barbara, CA 93106, USA
| | - Cyrus R Safinya
- Materials Department, University of California, Santa Barbara, CA 93106, USA; Physics Department, University of California, Santa Barbara, CA 93106, USA; Molecular, Cellular & Developmental Biology Department, University of California, Santa Barbara, CA 93106, USA.
| |
Collapse
|
49
|
The Impact of Liposomal Formulations on the Release and Brain Delivery of Methotrexate: An In Vivo Microdialysis Study. J Pharm Sci 2017; 106:2606-2613. [DOI: 10.1016/j.xphs.2017.03.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 03/10/2017] [Accepted: 03/10/2017] [Indexed: 01/16/2023]
|
50
|
A Biomimetic Microfluidic Tumor Microenvironment Platform Mimicking the EPR Effect for Rapid Screening of Drug Delivery Systems. Sci Rep 2017; 7:9359. [PMID: 28839211 PMCID: PMC5571192 DOI: 10.1038/s41598-017-09815-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 07/31/2017] [Indexed: 01/18/2023] Open
Abstract
Real-time monitoring of tumor drug delivery in vivo is a daunting challenge due to the heterogeneity and complexity of the tumor microenvironment. In this study, we developed a biomimetic microfluidic tumor microenvironment (bMTM) comprising co-culture of tumor and endothelial cells in a 3D environment. The platform consists of a vascular compartment featuring a network of vessels cultured with endothelial cells forming a complete lumen under shear flow in communication with 3D solid tumors cultured in a tumor compartment. Endothelial cell permeability to both small dye molecules and large liposomal drug carriers were quantified using fluorescence microscopy. Endothelial cell intercellular junction formation was characterized by immunostaining. Endothelial cell permeability significantly increased in the presence of either tumor cell conditioned media (TCM) or tumor cells. The magnitude of this increase in permeability was significantly higher in the presence of metastatic breast tumor cells as compared to non-metastatic ones. Immunostaining revealed impaired endothelial cell-cell junctions in the presence of either metastatic TCM or metastatic tumor cells. Our findings indicate that the bMTM platform mimics the tumor microenvironment including the EPR effect. This platform has a significant potential in applications such as cell-cell/cell-drug carrier interaction studies and rapid screening of cancer drug therapeutics/carriers.
Collapse
|