1
|
Yamamoto E, Nikko M, Miyatsuji M, Ando D, Miyazaki T, Koide T, Sato Y. Physicochemical profiling of nanomedicines using centrifugal field flow fractionation. Int J Pharm 2024; 663:124571. [PMID: 39128622 DOI: 10.1016/j.ijpharm.2024.124571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/18/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Nanomedicines comprise multiple components, and particle density is considered an important property that regulates the biodistribution of administered nanomedicines. The density of nanoparticles is characterized by centrifugal methods, such as analytical ultracentrifugation. Particle size and distribution are key physicochemical and quality attributes of nanomedicines. In this study, we developed a novel profiling method applicable to liposomes and lipid nanoparticles (LNPs), based on particle size and density, using centrifugal field-flow fractionation (CF3). We evaluated the elution profiles of PEGylated liposomes of different sizes with various doxorubicin (DOX)-loading amounts using CF3. This method was applied to evaluate the drug release of DOX-loaded liposomes, intra- and inter-batch variability, reconstitution reproducibility of AmBisome®, and elution characteristics of LNPs in COVID-19 vaccines (Comirnaty® and SpikevaxTM). The data obtained in the present study underscore the significance of the proposed methodology and highlight the importance of profiling and characterizing liposomes and LNPs using CF3 fractograms and a multi-angle light-scattering detector.
Collapse
Affiliation(s)
- Eiichi Yamamoto
- Division of Medical Devices, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-9501, Japan.
| | - Masataka Nikko
- Shimadzu Corporation, 1 Nishinokyo Kuwabara-cho, Nakagyo-ku, Kyoto 604-8511, Japan
| | - Megumi Miyatsuji
- Division of Drugs, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-9501, Japan
| | - Daisuke Ando
- Division of Drugs, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-9501, Japan
| | - Tamaki Miyazaki
- Division of Drugs, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-9501, Japan
| | - Tatsuo Koide
- Division of Drugs, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-9501, Japan
| | - Yoji Sato
- Division of Drugs, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-9501, Japan
| |
Collapse
|
2
|
Carretta A, Moscardini A, Signore G, Debellis D, Catalano F, Marotta R, Palmieri V, Tedeschi G, Scipioni L, Pozzi D, Caracciolo G, Beltram F, Cardarelli F. The supramolecular processing of liposomal doxorubicin hinders its therapeutic efficacy in cells. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200836. [PMID: 39050990 PMCID: PMC11268116 DOI: 10.1016/j.omton.2024.200836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/06/2024] [Accepted: 06/13/2024] [Indexed: 07/27/2024]
Abstract
The successful trajectory of liposome-encapsulated doxorubicin (e.g., Doxil, which has been approved by the U.S. Food and Drug Administration) as an anticancer nanodrug in clinical applications is contradicted by in vitro cell viability data that highlight its reduced efficacy in promoting cell death compared with non-encapsulated doxorubicin. No reports to date have provided a mechanistic explanation for this apparently discordant evidence. Taking advantage of doxorubicin intrinsic fluorescence and time-resolved optical microscopy, we analyze the uptake and intracellular processing of liposome-encapsulated doxorubicin (L-DOX) in several in vitro cellular models. Cell entry of L-DOX was found to lead to a rapid (seconds to minutes), energy- and temperature-independent release of crystallized doxorubicin nanorods into the cell cytoplasm, which then disassemble into a pool of fibril-shaped derivatives capable of crossing the cellular membrane while simultaneously releasing active drug monomers. Thus, a steady state is rapidly established in which the continuous supply of crystal nanorods from incoming liposomes is counteracted by a concentration-guided efflux in the extracellular medium of fibril-shaped derivatives and active drug monomers. These results demonstrate that liposome-mediated delivery is constitutively less efficient than isolated drug in establishing favorable conditions for drug retention in the cell. In addition to explaining previous contradictory evidence, present results impose careful rethinking of the synthetic identity of encapsulated anticancer drugs.
Collapse
Affiliation(s)
- Annalisa Carretta
- Scuola Normale Superiore, Laboratorio NEST, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Aldo Moscardini
- Scuola Normale Superiore, Laboratorio NEST, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Giovanni Signore
- Biochemistry Unit, Department of Biology, University of Pisa, via San Zeno 51, 56123 Pisa, Italy
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy
| | - Doriana Debellis
- Electron Microscopy Facility, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy
| | - Federico Catalano
- Electron Microscopy Facility, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy
| | - Roberto Marotta
- Electron Microscopy Facility, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy
| | - Valentina Palmieri
- Istituto dei Sistemi Complessi ISC CNR, Via dei Taurini 19, 00185 Rome, Italy
| | - Giulia Tedeschi
- Laboratory for Fluorescence Dynamics, Biomedical Engineering Department, University of California, Irvine, Irvine, CA, USA
| | - Lorenzo Scipioni
- Laboratory for Fluorescence Dynamics, Biomedical Engineering Department, University of California, Irvine, Irvine, CA, USA
| | - Daniela Pozzi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Giulio Caracciolo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Fabio Beltram
- Scuola Normale Superiore, Laboratorio NEST, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Francesco Cardarelli
- Scuola Normale Superiore, Laboratorio NEST, Piazza San Silvestro 12, 56127 Pisa, Italy
| |
Collapse
|
3
|
Lim S, Kwak M, Kang J, Cesaire M, Tang K, Robey RW, Frye WJE, Karim B, Butcher D, Lizak MJ, Dalmage M, Foster B, Nuechterlein N, Eberhart C, Cimino PJ, Gottesman MM, Jackson S. Ibrutinib disrupts blood-tumor barrier integrity and prolongs survival in rodent glioma model. Acta Neuropathol Commun 2024; 12:56. [PMID: 38589905 PMCID: PMC11003129 DOI: 10.1186/s40478-024-01763-6] [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: 11/20/2023] [Accepted: 03/24/2024] [Indexed: 04/10/2024] Open
Abstract
In malignant glioma, cytotoxic drugs are often inhibited from accessing the tumor site due to the blood-tumor barrier (BTB). Ibrutinib, FDA-approved lymphoma agent, inhibits Bruton tyrosine kinase (BTK) and has previously been shown to independently impair aortic endothelial adhesion and increase rodent glioma model survival in combination with cytotoxic therapy. Yet additional research is required to understand ibrutinib's effect on BTB function. In this study, we detail baseline BTK expression in glioma cells and its surrounding vasculature, then measure endothelial junctional expression/function changes with varied ibrutinib doses in vitro. Rat glioma cells and rodent glioma models were treated with ibrutinib alone (1-10 µM and 25 mg/kg) and in combination with doxil (10-100 µM and 3 mg/kg) to assess additive effects on viability, drug concentrations, tumor volume, endothelial junctional expression and survival. We found that ibrutinib, in a dose-dependent manner, decreased brain endothelial cell-cell adhesion over 24 h, without affecting endothelial cell viability (p < 0.005). Expression of tight junction gene and protein expression was decreased maximally 4 h after administration, along with inhibition of efflux transporter, ABCB1, activity. We demonstrated an additive effect of ibrutinib with doxil on rat glioma cells, as seen by a significant reduction in cell viability (p < 0.001) and increased CNS doxil concentration in the brain (56 ng/mL doxil alone vs. 74.6 ng/mL combination, p < 0.05). Finally, Ibrutinib, combined with doxil, prolonged median survival in rodent glioma models (27 vs. 16 days, p < 0.0001) with brain imaging showing a - 53% versus - 75% volume change with doxil alone versus combination therapy (p < 0.05). These findings indicate ibrutinib's ability to increase brain endothelial permeability via junctional disruption and efflux inhibition, to increase BTB drug entry and prolong rodent glioma model survival. Our results motivate the need to identify other BTB modifiers, all with the intent of improving survival and reducing systemic toxicities.
Collapse
Affiliation(s)
- Sanghee Lim
- Develomental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Building 10, Room 7D45, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Minhye Kwak
- Develomental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Building 10, Room 7D45, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Jeonghan Kang
- Develomental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Building 10, Room 7D45, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Melissa Cesaire
- Develomental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Building 10, Room 7D45, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Kayen Tang
- Develomental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Building 10, Room 7D45, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Robert W Robey
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, 20892, USA
| | - William J E Frye
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, 20892, USA
| | - Baktiar Karim
- Molecular Histopathology Laboratory, Frederick National Laboratory, Leidos Biomedical Research, Frederick, MD, 21702, USA
| | - Donna Butcher
- Molecular Histopathology Laboratory, Frederick National Laboratory, Leidos Biomedical Research, Frederick, MD, 21702, USA
| | - Martin J Lizak
- NIH MRI Research Facility and Mouse Imaging Facility, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Bethesda, MD, 20814, USA
| | - Mahalia Dalmage
- Develomental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Building 10, Room 7D45, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Brandon Foster
- Develomental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Building 10, Room 7D45, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Nicholas Nuechterlein
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Charles Eberhart
- Neuropathology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Bethesda, MD, 20892, USA
| | - Patrick J Cimino
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael M Gottesman
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, 20892, USA
| | - Sadhana Jackson
- Develomental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Building 10, Room 7D45, 10 Center Drive, Bethesda, MD, 20892, USA.
| |
Collapse
|
4
|
Shimoyama S, Okada K, Kimura T, Morohashi Y, Nakayama S, Kemmochi S, Makita-Suzuki K, Matulonis UA, Mori M. FF-10850, a Novel Liposomal Topotecan Achieves Superior Antitumor Activity via Macrophage- and Ammonia-Mediated Payload Release in the Tumor Microenvironment. Mol Cancer Ther 2023; 22:1454-1464. [PMID: 37683276 PMCID: PMC10690090 DOI: 10.1158/1535-7163.mct-23-0099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 08/03/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023]
Abstract
Topotecan, an approved treatment for refractory or recurrent ovarian cancer, has clinical limitations such as rapid clearance and hematologic toxicity. To overcome these limitations and maximize clinical benefit, we designed FF-10850, a dihydrosphingomyelin-based liposomal topotecan. FF-10850 demonstrated superior antitumor activity to topotecan in ovarian cancer cell line-based xenograft models, as well as in a clinically relevant DF181 platinum-refractory ovarian cancer patient-derived xenograft model. The safety profile was also improved with mitigation of hematologic toxicity. The improved antitumor activity and safety profile are achieved via its preferential accumulation and payload release triggered in the tumor microenvironment. Our data indicate that tumor-associated macrophages internalize FF-10850, resulting in complete payload release. The release mechanism also appears to be mediated by high ammonia concentration resulting from glutaminolysis, which is activated by tumor metabolic reprogramming. In ammonia-rich conditions, FF-10850 released payload more rapidly and to a greater extent than liposomal doxorubicin, a currently approved treatment for ovarian cancer. FF-10850 significantly enhanced antitumor activity in combination with carboplatin or PARP inhibitor without detrimental effects on body weight in murine xenograft models, and demonstrated synergistic antitumor activity combined with anti-PD-1 antibody with the development of tumor antigen-specific immunity. These results support phase I investigation of FF-10850 for the treatment of solid tumors including ovarian cancer (NCT04047251), and further evaluation in combination settings.
Collapse
Affiliation(s)
| | - Ken Okada
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, Kanagawa, Japan
| | - Toshifumi Kimura
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, Kanagawa, Japan
| | - Yasushi Morohashi
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, Kanagawa, Japan
| | - Shinji Nakayama
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, Kanagawa, Japan
| | - Sayaka Kemmochi
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, Kanagawa, Japan
| | - Keiko Makita-Suzuki
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, Kanagawa, Japan
| | - Ursula A. Matulonis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mikinaga Mori
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, Kanagawa, Japan
| |
Collapse
|
5
|
Watanabe A, Murayama S, Karasawa K, Yamamoto E, Morikawa S, Takita R, Murata S, Kato M. A Simple and Easy Method of Monitoring Doxorubicin Release from a Liposomal Drug Formulation in the Serum Using Fluorescence Spectroscopy. Chem Pharm Bull (Tokyo) 2019; 67:367-371. [PMID: 30930441 DOI: 10.1248/cpb.c18-00868] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Formulation of a drug as liposomes facilitates its delivery to the disease target. Rightly, liposomes are gaining popularity in the medical field. In order for the drug to show efficacy, release of the encapsulated drug from the liposome at the target site is required. However, the release is affected by the permeability of the lipid bilayer of the liposome, and it is important to examine the effect of the surrounding environment on the permeability. In this study, we showed the usefulness of fluorescence analysis, especially fluorescence fingerprint, for a rapid and simple monitoring of release of an encapsulated anticancer drug (doxorubicin) from its liposomal formulation (DOXIL). Our result indicated that the release is accelerated by the existence of membrane permeable ions, such as tris(hydroxymethyl)aminomethane, and blood proteins like albumin. Hence, monitoring of doxorubicin release by fluorescence analysis is useful for the efficacy evaluation of DOXIL in a biomimetic environment.
Collapse
Affiliation(s)
- Ayako Watanabe
- One-Stop Sharing Facility Center for Future Drug Discoveries, Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Shuhei Murayama
- Devision of Bioanalytical Chemistry, School of Pharmacy, Showa University
| | - Koji Karasawa
- Devision of Bioanalytical Chemistry, School of Pharmacy, Showa University
| | - Eiichi Yamamoto
- Analytical Research, Pharmaceutical Science & Technology Unit, Medicine Development Center, Eisai Co., Ltd
| | | | - Ryo Takita
- One-Stop Sharing Facility Center for Future Drug Discoveries, Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Shigeo Murata
- One-Stop Sharing Facility Center for Future Drug Discoveries, Graduate School of Pharmaceutical Sciences, The University of Tokyo.,Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Masaru Kato
- One-Stop Sharing Facility Center for Future Drug Discoveries, Graduate School of Pharmaceutical Sciences, The University of Tokyo.,Devision of Bioanalytical Chemistry, School of Pharmacy, Showa University
| |
Collapse
|
6
|
Abstract
Over the past few decades, liposome drug delivery systems (liposome DDS) have attracted much attention as the most advanced DDS. Efficacy and toxicity profiles of liposomes are based on their characteristic pharmacokinetics, drug release, and disposition after administration. Many attempts have been made to develop these systems especially as liposomal anti-cancer drugs. In the development of liposome DDS, identification of critical quality attributes and establishment of a control strategy to ensure consistent drug product quality are crucial. Among the quality attributes, particle size, drug encapsulation, and drug release from liposomes would affect their in vivo pharmacokinetic and pharmacodynamic properties. Thus these features need to be evaluated with appropriate analytical methods to confirm the quality and performance of the drug products. This article focuses on drug release from liposomes and reviews the effects of physicochemical properties of loaded drugs on release, simulation of drug release from liposomes, and design of a simulated body fluid for drug release assay of drug products.
Collapse
Affiliation(s)
- Eiichi Yamamoto
- Analytical Research, Pharmaceutical Science & Technology Unit, Medicine Development Center, Eisai Co., Ltd
| |
Collapse
|