1
|
Ischyropoulou M, Sabljo K, Schneider L, Niemeyer CM, Napp J, Feldmann C, Alves F. High-Load Gemcitabine Inorganic-Organic Hybrid Nanoparticles as an Image-Guided Tumor-Selective Drug-Delivery System to Treat Pancreatic Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2305151. [PMID: 37587542 DOI: 10.1002/adma.202305151] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/15/2023] [Indexed: 08/18/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a devastating prognosis without effective treatment options. Thus, there is an urgent need for more effective and safe therapies. Here, inorganic-organic hybrid nanoparticles (GMP-IOH-NPs) are presented as a novel drug-delivery system for the selective delivery of extraordinarily high concentrations of gemcitabine monophosphate (GMP), not only to the primary tumor but also to metastatic sites. GMP-IOH-NPs have a composition of [ZrO]2+ [GMP]2 - with GMP as drug anion (76% of total IOH-NP mass). Multiscale fluorescence imaging confirms an efficient uptake in tumor cells, independent of the activity of the human-equilibrative-nucleoside transporter (hENT1), being responsible for gemcitabine (GEM) transport into cells and a key factor for GEM resistance. Delivering already phosphorylated GMP via GMP-IOH-NPs into tumor cells also allows the cellular resistance induced by the downregulation of deoxycytidine kinase to be overcome. GMP-IOH-NPs show high accumulation in tumor lesions and only minor liver trapping when given intraperitoneally. GMP-IOH-NPs result in a higher antitumor efficacy compared to free GEM, which is further enhanced applying cetuximab-functionalized GMP-CTX-IOH-NPs. By maximizing the therapeutic benefits with high drug load, tumor-specific delivery, minimizing undesired side effects, overcoming mechanisms of chemoresistance, and preventing systemic GEM inactivation, GMP-IOH-NPs are anticipated to have a high chance to significantly improve current PDAC-patient outcome.
Collapse
Affiliation(s)
- Myrto Ischyropoulou
- Department of Diagnostic and Interventional Radiology, University Medical Center Goettingen (UMG), Robert-Koch-Strasse 40, 37075, Goettingen, Germany
- Max Planck Institute for Multidisciplinary Sciences (MPI-NAT), Hermann-Rein-Strasse 3, 37075, Goettingen, Germany
| | - Kristina Sabljo
- Institute of Inorganic Chemistry, Karlsruhe Institute of Technology (KIT), Engesserstrasse 15, 76131, Karlsruhe, Germany
| | - Leonie Schneider
- Institute for Biological Interfaces 1, Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Christof M Niemeyer
- Institute for Biological Interfaces 1, Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Joanna Napp
- Department of Diagnostic and Interventional Radiology, University Medical Center Goettingen (UMG), Robert-Koch-Strasse 40, 37075, Goettingen, Germany
- Max Planck Institute for Multidisciplinary Sciences (MPI-NAT), Hermann-Rein-Strasse 3, 37075, Goettingen, Germany
| | - Claus Feldmann
- Institute of Inorganic Chemistry, Karlsruhe Institute of Technology (KIT), Engesserstrasse 15, 76131, Karlsruhe, Germany
| | - Frauke Alves
- Department of Diagnostic and Interventional Radiology, University Medical Center Goettingen (UMG), Robert-Koch-Strasse 40, 37075, Goettingen, Germany
- Max Planck Institute for Multidisciplinary Sciences (MPI-NAT), Hermann-Rein-Strasse 3, 37075, Goettingen, Germany
- Department of Haematology and Medical Oncology, University Medical Center Goettingen (UMG), Robert-Koch-Strasse 40, 37075, Goettingen, Germany
| |
Collapse
|
2
|
Raza F, Evans L, Motallebi M, Zafar H, Pereira-Silva M, Saleem K, Peixoto D, Rahdar A, Sharifi E, Veiga F, Hoskins C, Paiva-Santos AC. Liposome-based diagnostic and therapeutic applications for pancreatic cancer. Acta Biomater 2023; 157:1-23. [PMID: 36521673 DOI: 10.1016/j.actbio.2022.12.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022]
Abstract
Pancreatic cancer is one of the harshest and most challenging cancers to treat, often labeled as incurable. Chemotherapy continues to be the most popular treatment yet yields a very poor prognosis. The main barriers such as inefficient drug penetration and drug resistance, have led to the development of drug carrier systems. The benefits, ease of fabrication and modification of liposomes render them as ideal future drug delivery systems. This review delves into the versatility of liposomes to achieve various mechanisms of treatment for pancreatic cancer. Not only are there benefits of loading chemotherapy drugs and targeting agents onto liposomes, as well as mRNA combined therapy, but liposomes have also been exploited for immunotherapy and can be programmed to respond to photothermal therapy. Multifunctional liposomal formulations have demonstrated significant pre-clinical success. Functionalising drug-encapsulated liposomes has resulted in triggered drug release, specific targeting, and remodeling of the tumor environment. Suppressing tumor progression has been achieved, due to their ability to more efficiently and precisely deliver chemotherapy. Currently, no multifunctional surface-modified liposomes are clinically approved for pancreatic cancer thus we aim to shed light on the trials and tribulations and progress so far, with the hope for liposomal therapy in the future and improved patient outcomes. STATEMENT OF SIGNIFICANCE: Considering that conventional treatments for pancreatic cancer are highly associated with sub-optimal performance and systemic toxicity, the development of novel therapeutic strategies holds outmost relevance for pancreatic cancer management. Liposomes are being increasingly considered as promising nanocarriers for providing not only an early diagnosis but also effective, highly specific, and safer treatment, improving overall patient outcome. This manuscript is the first in the last 10 years that revises the advances in the application of liposome-based formulations in bioimaging, chemotherapy, phototherapy, immunotherapy, combination therapies, and emergent therapies for pancreatic cancer management. Prospective insights are provided regarding several advantages resulting from the use of liposome technology in precision strategies, fostering new ideas for next-generation diagnosis and targeted therapies of pancreatic cancer.
Collapse
Affiliation(s)
- Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Lauren Evans
- Pure and Applied Chemistry, University of Strathclyde, 99 George Street, Glasgow, G1 1RD, UK
| | - Mahzad Motallebi
- Immunology Board for Transplantation And Cell-based Therapeutics (Immuno_TACT), Universal Scientific Education and Research Network (USERN), Tehran 7616911319, Iran; Nanomedicine Research Association (NRA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hajra Zafar
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Miguel Pereira-Silva
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| | - Kalsoom Saleem
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad 45320, Pakistan
| | - Diana Peixoto
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol 98613-35856, Iran
| | - Esmaeel Sharifi
- Cancer Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Francisco Veiga
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| | - Clare Hoskins
- Pure and Applied Chemistry, University of Strathclyde, 99 George Street, Glasgow, G1 1RD, UK
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal.
| |
Collapse
|
3
|
David KI, Ravikumar TS, Sethuraman S, Krishnan UM. Investigations of an organic-inorganic nanotheranostic hybrid for pancreatic cancer therapy using cancer-in-a-dish and in vivomodels. Biomed Mater 2022; 18. [PMID: 36270604 DOI: 10.1088/1748-605x/ac9cb2] [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: 11/19/2021] [Accepted: 10/21/2022] [Indexed: 12/14/2022]
Abstract
The incidence of highly aggressive pancreatic cancer is increasing across the globe and is projected to increase to 18.6% by 2050. The mortality rate for this form of cancer is very high and the 5 y relative survival rate is only about 9%-10%. The 3D pancreatic cancer microenvironment exerts a major influence on the poor survival rate. A key factor is the prevention of the penetration of the chemotherapeutic drugs in the three-dimensional (3D) microenvironment leading to the development of chemoresistance which is a major contributor to the survival rates. Hence,in vitrostudies using 3D cultures represent a better approach to understand the effect of therapeutic formulations on the cancer cells when compared to conventional 2D cultures. In the present study, we have explored three different conditions for the development of a 3D pancreatic tumour spheroid model from MiaPaCa-2 and PanC1 cells cultured for 10 days using Matrigel matrix. This optimized spheroid model was employed to evaluate a multi-functional nanotheranostic system fabricated using chitosan nanoparticles co-encapsulated with the chemotherapeutic agent gemcitabine and gold-capped iron oxide nanoparticles for multimodal imaging. The effect of the single and multiple-dose regimens of the theranostic system on the viability of 3D spheroids formed from the two pancreatic cancer cell lines was studied. It was observed that the 3D tumour spheroids cultured for 10 days exhibited resistance towards free gemcitabine drug, unlike the 2D culture. The administration of the multifunctional nanotheranostic system on alternate days effectively reduced the cancer cell viability after five doses to about 20% when compared with other groups. The repeated doses of the nanotheranostic system were found to be more effective than the single dose. Cell line-based differences in internalization of the carrier was also reflected in their response to the nanocarrier with PanC1 showing better sensitivity to the treatment.In vivostudies revealed that the combination of gemcitabine and magnetic field induced hypothermia produced superior regression in cancer when compared with the chemotherapeutic agent alone by a combination of activating the pro-apoptotic pathway and heat-induced necrosis. Our results reveal that this multi-functional system holds promise to overcome the current challenges to treat pancreatic cancers.
Collapse
Affiliation(s)
- Karolyn Infanta David
- Centre for Nanotechnology and Advanced Biomaterials, SASTRA Deemed University, Thanjavur, TamilNadu 613401, India.,School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, TamilNadu 613401, India
| | - T S Ravikumar
- Formerly at Sri Venkateswara Institute of Medical Sciences (SVIMS) Tirupati 517507, India
| | - Swaminathan Sethuraman
- Centre for Nanotechnology and Advanced Biomaterials, SASTRA Deemed University, Thanjavur, TamilNadu 613401, India.,School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, TamilNadu 613401, India
| | - Uma Maheswari Krishnan
- Centre for Nanotechnology and Advanced Biomaterials, SASTRA Deemed University, Thanjavur, TamilNadu 613401, India.,School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, TamilNadu 613401, India.,School of Arts, Sciences, Humanities and Education, SASTRA Deemed University, Thanjavur, TamilNadu 613401, India
| |
Collapse
|
4
|
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
|
5
|
Hoogevest P, Tiemessen H, Metselaar JM, Drescher S, Fahr A. The Use of Phospholipids to Make Pharmaceutical Form Line Extensions. EUR J LIPID SCI TECH 2021. [DOI: 10.1002/ejlt.202000297] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Peter Hoogevest
- Phospholipid Research Center Im Neuenheimer Feld 515 Heidelberg 69120D‐69120 Germany
| | - Harry Tiemessen
- Technical & Research Development PHAD PDU Specialty Novartis Campus Physical Garden (WSJ 177) 2.14 Basel CH‐4002 Switzerland
| | - Josbert M. Metselaar
- Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic Aachen D‐52074 Germany
- Institute for Biomedical Engineering, Faculty of Medicine RWTH Aachen University Aachen D‐52074 Germany
| | - Simon Drescher
- Phospholipid Research Center Im Neuenheimer Feld 515 Heidelberg D‐69120 Germany
| | - Alfred Fahr
- Professor Emeritus, Pharmaceutical Technology Friedrich‐Schiller‐University Jena Jena Germany
| |
Collapse
|
6
|
Fattahi N, Shahbazi MA, Maleki A, Hamidi M, Ramazani A, Santos HA. Emerging insights on drug delivery by fatty acid mediated synthesis of lipophilic prodrugs as novel nanomedicines. J Control Release 2020; 326:556-598. [PMID: 32726650 DOI: 10.1016/j.jconrel.2020.07.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 12/25/2022]
Abstract
Many drug molecules that are currently in the market suffer from short half-life, poor absorption, low specificity, rapid degradation, and resistance development. The design and development of lipophilic prodrugs can provide numerous benefits to overcome these challenges. Fatty acids (FAs), which are lipophilic biomolecules constituted of essential components of the living cells, carry out many necessary functions required for the development of efficient prodrugs. Chemical conjugation of FAs to drug molecules may change their pharmacodynamics/pharmacokinetics in vivo and even their toxicity profile. Well-designed FA-based prodrugs can also present other benefits, such as improved oral bioavailability, promoted tumor targeting efficiency, controlled drug release, and enhanced cellular penetration, leading to improved therapeutic efficacy. In this review, we discuss diverse drug molecules conjugated to various unsaturated FAs. Furthermore, various drug-FA conjugates loaded into various nanostructure delivery systems, including liposomes, solid lipid nanoparticles, emulsions, nano-assemblies, micelles, and polymeric nanoparticles, are reviewed. The present review aims to inspire readers to explore new avenues in prodrug design based on the various FAs with or without nanostructured delivery systems.
Collapse
Affiliation(s)
- Nadia Fattahi
- Department of Chemistry, Faculty of Science, University of Zanjan, P.O. Box 45195-313, Zanjan, Iran; Trita Nanomedicine Research Center (TNRC), Trita Third Millennium Pharmaceuticals, 45331-55681 Zanjan, Iran
| | - Mohammad-Ali Shahbazi
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran; Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Aziz Maleki
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran; Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mehrdad Hamidi
- Trita Nanomedicine Research Center (TNRC), Trita Third Millennium Pharmaceuticals, 45331-55681 Zanjan, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran; Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Ali Ramazani
- Department of Chemistry, Faculty of Science, University of Zanjan, P.O. Box 45195-313, Zanjan, Iran; Research Institute of Modern Biological Techniques (RIMBT), University of Zanjan, P.O. Box 45195-313, Zanjan, Iran
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland; Helsinki Institute of Life Science (HiLIFE), Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland.
| |
Collapse
|
7
|
Mu Y, Wang D, Bie L, Luo S, Mu X, Zhao Y. Glypican-1-targeted and gemcitabine-loaded liposomes enhance tumor-suppressing effect on pancreatic cancer. Aging (Albany NY) 2020; 12:19585-19596. [PMID: 33035197 PMCID: PMC7732280 DOI: 10.18632/aging.103918] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 07/25/2020] [Indexed: 01/24/2023]
Abstract
Liposomes (LPs) as promising drug delivery systems are widely applied in cancer therapy. This study aimed to investigate the effect of glypican-1 (GPC1)-targeted and gemcitabine (GEM)-loaded LP [GPC1-LP (GEM)] on cell proliferation and apoptosis in PANC-1s, as well as on orthotopic pancreatic cancer (PDAC) mice. The GPC1-LP (GEM) and LP (GEM) was prepared, and then the size distribution of GPC1-LP (GEM) was analyzed by dynamic light scattering (DLS). In vitro drug release assay of GPC1-LP (GEM) and LP (GEM) was performed, and the expression of GPC1 in PANC1 cells was detected as well. Next, the effects of free GEM, LP (GEM) and GPC1-LP (GEM) on cell viability, clone number, and apoptosis, as well as the expression of proteins associated with apoptosis were measured in 239T and PANC-1 cells. Furthermore, the body weight and tumor size of orthotopic PDAC mice were evaluated following the treatment of free GEM, LP (GEM) or GPC1-LP (GEM). LP (GEM) and GPC1-LP (GEM) were successfully prepared with a successful GEM release within 24 h. In addition, GPC1 was positively expressed in PANC-1 cells but not 293T cells. These findings provided more insights into the anti-tumor potential for the biomedical application of GPC1-LP (GEM) in PDAC.
Collapse
Affiliation(s)
- Yu Mu
- Department of Oncology, Affiliated Cancer Hospital of Zhengzhou University / Henan Cancer Hospital, Zhengzhou, China
| | - Dezhi Wang
- East China Normal University, Shanghai, China
| | - Liangyu Bie
- Department of Oncology, Affiliated Cancer Hospital of Zhengzhou University / Henan Cancer Hospital, Zhengzhou, China
| | - Suxia Luo
- Department of Oncology, Affiliated Cancer Hospital of Zhengzhou University / Henan Cancer Hospital, Zhengzhou, China
| | - Xiaoqian Mu
- Department of Oncology, Affiliated Cancer Hospital of Zhengzhou University / Henan Cancer Hospital, Zhengzhou, China
| | - Yanqiu Zhao
- Department of Oncology, Affiliated Cancer Hospital of Zhengzhou University / Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
8
|
Production of drug-releasing biodegradable microporous scaffold impregnated with gemcitabine using a CO2 foaming process. J CO2 UTIL 2020. [DOI: 10.1016/j.jcou.2020.101227] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
9
|
Yu J, Zhou S, Li J, Wang Y, Su Y, Chi D, Wang J, Wang X, He Z, Lin G, Liu D, Wang Y. Simple weak-acid derivatives of paclitaxel for remote loading into liposomes and improved therapeutic effects. RSC Adv 2020; 10:27676-27687. [PMID: 35516912 PMCID: PMC9055615 DOI: 10.1039/d0ra03190a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 05/27/2020] [Indexed: 12/05/2022] Open
Abstract
Liposomes are among the most successful nanocarriers; several products have been marketed, all of which were prepared by active loading methods. However, poorly water-soluble drugs without ionizable groups are usually incorporated into the lipid bi-layer of liposomes by passive loading methods, with serious drug leakage during blood circulation. Furthermore, there have been few improvements in their anti-cancer activity and safety. Herein, we designed and synthesized three weak-acid modified paclitaxel (PTX) derivatives with a one-step reaction for the remote loading of liposomal formulations. By comparison, PTX-succinic acid liposomes (PTX-SA LPs) exhibited the highest encapsulation efficiency (97.2 ± 1.8%) and drug loading (8.84 ± 0.16%); meanwhile, there was almost no change in their particle size or zeta potential within one month. Furthermore, compared with Taxol®, the PTX-SA LPs showed a 4.35-fold prolonged half-time, enhanced tumor accumulation, and an increased maximum tolerated dose (MTD) of more than 30 mg kg−1. As a result, the PTX-SA LPs displayed significantly improved in vivo anti-cancer efficacy in comparison with Taxol®. Therefore, weak-acid modification is proved to be a simple and effective method to achieve remote loading and high encapsulation efficiency of poorly soluble drugs, showing great potential for clinical application. A remote loading liposomal formulation of weak-acid paclitaxel derivative with high encapsulation efficiency and high drug loading, improved therapeutic efficiency and negligible toxicity.![]()
Collapse
Affiliation(s)
- Jiang Yu
- Wuya College of Innovation, Shenyang Pharmaceutical University Shenyang Liaoning 110016 P. R. China +86-24-23986325 +86-24-23986325
| | - Shuang Zhou
- Wuya College of Innovation, Shenyang Pharmaceutical University Shenyang Liaoning 110016 P. R. China +86-24-23986325 +86-24-23986325
| | - Jinbo Li
- Wuya College of Innovation, Shenyang Pharmaceutical University Shenyang Liaoning 110016 P. R. China +86-24-23986325 +86-24-23986325
| | - Yingli Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University Shenyang Liaoning 110016 P. R. China +86-24-23986325 +86-24-23986325
| | - Yujiao Su
- Wuya College of Innovation, Shenyang Pharmaceutical University Shenyang Liaoning 110016 P. R. China +86-24-23986325 +86-24-23986325
| | - Dongxu Chi
- Department of Pharmaceutics, Shenyang Pharmaceutical University Shenyang Liaoning 110016 P. R. China
| | - Jiamei Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University Shenyang Liaoning 110016 P. R. China +86-24-23986325 +86-24-23986325
| | - Xue Wang
- Department of Pharmaceutics, Shenyang Pharmaceutical University Shenyang Liaoning 110016 P. R. China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University Shenyang Liaoning 110016 P. R. China +86-24-23986325 +86-24-23986325
| | - Guimei Lin
- School of Pharmaceutical Science, Shandong University 44 Wenhuaxi Road Jinan 250012 China
| | - Dan Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University Shenyang 110016 Liaoning P. R. China +86-24-4352-0218 +86-24-4352-0218
| | - Yongjun Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University Shenyang Liaoning 110016 P. R. China +86-24-23986325 +86-24-23986325
| |
Collapse
|
10
|
Biodistribution and Pharmacokinetic Study of Gemcitabine Hydrochloride Loaded Biocompatible Iron-Based Metal Organic Framework. J Inorg Organomet Polym Mater 2019. [DOI: 10.1007/s10904-019-01417-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
11
|
Samanta K, Setua S, Kumari S, Jaggi M, Yallapu MM, Chauhan SC. Gemcitabine Combination Nano Therapies for Pancreatic Cancer. Pharmaceutics 2019; 11:E574. [PMID: 31689930 PMCID: PMC6920852 DOI: 10.3390/pharmaceutics11110574] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 10/28/2019] [Accepted: 10/28/2019] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer is one of the deadliest causes of cancer-related death in the United States, with a 5-year overall survival rate of 6 to 8%. These statistics suggest that immediate medical attention is needed. Gemcitabine (GEM) is the gold standard first-line single chemotherapy agent for pancreatic cancer but, after a few months, cells develop chemoresistance. Multiple clinical and experimental investigations have demonstrated that a combination or co-administration of other drugs as chemotherapies with GEM lead to superior therapeutic benefits. However, such combination therapies often induce severe systemic toxicities. Thus, developing strategies to deliver a combination of chemotherapeutic agents more securely to patients is needed. Nanoparticle-mediated delivery can offer to load a cocktail of drugs, increase stability and availability, on-demand and tumor-specific delivery while minimizing chemotherapy-associated adverse effects. This review discusses the available drugs being co-administered with GEM and the limitations associated during the process of co-administration. This review also helps in providing knowledge of the significant number of delivery platforms being used to overcome problems related to gemcitabine-based co-delivery of other chemotherapeutic drugs, thereby focusing on how nanocarriers have been fabricated, considering the modes of action, targeting receptors, pharmacology of chemo drugs incorporated with GEM, and the differences in the physiological environment where the targeting is to be done. This review also documents the focus on novel mucin-targeted nanotechnology which is under development for pancreatic cancer therapy.
Collapse
Affiliation(s)
- Kamalika Samanta
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Saini Setua
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Sonam Kumari
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Meena Jaggi
- Department of Immunology and Microbiology, Institute for Cancer Immunotherapy, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78503, USA.
| | - Murali M Yallapu
- Department of Immunology and Microbiology, Institute for Cancer Immunotherapy, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78503, USA.
| | - Subhash C Chauhan
- Department of Immunology and Microbiology, Institute for Cancer Immunotherapy, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78503, USA.
| |
Collapse
|
12
|
Wahgiman NA, Salim N, Abdul Rahman MB, Ashari SE. Optimization of nanoemulsion containing gemcitabine and evaluation of its cytotoxicity towards human fetal lung fibroblast (MRC5) and human lung carcinoma (A549) cells. Int J Nanomedicine 2019; 14:7323-7338. [PMID: 31686809 PMCID: PMC6751780 DOI: 10.2147/ijn.s212635] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 07/17/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Gemcitabine (GEM) is a chemotherapeutic agent, which is known to battle cancer but challenging due to its hydrophilic nature. Nanoemulsion is water-in-oil (W/O) nanoemulsion shows potential as a carrier system in delivering gemcitabine to the cancer cell. METHODS The behaviour of GEM in MCT/surfactants/NaCl systems was studied in the ternary system at different ratios of Tween 80 and Span 80. The system with surfactant ratio 3:7 of Tween 80 and Span 80 was chosen for further study on the preparation of nanoemulsion formulation due to the highest isotropic region. Based on the selected ternary phase diagram, a composition of F1 was chosen and used for optimization by using the D-optimal mixture design. The interaction variables between medium chain triglyceride (MCT), surfactant mixture Tween 80: Span 80 (ratio 3:7), 0.9 % sodium chloride solution and gemcitabine were evaluated towards particle size as a response. RESULTS The results showed that NaCl solution and GEM gave more effects on particle size, polydispersity index and zeta potential of 141.57±0.05 nm, 0.168 and -37.10 mV, respectively. The optimized nanoemulsion showed good stability (no phase separation) against centrifugation test and storage at three different temperatures. The in vitro release of gemcitabine at different pH buffer solution was evaluated. The results showed the release of GEM in buffer pH 6.5 (45.19%) was higher than GEM in buffer pH 7.4 (13.62%). The cytotoxicity study showed that the optimized nanoemulsion containing GEM induced cytotoxicity towards A549 cell and at the same time reduced cytotoxicity towards MRC5 when compared to the control (GEM solution).
Collapse
Affiliation(s)
- Nadiatul Atiqah Wahgiman
- Integrated Chemical BioPhysics Research, Faculty of Science, University Putra Malaysia (UPM), Serdang, Selangor43400, Malaysia
| | - Norazlinaliza Salim
- Integrated Chemical BioPhysics Research, Faculty of Science, University Putra Malaysia (UPM), Serdang, Selangor43400, Malaysia
- Centre of Foundation Studies for Agricultural Science, University Putra Malaysia (UPM), Serdang, Selangor43400, Malaysia
| | - Mohd Basyaruddin Abdul Rahman
- Integrated Chemical BioPhysics Research, Faculty of Science, University Putra Malaysia (UPM), Serdang, Selangor43400, Malaysia
| | - Siti Efliza Ashari
- Integrated Chemical BioPhysics Research, Faculty of Science, University Putra Malaysia (UPM), Serdang, Selangor43400, Malaysia
- Centre of Foundation Studies for Agricultural Science, University Putra Malaysia (UPM), Serdang, Selangor43400, Malaysia
| |
Collapse
|
13
|
Hassan T, Jinho P, Hytham H. G, Masters AR, Abdel-Aleem JA, Abdelrahman SI, Abdelrahman AA, Lyle LT, Yeo Y. Development of Liposomal Gemcitabine with High Drug Loading Capacity. Mol Pharm 2019; 16:2858-2871. [PMID: 31136710 PMCID: PMC6662591 DOI: 10.1021/acs.molpharmaceut.8b01284] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Liposomes are widely used for systemic delivery of chemotherapeutic agents to reduce their nonspecific side effects. Gemcitabine (Gem) makes a great candidate for liposomal encapsulation due to the short half-life and nonspecific side effects; however, it has been difficult to achieve liposomal Gem with high drug loading capacity. Remote loading, which uses a transmembrane pH gradient to induce an influx of drug and locks the drug in the core as a sulfate complex, does not serve Gem as efficiently as doxorubicin (Dox) due to the low p Ka value of Gem. Existing studies have attempted to improve Gem loading capacity in liposomes by employing lipophilic Gem derivatives or creating a high-concentration gradient for active loading into the hydrophilic cores (small volume loading). In this study, we combine the remote loading approach and small volume loading or hypertonic loading, a new approach to induce the influx of Gem into the preformed liposomes by high osmotic pressure, to achieve a Gem loading capacity of 9.4-10.3 wt % in contrast to 0.14-3.8 wt % of the conventional methods. Liposomal Gem showed a good stability during storage, sustained-release over 120 h in vitro, enhanced cellular uptake, and improved cytotoxicity as compared to free Gem. Liposomal Gem showed a synergistic effect with liposomal Dox on Huh7 hepatocellular carcinoma cells. A mixture of liposomal Gem and liposomal Dox delivered both drugs to the tumor more efficiently than a free drug mixture and showed a relatively good anti-tumor effect in a xenograft model of hepatocellular carcinoma. This study shows that bioactive liposomal Gem with high drug loading capacity can be produced by remote loading combined with additional approaches to increase drug influx into the liposomes.
Collapse
Affiliation(s)
- Tamam Hassan
- Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Park Jinho
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Gadalla Hytham H.
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Andrea R. Masters
- Clinical Pharmacology Analytical Core, Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jelan A. Abdel-Aleem
- Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Sayed I. Abdelrahman
- Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Aly A. Abdelrahman
- Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - L. Tiffany Lyle
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| | - Yoon Yeo
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
14
|
Ray P, Alhalhooly L, Ghosh A, Choi Y, Banerjee S, Mallik S, Banerjee S, Quadir M. Size-Transformable, Multifunctional Nanoparticles from Hyperbranched Polymers for Environment-Specific Therapeutic Delivery. ACS Biomater Sci Eng 2019; 5:1354-1365. [DOI: 10.1021/acsbiomaterials.8b01608] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
| | | | - Arnab Ghosh
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri 64128, United States
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | | | - Sushanta Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri 64128, United States
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | | | - Snigdha Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri 64128, United States
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | | |
Collapse
|
15
|
Rodallec A, Sicard G, Fanciullino R, Benzekry S, Lacarelle B, Milano G, Ciccolini J. Turning cold tumors into hot tumors: harnessing the potential of tumor immunity using nanoparticles. Expert Opin Drug Metab Toxicol 2018; 14:1139-1147. [PMID: 30354685 DOI: 10.1080/17425255.2018.1540588] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Immune checkpoint inhibitors have considerably changed the landscape of oncology. However apart from world-acclaimed success stories limited to melanoma and lung cancer, many solid tumors failed to respond to immune checkpoint inhibitors due to limited immunogenicity, unfavorable tumor micro-environments (TME), lack of infiltrating T lymphocytes or increases in Tregs. Areas covered: Combinatorial strategies are foreseen as the future of immunotherapy and using cytotoxics or modulating agents is expected to boost the efficacy of immune checkpoint inhibitors. In this respect, nanoparticles displaying unique pharmacokinetic features such as tumor targeting properties, optimal payload delivery and long-lasting interferences with TME, are promising candidates for such combinations. This review covers the basis, expectancies, limits and pitfalls of future combination between nanoparticles and immune check point inhibitors. Expert opinion: Nanoparticles allow optimal delivery of variety of payloads in tumors while sparing healthy tissue, thus triggering immunogenic cell death. Depleting tumor stroma could further help immune cells and monoclonal antibodies to better circulate in the TME, plus immune-modulating properties of the charged cytotoxics. Finally, nanoparticles themselves present immunogenicity and antigenicity likely to boost immune response at the tumor level.
Collapse
Affiliation(s)
- Anne Rodallec
- a SMARTc Unit, Centre de Recherche en Cancérologie de Marseille UMR Inserm U1068 , Aix Marseille University , Marseille , France
| | - Guillaume Sicard
- a SMARTc Unit, Centre de Recherche en Cancérologie de Marseille UMR Inserm U1068 , Aix Marseille University , Marseille , France
| | - Raphaelle Fanciullino
- a SMARTc Unit, Centre de Recherche en Cancérologie de Marseille UMR Inserm U1068 , Aix Marseille University , Marseille , France
| | | | - Bruno Lacarelle
- a SMARTc Unit, Centre de Recherche en Cancérologie de Marseille UMR Inserm U1068 , Aix Marseille University , Marseille , France
| | - Gerard Milano
- c EA666 Oncopharmacology Unit , Centre Antoine Lacassagne , Nice , France
| | - Joseph Ciccolini
- a SMARTc Unit, Centre de Recherche en Cancérologie de Marseille UMR Inserm U1068 , Aix Marseille University , Marseille , France
| |
Collapse
|
16
|
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
|
17
|
Yang W, Hu Q, Xu Y, Liu H, Zhong L. Antibody fragment-conjugated gemcitabine and paclitaxel-based liposome for effective therapeutic efficacy in pancreatic cancer. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 89:328-335. [PMID: 29752104 DOI: 10.1016/j.msec.2018.04.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 12/27/2017] [Accepted: 04/09/2018] [Indexed: 12/25/2022]
Abstract
In this study, we have developed an antibody fragment (AF)-conjugated gemcitabine (GEM) and paclitaxel (PTX)-loaded liposome (AF-GPL) to enhance the therapeutic efficacy in pancreatic cancer treatment. The maleimide-thiol chemistry was utilized to conjugate AF on the liposome surface. The dual-drug loaded liposome was nanosized and exhibited a controlled release of both the drugs. Importantly, two drugs have different release pattern over a period of time. The AF-conjugated liposome showed enhanced cellular uptake in pancreatic cancer cells compared to that of non-targeted liposome. Two-fold higher internalization of particles might increase the intracellular concentration of anticancer drugs that might further increase the therapeutic efficacy in pancreatic cancer cells. AF-GPL showed significantly higher cytotoxic effect in pancreatic cancer cell compared to that of non-targeted GPL. The IC50 value of GEM, PTX, GPL and AF-GPL were 5.9 μg/ml, 4.2 μg/ml, 1.92 μg/ml, and 0.45 μg/ml, respectively. Consistently, AF-GPL (4.12) showed significantly higher ratio of Bax/Bcl-2 compared to that of non-targeted GPL (2.8). Importantly, AF-GPL induced a significant apoptosis of cancer cells with predominant amount of cells in late apoptosis cells. Overall, AF-conjugated nanosystem could potentially improve the therapeutic efficacy in pancreatic cancers.
Collapse
Affiliation(s)
- Wentao Yang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang University, No.1 Minde Road, Nanchang, China
| | - Qian Hu
- Department of Emergency, The Second Affiliated Hospital of Nanchang University, No.1 Minde Road, Nanchang, China
| | - Yanmei Xu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, No.1 Minde Road, Nanchang, China
| | - Hailang Liu
- Department of Organ Transplant, The Second Affiliated Hospital of Nanchang University, No.1 Minde Road, Nanchang, China
| | - Lin Zhong
- Department of Organ Transplant, The Second Affiliated Hospital of Nanchang University, No.1 Minde Road, Nanchang, China.
| |
Collapse
|
18
|
Ho L, Bokharaei M, Li SD. Current update of a thermosensitive liposomes composed of DPPC and Brij78. J Drug Target 2018; 26:407-419. [DOI: 10.1080/1061186x.2017.1419361] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Laurence Ho
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Mehrdad Bokharaei
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Shyh-Dar Li
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
19
|
Birhanu G, Javar HA, Seyedjafari E, Zandi-Karimi A. Nanotechnology for delivery of gemcitabine to treat pancreatic cancer. Biomed Pharmacother 2017; 88:635-643. [PMID: 28142120 DOI: 10.1016/j.biopha.2017.01.071] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 01/02/2017] [Accepted: 01/12/2017] [Indexed: 12/21/2022] Open
Abstract
Pancreatic cancer (PC) is one of the most deadly and quickly fatal human cancers with a 5-year mortality rate close to 100%. Its prognosis is very poor, mainly because of its hostile biological behavior and late onset of symptoms for clinical diagnosis; these bring limitations on therapeutic interventions. Factors contributing for the difficulties in treating PC include: high rate of drug resistance, fast metastasis to different organs, poor prognosis and relapse of the tumor after therapy. After being approved by US FDA 1997, Gemcitabine (Gem) is the first line and the gold standard drug for all stages of advanced PC till now. However, its efficacy is unsatisfactory, mainly due to; its chemical instability and poor cellular uptake, resulting in an extremely short half-life and low bioavailability. To solve this drawbacks and increase the therapeutic outcome important progress has been achieved in the field of nanotechnology and offers a promising and effective alternative. This review mainly focus on the most commonly investigated nanoparticle (NP) delivery systems of Gem for PC treatment and the latest progresses achieved. Novel nanocarriers with better tumor targeting efficiencies and maximum treatment outcome to treat this deadly due are given much attention.
Collapse
Affiliation(s)
- Gebremariam Birhanu
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, International Campus (TUMS-IC), Tehran, Iran; School of Pharmacy, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Hamid Akbari Javar
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Seyedjafari
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran.
| | - Ali Zandi-Karimi
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| |
Collapse
|
20
|
Kattel K, Mondal G, Lin F, Kumar V, Mahato RI. Biodistribution of Self-Assembling Polymer-Gemcitabine Conjugate after Systemic Administration into Orthotopic Pancreatic Tumor Bearing Mice. Mol Pharm 2016; 14:1365-1372. [PMID: 27798825 DOI: 10.1021/acs.molpharmaceut.6b00929] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Therapeutic efficacy of gemcitabine (GEM) is severely limited due to its rapid metabolism by enzymatic deamination in vivo. We recently determined its therapeutic efficacy before (F-GEM) and after conjugation to poly(ethylene glycol)-block-poly(2-methyl-2-carboxyl-propylene carbonate) (mPEG-b-PCC-g-GEM-g-DC, abbreviated as P-GEM) in subcutaneous and orthotopic pancreatic tumor bearing mice. In this study, pharmacokinetic (PK) parameters and biodistribution profiles of F-GEM and P-GEM were determined after intravenous injection into orthotopic pancreatic tumor bearing NSG mice. To assess the short-term toxicity, the levels of hematological, hepatic, and renal injury markers were measured after 24 h postadministration into these mice. P-GEM was distributed to all the major organs, with higher accumulation in the liver, spleen, and tumor compared to F-GEM. Area under the curve (AUC), elimination half-life (t1/2), and mean residence time (MRT) of P-GEM treated group were significantly higher compared to those of F-GEM treated group: 246,425 ± 1605 vs 83,591 ± 1844 ng/mL × h as AUC, 5.77 ± 2.02 vs 1.99 ± 0.09 h as t1/2, and 4.45 ± 0.15 vs 1.12 ± 0.13 h as MRT. Further, P-GEM exhibited negligible systemic toxicity as evidenced by almost similar alanine aminotransferase (ALT) and aspartate aminotransferase (AST) values for both P-GEM and F-GEM. These results suggest that P-GEM protects GEM from degradation and provides sustained drug release, resulting in enhanced GEM delivery to the tumor by more than 2.5-fold compared to F-GEM. Hence, P-GEM is a promising gemcitabine conjugated polymeric micelle for treating pancreatic cancer.
Collapse
Affiliation(s)
- Krishna Kattel
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| | - Goutam Mondal
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| | - Feng Lin
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| | - Virender Kumar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| |
Collapse
|
21
|
Khare V, Singh A, Mahajan G, Alam N, Kour S, Gupta M, Kumar A, Singh G, Singh SK, Saxena AK, Mondhe DM, Gupta PN. Long-circulatory nanoparticles for gemcitabine delivery: Development and investigation of pharmacokinetics and in-vivo anticancer efficacy. Eur J Pharm Sci 2016; 92:183-93. [DOI: 10.1016/j.ejps.2016.07.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 06/14/2016] [Accepted: 07/08/2016] [Indexed: 02/06/2023]
|
22
|
Karaca M, Dutta R, Ozsoy Y, Mahato RI. Micelle Mixtures for Coadministration of Gemcitabine and GDC-0449 To Treat Pancreatic Cancer. Mol Pharm 2016; 13:1822-32. [PMID: 26981724 DOI: 10.1021/acs.molpharmaceut.5b00971] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Hedgehog (Hh) signaling plays an important role in the development and metastasis of pancreatic ductal adenocarcinoma (PDAC). Although gemcitabine (GEM) has been used as a first-line therapy for PDAC, its rapid metabolism and short plasma half-life restrict its use as a single chemotherapy. Combination therapy with more than one drug is a promising approach for treating cancer. Herein, we report the use of methoxy poly(ethylene glycol)-block-poly(2-methyl-2-carboxyl-propylene carbonate)-graft-dodecanol (mPEG-b-PCC-g-DC) copolymer for conjugating GEM and encapsulating a Hh inhibitor, vismodegib (GDC-0449), into its hydrophobic core for treating PDAC. Our objective was to determine whether the micelle mixtures of these two drugs could show better response in inhibiting Hh signaling pathway and restraining the proliferation and metastasis of pancreatic cancer. The in vivo stability of GEM significantly increased after conjugation, which resulted in its increased antitumor efficacy. Almost 80% of encapsulated GDC-0449 and 19% conjugated GEM were released in vitro at pH 5.5 in 48 h in a sustained manner. The invasion, migration, and colony forming features of MIA PaCa-2 cells were significantly inhibited by micelle mixture carrying GEM and GDC-0449. Remarkable increase in PARP cleavage and Bax proved increased apoptosis by this combination formulation compared to individual micelles. This combination therapy efficiently inhibited tumor growth, increased apoptosis, reduced Hh ligands PTCH-1 and Gli-1, and lowered EMT-activator ZEB-1 when injected to athymic nude mice bearing subcutaneous tumor generated using MIA PaCa-2 cells compared to monotherapy as observed from immunohistochemical analysis. In conclusion, micelle mixtures carrying GEM and GDC-0449 have the potential to treat pancreatic cancer.
Collapse
Affiliation(s)
- Melek Karaca
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States.,Department of Pharmaceutical Technology, Faculty of Pharmacy, Istanbul University , Istanbul, Turkey
| | - Rinku Dutta
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| | - Yildiz Ozsoy
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Istanbul University , Istanbul, Turkey
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| |
Collapse
|
23
|
Khare V, Sakarchi WA, Gupta PN, Curtis ADM, Hoskins C. Synthesis and characterization of TPGS–gemcitabine prodrug micelles for pancreatic cancer therapy. RSC Adv 2016. [DOI: 10.1039/c6ra09347g] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Evaluation of a novel polymer-drug conjugate formulation in pancreatic cancer.
Collapse
Affiliation(s)
- Vaibhav Khare
- Institute of Science and Technology in Medicine
- Keele University
- Keele
- UK
- Formulation and Drug Delivery Division
| | | | - Prem N. Gupta
- Formulation and Drug Delivery Division
- CSIR-Indian Institute of Integrative Medicine
- India 180001
| | | | - Clare Hoskins
- Institute of Science and Technology in Medicine
- Keele University
- Keele
- UK
| |
Collapse
|
24
|
May JP, Undzys E, Roy A, Li SD. Synthesis of a Gemcitabine Prodrug for Remote Loading into Liposomes and Improved Therapeutic Effect. Bioconjug Chem 2015; 27:226-37. [DOI: 10.1021/acs.bioconjchem.5b00619] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Jonathan P. May
- Faculty
of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
- Drug
Discovery Program, Ontario Institute for Cancer Research, Toronto, Ontario M5G 0A3, Canada
| | - Elijus Undzys
- Drug
Discovery Program, Ontario Institute for Cancer Research, Toronto, Ontario M5G 0A3, Canada
| | - Aniruddha Roy
- Faculty
of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
- Drug
Discovery Program, Ontario Institute for Cancer Research, Toronto, Ontario M5G 0A3, Canada
| | - Shyh-Dar Li
- Faculty
of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
- Drug
Discovery Program, Ontario Institute for Cancer Research, Toronto, Ontario M5G 0A3, Canada
| |
Collapse
|
25
|
Huang HC, Mallidi S, Liu J, Chiang CT, Mai Z, Goldschmidt R, Ebrahim-Zadeh N, Rizvi I, Hasan T. Photodynamic Therapy Synergizes with Irinotecan to Overcome Compensatory Mechanisms and Improve Treatment Outcomes in Pancreatic Cancer. Cancer Res 2015; 76:1066-77. [PMID: 26719532 DOI: 10.1158/0008-5472.can-15-0391] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Accepted: 12/10/2015] [Indexed: 12/18/2022]
Abstract
The ability of tumor cells to adapt to therapeutic regimens by activating alternative survival and growth pathways remains a major challenge in cancer therapy. Therefore, the most effective treatments will involve interactive strategies that target multiple nonoverlapping pathways while eliciting synergistic outcomes and minimizing systemic toxicities. Nanoliposomal irinotecan is approved by the FDA for gemcitabine-refractory metastatic pancreatic cancer. However, the full potential of irinotecan treatment is hindered by several cancer cell survival mechanisms, including ATP-binding cassette G2 (ABCG2) transporter-mediated irinotecan efflux from cells. Here, we demonstrate that benzoporphyrin derivative-based photodynamic therapy (PDT), a photochemical cytotoxic modality that activates the apoptotic pathway, reduced ABCG2 expression to increase intracellular irinotecan levels in pancreatic cancer. Moreover, we show that PDT inhibited survivin expression. Although PDT potentiated irinotecan treatment, we also demonstrate that irinotecan reduced the tumoral expression of monocarboxylate transporter 4, which was upregulated by PDT. Notably, using orthotopic xenograft models, we demonstrate that combination of single low-dose PDT and a subclinical dose of nanoliposomal irinotecan synergistically inhibited tumor growth by 70% for 3 weeks compared with 25% reduction after either monotherapies. Our findings offer new opportunities for the clinical translation of PDT and irinotecan combination therapy for effective pancreatic cancer treatment.
Collapse
Affiliation(s)
- Huang-Chiao Huang
- Wellman Center for Photomedicine, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts. Department of Dermatology, MGH, Boston, Massachusetts
| | - Srivalleesha Mallidi
- Wellman Center for Photomedicine, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts. Department of Dermatology, MGH, Boston, Massachusetts
| | - Joyce Liu
- Wellman Center for Photomedicine, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts. Department of Dermatology, MGH, Boston, Massachusetts
| | - Chun-Te Chiang
- Wellman Center for Photomedicine, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts. Department of Dermatology, MGH, Boston, Massachusetts
| | - Zhiming Mai
- Wellman Center for Photomedicine, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts. Department of Dermatology, MGH, Boston, Massachusetts
| | - Ruth Goldschmidt
- Wellman Center for Photomedicine, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts. Department of Dermatology, MGH, Boston, Massachusetts
| | - Neema Ebrahim-Zadeh
- Wellman Center for Photomedicine, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts. Department of Dermatology, MGH, Boston, Massachusetts
| | - Imran Rizvi
- Wellman Center for Photomedicine, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts. Department of Dermatology, MGH, Boston, Massachusetts
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts. Department of Dermatology, MGH, Boston, Massachusetts. Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, Massachusetts.
| |
Collapse
|
26
|
Mondal G, Kumar V, Shukla SK, Singh PK, Mahato RI. EGFR-Targeted Polymeric Mixed Micelles Carrying Gemcitabine for Treating Pancreatic Cancer. Biomacromolecules 2015; 17:301-13. [PMID: 26626700 DOI: 10.1021/acs.biomac.5b01419] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The objective of this study was to design GE11 peptide (YHWYGYTPQNVI) linked micelles of poly(ethylene glycol)-block-poly(2-methyl-2-carboxyl-propylene carbonate-graft-gemcitabine-graft-dodecanol (PEG-b-PCC-g-GEM-g-DC) for enhanced stability and target specificity of gemcitabine (GEM) to EGFR-positive pancreatic cancer cells. GE11-PEG-PCD/mPEG-b-PCC-g-GEM-g-DC mixed micelles showed EGFR-dependent enhanced cellular uptake, and cytotoxicity as compared to scrambled peptide HW12-PEG-PCD/mPEG-b-PCC-g-GEM-g-DC mixed micelles and unmodified mPEG-b-PCC-g-GEM-g-DC micelles. Importantly, GE11-linked mixed micelles preferentially accumulated in orthotopic pancreatic tumor and tumor vasculature at 24 h post systemic administration. GE11-linked mixed micelles inhibited orthotopic pancreatic tumor growth compared to HW12-linked mixed micelles, unmodified mPEG-b-PCC-g-GEM-g-DC micelles, and free GEM formulations. Tumor growth inhibition was mediated by apoptosis of tumor cells and endothelial cells as determined by immunohistochemical staining. In summary, GE11-linked mixed micelles is a promising approach to treat EGFR overexpressing cancers.
Collapse
Affiliation(s)
- Goutam Mondal
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| | - Virender Kumar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| | - Surendra K Shukla
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center , Omaha, Nebraska United States
| | - Pankaj K Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center , Omaha, Nebraska United States
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| |
Collapse
|
27
|
Raynor A, Jantscheff P, Ross T, Schlesinger M, Wilde M, Haasis S, Dreckmann T, Bendas G, Massing U. Saturated and mono-unsaturated lysophosphatidylcholine metabolism in tumour cells: a potential therapeutic target for preventing metastases. Lipids Health Dis 2015; 14:69. [PMID: 26162894 PMCID: PMC4499168 DOI: 10.1186/s12944-015-0070-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 06/29/2015] [Indexed: 01/21/2023] Open
Abstract
Background Metastasis is the leading cause of mortality in malignant diseases. Patients with metastasis often show reduced Lysophosphatidylcholine (LysoPC) plasma levels and treatment of metastatic tumour cells with saturated LysoPC species reduced their metastatic potential in vivo in mouse experiments. To provide a first insight into the interplay of tumour cells and LysoPC, the interactions of ten solid epithelial tumour cell lines and six leukaemic cell lines with saturated and mono-unsaturated LysoPC species were explored. Methods LysoPC metabolism by the different tumour cells was investigated by a combination of cell culture assays, GC and MS techniques. Functional consequences of changed membrane properties were followed microscopically by detecting lateral lipid diffusion or cellular migration. Experimental metastasis studies in mice were performed after pretreatment of B16.F10 melanoma cells with LysoPC and FFA, respectively. Results In contrast to the leukaemic cells, all solid tumour cells show a very fast extracellular degradation of the LysoPC species to free fatty acids (FFA) and glycerophosphocholine. We provide evidence that the formerly LysoPC bound FFA were rapidly incorporated into the cellular phospholipids, thereby changing the FA-compositions accordingly. A massive increase of the neutral lipid amount was observed, inducing the formation of lipid droplets. Saturated LysoPC and to a lesser extent also mono-unsaturated LysoPC increased the cell membrane rigidity, which is assumed to alter cellular functions involved in metastasis. According to that, saturated and mono-unsaturated LysoPC as well as the respective FFA reduced the metastatic potential of B16.F10 cells in mice. Application of high doses of liposomes mainly consisting of saturated PC was shown to be a suitable way to strongly increase the plasma level of saturated LysoPC in mice. Conclusion These data show that solid tumours display a high activity to hydrolyse LysoPC followed by a very rapid uptake of the resulting FFA; a mechanistic model is provided. In contrast to the physiological mix of LysoPC species, saturated and mono-unsaturated LysoPC alone apparently attenuate the metastatic activity of tumours and the artificial increase of saturated and mono-unsaturated LysoPC in plasma appears as novel therapeutic approach to interfere with metastasis.
Collapse
Affiliation(s)
- Anna Raynor
- Department of Lipids & Liposomes, Tumor Biology Center, Clinical Research, Breisacher Str. 117, 79106, Freiburg, Germany.
| | | | - Thomas Ross
- Department of Pharmaceutical Chemistry, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany.
| | - Martin Schlesinger
- Department of Pharmaceutical Chemistry, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany.
| | - Maurice Wilde
- Department of Lipids & Liposomes, Tumor Biology Center, Clinical Research, Breisacher Str. 117, 79106, Freiburg, Germany. .,Institute for Pharmaceutical Sciences, University of Freiburg, Albertstr. 25, 79104, Freiburg, Germany.
| | - Sina Haasis
- Department of Lipids & Liposomes, Tumor Biology Center, Clinical Research, Breisacher Str. 117, 79106, Freiburg, Germany. .,Institute for Pharmaceutical Sciences, University of Freiburg, Albertstr. 25, 79104, Freiburg, Germany.
| | - Tim Dreckmann
- Department of Lipids & Liposomes, Tumor Biology Center, Clinical Research, Breisacher Str. 117, 79106, Freiburg, Germany. .,Institute for Pharmaceutical Sciences, University of Freiburg, Albertstr. 25, 79104, Freiburg, Germany.
| | - Gerd Bendas
- Department of Pharmaceutical Chemistry, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany.
| | - Ulrich Massing
- Department of Lipids & Liposomes, Tumor Biology Center, Clinical Research, Breisacher Str. 117, 79106, Freiburg, Germany. .,Institute for Pharmaceutical Sciences, University of Freiburg, Albertstr. 25, 79104, Freiburg, Germany.
| |
Collapse
|
28
|
Dasa SSK, Suzuki R, Gutknecht M, Brinton LT, Tian Y, Michaelsson E, Lindfors L, Klibanov AL, French BA, Kelly KA. Development of target-specific liposomes for delivering small molecule drugs after reperfused myocardial infarction. J Control Release 2015; 220:556-567. [PMID: 26122651 DOI: 10.1016/j.jconrel.2015.06.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 06/02/2015] [Accepted: 06/14/2015] [Indexed: 01/19/2023]
Abstract
Although reperfusion is essential in restoring circulation to ischemic myocardium, it also leads to irreversible events including reperfusion injury, decreased cardiac function and ultimately scar formation. Various cell types are involved in the multi-phase repair process including inflammatory cells, vascular cells and cardiac fibroblasts. Therapies targeting these cell types in the infarct border zone can improve cardiac function but are limited by systemic side effects. The aim of this work was to develop liposomes with surface modifications to include peptides with affinity for cell types present in the post-infarct myocardium. To identify peptides specific for the infarct/border zone, we used in vivo phage display methods and an optical imaging approach: fluorescence molecular tomography (FMT). We identified peptides specific for cardiomyocytes, endothelial cells, myofibroblasts, and c-Kit + cells present in the border zone of the remodeling infarct. These peptides were then conjugated to liposomes and in vivo specificity and pharmacokinetics were determined. As a proof of concept, cardiomyocyte specific (I-1) liposomes were used to deliver a PARP-1 (poly [ADP-ribose] polymerase 1) inhibitor: AZ7379. Using a targeted liposomal approach, we were able to increase AZ7379 availability in the infarct/border zone at 24h post-injection as compared with free AZ7379. We observed ~3-fold higher efficiency of PARP-1 inhibition when all cell types were assessed using I-1 liposomes as compared with negative control peptide liposomes (NCP). When analyzed further, I-1 liposomes had 9-fold and 1.5-fold higher efficiencies in cardiomyocytes and macrophages, respectively, as compared with NCP liposomes. In conclusion, we have developed a modular drug delivery system that can be targeted to cell types of therapeutic interest in the infarct border zone.
Collapse
Affiliation(s)
- Siva Sai Krishna Dasa
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Ryo Suzuki
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA; Laboratory of Drug and Gene Delivery System, Faculty of Pharma-Sciences, Teikyo University, Tokyo, Japan
| | - Michael Gutknecht
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Lindsey T Brinton
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Yikui Tian
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | | | | | - Alexander L Klibanov
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA; Division of Cardiovascular Medicine, University of Virginia, Charlottesville, VA, USA
| | - Brent A French
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA; Division of Cardiovascular Medicine, University of Virginia, Charlottesville, VA, USA
| | - Kimberly A Kelly
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
29
|
Utama RH, Jiang Y, Zetterlund PB, Stenzel MH. Biocompatible Glycopolymer Nanocapsules via Inverse Miniemulsion Periphery RAFT Polymerization for the Delivery of Gemcitabine. Biomacromolecules 2015; 16:2144-56. [PMID: 26027950 DOI: 10.1021/acs.biomac.5b00545] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Encapsulation of hydrophilic cancer drugs in polymeric nanocapsules was achieved in a one-pot process via the inverse miniemulsion periphery RAFT polymerization (IMEPP) approach. The chosen guest molecule was gemcitabine hydrochloride, which is used as the first-line treatment of pancreatic cancer. The resulting nanocapsules were confirmed to be ∼200 nm, with excellent encapsulation (∼96%) and loading (∼12%) efficiency. Postpolymerization reaction was successfully conducted to create glyocopolymer nanocapsules without any impact on the loads as well as the nanocapsules size or morphology. The loaded nanocapsules were specifically designed to be responsive in a reductive environment. This was confirmed by the successful disintegration of the nanocapsules in the presence of glutathione. The gemcitabine-loaded nanocapsules were tested in vitro against pancreatic cancer cells (AsPC-1), with the results showing an enhancement in the cytotoxicity by two fold due to selective accumulation and release of the nanocapsules within the cells. The results demonstrated the versatility of IMEPP as a tool to synthesize functionalized, loaded-polymeric nanocapsules suitable for drug-delivery application.
Collapse
Affiliation(s)
- Robert H Utama
- ‡Centre for Advanced Macromolecular Design (CAMD), School of Chemistry, University of New South Wales, Sydney 2052, Australia
| | - Yanyan Jiang
- †Centre for Advanced Macromolecular Design (CAMD), School of Chemical Engineering, University of New South Wales, Sydney 2052, Australia.,‡Centre for Advanced Macromolecular Design (CAMD), School of Chemistry, University of New South Wales, Sydney 2052, Australia
| | - Per B Zetterlund
- †Centre for Advanced Macromolecular Design (CAMD), School of Chemical Engineering, University of New South Wales, Sydney 2052, Australia
| | - Martina H Stenzel
- †Centre for Advanced Macromolecular Design (CAMD), School of Chemical Engineering, University of New South Wales, Sydney 2052, Australia.,‡Centre for Advanced Macromolecular Design (CAMD), School of Chemistry, University of New South Wales, Sydney 2052, Australia
| |
Collapse
|
30
|
Kirui DK, Celia C, Molinaro R, Bansal SS, Cosco D, Fresta M, Shen H, Ferrari M. Mild hyperthermia enhances transport of liposomal gemcitabine and improves in vivo therapeutic response. Adv Healthc Mater 2015; 4:1092-103. [PMID: 25721343 PMCID: PMC4433418 DOI: 10.1002/adhm.201400738] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Revised: 01/17/2015] [Indexed: 12/11/2022]
Abstract
Obstructive biological barriers limit the transport and efficacy of cancer nanotherapeutics. Creative manipulation of tumor microenvironment provides promising avenues towards improving chemotherapeutic response. Such strategies include the use of mechanical stimuli to overcome barriers, and increase drug delivery and therapeutic efficacy. The rational use of gold nanorod-mediated mild hyperthermia treatment (MHT) alters tumor transport properties, increases liposomal gemcitabine (Gem Lip) delivery, and antitumor efficacy in pancreatic cancer CAPAN-1 tumor model. MHT treatment leads to a threefold increase in accumulation of 80-nm liposomes and enhances spatial interstitial distribution. I.v. injection of Gem Lip and MHT treatment lead to a threefold increase in intratumor gemcitabine concentration compared to chemotherapeutic infusion alone. Furthermore, combination of MHT treatment with infusion of 12 mg kg(-1) Gem Lip leads to a twofold increase in therapeutic efficacy and inhibition of CAPAN-1 tumor growth when compared to equimolar chemotherapeutic treatment alone. Enhanced therapeutic effect is confirmed by reduction in tumor size and increase in apoptotic index where MHT treatment combined with 12 mg kg(-1) Gem Lip achieves similar therapeutic efficacy as the use of 60 mg kg(-1) free gemcitabine. In conclusion, improvements in vivo efficacy are demonstrated resulting from MHT treatment that overcome transport barriers, promote delivery, improve efficacy of nanomedicines.
Collapse
Affiliation(s)
- Dickson K Kirui
- Department of NanoMedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas, 77030, USA
| | - Christian Celia
- Department of NanoMedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas, 77030, USA
- Department of Pharmacy, University of Chieti – Pescara “G. d’Annunzio”, Chieti, 66013, Italy
| | - Roberto Molinaro
- Department of NanoMedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas, 77030, USA
- Department of Health Sciences, University of Catanzaro “Magna Graecia”, Germaneto – Catanzaro, 88100, Italy
| | - Shyam S. Bansal
- Department of NanoMedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas, 77030, USA
| | - Donato Cosco
- Department of Health Sciences, University of Catanzaro “Magna Graecia”, Germaneto – Catanzaro, 88100, Italy
| | - Massimo Fresta
- Department of Health Sciences, University of Catanzaro “Magna Graecia”, Germaneto – Catanzaro, 88100, Italy
| | - Haifa Shen
- Department of NanoMedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas, 77030, USA
- Department of Cell and Development Biology, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Mauro Ferrari
- Department of NanoMedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas, 77030, USA
- Department of Biomedical Engineering in Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| |
Collapse
|
31
|
Longitudinal Bioluminescence Imaging of Primary Versus Abdominal Metastatic Tumor Growth in Orthotopic Pancreatic Tumor Models in NSG Mice. Pancreas 2015; 44:64-75. [PMID: 25406955 PMCID: PMC4262664 DOI: 10.1097/mpa.0000000000000238] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES The purpose of the present study was to develop and validate noninvasive bioluminescence imaging methods for differentially monitoring primary and abdominal metastatic tumor growth in mouse orthotopic models of pancreatic cancer. METHODS A semiautomated maximum entropy segmentation method was implemented for the primary tumor region of interest, and a rule-based method for manually drawing a region of interest for the abdominal metastatic region was developed for monitoring tumor growth in orthotopic models of pancreatic cancer. The 2 region-of-interest methods were validated by having 2 observers independently segment Panc-1 tumors, and the results were compared with the number of mesenteric lymph node nodules and histopathologic assessment of liver metastases. The findings were extended to orthotopic tumors of the more metastatic MIA PaCa-2 and AsPC-1 cells where separate groups of animals were implanted with different numbers of cells. RESULTS The results demonstrated that the segmentation methods were highly reliable, reproducible, and robust and allowed statistically significant discrimination in the growth rates of primary and abdominal metastatic tumors of different cell lines implanted with different numbers of cells. CONCLUSIONS The present results demonstrate that primary tumors and abdominal metastatic foci in orthotopic pancreatic cancer models can be reliably quantified separately and noninvasively over time with bioluminescence imaging.
Collapse
|
32
|
|
33
|
Core-shell nanoparticulate formulation of gemcitabine: lyophilization, stability studies, and in vivo evaluation. Drug Deliv Transl Res 2014; 4:439-51. [DOI: 10.1007/s13346-014-0206-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
34
|
Zhang J, Miao L, Guo S, Zhang Y, Zhang L, Satterlee A, Kim WY, Huang L. Synergistic anti-tumor effects of combined gemcitabine and cisplatin nanoparticles in a stroma-rich bladder carcinoma model. J Control Release 2014; 182:90-6. [PMID: 24637468 DOI: 10.1016/j.jconrel.2014.03.016] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 02/20/2014] [Accepted: 03/09/2014] [Indexed: 10/25/2022]
Abstract
Tumors grown in a stroma-rich mouse model resembling clinically advanced bladder carcinoma with UMUC3 and NIH 3T3 cells have high levels of fibroblasts and an accelerated tumor growth rate. We used this model to investigate the synergistic effect of combined gemcitabine monophosphate (GMP) nanoparticles and Cisplatin nanoparticles (Combo NP) on tumor-associated fibroblasts (TAFs). A single injection of Combo NP had synergistic anti-tumor effects while the same molar ratio of combined GMP and Cisplatin delivered as free drug (Combo Free) fell outside of the synergistic range. Combo NP nearly halted tumor growth with little evidence of general toxicity while Combo Free had only a modest inhibitory effect at 16mg/kg GMP and 1.6mg/kg Cisplatin. Combo NP increased levels of apoptosis within the tumor by approximately 1.3 folds (TUNEL analysis) and decreased α-SMA-positive fibroblast recruitment by more than 87% (immunofluorescence) after multiple injections compared with Combo Free, GMP NP or Cisplatin NP alone. The TAF-targeting capability of Combo NP was evaluated by double staining for TUNEL and α-SMA at various time points after a single injection. On day one after injection, 57% of the TUNEL-positive cells were identified as α-SMA-positive fibroblasts. By day four, tumor stroma was 85% depleted and 87% of the remaining TAFs were TUNEL-positive. Combo NP-treated tumors became 2.75 folds more permeable than those treated with Combo Free as measured by Evans Blue. We conclude that the antineoplastic effect of Combo NP works by first targeting TAFs and is more effective as an anti-tumor therapy than Combo Free, GMP NP or Cisplatin NP alone.
Collapse
Affiliation(s)
- Jing Zhang
- Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Lei Miao
- Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Shutao Guo
- Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yuan Zhang
- Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lu Zhang
- Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Andrew Satterlee
- Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - William Y Kim
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Leaf Huang
- Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
35
|
Gemcitabine Treatment of Rat Soft Tissue Sarcoma with Phosphatidyldiglycerol-Based Thermosensitive Liposomes. Pharm Res 2014; 31:2276-86. [DOI: 10.1007/s11095-014-1322-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 01/28/2014] [Indexed: 01/12/2023]
|
36
|
May JP, Ernsting MJ, Undzys E, Li SD. Thermosensitive liposomes for the delivery of gemcitabine and oxaliplatin to tumors. Mol Pharm 2013; 10:4499-508. [PMID: 24152292 DOI: 10.1021/mp400321e] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The majority of ultrafast temperature sensitive liposome (uTSL) formulations reported in the literature deliver the highly membrane permeable drug, doxorubicin (DOX). Here we report on the study of the uTSL formulation, HaT (Heat activated cytoToxic, composed of the phospholipid DPPC and the surfactant Brij78) loaded with the water-soluble, but poorly membrane permeable anticancer drugs, gemcitabine (GEM) and oxaliplatin (OXA). The HaT formulation displayed ultrafast release of these drugs in response to temperature, whereas attempts with LTSL (Lyso-lipid Temperature Sensitive Liposome, composed of DPPC, MSPC, and DSPE-PEG) were unsuccessful. HaT-GEM and HaT-OXA both released >80% of the encapsulated drug within 2 min at 40-42 °C, with <5% drug leakage at 37 °C after 30 min in serum. The pharmacokinetic profile of both drugs was improved by formulating with HaT relative to the free drug, with clearance reduced by 50-fold for GEM and 3-fold for OXA. HaT-GEM and HaT-OXA both displayed improved drug uptake in the heated tumor relative to the unheated tumor (by 9-fold and 3-fold, respectively). In particular, HaT-GEM showed 25-fold improved delivery to the heated tumor relative to free GEM and significantly enhanced antitumor efficacy with complete tumor regression after a single dose of HaT-GEM. These data suggest that uTSL technology can also be used to deliver nonmembrane permeable drugs via an intravascular ultrafast release mechanism to great effect.
Collapse
Affiliation(s)
- Jonathan P May
- Drug Delivery and Formulation, Drug Discovery Program, Ontario Institute for Cancer Research , 101 College Street, Suite 800, Toronto, Ontario, M5G 0A3, Canada
| | | | | | | |
Collapse
|
37
|
Chitkara D, Mittal A, Behrman SW, Kumar N, Mahato RI. Self-assembling, amphiphilic polymer-gemcitabine conjugate shows enhanced antitumor efficacy against human pancreatic adenocarcinoma. Bioconjug Chem 2013; 24:1161-73. [PMID: 23758084 DOI: 10.1021/bc400032x] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The therapeutic efficacy of gemcitabine is severely compromised due to its rapid plasma metabolism. Moreover, its hydrophilicity poses a challenge for its efficient entrapment in nanosized delivery systems and to provide a sustained release profile. In this study, gemcitabine was covalently conjugated to poly(ethylene glycol)-block-poly(2-methyl-2-carboxyl-propylene carbonate) (PEG-PCC) which could self-assemble into micelles of 23.6 nm. These micelles afforded protection to gemcitabine from plasma metabolism as evident by negligible amount of gemcitabine and its metabolite dFdU detected in the plasma after 24 h. A controlled release of gemcitabine from the micelles was observed with 53.89% drug release in 10 days in the presence of protease enzyme Cathepsin B. Gemcitabine conjugated micelles were cytotoxic, showed internalization, and induced cell apoptosis in MIA PaCa-2 and L3.6pl pancreatic cancer cell lines. These micelles efficiently inhibited tumor growth when injected intravenously into MIA PaCa-2 cell derived xenograft tumor bearing NSG mice at a dose of 40 mg/kg in terms of reduced tumor volume and tumor weight (0.38 g vs 0.58 g). TUNEL assay revealed that gemcitabine conjugated micelles induced a much higher extent of apoptosis in the tumor tissues compared to free gemcitabine. In conclusion, gemcitabine conjugated micelles were able to enhance the drug payload, protect it from rapid plasma metabolism, and provide a sustained release and showed enhanced antitumor activity, and thus have the potential to provide a better therapeutic alternative for treating pancreatic cancer.
Collapse
Affiliation(s)
- Deepak Chitkara
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | | | | | | | | |
Collapse
|
38
|
Tuli R, Armour M, Surmak A, Reyes J, Iordachita I, Patterson M, Wong J. Accuracy of off-line bioluminescence imaging to localize targets in preclinical radiation research. Radiat Res 2013; 179:416-21. [PMID: 23578189 DOI: 10.1667/rr2999.2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In this study, we investigated the accuracy of using off-line bioluminescence imaging (BLI) and tomography (BLT) to guide irradiation of small soft tissue targets on a small animal radiation research platform (SARRP) with on-board cone beam CT (CBCT) capability. A small glass bulb containing BL cells was implanted as a BL source in the abdomen of 11 mouse carcasses. Bioluminescence imaging and tomography were acquired for each carcass. Six carcasses were setup visually without immobilization and 5 were restrained in position with tape. All carcasses were setup in treatment position on the SARRP where the centroid position of the bulb on CBCT was taken as "truth". In the 2D visual setup, the carcass was setup by aligning the point of brightest luminescence with the vertical beam axis. In the CBCT assisted setup, the pose of the carcass on CBCT was aligned with that on the 2D BL image for setup. For both 2D setup methods, the offset of the bulb centroid on CBCT from the vertical beam axis was measured. In the BLT-CBCT fusion method, the 3D torso on BLT and CBCT was registered and the 3D offset of the respective source centroids was calculated. The setup results were independent of the carcass being immobilized or not due to the onset of rigor mortis. The 2D offset of the perceived BL source position from the CBCT bulb position was 2.3 mm ± 1.3 mm. The 3D offset between BLT and CBCT was 1.5 mm ± 0.9 mm. Given the rigidity of the carcasses, the setup results represent the best that can be achieved with off-line 2D BLI and 3D BLT. The setup uncertainty would require the use of undesirably large margin of 4-5 mm. The results compel the implementation of on-board BLT capability on the SARRP to eliminate setup error and to improve BLT accuracy.
Collapse
Affiliation(s)
- Richard Tuli
- Department of Radiation Oncology, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA.
| | | | | | | | | | | | | |
Collapse
|
39
|
Biocompatible gemcitabine-based nanomedicine engineered by Flow Focusing for efficient antitumor activity. Int J Pharm 2013; 443:103-9. [PMID: 23299085 DOI: 10.1016/j.ijpharm.2012.12.048] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 12/21/2012] [Accepted: 12/30/2012] [Indexed: 11/22/2022]
Abstract
We investigated the incorporation of gemcitabine into a colloidal carrier based on the biodegradable and biocompatible poly(d,l-lactide-co-glycolide) (PLGA) to optimize its anticancer activity. Two synthesis techniques (double emulsion/solvent evaporation, and Flow Focusing) were compared in terms of particle geometry, electrophoretic properties (surface charge), gemcitabine vehiculization capabilities (drug loading and release), blood compatibility, and in vitro antitumor activity. To the best of our knowledge, the second formulation methodology (Flow Focusing) has never been applied to the synthesis of gemcitabine-loaded PLGA particles. With the aim of achieving the finest (nano)formulation, experimental parameters associated to these preparation procedures were analyzed. The electrokinetics of the particles suggested that the chemotherapy agent was incorporated into the polymeric matrix. Blood compatibility was demonstrated in vitro. Flow Focusing led to a more appropriate geometry, higher gemcitabine loading and a sustained release profile. In addition, the cytotoxicity of gemcitabine-loaded particles prepared by Flow Focusing was tested in MCF-7 human breast adenocarcinoma cells, showing significantly greater antitumor activity compared to the free drug and to the gemcitabine-loaded particles synthesized by double emulsion/solvent evaporation. Thus, it has been identified the more adequate formulation conditions in the engineering of gemcitabine-loaded PLGA nanoparticles for the effective treatment of tumours.
Collapse
|
40
|
Brullé L, Vandamme M, Riès D, Martel E, Robert E, Lerondel S, Trichet V, Richard S, Pouvesle JM, Le Pape A. Effects of a non thermal plasma treatment alone or in combination with gemcitabine in a MIA PaCa2-luc orthotopic pancreatic carcinoma model. PLoS One 2012; 7:e52653. [PMID: 23300736 PMCID: PMC3530450 DOI: 10.1371/journal.pone.0052653] [Citation(s) in RCA: 132] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 11/19/2012] [Indexed: 12/01/2022] Open
Abstract
Pancreatic tumors are the gastrointestinal cancer with the worst prognosis in humans and with a survival rate of 5% at 5 years. Nowadays, no chemotherapy has demonstrated efficacy in terms of survival for this cancer. Previous study focused on the development of a new therapy by non thermal plasma showed significant effects on tumor growth for colorectal carcinoma and glioblastoma. To allow targeted treatment, a fibered plasma (Plasma Gun) was developed and its evaluation was performed on an orthotopic mouse model of human pancreatic carcinoma using a MIA PaCa2-luc bioluminescent cell line. The aim of this study was to characterize this pancreatic carcinoma model and to determine the effects of Plasma Gun alone or in combination with gemcitabine. During a 36 days period, quantitative BLI could be used to follow the tumor progression and we demonstrated that plasma gun induced an inhibition of MIA PaCa2-luc cells proliferation in vitro and in vivo and that this effect could be improved by association with gemcitabine possibly thanks to its radiosensitizing properties.
Collapse
Affiliation(s)
- Laura Brullé
- Centre d'Imagerie du Petit Animal-CIPA TAAM, UPS44 CNRS, Orléans, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Federico C, Morittu VM, Britti D, Trapasso E, Cosco D. Gemcitabine-loaded liposomes: rationale, potentialities and future perspectives. Int J Nanomedicine 2012; 7:5423-36. [PMID: 23139626 PMCID: PMC3490684 DOI: 10.2147/ijn.s34025] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
This review describes the strategies used in recent years to improve the biopharmaceutical properties of gemcitabine, a nucleoside analog deoxycytidine antimetabolite characterized by activity against many kinds of tumors, by means of liposomal devices. The main limitation of using this active compound is the rapid inactivation of deoxycytidine deaminase following administration in vivo. Consequently, different strategies based on its encapsulation/complexation in innovative vesicular colloidal carriers have been investigated, with interesting results in terms of increased pharmacological activity, plasma half-life, and tumor localization, in addition to decreased side effects. This review focuses on the specific approaches used, based on the encapsulation of gemcitabine in liposomes, with particular attention to the results obtained during the last 5 years. These approaches represent a valid starting point in the attempt to obtain a novel, commercializable drug formulation as already achieved for liposomal doxorubicin (Doxil®, Caelyx®).
Collapse
Affiliation(s)
- Cinzia Federico
- Department of Health Sciences, Building of BioSciences, University Magna Græcia of Catanzaro, Campus Universitario S Venuta, Germaneto, Italy.
| | | | | | | | | |
Collapse
|
42
|
Papa AL, Basu S, Sengupta P, Banerjee D, Sengupta S, Harfouche R. Mechanistic studies of Gemcitabine-loaded nanoplatforms in resistant pancreatic cancer cells. BMC Cancer 2012; 12:419. [PMID: 22998550 PMCID: PMC3543259 DOI: 10.1186/1471-2407-12-419] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 09/20/2012] [Indexed: 01/06/2023] Open
Abstract
Background Pancreatic cancer remains the deadliest of all cancers, with a mortality rate of 91%. Gemcitabine is considered the gold chemotherapeutic standard, but only marginally improves life-span due to its chemical instability and low cell penetrance. A new paradigm to improve Gemcitabine’s therapeutic index is to administer it in nanoparticles, which favour its delivery to cells when under 500 nm in diameter. Although promising, this approach still suffers from major limitations, as the choice of nanovector used as well as its effects on Gemcitabine intracellular trafficking inside pancreatic cancer cells remain unknown. A proper elucidation of these mechanisms would allow for the elaboration of better strategies to engineer more potent Gemcitabine nanotherapeutics against pancreatic cancer. Methods Gemcitabine was encapsulated in two types of commonly used nanovectors, namely poly(lactic-co-glycolic acid) (PLGA) and cholesterol-based liposomes, and their physico-chemical parameters assessed in vitro. Their mechanisms of action in human pancreatic cells were compared with those of the free drug, and with each others, using cytotoxity, apoptosis and ultrastructural analyses. Results Physico-chemical analyses of both drugs showed high loading efficiencies and sizes of less than 200 nm, as assessed by dynamic light scattering (DLS) and transmission electron microscopy (TEM), with a drug release profile of at least one week. These profiles translated to significant cytotoxicity and apoptosis, as well as distinct intracellular trafficking mechanisms, which were most pronounced in the case of PLGem showing significant mitochondrial, cytosolic and endoplasmic reticulum stresses. Conclusions Our study demonstrates how the choice of nanovector affects the mechanisms of drug action and is a crucial determinant of Gemcitabine intracellular trafficking and potency in pancreatic cancer settings.
Collapse
Affiliation(s)
- Anne-Laure Papa
- BWH-HST Center for Biomedical Engineering, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | | | | | | | | | | |
Collapse
|
43
|
Development of a novel preclinical pancreatic cancer research model: bioluminescence image-guided focal irradiation and tumor monitoring of orthotopic xenografts. Transl Oncol 2012; 5:77-84. [PMID: 22496923 DOI: 10.1593/tlo.11316] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Revised: 12/07/2011] [Accepted: 12/07/2011] [Indexed: 02/07/2023] Open
Abstract
PURPOSE We report on a novel preclinical pancreatic cancer research model that uses bioluminescence imaging (BLI)-guided irradiation of orthotopic xenograft tumors, sparing of surrounding normal tissues, and quantitative, noninvasive longitudinal assessment of treatment response. MATERIALS AND METHODS Luciferase-expressing MiaPaCa-2 pancreatic carcinoma cells were orthotopically injected in nude mice. BLI was compared to pathologic tumor volume, and photon emission was assessed over time. BLI was correlated to positron emission tomography (PET)/computed tomography (CT) to estimate tumor dimensions. BLI and cone-beam CT (CBCT) were used to compare tumor centroid location and estimate setup error. BLI and CBCT fusion was performed to guide irradiation of tumors using the small animal radiation research platform (SARRP). DNA damage was assessed by γ-H2Ax staining. BLI was used to longitudinally monitor treatment response. RESULTS Bioluminescence predicted tumor volume (R = 0.8984) and increased linearly as a function of time up to a 10-fold increase in tumor burden. BLI correlated with PET/CT and necropsy specimen in size (P < .05). Two-dimensional BLI centroid accuracy was 3.5 mm relative to CBCT. BLI-guided irradiated pancreatic tumors stained positively for γ-H2Ax, whereas surrounding normal tissues were spared. Longitudinal assessment of irradiated tumors with BLI revealed significant tumor growth delay of 20 days relative to controls. CONCLUSIONS We have successfully applied the SARRP to a bioluminescent, orthotopic preclinical pancreas cancer model to noninvasively: 1) allow the identification of tumor burden before therapy, 2) facilitate image-guided focal radiation therapy, and 3) allow normalization of tumor burden and longitudinal assessment of treatment response.
Collapse
|
44
|
Yang F, Jin C, Jiang Y, Li J, Di Y, Ni Q, Fu D. Liposome based delivery systems in pancreatic cancer treatment: from bench to bedside. Cancer Treat Rev 2011; 37:633-642. [PMID: 21330062 DOI: 10.1016/j.ctrv.2011.01.006] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Revised: 01/15/2011] [Accepted: 01/21/2011] [Indexed: 12/12/2022]
Abstract
Despite rapid advances in cancer diagnosis and treatment, pancreatic cancer remains one of the most difficult human malignancies to be treated, with a mortality rate nearly equal to its incidence. Although gemcitabine has been established as the standard first-line treatment for advanced pancreatic cancer, gemcitabine-based combination chemotherapy showed either marginal or no improvement in survival. Developments in liposomal delivery systems have facilitated the targeting of specific agents for cancer treatment. Such systems could be developed as platforms for future multi-functional theranostic nanodevices tailor-made for the combined detection of early cancer and functional drug delivery. We systemically review liposome based drug-delivery systems, which can provide improved pharmacokinetics, reduced side effects and potentially increased tumor uptake, for pancreatic cancer therapy. Novel liposomal formulations allowing for higher tumor targeting efficiencies and used in current clinical trials to treat this challenging disease are emphasized.
Collapse
Affiliation(s)
- Feng Yang
- Pancreatic Disease Institute, Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai, China.
| | | | | | | | | | | | | |
Collapse
|
45
|
Celia C, Cosco D, Paolino D, Fresta M. Gemcitabine-loaded innovative nanocarriers vs GEMZAR: biodistribution, pharmacokinetic features and in vivo antitumor activity. Expert Opin Drug Deliv 2011; 8:1609-29. [PMID: 22077480 DOI: 10.1517/17425247.2011.632630] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Gemcitabine, an anticancer drug, is a nucleoside analog deoxycytidine antimetabolite, which acts against a wide range of solid tumors. The limitation of gemcitabine is its rapid inactivation by the deoxycytidine deaminase enzyme following its in vivo administration. AREAS COVERED One of the most promising new approaches for improving the biopharmaceutical properties of gemcitabine is the use of innovative drug delivery devices. This review explains the current status of gemcitabine drug delivery, which has been under development over the past 5 years, with particular emphasis on liposomal delivery. In addition, the use of novel supramolecular vesicular aggregates (SVAs), polymeric nanoparticles and squalenoylation were treated as interesting innovative approaches for the administration of the nucleoside analog. EXPERT OPINION Different colloidal systems containing gemcitabine have been realized, with the aim of providing important potential advancements through traditional ways of therapy. A possible future commercialization of modified gemcitabine is desirable, as was true in the case of liposomal doxorubicin (Doxil(®), Caely(®)).
Collapse
Affiliation(s)
- Christian Celia
- The Methodist Hospital Research Institute, Department of Nanomedicine, 6670 Bertner St, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
46
|
Jantscheff P, Esser N, Geipel A, Woias P, Ziroli V, Goldschmidtboing F, Massing U. Metastasizing, Luciferase Transduced MAT‑Lu Rat Prostate Cancer Models: Follow up of Bolus and Metronomic Therapy with Doxorubicin as Model Drug. Cancers (Basel) 2011; 3:2679-95. [PMID: 24212827 PMCID: PMC3757437 DOI: 10.3390/cancers3022679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 05/16/2011] [Accepted: 06/16/2011] [Indexed: 11/24/2022] Open
Abstract
The most fatal outcomes of prostate carcinoma (PCa) result from hormone-refractory variants of the tumor, especially from metastatic spread rather than from primary tumor burden. The goal of the study was to establish and apply rat MAT-Lu prostate cancer tumor models for improved non-invasive live follow up of tumor growth and metastasis by in vivo bioluminescence. We established luciferase transduced MAT-Lu rat PCa cells and studied tumor growth and metastatic processes in an ectopic as well as orthotopic setting. An intravenous bolus treatment with doxorubicin was used to demonstrate the basic applicability of in vivo imaging to follow up therapeutic intervention in these models. In vitro analysis of tissue homogenates confirmed major metastatic spread of subcutaneous tumors into the lung. Our sensitive method, however, for the first time detects metastasis also in lymph node (11/24), spleen (3/24), kidney (4/24), liver (5/24), and bone tissue (femur or spinal cord - 5/20 and 12/20, respectively). Preliminary data of orthotopic implantation (three animals) showed metastatic invasion to investigated organs in all animals but with varying preference (e.g., to lymph nodes). Intravenous bolus treatment of MAT-Lu PCa with doxorubicin reduced subcutaneous tumor growth by about 50% and the number of animals affected by metastatic lesions in lymph nodes (0/4), lung (3/6) or lumbar spine (0/2), as determined by in vivo imaging and in vitro analysis. Additionally, the possible applicability of the luciferase transduced MAT-Lu model(s) to study basic principles of metronomic therapies via jugular vein catheter, using newly established active microport pumping systems, is presented.
Collapse
Affiliation(s)
- Peter Jantscheff
- Tumour Biology Center, Clinical Research, Department Lipids & Liposomes, Breisacher Str.117, D-79106 Freiburg, Germany; E-Mails: (V.Z.); (U.M.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +49-761-206-1880; Fax: +49-761-206-261-1880
| | - Norbert Esser
- ProQinase GmbH, Breisacher Str. 117, D-79106 Freiburg, Germany; E-Mail:
| | - Andreas Geipel
- Laboratory for Design of Microsystems, Department of Microsystems Engineering (IMTEK), Georges-Köhler-Allee 106, D-79110 Freiburg, Germany; E-Mails: (A.G.); (P.W.); (F.G.)
| | - Peter Woias
- Laboratory for Design of Microsystems, Department of Microsystems Engineering (IMTEK), Georges-Köhler-Allee 106, D-79110 Freiburg, Germany; E-Mails: (A.G.); (P.W.); (F.G.)
| | - Vittorio Ziroli
- Tumour Biology Center, Clinical Research, Department Lipids & Liposomes, Breisacher Str.117, D-79106 Freiburg, Germany; E-Mails: (V.Z.); (U.M.)
| | - Frank Goldschmidtboing
- Laboratory for Design of Microsystems, Department of Microsystems Engineering (IMTEK), Georges-Köhler-Allee 106, D-79110 Freiburg, Germany; E-Mails: (A.G.); (P.W.); (F.G.)
| | - Ulrich Massing
- Tumour Biology Center, Clinical Research, Department Lipids & Liposomes, Breisacher Str.117, D-79106 Freiburg, Germany; E-Mails: (V.Z.); (U.M.)
| |
Collapse
|
47
|
Lysophosphatidylcholine Pretreatment Reduces VLA-4 and P-Selectin–Mediated B16.F10 Melanoma Cell Adhesion In vitro and Inhibits Metastasis-Like Lung Invasion In vivo. Mol Cancer Ther 2011; 10:186-97. [DOI: 10.1158/1535-7163.mct-10-0474] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
48
|
CXCL12 Mediates Immunosuppression in the Lymphoma Microenvironment after Allogeneic Transplantation of Hematopoietic Cells. Cancer Res 2010; 70:10170-81. [DOI: 10.1158/0008-5472.can-10-1943] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
49
|
Elsadek B, Graeser R, Esser N, Schäfer-Obodozie C, Tsurumi C, Abu Ajaj K, Warnecke A, Unger C, Saleem T, Kratz F. In vivo evaluation of a novel albumin-binding prodrug of doxorubicin in an orthotopic mouse model of prostate cancer (LNCaP). Prostate Cancer Prostatic Dis 2010; 14:14-21. [PMID: 21042336 DOI: 10.1038/pcan.2010.43] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
PSA, which is overexpressed in prostate carcinoma, represents a molecular target for selectively releasing an anticancer agent from a prodrug formulation. In this study, we report on the in vivo antitumor efficacy of an efficacious albumin-binding prodrug of doxorubicin (PSA9) that incorporates p-aminobenzyloxycarbonyl (PABC) as a self-immolative spacer in addition to the heptapeptide, Arg-Ser-Ser-Tyr-Tyr-Ser-Leu, which serves as a substrate for PSA. The prodrug is cleaved very efficiently by PSA releasing H-Ser-Leu-PABC-doxorubicin and subsequently doxorubicin in PSA-positive cell lysates and prostate tumor homogenates as the final cleavage product. PSA9 at 3 × 6 mg kg(-1) doxorubicin equivalents (intravenous) was compared with conventional doxorubicin at equitoxic doses (at 3 × 3 mg kg(-1); intravenous) in an orthotopic mouse model of prostate cancer using LNCaP lentiviral luciferase-neomycin cells transduced with luciferase. Whereas doxorubicin did not show any efficacy against the primary tumor or metastases, the prodrug reduced the primary tumor by 30-50% and circulating PSA levels, and in addition, showed a pronounced reduction in lung and bone metastases by ∼77% and ∼96%, respectively, and a positive trend regarding the activity against liver and lymph-node metastases compared with control and doxorubicin-treated animals. The incorporation of PABC as a self-immolative spacer together with a PSA substrate demonstrates superior antitumor effects over doxorubicin attributed to an efficient cleavage by PSA releasing doxorubicin as the final active agent in prostate tumor homogenates. Using this approach for developing effective prodrugs against prostate cancer, is worthy of further preclinical optimization.
Collapse
Affiliation(s)
- B Elsadek
- Division of Macromolecular Prodrugs, Tumor Biology Center, Freiburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Folate-targeted supramolecular vesicular aggregates based on polyaspartyl-hydrazide copolymers for the selective delivery of antitumoral drugs. Biomaterials 2010; 31:7340-54. [PMID: 20609469 DOI: 10.1016/j.biomaterials.2010.05.060] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Accepted: 05/21/2010] [Indexed: 11/20/2022]
Abstract
Supramolecular vesicular aggregates (SVAs) have the advantage of combining the safe and biocompatible properties of colloidal vesicular carriers based on phospholipids with those of polymeric materials, i.e. polyaspartyl-hydrazide (PAHy) copolymers. To provide SVAs with a certain tumour selectivity, folate moieties were chemically conjugated to PAHy copolymers. Physicochemical properties (mean sizes, polydispersity index and zeta potential) of folate-targeted SVAs (FT-SVAs) loaded with gemcitabine were evaluated. The antiproliferative and anticancer activity of gemcitabine-loaded FT-SVAs was evaluated against two cancer cell lines, i.e. MCF-7 cells which over-express the folate receptor and the BxPC-3 cells, which do not over-express this receptor. Gemcitabine-loaded FT-SVAs showed a significantly (p < 0.001) greater and more specific in vitro anticancer activity with respect to both the free drug and the drug-loaded conventional liposomes or untargeted SVAs. Confocal microscopy, flow cytometry analysis and beta-scintillation highlighted that FT-SVAs were able to interact with MCF-7 cells after just 3 h and to increase the amount internalization in cells over-expressing the folate receptor. The in vivo biodistribution and pharmacokinetic experiments showed that gemcitabine-loaded SVAs and FT-SVAs were removed from the circulatory system at a slower rate than the native drug and a prolonged gemcitabine plasma concentration was observed for up to 16 h. SVAs were accumulated mainly in the lungs, spleen and kidneys, while FT-SVAs were also up taken by brain. These interesting and stimulating results suggest the existence of a possible in vivo application of SVAs and encourage the use of folate as a targeting agent in anticancer therapy.
Collapse
|