1
|
Silant’ev VE, Belousov AS, Trukhin FO, Struppul NE, Shmelev ME, Patlay AA, Shatilov RA, Kumeiko VV. Rational Design of Pectin-Chitosan Polyelectrolyte Nanoparticles for Enhanced Temozolomide Delivery in Brain Tumor Therapy. Biomedicines 2024; 12:1393. [PMID: 39061967 PMCID: PMC11273711 DOI: 10.3390/biomedicines12071393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Conventional chemotherapeutic approaches currently used for brain tumor treatment have low efficiency in targeted drug delivery and often have non-target toxicity. Development of stable and effective drug delivery vehicles for the most incurable diseases is one of the urgent biomedical challenges. We have developed polymer nanoparticles (NPs) with improved temozolomide (TMZ) delivery for promising brain tumor therapy, performing a rational design of polyelectrolyte complexes of oppositely charged polysaccharides of cationic chitosan and anionic pectin. The NPs' diameter (30 to 330 nm) and zeta-potential (-29 to 73 mV) varied according to the initial mass ratios of the biopolymers. The evaluation of nanomechanical parameters of native NPs demonstrated changes in Young's modulus from 58 to 234 kPa and adhesion from -0.3 to -3.57 pN. Possible mechanisms of NPs' formation preliminary based on ionic interactions between ionogenic functional groups were proposed by IR spectroscopy and dynamic rheology. The study of the parameters and kinetics of TMZ sorption made it possible to identify compounds that most effectively immobilize and release the active substance in model liquids that simulate the internal environment of the body. A polyelectrolyte carrier based on an equal ratio of pectin-chitosan (0.1% by weight) was selected as the most effective for the delivery of TMZ among a series of obtained NPs, which indicates a promising approach to the treatment of brain tumors.
Collapse
Affiliation(s)
- Vladimir E. Silant’ev
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia; (A.S.B.); (F.O.T.); (N.E.S.); (M.E.S.); (A.A.P.); (R.A.S.)
- Laboratory of Electrochemical Processes, Institute of Chemistry, Far Eastern Branch of Russian Academy of Sciences, Vladivostok 690022, Russia
| | - Andrei S. Belousov
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia; (A.S.B.); (F.O.T.); (N.E.S.); (M.E.S.); (A.A.P.); (R.A.S.)
| | - Fedor O. Trukhin
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia; (A.S.B.); (F.O.T.); (N.E.S.); (M.E.S.); (A.A.P.); (R.A.S.)
| | - Nadezhda E. Struppul
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia; (A.S.B.); (F.O.T.); (N.E.S.); (M.E.S.); (A.A.P.); (R.A.S.)
| | - Mikhail E. Shmelev
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia; (A.S.B.); (F.O.T.); (N.E.S.); (M.E.S.); (A.A.P.); (R.A.S.)
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch of Russian Academy of Sciences, Vladivostok 690041, Russia
| | - Aleksandra A. Patlay
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia; (A.S.B.); (F.O.T.); (N.E.S.); (M.E.S.); (A.A.P.); (R.A.S.)
| | - Roman A. Shatilov
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia; (A.S.B.); (F.O.T.); (N.E.S.); (M.E.S.); (A.A.P.); (R.A.S.)
| | - Vadim V. Kumeiko
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia; (A.S.B.); (F.O.T.); (N.E.S.); (M.E.S.); (A.A.P.); (R.A.S.)
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch of Russian Academy of Sciences, Vladivostok 690041, Russia
| |
Collapse
|
2
|
Hennigan K, Lavik E. Nature vs. Manmade: Comparing Exosomes and Liposomes for Traumatic Brain Injury. AAPS J 2023; 25:83. [PMID: 37610471 DOI: 10.1208/s12248-023-00849-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 08/08/2023] [Indexed: 08/24/2023] Open
Abstract
Traumatic brain injury (TBI) of all severities is a significant public health burden, causing a range of effects that can lead to death or a diminished quality of life. Liposomes and mesenchymal stem cell-derived exosomes are two drug delivery agents with potential to be leveraged in the treatment of TBI by increasing the efficacy of drug therapies as well as having additional therapeutic effects. They exhibit several physical similarities, but key differences affect their performances as nanocarriers. Liposomes can be produced commercially at scale, and liposomes achieve higher encapsulation efficiency. Meanwhile, the intrinsic cargo and targeting moieties of exosomes, which liposomes lack, give exosomes a greater ability to facilitate neural regeneration, and exosomes do not trigger the infusion reactions that liposomes can. However, there are concerns about both exosomes and liposomes regarding interactions with tumors. The same routes of administration can be used for both exosomes and liposomes, resulting in somewhat different distribution throughout the body. While the effect of the nanocarrier type on accumulation in the brain is not concrete, targeting leads to increased accumulation of both exosomes and liposomes in the brain, upon which on-demand release can be used for both drug deliverers. Although neither have been applied to TBI in humans, preclinical trials have shown their immense potential, as have clinical trials pertaining to other brain injuries and conditions. While questions remain, research thus far shows that the various differences make exosomes a better choice of nanocarrier for TBI.
Collapse
Affiliation(s)
- Kate Hennigan
- Marriotts Ridge High School, Ellicott City, Maryland, 21042, USA
| | - Erin Lavik
- University of Maryland, Baltimore County, 1000 Hilltop Circle, Baltimore, Maryland, 21250, USA.
| |
Collapse
|
3
|
Mescheryakova SA, Matlakhov IS, Strokin PD, Drozd DD, Goryacheva IY, Goryacheva OA. Fluorescent Alloyed CdZnSeS/ZnS Nanosensor for Doxorubicin Detection. BIOSENSORS 2023; 13:596. [PMID: 37366961 DOI: 10.3390/bios13060596] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023]
Abstract
Doxorubicin (DOX) is widely used in chemotherapy as an anti-tumor drug. However, DOX is highly cardio-, neuro- and cytotoxic. For this reason, the continuous monitoring of DOX concentrations in biofluids and tissues is important. Most methods for the determination of DOX concentrations are complex and costly, and are designed to determine pure DOX. The purpose of this work is to demonstrate the capabilities of analytical nanosensors based on the quenching of the fluorescence of alloyed CdZnSeS/ZnS quantum dots (QDs) for operative DOX detection. To maximize the nanosensor quenching efficiency, the spectral features of QDs and DOX were carefully studied, and the complex nature of QD fluorescence quenching in the presence of DOX was shown. Using optimized conditions, turn-off fluorescence nanosensors for direct DOX determination in undiluted human plasma were developed. A DOX concentration of 0.5 µM in plasma was reflected in a decrease in the fluorescence intensity of QDs, stabilized with thioglycolic and 3-mercaptopropionic acids, for 5.8 and 4.4 %, respectively. The calculated Limit of Detection values were 0.08 and 0.03 μg/mL using QDs, stabilized with thioglycolic and 3-mercaptopropionic acids, respectively.
Collapse
Affiliation(s)
- Svetlana A Mescheryakova
- Department of General and Inorganic Chemistry, Chemistry Institute, Saratov State University Named after N.G. Chernyshevsky, Astrakhanskaya 83, 410012 Saratov, Russia
| | - Ivan S Matlakhov
- Department of General and Inorganic Chemistry, Chemistry Institute, Saratov State University Named after N.G. Chernyshevsky, Astrakhanskaya 83, 410012 Saratov, Russia
| | - Pavel D Strokin
- Department of General and Inorganic Chemistry, Chemistry Institute, Saratov State University Named after N.G. Chernyshevsky, Astrakhanskaya 83, 410012 Saratov, Russia
| | - Daniil D Drozd
- Department of General and Inorganic Chemistry, Chemistry Institute, Saratov State University Named after N.G. Chernyshevsky, Astrakhanskaya 83, 410012 Saratov, Russia
| | - Irina Yu Goryacheva
- Department of General and Inorganic Chemistry, Chemistry Institute, Saratov State University Named after N.G. Chernyshevsky, Astrakhanskaya 83, 410012 Saratov, Russia
| | - Olga A Goryacheva
- Department of General and Inorganic Chemistry, Chemistry Institute, Saratov State University Named after N.G. Chernyshevsky, Astrakhanskaya 83, 410012 Saratov, Russia
| |
Collapse
|
4
|
Dai L, Li S, Hao Q, Zhou R, Zhou H, Lei W, Kang H, Wu H, Li Y, Ma X. Low-density lipoprotein: a versatile nanoscale platform for targeted delivery. NANOSCALE ADVANCES 2023; 5:1011-1022. [PMID: 36798503 PMCID: PMC9926902 DOI: 10.1039/d2na00883a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/06/2023] [Indexed: 06/18/2023]
Abstract
Low-density lipoprotein (LDL) is a small lipoprotein that plays a vital role in controlling lipid metabolism. LDL has a delicate nanostructure with unique physicochemical properties: superior payload capacity, long residence time in circulation, excellent biocompatibility, smaller size, and natural targeting. In recent decades, the superiority and feasibility of LDL particles as targeted delivery carriers have attracted much attention. In this review, we introduce the structure, composition, advantages, defects, and reconstruction of LDL delivery systems, summarize their research status and progress in targeted diagnosis and therapy, and finally look forward to the clinical application of LDL as an effective delivery vehicle.
Collapse
Affiliation(s)
- Luyao Dai
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University Xi'an Shaanxi 710061 China
- Department of Biophysics, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University Health Science Center Xi'an Shaanxi 710061 China
| | - Shuaijun Li
- Department of Biophysics, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University Health Science Center Xi'an Shaanxi 710061 China
| | - Qian Hao
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University Xi'an Shaanxi 710061 China
- Department of Biophysics, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University Health Science Center Xi'an Shaanxi 710061 China
| | - Ruina Zhou
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University Xi'an Shaanxi 710061 China
- Department of Biophysics, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University Health Science Center Xi'an Shaanxi 710061 China
| | - Hui Zhou
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University Xi'an Shaanxi 710061 China
- Department of Biophysics, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University Health Science Center Xi'an Shaanxi 710061 China
| | - Wenxi Lei
- Department of Biophysics, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University Health Science Center Xi'an Shaanxi 710061 China
| | - Huafeng Kang
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University Xi'an Shaanxi 710061 China
| | - Hao Wu
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University Xi'an Shaanxi 710061 China
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis Sacramento CA 95817 USA
- Department of Biophysics, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University Health Science Center Xi'an Shaanxi 710061 China
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis Sacramento CA 95817 USA
| | - Xiaobin Ma
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University Xi'an Shaanxi 710061 China
| |
Collapse
|
5
|
Adekeye AO, Needham D, Rahman R. Low-Density Lipoprotein Pathway Is a Ubiquitous Metabolic Vulnerability in High Grade Glioma Amenable for Nanotherapeutic Delivery. Pharmaceutics 2023; 15:pharmaceutics15020599. [PMID: 36839921 PMCID: PMC9958636 DOI: 10.3390/pharmaceutics15020599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/12/2023] Open
Abstract
Metabolic reprogramming, through increased uptake of cholesterol in the form of low-density lipoproteins (LDL), is one way by which cancer cells, including high grade gliomas (HGG), maintain their rapid growth. In this study, we determined LDL receptor (LDLR) expression in HGGs using immunohistochemistry on tissue microarrays from intra- and inter tumour regions of 36 adult and 133 paediatric patients to confirm LDLR as a therapeutic target. Additionally, we analysed expression levels in three representative cell line models to confirm their future utility to test LDLR-targeted nanoparticle uptake, retention, and cytotoxicity. Our data show widespread LDLR expression in adult and paediatric cohorts, but with significant intra-tumour variation observed between the core and either rim or invasive regions of adult HGG. Expression was independent of paediatric tumour grade or identified clinicopathological factors. LDLR-expressing tumour cells localized preferentially within perivascular niches, also with significant adult intra-tumour variation. We demonstrated variable levels of LDLR expression in all cell lines, confirming their suitability as models to test LDLR-targeted nanotherapy delivery. Overall, our study reveals the LDLR pathway as a ubiquitous metabolic vulnerability in high grade gliomas across all ages, amenable to future consideration of LDL-mediated nanoparticle/drug delivery to potentially circumvent tumour heterogeneity.
Collapse
Affiliation(s)
- Adenike O. Adekeye
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
| | - David Needham
- Department of Mechanical Engineering and Material Science, School of Engineering, Duke University, Durham, NC 27708, USA
| | - Ruman Rahman
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
- Correspondence:
| |
Collapse
|
6
|
Ziganshina AY, Mansurova EE, Voloshina AD, Lyubina AP, Amerhanova SK, Shulaeva MM, Nizameev IR, Kadirov MK, Bakhtiozina LR, Semenov VE, Antipin IS. Thymine-Modified Nanocarrier for Doxorubicin Delivery in Glioblastoma Cells. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020551. [PMID: 36677608 PMCID: PMC9864328 DOI: 10.3390/molecules28020551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023]
Abstract
Brain tumor glioblastoma is one of the worst types of cancer. The blood-brain barrier prevents drugs from reaching brain cells and shields glioblastoma from treatment. The creation of nanocarriers to improve drug delivery and internalization effectiveness may be the solution to this issue. In this paper, we report on a new nanocarrier that was developed to deliver the anticancer drug doxorubicin to glioblastoma cells. The nanocarrier was obtained by nanoemulsion polymerization of diallyl disulfide with 1-allylthymine. Diallyl disulfide is a redox-sensitive molecule involved in redox cell activities, and thymine is a uracil derivative and one of the well-known bioactive compounds that can enhance the pharmacological activity of doxorubicin. Doxorubicin was successfully introduced into the nanocarrier with a load capacity of about 4.6%. Biological studies showed that the doxorubicin nanocarrier composition is far more cytotoxic to glioblastoma cells (T98G) than it is to cancer cells (M-HeLa) and healthy cells (Chang liver). The nanocarrier improves the penetration of doxorubicin into T98G cells and accelerates the cells' demise, as is evident from flow cytometry and fluorescence microscopy data. The obtained nanocarrier, in our opinion, is a promising candidate for further research in glioblastoma therapy.
Collapse
Affiliation(s)
- Albina Y. Ziganshina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Arbuzov Str. 8, 420088 Kazan, Russia
- Correspondence:
| | - Elina E. Mansurova
- Alexander Butlerov Institute of Chemistry, Kazan Federal University, Lobachevsky Str. 1/29, 420008 Kazan, Russia
| | - Alexandra D. Voloshina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Arbuzov Str. 8, 420088 Kazan, Russia
| | - Anna P. Lyubina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Arbuzov Str. 8, 420088 Kazan, Russia
| | - Syumbelya K. Amerhanova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Arbuzov Str. 8, 420088 Kazan, Russia
| | - Marina M. Shulaeva
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Arbuzov Str. 8, 420088 Kazan, Russia
| | - Irek R. Nizameev
- Department of Nanotechnologies in Electronics, Kazan National Research Technical University Named after A. N. Tupolev—KAI, 10, K. Marx Str., 420111 Kazan, Russia
| | - Marsil K. Kadirov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Arbuzov Str. 8, 420088 Kazan, Russia
| | - Leysan R. Bakhtiozina
- Alexander Butlerov Institute of Chemistry, Kazan Federal University, Lobachevsky Str. 1/29, 420008 Kazan, Russia
| | - Vyacheslav E. Semenov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Arbuzov Str. 8, 420088 Kazan, Russia
| | - Igor S. Antipin
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Arbuzov Str. 8, 420088 Kazan, Russia
| |
Collapse
|
7
|
Ghosh P, Tiwari H, Lakkakula J, Roy A, Emran TB, Rashid S, Alghamdi S, Rajab BS, Almehmadi M, Allahyani M, Aljuaid A, Alsaiari AA, Sharma R, Babalghith AO. A decade's worth of impact: Dox loaded liposomes in anticancer activity. MATERIALS TODAY ADVANCES 2022; 16:100313. [DOI: 10.1016/j.mtadv.2022.100313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
8
|
Jaragh-Alhadad L, Samir M, Harford TJ, Karnik S. Low-density lipoprotein encapsulated thiosemicarbazone metal complexes is active targeting vehicle for breast, lung, and prostate cancers. Drug Deliv 2022; 29:2206-2216. [PMID: 35815732 PMCID: PMC9278447 DOI: 10.1080/10717544.2022.2096713] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/21/2022] [Accepted: 06/27/2022] [Indexed: 01/03/2023] Open
Abstract
Cancer is a leading cause of death worldwide and affects society in terms of the number of lives lost. Current cancer treatments are based on conventional chemotherapy which is nonspecific in targeting cancer. Therefore, intensive efforts are underway to better target cancer-specific cells while minimizing the side effects on healthy tissues by using LDL particles as active drug delivery vehicles. The goal is to encapsulate anticancer agents thiosemicarbazone metal-ligand complexes into LDL particles to increase the cytotoxic effect of the agent by internalization through LDL receptors into MCF7, A549, and C42 cancer cell lines as segregate models for biological evaluations targeting tubulin. Zeta potential data of LDL-particles encapsulated anticancer agents showed an acceptable diameter range between 66-91 nm and uniform particle morphology. The results showed cell proliferation reduction in all tested cell lines. The IC50 values of LDL encapsulated thiosemicarbazone metal-ligand complexes treated with MCF7, A549, and C42 ranged between 1.18-6.61 µM, 1.17-9.66 µM, and 1.01-6.62 µM, respectively. Western blot analysis showed a potent decrease in tubulin expression when the cell lines were treated with LDL particles encapsulated with thiosemicarbazone metal-ligand complexes as anticancer agents. In conclusion, the data provide strong evidence that LDL particles are used as an active drug delivery strategy for cancer therapy.
Collapse
Affiliation(s)
- Laila Jaragh-Alhadad
- Department of Chemistry, Faculty of Science, Kuwait University, Kuwait, Safat, Kuwait
- Cardiovascular and Metabolic Sciences Department, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| | - Mayada Samir
- Department of Chemistry, Faculty of Science, Kuwait University, Kuwait, Safat, Kuwait
| | - Terri J. Harford
- Cardiovascular and Metabolic Sciences Department, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| | - Sadashiva Karnik
- Cardiovascular and Metabolic Sciences Department, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
- Cleveland Clinic Learner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
9
|
Surface-modified lipid nanocarriers for crossing the blood-brain barrier (BBB): a current overview of active targeting in brain diseases. Colloids Surf B Biointerfaces 2022; 221:112999. [DOI: 10.1016/j.colsurfb.2022.112999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/26/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022]
|
10
|
Brookes A, Ji L, Bradshaw TD, Stocks M, Gray D, Butler J, Gershkovich P. Is Oral Lipid-Based Delivery for Drug Targeting to the Brain Feasible? Eur J Pharm Biopharm 2022; 172:112-122. [PMID: 35149190 DOI: 10.1016/j.ejpb.2022.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/21/2022] [Accepted: 02/05/2022] [Indexed: 12/12/2022]
Abstract
This review outlines the feasibility of oral lipid-based targeted delivery of drugs to the brain, including permeation of the central nervous system's (CNS) protective blood-brain barrier (BBB). The structure of the BBB and disruption caused by varying disease states highlights the need for disease-specific approaches to alter permeation. Disruption during disease state, and the effects of certain molecules on the barrier, demonstrate the possibility of exploiting such BBB disruption for drug delivery. Many administration methods can be used to target the brain, but oral administration is considered ideal for chronic, long-term illnesses. Several lipids that have been shown to facilitate drug delivery into the brain after systemic administration, but could also be delivered orally are discussed, including oleic acid, triolein, alkylglycerol, and conjugates of linoleic and myristic acids. Current data reveal the potential for the use of such lipids as part of oral formulations for delivery to the brain by reaching sufficient plasma levels after administration to increase the permeability of the BBB. However, gaps in the literature remain regarding the concentrations and form of most lipids required to produce the desired effects. The use of lipids via oral delivery for brain targeting has not been investigated thoroughly enough to determine with certainty if similar permeability-enhancing effects would be observed as for parenteral administration. In conclusion, further research to fill research gaps is needed, but the limited evidence suggests that oral lipid-based drug delivery for brain targeting is potentially feasible.
Collapse
Affiliation(s)
- Alice Brookes
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD
| | - Liuhang Ji
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD
| | - Tracey D Bradshaw
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD
| | - Michael Stocks
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD
| | - David Gray
- Division of Food, Nutrition and Dietetics, University of Nottingham, Sutton Bonington Campus, Loughborough, Leicestershire, UK, LE12 5RD
| | - James Butler
- GlaxoSmithKline Research and Development, Park Road, Ware, Hertfordshire, UK, SG12 0DP
| | - Pavel Gershkovich
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD.
| |
Collapse
|
11
|
Blood-Brain Barrier in Brain Tumors: Biology and Clinical Relevance. Int J Mol Sci 2021; 22:ijms222312654. [PMID: 34884457 PMCID: PMC8657947 DOI: 10.3390/ijms222312654] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/13/2021] [Accepted: 11/18/2021] [Indexed: 12/13/2022] Open
Abstract
The presence of barriers, such as the blood–brain barrier (BBB) and brain–tumor barrier (BTB), limits the penetration of antineoplastic drugs into the brain, resulting in poor response to treatments. Many techniques have been developed to overcome the presence of these barriers, including direct injections of substances by intranasal or intrathecal routes, chemical modification of drugs or constituents of BBB, inhibition of efflux pumps, physical disruption of BBB by radiofrequency electromagnetic radiation (EMP), laser-induced thermal therapy (LITT), focused ultrasounds (FUS) combined with microbubbles and convection enhanced delivery (CED). However, most of these strategies have been tested only in preclinical models or in phase 1–2 trials, and none of them have been approved for treatment of brain tumors yet. Concerning the treatment of brain metastases, many molecules have been developed in the last years with a better penetration across BBB (new generation tyrosine kinase inhibitors like osimertinib for non-small-cell lung carcinoma and neratinib/tucatinib for breast cancer), resulting in better progression-free survival and overall survival compared to older molecules. Promising studies concerning neural stem cells, CAR-T (chimeric antigen receptors) strategies and immunotherapy with checkpoint inhibitors are ongoing.
Collapse
|
12
|
Optimizing the Design of Blood-Brain Barrier-Penetrating Polymer-Lipid-Hybrid Nanoparticles for Delivering Anticancer Drugs to Glioblastoma. Pharm Res 2021; 38:1897-1914. [PMID: 34655006 DOI: 10.1007/s11095-021-03122-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/07/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Chemotherapy for glioblastoma multiforme (GBM) remains ineffective due to insufficient penetration of therapeutic agents across the blood-brain barrier (BBB) and into the GBM tumor. Herein, is described, the optimization of the lipid composition and fabrication conditions for a BBB- and tumor penetrating terpolymer-lipid-hybrid nanoparticle (TPLN) for delivering doxorubicin (DOX) to GBM. METHODS The composition of TPLNs was first screened using different lipids based on nanoparticle properties and in vitro cytotoxicity by using 23 full factorial experimental design. The leading DOX loaded TPLNs (DOX-TPLN) were prepared by further optimization of conditions and used to study cellular uptake mechanisms, in vitro cytotoxicity, three-dimensional (3D) glioma spheroid penetration, and in vivo biodistribution in a murine orthotopic GBM model. RESULTS Among various lipids studied, ethyl arachidate (EA) was found to provide excellent nanoparticle properties e.g., size, polydispersity index (PDI), zeta potential, encapsulation efficiency, drug loading, and colloidal stability, and highest anticancer efficacy for DOX-TPLN. Further optimized EA-based TPLNs were prepared with an optimal particle size (103.8 ± 33.4 nm) and PDI (0.208 ± 0.02). The resultant DOX-TPLNs showed ~ sevenfold higher efficacy than free DOX against human GBM U87-MG-RED-FLuc cells in vitro. The interaction between the TPLNs and the low-density lipoprotein receptors also facilitated cellular uptake, deep penetration into 3D glioma spheroids, and accumulation into the in vivo brain tumor regions of DOX-TPLNs. CONCLUSION This work demonstrated that the TPLN system can be optimized by rational selection of lipid type, lipid content, and preparation conditions to obtain DOX-TPLN with enhanced anticancer efficacy and GBM penetration and accumulation.
Collapse
|
13
|
Garofalo T, Misasi R, Preta G. Editorial: Targeting Lipid Rafts as a Strategy Against Infection and Cancer. Front Cell Dev Biol 2021; 9:748905. [PMID: 34552936 PMCID: PMC8450352 DOI: 10.3389/fcell.2021.748905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 08/13/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Tina Garofalo
- Department of Experimental Medicine, "Sapienza" University, Rome, Italy
| | - Roberta Misasi
- Department of Experimental Medicine, "Sapienza" University, Rome, Italy
| | - Giulio Preta
- Institute of Biochemistry, Life Science Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
14
|
Han L, Jiang C. Evolution of blood-brain barrier in brain diseases and related systemic nanoscale brain-targeting drug delivery strategies. Acta Pharm Sin B 2021; 11:2306-2325. [PMID: 34522589 PMCID: PMC8424230 DOI: 10.1016/j.apsb.2020.11.023] [Citation(s) in RCA: 143] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/30/2020] [Accepted: 10/09/2020] [Indexed: 02/07/2023] Open
Abstract
Blood–brain barrier (BBB) strictly controls matter exchange between blood and brain, and severely limits brain penetration of systemically administered drugs, resulting in ineffective drug therapy of brain diseases. However, during the onset and progression of brain diseases, BBB alterations evolve inevitably. In this review, we focus on nanoscale brain-targeting drug delivery strategies designed based on BBB evolutions and related applications in various brain diseases including Alzheimer's disease, Parkinson's disease, epilepsy, stroke, traumatic brain injury and brain tumor. The advances on optimization of small molecules for BBB crossing and non-systemic administration routes (e.g., intranasal treatment) for BBB bypassing are not included in this review.
Collapse
Key Words
- AD, Alzheimer's disease
- AMT, alpha-methyl-l-tryptophan
- Aβ, amyloid beta
- BACE1, β-secretase 1
- BBB, blood–brain barrier
- BDNF, brain derived neurotrophic factor
- BTB, blood–brain tumor barrier
- Blood–brain barrier
- Brain diseases
- Brain-targeting
- CMT, carrier-mediated transportation
- DTPA-Gd, Gd-diethyltriaminepentaacetic acid
- Drug delivery systems
- EPR, enhanced permeability and retention
- GLUT1, glucose transporter-1
- Gd, gadolinium
- ICAM-1, intercellular adhesion molecule-1
- KATP, ATP-sensitive potassium channels
- KCa, calcium-dependent potassium channels
- LAT1, L-type amino acid transporter 1
- LDL, low density lipoprotein
- LDLR, LDL receptor
- LFA-1, lymphocyte function associated antigen-1
- LRP1, LDLR-related protein 1
- MFSD2A, major facilitator superfamily domain-containing protein 2a
- MMP9, metalloproteinase-9
- MRI, magnetic resonance imaging
- NPs, nanoparticles
- Nanoparticles
- P-gp, P-glycoprotein
- PD, Parkinson's disease
- PEG, polyethyleneglycol
- PEG-PLGA, polyethyleneglycol-poly(lactic-co-glycolic acid)
- PLGA, poly(lactic-co-glycolic acid)
- PSMA, prostate-specific membrane antigen
- RAGE, receptor for advanced glycosylation end products
- RBC, red blood cell
- RMT, receptor-mediated transcytosis
- ROS, reactive oxygen species
- TBI, traumatic brain injury
- TJ, tight junction
- TfR, transferrin receptor
- VEGF, vascular endothelial growth factor
- ZO1, zona occludens 1
- siRNA, short interfering RNA
- tPA, tissue plasminogen activator
Collapse
Affiliation(s)
- Liang Han
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
- Corresponding author. Tel./fax: +86 512 65882089.
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 200032, China
| |
Collapse
|
15
|
Crossing the blood-brain barrier: A review on drug delivery strategies using colloidal carrier systems. Neurochem Int 2021; 147:105017. [PMID: 33887377 DOI: 10.1016/j.neuint.2021.105017] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/27/2021] [Accepted: 03/06/2021] [Indexed: 02/05/2023]
Abstract
The blood-brain barrier represents the major challenge for delivering drugs to the central nervous system (CNS). It separates the blood circulation from the brain tissue, thereby protecting the CNS and maintaining its ion homeostasis. Unfortunately, most drugs are not able to cross this barrier in vivo despite promising in vitro results. One approach to solve this problem is the delivery of drugs via surface modified nanocarrier systems. This review will give an overview on currently tested systems, mainly liposomes and solid nanoparticles and inform about new developments.
Collapse
|
16
|
LDL receptors and their role in targeted therapy for glioma: a review. Drug Discov Today 2021; 26:1212-1225. [PMID: 33609780 DOI: 10.1016/j.drudis.2021.02.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/15/2021] [Accepted: 02/06/2021] [Indexed: 11/22/2022]
Abstract
Gliomas are highly lethal forms of cancers occurring in the brain. Delivering the drugs into the brain is a major challenge to the treatment of gliomas because of the highly selectively permeable blood-brain barrier (BBB). Tapping the potential of receptor-mediated drug delivery systems using targeted nanoparticles (NPs) is a sought-after step forward toward successful glioma treatment. Several receptors are the focus of research for application in drug delivery. Low-density lipoprotein receptors (LDLR) are abundantly expressed in both healthy brains and diseased brains with a disrupted BBB. In this review, we discuss the LDLR and the types of NPs that have been used to target the brain via this receptor.
Collapse
|
17
|
Di L, Maiseyeu A. Low-density lipoprotein nanomedicines: mechanisms of targeting, biology, and theranostic potential. Drug Deliv 2021; 28:408-421. [PMID: 33594923 PMCID: PMC7894439 DOI: 10.1080/10717544.2021.1886199] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Native nanostructured lipoproteins such as low- and high-density lipoproteins (LDL and HDL) are powerful tools for the targeted delivery of drugs and imaging agents. While the cellular recognition of well-known HDL-based carriers occurs via interactions with an HDL receptor, the selective delivery and uptake of LDL particles by target cells are more complex. The most well-known mode of LDL-based delivery is via the interaction between apolipoprotein B (Apo-B) - the main protein of LDL - and the low-density lipoprotein receptor (LDLR). LDLR is expressed in the liver, adipocytes, and macrophages, and thus selectively delivers LDL carriers to these cells and tissues. Moreover, the elevated expression of LDLR in tumor cells indicates a role for LDL in the targeted delivery of chemotherapy drugs. In addition, chronic inflammation associated with hypercholesterolemia (i.e., high levels of endogenous LDL) can be abated by LDL carriers, which outcompete the deleterious oxidized LDL for uptake by macrophages. In this case, synthetic LDL nanocarriers act as 'eat-me' signals and exploit mechanisms of native LDL uptake for targeted drug delivery and imaging. Lastly, recent studies have shown that the delivery of LDL-based nanocarriers to macrophages via fluid-phase pinocytosis is a promising tool for atherosclerosis imaging. Hence, the present review summarizes the use of natural and synthetic LDL-based carriers for drug delivery and imaging and discusses various mechanisms of targeting.
Collapse
Affiliation(s)
- Lin Di
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Clevehand, OH, USA
| | - Andrei Maiseyeu
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Clevehand, OH, USA
| |
Collapse
|
18
|
Nanotechnology and Nanocarrier-Based Drug Delivery as the Potential Therapeutic Strategy for Glioblastoma Multiforme: An Update. Cancers (Basel) 2021; 13:cancers13020195. [PMID: 33430494 PMCID: PMC7827410 DOI: 10.3390/cancers13020195] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/20/2020] [Accepted: 01/04/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Glioblastoma multiforme (GBM) are among the most lethal tumors. The highly invasive nature and presence of GBM stem cells, as well as the blood brain barrier (BBB) which limits chemotherapeutic drugs from entering the tumor mass, account for the high chance of treatment failure. Recent developments have found that nanoparticles can be conjugated to liposomes, dendrimers, metal irons, or polymeric micelles, which enhance the drug-loaded compounds to efficiently penetrate the BBB, thus offering new possibilities for overcoming GBM stem cell-mediated resistance to chemotherapy and radiation therapy. In addition, there have been new emerging strategies that use nanocarriers for successful GBM treatment in animal models. This review highlights the recent development of nanotechnology and nanocarrier-based drug delivery for treatment of GBMs, which may be a promising therapeutic strategy for this tumor entity. Abstract Glioblastoma multiforme (GBM) is the most common and malignant brain tumor with poor prognosis. The heterogeneous and aggressive nature of GBMs increases the difficulty of current standard treatment. The presence of GBM stem cells and the blood brain barrier (BBB) further contribute to the most important compromise of chemotherapy and radiation therapy. Current suggestions to optimize GBM patients’ outcomes favor controlled targeted delivery of chemotherapeutic agents to GBM cells through the BBB using nanoparticles and monoclonal antibodies. Nanotechnology and nanocarrier-based drug delivery have recently gained attention due to the characteristics of biosafety, sustained drug release, increased solubility, and enhanced drug bioactivity and BBB penetrability. In this review, we focused on recently developed nanoparticles and emerging strategies using nanocarriers for the treatment of GBMs. Current studies using nanoparticles or nanocarrier-based drug delivery system for treatment of GBMs in clinical trials, as well as the advantages and limitations, were also reviewed.
Collapse
|
19
|
SPION and doxorubicin-loaded polymeric nanocarriers for glioblastoma theranostics. Drug Deliv Transl Res 2021; 11:515-523. [PMID: 33405212 DOI: 10.1007/s13346-020-00880-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2020] [Indexed: 12/12/2022]
Abstract
Glioma is a type of cancer with a very poor prognosis with a survival of around 15 months in the case of glioblastoma multiforme (GBM). In order to advance in personalized medicine, we developed polymeric nanoparticles (PNP) loaded with both SPION (superparamagnetic iron oxide nanoparticles) and doxorubicin (DOX). The former being used for its potential to accumulate the PNP in the tumor under a strong magnetic field and the later for its therapeutic potential. The emulsion solvent and evaporation method was selected to develop monodisperse PNP with high loading efficiency in both SPION and DOX. Once injected in mice, a significant accumulation of the PNP was observed within the tumoral tissue under static magnetic field as observed by MRI leading to a reduction of tumor growth rate.
Collapse
|
20
|
Preta G. New Insights Into Targeting Membrane Lipids for Cancer Therapy. Front Cell Dev Biol 2020; 8:571237. [PMID: 32984352 PMCID: PMC7492565 DOI: 10.3389/fcell.2020.571237] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/13/2020] [Indexed: 12/15/2022] Open
Abstract
Modulation of membrane lipid composition and organization is currently developing as an effective therapeutic strategy against a wide range of diseases, including cancer. This field, known as membrane-lipid therapy, has risen from new discoveries on the complex organization of lipids and between lipids and proteins in the plasma membranes. Membrane microdomains present in the membrane of all eukaryotic cells, known as lipid rafts, have been recognized as an important concentrating platform for protein receptors involved in the regulation of intracellular signaling, apoptosis, redox balance and immune response. The difference in lipid composition between the cellular membranes of healthy cells and tumor cells allows for the development of novel therapies based on targeting membrane lipids in cancer cells to increase sensitivity to chemotherapeutic agents and consequently defeat multidrug resistance. In the current manuscript strategies based on influencing cholesterol/sphingolipids content will be presented together with innovative ones, more focused in changing biophysical properties of the membrane bilayer without affecting the composition of its constituents.
Collapse
Affiliation(s)
- Giulio Preta
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
21
|
Azarmi M, Maleki H, Nikkam N, Malekinejad H. Transcellular brain drug delivery: A review on recent advancements. Int J Pharm 2020; 586:119582. [DOI: 10.1016/j.ijpharm.2020.119582] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 06/08/2020] [Accepted: 06/23/2020] [Indexed: 02/07/2023]
|
22
|
Ricca A, Cascino F, Morena F, Martino S, Gritti A. In vitro Validation of Chimeric β-Galactosylceramidase Enzymes With Improved Enzymatic Activity and Increased Secretion. Front Mol Biosci 2020; 7:167. [PMID: 32850960 PMCID: PMC7396597 DOI: 10.3389/fmolb.2020.00167] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/01/2020] [Indexed: 12/27/2022] Open
Abstract
Globoid Cell Leukodystrophy (GLD) is a lysosomal storage disease (LSD) caused by inherited defects of the β-galactosylceramidase (GALC) gene. The infantile forms display a rapid and aggressive central and peripheral nervous system (CNS and PNS) dysfunction. No treatments are available for GLD patients. Effective gene therapy (GT) strategies for GLD require a safe and widespread delivery of the functional GALC enzyme to all affected tissues/organs, and particularly to the CNS. The use of chimeric lysosomal enzymes with increased secretion and enhanced transport across the blood-brain barrier (BBB) that boost the efficacy of GT approaches in pre-clinical models of similar neurodegenerative LSDs may benefit GLD as well. Here, we tested the safety and biological efficacy of chimeric GALC enzymes engineered to express an alternative signal peptide (iduronate-2-sulfatase - IDSsp) and the low-density lipoprotein receptor (LDLr)-binding domain from the Apolipoprotein E II (ApoE II) in GLD murine neural and hematopoietic stem/progenitor cells and progeny, which are relevant cells types in the context of in vivo and ex vivo GT platforms. We show that the lentiviral vector-mediated expression of the chimeric GALC enzymes is safe and leads to supranormal enzymatic activity in both neural and hematopoietic cells. The IDSsp.GALC shows enhanced expression and secretion in comparison to the unmodified GALC. The chimeric GALC enzymes produced by LV-transduced cells reduce intracellular galactosylceramide (GalCer) storage and effectively cross-correct GLD murine neurons and glial cells, indicating that the transgenic enzymes are delivered to lysosomes, efficiently secreted, and functional. Of note, the expression of LDLr and LDLr-related proteins in GLD neurons and glial cells supports the exploitation of this system to enhance the GALC supply in affected CNS cells and tissues. These in vitro studies support the use of chimeric GALC enzymes to develop novel and more effective GT approaches for GLD.
Collapse
Affiliation(s)
- Alessandra Ricca
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Federica Cascino
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
23
|
Salaroglio IC, Abate C, Rolando B, Battaglia L, Gazzano E, Colombino E, Costamagna C, Annovazzi L, Mellai M, Berardi F, Capucchio MT, Schiffer D, Riganti C. Validation of Thiosemicarbazone Compounds as P-Glycoprotein Inhibitors in Human Primary Brain-Blood Barrier and Glioblastoma Stem Cells. Mol Pharm 2019; 16:3361-3373. [PMID: 31265310 DOI: 10.1021/acs.molpharmaceut.9b00018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
P-glycoprotein (Pgp) is highly expressed on blood-brain barrier (BBB) and glioblastoma (GB) cells, particularly on cancer stem cells (SC). Pgp recognizes a broad spectrum of substrates, limiting the therapeutic efficacy of several chemotherapeutic drugs in eradicating GB SC. Finding effective and safe inhibitors of Pgp that improve drug delivery across the BBB and target GB SC is open to investigation. We previously identified a series of thiosemicarbazone compounds that inhibit Pgp with an EC50 in the nanomolar range, and herein, we investigate the efficacy of three of them in bypassing Pgp-mediated drug efflux in primary human BBB and GB cells. At 10 nM, the compounds were not cytotoxic for the brain microvascular endothelial hCMEC/D3 cell line, but they markedly enhanced the permeability of the Pgp-substrate doxorubicin through the BBB. Thiosemicarbazone derivatives increased doxorubicin uptake in GB, with greater effects in the Pgp-rich SC clones than in the differentiated clones derived from the same tumor. All compounds increased intratumor doxorubicin accumulation and consequent toxicity in GB growing under competent BBB, producing significant killing of GB SC. The compounds crossed the BBB monolayer. The most stable derivative, 10a, had a half-life in serum of 4.2 h. The coadministration of doxorubicin plus 10a significantly reduced the growth of orthotopic GB-SC xenografts, without eliciting toxic side effects. Our work suggests that the thiosemicarbazone compounds are able to transform doxorubicin, a prototype BBB-impermeable drug, into a BBB-permeable drug. Bypassing Pgp-mediated drug efflux in both BBB and GB SC, thiosemicarbazones might increase the success of chemotherapy in targeting GB SC, which represent the most aggressive and difficult components to eradicate.
Collapse
Affiliation(s)
- Iris Chiara Salaroglio
- Dipartimento di Oncologia , Università di Torino , via Santena 5/bis , 10126 Torino , Italy
| | - Carmen Abate
- Dipartimento di Farmacia-Scienze del Farmaco , Università di Bari "Aldo Moro" , Via Orabona 4 , 70125 Bari , Italy
| | - Barbara Rolando
- Dipartimento di Scienza e Tecnologia del Farmaco , Università di Torino , via Pietro Giuria 9 , 10125 Torino , Italy
| | - Luigi Battaglia
- Dipartimento di Scienza e Tecnologia del Farmaco , Università di Torino , via Pietro Giuria 9 , 10125 Torino , Italy
| | - Elena Gazzano
- Dipartimento di Oncologia , Università di Torino , via Santena 5/bis , 10126 Torino , Italy
| | - Elena Colombino
- Dipartimento di Scienze Veterinarie , Università di Torino , Largo Braccini 2 , 10095 Grugliasco , Italy
| | - Costanzo Costamagna
- Dipartimento di Oncologia , Università di Torino , via Santena 5/bis , 10126 Torino , Italy
| | - Laura Annovazzi
- Centro Ricerche , Fondazione Policlinico di Monza , via Pietro Micca 29 , 13100 Vercelli , Italy
| | - Marta Mellai
- Dipartimento di Scienze della Salute , Università del Piemonte Orientale , corso Mazzini 18 , 28100 Novara , Italy
| | - Francesco Berardi
- Dipartimento di Farmacia-Scienze del Farmaco , Università di Bari "Aldo Moro" , Via Orabona 4 , 70125 Bari , Italy
| | - Maria Teresa Capucchio
- Dipartimento di Scienze Veterinarie , Università di Torino , Largo Braccini 2 , 10095 Grugliasco , Italy
| | - Davide Schiffer
- Dipartimento di Neuroscienze , Università di Torino , via Cherasco 15 , 10126 Torino , Italy
| | - Chiara Riganti
- Dipartimento di Oncologia , Università di Torino , via Santena 5/bis , 10126 Torino , Italy
| |
Collapse
|
24
|
Moura RP, Martins C, Pinto S, Sousa F, Sarmento B. Blood-brain barrier receptors and transporters: an insight on their function and how to exploit them through nanotechnology. Expert Opin Drug Deliv 2019; 16:271-285. [PMID: 30767695 DOI: 10.1080/17425247.2019.1583205] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION The blood-brain barrier (BBB) is a highly limiting barrier that prevents the brain from contacting with several circulating molecules, including harmful agents. However, certain systemic nutrients and macromolecules are able to cross the BBB and reach the brain parenchyma, involving the interaction with multiple receptors and/or transporters at the BBB surface. Nanotechnology allows the creation of drug vehicles, functionalized with targeting ligands for binding specific BBB receptors and/or transporters, hence triggering the transport through this biobarrier. AREAS COVERED This review focuses the BBB receptors/transporters to be exploited in regard to their overall structure and biologic function, as well as their role in the development of strategies envisaging drug delivery to the brain. Then, the interplay between the targeting of these BBB receptors/transporters and nanotechnology is explored, as they can increase by several-fold the effectiveness of brain-targeted therapies. EXPERT OPINION Nanomedicine may be particularly useful in brain drug delivery, mainly due to the possibility of functionalizing nanoparticles to target specific receptors/transporters. Since the BBB is endowed with numerous receptors and transporters responsible for regulating the proper metabolic activity of the brain, their targeting can be a promising bypass strategy to circumvent the hurdle that the BBB represents for brain drug delivery.
Collapse
Affiliation(s)
- Rui Pedro Moura
- a CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde , Gandra , Portugal
| | - Cláudia Martins
- b I3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto , Porto , Portugal.,c INEB - Instituto de Engenharia Biomédica , Universidade do Porto , Porto , Portugal.,d ICBAS - Instituto de Ciências Biomédicas Abel Salazar , Universidade do Porto , Porto , Portugal
| | - Soraia Pinto
- b I3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto , Porto , Portugal.,c INEB - Instituto de Engenharia Biomédica , Universidade do Porto , Porto , Portugal
| | - Flávia Sousa
- a CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde , Gandra , Portugal.,b I3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto , Porto , Portugal.,c INEB - Instituto de Engenharia Biomédica , Universidade do Porto , Porto , Portugal.,d ICBAS - Instituto de Ciências Biomédicas Abel Salazar , Universidade do Porto , Porto , Portugal
| | - Bruno Sarmento
- a CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde , Gandra , Portugal.,b I3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto , Porto , Portugal.,c INEB - Instituto de Engenharia Biomédica , Universidade do Porto , Porto , Portugal
| |
Collapse
|
25
|
Guevara J, Romo J, Hernandez E, Guevara NV. Identification of Receptor Ligands in Apo B100 Reveals Potential Functional Domains. Protein J 2019; 37:548-571. [PMID: 30259240 DOI: 10.1007/s10930-018-9792-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
LDL, VLDL and other members of the low-density lipoparticles (LLPs) enter cells through a large family of receptors. The actual receptor ligand(s) in apolipoprotein B100, one of the main proteins of LLP, remain(s) unknown. The objective of this study was to identify true receptor ligand(s) in apo B100, a molecule of 4563 residues. Apo B100 contains 33 analogues of Cardin-Weintraub arginine/lysine-based receptor ligand motifs and shares key lysine motifs and sequence similarity with the LDL receptor-associated protein, MESD, and heat shock proteins. Eleven FITC-labeled synthetic peptides of 21-42 residues, with at least one ligand, were tested for binding and internalization using HeLa cells. All peptides bind but display different binding capacities and patterns. Peptides B0013, B0582, B2366, and B2932 mediate endocytosis and appear in distinct sites in the cytoplasm. B0708 and B3181 bind and remain on the cell surface as aggregates/clusters. Peptides B3119 (Site A) and B3347 (Site B), the putative ligands, showed low binding and no cell entry capacity. Apo B100 regions in this study share similarities with related proteins of known function including chaperone proteins and Apo BEC stimulating protein, and not directly related proteins, e.g., the DNA-binding domain of interferon regulatory factors, MSX2-interacting protein, and snake venom Zinc metalloproteinase-disintegrin-like proteins.
Collapse
Affiliation(s)
- Juan Guevara
- Biophysics Research Laboratory, Department of Physics and Astronomy, The University of Texas Rio Grande Valley, One West University Blvd, Brownsville, TX, 78520, USA
| | - Jamie Romo
- Biophysics Research Laboratory, Department of Physics and Astronomy, The University of Texas Rio Grande Valley, One West University Blvd, Brownsville, TX, 78520, USA
| | - Ernesto Hernandez
- Biophysics Research Laboratory, Department of Physics and Astronomy, The University of Texas Rio Grande Valley, One West University Blvd, Brownsville, TX, 78520, USA
| | - Natalia Valentinova Guevara
- Biophysics Research Laboratory, Department of Physics and Astronomy, The University of Texas Rio Grande Valley, One West University Blvd, Brownsville, TX, 78520, USA.
| |
Collapse
|
26
|
Wu Y, Wang S, Shang L, Zhang H, Qin J, Ren Y, Ye T. Effect of borneol as a penetration enhancer on brain targeting of nanoliposomes: facilitate direct delivery to neurons. Nanomedicine (Lond) 2018; 13:2709-2727. [DOI: 10.2217/nnm-2018-0282] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Aim: This study is aimed to evaluate borneol as a penetration enhancer to improve brain target of nanoliposome. Materials & methods: Effects of borneol on pharmacokinetics, targeting efficiency, brain subareas distribution and neuron-targeting level and pathway were studied by fluorescence spectrophotometry and immunofluorescence. Results: Borneol did not influence physicochemical property of doxorubicin hydrochloride nanoliposome (Dox-nanoLips). Co-administration of Dox-nanoLips with borneol elevated brain-target efficiency due to selective distribution increase in the cerebral cortex and hippocampus without difference in contralateral hemisphere. Borneol improved neuronal-targeting level of Dox-nanoLips in the cortex, CA3 and dentate gyrus regions via opening tight junctions of blood–brain barrier and then bypassing astrocyte. Conclusion: Borneol is potential to be a promising penetration enhancer for nanocarrier to target neurons.
Collapse
Affiliation(s)
- Yue Wu
- Department of Pharmaceutics, School of Chinese Medicines, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Shujun Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Lei Shang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Medical College, Shenyang, PR China
| | - Haifeng Zhang
- Department of Pharmacology, School of Life Science & Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Jianxiu Qin
- Department of Pharmaceutics, School of Chinese Medicines, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Yating Ren
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Tiantian Ye
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China
| |
Collapse
|
27
|
Ivask A, Pilkington EH, Blin T, Käkinen A, Vija H, Visnapuu M, Quinn JF, Whittaker MR, Qiao R, Davis TP, Ke PC, Voelcker NH. Uptake and transcytosis of functionalized superparamagnetic iron oxide nanoparticles in an in vitro blood brain barrier model. Biomater Sci 2018; 6:314-323. [PMID: 29239410 DOI: 10.1039/c7bm01012e] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Two major hurdles in nanomedicine are the limited strategies for synthesizing stealth nanoparticles and the poor efficacy of the nanoparticles in translocating across the blood brain barrier (BBB). Here we examined the uptake and transcytosis of iron oxide nanoparticles (IONPs) grafted with biomimetic phosphorylcholine (PC) brushes in an in vitro BBB model system, and compared them with bare, PEG or PC-PEG mixture grafted IONPs. Hyperspectral imaging indicated IONP co-localization with cells. Quantitative analysis with total reflection X-ray fluorescence spectrometry showed that after 24 h, 78% of PC grafted, 68-69% of PEG or PC-PEG grafted, and 30% of bare IONPs were taken up by the BBB. Transcytosis of IONPs was time-dependent and after 24 h, 16-17% of PC or PC-PEG mixture grafted IONPs had passed the BBB model, significantly more than PEG grafted or bare IONPs. These findings point out that grafting of IONPs with PC is a viable strategy for improving the uptake and transcytosis of nanoparticles.
Collapse
Affiliation(s)
- Angela Ivask
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Salaroglio IC, Gazzano E, Kopecka J, Chegaev K, Costamagna C, Fruttero R, Guglielmo S, Riganti C. New Tetrahydroisoquinoline Derivatives Overcome Pgp Activity in Brain-Blood Barrier and Glioblastoma Multiforme in Vitro. Molecules 2018; 23:molecules23061401. [PMID: 29890725 PMCID: PMC6099747 DOI: 10.3390/molecules23061401] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 06/07/2018] [Accepted: 06/07/2018] [Indexed: 11/16/2022] Open
Abstract
P-glycoprotein (Pgp) determines resistance to a broad spectrum of drugs used against glioblastoma multiforme (GB). Indeed, Pgp is highly expressed in GB stem cells and in the brain-blood barrier (BBB), the peculiar endothelium surrounding the brain. Inhibiting Pgp activity in the BBB and GB is still an open challenge. Here, we tested the efficacy of a small library of tetrahydroisoquinoline derivatives with an EC50 for Pgp ≤ 50 nM, in primary human BBB cells and in patient-derived GB samples, from which we isolated differentiated/adherent cells (AC, i.e., Pgp-negative/doxorubicin-sensitive cells) and stem cells (neurospheres, NS, i.e., Pgp-positive/doxorubicin-resistant cells). Three compounds used at 1 nM increased the delivery of doxorubicin, a typical substrate of Pgp, across BBB monolayer, without altering the expression and activity of other transporters. The compounds increased the drug accumulation within NS, restoring doxorubicin-induced necrosis and apoptosis, and reducing cell viability. In co-culture systems, the compounds added to the luminal face of BBB increased the delivery of doxorubicin to NS growing under BBB and rescued the drug’s cytotoxicity. Our work identified new ligands of Pgp active at low nanomolar concentrations. These compounds reduce Pgp activity in BBB and GB and improve in vitro chemotherapy efficacy in this tumor.
Collapse
Affiliation(s)
| | - Elena Gazzano
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Torino Italy.
| | - Joanna Kopecka
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Torino Italy.
| | - Konstantin Chegaev
- Department of Drug Science and Technology, University of Torino, via Pietro Giuria 9, 10125, Torino, Italy.
| | - Costanzo Costamagna
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Torino Italy.
| | - Roberta Fruttero
- Department of Drug Science and Technology, University of Torino, via Pietro Giuria 9, 10125, Torino, Italy.
| | - Stefano Guglielmo
- Department of Drug Science and Technology, University of Torino, via Pietro Giuria 9, 10125, Torino, Italy.
| | - Chiara Riganti
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Torino Italy.
| |
Collapse
|
29
|
Gazzano E, Lazzarato L, Rolando B, Kopecka J, Guglielmo S, Costamagna C, Chegaev K, Riganti C. Mitochondrial Delivery of Phenol Substructure Triggers Mitochondrial Depolarization and Apoptosis of Cancer Cells. Front Pharmacol 2018; 9:580. [PMID: 29915539 PMCID: PMC5994430 DOI: 10.3389/fphar.2018.00580] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 05/15/2018] [Indexed: 12/16/2022] Open
Abstract
Antitumor chemotherapy remains one of the most important challenge of the medicinal chemistry. Emerging research in chemotherapy is focused on exploiting the biochemical differences between cancer cell and normal cell metabolism in order to reduce the side effects and increase antitumor therapy efficacy. The higher mitochondrial transmembrane potential of cancer cells compared to not-transformed cells favors the intra-mitochondrial accumulation of cationic drugs in the former. This feature could be exploited to allow selective delivery of antineoplastic drugs to the cancer cells. In this work we designed and synthetized phenol derivatives joined to the triphenylphosphonium (TPP) cation, a well-known vector for mitochondrial targeting. Two designed phenol TPP-derivatives 1 and 2 show remarkable cytotoxic activity against different cancer cell lines, but were less toxic against normal cells. The differential cytotoxicity relied on the higher mitochondrial biogenesis and oxidative-phosphorylation metabolism of the former. By reducing mitochondrial mass and energetic metabolism, and increasing at the same time the levels of intra-mitochondrial reactive oxygen species, phenol TPP-derivatives 1 and 2 induced mitochondria depolarization and triggered a caspase 9/3-mediated apoptosis, limited to cancer cells. This work provides the rationale to further develop phenol TPP-derivatives targeting mitochondria as new and selective anticancer tools.
Collapse
Affiliation(s)
- Elena Gazzano
- Department of Oncology, University of Turin, Turin, Italy
| | - Loretta Lazzarato
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Barbara Rolando
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Joanna Kopecka
- Department of Oncology, University of Turin, Turin, Italy
| | - Stefano Guglielmo
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | | | - Konstantin Chegaev
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Chiara Riganti
- Department of Oncology, University of Turin, Turin, Italy
| |
Collapse
|
30
|
Chattaragada MS, Riganti C, Sassoe M, Principe M, Santamorena MM, Roux C, Curcio C, Evangelista A, Allavena P, Salvia R, Rusev B, Scarpa A, Cappello P, Novelli F. FAM49B, a novel regulator of mitochondrial function and integrity that suppresses tumor metastasis. Oncogene 2018; 37:697-709. [PMID: 29059164 PMCID: PMC5808099 DOI: 10.1038/onc.2017.358] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 08/07/2017] [Accepted: 08/12/2017] [Indexed: 12/13/2022]
Abstract
Mitochondrial dysregulation plays a central role in cancers and drives reactive oxygen species (ROS)-dependent tumor progression. We investigated the pro-tumoral roles of mitochondrial dynamics and altered intracellular ROS levels in pancreatic ductal adenocarcinoma (PDAC). We identified 'family with sequence similarity 49 member B' (FAM49B) as a mitochondria-localized protein that regulates mitochondrial fission and cancer progression. Silencing FAM49B in PDAC cells resulted in increased fission and mitochondrial ROS generation, which enhanced PDAC cell proliferation and invasion. Notably, FAM49B expression levels in PDAC cells were downregulated by the tumor microenvironment. Overall, the results of this study show that FAM49B acts as a suppressor of cancer cell proliferation and invasion in PDAC by regulating tumor mitochondrial redox reactions and metabolism.
Collapse
Affiliation(s)
- M S Chattaragada
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Center for Experimental Research and Medical Studies, Azienda Universitaria Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy
| | - C Riganti
- Department of Oncology, University of Turin, Turin, Italy
| | - M Sassoe
- Department of Neurosciences, ‘Rita Levi Montalcini’, University of Turin, Turin, Italy
| | - M Principe
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Center for Experimental Research and Medical Studies, Azienda Universitaria Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy
| | - M M Santamorena
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Center for Experimental Research and Medical Studies, Azienda Universitaria Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy
| | - C Roux
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Center for Experimental Research and Medical Studies, Azienda Universitaria Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy
| | - C Curcio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Center for Experimental Research and Medical Studies, Azienda Universitaria Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy
| | - A Evangelista
- Department of Clinical Epidemiology, Azienda Universitaria Ospedaliera Città della Salute e della Scienza and CPO Piemonte, Turin, Italy
| | - P Allavena
- Laboratory of Molecular Gastroenterology, Department of Gastroenterology, Humanitas Clinical and Research Center, Milan, Italy
| | - R Salvia
- Department of Surgery, The Pancreas Institute, University and Hospital Trust of Verona, Verona, Italy
| | - B Rusev
- Department of Pathology and Public Health & ARC-NET Research Center, University and Hospital Trust of Verona, Verona, Italy
| | - A Scarpa
- Department of Pathology and Public Health & ARC-NET Research Center, University and Hospital Trust of Verona, Verona, Italy
| | - P Cappello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Center for Experimental Research and Medical Studies, Azienda Universitaria Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy
- Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - F Novelli
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Center for Experimental Research and Medical Studies, Azienda Universitaria Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy
- Molecular Biotechnology Center, University of Turin, Turin, Italy
| |
Collapse
|
31
|
Kumar V, Kumar V, McGuire T, Coulter DW, Sharp JG, Mahato RI. Challenges and Recent Advances in Medulloblastoma Therapy. Trends Pharmacol Sci 2017; 38:1061-1084. [PMID: 29061299 DOI: 10.1016/j.tips.2017.09.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 09/19/2017] [Accepted: 09/25/2017] [Indexed: 10/18/2022]
Abstract
Medulloblastoma (MB) is the most common childhood brain tumor, which occurs in the posterior fossa. MB tumors are highly heterogeneous and have diverse genetic make-ups, with differential microRNA (miRNA) expression profiles and variable prognoses. MB can be classified into four subgroups, each with different origins, pathogenesis, and potential therapeutic targets. miRNA and small-molecule targeted therapies have emerged as a potential new therapeutic paradigm in MB treatment. However, the development of chemoresistance due to surviving cancer stem cells and dysregulation of miRNAs remains a challenge. Combination therapies using multiple drugs and miRNAs could be effective approaches. In this review we discuss various MB subtypes, barriers, and novel therapeutic options which may be less toxic than current standard treatments.
Collapse
Affiliation(s)
- Vinod Kumar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Virender Kumar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Timothy McGuire
- Department of Pharmacy Practice, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Donald W Coulter
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - John G Sharp
- Department of Genetics, Cell Biology, and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
32
|
Játiva P, Ceña V. Use of nanoparticles for glioblastoma treatment: a new approach. Nanomedicine (Lond) 2017; 12:2533-2554. [DOI: 10.2217/nnm-2017-0223] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GBM) is a very aggressive CNS tumor with poor prognosis. Current treatment lacks efficacy indicating that new therapeutic approaches are needed. One of these new approaches is based on the use of nanoparticles (NPs) to deliver different cargos (antitumoral drugs or genetic materials) to tumoral cells. This review covers the signaling pathways altered in GBM cells to understand the rationale behind choosing new therapeutic targets and recent advances in the use of different NPs to deliver to GBM cells, both in vitro and in vivo, different therapeutic molecules. A special focus is placed on the effect of NPs on orthotopic brain tumors since this animal model represents the optimal model for translational purposes.
Collapse
Affiliation(s)
- Pablo Játiva
- Unidad Asociada Neurodeath, Universidad de Castilla-La Mancha, Albacete, Spain
- CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| | - Valentín Ceña
- Unidad Asociada Neurodeath, Universidad de Castilla-La Mancha, Albacete, Spain
- CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
33
|
Wang CW, Huang CC, Chou PH, Chang YP, Wei S, Guengerich FP, Chou YC, Wang SF, Lai PS, Souček P, Ueng YF. 7-ketocholesterol and 27-hydroxycholesterol decreased doxorubicin sensitivity in breast cancer cells: estrogenic activity and mTOR pathway. Oncotarget 2017; 8:66033-66050. [PMID: 29029490 PMCID: PMC5630390 DOI: 10.18632/oncotarget.19789] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 06/27/2017] [Indexed: 11/30/2022] Open
Abstract
Hypercholesterolemia is one of the risk factors for poor outcome in breast cancer therapy. To elucidate the influence of the main circulating oxysterols, cholesterol oxidation products, on the cell-killing effect of doxorubicin, cells were exposed to oxysterols at a subtoxic concentration. When cells were exposed to oxysterols in fetal bovine serum-supplemented medium, 7-ketocholesterol (7-KC), but not 27-hydroxycholesterol (27-HC), decreased the cytotoxicity of doxorubicin in MCF-7 (high estrogen receptor (ER)α/ERβ ratio) cells and the decreased cytotoxicity was restored by the P-glycoprotein inhibitor verapamil. 7-KC stimulated the efflux function of P-glycoprotein and reduced intracellular doxorubicin accumulation in MCF-7 but not in ERα(-) MDA-MB-231 and the resistant MCF-7/ADR cells. In MCF-7 cells, 7-KC increased the mRNA and protein levels of P-glycoprotein. The 7-KC-suppressed doxorubicin accumulation was restored by the fluvestrant and ERα knockdown. In a yeast reporter assay, the ERα activation by 7-KC was more potent than 27-HC. 7-KC, but not 27-HC, stimulated the expression of an ER target, Trefoil factor 1 in MCF-7 cells. When charcoal-stripped fetal bovine serum was used, both 7-KC and 27-HC induced Trefoil factor 1 expression and reduced doxorubicin accumulation in MCF-7 cells. 7-KC-reduced doxorubicin accumulation could be reversed by inhibitors of phosphatidylinositol 3-kinase, Akt, and mammalian target of rapamycin (mTOR). These findings demonstrate that 7-KC decreases the cytotoxicity of doxorubicin through the up-regulation of P-glycoprotein in an ERα- and mTOR-dependent pathway. The 7-KC- and 27-HC-elicited estrogenic effects are crucial in the P-glycoprotein induction in breast cancer cells.
Collapse
Affiliation(s)
- Chun-Wei Wang
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan, R.O.C.,Institute of Biopharmaceutical Sciences, School of Pharmacy, National Yang-Ming University, Taipei, Taiwan, R.O.C
| | - Chiung-Chiao Huang
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan, R.O.C
| | - Pei-Hsin Chou
- Department of Environmental Engineering, National Chung-Kung University, Tainan, Taiwan, R.O.C
| | - Yu-Ping Chang
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan, R.O.C
| | - Shouzuo Wei
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | - Yueh-Ching Chou
- Department of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan, R.O.C.,Department of Pharmacy, Taipei Veterans General Hospital, Taipei, Taiwan, R.O.C.,Department of Pharmacy, Taipei Medical University, Taipei, Taiwan, R.O.C
| | - Sheng-Fan Wang
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan, R.O.C.,Department of Pharmacy, Taipei Veterans General Hospital, Taipei, Taiwan, R.O.C
| | - Ping-Shan Lai
- Department of Chemistry, College of Science, National Chung-Hsin University, Taichung, Taiwan, R.O.C
| | - Pavel Souček
- Department of Toxicogenomics, National Institute of Public Health, Prague, Czech Republic
| | - Yune-Fang Ueng
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan, R.O.C.,Institute of Biopharmaceutical Sciences, School of Pharmacy, National Yang-Ming University, Taipei, Taiwan, R.O.C.,Department of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan, R.O.C.,Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan, R.O.C
| |
Collapse
|
34
|
Nanoparticles and targeted drug delivery in cancer therapy. Immunol Lett 2017; 190:64-83. [PMID: 28760499 DOI: 10.1016/j.imlet.2017.07.015] [Citation(s) in RCA: 267] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 07/04/2017] [Accepted: 07/26/2017] [Indexed: 12/11/2022]
Abstract
Surgery, chemotherapy, radiotherapy, and hormone therapy are the main common anti-tumor therapeutic approaches. However, the non-specific targeting of cancer cells has made these approaches non-effective in the significant number of patients. Non-specific targeting of malignant cells also makes indispensable the application of the higher doses of drugs to reach the tumor region. Therefore, there are two main barriers in the way to reach the tumor area with maximum efficacy. The first, inhibition of drug delivery to healthy non-cancer cells and the second, the direct conduction of drugs into tumor site. Nanoparticles (NPs) are the new identified tools by which we can deliver drugs into tumor cells with minimum drug leakage into normal cells. Conjugation of NPs with ligands of cancer specific tumor biomarkers is a potent therapeutic approach to treat cancer diseases with the high efficacy. It has been shown that conjugation of nanocarriers with molecules such as antibodies and their variable fragments, peptides, nucleic aptamers, vitamins, and carbohydrates can lead to effective targeted drug delivery to cancer cells and thereby cancer attenuation. In this review, we will discuss on the efficacy of the different targeting approaches used for targeted drug delivery to malignant cells by NPs.
Collapse
|
35
|
Morris ME, Rodriguez-Cruz V, Felmlee MA. SLC and ABC Transporters: Expression, Localization, and Species Differences at the Blood-Brain and the Blood-Cerebrospinal Fluid Barriers. AAPS JOURNAL 2017; 19:1317-1331. [PMID: 28664465 DOI: 10.1208/s12248-017-0110-8] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/05/2017] [Indexed: 12/11/2022]
Abstract
The blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier (BCSFB) separate the brain and cerebrospinal fluid (CSF) from the systemic circulation and represent a barrier to the uptake of both endogenous compounds and xenobiotics into the brain. For compounds whose passive diffusion is limited due to their ionization or hydrophilicity, membrane transporters can facilitate their uptake across the BBB or BCSFB. Members of the solute carrier (SLC) and ATP-binding case (ABC) families are present on these barriers. Differences exist in the localization and expression of transport proteins between the BBB and BCSFB, resulting in functional differences in transport properties. This review focuses on the expression, membrane localization, and different isoforms present at each barrier. Diseases that affect the central nervous system including brain tumors, HIV, Alzheimer's disease, Parkinson's disease, and stroke affect the integrity and expression of transporters at the BBB and BCSFB and will be briefly reviewed.
Collapse
Affiliation(s)
- Marilyn E Morris
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York at Buffalo, Buffalo, New York, 14214-8033, USA.
| | - Vivian Rodriguez-Cruz
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York at Buffalo, Buffalo, New York, 14214-8033, USA
| | - Melanie A Felmlee
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J Long School of Pharmacy and Health Sciences, University of the Pacific, 3601 Pacific Ave, Stockton, California, 95211, USA
| |
Collapse
|
36
|
Beg S, Jain S, Kushwah V, Bhatti GK, Sandhu PS, Katare OP, Singh B. Novel surface-engineered solid lipid nanoparticles of rosuvastatin calcium for low-density lipoprotein-receptor targeting: a Quality by Design-driven perspective. Nanomedicine (Lond) 2017; 12:333-356. [PMID: 28093941 DOI: 10.2217/nnm-2016-0336] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM The present studies describe Quality by Design-oriented development and characterization of surface-engineered solid lipid nanoparticles (SLNs) of rosuvastatin calcium for low density lipoprotein-receptor targeting. MATERIALS & METHODS SLNs were systematically prepared employing Compritol 888 and Tween-80. Surface modification of SLNs was accomplished with Phospholipon 90G and DSPE-mPEG-2000 as the ligands for specific targeting to the low density lipoprotein-receptors. SLNs were evaluated for size, potential, entrapment, drug release performance and gastric stability. Also, the formulations were evaluated for cellular cytotoxicity, uptake and permeability, pharmacokinetic, pharmacodynamic and biochemical studies. RESULTS & CONCLUSION Overall, the studies ratified enhanced biopharmaceutical performance of the surface-engineered SLNs of rosuvastatin as a novel approach for the management of hyperlipidemia-like conditions.
Collapse
Affiliation(s)
- Sarwar Beg
- University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Studies, Panjab University, Chandigarh 160 014, India
| | - Sanyog Jain
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research, Mohali 160 062, Punjab, India
| | - Varun Kushwah
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research, Mohali 160 062, Punjab, India
| | - Gurjit Kaur Bhatti
- UGC-Centre of Excellence in Applications of Nanomaterials, Nanoparticles & Nanocomposites (Biomedical Sciences), Panjab University, Chandigarh 160 014, India
| | - Premjeet Singh Sandhu
- UGC-Centre of Excellence in Applications of Nanomaterials, Nanoparticles & Nanocomposites (Biomedical Sciences), Panjab University, Chandigarh 160 014, India
| | - O P Katare
- University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Studies, Panjab University, Chandigarh 160 014, India
| | - Bhupinder Singh
- University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Studies, Panjab University, Chandigarh 160 014, India.,UGC-Centre of Excellence in Applications of Nanomaterials, Nanoparticles & Nanocomposites (Biomedical Sciences), Panjab University, Chandigarh 160 014, India
| |
Collapse
|
37
|
Aparicio-Blanco J, Martín-Sabroso C, Torres-Suárez AI. In vitro screening of nanomedicines through the blood brain barrier: A critical review. Biomaterials 2016; 103:229-255. [PMID: 27392291 DOI: 10.1016/j.biomaterials.2016.06.051] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 06/14/2016] [Accepted: 06/20/2016] [Indexed: 12/16/2022]
Abstract
The blood-brain barrier accounts for the high attrition rate of the treatments of most brain disorders, which therefore remain one of the greatest health-care challenges of the twenty first century. Against this background of hindrance to brain delivery, nanomedicine takes advantage of the assembly at the nanoscale of available biomaterials to provide a delivery platform with potential to raising brain levels of either imaging or therapeutic agents. Nevertheless, to prevent later failure due to ineffective drug levels at the target site, researchers have been endeavoring to develop a battery of in vitro screening procedures that can predict earlier in the drug discovery process the ability of these cutting-edge drug delivery platforms to cross the blood-brain barrier for biomedical purposes. This review provides an in-depth analysis of the currently available in vitro blood-brain barrier models (both cell-based and non-cell-based) with the focus on their suitability for understanding the biological brain distribution of forthcoming nanomedicines. The relationship between experimental factors and underlying physiological assumptions that would ultimately lead to a more predictive capacity of their in vivo performance, and those methods already assayed for the evaluation of the brain distribution of nanomedicines are comprehensively discussed.
Collapse
Affiliation(s)
- Juan Aparicio-Blanco
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Complutense University, 28040, Madrid, Spain
| | - Cristina Martín-Sabroso
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Complutense University, 28040, Madrid, Spain
| | - Ana-Isabel Torres-Suárez
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Complutense University, 28040, Madrid, Spain; University Institute of Industrial Pharmacy, Complutense University, 28040, Madrid, Spain.
| |
Collapse
|
38
|
On NH, Yathindranath V, Sun Z, Miller DW. Pathways for Drug Delivery to the Central Nervous System. Drug Deliv 2016. [DOI: 10.1002/9781118833322.ch16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
39
|
Mfsd2a-based pharmacological strategies for drug delivery across the blood–brain barrier. Pharmacol Res 2016; 104:124-31. [DOI: 10.1016/j.phrs.2015.12.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 12/20/2015] [Accepted: 12/20/2015] [Indexed: 12/20/2022]
|
40
|
Novel Antitransferrin Receptor Antibodies Improve the Blood-Brain Barrier Crossing Efficacy of Immunoliposomes. J Pharm Sci 2016; 105:276-83. [PMID: 26852859 DOI: 10.1016/j.xphs.2015.11.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 10/23/2015] [Accepted: 10/28/2015] [Indexed: 12/22/2022]
Abstract
Surface functionalization with antitransferrin receptor (TfR) mAbs has been suggested as the strategy to enhance the transfer of nanoparticles (NPs) across the blood-brain barrier (BBB) and to carry nonpermeant drugs from the blood into the brain. However, the efficiency of BBB crossing is currently too poor to be used in vivo. In the present investigation, we compared 6 different murine mAbs specific for different epitopes of the human TfR to identify the best performing one for the functionalization of NPs. For this purpose, we compared the ability of mAbs to cross an in vitro BBB model made of human brain capillary endothelial cells (hCMEC/D3). Liposomes functionalized with the best performing mAb (MYBE/4C1) were uptaken, crossed the BBB in vitro, and facilitated the BBB in vitro passage of doxorubicin, an anticancer drug, 3.9 folds more than liposomes functionalized with a nonspecific IgG, as assessed by confocal microscopy, radiochemical techniques, and fluorescence, and did not modify the cell monolayer structural or functional properties. These results show that MYBE/4C1 antihuman TfR mAb is a powerful resource for the enhancement of BBB crossing of NPs and is therefore potentially useful in the treatment of neurologic diseases and disorders including brain carcinomas.
Collapse
|
41
|
Tagami T, Taki M, Ozeki T. Nanoparticulate Drug Delivery Systems to Overcome the Blood–Brain Barrier. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2016. [DOI: 10.1007/978-1-4939-3121-7_17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
42
|
Gaudin A, Andrieux K, Couvreur P. Nanomedicines and stroke: Toward translational research. J Drug Deliv Sci Technol 2015. [DOI: 10.1016/j.jddst.2015.07.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
43
|
Ung N, Yang I. Nanotechnology to augment immunotherapy for the treatment of glioblastoma multiforme. J Neurooncol 2015; 123:473-81. [DOI: 10.1007/s11060-015-1814-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 05/20/2015] [Indexed: 12/30/2022]
|
44
|
Nanoparticle-mediated growth factor delivery systems: A new way to treat Alzheimer's disease. J Control Release 2015; 206:187-205. [DOI: 10.1016/j.jconrel.2015.03.024] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/19/2015] [Accepted: 03/20/2015] [Indexed: 01/03/2023]
|
45
|
Roundabout 4 regulates blood-tumor barrier permeability through the modulation of ZO-1, Occludin, and Claudin-5 expression. J Neuropathol Exp Neurol 2015; 74:25-37. [PMID: 25470344 DOI: 10.1097/nen.0000000000000146] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The blood-tumor barrier (BTB) restricts the delivery of chemotherapeutic drug molecules to tumor tissues. We found that the endothelial cell (EC) receptor molecule Roundabout 4 (Robo4) is endogenously expressed in human brain microvascular ECs and that it is upregulated in a BTB model of glioma cocultured ECs. Knockdown of Robo4 in this BTB model increased permeability; short hairpin RNA targeting Robo4 (shRobo4) led to decreased transendothelial electric resistance values, increased BTB permeability, and downregulated expression of the EC tight junction proteins ZO-1, occludin, and claudin-5. Roundabout 4 influenced BTB permeability via binding with its ligand, Slit2. Short hairpin RNA targeting Robo4 also increased matrix metalloproteinase-9 (MMP-9) activity and expression in glioma cocultured ECs; pretreatment with the MMP inhibitor GM6001 partially blocked the effects of shRobo4 on the transendothelial electric resistance values and ZO-1 and occludin expression. Short hairpin RNA targeting Robo4 also upregulated the phosphorylation of Src and Erk1/2; the Src inhibitor PP2 and the Erk1/2 inhibitor PD98059 blocked shRobo4-mediated alteration in ZO-1 and occludin expression. Together, our results indicate that knockdown of Robo4 increased BTB permeability by reducing EC tight junction protein expression, and that the Src-Erk1/2-MMP-9 signal pathways are involved in this process. Thus, Robo4 may represent a useful future therapeutic target for enhancing BTB permeability.
Collapse
|
46
|
Chen J, Xu ZY, Wang F. Association between DNA methylation and multidrug resistance in human glioma SHG-44 cells. Mol Med Rep 2014; 11:43-52. [PMID: 25333456 PMCID: PMC4237088 DOI: 10.3892/mmr.2014.2690] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 06/11/2014] [Indexed: 01/03/2023] Open
Abstract
The aim of the present study was to evaluate the association between DNA methylation and multidrug resistance (MDR) in glioma and identify novel effectors responsible for MDR in human gliomas. An MDR glioma cell line, SGH-44/ADM, was developed using adriamycin (ADM) impulse treatment. Cryopreservation, recovery and withdrawal were performed to evaluate the stability of SGH-44/ADM cells. The adherence rate and cellular morphology were observed by microscopy, and the cell growth curve and doubling time were determined. DNA methylation was analyzed using a methylated DNA immunoprecipitation microarray chip (MeDIP-Chip). The cell cycle, Rh123 ingestion and exudation, and SGH-44/ADM apoptosis were analyzed by flow cytometry. SGH-44/ADM cells showed little difference as compared with parental cells, except that SGH-44/ADM cells were bigger in size with a wizened nucleus. Compared to SGH-44 cells, a larger proportion of SGH-44/ADM cells remained in G1 and S phase, as measured by flow cytometry. The MDR of SGH-44/ADM was associated with the upregulation of multi-drug resistance 1, prostaglandin-endoperoxide synthase 2 (COX-2); protein kinase C α (PKCα); however, the expression of these genes was not associated with DNA methylation. In the MeDIP-Chip analysis, 74 functions were markedly enhanced, and seven significant pathways were observed. Genes including SNAP47, ARRB2, PARD6B, TGFB1, VPS4B and CBLB were identified by gene ontology analysis. The predominant molecular mechanism of MDR in SGH-44/ADM cells was identified as exocytosis and efflux. The expression of COX-2, PKCα and P-glycoprotein (Pgp) was not found to be associated with DNA methylation. Genes including SNAP47, VAMP4 and VAMP3 may serve as the downstream effectors of Pgp, COX-2 or PKCα; however, further experiments are required to verify these observations.
Collapse
Affiliation(s)
- Jin Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Zhong-Ye Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Feng Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
47
|
Pinzón-Daza ML, Salaroglio IC, Kopecka J, Garzòn R, Couraud PO, Ghigo D, Riganti C. The cross-talk between canonical and non-canonical Wnt-dependent pathways regulates P-glycoprotein expression in human blood-brain barrier cells. J Cereb Blood Flow Metab 2014; 34:1258-69. [PMID: 24896565 PMCID: PMC4126086 DOI: 10.1038/jcbfm.2014.100] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 03/19/2014] [Accepted: 04/13/2014] [Indexed: 12/11/2022]
Abstract
In this work, we investigate if and how transducers of the 'canonical' Wnt pathway, i.e., Wnt/glycogen synthase kinase 3 (GSK3)/β-catenin, and transducers of the 'non-canonical' Wnt pathway, i.e., Wnt/RhoA/RhoA kinase (RhoAK), cooperate to control the expression of P-glycoprotein (Pgp) in blood-brain barrier (BBB) cells. By analyzing human primary brain microvascular endothelial cells constitutively activated for RhoA, silenced for RhoA or treated with the RhoAK inhibitor Y27632, we found that RhoAK phosphorylated and activated the protein tyrosine phosphatase 1B (PTP1B), which dephosphorylated tyrosine 216 of GSK3, decreasing the GSK3-mediated inhibition of β-catenin. By contrast, the inhibition of RhoA/RhoAK axis prevented the activation of PTP1B, enhanced the GSK3-induced phosphorylation and ubiquitination of β-catenin, and reduced the β-catenin-driven transcription of Pgp. The RhoAK inhibition increased the delivery of Pgp substrates like doxorubicin across the BBB and improved the doxorubicin efficacy against glioblastoma cells co-cultured under a BBB monolayer. Our data demonstrate that in human BBB cells the expression of Pgp is controlled by a cross-talk between canonical and non-canonical Wnt pathways. The disruption of this cross-talk, e.g., by inhibiting RhoAK, downregulates Pgp and increases the delivery of Pgp substrates across the BBB.
Collapse
Affiliation(s)
- Martha L Pinzón-Daza
- 1] Department of Oncology, School of Medicine, University of Turin, Turin, Italy [2] Unidad de Bioquímica, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia
| | - Iris C Salaroglio
- Department of Oncology, School of Medicine, University of Turin, Turin, Italy
| | - Joanna Kopecka
- Department of Oncology, School of Medicine, University of Turin, Turin, Italy
| | - Ruth Garzòn
- Unidad de Bioquímica, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia
| | - Pierre-Olivier Couraud
- Institut Cochin, Centre National de la Recherche Scientifique UMR 8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U567, Université René Descartes, Paris, France
| | - Dario Ghigo
- 1] Department of Oncology, School of Medicine, University of Turin, Turin, Italy [2] Center for Experimental Research and Medical Studies, University of Turin, Turin, Italy
| | - Chiara Riganti
- 1] Department of Oncology, School of Medicine, University of Turin, Turin, Italy [2] Center for Experimental Research and Medical Studies, University of Turin, Turin, Italy
| |
Collapse
|
48
|
Pedrini I, Gazzano E, Chegaev K, Rolando B, Marengo A, Kopecka J, Fruttero R, Ghigo D, Arpicco S, Riganti C. Liposomal nitrooxy-doxorubicin: one step over caelyx in drug-resistant human cancer cells. Mol Pharm 2014; 11:3068-79. [PMID: 25057799 DOI: 10.1021/mp500257s] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In this work we prepared and characterized two liposomal formulations of a semisynthetic nitric oxide (NO)-releasing doxorubicin (Dox), called nitrooxy-Dox (NitDox), which we previously demonstrated to be cytotoxic in Dox-resistant human colon cancer cells. Liposomes with 38.2% (Lip A) and 19.1% (Lip B) cholesterol were synthesized: both formulations had similar size and zeta potential values and caused the same intracellular distribution of free NitDox, but Lip B accumulated and released NitDox more efficiently. In Dox-resistant human colon cancer cells, Lip A and Lip B exhibited a more favorable kinetics of drug uptake and NO release, and a stronger cytotoxicity than Dox and free NitDox. While Caelyx, one of the liposomal Dox formulations approved for breast and ovary tumors treatment, was ineffective in Dox-resistant breast/ovary cancer cells, Lip B, and to a lesser extent Lip A, still exerted a significant cytotoxicity in these cells. This event was accompanied in parallel by a higher release of NO, which caused nitration of P-glycoprotein (Pgp) and multidrug resistance related protein 1 (MRP1), two transporters involved in Dox efflux, and impaired their pump activity. By doing so, the efflux kinetics of Dox after treatment with Lip B was markedly slowed down and the intracellular accumulation of Dox was increased in breast and ovary drug-resistant cells. We propose these liposomal formulations of NitDox as new tools with a specific indication for tumors overexpressing Pgp and MRP1.
Collapse
Affiliation(s)
- Isabella Pedrini
- Department of Drug Science and Technology, University of Torino , via Pietro Giuria 9, 10125 Torino, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Battaglia L, Gallarate M, Peira E, Chirio D, Muntoni E, Biasibetti E, Capucchio MT, Valazza A, Panciani PP, Lanotte M, Schiffer D, Annovazzi L, Caldera V, Mellai M, Riganti C. Solid lipid nanoparticles for potential doxorubicin delivery in glioblastoma treatment: preliminary in vitro studies. J Pharm Sci 2014; 103:2157-2165. [PMID: 24824141 DOI: 10.1002/jps.24002] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 04/08/2014] [Accepted: 04/14/2014] [Indexed: 01/23/2023]
Abstract
The major obstacle to glioblastoma pharmacological therapy is the overcoming of the blood-brain barrier (BBB). In literature, several strategies have been proposed to overcome the BBB: in this experimental work, solid lipid nanoparticles (SLN), prepared according to fatty acid coacervation technique, are proposed as the vehicle for doxorubicin (Dox), to enhance its permeation through an artificial model of BBB. The in vitro cytotoxicity of Dox-loaded SLN has been measured on three different commercial and patient-derived glioma cell lines. Dox was entrapped within SLN thanks to hydrophobic ion pairing with negatively charged surfactants, used as counterions. Results indicate that Dox entrapped in SLN maintains its cytotoxic activity toward glioma cell lines; moreover, its permeation through hCMEC/D3 cell monolayer, assumed as a model of the BBB, was increased when the drug was entrapped in SLN. In conclusion, SLN proved to be a promising vehicle for the delivery of Dox to the brain in glioblastoma treatment.
Collapse
Affiliation(s)
- Luigi Battaglia
- Università degli Studi di Torino, Dipartimento di Scienza e Tecnologia del Farmaco, Torino, Italy.
| | - Marina Gallarate
- Università degli Studi di Torino, Dipartimento di Scienza e Tecnologia del Farmaco, Torino, Italy
| | - Elena Peira
- Università degli Studi di Torino, Dipartimento di Scienza e Tecnologia del Farmaco, Torino, Italy
| | - Daniela Chirio
- Università degli Studi di Torino, Dipartimento di Scienza e Tecnologia del Farmaco, Torino, Italy
| | - Elisabetta Muntoni
- Università degli Studi di Torino, Dipartimento di Scienza e Tecnologia del Farmaco, Torino, Italy
| | - Elena Biasibetti
- Università degli Studi di Torino, Dipartimento di Scienze Veterinarie, Grugliasco, Italy
| | - Maria Teresa Capucchio
- Università degli Studi di Torino, Dipartimento di Scienze Veterinarie, Grugliasco, Italy
| | - Alberto Valazza
- Università degli Studi di Torino, Dipartimento di Scienze Veterinarie, Grugliasco, Italy
| | - Pier Paolo Panciani
- Università degli Studi di Torino, Dipartimento di Neuroscienze, Torino, Italy
| | - Michele Lanotte
- Università degli Studi di Torino, Dipartimento di Neuroscienze, Torino, Italy
| | - Davide Schiffer
- Centro di NeuroBioOncologia, Policlinico di Monza, Vercelli, Italy
| | - Laura Annovazzi
- Centro di NeuroBioOncologia, Policlinico di Monza, Vercelli, Italy
| | | | - Marta Mellai
- Centro di NeuroBioOncologia, Policlinico di Monza, Vercelli, Italy
| | - Chiara Riganti
- Università degli Studi di Torino, Dipartimento di Oncologia, Orbassano, Italy
| |
Collapse
|
50
|
Liu L, Zuo LF, Guo JW. ABCG2 gene amplification and expression in esophageal cancer cells with acquired adriamycin resistance. Mol Med Rep 2014; 9:1299-304. [PMID: 24535197 DOI: 10.3892/mmr.2014.1949] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 01/29/2014] [Indexed: 11/06/2022] Open
Abstract
Resistance to chemotherapeutic agents is the main reason for treatment failure in patients with cancer. The primary mechanism of multidrug resistance (MDR) is the overexpression of drug efflux transporters, including ATP‑binding cassette transporter G2 (ABCG2). To the best of our knowledge, the MDR mechanisms of esophageal cancer have not been described. An adriamycin (ADM)-resistant subline, Eca109/ADM, was generated from the Eca109 esophageal cancer cell line by a stepwise selection in ADM from 0.002 to 0.02 ng/µl. The resulting subline, designated Eca109/ADM, revealed a 3.29-fold resistance against ADM compared with the Eca109 cell line. The ABCG2 gene expression in the Eca109/ADM cells was increased compared with that of the Eca109 cells. The cellular properties of the Eca109/ADM cells were detected by reverse transcription polymerase chain reaction (RT-PCR), flow cytometry and western blotting. The ABCG2 expression levels were detected by RT-PCR and flow cytometry, and the drug efflux effect was detected by flow cytometry. The present study detected the correlation between ABCG2 and the multidrug resistance of esophageal cancer. ABCG2 gene expression and the drug efflux effect of the Eca109/ADM cells were increased compared with those of the Eca109 cells. Collectively, the results of this study indicated that the overexpression of ABCG2 in the Eca109/ADM cells resulted in drug efflux, which may be responsible for the development of esophageal cancer MDR.
Collapse
Affiliation(s)
- Liang Liu
- Department of Flow Cytometry Analysis, Tumor Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Lian Fu Zuo
- Department of Flow Cytometry Analysis, Tumor Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Jian Wen Guo
- Department of Flow Cytometry Analysis, Tumor Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|