1
|
Verma R, Zeyaullah M, Singh V, Saxena PS, Koch B, Kumar M. Chitosan-Functionalized Fluorescent Calcium Carbonate Nanoparticle Loaded with Methotrexate: Future Theranostics for Triple Negative Breast Cancer. ACS Biomater Sci Eng 2025. [PMID: 39883480 DOI: 10.1021/acsbiomaterials.4c02390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Herein, fluorescent calcium carbonate nanoclusters encapsulated with methotrexate (Mtx) and surface functionalized with chitosan (25 nm) (@Calmat) have been developed for the imaging and treatment of triple-negative breast cancer (TNBC). These biocompatible, pH-sensitive nanoparticles demonstrate significant potential for targeted therapy and diagnostic applications. The efficacy of nanoparticles (NPs) was evaluated in MDA-MB-231 TNBC cell lines. The enhanced permeability and retention effect facilitated the accumulation of NPs, in tumor-bearing rats, as confirmed by in vivo fluorescence imaging. Treatment with @Calmat resulted in a marked reduction in pro-inflammatory cytokines, with levels of IL-6 (1225 ± 67 pg/mL), IL-1β (379 ± 69 pg/mL), and TNF-α (14.1 ± 2 pg/mL), in contrast to the diseased control group (IL-6: 2223 ± 99; IL-1β: 1632 ± 90; TNF-α: 40 ± 3 pg/mL). A similar trend was observed for liver and kidney function biomarkers. Mechanistic studies revealed that @Calmat treatment activates the Bax/Bcl-2 signaling pathway, leading to cell cycle arrest in the G1 phase and subsequent late-phase apoptosis. As a result, the tumor inhibition rate reached 88%, with 80% of treated rats surviving beyond 100 days. These findings highlight the strong potential of @Calmat as a dual-function theranostic agent for the management of TNBC.
Collapse
Affiliation(s)
- Rinki Verma
- School of Biomedical Engineering, IIT (BHU), Varanasi 221005, India
| | - Md Zeyaullah
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Virendra Singh
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Preeti Suman Saxena
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Biplob Koch
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Manoj Kumar
- Nano 2 Micro Material Design Lab, Department of Chemical Engineering and Technology, IIT (BHU), Varanasi 221005, India
| |
Collapse
|
2
|
Bazzazan MA, Fattollazadeh P, Keshavarz Shahbaz S, Rezaei N. Polymeric nanoparticles as a promising platform for treating triple-negative breast cancer: Current status and future perspectives. Int J Pharm 2024; 664:124639. [PMID: 39187034 DOI: 10.1016/j.ijpharm.2024.124639] [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: 05/31/2024] [Revised: 08/04/2024] [Accepted: 08/23/2024] [Indexed: 08/28/2024]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive subtype of breast cancer that lacks expression of estrogen, progesterone, and HER2 receptor targets for therapy. Polymeric nanoparticles help address the challenges in treating TNBC by enabling tailored and targeted drug delivery. Biocompatible polymeric nanoparticles leverage enhanced tumor permeability for site-specific accumulation and ligand-mediated active targeting to boost specificity. Controlled, sustained intratumorally release of encapsulated chemotherapies, such as paclitaxel and curcumin, improves antitumor efficacy as demonstrated through preclinical TNBC models. However, the practical application of these nanomedicines still has room for improvement. Advancing personalized nanoparticle platforms that align treatments to TNBC's expanding molecular subtypes shows promise. Expanding the polymer range through novel copolymers or drug conjugates may improve tumor penetration, stability, and drug encapsulation. Incorporating gene therapies, imaging agents, or triggering stimuli responsiveness into polymeric nanoparticles can also overcome innate and acquired drug resistance in TNBC while monitoring outcomes. This article reviews the different types of nanoparticles used to treat TNBC and the different mechanisms of nanoparticles that can deliver drugs to tumor cells. Collaboration across different disciplines aimed at developing combination therapies, immuno-oncology, tumor-targeting ligands, and translating preclinical safety/efficacy via scalable manufacturing practices is essential. Well-designed polymeric nanoparticles offer immense potential for patient-centric TNBC treatment, but continued optimization across bench to bedside efforts is critical for clinical realization and transforming patient outcomes.
Collapse
Affiliation(s)
- Mohammad Amin Bazzazan
- Student Research Committee, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical Science, Qazvin, Iran
| | - Pouriya Fattollazadeh
- Student Research Committee, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical Science, Qazvin, Iran
| | - Sanaz Keshavarz Shahbaz
- USERN Office, Qazvin University of Medical Science, Qazvin, Iran; Cellular and Molecular Research Center, Research Institute for Prevention of Noncommunicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran.
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran; Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
3
|
Krishnan J, Poomalai P, Ravichandran A, Reddy A, Sureshkumar R. A Concise Review on Effect of PEGylation on the Properties of Lipid-Based Nanoparticles. Assay Drug Dev Technol 2024; 22:246-264. [PMID: 38828531 DOI: 10.1089/adt.2024.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024] Open
Abstract
Nanoparticle-based drug delivery systems have emerged as promising platforms for enhancing therapeutic efficacy while minimizing off-target effects. Among various strategies employed to optimize these systems, polyethylene glycol (PEG) modification, known as PEGylation-the covalent attachment of PEG to nanoparticles, has gained considerable attention for its ability to impart stealth properties to nanoparticles while also extending circulation time and improving biocompatibility. PEGylation extends to different drug delivery systems, in specific, nanoparticles for targeting cancer cells, where the concentration of drug in the cancer cells is improved by virtue of PEGylation. The primary challenge linked to PEGylation lies in its confirmation. Numerous research findings provide comprehensive insights into selecting PEG for various PEGylation methods. In this review, we have endeavored to consolidate the outcomes concerning the choice of PEG and diverse PEGylation techniques.
Collapse
Affiliation(s)
- Janesha Krishnan
- Department of Pharmaceutics, Center for Nano Engineering Science & Technology (C-NEST), JSS Academy of Higher Education and Research, JSS College of Pharmacy, Ooty, India
| | - Praveena Poomalai
- Department of Pharmaceutics, Center for Nano Engineering Science & Technology (C-NEST), JSS Academy of Higher Education and Research, JSS College of Pharmacy, Ooty, India
| | - Ashwin Ravichandran
- Department of Pharmaceutics, Center for Nano Engineering Science & Technology (C-NEST), JSS Academy of Higher Education and Research, JSS College of Pharmacy, Ooty, India
| | - Aishwarya Reddy
- Department of Pharmaceutics, Center for Nano Engineering Science & Technology (C-NEST), JSS Academy of Higher Education and Research, JSS College of Pharmacy, Ooty, India
| | - Raman Sureshkumar
- Department of Pharmaceutics, Center for Nano Engineering Science & Technology (C-NEST), JSS Academy of Higher Education and Research, JSS College of Pharmacy, Ooty, India
| |
Collapse
|
4
|
Kumari L, Mishra L, Patel P, Sharma N, Gupta GD, Kurmi BD. Emerging targeted therapeutic strategies for the treatment of triple-negative breast cancer. J Drug Target 2023; 31:889-907. [PMID: 37539789 DOI: 10.1080/1061186x.2023.2245579] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 07/30/2023] [Indexed: 08/05/2023]
Abstract
Triple-negative breast cancer (TNBC), a subtype of breast cancer that lacks expression of oestrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER-2), has clinical features including a high degree of invasiveness, an elevated risk of metastasis, tendency to relapse, and poor prognosis. It constitutes around 10-15% of all breast cancer, and having heredity of BRCA1 mutated breast cancer could be a reason for the occurrence of TNBC in women. Overexpression of cellular and molecular targets, i.e. CD44 receptor, EGFR receptor, Folate receptor, Transferrin receptor, VEGF receptor, and Androgen receptor, have emerged as promising targets for treating TNBC. Signalling pathways such as Notch signalling and PI3K/AKT/mTOR also play a significant role in carrying out and managing crucial pro-survival and pro-growth cellular processes that can be utilised for targeted therapy against triple-negative breast cancer. This review sheds light on various targeting strategies, including cellular and molecular targets, signalling pathways, poly (ADP-ribose) polymerase inhibitors, antibody-drug conjugates, and immune checkpoint inhibitors PARP, immunotherapy, ADCs have all found a place in the current TNBC therapeutic paradigm. The role of photothermal therapy (PTT) and photodynamic therapy (PDT) has also been explored briefly.
Collapse
Affiliation(s)
- Lakshmi Kumari
- Department of Pharmaceutics, ISF College Pharmacy, Moga, Punjab, India
| | - Lopamudra Mishra
- Department of Pharmaceutics, ISF College Pharmacy, Moga, Punjab, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College Pharmacy, Moga, Punjab, India
| | - Nitin Sharma
- Department of Pharmaceutics, ISF College Pharmacy, Moga, Punjab, India
| | | | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College Pharmacy, Moga, Punjab, India
| |
Collapse
|
5
|
Santos JF, Azevedo R, Prudêncio M, Marques F, Le Gal Y, Lorcy D, Fernandes C. Block Copolymer Micelles Encapsulating Au(III) Bis(Dithiolene) Complexes as Promising Nanostructures with Antiplasmodial Activity. Pharmaceutics 2023; 15:pharmaceutics15031030. [PMID: 36986890 PMCID: PMC10058554 DOI: 10.3390/pharmaceutics15031030] [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: 01/20/2023] [Revised: 03/15/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Block copolymer micelles (BCMs) can be used to improve the solubility of lipophilic drugs and increase their circulation half-life. Hence, BCMs assembled from MePEG-b-PCL were evaluated as drug delivery systems of gold(III) bis(dithiolene) complexes (herein AuS and AuSe) to be employed as antiplasmodial drugs. These complexes exhibited remarkable antiplasmodial activity against liver stages of the Plasmodiumberghei parasite, and low toxicity in a model of zebrafish embryos. To improve the complexes' solubility, BCMs were loaded with AuS, AuSe, and the reference drug primaquine (PQ). PQ-BCMs (Dh = 50.9 ± 2.8 nm), AuSe-BCMs (Dh = 87.1 ± 9.7 nm), and AuS-BCMs (Dh = 72.8 ± 3.1 nm) were obtained with a loading efficiency of 82.5%, 55.5%, and 77.4%, respectively. HPLC analysis and UV-Vis spectrophotometry showed that the compounds did not suffer degradation after encapsulation in BCMs. In vitro release studies suggest that AuS/AuSe-BCMs present a more controlled release compared with PQ-loaded BCMs. The antiplasmodial hepatic activity of the drugs was assessed in vitro and results indicate that both complexes present higher inhibitory activity than PQ, although encapsulated AuS and AuSe presented lower activity than their non-encapsulated counterparts. Nevertheless, these results suggest that the use of BCMs as delivery vehicles for lipophilic metallodrugs, particularly AuS and AuSe, could enable the controlled release of complexes and improve their biocompatibility, constituting a promising alternative to conventional antimalarial treatments.
Collapse
Affiliation(s)
- Joana F Santos
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, km 139.7, 2695-066 Bobadela, Portugal
| | - Raquel Azevedo
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Miguel Prudêncio
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Fernanda Marques
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, km 139.7, 2695-066 Bobadela, Portugal
- Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, km 139.7, 2695-066 Bobadela, Portugal
| | - Yann Le Gal
- Institut des Sciences Chimiques de Rennes-UMR 6226, Université de Rennes, CNRS, ISCR, F-35000 Rennes, France
| | - Dominique Lorcy
- Institut des Sciences Chimiques de Rennes-UMR 6226, Université de Rennes, CNRS, ISCR, F-35000 Rennes, France
| | - Célia Fernandes
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, km 139.7, 2695-066 Bobadela, Portugal
- Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, km 139.7, 2695-066 Bobadela, Portugal
| |
Collapse
|
6
|
Guha L, Bhat IA, Bashir A, Rahman JU, Pottoo FH. Nanotechnological Approaches for the Treatment of Triple-Negative Breast Cancer: A Comprehensive Review. Curr Drug Metab 2022; 23:781-799. [PMID: 35676850 DOI: 10.2174/1389200223666220608144551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/01/2022] [Accepted: 03/10/2022] [Indexed: 01/05/2023]
Abstract
Breast cancer is the most prevalent cancer in women around the world, having a sudden spread nowadays because of the poor sedentary lifestyle of people. Comprising several subtypes, one of the most dangerous and aggressive ones is triple-negative breast cancer or TNBC. Even though conventional surgical approaches like single and double mastectomy and preventive chemotherapeutic approaches are available, they are not selective to cancer cells and are only for symptomatic treatment. A new branch called nanotechnology has emerged in the last few decades that offers various novel characteristics, such as size in nanometric scale, enhanced adherence to multiple targeting moieties, active and passive targeting, controlled release, and site-specific targeting. Among various nanotherapeutic approaches like dendrimers, lipid-structured nanocarriers, carbon nanotubes, etc., nanoparticle targeted therapeutics can be termed the best among all for their specific cytotoxicity to cancer cells and increased bioavailability to a target site. This review focuses on the types and molecular pathways involving TNBC, existing treatment strategies, various nanotechnological approaches like exosomes, carbon nanotubes, dendrimers, lipid, and carbon-based nanocarriers, and especially various nanoparticles (NPs) like polymeric, photodynamic, peptide conjugated, antibody-conjugated, metallic, inorganic, natural product capped, and CRISPR based nanoparticles already approved for treatment or are under clinical and pre-clinical trials for TNBC.
Collapse
Affiliation(s)
- Lahanya Guha
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Mohali, S.A.S Nagar, Punjab 160062, India
| | - Ishfaq Ahmad Bhat
- Northern Railway Hospital, Sri Mata Vaishno Devi, Katra, Reasi 182320, India
| | - Aasiya Bashir
- Department of Pharmaceutical Sciences, Faculty of Applied Sciences and Technology, University of Kashmir, Hazratbal, Srinagar-190006, J&K, India
| | - Jawad Ur Rahman
- Department of Microbiology, College of Medicine, Imam Abdulrahman Bin Faisal University, P.O.BOX 1982, Dammam 31441, Saudi Arabia
| | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O.BOX 1982, Dammam 31441, Saudi Arabia
| |
Collapse
|
7
|
Moret F, Conte C, Esposito D, Dal Poggetto G, Avitabile C, Ungaro F, Tiso N, Romanelli A, Laurienzo P, Reddi E, Quaglia F. Biodegradable nanoparticles combining cancer cell targeting and anti-angiogenic activity for synergistic chemotherapy in epithelial cancer. Drug Deliv Transl Res 2022; 12:2488-2500. [PMID: 34973132 PMCID: PMC9458690 DOI: 10.1007/s13346-021-01090-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2021] [Indexed: 12/12/2022]
Abstract
A biodegradable engineered nanoplatform combining anti-angiogenic activity and targeting of cancer cells to improve the anticancer activity of docetaxel (DTX) is here proposed. Indeed, we have developed biodegradable nanoparticles (NPs) of poly(ethylene glycol)-poly(ε-caprolactone), exposing on the surface both folate motifs (Fol) for recognition in cells overexpressing Folate receptor-α (FRα) and the anti-angiogenic hexapeptide aFLT1. NPs showed a size around 100 nm, the exposure of 60% of Fol moieties on the surface, and the ability to entrap DTX and sustain its release with time. NPs were stable in simulated biological fluids and slightly interacted with Fetal Bovine serum, especially in the formulation decorated with Fol and aFLT1. The presence of Fol on NPs did not impair the anti-angiogenic activity of aFLT1, as assessed by in vitro tube formation assay in HUVEC endothelial cells. In both 2D and 3D KB cell cultures in vitro, the cytotoxicity of DTX loaded in NPs was not significantly affected by Fol/aFLT1 double decoration compared to free DTX. Remarkably, NPs distributed differently in 3D multicellular spheroids of FRα-positive KB cancer cells depending on the type of ligand displayed on the surface. In particular, NPs unmodified on the surface were randomly distributed in the spheroid, whereas the presence of Fol promoted the accumulation in the outer rims of the spheroid. Finally, NPs with Fol and aFLT1 gave a uniform distribution throughout the spheroid structure. When tested in zebrafish embryos xenografted with KB cells, NPs displaying Fol/aFLT1 reduced DTX systemic toxicity and inhibited the growth of the tumor mass and associated vasculature synergistically. Overall, nanotechnology offers excellent ground for combining therapeutic concepts in cancer, paving the way to novel multifunctional nanopharmaceuticals decorated with bioactive elements that can significantly improve therapeutic outcomes.
Collapse
Affiliation(s)
- Francesca Moret
- Department of Biology, University of Padova, Padova, 35121, Italy
| | - Claudia Conte
- Department of Pharmacy, University of Napoli Federico II, Napoli, 80131, Italy
| | - Diletta Esposito
- Department of Pharmacy, University of Napoli Federico II, Napoli, 80131, Italy
| | | | | | - Francesca Ungaro
- Department of Pharmacy, University of Napoli Federico II, Napoli, 80131, Italy
| | - Natascia Tiso
- Department of Biology, University of Padova, Padova, 35121, Italy
| | - Alessandra Romanelli
- Department of Pharmaceutical Sciences, University of Milan, Milano, 20133, Italy
| | - Paola Laurienzo
- Institute for Polymers, Composites and Biomaterials, CNR, Pozzuoli, 80078, Italy
| | - Elena Reddi
- Department of Biology, University of Padova, Padova, 35121, Italy.
| | - Fabiana Quaglia
- Department of Pharmacy, University of Napoli Federico II, Napoli, 80131, Italy. .,Institute for Polymers, Composites and Biomaterials, CNR, Pozzuoli, 80078, Italy.
| |
Collapse
|
8
|
Conte C, Monteiro PF, Gurnani P, Stolnik S, Ungaro F, Quaglia F, Clarke P, Grabowska A, Kavallaris M, Alexander C. Multi-component bioresponsive nanoparticles for synchronous delivery of docetaxel and TUBB3 siRNA to lung cancer cells. NANOSCALE 2021; 13:11414-11426. [PMID: 34160534 DOI: 10.1039/d1nr02179f] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Bioresponsive nanoparticles (NPs) are of interest for anticancer nanomedicines, owing to the possibility to 'design in' selective modulation of drug release at target sites. Here we describe the double emulsion formulation of redox-responsive NPs based on modified polyethylene glycol (PEG)-co-poly(lactic-co-glycolic acid) (PLGA) block copolymers and oligo (β-aminoesters) (OBAE), both of which contained disulfide linkages, for the co-delivery of a cytotoxic small molecule drug and a nucleic acid. In particular, we focused our attention on docetaxel (DTX) and a siRNA against TUBB3, a gene that encodes for βIII-tubulin, in order to have a synergistic effect in the treatment of lung cancer. Spherical NPs of around 150 nm with negative zeta potential and high loading efficiencies of both drugs were obtained. Stability and release studies showed "on demand" drug release under reducing conditions. Unloaded NPs containing PEG-disulfide-PLGA and OBAE were well-tolerated by lung cancer cells, thus masking the intrinsic cytotoxicity of OBAE, while for intracellular siRNA delivery, redox responsive NPs demonstrated a higher cell internalization with a preferential cytoplasmic accumulation of siRNA, with a subsequent fast gene-silencing efficiency. The viability of cells treated with combined DTX/TUBB3-siRNA NPs significantly decreased as compared to NPs loaded only with DTX, thus showing an efficient combined anticancer effect, due to a substantial reduction of β-tubulin expression. Finally, in an in vivo feasibility study employing an orthotopic lung cancer model, NPs formulated with an anti-luciferase siRNA distributed throughout the lungs following oro-tracheal administration, and demonstrated effective gene knockdown and no apparent cytotoxicity. Taken together, these results show that the double emulsion formulated redox responsive PEG-PLGA and OBAE systems represent a promising new therapeutic approach for the local combined chemo- and gene-therapy of lung cancer.
Collapse
Affiliation(s)
- Claudia Conte
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, NG7 2RD, UK. and Drug Delivery Laboratory, Department of Pharmacy, University of Napoli Federico II, 80131 Napoli, Italy
| | - Patrícia F Monteiro
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, NG7 2RD, UK.
| | - Pratik Gurnani
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, NG7 2RD, UK.
| | - Snow Stolnik
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, NG7 2RD, UK.
| | - Francesca Ungaro
- Drug Delivery Laboratory, Department of Pharmacy, University of Napoli Federico II, 80131 Napoli, Italy
| | - Fabiana Quaglia
- Drug Delivery Laboratory, Department of Pharmacy, University of Napoli Federico II, 80131 Napoli, Italy
| | - Philip Clarke
- The BioDiscovery Institute and Faculty of Medicine & Health Sciences, University of Nottingham, NG7 2RD, UK
| | - Anna Grabowska
- The BioDiscovery Institute and Faculty of Medicine & Health Sciences, University of Nottingham, NG7 2RD, UK
| | - Maria Kavallaris
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, NSW 2052, Australia and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and Australian Centre for NanoMedicine, UNSW Sydney, NSW 2052, Australia and School of Women's and Children's Health, Faculty of Medicine, UNSW Sydney, NSW, Australia 2052
| | - Cameron Alexander
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, NG7 2RD, UK.
| |
Collapse
|
9
|
|
10
|
Taresco V, Abelha TF, Cavanagh RJ, Vasey CE, Anane‐Adjei AB, Pearce AK, Monteiro PF, Spriggs KA, Clarke P, Ritchie A, Martin S, Rahman R, Grabowska AM, Ashford MB, Alexander C. Functionalized Block Co‐Polymer Pro‐Drug Nanoparticles with Anti‐Cancer Efficacy in 3D Spheroids and in an Orthotopic Triple Negative Breast Cancer Model. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000103] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Vincenzo Taresco
- School of Pharmacy University Park University of Nottingham Nottingham NG7 2RD UK
| | - Thais F. Abelha
- School of Pharmacy University Park University of Nottingham Nottingham NG7 2RD UK
| | - Robert J. Cavanagh
- School of Pharmacy University Park University of Nottingham Nottingham NG7 2RD UK
| | - Catherine E. Vasey
- School of Pharmacy University Park University of Nottingham Nottingham NG7 2RD UK
| | | | - Amanda K. Pearce
- School of Pharmacy University Park University of Nottingham Nottingham NG7 2RD UK
- School of Chemistry University of Birmingham Birmingham B15 2TT UK
| | - Patrícia F. Monteiro
- School of Pharmacy University Park University of Nottingham Nottingham NG7 2RD UK
| | - Keith A. Spriggs
- School of Pharmacy University Park University of Nottingham Nottingham NG7 2RD UK
| | - Philip Clarke
- AstraZeneca Pharmaceutical Sciences Innovative Medicines Silk Court Business Park Macclesfield Cheshire SK10 2NA UK
| | - Alison Ritchie
- AstraZeneca Pharmaceutical Sciences Innovative Medicines Silk Court Business Park Macclesfield Cheshire SK10 2NA UK
| | - Stewart Martin
- AstraZeneca Pharmaceutical Sciences Innovative Medicines Silk Court Business Park Macclesfield Cheshire SK10 2NA UK
| | - Ruman Rahman
- AstraZeneca Pharmaceutical Sciences Innovative Medicines Silk Court Business Park Macclesfield Cheshire SK10 2NA UK
| | - Anna M. Grabowska
- AstraZeneca Pharmaceutical Sciences Innovative Medicines Silk Court Business Park Macclesfield Cheshire SK10 2NA UK
| | - Marianne B. Ashford
- Division of Cancer and Stem Cells Faculty of Medicine & Health Sciences University of Nottingham Nottingham NG7 2RD UK
| | - Cameron Alexander
- School of Pharmacy University Park University of Nottingham Nottingham NG7 2RD UK
| |
Collapse
|
11
|
Jain V, Kumar H, Anod HV, Chand P, Gupta NV, Dey S, Kesharwani SS. A review of nanotechnology-based approaches for breast cancer and triple-negative breast cancer. J Control Release 2020; 326:628-647. [PMID: 32653502 DOI: 10.1016/j.jconrel.2020.07.003] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/03/2020] [Accepted: 07/04/2020] [Indexed: 12/24/2022]
Abstract
Breast cancer (BC) is one of the most prevalent cancers in women. Triple-negative breast cancer (TNBC) in which the three major receptors i.e. estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2), are absent is known to express the most aggressive phenotype and increased metastasis which results in the development of resistance to chemotherapy. It offers various therapeutic advantages in treating BC and TNBC. Nanotechnology offers various unique characteristics such as small size (nanometric), active and passive targeting, and the ability to attach multiple targeting moieties, controlled release, and site-specific targeting. This review focuses on conventional drug therapies, recent treatment strategies, and unique therapeutic approaches available for BC and TNBC. The role of breast cancer stem cells in the recurrence of BC and TNBC has also been highlighted. Several chemotherapeutic agents delivered using nanocarriers such as polymeric nanoparticles/micelles, metallic/inorganic NPs, and lipid-based NPs (Liposome, solid-lipid nanoparticles (SLNs), and nanostructured lipid carriers (NLCs)), etc. with excellent responses in the treatment of BC/TNBC along with breast cancer stem cells have been discussed in details. Moreover, the application of nanomedicine including CRISPR nanoparticle, exosomes for the treatment of BC/TNBC and other molecular targets available such as poly (ADP-ribose) polymerase (PARP), epidermal growth factor receptor (EGFR), Vascular endothelial growth factor (VEGF), etc. for further exploration have also been discussed.
Collapse
Affiliation(s)
- Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India.
| | - Hitesh Kumar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India
| | - Haritha V Anod
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India
| | - Pallavi Chand
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India
| | - N Vishal Gupta
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India
| | - Surajit Dey
- College of Pharmacy, Roseman University of Health Sciences, Henderson, NV, USA
| | | |
Collapse
|
12
|
Jing W, Ma JT, Han CB. Metastatic Breast Cancer Coexisting With HER-2 Amplification and EGFR Exon 19 Deletion Benefits From EGFR-TKI Therapy: A Case Report. Front Oncol 2020; 10:771. [PMID: 32547945 PMCID: PMC7274020 DOI: 10.3389/fonc.2020.00771] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 04/21/2020] [Indexed: 11/13/2022] Open
Abstract
Background: Patients with different molecular subtypes of breast cancers have different recurrence risks and prognoses. Clinical support and evidence to guide management are absent for patients with breast cancer coexisting with HER-2 amplification and EGFR mutations. Case presentation: We report a case of breast cancer coexisting with HER-2 amplification and EGFR exon 19 deletion (E19 del). The patient presented with solitary pulmonary nodule and enlargement of hilar and mediastinal lymph nodes 2 years after radical mastectomy. Biopsy of the subcarinal lymph node showed suspected adenocarcinoma. The specimen was too small for further immunohistochemistry, but an EGFR E19 del was discovered. Due to the primary diagnosis of EGFR-mutant lung adenocarcinoma, EGFR-TKI gefitinib was administered and resulted in 1 year of stable disease until the patient developed progression in the right pulmonary nodule with new metastatic cervical lymph nodes. According to histopathological findings of re-biopsy of the pulmonary nodule and left cervical and subcarinal lymph nodes, the patient was diagnosed with breast cancer with lung metastasis and multiple lymph node metastases. The patient received multiple anti-HER-2-targeted therapies (trastuzumab for 9.7 months, lapatinib for 9 months, and pyrotinib for 4+ months) and survived for more than 36 months after lung metastasis. Conclusions: This case suggested that breast cancer coexisting with HER-2 amplification and EGFR E19 del may be driven by both HER-2 and EGFR signaling pathways, and patients can benefit from EGFR-TKI and anti-HER-2 therapy.
Collapse
Affiliation(s)
- Wei Jing
- Department of Clinical Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jie-Tao Ma
- Department of Clinical Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Cheng-Bo Han
- Department of Clinical Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
13
|
Babu Varukattu N, Lin W, Vivek R, Rejeeth C, Sabarathinam S, Yao Z, Zhang H. Targeted and Intrinsic Activity of HA-Functionalized PEI-Nanoceria as a Nano Reactor in Potential Triple-Negative Breast Cancer Treatment. ACS APPLIED BIO MATERIALS 2020; 3:186-196. [PMID: 35019435 DOI: 10.1021/acsabm.9b00577] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Although there has been considerable achievement in the field of breast cancer therapeutics, tackling the disturbing issue of highly potent triple-negative breast cancer (TNBC) still remains a hurdle in cancer therapeutics. Here, for the first time we propose a poly(ethylenimine) (PEI)-mediated approach for the synthesis of hyaluronic acid (HA) tagged cerium oxide nanoparticles (CePEI-NPs) as a therapeutic agent in TNBC. Primarily, the formulated HA-CePEI-NPs upon treatment displayed superior anticancer effect by exhibiting the loss of mitochondrial membrane potential (MMP). These particles acted as a nano reactor by the generation of reactive oxygen species (ROS) during the treatment. We further evaluated the caspase activity which divulgated the activation of caspases-3 and -9 while there was a decrease in the level of Bcl-2. The treatment also resulted in the release of cytochrome c (Cyt c), and in addition, features such as pynknosis and G2/M phase arrest were also noted. Hence the nano reactor property of nano ceria in activating mitochondrial-mediated intrinsic apoptosis highlights its promising role as a nano drug for therapeutic applications in TNBC.
Collapse
Affiliation(s)
- Nipun Babu Varukattu
- Cancer Research Center, Shantou University Medical College, Shantou, Guangdong 515041,China
| | - Wan Lin
- Cancer Research Center, Shantou University Medical College, Shantou, Guangdong 515041,China
| | - Raju Vivek
- Bio-nano Therapeutics Research Laboratory, School of Life Sciences, Department of Zoology, Bharathiar University, Coimbatore 641046, India
| | - Chandrababu Rejeeth
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shanmugam Sabarathinam
- Bioprocess and Biomaterials Laboratory, Department of Microbial Biotechnology, Bharathiar University, Coimbatore 641046, India
| | - Zhimeng Yao
- Institute of Precision Cancer and Pathology, Jinan University Medical College, Guangzhou, Guangdong 510632, China
| | - Hao Zhang
- Institute of Precision Cancer and Pathology, Jinan University Medical College, Guangzhou, Guangdong 510632, China
- Cancer Research Center, Shantou University Medical College, Shantou, Guangdong 515041,China
| |
Collapse
|
14
|
Costabile G, Provenzano R, Azzalin A, Scoffone VC, Chiarelli LR, Rondelli V, Grillo I, Zinn T, Lepioshkin A, Savina S, Miro A, Quaglia F, Makarov V, Coenye T, Brocca P, Riccardi G, Buroni S, Ungaro F. PEGylated mucus-penetrating nanocrystals for lung delivery of a new FtsZ inhibitor against Burkholderia cenocepacia infection. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 23:102113. [PMID: 31669084 DOI: 10.1016/j.nano.2019.102113] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 09/16/2019] [Accepted: 10/05/2019] [Indexed: 01/15/2023]
Abstract
C109 is a potent but poorly soluble FtsZ inhibitor displaying promising activity against Burkholderia cenocepacia, a high-risk pathogen for cystic fibrosis (CF) sufferers. To harness C109 for inhalation, we developed nanocrystal-embedded dry powders for inhalation suspension consisting in C109 nanocrystals stabilized with D-α-tocopheryl polyethylene glycol 1000 succinate (TPGS) embedded in hydroxypropyl-β-cyclodextrin (CD). The powders could be safely re-dispersed in water for in vitro aerosolization. Owing to the presence of a PEG shell, the rod shape and the peculiar aspect ratio, C109 nanocrystals were able to diffuse through artificial CF mucus. The promising technological features were completed by encouraging in vitro/in vivo effects. The formulations displayed no toxicity towards human bronchial epithelial cells and were active against planktonic and sessile B. cenocepacia strains. The efficacy of C109 nanosuspensions in combination with piperacillin was confirmed in a Galleria mellonella infection model, strengthening their potential for combined therapy of B. cenocepacia lung infections.
Collapse
Affiliation(s)
| | - Romina Provenzano
- Department of Pharmacy, University of Napoli "Federico II", Napoli, Italy
| | - Alberto Azzalin
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Viola Camilla Scoffone
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Laurent R Chiarelli
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Valeria Rondelli
- Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Segrate, (MI), Italy
| | | | - Thomas Zinn
- ESRF-The European Synchrotron, Grenoble, France
| | - Alexander Lepioshkin
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences, Moscow, Russia
| | - Svetlana Savina
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences, Moscow, Russia
| | - Agnese Miro
- Department of Pharmacy, University of Napoli "Federico II", Napoli, Italy
| | - Fabiana Quaglia
- Department of Pharmacy, University of Napoli "Federico II", Napoli, Italy
| | - Vadim Makarov
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences, Moscow, Russia
| | - Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Paola Brocca
- Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Segrate, (MI), Italy
| | - Giovanna Riccardi
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Silvia Buroni
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy.
| | - Francesca Ungaro
- Department of Pharmacy, University of Napoli "Federico II", Napoli, Italy.
| |
Collapse
|
15
|
Pellosi DS, Paula LB, de Melo MT, Tedesco AC. Targeted and Synergic Glioblastoma Treatment: Multifunctional Nanoparticles Delivering Verteporfin as Adjuvant Therapy for Temozolomide Chemotherapy. Mol Pharm 2019; 16:1009-1024. [PMID: 30698450 DOI: 10.1021/acs.molpharmaceut.8b01001] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Despite advances in cancer therapies, glioblastoma multiforme treatment remains inefficient due to the brain-blood barrier (BBB) inhibitory activity and to the low temozolomide (TMZ) chemotherapeutic selectivity. To improve therapeutic outcomes, in this work we propose two strategies, (i) photodynamic therapy (PDT) as adjuvant treatment and (ii) engineering of multifunctional theranostic/targeted nanoparticles ( m-NPs) that integrate biotin as a targeting moiety with rhodamine-B as a theranostic agent in pluronic P85/F127 copolymers. These smart m-NPs can surmount the BBB and coencapsulate multiple cargoes under optimized conditions. Overall, the present study conducts a rational m-NP design, characterization, and optimizes the formulation conditions. Confocal microscopy studies on T98-G, U87-MG, and U343 glioblastoma cells and on NIH-3T3 normal fibroblast cells show that the m-NPs and the encapsulated drugs are selectively taken up by tumor cells presenting a broad intracellular distribution. The formulations display no toxicity in the absence of light and are not toxic to healthy cells, but they exert a robust synergic action in cancer cells in the case of concomitant PDT/TMZ treatment, especially at low TMZ concentrations and higher light doses, as demonstrated by nonlinear dose-effect curves based on the Chou-Talalay method. The results evidenced different mechanisms of action related to the disjoint cell cycle phases at the optimal PDT/TMZ ratio. This effect favors synergism between the PDT and the chemotherapy with TMZ, enhances the antiproliferative effect, and overcomes cross-resistance mechanisms. These results point out that m-NP-based PDT adjuvant therapy is a promising strategy to improve TMZ-based glioblastoma multiforme treatments.
Collapse
Affiliation(s)
- Diogo S Pellosi
- Laboratory of Hybrid Materials, Department of Chemistry , Federal University of São Paulo , Diadema 04021-001 , Brazil.,Center of Nanotechnology and Tissue Engineering, Photobiology and Photomedicine Research Group, Department of Chemistry FFCLRP , São Paulo University , Ribeirão Preto , Brazil
| | - Leonardo B Paula
- Center of Nanotechnology and Tissue Engineering, Photobiology and Photomedicine Research Group, Department of Chemistry FFCLRP , São Paulo University , Ribeirão Preto , Brazil
| | - Maryanne T de Melo
- Center of Nanotechnology and Tissue Engineering, Photobiology and Photomedicine Research Group, Department of Chemistry FFCLRP , São Paulo University , Ribeirão Preto , Brazil
| | - Antonio C Tedesco
- Center of Nanotechnology and Tissue Engineering, Photobiology and Photomedicine Research Group, Department of Chemistry FFCLRP , São Paulo University , Ribeirão Preto , Brazil
| |
Collapse
|
16
|
Pawar A, Prabhu P. Nanosoldiers: A promising strategy to combat triple negative breast cancer. Biomed Pharmacother 2019; 110:319-341. [DOI: 10.1016/j.biopha.2018.11.122] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/10/2018] [Accepted: 11/25/2018] [Indexed: 12/16/2022] Open
|
17
|
Gaio E, Conte C, Esposito D, Miotto G, Quaglia F, Moret F, Reddi E. Co-delivery of Docetaxel and Disulfonate Tetraphenyl Chlorin in One Nanoparticle Produces Strong Synergism between Chemo- and Photodynamic Therapy in Drug-Sensitive and -Resistant Cancer Cells. Mol Pharm 2018; 15:4599-4611. [PMID: 30148955 DOI: 10.1021/acs.molpharmaceut.8b00597] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cancer therapies based on the combinations of different drugs and/or treatment modalities are emerging as important strategies for increasing efficacy and cure, decreasing unwanted toxicity, and overcoming drug resistance, provided that optimized drug concentration ratios are delivered into the target tissue. To these purposes, delivery systems such as nanoparticles (NPs) offer the unique opportunity to finely tune the drug loading and the release rate of drug combinations in the target tissues. Here, we propose double-layered polymeric NPs for the delivery of the chemotherapeutic docetaxel (DTX) and the photosensitizer disulfonate tetraphenyl chlorin (TPCS2a) coated with hyaluronic acid (HA), which allows cell targeting via CD44 receptors. The simultaneous delivery of the two drugs aims at killing DTX-sensitive (HeLa-P, MDA-MB-231) and DTX-resistant (HeLa-R) cancer cells by combining chemotherapy and photodynamic therapy (PDT). Using the Chou and Talalay method that analyses drug interactions and calculates combination index (CI) using the median-effect principle, we compared the efficiency of DTX chemotherapy combined with TPCS2a-PDT for drugs delivered in the standard solvents, coloaded in the same NP (DTX/TPCS2a-NP) or loaded in separate NPs (DTX-NPs + TPCS2a-NPs). Along with the drug interaction studies, we gained insight into cell death mechanisms after combo-therapy and into the extent of TPCS2a intracellular uptake and localization. In all cell lines considered, the analysis of the viability data revealed synergistic drug/treatment interaction especially when DTX and TPCS2a were delivered to cells coloaded in the same NPs despite the reduced PS uptake measured in the presence of the delivery systems. In fact, while the combinations of the free drugs or drugs in separate NPs gave slight synergism (CI < 1) only at doses killing more than 50% of the cells, the combination of drugs in one NPs gave high synergism also at doses killing 10-20% of the cells. Furthermore, the DTX dose in the combination DTX/TPCS2a-NPs could be reduced by ∼2.6- and 10.7-fold in HeLa-P and MDA-MB-231, respectively. Importantly, drug codelivery in NPs was very efficient in inducing cell mortality also in DTX resistant HeLa-R cells overexpressing P-glycoprotein 1 in which the dose of the chemotherapeutic can be reduced by more than 100 times using DTX/TPCS2a-NPs. Overall, our data demonstrate that the protocol for the preparation of HA-targeted double layer polymeric NPs allows to control the concentration ratio of coloaded drugs and the delivery of the transported drugs for obtaining a highly synergistic interaction combining DTX-chemotherapy and TPCS2a-PDT.
Collapse
Affiliation(s)
- Elisa Gaio
- Cell Biology Unit, Department of Biology , University of Padova , Padova , Italy
| | - Claudia Conte
- Drug Delivery Laboratory, Department of Pharmacy , University of Napoli Federico II , Napoli , Italy
| | - Diletta Esposito
- Drug Delivery Laboratory, Department of Pharmacy , University of Napoli Federico II , Napoli , Italy
| | - Giovanni Miotto
- Department of Molecular Medicine , University of Padova , Padova , Italy
| | - Fabiana Quaglia
- Drug Delivery Laboratory, Department of Pharmacy , University of Napoli Federico II , Napoli , Italy
| | - Francesca Moret
- Cell Biology Unit, Department of Biology , University of Padova , Padova , Italy
| | - Elena Reddi
- Cell Biology Unit, Department of Biology , University of Padova , Padova , Italy
| |
Collapse
|
18
|
Zhong P, Gu X, Cheng R, Deng C, Meng F, Zhong Z. α vβ 3 integrin-targeted micellar mertansine prodrug effectively inhibits triple-negative breast cancer in vivo. Int J Nanomedicine 2017; 12:7913-7921. [PMID: 29138558 PMCID: PMC5667790 DOI: 10.2147/ijn.s146505] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Antibody-mertansine (DM1) conjugates (AMCs) are among the very few active targeting therapeutics that are approved or clinically investigated for treating various cancers including metastatic breast cancer. However, none of the AMCs are effective for the treatment of triple-negative breast cancers (TNBCs). Here, we show that cRGD-decorated, redox-activatable micellar mertansine prodrug (cRGD-MMP) can effectively target and deliver DM1 to αvβ3 integrin overexpressing MDA-MB-231 TNBC xenografts in nude mice, resulting in potent tumor growth inhibition. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays showed that cRGD-MMP had obvious targetability to MDA-MB-231 cells with a low half-maximal inhibitory concentration (IC50) of 0.18 μM, which was close to that of free DM1 and 2.2-fold lower than that of micellar mertansine prodrug (MMP; nontargeting control). The confocal microscopy studies demonstrated that cRGD-MMP mediated a clearly more efficient cellular uptake and intracellular release of doxorubicin (used as a fluorescent anticancer drug model) in MDA-MB-231 cells. Notably, cRGD-MMP loaded with 1,1′-dioctadecyltetramethyl indotricarbocyanine iodide (DiR; a hydrophobic near-infrared dye) was shown to quickly accumulate in the MDA-MB-231 tumor with strong DiR fluorescence from 2 to 24 h post injection. MMP loaded with DiR could also accumulate in the tumor, although significantly less than cRGD-MMP. The biodistribution studies revealed a high DM1 accumulation of 8.1%ID/g in the tumor for cRGD-MMP at 12 h post injection. The therapeutic results demonstrated that cRGD-MMP effectively suppressed MDA-MB-231 tumor growth at 1.6 mg DM1 equiv./kg without causing noticeable side effects, as shown by little body weight loss and histological analysis. This MMP has appeared as a promising platform for potent treatment of TNBCs.
Collapse
Affiliation(s)
- Ping Zhong
- Biomedical Polymers Laboratory.,Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, China
| | - Xiaolei Gu
- Biomedical Polymers Laboratory.,Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, China
| | - Ru Cheng
- Biomedical Polymers Laboratory.,Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, China
| | - Chao Deng
- Biomedical Polymers Laboratory.,Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, China
| | - Fenghua Meng
- Biomedical Polymers Laboratory.,Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory.,Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, China
| |
Collapse
|
19
|
Radiolabeled block copolymer micelles for image-guided drug delivery. Int J Pharm 2016; 515:692-701. [DOI: 10.1016/j.ijpharm.2016.11.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 10/07/2016] [Accepted: 11/02/2016] [Indexed: 01/02/2023]
|
20
|
Pellosi DS, Calori IR, de Paula LB, Hioka N, Quaglia F, Tedesco AC. Multifunctional theranostic Pluronic mixed micelles improve targeted photoactivity of Verteporfin in cancer cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 71:1-9. [PMID: 27987651 DOI: 10.1016/j.msec.2016.09.064] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 09/13/2016] [Accepted: 09/27/2016] [Indexed: 01/04/2023]
Abstract
Nanotechnology development provides new strategies to treat cancer by integration of different treatment modalities in a single multifunctional nanoparticle. In this scenario, we applied the multifunctional Pluronic P123/F127 mixed micelles for Verteporfin-mediated photodynamic therapy in PC3 and MCF-7 cancer cells. Micelles functionalization aimed the targeted delivery by the insertion of biotin moiety on micelle surface and fluorescence image-based through rhodamine-B dye conjugation in the polymer chains. Multifunctional Pluronics formed spherical nanoparticulated micelles that efficiently encapsulated the photosensitizer Verteporfin maintaining its favorable photophysical properties. Lyophilized formulations were stable at least for 6months and readily reconstituted in aqueous media. The multifunctional micelles were stable in protein-rich media due to the dual Pluronic mixed micelles characteristic: high drug loading capacity provided by its micellar core and high kinetic stability due its biocompatible shell. Biotin surface functionalized micelles showed higher internalization rates due biotin-mediated endocytosis, as demonstrated by competitive cellular uptake studies. Rhodamine B-tagged micelles allowed monitoring cellular uptake and intracellular distribution of the formulations. Confocal microscopy studies demonstrated a larger intracellular distribution of the formulation and photosensitizer, which could drive Verteporfin to act on multiple cell sites. Formulations were not toxic in the dark condition, but showed high Verteporfin-induced phototoxicity against both cancer cell lines at low drug and light doses. These results point Verteporfin-loaded multifunctional micelles as a promising tool to further developments in photodynamic therapy of cancer.
Collapse
Affiliation(s)
- Diogo Silva Pellosi
- Laboratory of Phobiology and photomdicine, Department of Chemistry (FFCLRP), University of São Paulo, Av. dos Bandeirantes 3900, 14040-901, Vila Monte Alegre, Ribeirão Preto, Brazil
| | - Italo Rodrigo Calori
- Research Nucleus of Photodynamic Therapy, Department of Chemistry, State University of Maringá, Av. Colombo 5790, 97020-900 Maringá, Brazil
| | - Leonardo Barcelos de Paula
- Laboratory of Phobiology and photomdicine, Department of Chemistry (FFCLRP), University of São Paulo, Av. dos Bandeirantes 3900, 14040-901, Vila Monte Alegre, Ribeirão Preto, Brazil
| | - Noboru Hioka
- Research Nucleus of Photodynamic Therapy, Department of Chemistry, State University of Maringá, Av. Colombo 5790, 97020-900 Maringá, Brazil
| | - Fabiana Quaglia
- Laboratory of Drug Delivery, Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesanto 49, 80131 Napoli, Italy
| | - Antonio Claudio Tedesco
- Laboratory of Phobiology and photomdicine, Department of Chemistry (FFCLRP), University of São Paulo, Av. dos Bandeirantes 3900, 14040-901, Vila Monte Alegre, Ribeirão Preto, Brazil.
| |
Collapse
|
21
|
Pellosi DS, Moret F, Fraix A, Marino N, Maiolino S, Gaio E, Hioka N, Reddi E, Sortino S, Quaglia F. Pluronic ® P123/F127 mixed micelles delivering sorafenib and its combination with verteporfin in cancer cells. Int J Nanomedicine 2016; 11:4479-4494. [PMID: 27660441 PMCID: PMC5019320 DOI: 10.2147/ijn.s103344] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Here, we developed Pluronic® P123/F127 (poloxamer) mixed micelles for the intravenous delivery of the anticancer drug sorafenib (SRB) or its combination with verteporfin (VP), a photosensitizer for photodynamic therapy that should complement well the cytotoxicity profile of the chemotherapeutic. SRB loading inside the core of micelles was governed by the drug:poloxamer weight ratio, while in the case of the SRB-VP combination, a mutual interference between the two drugs occurred and only specific ratios could ensure maximum loading efficiency. Coentrapment of SRB did not alter the photophysical properties of VP, confirming that SRB did not participate in any bimolecular process with the photosensitizer. Fluorescence resonance energy-transfer measurement of micelles in serum protein-containing cell-culture medium demonstrated the excellent stability of the system in physiologically relevant conditions. These results were in line with the results of the release study showing a release rate of both drugs in the presence of proteins slower than in phosphate buffer. SRB release was sustained, while VP remained substantially entrapped in the micelle core. Cytotoxicity studies in MDA-MB231 cells revealed that at 24 hours, SRB-loaded micelles were more active than free SRB only at very low SRB concentrations, while at 24+24 hours a prolonged cytotoxic effect of SRB-loaded micelles was observed, very likely mediated by the block in the S phase of the cell cycle. The combination of SRB with VP under light exposure was less cytotoxic than both the free combination and VP-loaded micelles + SRB-loaded micelles combination. This behavior was clearly explainable in terms of micelle uptake and intracellular localization. Besides the clear advantage of delivering SRB in poloxamer micelles, our results provide a clear example that each photochemotherapeutic combination needs detailed investigations on their particular interaction, and no generalization on enhanced cytotoxic effects should be derived a priori.
Collapse
Affiliation(s)
- Diogo Silva Pellosi
- Research Nucleus of Photodynamic Therapy, Chemistry Department, State University of Maringá, Maringá, Brazil
- Drug Delivery Laboratory, Department of Pharmacy, University of Naples Federico II, Naples
| | - Francesca Moret
- Cell Biology Unit, Department of Biology, University of Padova, Padua
| | - Aurore Fraix
- Laboratory of Photochemistry, Department of Drug Sciences, University of Catania, Catania, Italy
| | - Nino Marino
- Laboratory of Photochemistry, Department of Drug Sciences, University of Catania, Catania, Italy
| | - Sara Maiolino
- Drug Delivery Laboratory, Department of Pharmacy, University of Naples Federico II, Naples
| | - Elisa Gaio
- Cell Biology Unit, Department of Biology, University of Padova, Padua
| | - Noboru Hioka
- Research Nucleus of Photodynamic Therapy, Chemistry Department, State University of Maringá, Maringá, Brazil
| | - Elena Reddi
- Cell Biology Unit, Department of Biology, University of Padova, Padua
| | - Salvatore Sortino
- Laboratory of Photochemistry, Department of Drug Sciences, University of Catania, Catania, Italy
| | - Fabiana Quaglia
- Drug Delivery Laboratory, Department of Pharmacy, University of Naples Federico II, Naples
| |
Collapse
|
22
|
Patel K, Doddapaneni R, Chowdhury N, Boakye CH, Behl G, Singh M. Tumor stromal disrupting agent enhances the anticancer efficacy of docetaxel loaded PEGylated liposomes in lung cancer. Nanomedicine (Lond) 2016; 11:1377-92. [PMID: 27171485 DOI: 10.2217/nnm.16.37] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
AIM Therapeutic efficacy of anticancer nanomedicine is compromised by tumor stromal barriers. The present study deals with the development of docetaxel loaded PEGylated liposomes (DTXPL) and to investigate the effect of tumor stroma disrupting agent, telmisartan, on anticancer efficacy of DTXPL. METHODS DTXPL was prepared using proprietary modified hydration method. Effect of oral telmisartan treatment on tumor uptake of coumarin-6 liposomes and anticancer efficacy of DTXPL was evaluated in orthotopic xenograft lung tumor bearing mice. RESULTS DTXPL (105.7 ± 3.8 nm) showed very high physical stability, negligible hemolysis, 428% enhancement in bioavailability with significantly higher intratumoral uptake. Marked reduction in collagen-I, MMP2/9 and lung tumor weight were observed in DTXPL+telmisartan group. CONCLUSION Combination of DTXPL with telmisartan could significantly enhance clinical outcome in lung cancer.
Collapse
Affiliation(s)
- Ketan Patel
- College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Ravi Doddapaneni
- College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Nusrat Chowdhury
- College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Cedar Ha Boakye
- College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Gautam Behl
- College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Mandip Singh
- College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| |
Collapse
|
23
|
Conte C, Fotticchia I, Tirino P, Moret F, Pagano B, Gref R, Ungaro F, Reddi E, Giancola C, Quaglia F. Cyclodextrin-assisted assembly of PEGylated polyester nanoparticles decorated with folate. Colloids Surf B Biointerfaces 2016; 141:148-157. [DOI: 10.1016/j.colsurfb.2016.01.035] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Revised: 11/27/2015] [Accepted: 01/19/2016] [Indexed: 01/28/2023]
|
24
|
Sorolla A, Ho D, Wang E, Evans CW, Ormonde CFG, Rashwan R, Singh R, Iyer KS, Blancafort P. Sensitizing basal-like breast cancer to chemotherapy using nanoparticles conjugated with interference peptide. NANOSCALE 2016; 8:9343-53. [PMID: 27089946 DOI: 10.1039/c5nr08331a] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Basal-like breast cancers are highly aggressive malignancies associated with very poor prognosis. Although these cancers may initially respond to first-line treatment, they become highly resistant to standard chemotherapy in the metastatic setting. Chemotherapy resistance in basal-like breast cancers is associated with highly selective overexpression of the homeobox transcription factor Engrailed 1 (EN1). Herein, we propose a novel therapeutic strategy using poly(glycidyl methacrylate) nanoparticles decorated with poly(acrylic acid) that enable dual delivery of docetaxel and interference peptides designed to block or inhibit EN1 (EN1-iPep). We demonstrate that EN1-iPep is highly selective in inducing apoptotic cell death in basal-like cancer cells with negligible effects in a non-neoplastic human mammary epithelial cell line. Furthermore, we show that treatment with EN1-iPep results in a highly synergistic pharmacological interaction with docetaxel in inhibiting cancer cell growth. The incorporation of these two agents in a single nanoformulation results in greater anticancer efficacy than current nanoparticle-based treatments used in the clinical setting.
Collapse
Affiliation(s)
- A Sorolla
- Cancer Epigenetics, Harry Perkins Institute of Medical Research, Nedlands, WA 6009, Australia.
| | - D Ho
- School of Chemistry & Biochemistry, The University of Western Australia, Crawley, WA 6009, Australia.
| | - E Wang
- Cancer Epigenetics, Harry Perkins Institute of Medical Research, Nedlands, WA 6009, Australia.
| | - C W Evans
- School of Chemistry & Biochemistry, The University of Western Australia, Crawley, WA 6009, Australia.
| | - C F G Ormonde
- School of Chemistry & Biochemistry, The University of Western Australia, Crawley, WA 6009, Australia.
| | - R Rashwan
- Cancer Epigenetics, Harry Perkins Institute of Medical Research, Nedlands, WA 6009, Australia.
| | - R Singh
- School of Chemistry & Biochemistry, The University of Western Australia, Crawley, WA 6009, Australia.
| | - K Swaminathan Iyer
- School of Chemistry & Biochemistry, The University of Western Australia, Crawley, WA 6009, Australia.
| | - P Blancafort
- Cancer Epigenetics, Harry Perkins Institute of Medical Research, Nedlands, WA 6009, Australia.
| |
Collapse
|
25
|
Mogoşanu GD, Grumezescu AM, Bejenaru C, Bejenaru LE. Polymeric protective agents for nanoparticles in drug delivery and targeting. Int J Pharm 2016; 510:419-29. [PMID: 26972379 DOI: 10.1016/j.ijpharm.2016.03.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 03/09/2016] [Indexed: 01/08/2023]
Abstract
Surface modification/functionalization of nanoparticles (NPs) using polymeric protective agents is an issue of great importance and actuality for drug delivery and targeting. Improving the blood circulation half-life of surface-protected nanocarriers is closely related to the elimination of main biological barriers and limiting factors (protein absorption and opsonization), due to the phagocytic activity of reticuloendothelial system. For passive or active targeted delivery, in biomedical area, surface-functionalized NPs with tissue-recognition ligands were designed and optimized as a result of modern research techniques. Also, multi-functionalized nanostructures are characterized by enhanced bioavailability, efficacy, targeted localization, active cellular uptake, and low side effects. Surface-protected NPs are obtained from biocompatible, biodegradable and less toxic natural polymers (dextran, β-cyclodextrin, chitosan, hyaluronic acid, heparin, gelatin) or synthetic polymers, such as poly(lactic acid), poly(lactic-co-glycolic) acid, poly(ε-caprolactone) and poly(alkyl cyanoacrylates). PEGylation is one of the most important functionalization methods providing steric stabilization, long circulating and 'stealth' properties for both polymeric and inorganic-based nanosystems. In addition, for their antimicrobial, antiviral and antitumor effects, cutting-edge researches in the field of pharmaceutical nanobiotechnology highlighted the importance of noble metal (platinum, gold, silver) NPs decorated with biopolymers.
Collapse
Affiliation(s)
- George Dan Mogoşanu
- Department of Pharmacognosy & Phytotherapy, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
| | - Alexandru Mihai Grumezescu
- Department of Science and Engineering of Oxidic Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, Politehnica University of Bucharest, 1-7 Polizu Street, 011061 Bucharest, Romania.
| | - Cornelia Bejenaru
- Department of Vegetal & Animal Biology, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
| | - Ludovic Everard Bejenaru
- Department of Pharmacognosy & Phytotherapy, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
| |
Collapse
|
26
|
Eloy JO, Petrilli R, Topan JF, Antonio HMR, Barcellos JPA, Chesca DL, Serafini LN, Tiezzi DG, Lee RJ, Marchetti JM. Co-loaded paclitaxel/rapamycin liposomes: Development, characterization and in vitro and in vivo evaluation for breast cancer therapy. Colloids Surf B Biointerfaces 2016; 141:74-82. [PMID: 26836480 DOI: 10.1016/j.colsurfb.2016.01.032] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 01/14/2016] [Accepted: 01/19/2016] [Indexed: 12/27/2022]
Abstract
Paclitaxel and rapamycin have been reported to act synergistically to treat breast cancer. Albeit paclitaxel is available for breast cancer treatment, the most commonly used formulation in the clinic presents side effects, limiting its use. Furthermore, both drugs present pharmacokinetics drawbacks limiting their in vivo efficacy and clinic combination. As an alternative, drug delivery systems, particularly liposomes, emerge as an option for drug combination, able to simultaneously deliver co-loaded drugs with improved therapeutic index. Therefore, the purpose of this study is to develop and characterize a co-loaded paclitaxel and rapamycin liposome and evaluate it for breast cancer efficacy both in vitro and in vivo. Results showed that a SPC/Chol/DSPE-PEG (2000) liposome was able to co-encapsulate paclitaxel and rapamycin with suitable encapsulation efficiency values, nanometric particle size, low polydispersity and neutral zeta potential. Taken together, FTIR and thermal analysis evidenced drug conversion to the more bioavailable molecular and amorphous forms, respectively, for paclitaxel and rapamycin. The pegylated liposome exhibited excellent colloidal stability and was able to retain drugs encapsulated, which were released in a slow and sustained fashion. Liposomes were more cytotoxic to 4T1 breast cancer cell line than the free drugs and drugs acted synergistically, particularly when co-loaded. Finally, in vivo therapeutic evaluation carried out in 4T1-tumor-bearing mice confirmed the in vitro results. The co-loaded paclitaxel/rapamycin pegylated liposome better controlled tumor growth compared to the solution. Therefore, we expect that the formulation developed herein might be a contribution for future studies focusing on the clinical combination of paclitaxel and rapamycin.
Collapse
Affiliation(s)
- Josimar O Eloy
- College of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Avenida do Cafe s/n, 14040-903 Ribeirao Preto, SP, Brazil; College of Pharmacy, The Ohio State University, Columbus, 500W 12th Ave, Columbus, OH 43210, United States
| | - Raquel Petrilli
- College of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Avenida do Cafe s/n, 14040-903 Ribeirao Preto, SP, Brazil; College of Pharmacy, The Ohio State University, Columbus, 500W 12th Ave, Columbus, OH 43210, United States
| | - José Fernando Topan
- College of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Avenida do Cafe s/n, 14040-903 Ribeirao Preto, SP, Brazil
| | - Heriton Marcelo Ribeiro Antonio
- School of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Avenida Bandeirantes s/n, 14040-040 Ribeirao Preto, SP, Brazil
| | - Juliana Palma Abriata Barcellos
- College of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Avenida do Cafe s/n, 14040-903 Ribeirao Preto, SP, Brazil
| | - Deise L Chesca
- School of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Avenida Bandeirantes s/n, 14040-040 Ribeirao Preto, SP, Brazil
| | - Luciano Neder Serafini
- School of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Avenida Bandeirantes s/n, 14040-040 Ribeirao Preto, SP, Brazil
| | - Daniel G Tiezzi
- School of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Avenida Bandeirantes s/n, 14040-040 Ribeirao Preto, SP, Brazil
| | - Robert J Lee
- College of Pharmacy, The Ohio State University, Columbus, 500W 12th Ave, Columbus, OH 43210, United States
| | - Juliana Maldonado Marchetti
- College of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Avenida do Cafe s/n, 14040-903 Ribeirao Preto, SP, Brazil.
| |
Collapse
|
27
|
Yu K, Zhao J, Zhang Z, Gao Y, Zhou Y, Teng L, Li Y. Enhanced delivery of Paclitaxel using electrostatically-conjugated Herceptin-bearing PEI/PLGA nanoparticles against HER-positive breast cancer cells. Int J Pharm 2015; 497:78-87. [PMID: 26617314 DOI: 10.1016/j.ijpharm.2015.11.033] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 10/29/2015] [Accepted: 11/20/2015] [Indexed: 11/17/2022]
Abstract
We have developed a novel nanoparticle delivery system fabricated from polyethylenimine (PEI) and poly(d,l-lactide-co-glycolide) (PLGA), which were able to deliver the chemotherapeutic agent Paclitaxel, while the biomacromolecule Herceptin acted as a targeting ligand that was conjugated onto the surfaces of the nanoparticles via electrostatic interactions. In this study, these electrostatically-conjugated Herceptin-bearing PEI/PLGA nanoparticles (eHER-PPNs) were optimized and employed as vectors to target HER2-positive breast cancer cells. The eHER-PPNs had an average diameter of ∼ 280 nm and a neutral surface charge (1.00 ± 0.73 mV), which remained stable under physiological conditions. The anticancer effects of eHER-PPNs were investigated in HER2-positive BT474 cells and HER2-negative MCF7 cells. The eHER-PPNs showed enhanced cytotoxicity that was dependent on the receptor expression levels and the incubation time. These conjugated nanoparticles deliver Paclitaxel more efficiently (p<0.001) than unmodified PPNs, Herceptin and the combined effects of these two monotherapies. Furthermore, the chemically-conjugated Herceptin-bearing PEI/PLGA nanoparticles (cHER-PPNs) were fabricated as a comparison. The eHER-PPNs exhibited lower cell viability (46.7%) than that of cHER-PPNs (65.1%). The targeting ability of eHER-PPNs was demonstrated through confocal microscopy images and flow cytometry, which showed that eHER-PPNs displayed higher cellular uptake efficiency (p<0.001) in comparison with cHER-PPNs. Therefore, eHER-PPNs could provide promising platforms for the delivery of therapeutic drugs against HER2-positive breast cancers.
Collapse
Affiliation(s)
- Kongtong Yu
- School of Life Sciences, Jilin University, Qianjin Street No.2699, Changchun, Jilin Province 130012, China
| | - Jinlong Zhao
- School of Life Sciences, Jilin University, Qianjin Street No.2699, Changchun, Jilin Province 130012, China
| | - Zunkai Zhang
- School of Life Sciences, Jilin University, Qianjin Street No.2699, Changchun, Jilin Province 130012, China
| | - Yin Gao
- School of Life Sciences, Jilin University, Qianjin Street No.2699, Changchun, Jilin Province 130012, China
| | - Yulin Zhou
- School of Life Sciences, Jilin University, Qianjin Street No.2699, Changchun, Jilin Province 130012, China
| | - Lesheng Teng
- School of Life Sciences, Jilin University, Qianjin Street No.2699, Changchun, Jilin Province 130012, China.
| | - Youxin Li
- School of Life Sciences, Jilin University, Qianjin Street No.2699, Changchun, Jilin Province 130012, China.
| |
Collapse
|
28
|
Mendes TFS, Kluskens LD, Rodrigues LR. Triple Negative Breast Cancer: Nanosolutions for a Big Challenge. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2015; 2:1500053. [PMID: 27980912 PMCID: PMC5115335 DOI: 10.1002/advs.201500053] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 06/03/2015] [Indexed: 05/11/2023]
Abstract
Triple negative breast cancer (TNBC) is a particular immunopathological subtype of breast cancer that lacks expression of estrogen and progesterone receptors (ER/PR) and amplification of the human epidermal growth factor receptor 2 (HER2) gene. Characterized by aggressive and metastatic phenotypes and high rates of relapse, TNBC is the only breast cancer subgroup still lacking effective therapeutic options, thus presenting the worst prognosis. The development of targeted therapies, as well as early diagnosis methods, is vital to ensure an adequate and timely therapeutic intervention in patients with TNBC. This review intends to discuss potentially emerging approaches for the diagnosis and treatment of TNBC patients, with a special focus on nano-based solutions that actively target these particular tumors.
Collapse
Affiliation(s)
| | - Leon D Kluskens
- Centre of Biological Engineering University of Minho 4710-057 Braga Portugal
| | | |
Collapse
|
29
|
Tran TH, Ramasamy T, Choi JY, Nguyen HT, Pham TT, Jeong JH, Ku SK, Choi HG, Yong CS, Kim JO. Tumor-targeting, pH-sensitive nanoparticles for docetaxel delivery to drug-resistant cancer cells. Int J Nanomedicine 2015; 10:5249-62. [PMID: 26346426 PMCID: PMC4552257 DOI: 10.2147/ijn.s89584] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The attachment of polyethylene glycol (PEG) increases the circulation time of drug-containing nanoparticles; however, this also negatively affects cellular uptake. To overcome this problem, unique lipid polymer hybrid (LPH) nanoparticles were developed with a pH-responsive PEG layer that detached prior to cell uptake. Docetaxel (DTX) was incorporated into the lipid core of the nanoparticles, which was then shielded with the pH-responsive block co-polymer polyethylene glycol-b-polyaspartic acid (PEG-b-PAsp) using a modified emulsion method. The optimized LPH nanoparticles were ~200 nm and had a narrow size distribution. Drug release from DTX-loaded LPH (DTX-LPH) nanoparticles was pH-sensitive, which is beneficial for tumor targeting. More importantly, DTX-LPH nanoparticles were able to effectively induce apoptosis in cancer cells. The negative surface charge and PEG shell of vehicle remarkably enhanced the blood circulation and physiological activity of DTX-LPH nanoparticles compared with that of free DTX. The nanoparticles were also found to reduce the size of tumors in tumor-bearing xenograft mice. The in vivo anticancer effect of DTX-LPH nanoparticles was further confirmed by the elevated levels of caspase-3 and poly ADP ribose polymerase found in the tumors after treatment. Thus, the results suggest that this novel LPH system could be an effective new treatment for cancer.
Collapse
Affiliation(s)
- Tuan Hiep Tran
- College of Pharmacy, Yeungnam University, Dae-Dong, South Korea
| | | | - Ju Yeon Choi
- College of Pharmacy, Yeungnam University, Dae-Dong, South Korea
| | | | - Thanh Tung Pham
- College of Pharmacy, Yeungnam University, Dae-Dong, South Korea
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, Dae-Dong, South Korea
| | - Sae Kwang Ku
- College of Korean Medicine, Daegu Haany University, Gyeongsan, South Korea
| | - Han-Gon Choi
- College of Pharmacy, Hanyang University, Hanyangdaehak-ro, Sangnok-gu, Ansan, South Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, Dae-Dong, South Korea
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Dae-Dong, South Korea
| |
Collapse
|
30
|
Guo P, Song S, Li Z, Tian Y, Zheng J, Yang X, Pan W. In vitro and in vivo evaluation of APRPG-modified angiogenic vessel targeting micelles for anticancer therapy. Int J Pharm 2015; 486:356-66. [DOI: 10.1016/j.ijpharm.2015.03.067] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 02/27/2015] [Accepted: 03/30/2015] [Indexed: 12/18/2022]
|
31
|
Maiolino S, Russo A, Pagliara V, Conte C, Ungaro F, Russo G, Quaglia F. Biodegradable nanoparticles sequentially decorated with Polyethyleneimine and Hyaluronan for the targeted delivery of docetaxel to airway cancer cells. J Nanobiotechnology 2015; 13:29. [PMID: 25888948 PMCID: PMC4424546 DOI: 10.1186/s12951-015-0088-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 03/19/2015] [Indexed: 12/31/2022] Open
Abstract
Background Novel polymeric nanoparticles (NPs) specifically designed for delivering chemotherapeutics in the body and aimed at improving treatment activity and selectivity, cover a very relevant area in the field of nanomedicine. Here, we describe how to build a polymer shell of Hyaluronan (HA) and Polyethyleneimine (PEI) on biodegradable NPs of poly(lactic-co-glycolic) acid (PLGA) through electrostatic interactions and to achieve NPs with unique features of sustained delivery of a docetaxel (DTX) drug cargo as well as improved intracellular uptake. Results A stable PEI or HA/PEI shell could be obtained by careful selection of layering conditions. NPs with exquisite stability in salt and protein-rich media, with size and surface charge matching biological requirements for intravenous injection and endowed with sustained DTX release could be obtained. Cytotoxicity, uptake and activity of both PLGA/PEI/HA and PLGA/PEI NPs were evaluated in CD44(+) (A549) and CD44(−) (Calu-3) lung cancer cells. In fact, PEI-coated NPs can be formed after degradation/dissociation of the surface HA because of the excess hyaluronidases overexpressed in tumour interstitium. There was no statistically significant cytotoxic effect of PLGA/PEI/HA and PLGA/PEI NPs in both cell lines, thus suggesting that introduction of PEI in NP shell was not hampered by its intrinsic toxicity. Intracellular trafficking of NPs fluorescently labeled with Rhodamine (RHO) (RHO-PLGA/PEI/HA and RHO-PLGA/PEI NPs) demonstrated an increased time-dependent uptake only for RHO-PLGA/PEI/HA NPs in A549 cells as compared to Calu-3 cells. As expected, RHO-PLGA/PEI NP uptake in A549 cells was comparable to that observed in Calu-3 cells. RHO-PLGA/PEI/HA NPs internalized into A549 cells showed a preferential perinuclear localization. Cytotoxicity data in A549 cells suggested that DTX delivered through PLGA/PEI/HA NPs exerted a more potent antiproliferative activity than free DTX. Furthermore, DTX-PLGA/PEI NPs, as hypothetical result of hyaluronidase-mediated degradation in tumor interstitium, were still able to improve the cytotoxic activity of free DTX. Conclusions Taken together, results lead us to hypothesize that biodegradable NPs coated with a PEI/HA shell represent a very promising system to treat CD44 overexpressing lung cancer. In principle, this novel nanocarrier can be extended to different single drugs and drug combinations taking advantage of the shell and core properties. Electronic supplementary material The online version of this article (doi:10.1186/s12951-015-0088-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sara Maiolino
- Laboratory of Drug Delivery, Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, Napoli, 80131, Italy.
| | - Annapina Russo
- Laboratory of Biochemistry, Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, Napoli, 80131, Italy.
| | - Valentina Pagliara
- Laboratory of Biochemistry, Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, Napoli, 80131, Italy.
| | - Claudia Conte
- Laboratory of Drug Delivery, Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, Napoli, 80131, Italy.
| | - Francesca Ungaro
- Laboratory of Drug Delivery, Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, Napoli, 80131, Italy.
| | - Giulia Russo
- Laboratory of Biochemistry, Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, Napoli, 80131, Italy.
| | - Fabiana Quaglia
- Laboratory of Drug Delivery, Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, Napoli, 80131, Italy.
| |
Collapse
|