1
|
Szupryczyński K, Czeleń P, Jeliński T, Szefler B. What is the Reason That the Pharmacological Future of Chemotherapeutics in the Treatment of Lung Cancer Could Be Most Closely Related to Nanostructures? Platinum Drugs in Therapy of Non-Small and Small Cell Lung Cancer and Their Unexpected, Possible Interactions. The Review. Int J Nanomedicine 2024; 19:9503-9547. [PMID: 39296940 PMCID: PMC11410046 DOI: 10.2147/ijn.s469217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/19/2024] [Indexed: 09/21/2024] Open
Abstract
Over the course of several decades, anticancer treatment with chemotherapy drugs for lung cancer has not changed significantly. Unfortunately, this treatment prolongs the patient's life only by a few months, causing many side effects in the human body. It has also been proven that drugs such as Cisplatin, Carboplatin, Oxaliplatin and others can react with other substances containing an aromatic ring in which the nitrogen atom has a free electron group in its structure. Thus, such structures may have a competitive effect on the nucleobases of DNA. Therefore, scientists are looking not only for new drugs, but also for new alternative ways of delivering the drug to the cancer site. Nanotechnology seems to be a great hope in this matter. Creating a new nanomedicine would reduce the dose of the drug to an absolute minimum, and thus limit the toxic effect of the drug; it would allow for the exclusion of interactions with competitive compounds with a structure similar to nucleobases; it would also permit using the so-called targeted treatment and bypassing healthy cells; it would allow for the introduction of other treatment options, such as radiotherapy directly to the cancer site; and it would provide diagnostic possibilities. This article is a review that aims to systematize the knowledge regarding the anticancer treatment of lung cancer, but not only. It shows the clear possibility of interactions of chemotherapeutics with compounds competitive to the nitrogenous bases of DNA. It also shows the possibilities of using nanostructures as potential Platinum drug carriers, and proves that nanomedicine can easily become a new medicinal product in personalized medicine.
Collapse
Affiliation(s)
- Kamil Szupryczyński
- Doctoral School of Medical and Health Sciences, Faculty of Pharmacy, Collegium Medicum, Nicolaus, Copernicus University, Bydgoszcz, Poland
| | - Przemysław Czeleń
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Tomasz Jeliński
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Beata Szefler
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| |
Collapse
|
2
|
Kanwal S, Naveed M, Arshad A, Arshad A, Firdous F, Faisal A, Yameen B. Reduction-Sensitive Dextran-Paclitaxel Polymer-Drug Conjugate: Synthesis, Self-Assembly into Nanoparticles, and In Vitro Anticancer Efficacy. Bioconjug Chem 2021; 32:2516-2529. [PMID: 34762796 DOI: 10.1021/acs.bioconjchem.1c00492] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Delivery systems that can encapsulate a precise amount of drug and offer a spatiotemporally controlled drug release are being actively sought for safe yet effective cancer therapy. Compared to polymer nanoparticle (NP)-based delivery systems that rely on physical drug encapsulation, NPs derived from stimuli-sensitive covalent polymer-drug conjugates (PDCs) have emerged as promising alternatives offering precise control over drug dosage and spatiotemporal drug release. Herein, we report a reduction-sensitive PDC "Dex-SS-PTXL" synthesized by conjugating dextran and paclitaxel (PTXL) through a disulfide bond-bearing linker. The synthesized Dex-SS-PTXL PDC with a precise degree of substitution in terms of the percentage of repeat units of dextran covalently conjugated to PTXL (27 ± 0.6%) and the amount of drug carried by the PDC (39 ± 1.4 wt %) was found to self-assemble into spherical NPs with an average size of 110 ± 34 nm and a ζ-potential of -14.09 ± 8 mV. The reduction-sensitive Dex-SS-PTXL NPs were found to release PTXL exclusively in response to the reducing agent concentration reflective of the intracellular reducing environment of the tumor cells. Challenging BT-549 and MCF-7 cells with Dex-SS-PTXL NPs revealed significant cytotoxicity, while the IC50 values and the mode of action (mitotic arrest) of Dex-SS-PTXL NPs were found to be comparable to those of free PTXL, highlighting the active nature of the intracellularly released drug. The developed PDC with its unique ability to self-assemble into NPs and stimuli-responsive drug release can enhance the success of the NP-based drug delivery systems during clinical translation.
Collapse
Affiliation(s)
- Sidra Kanwal
- Department of Chemistry and Chemical Engineering, Syed Babar Ali School of Science and Engineering (SBASSE), Lahore University of Management Sciences (LUMS), Lahore 54792, Pakistan
| | - Muhammad Naveed
- Cancer Therapeutics Laboratory, Department of Biology, Syed Babar Ali School of Science and Engineering (SBASSE), Lahore University of Management Sciences (LUMS), Lahore 54792, Pakistan
| | - Ali Arshad
- Department of Chemistry and Chemical Engineering, Syed Babar Ali School of Science and Engineering (SBASSE), Lahore University of Management Sciences (LUMS), Lahore 54792, Pakistan
| | - Azka Arshad
- Department of Chemistry and Chemical Engineering, Syed Babar Ali School of Science and Engineering (SBASSE), Lahore University of Management Sciences (LUMS), Lahore 54792, Pakistan
| | - Farhat Firdous
- Department of Chemistry and Chemical Engineering, Syed Babar Ali School of Science and Engineering (SBASSE), Lahore University of Management Sciences (LUMS), Lahore 54792, Pakistan
| | - Amir Faisal
- Cancer Therapeutics Laboratory, Department of Biology, Syed Babar Ali School of Science and Engineering (SBASSE), Lahore University of Management Sciences (LUMS), Lahore 54792, Pakistan
| | - Basit Yameen
- Department of Chemistry and Chemical Engineering, Syed Babar Ali School of Science and Engineering (SBASSE), Lahore University of Management Sciences (LUMS), Lahore 54792, Pakistan
| |
Collapse
|
3
|
Lee CR, Kim GG, Park SB, Kim SW. Synthesis of Hyaluronic Acid-Conjugated Fe 3O 4@CeO 2 Composite Nanoparticles for a Target-Oriented Multifunctional Drug Delivery System. MICROMACHINES 2021; 12:mi12091018. [PMID: 34577662 PMCID: PMC8466504 DOI: 10.3390/mi12091018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/20/2021] [Accepted: 08/24/2021] [Indexed: 11/16/2022]
Abstract
This study is based on the principle that superparamagnetic iron oxide nanoparticles (Fe3O4) can be used to target a specific area given that their magnetic properties emerge when an external magnetic field is applied. Cerium oxide (CeO2), which causes oxidative stress by generating reactive oxygen species (ROS) in the environment of tumor cells, was synthesized on the surface of superparamagnetic iron oxide nanoparticles to produce nanoparticles that selectively kill cancer cells. In addition, hyaluronic acid (HA) was coated on the cerium's surface to target CD44-overexpressing tumor cells, and natZr was chelated on the Fe3O4@CeO2 surface to show the usefulness of labeling the radioisotope 89Zr (T1/2 = 3.3 d). The synthesis of Fe3O4@CeO2 was confirmed by Fourier Transform-Infrared Spectroscopy (FT-IR), X-ray Diffraction (XRD) and Field Emission-Transmission Electron Microscope (FE-TEM). The coating of HA was confirmed by FT-IR, X-ray Photoelectron. Spectroscopy (XPS), FE-TEM, Energy-Dispersive X-ray Spectroscopy (EDS) and Thermogravimetric Analysis (TGA)/Differential Scanning Calorimetry (DSC). The sizes of the prepared nanoparticles were confirmed through FE-TEM and Field Emission-Scanning Electron (FE-SEM) (sizes of 15 to 30 nm), and it was confirmed that natZr was introduced onto the surface of the nanoparticles using EDS. The particle size of the dispersed material was limited through Dynamic Light Scattering (DLS) to about 148 nm in aqueous solution, which was suitable for the (enhanced permeation and retention) EPR effect. It was confirmed that the HA-coated nanoparticles have good dispersibility. Finally, a cytotoxicity evaluation confirmed the ability of CeO2 to generate ROS and target the delivery of HA. In conclusion, Fe3O4@CeO2 can effectively inhibit cancer cells through the activity of cerium oxide in the body when synthesized in nano-sized superparamagnetic coral iron that has magnetic properties. Subsequently, by labeling the radioactive isotope 89Zr, it is possible to create a theranostic drug delivery system that can be used for cancer diagnosis.
Collapse
Affiliation(s)
- Chang Ryong Lee
- Department of Advanced Materials Chemistry, Dongguk University, Gyeongju 38066, Korea; (C.R.L.); (G.G.K.)
| | - Gun Gyun Kim
- Department of Advanced Materials Chemistry, Dongguk University, Gyeongju 38066, Korea; (C.R.L.); (G.G.K.)
| | - Sung Bum Park
- Department of Safety Engineering, Dongguk University, Gyeongju 38066, Korea;
| | - Sang Wook Kim
- Department of Advanced Materials Chemistry, Dongguk University, Gyeongju 38066, Korea; (C.R.L.); (G.G.K.)
- Correspondence: ; Tel.: +82-54-770-2216
| |
Collapse
|
4
|
Clinical Milestones in Nanotherapeutics: Current Status and Future Prospects. NANOMATERIALS: EVOLUTION AND ADVANCEMENT TOWARDS THERAPEUTIC DRUG DELIVERY (PART II) 2021:194-245. [DOI: 10.2174/9781681088235121010009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
5
|
Cao S, Lin C, Liang S, Tan CH, Er Saw P, Xu X. Enhancing Chemotherapy by RNA Interference. BIO INTEGRATION 2020. [DOI: 10.15212/bioi-2020-0003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Abstract Small interfering RNA (siRNA) has shown tremendous potential for treating human diseases in the past decades. siRNA can selectively silence a pathological pathway through the targeting and degradation of a specific mRNA, significantly reducing the off-target side
effects of anticancer drugs. However, the poor pharmacokinetics of RNA significantly restricted the clinical use of RNAi technology. In this review, we examine in-depth the siRNA therapeutics currently in preclinical and clinical trials, multiple challenges faced in siRNA therapy, feasibility
of siRNA treatment with anticancer drugs in combined with siRNA in nanoparticles or modified to be parental drugs, sequential therapy of siRNA treatment prior to drug treatment with siRNA and drugs loaded in nanoparticles. We focused on the combinatorial activation of apoptosis by different
pathways, namely Bcl-2, survivin, and Pgp protein. Taken together, this review would serve to establish the pathway of effective and efficient combination therapy of siRNA and drugs as a new strategy.
Collapse
Affiliation(s)
- Shuwen Cao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chunhao Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shunung Liang
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Baiyun District, Guangzhou, China
| | - Chee Hwee Tan
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Baiyun District, Guangzhou, China
| | - Phei Er Saw
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoding Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
6
|
Hathout RM, Metwally AA, Woodman TJ, Hardy JG. Prediction of Drug Loading in the Gelatin Matrix Using Computational Methods. ACS OMEGA 2020; 5:1549-1556. [PMID: 32010828 PMCID: PMC6990624 DOI: 10.1021/acsomega.9b03487] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 12/31/2019] [Indexed: 05/05/2023]
Abstract
The delivery of drugs is a topic of intense research activity in both academia and industry with potential for positive economic, health, and societal impacts. The selection of the appropriate formulation (carrier and drug) with optimal delivery is a challenge investigated by researchers in academia and industry, in which millions of dollars are invested annually. Experiments involving different carriers and determination of their capacity for drug loading are very time-consuming and therefore expensive; consequently, approaches that employ computational/theoretical chemistry to speed have the potential to make hugely beneficial economic, environmental, and health impacts through savings in costs associated with chemicals (and their safe disposal) and time. Here, we report the use of computational tools (data mining of the available literature, principal component analysis, hierarchical clustering analysis, partial least squares regression, autocovariance calculations, molecular dynamics simulations, and molecular docking) to successfully predict drug loading into model drug delivery systems (gelatin nanospheres). We believe that this methodology has the potential to lead to significant change in drug formulation studies across the world.
Collapse
Affiliation(s)
- Rania M. Hathout
- Department
of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
- E-mail: (R.M.H.)
| | - AbdelKader A. Metwally
- Department
of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
- Department
of Pharmaceutics, Faculty of Pharmacy, Health Sciences Center, Kuwait University, Kuwait 90805, Kuwait
| | - Timothy J. Woodman
- Department
of Pharmacy and Pharmacology, University
of Bath, Bath BA2 7AY, U.K
| | - John G. Hardy
- Department
of Chemistry, Lancaster University, Lancaster, Lancashire LA1 4YB, U.K
- Materials
Science Institute, Lancaster University, Lancaster, Lancashire LA1 4YB, U.K
- E-mail; (J.G.H.)
| |
Collapse
|
7
|
Anaya-Ruiz M, Bandala C, Landeta G, Martínez-Morales P, Zumaquero-Rios JL, Sarracent-Pérez J, Pérez-Santos M. Nanostructured Systems in Advanced Drug Targeting for the Cancer Treatment: Recent Patents. Recent Pat Anticancer Drug Discov 2019; 14:85-94. [PMID: 30381087 DOI: 10.2174/1574892813666181031154146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 09/23/2018] [Accepted: 10/30/2018] [Indexed: 02/05/2023]
Abstract
BACKGROUND Cancer is one of the leading causes of death in the world and it is necessary to develop new strategies for its treatment because most therapies have limited access to many types of tumors, as well as low therapeutic efficacy and high toxicity. OBJECTIVE The present research aims to identify recent patents of drug delivery nanostructured systems that may have application in improving cancer treatment. METHODS Recent patents regarding the drug delivery nanostructured systems for cancer treatment were obtained from the patent databases of the six main patent offices of the world: United States Patent and Trademark Office, European Patent Office, World Intellectual Property Organization, Japan Patent Office, State Intellectual Property Office of China and Korean Intellectual Property Office. RESULTS A total of 1710 patent documents from 1998 to 2017 including "drug delivery nanostructured systems for cancer treatment" were retrieved. The top five countries in patent share were USA, China, South Korea, Canada and Germany. The universities and enterprises of USA had the highest amount of patents followed by institutions from China. CONCLUSION There is a strong tendency for the development of new nanostructured systems for the release of drugs; particularly, in recent years, the development of nanoparticles has focused on nanodiscs, gold nanoparticles and immunoliposomes.
Collapse
Affiliation(s)
- Maricruz Anaya-Ruiz
- Cell Biology Laboratory, Biomedical Research Center of the East, Mexican Institute of Social Security, Metepec, Puebla CP 74360, Mexico
| | - Cindy Bandala
- Department of Neuurosciences, National Institute of Rehabilitation, City of Mexico, Mexico
| | - Gerardo Landeta
- Coordination of Applied Research, University Center for Linkage and Transfer of Technology, Benemérita Autonomous University of Puebla, Puebla CP 72570, Mexico
| | - Patricia Martínez-Morales
- Laboratory of Molecular Biology, Center for Biomedical Research of the East, Mexican Institute of Social Security / CONACYT, Metepec, Puebla CP 74360, Mexico
| | - Jose L Zumaquero-Rios
- Laboratory of Parasitology and Vectors, Faculty of Biology, Benemérita Universidad Autonomous University of Puebla, Puebla CP 72570, Mexico
| | - Jorge Sarracent-Pérez
- Laboratory of Parasitology, Institute of Tropical Medicine "Pedro Kouri ", Havana CP 11400, Cuba
| | - Martín Pérez-Santos
- Technology Marketing Office, University Link Center and Technology Transfer, Benemerita Universidad Autonoma de Puebla, Puebla CP 72570, Mexico
| |
Collapse
|
8
|
He J, Li C, Ding L, Huang Y, Yin X, Zhang J, Zhang J, Yao C, Liang M, Pirraco RP, Chen J, Lu Q, Baldridge R, Zhang Y, Wu M, Reis RL, Wang Y. Tumor Targeting Strategies of Smart Fluorescent Nanoparticles and Their Applications in Cancer Diagnosis and Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1902409. [PMID: 31369176 DOI: 10.1002/adma.201902409] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/30/2019] [Indexed: 06/10/2023]
Abstract
Advantages such as strong signal strength, resistance to photobleaching, tunable fluorescence emissions, high sensitivity, and biocompatibility are the driving forces for the application of fluorescent nanoparticles (FNPs) in cancer diagnosis and therapy. In addition, the large surface area and easy modification of FNPs provide a platform for the design of multifunctional nanoparticles (MFNPs) for tumor targeting, diagnosis, and treatment. In order to obtain better targeting and therapeutic effects, it is necessary to understand the properties and targeting mechanisms of FNPs, which are the foundation and play a key role in the targeting design of nanoparticles (NPs). Widely accepted and applied targeting mechanisms such as enhanced permeability and retention (EPR) effect, active targeting, and tumor microenvironment (TME) targeting are summarized here. Additionally, a freshly discovered targeting mechanism is introduced, termed cell membrane permeability targeting (CMPT), which improves the tumor-targeting rate from less than 5% of the EPR effect to more than 50%. A new design strategy is also summarized, which is promising for future clinical targeting NPs/nanomedicines design. The targeting mechanism and design strategy will inspire new insights and thoughts on targeting design and will speed up precision medicine and contribute to cancer therapy and early diagnosis.
Collapse
Affiliation(s)
- Jiuyang He
- Tumor Precision Targeting Research Center, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Chenchen Li
- Tumor Precision Targeting Research Center, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
| | - Lin Ding
- Tumor Precision Targeting Research Center, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
- Department of Biological Chemistry, The University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yanan Huang
- Tumor Precision Targeting Research Center, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
| | - Xuelian Yin
- Tumor Precision Targeting Research Center, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
| | - Junfeng Zhang
- Tumor Precision Targeting Research Center, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
| | - Jian Zhang
- Universal Medical Imaging Diagnostic Research Center, Shanghai, 200233, P. R. China
| | - Chenjie Yao
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
- Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, 02115, USA
| | - Minmin Liang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Rogério P Pirraco
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's PT Government Associate Lab, 4805, Braga/Guimarães, Portugal
| | - Jie Chen
- Tumor Precision Targeting Research Center, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
| | - Quan Lu
- Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, 02115, USA
| | - Ryan Baldridge
- Department of Biological Chemistry, The University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yong Zhang
- Tumor Precision Targeting Research Center, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
- Department of Biomedical Engineering, National University of Singapore, Singapore, 119077, Singapore
| | - Minghong Wu
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's PT Government Associate Lab, 4805, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017, Barco, Guimarães, Portugal
| | - Yanli Wang
- Tumor Precision Targeting Research Center, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
- Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, 02115, USA
| |
Collapse
|
9
|
Jain AK, Thareja S. In vitro and in vivo characterization of pharmaceutical nanocarriers used for drug delivery. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:524-539. [DOI: 10.1080/21691401.2018.1561457] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Akhlesh K. Jain
- Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh, India
| | - Suresh Thareja
- Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh, India
| |
Collapse
|
10
|
Noury M, López J. Nanomedicine and personalised medicine: understanding the personalisation of health care in the molecular era. SOCIOLOGY OF HEALTH & ILLNESS 2017; 39:547-565. [PMID: 27782304 DOI: 10.1111/1467-9566.12502] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Globally supported by public policy and investment, nanomedicine is presented as an ongoing medical revolution that will radically change the practice of health care from diagnostic to therapeutic, and everything in between. One of nanomedicine's major promises is that of personalised medicine, enabling diagnostics and therapeutics tailored to individual needs and developing a truly 'patient-friendly' medical approach. Based on qualitative interviews with nanomedicine researchers in Canada, this article explores the emerging concept of personalised medicine as it becomes entangled with nanomedical research. More precisely, drawing on insights from science studies and the sociology of expectations, it analyses researchers' perceptions of personalised medicine in the cutting edge of current nanomedicine research. Two perceptions of personalisation are identified; a molecular conception of individuality and a technical conception of personalisation. The article concludes by examining the relationship between the two conceptions and contrasts them with the normative reflex of a more expansive conception of personalised medicine.
Collapse
Affiliation(s)
- Mathieu Noury
- Université du Québec en Abitibi-Témiscamingue, Rouyn-Noranda, Quebec, Canada
| | | |
Collapse
|
11
|
Doi Y, Abu Lila AS, Matsumoto H, Okada T, Shimizu T, Ishida T. Improvement of intratumor microdistribution of PEGylated liposome via tumor priming by metronomic S-1 dosing. Int J Nanomedicine 2016; 11:5573-5582. [PMID: 27822036 PMCID: PMC5087787 DOI: 10.2147/ijn.s119069] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The efficient delivery of nanocarrier-based cancer therapeutics into tumor tissue is problematic. Structural abnormalities, tumor vasculature heterogeneity, and elevated intratumor pressure impose barriers against the preferential accumulation of nanocarrier-based cancer therapeutics within tumor tissues and, consequently, compromise their therapeutic efficacy. Recently, we have reported that metronomic S-1, orally available tegafur formulation, dosing synergistically augmented the therapeutic efficacy of oxaliplatin (l-OHP)-containing PEGylated liposome without increasing the toxicity in animal model. However, the exact mechanism behind such synergistic effect was not fully elucidated. In this study, therefore, we tried to shed the light on the contributions of metronomic S-1 dosing to the enhanced accumulation and/or spatial distribution of PEGylated liposome within tumor tissue. Tumor priming with metronomic S-1 treatment induced a potent apoptotic response against both angiogenic endothelial cells and tumor cells adjacent to tumor blood vessels, resulting in enhanced tumor blood flow via transient normalization of tumor vasculature, along with alleviation of intratumor pressure. Such a change in the tumor microenvironment imparted by S-1 treatment allows efficient delivery of PEGylated liposome to tumor tissue and permits their deep penetration/distribution into the tumor mass. Such a priming effect of S-1 dosing can be exploited as a promising strategy to enhance the therapeutic efficacy of nanocarrier-based cancer therapeutics suffering from inadequate/heterogeneous delivery to tumor tissues.
Collapse
Affiliation(s)
- Yusuke Doi
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Amr S Abu Lila
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt; Department of Pharmaceutics, Faculty of Pharmacy, Hail University, Hail, Saudi Arabia
| | - Haruna Matsumoto
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Tomoko Okada
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Taro Shimizu
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| |
Collapse
|
12
|
Sun L, Xiong X, Zou Q, Ouyang P, Burkhardt C, Krastev R. Design of intelligent chitosan/heparin hollow microcapsules for drug delivery. J Appl Polym Sci 2016. [DOI: 10.1002/app.44425] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Lili Sun
- College of Biotechnology and Pharmaceutical Engineering; Nanjing Tech University; 211816 Nanjing China
| | - Xin Xiong
- NMI Natural and Medical Sciences Institute at the University of Tübingen; 72770 Reutlingen Germany
| | - Qiaogen Zou
- School of Pharmaceutical Sciences; Nanjing Tech University; 211816 Nanjing China
| | - Pingkai Ouyang
- College of Biotechnology and Pharmaceutical Engineering; Nanjing Tech University; 211816 Nanjing China
| | - Claus Burkhardt
- NMI Natural and Medical Sciences Institute at the University of Tübingen; 72770 Reutlingen Germany
| | - Rumen Krastev
- NMI Natural and Medical Sciences Institute at the University of Tübingen; 72770 Reutlingen Germany
- Faculty of Applied Chemistry; Reutlingen University; 72762 Reutlingen Germany
| |
Collapse
|
13
|
Nakamura Y, Mochida A, Choyke PL, Kobayashi H. Nanodrug Delivery: Is the Enhanced Permeability and Retention Effect Sufficient for Curing Cancer? Bioconjug Chem 2016; 27:2225-2238. [PMID: 27547843 DOI: 10.1021/acs.bioconjchem.6b00437] [Citation(s) in RCA: 624] [Impact Index Per Article: 69.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Nanotechnology offers several attractive design features that have prompted its exploration for cancer diagnosis and treatment. Nanosized drugs have a large loading capacity, the ability to protect the payload from degradation, a large surface on which to conjugate targeting ligands, and controlled or sustained release. Nanosized drugs also leak preferentially into tumor tissue through permeable tumor vessels and are then retained in the tumor bed due to reduced lymphatic drainage. This process is known as the enhanced permeability and retention (EPR) effect. However, while the EPR effect is widely held to improve delivery of nanodrugs to tumors, it in fact offers less than a 2-fold increase in nanodrug delivery compared with critical normal organs, resulting in drug concentrations that are not sufficient for curing most cancers. In this Review, we first overview various barriers for nanosized drug delivery with an emphasis on the capillary wall's resistance, the main obstacle to delivering drugs. Then, we discuss current regulatory issues facing nanomedicine. Finally, we discuss how to make the delivery of nanosized drugs to tumors more effective by building on the EPR effect.
Collapse
Affiliation(s)
- Yuko Nakamura
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, Maryland 20892-1088, United States
| | - Ai Mochida
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, Maryland 20892-1088, United States
| | - Peter L Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, Maryland 20892-1088, United States
| | - Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, Maryland 20892-1088, United States
| |
Collapse
|
14
|
Kobayashi H, Choyke PL. Super enhanced permeability and retention (SUPR) effects in tumors following near infrared photoimmunotherapy. NANOSCALE 2016; 8:12504-9. [PMID: 26443992 PMCID: PMC4824660 DOI: 10.1039/c5nr05552k] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
To date, the delivery of nano-sized therapeutic agents to cancers largely relies on enhanced permeability and retention (EPR) effects that are caused by the leaky nature of cancer vasculature. However, nano-sized agents delivered in this way have demonstrated limited success in oncology due to the relatively small magnitude of the EPR effect. For achieving superior delivery of nano-sized agents, super-enhanced permeability and retention (SUPR) effects are needed. Near infrared photo-immunotherapy (NIR-PIT) is a recently reported therapy that treats tumors with light therapy and subsequently causes an increase in nano-drug delivery up to 24-fold compared with untreated tumors in which only the EPR effect is present. SUPR effects could enhance delivery into tumor beds of a wide variety of nano-sized agents including particles, antibodies, and protein binding small molecular agents. Therefore, taking advantage of the SUPR effects after NIR-PIT may be a promising avenue to utilize a wide variety of nano-drugs in a highly effective manner.
Collapse
Affiliation(s)
- Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | |
Collapse
|
15
|
Chen X, Liu L, Jiang C. Charge-reversal nanoparticles: novel targeted drug delivery carriers. Acta Pharm Sin B 2016; 6:261-7. [PMID: 27471667 PMCID: PMC4951588 DOI: 10.1016/j.apsb.2016.05.011] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 05/15/2016] [Accepted: 05/16/2016] [Indexed: 01/31/2023] Open
Abstract
Spurred by significant progress in materials chemistry and drug delivery, charge-reversal nanocarriers are being developed to deliver anticancer formulations in spatial-, temporal- and dosage-controlled approaches. Charge-reversal nanoparticles can release their drug payload in response to specific stimuli that alter the charge on their surface. They can elude clearance from the circulation and be activated by protonation, enzymatic cleavage, or a molecular conformational change. In this review, we discuss the physiological basis for, and recent advances in the design of charge-reversal nanoparticles that are able to control drug biodistribution in response to specific stimuli, endogenous factors (changes in pH, redox gradients, or enzyme concentration) or exogenous factors (light or thermos-stimulation).
Collapse
Key Words
- Abs, integrin aVb3 mAbs
- B-PDEAEA, poly[(2-acryloyl) ethyl (p-boronic acid benzyl) diethylammonium bromide]
- BPS, bridged polysilsesquioxanexerogel
- BSA, bovine serum albumin
- CA4, combretastatin A4
- CAPL, charge reversible pullulan-based
- CHPNH2, cationic cholesteryl group–bearing pullulans
- CMC, carboxymethyl cellulose
- CPLAs, cationic polylactides
- Cancer therapy
- Charge-reversal nanoparticles
- Cit, citraconic anhydride
- Cya, cysteamine hydrochloride
- DAP, 2,3-diamino-propionate
- DCL, dimethyl maleamidic acid-ε-caprolactone
- DDS, drug delivery system
- DM, dimyristeroyl
- DMA, 2,3-dimethylmaleic anhydride
- DMPA, dimethylol propionic acid
- DOX, doxorubicin
- Drug delivery carriers
- FITC, fluorescein isothiocyanate
- GO, graphene oxide
- GSH, glutathione
- Glu, glutamic acid
- HCC, hepatocellular carcinoma
- HEP, 1,4-bis(2-hydroxyethyl) piperazine
- HMP, p-hydroxylmethylenephenol
- His, histidine
- MG, microgels
- MMPs, matrix metalloproteinases
- MNP, magnetic nanoparticles
- NPs, nanoparticles
- Nanotechnology
- PAEP, poly(allyl ethylene phosphate)
- PAH, poly(allylamine) hydrochloride
- PBAE, poly(β-amino ester)
- PCL, poly(ε-caprolactone)
- PDADMAC, poly(diallyldimethylammonium chloride)
- PEG, polyethylene glycol
- PEI, polyethylenimine
- PEO, poly(ethylene oxide)
- PK, protein kinase
- PLA, ploylactic acid
- PLGA, poly(lactic-co-glycolic acid)
- PLL, poly(l-lysine)
- PMA, poly(methacrylic acid)
- PS, pH sensitive
- PSS, poly(sodium 4-styrenesulfonate)
- PSSS, poly(styrene-co-4-styrene-sulfonate)
- PTX, paclitaxel
- PU, polyurethane
- PVPON, poly(N-vinylpyrrolidone)
- ROS, reactive oxygen species
- SOD, superoxide dismutase
- Stimuli responsive
- TMA, 2-(mercaptoethyl) trimethylammonium chloride
- TUNA, thioundecyl-tetraethyleneglycolester-o-nitrobenzy-lethyldimethyl ammonium bromide
- pA-F, fluorescein-labeled polyanion
Collapse
Affiliation(s)
- Xinli Chen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Lisha Liu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| |
Collapse
|
16
|
Coelho SC, Almeida GM, Santos-Silva F, Pereira MC, Coelho MAN. Enhancing the efficiency of bortezomib conjugated to pegylated gold nanoparticles: an in vitro study on human pancreatic cancer cells and adenocarcinoma human lung alveolar basal epithelial cells. Expert Opin Drug Deliv 2016; 13:1075-81. [PMID: 27087021 DOI: 10.1080/17425247.2016.1178234] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Gold nanoparticles have become promising vectors for cancer diagnosis and treatment. The present study investigates the effect of bortezomib (BTZ), a proteasome inhibitor, conjugated with pegylated gold nanoparticles (PEGAuNPs) in pancreatic and lung cancer cells. METHODS Synthesized gold nanoparticles (PEGAuNPs) were conjugated with bortezomib antitumor drug. We investigated the cytotoxicity induced by BTZ conjugated with functionalized gold nanoparticles in vitro, in the human pancreatic (S2-013) and lung (A549) cancer cell lines. RESULTS We found an efficient of conjugation of BTZ with PEGAuNPs. In vitro assays showed that after 72 h' incubation with PEGAuNPs-BTZ cancer cells revealed alterations in morphology; also for S2-013 and A549 cancer cells, the IC50 value of free BTZ is respectively 1.5 and 4.3 times higher than the IC50 value of PEGAuNPs-BTZ. Furthermore, for TERT-HPNE, the IC50 value is around 63 times lower for free BTZ than the conjugated nanovehicle. Cell growth inhibition results showed a remarkable enhancement in the effect of BTZ when conjugated with AuNPs. CONCLUSIONS Our findings showed that conjugation with PEGAuNPs enhance the BTZ growth-inhibition effect on human cancer cells (S2-013 and A549) and decreases its toxicity against normal cells (TERT-HPNE).
Collapse
Affiliation(s)
- Sílvia Castro Coelho
- a LEPABE, Department of Chemical Engineering, Faculty of Engineering , University of Porto , Porto , Portugal
| | - Gabriela M Almeida
- b Instituto de Investigação e Inovação em Saúde , Universidade do Porto , Porto , Portugal.,c Expression Regulation in Cancer Group , IPATIMUP , Porto , Portugal
| | - Filipe Santos-Silva
- b Instituto de Investigação e Inovação em Saúde , Universidade do Porto , Porto , Portugal.,d Public Awareness of Cancer Unit , IPATIMUP , Porto , Portugal.,e Faculty of Medicine , University of Porto , Porto , Portugal.,f Department of Biochemistry and Molecular Biology, Eppley Institute , University of Nebraska Medical Center , Omaha , NE , USA
| | - Maria Carmo Pereira
- a LEPABE, Department of Chemical Engineering, Faculty of Engineering , University of Porto , Porto , Portugal
| | - Manuel A N Coelho
- a LEPABE, Department of Chemical Engineering, Faculty of Engineering , University of Porto , Porto , Portugal
| |
Collapse
|
17
|
Nanoparticles in radiation oncology: From bench-side to bedside. Cancer Lett 2016; 375:256-262. [PMID: 26987625 DOI: 10.1016/j.canlet.2016.03.011] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 03/07/2016] [Accepted: 03/08/2016] [Indexed: 12/13/2022]
Abstract
Nanoparticles (NP) are "in vogue" in medical research. Pre-clinical studies accumulate evidence of NP enhancing radiation therapy. On one hand, NP, selected for their intrinsic physicochemical characteristics, are radio-sensitizers. Thus, when NP accumulate in cancer cells, they increase the radiation absorption coefficient specifically in tumour tissue, sparing healthy surrounding tissue from toxicity. On the other hand, NP, by being drug vectors, can carry radio-sensitizer therapeutics to cancer cells. Finally, NP present theranostic effects. Indeed they are used in imaging as contrast agents. NP therefore can be multi-tasking and have promising prospect in radiotherapy field. In spite of the numerous encouraging preclinical evidence, the very small number of clinical trials investigating NP possible involvement in the radiotherapy clinical practice suggests a physicians' unwillingness. Many prerequisites seem necessary including define biological mechanisms of NP radiosensitization pathways and of NP clearance. NP biocompatibility and toxicities should be better investigated to select, among the extensive range of possible systems, the harmless and most efficient one, and to finally come to a safe and successful clinical use. The present review focuses on the various interests of NP in the radiotherapy area and proposes a discussion about their role in the future clinical practice.
Collapse
|
18
|
Supramolecular nanoscale assemblies for cancer diagnosis and therapy. J Control Release 2015; 213:152-167. [PMID: 26160308 DOI: 10.1016/j.jconrel.2015.06.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/26/2015] [Accepted: 06/27/2015] [Indexed: 02/08/2023]
Abstract
Nanocarriers based on polymers, metals and lipids have been extensively developed for cancer therapy and diagnosis due to their ability to enhance drug accumulation in cancer cells and decrease undesired drug toxicity in healthy tissues. Overcoming multidrug resistance by designing proper drug nanocarriers will improve outcome of existing oncologic treatments such as chemotherapy and radiotherapy. In this article the relation between physicochemical properties and capacity of a nanosystem to deliver therapeutic agents into pathological sites is discussed. Most promising examples of drug delivery systems are reviewed, and, in particular, the design of a carbohydrate based matrix with entrapped gold nanoparticles is highlighted.
Collapse
|
19
|
Kobayashi H, Turkbey B, Watanabe R, Choyke PL. Cancer drug delivery: considerations in the rational design of nanosized bioconjugates. Bioconjug Chem 2014; 25:2093-100. [PMID: 25385142 PMCID: PMC4275162 DOI: 10.1021/bc500481x] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
![]()
In
order to efficiently deliver anticancer agents to tumors, biocompatible
nanoparticles or bioconjugates, including antibody–drug conjugates
(ADCs), have recently been designed, synthesized, and tested, some
even in clinical trials. Controlled delivery can be enhanced by changing
specific design characteristics of the bioconjugate such as its size,
the nature of the payload, and the surface features. The delivery
of macromolecular drugs to cancers largely relies on the leaky nature
of the tumor vasculature compared with healthy vessels in normal organs.
When administered intravenously, macromolecular bioconjugates and
nanosized agents tend to circulate for prolonged times, unless they
are small enough to be excreted by the kidney or stealthy enough to
evade the macrophage phagocytic system (MPS), formerly the reticulo-endothelial
system (RES). Therefore, macromolecular bioconjugates and nanosized
agents with long circulation times leak preferentially into tumor
tissue through permeable tumor vessels and are then retained in the
tumor bed due to reduced lymphatic drainage. This process is known
as the enhanced permeability and retention (EPR) effect. However,
success of cancer drug delivery only relying on the EPR effect is
still limited. To cure cancer patients, further improvement of drug
delivery is required by both designing superior agents and enhancing
EPR effects. In this Review, we describe the basis of macromolecular
or nanosized bioconjugate delivery into cancer tissue and discuss
current diagnostic methods for evaluating leakiness of the tumor vasculature.
Then, we discuss methods to augment conventional “permeability
and retention” effects for macromolecular or nanosized bioconjugates
in cancer tissue.
Collapse
Affiliation(s)
- Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, Maryland 20892, United States
| | | | | | | |
Collapse
|
20
|
Rhoda K, Choonara YE, Kumar P, Bijukumar D, du Toit LC, Pillay V. Potential nanotechnologies and molecular targets in the quest for efficient chemotherapy in ovarian cancer. Expert Opin Drug Deliv 2014; 12:613-34. [PMID: 25300775 DOI: 10.1517/17425247.2015.970162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Ovarian cancer, considered one of the most fatal gynecological cancers, goes largely undiagnosed until metastasis presents itself, usually once the patient is in the final stages and thus, too late for worthwhile therapy. Targeting this elusive disease in its early stages would improve the outcome for most patients, while the information generated thereof would increase the possibility of preventative mechanisms of therapy. AREAS COVERED This review discusses various molecular targets as possible moieties to be incorporated in a holistic drug delivery system or the more aptly termed 'theranostic' system. These molecular targets can be used for targeting, visualizing, diagnosing, and ultimately, treating ovarian cancer in its entirety. Currently implemented nanoframeworks, such as nanomicelles and nanoliposomes, are described and the effectiveness of nanostructures in tumor targeting, treatment functions, and overcoming the drug resistance challenge is discussed. EXPERT OPINION Novel nanotechnology strategies such as the development of nanoframeworks decorated with targeted ligands of a molecular nature may provide an efficient chemotherapy, especially when instituted in combination with imaging, diagnostic, and ultimately, therapeutic moieties. An imperative aspect of utilizing nanotechnology in the treatment of ovarian cancer is the flexibility of the drug delivery system and its ability to overcome standard obstacles such as: i) successfully treating the desired cells through direct targeting; ii) reducing toxicity levels of treatment by achieving direct targeting; and iii) delivery of targeted therapy using an efficient vehicle that is exceptionally degradable in response to a particular stimulus. The targeting of ovarian cancer in its early stages using imaging and diagnostic nanotechnology is an area that can be improved upon by combining therapeutic moieties with molecular biomarkers. The nanotechnology and molecular markers mentioned in this review have generally been used for either imaging or diagnostics, and have not yet been successfully implemented into bi-functional tools, which it is hoped, should eventually include a therapeutic aspect.
Collapse
Affiliation(s)
- Khadija Rhoda
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand , Johannesburg, 7 York Road, Parktown, 2193 , South Africa
| | | | | | | | | | | |
Collapse
|
21
|
Bexiga MG, Kelly C, Dawson KA, Simpson JC. RNAi-mediated inhibition of apoptosis fails to prevent cationic nanoparticle-induced cell death in cultured cells. Nanomedicine (Lond) 2014; 9:1651-64. [DOI: 10.2217/nnm.13.151] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background: Nanoparticles are increasingly being considered as a novel and potent tool for drug delivery, and, therefore, concerns regarding the safety of their use in humans are pertinent. It has been shown that nanoparticles displaying unsaturated amines at their surface are toxic to cells, but the molecular and cellular mechanisms elicited in this response have yet to be described. Aims: In this work we identify key proteins involved in the cytotoxicity of amine-modified polystyrene nanoparticles. We also demonstrate the suitability of RNAi to provide a molecular description of how nanoparticles and cells interact. Materials & methods: We have used a focused RNAi strategy in 1321N1 cells to identify key proteins involved in the cytotoxicity induced by amine-modified polystyrene nanoparticles. Results: We show that the apoptosome is central to the observed mechanism of toxicity and that, although the proapoptotic proteins BAX, BAK, BID, BIM and PUMA are critical modulators of the process, their cellular depletion is insufficient to protect cells from nanoparticle-induced cell death. Conclusion: We conclude that the apoptosome, together with proapoptotic proteins of the Bcl-2 family of proteins, is central to amine-modified polystyrene nanoparticle-induced cell death. We further demonstrate that RNAi is a powerful and suitable tool to study the effects of nanoparticles on cellular processes, in particular apoptosis. Original submitted 18 March 2013; Revised submitted 22 July 2013
Collapse
Affiliation(s)
- Mariana G Bexiga
- School of Biology & Environmental Science & Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
- Centre for Neuroscience & Cell Biology, University of Coimbra, Portugal
- Centre for BioNano Interactions, School of Chemistry & Chemical Biology, University College Dublin, Dublin 4, Ireland
| | - Ciara Kelly
- School of Biology & Environmental Science & Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Kenneth A Dawson
- Centre for BioNano Interactions, School of Chemistry & Chemical Biology, University College Dublin, Dublin 4, Ireland
| | - Jeremy C Simpson
- School of Biology & Environmental Science & Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
| |
Collapse
|
22
|
Saito N, Haniu H, Usui Y, Aoki K, Hara K, Takanashi S, Shimizu M, Narita N, Okamoto M, Kobayashi S, Nomura H, Kato H, Nishimura N, Taruta S, Endo M. Safe clinical use of carbon nanotubes as innovative biomaterials. Chem Rev 2014; 114:6040-79. [PMID: 24720563 PMCID: PMC4059771 DOI: 10.1021/cr400341h] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Indexed: 02/06/2023]
Affiliation(s)
- Naoto Saito
- Institute
for Biomedical Sciences, Shinshu University, Asahi 3-1-1, Matsumoto 390-8621, Japan
| | - Hisao Haniu
- Department
of Orthopaedic Surgery, Shinshu University
School of Medicine, Asahi
3-1-1, Matsumoto 390-8621, Japan
| | - Yuki Usui
- Department
of Orthopaedic Surgery, Shinshu University
School of Medicine, Asahi
3-1-1, Matsumoto 390-8621, Japan
- Research Center for Exotic Nanocarbons, and Faculty of Engineering, Shinshu University, Wakasato 4-17-1, Nagano 380-8553, Japan
| | - Kaoru Aoki
- Department
of Orthopaedic Surgery, Shinshu University
School of Medicine, Asahi
3-1-1, Matsumoto 390-8621, Japan
| | - Kazuo Hara
- Department
of Orthopaedic Surgery, Shinshu University
School of Medicine, Asahi
3-1-1, Matsumoto 390-8621, Japan
| | - Seiji Takanashi
- Department
of Orthopaedic Surgery, Shinshu University
School of Medicine, Asahi
3-1-1, Matsumoto 390-8621, Japan
| | - Masayuki Shimizu
- Department
of Orthopaedic Surgery, Shinshu University
School of Medicine, Asahi
3-1-1, Matsumoto 390-8621, Japan
| | - Nobuyo Narita
- Department
of Orthopaedic Surgery, Shinshu University
School of Medicine, Asahi
3-1-1, Matsumoto 390-8621, Japan
| | - Masanori Okamoto
- Department
of Orthopaedic Surgery, Shinshu University
School of Medicine, Asahi
3-1-1, Matsumoto 390-8621, Japan
| | - Shinsuke Kobayashi
- Department
of Orthopaedic Surgery, Shinshu University
School of Medicine, Asahi
3-1-1, Matsumoto 390-8621, Japan
| | - Hiroki Nomura
- Department
of Orthopaedic Surgery, Shinshu University
School of Medicine, Asahi
3-1-1, Matsumoto 390-8621, Japan
| | - Hiroyuki Kato
- Department
of Orthopaedic Surgery, Shinshu University
School of Medicine, Asahi
3-1-1, Matsumoto 390-8621, Japan
| | - Naoyuki Nishimura
- R&D
Center, Nakashima Medical Co. Ltd., Haga 5322, Kita-ku, Okayama 701-1221, Japan
| | - Seiichi Taruta
- Research Center for Exotic Nanocarbons, and Faculty of Engineering, Shinshu University, Wakasato 4-17-1, Nagano 380-8553, Japan
| | - Morinobu Endo
- Research Center for Exotic Nanocarbons, and Faculty of Engineering, Shinshu University, Wakasato 4-17-1, Nagano 380-8553, Japan
| |
Collapse
|
23
|
Kobayashi H, Watanabe R, Choyke PL. Improving conventional enhanced permeability and retention (EPR) effects; what is the appropriate target? Am J Cancer Res 2013; 4:81-9. [PMID: 24396516 PMCID: PMC3881228 DOI: 10.7150/thno.7193] [Citation(s) in RCA: 663] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 08/30/2013] [Indexed: 12/18/2022] Open
Abstract
Nano-sized therapeutic agents have several advantages over low molecular weight agents such as a larger loading capacity, the ability to protect the payload until delivery, more specific targeting due to multivalency and the opportunity for controlled/sustained release. However, the delivery of nano-sized agents into cancer tissue is problematic because it mostly relies on the enhanced permeability and retention (EPR) effect that depends on the leaky nature of the tumor vasculature and the prolonged circulation of nano-sized agents, allowing slow but uneven accumulation in the tumor bed. Delivery of nano-sized agents is dependent on several factors that influence the EPR effect; 1. Regional blood flow to the tumor, 2. Permeability of the tumor vasculature, 3. Structural barriers imposed by perivascular tumor cells and extracellular matrix, 4. Intratumoral pressure. In this review, these factors will be described and methods to enhance nano-agent delivery will be reviewed.
Collapse
|
24
|
Videira M, Arranja A, Rafael D, Gaspar R. Preclinical development of siRNA therapeutics: towards the match between fundamental science and engineered systems. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2013; 10:689-702. [PMID: 24333589 DOI: 10.1016/j.nano.2013.11.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 11/21/2013] [Accepted: 11/24/2013] [Indexed: 12/14/2022]
Abstract
UNLABELLED The evolution of synthetic RNAi faces the paradox of interfering with the human biological environment. Due to the fact that all cell physiological processes can be target candidates, silencing a precise biological pathway could be challenging if target selectivity is not properly addressed. Molecular biology has provided scientific tools to suppress some of the most critical issues in gene therapy, while setting the standards for siRNA clinical application. However, the protein down-regulation through the mRNA silencing is intimately related to the sequence-specific siRNA ability to interact accurately with the potential target. Moreover, its in vivo biological fate is highly dependent on the successful design of a vehicle able to overcome both extracellular and intracellular barriers. Anticipating a great deal of innovation, crucial to meet the challenges involved in the RNAi therapeutics, the present review intends to build up a synopsis on the delivery strategies currently developed. FROM THE CLINICAL EDITOR This review discusses recent progress and pertinent limiting factors related to the use of siRNA-s as efficient protein-specific "silencing" agents, focusing on targeted delivery not only to cells of interest, but to the proper intracellular destination.
Collapse
Affiliation(s)
- M Videira
- iMed.UL - Research Institute for Medicines and Pharmaceutical Sciences, Faculty of Pharmacy, University of Lisbon, Av. Professor Gama Pinto, 1649-003 Lisbon, Portugal.
| | - A Arranja
- iMed.UL - Research Institute for Medicines and Pharmaceutical Sciences, Faculty of Pharmacy, University of Lisbon, Av. Professor Gama Pinto, 1649-003 Lisbon, Portugal
| | - D Rafael
- iMed.UL - Research Institute for Medicines and Pharmaceutical Sciences, Faculty of Pharmacy, University of Lisbon, Av. Professor Gama Pinto, 1649-003 Lisbon, Portugal
| | - R Gaspar
- iMed.UL - Research Institute for Medicines and Pharmaceutical Sciences, Faculty of Pharmacy, University of Lisbon, Av. Professor Gama Pinto, 1649-003 Lisbon, Portugal
| |
Collapse
|
25
|
Pan S, Sardesai NP, Liu H, Rusling JF. Assessing DNA Damage from Enzyme-Oxidized Single-Walled Carbon Nanotubes. Toxicol Res (Camb) 2013; 2:375-378. [PMID: 24159372 DOI: 10.1039/c3tx50022e] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Peroxidase enzyme digests of oxidized single-wall carbon nanotubes (SWCNT) were shown to damage DNA in potentially genotoxic reactions for the first time using an electro-optical array with and without metabolic activation.
Collapse
Affiliation(s)
- Shenmin Pan
- Department of Chemistry, University of Connecticut
| | | | | | | |
Collapse
|
26
|
Alvarez-Lorenzo C, Concheiro A. From Drug Dosage Forms to Intelligent Drug-delivery Systems: a Change of Paradigm. SMART MATERIALS FOR DRUG DELIVERY 2013. [DOI: 10.1039/9781849736800-00001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The design of new drug-delivery systems (DDSs) able to regulate the moment and the rate at which the release should take place, and even to target the drug to specific tissues and cell compartments, has opened novel perspectives to improve the efficacy and safety of the therapeutic treatments. Ideally, the drug should only have access to its site of action and the release should follow the evolution of the disease or of certain biorhythms. The advances in the DDSs field are possible because of a better knowledge of the physiological functions and barriers to the drug access to the action site, but also due to the possibility of having “active” excipients that provide novel features. The joint work in a wide range of disciplines, comprising materials science, biomedical engineering and pharmaceutical technology, prompts the design and development of materials (lipids, polymers, hybrids) that can act as sensors of physiological parameters or external variables, and as actuators able to trigger or tune the release process. Such smart excipients lead to an advanced generation of DDSs designed as intelligent or stimuli-responsive. This chapter provides an overview of how the progress in DDSs is intimately linked to the evolution of the excipients, understood as a specific category of biomaterials. The phase transitions, the stimuli that can trigger them and the mechanisms behind the performance of the intelligent DDSs are analyzed as a whole, to serve as an introduction to the topics that are comprehensively discussed in the subsequent chapters of the book. A look to the future is also provided.
Collapse
Affiliation(s)
- C. Alvarez-Lorenzo
- Departamento de Farmacia y Tecnología Farmacéutica Facultad de Farmacia, Universidad de Santiago de Compostela, 15782-Santiago de Compostela Spain
| | - A. Concheiro
- Departamento de Farmacia y Tecnología Farmacéutica Facultad de Farmacia, Universidad de Santiago de Compostela, 15782-Santiago de Compostela Spain
| |
Collapse
|
27
|
Gasbarri C, Angelini G, Fontana A, De Maria P, Siani G, Giannicchi I, Cort AD. Kinetics of demetallation of a zinc–salophen complex into liposomes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1818:747-52. [DOI: 10.1016/j.bbamem.2011.10.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 09/07/2011] [Accepted: 10/18/2011] [Indexed: 01/10/2023]
|
28
|
Venturelli E, Fabbro C, Chaloin O, Ménard-Moyon C, Smulski CR, Da Ros T, Kostarelos K, Prato M, Bianco A. Antibody covalent immobilization on carbon nanotubes and assessment of antigen binding. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2011; 7:2179-2187. [PMID: 21608125 DOI: 10.1002/smll.201100137] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 03/01/2011] [Indexed: 05/30/2023]
Abstract
Controlling the covalent bonding of antibodies onto functionalized carbon nanotubes is a key step in the design and preparation of nanotube-based conjugates for targeting cancer cells. For this purpose, an anti-MUC1 antibody (Ab) is linked to both multi-walled (MWCNTs) and double-walled carbon nanotubes (DWCNTs) using different synthetic strategies. The presence of the Ab attached to the nanotubes is confirmed by gel electrophoresis and thermogravimetric analysis. Most importantly, molecular recognition of the antigen by surface plasmon resonance is able to determine similar Ab binding capacities for both Ab-DWCNTs and Ab-MWCNTs. These results are very relevant for the design of future receptor-targeting strategies using chemically functionalized carbon nanotubes.
Collapse
Affiliation(s)
- Enrica Venturelli
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Laboratoire d'Immunologie et Chimie Thérapeutiques, Strasbourg 67000, France
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Samoshina NM, Liu X, Brazdova B, Franz AH, Samoshin VV, Guo X. Fliposomes: pH-Sensitive Liposomes Containing a trans-2-morpholinocyclohexanol-Based Lipid That Performs a Conformational Flip and Triggers an Instant Cargo Release in Acidic Medium. Pharmaceutics 2011; 3:379-405. [PMID: 24310586 PMCID: PMC3857072 DOI: 10.3390/pharmaceutics3030379] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 06/07/2011] [Accepted: 06/30/2011] [Indexed: 02/02/2023] Open
Abstract
Incorporation of a pH-sensitive conformational switch into a lipid structure enables a drastic conformational flip upon protonation that disrupts the liposome membrane and causes rapid release of cargo specifically in areas of increased acidity. pH-sensitive liposomes containing the amphiphile (1) with trans-2-morpholinocyclohexanol conformational switch, a phospholipid, and a PEG-lipid conjugate were constructed and characterized. The optimized composition—1/POPC/PEG-ceramide (50/4/5)—could be stored at 4 °C and pH 7.4 for up to 1.5 years, and was stable in blood serum in vitro after 48 h at 37 °C. Liposomes loaded with ANTS/DPX or methotrexate demonstrated an unusually quick content release (in a few seconds) at pH below 5.5, which was independent of inter-liposome contact. The pH-titration curve for the liposome leakage paralleled the curve for the acid-induced conformational flip of 1 studied by 1H-NMR. Freeze-fracture electron microscopy images showed budding and division of the bilayer at pH 5.5. A plausible mechanism of pH-sensitivity involves an acid-triggered conformational flip of 1, shortening of lipid tails, and membrane perturbations, which cause the content leakage. The methotrexate-loaded liposomes demonstrated much higher cytotoxicity in HeLa cells than the free drug indicating that they can serve as viable drug delivery systems.
Collapse
Affiliation(s)
- Nataliya M. Samoshina
- Department of Pharmaceutics and Medicinal Chemistry, University of the Pacific, 751 Brookside Road, Stockton, CA 95211, USA
- Department of Chemistry, University of the Pacific, 3601 Pacific Ave., Stockton, CA 95211, USA
| | - Xin Liu
- Department of Chemistry, University of the Pacific, 3601 Pacific Ave., Stockton, CA 95211, USA
| | - Barbora Brazdova
- Department of Chemistry, University of the Pacific, 3601 Pacific Ave., Stockton, CA 95211, USA
| | - Andreas H. Franz
- Department of Chemistry, University of the Pacific, 3601 Pacific Ave., Stockton, CA 95211, USA
| | - Vyacheslav V. Samoshin
- Department of Chemistry, University of the Pacific, 3601 Pacific Ave., Stockton, CA 95211, USA
- Authors to whom correspondence should be addressed; E-Mails: (V.V.S.); (X.G.); Tel.: +1-209-946-2921 (V.V.S.); +1-209-946-2321 (X.G.); Fax: +1-209-946-2607 (V.V.S.)
| | - Xin Guo
- Department of Pharmaceutics and Medicinal Chemistry, University of the Pacific, 751 Brookside Road, Stockton, CA 95211, USA
- Authors to whom correspondence should be addressed; E-Mails: (V.V.S.); (X.G.); Tel.: +1-209-946-2921 (V.V.S.); +1-209-946-2321 (X.G.); Fax: +1-209-946-2607 (V.V.S.)
| |
Collapse
|
30
|
Metwally AA, Pourzand C, Blagbrough IS. Efficient Gene Silencing by Self-Assembled Complexes of siRNA and Symmetrical Fatty Acid Amides of Spermine. Pharmaceutics 2011; 3:125-40. [PMID: 24310492 PMCID: PMC3864230 DOI: 10.3390/pharmaceutics3020125] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 03/11/2011] [Accepted: 03/22/2011] [Indexed: 01/10/2023] Open
Abstract
Gene silencing by siRNA (synthetic dsRNA of 21-25 nucleotides) is a well established biological tool in gene expression studies and has a promising therapeutic potential for difficult-to-treat diseases. Five fatty acids of various chain length and oxidation state (C12:0, C18:0, C18:1, C18:2, C22:1) were conjugated to the naturally occurring polyamine, spermine, and evaluated for siRNA delivery and gene knock-down. siRNA delivery could not be related directly to gene silencing efficiency as N4,N9-dierucoyl spermine resulted in higher siRNA delivery compared to N4,N9-dioleoyl spermine. GFP silencing in HeLa cells showed that the unsaturated fatty acid amides are more efficient than saturated fatty acid amides, with N4,N9-dioleoyl spermine resulting in the most efficient gene silencing in the presence of serum. The alamarBlue cell viability assay showed that fatty acid amides of spermine have good viability (75%–85% compared to control) except N4,N9-dilauroyl spermine which resulted in low cell viability. These results prove that unsaturated fatty acid amides of spermine are efficient, non-toxic, non-viral vectors for siRNA mediated gene silencing.
Collapse
|