1
|
Chaudhary V, Kajla P, Lather D, Chaudhary N, Dangi P, Singh P, Pandiselvam R. Bacteriophages: a potential game changer in food processing industry. Crit Rev Biotechnol 2024; 44:1325-1349. [PMID: 38228500 DOI: 10.1080/07388551.2023.2299768] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 08/16/2023] [Accepted: 10/03/2023] [Indexed: 01/18/2024]
Abstract
In the food industry, despite the widespread use of interventions such as preservatives and thermal and non-thermal processing technologies to improve food safety, incidences of foodborne disease continue to happen worldwide, prompting the search for alternative strategies. Bacteriophages, commonly known as phages, have emerged as a promising alternative for controlling pathogenic bacteria in food. This review emphasizes the potential applications of phages in biological sciences, food processing, and preservation, with a particular focus on their role as biocontrol agents for improving food quality and preservation. By shedding light on recent developments and future possibilities, this review highlights the significance of phages in the food industry. Additionally, it addresses crucial aspects such as regulatory status and safety concerns surrounding the use of bacteriophages. The inclusion of up-to-date literature further underscores the relevance of phage-based strategies in reducing foodborne pathogenic bacteria's presence in both food and the production environment. As we look ahead, new phage products are likely to be targeted against emerging foodborne pathogens. This will further advance the efficacy of approaches that are based on phages in maintaining the safety and security of food.
Collapse
Affiliation(s)
- Vandana Chaudhary
- Department of Dairy Technology, College of Dairy Science and Technology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana, India
| | - Priyanka Kajla
- Department of Food Technology, Guru Jambheshwar University of Science and Technology, Hisar, Haryana, India
| | - Deepika Lather
- Department of Veterinary Pathology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana, India
| | - Nisha Chaudhary
- Department of Food Science and Technology, College of Agriculture, Agriculture University, Jodhpur, Rajasthan, India
| | - Priya Dangi
- Department of Food and Nutrition and Food Technology, Institute of Home Economics, University of Delhi, New Delhi, India
| | - Punit Singh
- Department of Mechanical Engineering, Institute of Engineering and Technology, GLA University Mathura, Mathura, Uttar Pradesh, India
| | - Ravi Pandiselvam
- Physiology, Biochemistry and Post-Harvest Technology Division, ICAR -Central Plantation Crops Research Institute, Kasaragod, Kerala, India
| |
Collapse
|
2
|
Rana A, Adhikary M, Singh PK, Das BC, Bhatnagar S. "Smart" drug delivery: A window to future of translational medicine. Front Chem 2023; 10:1095598. [PMID: 36688039 PMCID: PMC9846181 DOI: 10.3389/fchem.2022.1095598] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 11/28/2022] [Indexed: 01/05/2023] Open
Abstract
Chemotherapy is the mainstay of cancer treatment today. Chemotherapeutic drugs are non-selective and can harm both cancer and healthy cells, causing a variety of adverse effects such as lack of specificity, cytotoxicity, short half-life, poor solubility, multidrug resistance, and acquiring cancer stem-like characteristics. There is a paradigm shift in drug delivery systems (DDS) with the advent of smarter ways of targeted cancer treatment. Smart Drug Delivery Systems (SDDSs) are stimuli responsive and can be modified in chemical structure in response to light, pH, redox, magnetic fields, and enzyme degradation can be future of translational medicine. Therefore, SDDSs have the potential to be used as a viable cancer treatment alternative to traditional chemotherapy. This review focuses mostly on stimuli responsive drug delivery, inorganic nanocarriers (Carbon nanotubes, gold nanoparticles, Meso-porous silica nanoparticles, quantum dots etc.), organic nanocarriers (Dendrimers, liposomes, micelles), antibody-drug conjugates (ADC) and small molecule drug conjugates (SMDC) based SDDSs for targeted cancer therapy and strategies of targeted drug delivery systems in cancer cells.
Collapse
Affiliation(s)
- Abhilash Rana
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Meheli Adhikary
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Praveen Kumar Singh
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Bhudev C. Das
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India,Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh, India
| | - Seema Bhatnagar
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India,*Correspondence: Seema Bhatnagar,
| |
Collapse
|
3
|
New Advances in Biomedical Application of Polymeric Micelles. Pharmaceutics 2022; 14:pharmaceutics14081700. [PMID: 36015325 PMCID: PMC9416043 DOI: 10.3390/pharmaceutics14081700] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/29/2022] [Accepted: 08/07/2022] [Indexed: 12/20/2022] Open
Abstract
In the last decade, nanomedicine has arisen as an emergent area of medicine, which studies nanometric systems, namely polymeric micelles (PMs), that increase the solubility and the stability of the encapsulated drugs. Furthermore, their application in dermal drug delivery is also relevant. PMs present unique characteristics because of their unique core-shell architecture. They are colloidal dispersions of amphiphilic compounds, which self-assemble in an aqueous medium, giving a structure-type core-shell, with a hydrophobic core (that can encapsulate hydrophobic drugs), and a hydrophilic shell, which works as a stabilizing agent. These features offer PMs adequate steric protection and determine their hydrophilicity, charge, length, and surface density properties. Furthermore, due to their small size, PMs can be absorbed by the intestinal mucosa with the drug, and they transport the drug in the bloodstream until the therapeutic target. Moreover, PMs improve the pharmacokinetic profile of the encapsulated drug, present high load capacity, and are synthesized by a reproducible, easy, and low-cost method. In silico approaches have been explored to improve the physicochemical properties of PMs. Based on this, a computer-aided strategy was developed and validated to enable the delivery of poorly soluble drugs and established critical physicochemical parameters to maximize drug loading, formulation stability, and tumor exposure. Poly(2-oxazoline) (POx)-based PMs display unprecedented high loading concerning water-insoluble drugs and over 60 drugs have been incorporated in POx PMs. Among various stimuli, pH and temperature are the most widely studied for enhanced drug release at the site of action. Researchers are focusing on dual (pH and temperature) responsive PMs for controlled and improved drug release at the site of action. These dual responsive systems are mainly evaluated for cancer therapy as certain malignancies can cause a slight increase in temperature and a decrease in the extracellular pH around the tumor site. This review is a compilation of updated therapeutic applications of PMs, such as PMs that are based on Pluronics®, micelleplexes and Pox-based PMs in several biomedical applications.
Collapse
|
4
|
Lipid-Drug Conjugates and Nanoparticles for the Cutaneous Delivery of Cannabidiol. Int J Mol Sci 2022; 23:ijms23116165. [PMID: 35682847 PMCID: PMC9180973 DOI: 10.3390/ijms23116165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/23/2022] [Accepted: 05/30/2022] [Indexed: 02/01/2023] Open
Abstract
Lipid nanoparticles are currently used to deliver drugs to specific sites in the body, known as targeted therapy. Conjugates of lipids and drugs to produce drug-enriched phospholipid micelles have been proposed to increase the lipophilic character of drugs to overcome biological barriers. However, their applicability at the topical level is still minimal. Phospholipid micelles are amphiphilic colloidal systems of nanometric dimensions, composed of a lipophilic nucleus and a hydrophilic outer surface. They are currently used successfully as pharmaceutical vehicles for poorly water-soluble drugs. These micelles have high in vitro and in vivo stability and high biocompatibility. This review discusses the use of lipid-drug conjugates as biocompatible carriers for cutaneous application. This work provides a metadata analysis of publications concerning the conjugation of cannabidiol with lipids as a suitable approach and as a new delivery system for this drug.
Collapse
|
5
|
Kim J, She C, Potez M, Huang P, Wu Q, Prager BC, Qiu Z, Bao S, Rich JN, Liu JKC. Phage display targeting identifies EYA1 as a regulator of glioblastoma stem cell maintenance and proliferation. Stem Cells 2021; 39:853-865. [PMID: 33594762 PMCID: PMC10741052 DOI: 10.1002/stem.3355] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 01/20/2021] [Indexed: 11/06/2022]
Abstract
Glioblastoma (GBM) ranks among the most lethal of human malignancies with GBM stem cells (GSCs) that contribute to tumor growth and therapeutic resistance. Identification and isolation of GSCs continue to be a challenge, as definitive methods to purify these cells for study or targeting are lacking. Here, we leveraged orthogonal in vitro and in vivo phage display biopanning strategies to isolate a single peptide with GSC-specific binding properties. In silico analysis of this peptide led to the isolation of EYA1 (Eyes Absent 1), a tyrosine phosphatase and transcriptional coactivator. Validating the phage discovery methods, EYA1 was preferentially expressed in GSCs compared to differentiated tumor progeny. MYC is a central mediator of GSC maintenance but has been resistant to direct targeting strategies. Based on correlation and colocalization of EYA1 and MYC, we interrogated a possible interaction, revealing binding of EYA1 to MYC and loss of MYC expression upon targeting EYA1. Supporting a functional role for EYA1, targeting EYA1 expression decreased GSC proliferation, migration, and self-renewal in vitro and tumor growth in vivo. Collectively, our results suggest that phage display can identify novel therapeutic targets in stem-like tumor cells and that an EYA1-MYC axis represents a potential therapeutic paradigm for GBM.
Collapse
Affiliation(s)
- JongMyung Kim
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| | - Chunhua She
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| | - Marine Potez
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| | - Ping Huang
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Qiulian Wu
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037
| | - Briana C. Prager
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037
| | - Zhixin Qiu
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037
| | - Shideng Bao
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Jeremy N. Rich
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037
| | - James K. C. Liu
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612
- University of South Florida, Morsani College of Medicine, Tampa, FL
| |
Collapse
|
6
|
Gowsalya K, Yasothamani V, Vivek R. Emerging indocyanine green-integrated nanocarriers for multimodal cancer therapy: a review. NANOSCALE ADVANCES 2021; 3:3332-3352. [PMID: 36133722 PMCID: PMC9418715 DOI: 10.1039/d1na00059d] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/03/2021] [Indexed: 05/17/2023]
Abstract
Nanotechnology is a branch of science dealing with the development of new types of nanomaterials by several methods. In the biomedical field, nanotechnology is widely used in the form of nanotherapeutics. Therefore, the current biomedical research pays much attention to nanotechnology for the development of efficient cancer treatment. Indocyanine green (ICG) is a near-infrared tricarbocyanine dye approved by the Food and Drug Administration (FDA) for human clinical use. ICG is a biologically safe photosensitizer and it can kill tumor cells by producing singlet oxygen species and photothermal heat upon NIR irradiation. ICG has some limitations such as easy aggregation, rapid aqueous degradation, and a short half-life. To address these limitations, ICG is further formulated with nanoparticles. Therefore, ICG is integrated with organic nanomaterials (polymers, micelles, liposomes, dendrimers and protein), inorganic nanomaterials (magnetic, gold, mesoporous, calcium, and LDH based), and hybrid nanomaterials. The combination of ICG with nanomaterials provides highly efficient therapeutic effects. Nowadays, ICG is used for various biomedical applications, especially in cancer therapeutics. In this review, we mainly focus on ICG-based combined cancer nanotherapeutics for advanced cancer treatment.
Collapse
Affiliation(s)
- Karunanidhi Gowsalya
- Bio-Nano Therapeutics Research Laboratory, Cancer Research Program (CRP), School of Life Sciences, Department of Zoology, Bharathiar University Coimbatore-641 046 India
| | - Vellingiri Yasothamani
- Bio-Nano Therapeutics Research Laboratory, Cancer Research Program (CRP), School of Life Sciences, Department of Zoology, Bharathiar University Coimbatore-641 046 India
| | - Raju Vivek
- Bio-Nano Therapeutics Research Laboratory, Cancer Research Program (CRP), School of Life Sciences, Department of Zoology, Bharathiar University Coimbatore-641 046 India
| |
Collapse
|
7
|
Multifunctional polymeric micellar nanomedicine in the diagnosis and treatment of cancer. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 126:112186. [PMID: 34082985 DOI: 10.1016/j.msec.2021.112186] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 05/08/2021] [Accepted: 05/11/2021] [Indexed: 02/07/2023]
Abstract
Polymeric micelles are a prevalent topic of research for the past decade, especially concerning their fitting ability to deliver drug and diagnostic agents. This delivery system offers outstanding advantages, such as biocompatibility, high loading efficiency, water-solubility, and good stability in biological fluids, to name a few. The multifunctional polymeric micellar architect offers the added capability to adapt its surface to meet the looked-for clinical needs. This review cross-talks the recent reports, proof-of-concept studies, patents, and clinical trials that utilize polymeric micellar family architectures concerning cancer targeted delivery of anticancer drugs, gene therapeutics, and diagnostic agents. The manuscript also expounds on the underlying opportunities, allied challenges, and ways to resolve their bench-to-bedside translation for allied clinical applications.
Collapse
|
8
|
Alhakamy NA, Ahmed OAA, Fahmy UA, Md S. Development and In Vitro Evaluation of 2-Methoxyestradiol Loaded Polymeric Micelles for Enhancing Anticancer Activities in Prostate Cancer. Polymers (Basel) 2021; 13:884. [PMID: 33805675 PMCID: PMC7998642 DOI: 10.3390/polym13060884] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/07/2021] [Accepted: 03/10/2021] [Indexed: 12/15/2022] Open
Abstract
The present study aimed to formulate and optimize 2ME-loaded PMs (2ME-PMs) for enhancing the anticancer activity of 2ME in prostate cancer (PC). The 2ME-PMs were formulated using PEG-PLGA (PL), Tween 80 (TW80), and alpha-lipoic acid (ALA). The optimization was carried out using a Box-Behnken design with the PL, TW80, and ALA as the independent variables and particle size (PS) as the response. The formulation was optimized for the lowest possible PS, and the software suggested optimum formula with 100.282 mg, 2%, and 40 mg for PL, TW80, and ALA, respectively. The optimized PMs had spherical morphology with PS of 65.36 ± 2.2 nm, polydispersity index (PDI) of 0.273 ± 0.03, and entrapment efficiency of 65.23 ± 3.5%. The in vitro drug release was 76.3 ± 3.2% after 24 h. The cell line studies using PC-3 cells showed IC50 values of 18.75 and 54.41 µmol for 2ME-PM and 2ME, respectively. The estimation of tumor biomarkers was also carried out. The tumor biomarkers caspase-9 (17.38 ± 1.42 ng/mL), tumor protein P53 (p53) (1050.0 ± 40.9 pg/mL), nitric oxide (NO) (0.693 ± 0.03 pg/mL), interleukin-1β (IL-1β) (25.84 ± 2.23 pg/mL), nuclear factor kappa B (NF-kB) (0.719 ± 0.07 pg/mL), interleukin-6 (IL-6) (2.53 ± 0.16 folds), and cyclooxygenase-2 (COX-2) (3.04 ± 0.5 folds) were determined for 2ME-PMs and the results showed that these values changed significantly compared to those of 2ME. Overall, the results showed that the formulation of 2ME to 2ME-PMs enhances the anticancer effect. The exploration of the combined advantages of PEG, PLGA, ALA, and PMs in cancer therapy and the delivery of 2ME is the major importance of this research work. PEG reduces the elimination of 2ME, PLGA enhances 2ME loading, ALA has an inherent apoptotic effect, and PMs can efficiently target tumor cells.
Collapse
Affiliation(s)
- Nabil A. Alhakamy
- Department of Pharmaceutics, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (N.A.A.); (O.A.A.A.); (U.A.F.)
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Mohamed Saeed Tamer Chair for Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Osama A. A. Ahmed
- Department of Pharmaceutics, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (N.A.A.); (O.A.A.A.); (U.A.F.)
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Mohamed Saeed Tamer Chair for Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Usama A. Fahmy
- Department of Pharmaceutics, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (N.A.A.); (O.A.A.A.); (U.A.F.)
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Mohamed Saeed Tamer Chair for Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Shadab Md
- Department of Pharmaceutics, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (N.A.A.); (O.A.A.A.); (U.A.F.)
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Mohamed Saeed Tamer Chair for Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
9
|
Alven S, Aderibigbe BA. The Therapeutic Efficacy of Dendrimer and Micelle Formulations for Breast Cancer Treatment. Pharmaceutics 2020; 12:E1212. [PMID: 33333778 PMCID: PMC7765183 DOI: 10.3390/pharmaceutics12121212] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is among the most common types of cancer in women and it is the cause of a high rate of mortality globally. The use of anticancer drugs is the standard treatment approach used for this type of cancer. However, most of these drugs are limited by multi-drug resistance, drug toxicity, poor drug bioavailability, low water solubility, poor pharmacokinetics, etc. To overcome multi-drug resistance, combinations of two or more anticancer drugs are used. However, the combination of two or more anticancer drugs produce toxic side effects. Micelles and dendrimers are promising drug delivery systems that can overcome the limitations associated with the currently used anticancer drugs. They have the capability to overcome drug resistance, reduce drug toxicity, improve the drug solubility and bioavailability. Different classes of anticancer drugs have been loaded into micelles and dendrimers, resulting in targeted drug delivery, sustained drug release mechanism, increased cellular uptake, reduced toxic side effects of the loaded drugs with enhanced anticancer activity in vitro and in vivo. This review article reports the biological outcomes of dendrimers and micelles loaded with different known anticancer agents on breast cancer in vitro and in vivo.
Collapse
Affiliation(s)
| | - Blessing Atim Aderibigbe
- Department of Chemistry, University of Fort Hare, Alice Campus, Eastern Cape 5700, South Africa;
| |
Collapse
|
10
|
Oda CMR, Silva JDO, Fernandes RS, Braga AV, Machado RDR, Coelho MDM, Cassali GD, Reis DC, de Barros ALB, Leite EA. Encapsulating paclitaxel in polymeric nanomicelles increases antitumor activity and prevents peripheral neuropathy. Biomed Pharmacother 2020; 132:110864. [PMID: 33254426 DOI: 10.1016/j.biopha.2020.110864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/26/2020] [Accepted: 10/05/2020] [Indexed: 11/19/2022] Open
Abstract
Paclitaxel (PTX) has a great clinical significance as an antitumor drug, although several side effects are strongly dose-limiting. In this way, we prepared a PTX-loaded 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy (polyethylene glycol)-2000] polymeric micelles (PM/PTX) in an attempt to improve safety and effectiveness of conventional PTX formulation (CrEL/EtOH/PTX). In this study, we evaluated from both formulations: stability after dilution, hemocompatibility, cellular uptake, acute toxicity in healthy mice, antitumor activity, and toxicity after multiple-dose treatment. PM/PTX appeared to be more stable than CrEL/EtOH/PTX after dilution. PM/PTX did not exhibit hemolytic activity (values <1%), even at high concentrations. In vitro cellular uptake study indicated that polymeric micelles were able to deliver more PTX (5.8 %) than CrEL/EtOH (2.7 %) to 4T1 cells. In the acute toxicity evaluation in healthy mice, CrEL/EtOH/PTX (single dose of 20 mg/kg) induced peripheral neuropathy, which was not observed in PM/PTX group. Similar results were observed after tumor-bearing mice received a multiple-dose regimen (seven doses of 10 mg/kg). Worth mentioning, we also evaluated vehicles, and CrEL/EtOH alone was not capable of inducing neuropathic pain. Besides, PM/PTX exhibited a higher antitumor activity with an inhibition ratio approximately 1.5-fold higher than CrEL/EtOH/PTX group. This study suggested that PM/PTX is safer than CrEL/EtOH/PTX, and was able to improve the antitumor effectiveness in a 4T1 breast cancer model.
Collapse
Affiliation(s)
- Caroline Mari Ramos Oda
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil
| | - Juliana de Oliveira Silva
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil
| | - Renata Salgado Fernandes
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil
| | - Alysson Vinícius Braga
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil
| | - Renes de Resende Machado
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil
| | - Márcio de Matos Coelho
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil
| | - Geovanni Dantas Cassali
- Department of General Pathology, Biological Science Institute, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil
| | - Diego Carlos Reis
- Department of General Pathology, Biological Science Institute, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil
| | - André Luís Branco de Barros
- Department of Clinical and Toxicological Analyses, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil
| | - Elaine Amaral Leite
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
11
|
Xu H, Cao B, Li Y, Mao C. Phage nanofibers in nanomedicine: Biopanning for early diagnosis, targeted therapy, and proteomics analysis. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1623. [PMID: 32147974 DOI: 10.1002/wnan.1623] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/02/2020] [Accepted: 02/04/2020] [Indexed: 12/16/2022]
Abstract
Display of a peptide or protein of interest on the filamentous phage (also known as bacteriophage), a biological nanofiber, has opened a new route for disease diagnosis and therapy as well as proteomics. Earlier phage display was widely used in protein-protein or antigen-antibody studies. In recent years, its application in nanomedicine is becoming increasingly popular and encouraging. We aim to review the current status in this research direction. For better understanding, we start with a brief introduction of basic biology and structure of the filamentous phage. We present the principle of phage display and library construction method on the basis of the filamentous phage. We summarize the use of the phage displayed peptide library for selecting peptides with high affinity against cells or tissues. We then review the recent applications of the selected cell or tissue targeting peptides in developing new targeting probes and therapeutics to advance the early diagnosis and targeted therapy of different diseases in nanomedicine. We also discuss the integration of antibody phage display and modern proteomics in discovering new biomarkers or target proteins for disease diagnosis and therapy. Finally, we propose an outlook for further advancing the potential impact of phage display on future nanomedicine. This article is categorized under: Biology-Inspired Nanomaterials > Protein and Virus-Based Structures.
Collapse
Affiliation(s)
- Hong Xu
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma, USA
| | - Binrui Cao
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma, USA
| | - Yan Li
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma, USA
| | - Chuanbin Mao
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma, USA
| |
Collapse
|
12
|
Changalvaie B, Han S, Moaseri E, Scaletti F, Truong L, Caplan R, Cao A, Bouchard R, Truskett TM, Sokolov KV, Johnston KP. Indocyanine Green J Aggregates in Polymersomes for Near-Infrared Photoacoustic Imaging. ACS APPLIED MATERIALS & INTERFACES 2019; 11:46437-46450. [PMID: 31804795 DOI: 10.1021/acsami.9b14519] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Clinical translation of photoacoustic imaging (PAI) has been limited by the lack of near-infrared (NIR) contrast agents with low toxicity required for regulatory approval. Herein, J aggregates of indocyanine green (ICG) with strong NIR absorbance were encapsulated at high loadings within small 77 nm polymersomes (nanocapsules) composed of poly(lactide-co-glycolide-b-poly(ethylene glycol)) (PLGA-b-PEG) bilayers, thus enabling PAI of of breast and ovarian cancer cells with high specificity and a sensitivity at the level of ∼100 total cells. All of the major components of the polymersomes are FDA approved and used in the clinic. During formation of polymersomes with a water-in-oil-in-water double emulsion process, loss of ICG from the ICG J aggregates was minimized by coating them with a layer of branched polyethylenimine and by providing excess "sacrificial" ICG to adsorb at the oil-water interfaces. The encapsulated J aggregates were protected against dissociation by the polymersome shell for 24 h in 100% fetal bovine serum, after which the polymersomes biodegraded and the J aggregates dissociated to ICG monomers.
Collapse
Affiliation(s)
| | - Sangheon Han
- Department of Imaging Physics , MD Anderson Cancer Center , Houston , Texas 77030 , United States
- Department of Bioengineering , Rice University , Houston , Texas 77005 , United States
| | | | | | | | | | | | - Richard Bouchard
- Department of Imaging Physics , MD Anderson Cancer Center , Houston , Texas 77030 , United States
- Graduate School of Biomedical Sciences , The University of Texas MD Anderson Cancer Center , Houston , Texas 77030 , United States
| | | | - Konstantin V Sokolov
- Department of Imaging Physics , MD Anderson Cancer Center , Houston , Texas 77030 , United States
- Department of Bioengineering , Rice University , Houston , Texas 77005 , United States
| | | |
Collapse
|
13
|
Alhadlaq HA, Akhtar MJ, Ahamed M. Different cytotoxic and apoptotic responses of MCF-7 and HT1080 cells to MnO 2 nanoparticles are based on similar mode of action. Toxicology 2018; 411:71-80. [PMID: 30395893 DOI: 10.1016/j.tox.2018.10.023] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/17/2018] [Accepted: 10/31/2018] [Indexed: 01/08/2023]
Abstract
Manganese (IV) oxide nanoparticles (MnO2 NPs) are increasingly used in numerous applications. Multiple applications of MnO2 NPs, however, increase the human exposure and thus potential risk related to their toxicity. There is little information regarding the toxicity mechanisms of MnO2 NPs in human cells. In this study, we explored the toxic potential of MnO2 NPs in human breast cancer epithelial (MCF-7) and human fibrosarcoma epithelial (HT1080) cells in order to examine whether epithelial cells of different origins showed similar responses. Results demonstrated that MnO2 NPs induced cell viability reduction and membrane damage in both MCF-7 and HT1080 cells in a dose-dependent manner. MnO2 NPs were also found to induce pro-oxidants generation and antioxidants depletion in both cells. We further observed that MnO2 NPs induce apoptosis in both MCF-7 and HT1080 cells evident by altered regulation of apoptotic genes (p53, bax & bcl-2), cell cycle arrest and low mitochondrial membrane potential. Interestingly, we noticed that HT1080 cells were more susceptible to MnO2 NPs exposure than those of MCF-7 cells. This could be due to higher level of MnO2 NPs uptake into HT1080 cells as compared to MCF-7 cells. However, the mechanism of toxicity induced by MnO2 NPs in both MCF-7 and HT1080 cells was highly similar. This study warrants further research to delineate the underlying mechanisms of MnO2 NPs toxicity at in vivo level.
Collapse
Affiliation(s)
- Hisham A Alhadlaq
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh, Saudi Arabia; Department of Physics and Astronomy, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mohd Javed Akhtar
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh, Saudi Arabia
| | - Maqusood Ahamed
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
14
|
Oda CMR, de Barros ALB, Fernandes RS, Miranda SEM, Teixeira MX, Cardoso VN, Oliveira MC, Leite EA. Freeze-dried diethylenetriaminepentaacetic acid-functionalized polymeric micelles containing paclitaxel: A kit formulation for theranostic application in cancer. J Drug Deliv Sci Technol 2018. [DOI: 10.1016/j.jddst.2018.05.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
15
|
Landscape Phage: Evolution from Phage Display to Nanobiotechnology. Viruses 2018; 10:v10060311. [PMID: 29880747 PMCID: PMC6024655 DOI: 10.3390/v10060311] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/01/2018] [Accepted: 06/05/2018] [Indexed: 02/07/2023] Open
Abstract
The development of phage engineering technology has led to the construction of a novel type of phage display library-a collection of nanofiber materials with diverse molecular landscapes accommodated on the surface of phage particles. These new nanomaterials, called the "landscape phage", serve as a huge resource of diagnostic/detection probes and versatile construction materials for the preparation of phage-functionalized biosensors and phage-targeted nanomedicines. Landscape-phage-derived probes interact with biological threat agents and generate detectable signals as a part of robust and inexpensive molecular recognition interfaces introduced in mobile detection devices. The use of landscape-phage-based interfaces may greatly improve the sensitivity, selectivity, robustness, and longevity of these devices. In another area of bioengineering, landscape-phage technology has facilitated the development and testing of targeted nanomedicines. The development of high-throughput phage selection methods resulted in the discovery of a variety of cancer cell-associated phages and phage proteins demonstrating natural proficiency to self-assemble into various drug- and gene-targeting nanovehicles. The application of this new "phage-programmed-nanomedicines" concept led to the development of a number of cancer cell-targeting nanomedicine platforms, which demonstrated anticancer efficacy in both in vitro and in vivo experiments. This review was prepared to attract the attention of chemical scientists and bioengineers seeking to develop functionalized nanomaterials and use them in different areas of bioscience, medicine, and engineering.
Collapse
|
16
|
Liu P, Han L, Wang F, Li X, Petrenko VA, Liu A. Sensitive colorimetric immunoassay of Vibrio parahaemolyticus based on specific nonapeptide probe screening from a phage display library conjugated with MnO 2 nanosheets with peroxidase-like activity. NANOSCALE 2018; 10:2825-2833. [PMID: 29362753 DOI: 10.1039/c7nr06633c] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Pathogen detection continues to receive significant attention due to the harmful effects of pathogens on public health. Herein, specific nonapeptide-fusion proteins pVIII (pVIII fusion) were isolated from phage VQTVQIGSD (designated by the sequence of a fused foreign peptide), which was specifically screened from the f8/9 landscape phage library against Vibrio parahaemolyticus (V. parahaemolyticus) in a high-throughput way. The as-prepared V. parahaemolyticus-specific recognition element is cheaper and more available than antibodies. Further, a highly sensitive colorimetric immunoassay for V. parahaemolyticus was established using pVIII fusion as capture probes coupled with protein-templated MnO2 nanosheets (NSs) as signal probes. In the presence of a target bacterium, V. parahaemolyticus, a sandwich-type complex of pVIII fusion-V. parahaemolyticus-MnO2 NS@pVIII fusion was formed through specific recognition of pVIII fusion and V. parahaemolyticus. The signal probes (MnO2 NSs) could catalyze the reaction of 3,3',5,5'-tetramethylbenzidine and H2O2 to generate a colorimetric change. The proposed V. parahaemolyticus detection method demonstrated a wide detection range (20-104 colony-forming units (CFU) mL-1), low limit of detection (15 CFU mL-1), excellent selectivity, and high reliability for real marine samples, showing potential application in marine microbiological detection and control.
Collapse
Affiliation(s)
- Pei Liu
- Institute for Biosensing, and College of Chemistry & Chemical Engineering, Qingdao University, Qingdao 266071, China.
| | | | | | | | | | | |
Collapse
|
17
|
Synthesis and in vitro evaluation of a novel magnetic drug delivery system; proecological method for the preparation of CoFe2O4 nanostructures. J Mol Liq 2018. [DOI: 10.1016/j.molliq.2017.11.133] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
18
|
Qu X, Qiu P, Zhu Y, Yang M, Mao C. Guiding nanomaterials to tumors for breast cancer precision medicine: from tumor-targeting small-molecule discovery to targeted nanodrug delivery. NPG ASIA MATERIALS 2017; 9:e452. [PMID: 29657602 PMCID: PMC5898397 DOI: 10.1038/am.2017.196] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 08/27/2017] [Accepted: 09/21/2017] [Indexed: 05/03/2023]
Abstract
Precision medicine emphasizes patient-specific formulation for treatment of diseases, especially cancer. However, in targeted cancer treatment, because the expression level of tumor receptors in each patient varies even for the same type of cancer, the ligand/receptor-mediated approach does not seem promising for precision medicine. In this work, we demonstrated our strategy of using a phage display technique for breast cancer precision medicine. Using in vivo biopanning, we first selected an MCF-7 breast tumor-targeting peptide, then tested the effectiveness of the as-selected peptide in tumor homing and finally conjugated the peptide to a model photothermal drug, namely, gold nanorods, to achieve enhanced cancer killing efficacy. The peptides identified by the phage display technique can guide the drug to the tumors without the need to know the exact receptors on the tumor. This approach requires significantly less effort to explore patient-specific targeting molecules for precision medicine.
Collapse
Affiliation(s)
- Xuewei Qu
- Stephenson Life Sciences Research Center, Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, USA
| | - Penghe Qiu
- Stephenson Life Sciences Research Center, Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, USA
| | - Ye Zhu
- Stephenson Life Sciences Research Center, Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, USA
| | - Mingying Yang
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chuanbin Mao
- Stephenson Life Sciences Research Center, Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, USA
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
19
|
Valenzuela-Oses JK, García MC, Feitosa VA, Pachioni-Vasconcelos JA, Gomes-Filho SM, Lourenço FR, Cerize NN, Bassères DS, Rangel-Yagui CO. Development and characterization of miltefosine-loaded polymeric micelles for cancer treatment. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 81:327-333. [DOI: 10.1016/j.msec.2017.07.040] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/13/2017] [Accepted: 07/27/2017] [Indexed: 02/06/2023]
|
20
|
Autonomous self-navigating drug-delivery vehicles: from science fiction to reality. Ther Deliv 2017; 8:1063-1075. [DOI: 10.4155/tde-2017-0086] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Low efficacy of targeted nanomedicines in biological experiments enforced us to challenge the traditional concept of drug targeting and suggest a paradigm of ‘addressed self-navigating drug-delivery vehicles,’ in which affinity selection of targeting peptides and vasculature-directed in vivo phage screening is replaced by the migration selection, which explores ability of ‘promiscuous’ phages and their proteins to migrate through the tumor-surrounding cellular barriers, using a ‘hub and spoke’ delivery strategy, and penetrate into the tumor affecting the diverse tumor cell population. The ‘self-navigating’ drug-delivery paradigm can be used as a theoretical and technical platform in design of a novel generation of molecular medications and imaging probes for precise and personal medicine. [Formula: see text]
Collapse
|
21
|
Ju Z, Sun W. Drug delivery vectors based on filamentous bacteriophages and phage-mimetic nanoparticles. Drug Deliv 2017; 24:1898-1908. [PMID: 29191048 PMCID: PMC8241185 DOI: 10.1080/10717544.2017.1410259] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/16/2017] [Accepted: 11/23/2017] [Indexed: 12/11/2022] Open
Abstract
With the development of nanomedicine, a mass of nanocarriers have been exploited and utilized for targeted drug delivery, including liposomes, polymers, nanoparticles, viruses, and stem cells. Due to huge surface bearing capacity and flexible genetic engineering property, filamentous bacteriophage and phage-mimetic nanoparticles are attracting more and more attentions. As a rod-like bio-nanofiber without tropism to mammalian cells, filamentous phage can be easily loaded with drugs and directly delivered to the lesion location. In particular, chemical drugs can be conjugated on phage surface by chemical modification, and gene drugs can also be inserted into the genome of phage by recombinant DNA technology. Meanwhile, specific peptides/proteins displayed on the phage surface are able to conjugate with nanoparticles which will endow them specific-targeting and huge drug-loading capacity. Additionally, phage peptides/proteins can directly self-assemble into phage-mimetic nanoparticles which may be applied for self-navigating drug delivery nanovehicles. In this review, we summarize the production of phage particles, the identification of targeting peptides, and the recent applications of filamentous bacteriophages as well as their protein/peptide for targeting drug delivery in vitro and in vivo. The improvement of our understanding of filamentous bacteriophage and phage-mimetic nanoparticles will supply new tools for biotechnological approaches.
Collapse
Affiliation(s)
- Zhigang Ju
- Medicine College, Guiyang University of Chinese Medicine, Huaxi university town, Guiyang City, Guizhou Province, China
| | - Wei Sun
- Key Laboratory of Plant Physiology and Development Regulation, College of Life Science, Guizhou Normal University, Huaxi university town, Guiyang City, Guizhou Province, China
| |
Collapse
|
22
|
Gu W, Chen J, Patra P, Yang X, Gu Q, Wei L, Acker JP, Kong B. Nanoformulated water-soluble paclitaxel to enhance drug efficacy and reduce hemolysis side effect. J Biomater Appl 2017; 32:66-73. [PMID: 28504558 DOI: 10.1177/0885328217708458] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Surgery, chemotherapy, and radiotherapy are the three top cancer treatment modalities. Paclitaxel (PTX) is one of the most widely used chemotherapy drugs. However, its clinical applications have been significantly limited due to: (i) serious hemolysis effect of currently available commercial paclitaxel formulations and (ii) its water insolubility. An easy way to deliver paclitaxel by a new nanocarrier system using pluronic copolymers of P123/F68 and Sorbitan monopalmitate (Span 40) was reported in our previous research article. The characterization of the formulation and analysis of drug release and cellular uptake were also presented. In this article, we reported discoveries of our follow-up in vivo antitumor and in vitro hemolytic study discoveries. The experimental results showed that the nanoformulated PTX achieved much better tumor suppression performance while reducing hemolysis side effects. This newly formulated drug can significantly improve patient outcomes in cancer chemotherapy.
Collapse
Affiliation(s)
- Weiting Gu
- Qilu Hospital, Shandong University, Jinan, China
| | - Jie Chen
- Biomedical Engineering Department, University of Alberta, Edmonton, Alberta, Canada
| | - Prabir Patra
- Department of Biomedical Engineering, University of Bridgeport, Connecticut, USA
| | - Xiaoyan Yang
- Biomedical Engineering Department, University of Alberta, Edmonton, Alberta, Canada
| | - Quanrong Gu
- Biomedical Engineering Department, University of Alberta, Edmonton, Alberta, Canada
| | - Lingxuan Wei
- Qilu Hospital, Shandong University, Jinan, China
| | - Jason P Acker
- Centre for Innovation, Canadian Blood Service, Edmonton, Alberta, Canada
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Beihua Kong
- Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
23
|
Formulation of a Sustained Release Docetaxel Loaded Cockle Shell-Derived Calcium Carbonate Nanoparticles against Breast Cancer. Pharm Res 2017; 34:1193-1203. [DOI: 10.1007/s11095-017-2135-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 02/27/2017] [Indexed: 10/19/2022]
|
24
|
Oda CMR, Fernandes RS, de Araújo Lopes SC, de Oliveira MC, Cardoso VN, Santos DM, de Castro Pimenta AM, Malachias A, Paniago R, Townsend DM, Colletti PM, Rubello D, Alves RJ, de Barros ALB, Leite EA. Synthesis, characterization and radiolabeling of polymeric nano-micelles as a platform for tumor delivering. Biomed Pharmacother 2017; 89:268-275. [PMID: 28235689 DOI: 10.1016/j.biopha.2017.01.144] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 01/25/2017] [Indexed: 10/20/2022] Open
Abstract
The use of nanoparticles for diagnostic approaches leads to higher accumulation in the targeting tissue promoting a better signal-to-noise ratio and consequently, early tumor detection through scintigraphic techniques. Such approaches have inherent advantages, including the possibility of association with a variety of gamma-emitting radionuclides available, among them, Tecnethium-99m (99mTc). 99mTc is readily conjugated with nanoparticles using chelating agents, such as diethylenetriaminepentaacetic acid (DTPA). Leveraging this approach, we synthesized polymeric micelles (PM) consisting of 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethyleneglycol)-2000] (DSPE-mPEG2000) functionalized with DTPA for radiolabeling with 99mTc. Micelles made up of DSPE-mPEG2000 and DSPE-PEG2000-DTPA had a mean diameter of ∼10nm, as measured by DLS and SAXS techniques, and a zeta potential of -2.7±1.1mV. Radiolabeled micelles exhibited high radiochemical yields and stability. In vivo assays indicated long blood circulation time (456.3min). High uptake in liver, spleen and kidneys was observed in the biodistribution and imaging studies on healthy and tumor-bearing mice. In addition, a high tumor-to-muscle ratio was detected, which increased over time, showing accumulation of the PM in the tumor region. These findings indicate that this system is a promising platform for simultaneous delivery of therapeutic agents and diagnostic probes.
Collapse
Affiliation(s)
- Caroline Mari Ramos Oda
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Renata Salgado Fernandes
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Sávia Caldeira de Araújo Lopes
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Mônica Cristina de Oliveira
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Valbert Nascimento Cardoso
- Department of Clinical and Toxicological Analyses, Faculty of Pharmacy, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Daniel Moreira Santos
- Department of Biochemistry and Imunology, Biological Science Institute, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Adriano Monteiro de Castro Pimenta
- Department of Biochemistry and Imunology, Biological Science Institute, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Angelo Malachias
- Department of Physics, Exact Sciences Institute, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Rogério Paniago
- Department of Physics, Exact Sciences Institute, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Danyelle M Townsend
- Department of Drug Discovery and Pharmaceutical Sciences, Medical University of South Carolina, United States
| | - Patrick M Colletti
- Department of Nuclear Medicine, University of Southern California, Los Angeles, CA, United States
| | - Domenico Rubello
- Department of Nuclear Medicine, Imaging and Clinical Pathology, Santa Maria della Misericordia Hospital, Rovigo, Italy.
| | - Ricardo José Alves
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - André Luís Branco de Barros
- Department of Clinical and Toxicological Analyses, Faculty of Pharmacy, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Elaine Amaral Leite
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
25
|
Liu R, Li X, Xiao W, Lam KS. Tumor-targeting peptides from combinatorial libraries. Adv Drug Deliv Rev 2017; 110-111:13-37. [PMID: 27210583 DOI: 10.1016/j.addr.2016.05.009] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 05/10/2016] [Accepted: 05/11/2016] [Indexed: 02/07/2023]
Abstract
Cancer is one of the major and leading causes of death worldwide. Two of the greatest challenges in fighting cancer are early detection and effective treatments with no or minimum side effects. Widespread use of targeted therapies and molecular imaging in clinics requires high affinity, tumor-specific agents as effective targeting vehicles to deliver therapeutics and imaging probes to the primary or metastatic tumor sites. Combinatorial libraries such as phage-display and one-bead one-compound (OBOC) peptide libraries are powerful approaches in discovering tumor-targeting peptides. This review gives an overview of different combinatorial library technologies that have been used for the discovery of tumor-targeting peptides. Examples of tumor-targeting peptides identified from each combinatorial library method will be discussed. Published tumor-targeting peptide ligands and their applications will also be summarized by the combinatorial library methods and their corresponding binding receptors.
Collapse
Affiliation(s)
- Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA; University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Xiaocen Li
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA; University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Wenwu Xiao
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA; University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA; University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA; Division of Hematology & Oncology, Department of Internal Medicine, University of California Davis, Sacramento, CA 95817, USA
| |
Collapse
|
26
|
Gill KK, Kamal MM, Kaddoumi A, Nazzal S. EGFR targeted delivery of paclitaxel and parthenolide co-loaded in PEG-Phospholipid micelles enhance cytotoxicity and cellular uptake in non-small cell lung cancer cells. J Drug Deliv Sci Technol 2016. [DOI: 10.1016/j.jddst.2016.10.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
27
|
van der Zande M, Undas AK, Kramer E, Monopoli MP, Peters RJ, Garry D, Antunes Fernandes EC, Hendriksen PJ, Marvin HJP, Peijnenburg AA, Bouwmeester H. Different responses of Caco-2 and MCF-7 cells to silver nanoparticles are based on highly similar mechanisms of action. Nanotoxicology 2016; 10:1431-1441. [PMID: 27597447 DOI: 10.1080/17435390.2016.1225132] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The mode of action of silver nanoparticles (AgNPs) is suggested to be exerted through both Ag+ and AgNP dependent mechanisms. Ingestion is one of the major NP exposure routes, and potential effects are often studied using Caco-2 cells, a well-established model for the gut epithelium. MCF-7 cells are epithelial breast cancer cells with extensive well-characterized toxicogenomics profiles. In the present study, we aimed to gain a deeper understanding of the cellular molecular responses in Caco-2 and MCF-7 cells after AgNP exposure in order to evaluate whether epithelial cells derived from different tissues demonstrated similar responses. These insights could possibly reduce the size of cell panels for NP hazard identification screening purposes. AgNPs of 20, 30, 60, and 110 nm, and AgNO3 were exposed for 6 h and 24 h. AgNPs were shown to be taken up and dissolve intracellularly. Compared with MCF-7 cells, Caco-2 cells showed a higher sensitivity to AgNPs, slower gene expression kinetics and absence of NP size-dependent responses. However, on a molecular level, no significant differences were observed between the two cell types. Transcriptomic analysis showed that Ag(NP) exposure caused (oxidative) stress responses, possibly leading to cell death in both cell lines. There was no indication for effects specifically induced by AgNPs. Responses to AgNPs appeared to be induced by silver ions released from the AgNPs. In conclusion, differences in mRNA responses to AgNPs between Caco-2 and MCF-7 cells were mainly related to timing and magnitude, but not to a different underlying mechanism.
Collapse
Affiliation(s)
- Meike van der Zande
- a RIKILT - Wageningen University & Research Centre , Wageningen , The Netherlands and
| | - Anna K Undas
- a RIKILT - Wageningen University & Research Centre , Wageningen , The Netherlands and
| | - Evelien Kramer
- a RIKILT - Wageningen University & Research Centre , Wageningen , The Netherlands and
| | | | - Ruud J Peters
- a RIKILT - Wageningen University & Research Centre , Wageningen , The Netherlands and
| | - David Garry
- b University College Dublin , Dublin , Ireland
| | | | - Peter J Hendriksen
- a RIKILT - Wageningen University & Research Centre , Wageningen , The Netherlands and
| | - Hans J P Marvin
- a RIKILT - Wageningen University & Research Centre , Wageningen , The Netherlands and
| | - Ad A Peijnenburg
- a RIKILT - Wageningen University & Research Centre , Wageningen , The Netherlands and
| | - Hans Bouwmeester
- a RIKILT - Wageningen University & Research Centre , Wageningen , The Netherlands and
| |
Collapse
|
28
|
Double-targeted polymersomes and liposomes for multiple barrier crossing. Int J Pharm 2016; 511:946-56. [DOI: 10.1016/j.ijpharm.2016.08.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Revised: 07/31/2016] [Accepted: 08/02/2016] [Indexed: 01/09/2023]
|
29
|
Cerqueira-Coutinho C, Vidal LP, Pinto SR, Santos-Oliveira R. Drug metabolism: Comparison of biodistribution profile of holmium in three different compositions in healthy Wistar rats. Appl Radiat Isot 2016; 112:27-30. [DOI: 10.1016/j.apradiso.2016.02.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 02/11/2016] [Accepted: 02/23/2016] [Indexed: 12/30/2022]
|
30
|
Cerqueira-Coutinho C, Missailidis S, Alessandra-Perini J, Machado DE, Perini JA, Santos-Oliveira R. Comparison of biodistribution profile of monoclonal antibodies nanoparticles and aptamers in rats with breast cancer. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2016; 45:598-601. [DOI: 10.3109/21691401.2016.1163717] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
| | - Sotiris Missailidis
- Laboratory of Research in Pharmaceutical Sciences, Zona Oeste State University, Rio de Janeiro, Brazil
| | - Jéssica Alessandra-Perini
- Laboratory of Research in Pharmaceutical Sciences, Zona Oeste State University, Rio de Janeiro, Brazil
- Graduate Program in Public Health and Environment National School of Public Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Daniel Escorsim Machado
- Laboratory of Research in Pharmaceutical Sciences, Zona Oeste State University, Rio de Janeiro, Brazil
| | - Jamila Alessandra Perini
- Laboratory of Research in Pharmaceutical Sciences, Zona Oeste State University, Rio de Janeiro, Brazil
- Graduate Program in Public Health and Environment National School of Public Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- National Institute of Traumatology and Orthopedics, Rio de Janeiro, Brazil
| | - Ralph Santos-Oliveira
- Laboratory of Radiopharmacy and Nanoradiopharmaceuticals, Zona Oeste State University, Rio de Janeiro, Brazil
| |
Collapse
|
31
|
Gross AL, Gillespie JW, Petrenko VA. Promiscuous tumor targeting phage proteins. Protein Eng Des Sel 2016; 29:93-103. [PMID: 26764410 PMCID: PMC4753993 DOI: 10.1093/protein/gzv064] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 11/30/2015] [Accepted: 12/01/2015] [Indexed: 12/21/2022] Open
Abstract
Cancer cell-specific targeting ligands against numerous cancer cell lines have been selected previously and used as ligands for cell-specific delivery of chemotherapies and various nanomedicines. However, tumor heterogeneity is one recognized problem hampering clinical translation of targeted anti-cancer medicines. Therefore, a novel class of targeting ligands is required that recognize receptors expressed between a variety of cancer phenotypes, identified here as 'promiscuous' ligands. In this work, promiscuous phage fusion proteins were first identified by a novel selection scheme to enrich for pan-cancer cell binding abilities, as indicated by conserved structural motifs identified previously in other cancer types. Additionally, peptide sequences containing a combination of motifs were identified to modulate binding. A panel of phage fusion proteins was studied for their specificity and selectivity for lung and pancreatic cancer cells. Phage displaying the fusion peptides GSLEEVSTL or GEFDELMTM, the two predominate clones with greatest binding ability, were used to modify preformed, doxorubicin-loaded, liposomes. These modified liposomes increased cytotoxicity up to 8.1-fold in several cancer cell lines when compared with unmodified liposomal doxorubicin. Taken together, these data indicate that promiscuous phage proteins, selected against different cancer cell lines, can be used as targeting ligands for treatment of heterogeneous tumor populations.
Collapse
Affiliation(s)
- Amanda L Gross
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - James W Gillespie
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - Valery A Petrenko
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
32
|
Gillespie JW, Wei L, Petrenko VA. Selection of Lung Cancer-Specific Landscape Phage for Targeted Drug Delivery. Comb Chem High Throughput Screen 2016; 19:412-22. [PMID: 27095536 PMCID: PMC5066567 DOI: 10.2174/1386207319666160420141024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 03/01/2016] [Accepted: 03/30/2016] [Indexed: 12/24/2022]
Abstract
Cancer cell-specific diagnostic or therapeutic tools are commonly believed to significantly increase the success rate of cancer diagnosis and targeted therapies. To extend the repertoire of available cancer cell-specific phage fusion proteins and study their efficacy as navigating moieties, we used two landscape phage display libraries f8/8 and f8/9 displaying an 8- or 9-mer random peptide fusion to identify a panel of novel peptide families that are specific to Calu-3 cells. Using a phage capture assay, we showed that two of the selected phage clones, ANGRPSMT and VNGRAEAP (phage and their recombinant proteins are named by the sequence of the fusion peptide), are selective for the Calu-3 cell line in comparison to phenotypically normal lung epithelial cells and distribute into unique subcellular fractions.
Collapse
Affiliation(s)
| | | | - Valery A Petrenko
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA.
| |
Collapse
|
33
|
Henry KA, Arbabi-Ghahroudi M, Scott JK. Beyond phage display: non-traditional applications of the filamentous bacteriophage as a vaccine carrier, therapeutic biologic, and bioconjugation scaffold. Front Microbiol 2015; 6:755. [PMID: 26300850 PMCID: PMC4523942 DOI: 10.3389/fmicb.2015.00755] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 07/10/2015] [Indexed: 12/23/2022] Open
Abstract
For the past 25 years, phage display technology has been an invaluable tool for studies of protein-protein interactions. However, the inherent biological, biochemical, and biophysical properties of filamentous bacteriophage, as well as the ease of its genetic manipulation, also make it an attractive platform outside the traditional phage display canon. This review will focus on the unique properties of the filamentous bacteriophage and highlight its diverse applications in current research. Particular emphases are placed on: (i) the advantages of the phage as a vaccine carrier, including its high immunogenicity, relative antigenic simplicity and ability to activate a range of immune responses, (ii) the phage's potential as a prophylactic and therapeutic agent for infectious and chronic diseases, (iii) the regularity of the virion major coat protein lattice, which enables a variety of bioconjugation and surface chemistry applications, particularly in nanomaterials, and (iv) the phage's large population sizes and fast generation times, which make it an excellent model system for directed protein evolution. Despite their ubiquity in the biosphere, metagenomics work is just beginning to explore the ecology of filamentous and non-filamentous phage, and their role in the evolution of bacterial populations. Thus, the filamentous phage represents a robust, inexpensive, and versatile microorganism whose bioengineering applications continue to expand in new directions, although its limitations in some spheres impose obstacles to its widespread adoption and use.
Collapse
Affiliation(s)
- Kevin A. Henry
- Human Health Therapeutics Portfolio, National Research Council Canada, OttawaON, Canada
| | - Mehdi Arbabi-Ghahroudi
- Human Health Therapeutics Portfolio, National Research Council Canada, OttawaON, Canada
- School of Environmental Sciences, University of Guelph, GuelphON, Canada
- Department of Biology, Carleton University, OttawaON, Canada
| | - Jamie K. Scott
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BCCanada
- Faculty of Health Sciences, Simon Fraser University, BurnabyBC, Canada
| |
Collapse
|
34
|
Gill KK, Kaddoumi A, Nazzal S. PEG–lipid micelles as drug carriers: physiochemical attributes, formulation principles and biological implication. J Drug Target 2014; 23:222-31. [DOI: 10.3109/1061186x.2014.997735] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
35
|
Wang T, Yang S, Mei LA, Parmar CK, Gillespie JW, Praveen KP, Petrenko VA, Torchilin VP. Paclitaxel-loaded PEG-PE-based micellar nanopreparations targeted with tumor-specific landscape phage fusion protein enhance apoptosis and efficiently reduce tumors. Mol Cancer Ther 2014; 13:2864-75. [PMID: 25239936 PMCID: PMC4258532 DOI: 10.1158/1535-7163.mct-14-0052] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In an effort to improve the therapeutic index of cancer chemotherapy, we developed an advanced nanopreparation based on the combination of landscape phage display to obtain new targeting ligands with micellar nanoparticles for tumor targeting of water-insoluble neoplastic agents. With paclitaxel as a drug, this self-assembled nanopreparation composed of MCF-7-specific phage protein and polyethylene glycol-phosphatidylethanolamine (PEG-PE) micelles showed selective toxicity to target cancer cells rather than nontarget, non cancer cells in vitro. In vivo, the targeted phage micelles triggered a dramatic tumor reduction and extensive necrosis as a result of improved tumor delivery of paclitaxel. The enhanced anticancer effect was also verified by an enhanced apoptosis and reduced tumor cell proliferation following the treatment with the targeted micellar paclitaxel both in vitro and in vivo. The absence of hepatotoxicity and pathologic changes in tissue sections of vital organs, together with maintenance of overall health of mice following the treatment, further support its translational potential as an effective and safe chemotherapy for improved breast cancer treatment.
Collapse
Affiliation(s)
- Tao Wang
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, Massachusetts
| | - Shenghong Yang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Leslie A Mei
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, Massachusetts
| | - Chirag K Parmar
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, Massachusetts
| | | | - Kulkarni P Praveen
- Center for Translational Imaging, Northeastern University, Boston, Massachusetts
| | | | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, Massachusetts. King Abdulaziz University, Abdullah Sulayman, Jeddah, Saudi Arabia.
| |
Collapse
|
36
|
Chetprayoon P, Shima F, Matsusaki M, Akagi T, Akashi M. Sustainable Release of Paclitaxel from Biodegradable Poly(γ-glutamic acid) Nanoparticles for Treatment of Atherosclerosis. CHEM LETT 2014. [DOI: 10.1246/cl.140736] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Paninee Chetprayoon
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University
| | - Fumiaki Shima
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University
| | - Takami Akagi
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University
| | - Mitsuru Akashi
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University
| |
Collapse
|
37
|
Wang F, Liu P, Sun L, Li C, Petrenko VA, Liu A. Bio-mimetic nanostructure self-assembled from Au@Ag heterogeneous nanorods and phage fusion proteins for targeted tumor optical detection and photothermal therapy. Sci Rep 2014; 4:6808. [PMID: 25348392 PMCID: PMC4210868 DOI: 10.1038/srep06808] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 10/08/2014] [Indexed: 12/22/2022] Open
Abstract
Nanomaterials with near-infrared (NIR) absorption have been widely studied in cancer detection and photothermal therapy (PTT), while it remains a great challenge in targeting tumor efficiently with minimal side effects. Herein we report a novel multifunctional phage-mimetic nanostructure, which was prepared by layer-by-layer self-assembly of Au@Ag heterogenous nanorods (NRs) with rhodamine 6G, and specific pVIII fusion proteins. Au@Ag NRs, first being applied for PTT, exhibited excellent stability, cost-effectivity, biocompatibility and tunable NIR absorption. The fusion proteins were isolated from phage DDAGNRQP specifically selected from f8/8 landscape phage library against colorectal cancer cells in a high-throughput way. Considering the definite charge distribution and low molecular weight, phage fusion proteins were assembled on the negatively charged NR core by electrostatic interactions, exposing the N-terminus fused with DDAGNRQP peptide on the surface. The fluorescent images showed that assembled phage fusion proteins can direct the nanostructure into cancer cells. The nanostructure was more efficient than gold nanorods and silver nanotriangle-based photothermal agents and was capable of specifically ablating SW620 cells after 10 min illumination with an 808 nm laser in the light intensity of 4 W/cm(2). The prepared nanostructure would become an ideal reagent for simutaneously targeted optical imaging and PTT of tumor.
Collapse
Affiliation(s)
- Fei Wang
- Laboratory for Biosensing, Qingdao Institute of Bioenergy & Bioprocess Technology, and Key Laboratory of Biofuels, Chinese Academy of Sciences, 189 Songling Road, Qingdao 266101, China, and University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Pei Liu
- Laboratory for Biosensing, Qingdao Institute of Bioenergy & Bioprocess Technology, and Key Laboratory of Biofuels, Chinese Academy of Sciences, 189 Songling Road, Qingdao 266101, China, and University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Lin Sun
- Key Laboratory of Chemical Sensing & Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China
| | - Cuncheng Li
- Key Laboratory of Chemical Sensing & Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China
| | - Valery A. Petrenko
- Department of Pathobiology, Auburn University, 269 Greene Hall, Auburn, Alabama 36849-5519, United States
| | - Aihua Liu
- Laboratory for Biosensing, Qingdao Institute of Bioenergy & Bioprocess Technology, and Key Laboratory of Biofuels, Chinese Academy of Sciences, 189 Songling Road, Qingdao 266101, China, and University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| |
Collapse
|
38
|
Lu J, Zhao W, Huang Y, Liu H, Marquez R, Gibbs RB, Li J, Venkataramanan R, Xu L, Li S, Li S. Targeted delivery of Doxorubicin by folic acid-decorated dual functional nanocarrier. Mol Pharm 2014; 11:4164-78. [PMID: 25265550 PMCID: PMC4224520 DOI: 10.1021/mp500389v] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Doxorubicin (DOX) is one of the most commonly used antineoplastic agents, but its clinical application is oftentimes coupled with severe side effects. Selective delivery of DOX to tumors via nanosized drug carrier represents an attractive approach to this problem. Previously, we developed a dual functional nanomicellar carrier, PEG5K-embelin2 (PEG5K-EB2), which was able to deliver paclitaxel (PTX) selectively to tumors and to achieve an enhanced therapeutic effect. In the present study, we examined the utility of PEG5K-EB2 to deliver DOX to tumors. In addition, folic acid (FA) was coupled to the surface of the PEG5K-EB2 micelles (FA-PEG5K-EB2) to further improve the selective targetability of the system. DOX-loaded PEG5K-EB2 micelles were uniformly spherical particles with a diameter of approximately 20 nm. Incorporation of FA had minimal effect on the size of the particles. The DOX loading efficiency was as high as 91.7% and 93.5% for PEG5K-EB2 and FA-PEG5K-EB2, respectively. DOX formulated in PEG5K-EB2 micelles (with or without FA decoration) demonstrated sustained kinetics of DOX release compared to free DOX. FA-PEG5K-EB2 significantly facilitated the intracellular uptake of DOX over free DOX and PEGylated liposomal DOX (Doxil) in breast cancer cells, 4T1.2, and drug resistant cells, NCI/ADR-RES. P-gp ATPase assay showed that PEG5K-EB2 significantly inhibited the function of the P-gp efflux pump. The maximum tolerated dose of DOX-loaded PEG5K-EB2 micelles was 15 mg/kg in mice, which was 1.5-fold greater than that for free DOX. Pharmacokinetics (PK) and biodistribution studies showed that both types of DOX-loaded micelles, especially FA-PEG5K-EB2, were able to significantly prolong the blood circulation time of DOX and facilitate its preferential accumulation at the tumor tissue. Finally, DOX/PEG5K-EB2 mixed micelles demonstrated significantly enhanced tumor growth inhibitory effect with minimal toxicity in comparison to free DOX and Doxil and the antitumor activity was further enhanced after the decoration by folic acid. Our data suggest that FA-PEG5K-EB2 micelles represent a promising DOX delivery system that warrants more study in the future.
Collapse
Affiliation(s)
- Jianqin Lu
- Center for Pharmacogenetics, ‡Department of Pharmaceutical Sciences, School of Pharmacy, and §University of Pittsburgh Cancer Institute, University of Pittsburgh , Pittsburgh, Pennsylvania 15261, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Jhaveri AM, Torchilin VP. Multifunctional polymeric micelles for delivery of drugs and siRNA. Front Pharmacol 2014; 5:77. [PMID: 24795633 PMCID: PMC4007015 DOI: 10.3389/fphar.2014.00077] [Citation(s) in RCA: 276] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 03/31/2014] [Indexed: 12/18/2022] Open
Abstract
Polymeric micelles, self-assembling nano-constructs of amphiphilic copolymers with a core-shell structure have been used as versatile carriers for delivery of drugs as well as nucleic acids. They have gained immense popularity owing to a host of favorable properties including their capacity to effectively solubilize a variety of poorly soluble pharmaceutical agents, biocompatibility, longevity, high stability in vitro and in vivo and the ability to accumulate in pathological areas with compromised vasculature. Moreover, additional functions can be imparted to these micelles by engineering their surface with various ligands and cell-penetrating moieties to allow for specific targeting and intracellular accumulation, respectively, to load them with contrast agents to confer imaging capabilities, and incorporating stimuli-sensitive groups that allow drug release in response to small changes in the environment. Recently, there has been an increasing trend toward designing polymeric micelles which integrate a number of the above functions into a single carrier to give rise to “smart,” multifunctional polymeric micelles. Such multifunctional micelles can be envisaged as key to improving the efficacy of current treatments which have seen a steady increase not only in hydrophobic small molecules, but also in biologics including therapeutic genes, antibodies and small interfering RNA (siRNA). The purpose of this review is to highlight recent advances in the development of multifunctional polymeric micelles specifically for delivery of drugs and siRNA. In spite of the tremendous potential of siRNA, its translation into clinics has been a significant challenge because of physiological barriers to its effective delivery and the lack of safe, effective and clinically suitable vehicles. To that end, we also discuss the potential and suitability of multifunctional polymeric micelles, including lipid-based micelles, as promising vehicles for both siRNA and drugs.
Collapse
Affiliation(s)
- Aditi M Jhaveri
- Department of Pharmaceutical Sciences, Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University Boston, MA, USA
| | - Vladimir P Torchilin
- Department of Pharmaceutical Sciences, Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University Boston, MA, USA
| |
Collapse
|
40
|
Ren H, Gao C, Zhou L, Liu M, Xie C, Lu W. EGFR-targeted poly(ethylene glycol)-distearoylphosphatidylethanolamine micelle loaded with paclitaxel for laryngeal cancer: preparation, characterization and in vitro evaluation. Drug Deliv 2014; 22:785-94. [PMID: 24670093 DOI: 10.3109/10717544.2014.896057] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The objective of this study was to evaluate the potential of using polymeric micelles modified with a peptide (termed GE11) ligand of epidermal growth factor receptor as the targeted carriers to achieve increased accumulation in laryngeal cancer and enhanced intracellular delivery for the encapsulated anticancer drugs. Poly (ethylene glycol)-distearoylphosphatidylethanolamine (PEG-DSPE) micelles containing paclitaxel were prepared via film-hydration method followed by investigation of in vitro release of paclitaxel in phosphate-buffered saline. The average size of GE11-PEG-DSPE/paclitaxel micelle and mPEG-DSPE/paclitaxel were 35 ± 2.8 nm [the polydispersity index (PDI) = 0.207] and 28 ± 2.1 nm (PDI = 0.154), respectively. Micelles with or without GE11-modified had similar physicochemical properties. Transmission electron microscopy showed that the micelles were homogeneous and spherical in shape. Encapsulation efficiency and drug loading of the micelle were 74.11 ± 3.89% and 3.58 ± 2.82%, respectively. The in vitro targeting characteristic of GE11-modified micelles was investigated by observing the level of cellular uptake of fluorescent coumarin-6-loaded micelles on EGFR over-expressed human laryngeal cancer cell line Hep-2 and EGFR low-expressed human leukemic cell line U-937. Hep-2 cell proliferation was significantly inhibited by GE11-PEG-DSPE/paclitaxel micelle compared to mPEG-DSPE/paclitaxel micelle and Taxol in vitro. Our results suggested that GE11-PEG-DSPE micelle could be a promising strategy for enhancing paclitaxel's chemotherapeutic effects on EGFR over-expressed cancer cells.
Collapse
Affiliation(s)
- Henglei Ren
- a Department of Otolaryngology-Head and Neck Surgery , EENT Hospital, Fudan University , Shanghai , China and
| | - Chunli Gao
- a Department of Otolaryngology-Head and Neck Surgery , EENT Hospital, Fudan University , Shanghai , China and
| | - Liang Zhou
- a Department of Otolaryngology-Head and Neck Surgery , EENT Hospital, Fudan University , Shanghai , China and
| | - Min Liu
- b Department of Pharmaceutics , School of Pharmacy, Fudan University , Shanghai , China
| | - Cao Xie
- b Department of Pharmaceutics , School of Pharmacy, Fudan University , Shanghai , China
| | - Weiyue Lu
- b Department of Pharmaceutics , School of Pharmacy, Fudan University , Shanghai , China
| |
Collapse
|
41
|
Dong X, Qiu XC, Liu Q, Jia J. Bibliometric analysis of nanotechnology applied in oncology from 2002 to 2011. Tumour Biol 2014; 34:3273-8. [PMID: 23959469 DOI: 10.1007/s13277-013-1032-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 07/18/2013] [Indexed: 10/26/2022] Open
Abstract
Innovation in the last decade has endowed nanotechnology with an assortment of tools for drug delivery system, imaging, and sensing in cancer research. These rapidly emerging tools are indicative of a burgeoning field ready to expand into medical applications. The aim of this study is to analyze the applications of nanotechnology in oncology with bibliometric methods and evaluate development in this field. Literature search was performed using PubMed search engines with MeSH terms (all)--nanotechnology, nanomedicine, nanoparticle, nanocapsules, micellar systems, and oncology or cancer or neoplasms. Within 2,543 articles from 2002 to 2011 in over 50 medical magazines from over 30 countries, we did a series analysis on these articles' countries, keywords, and authors. Our results show that articles in nanotechnology in oncology are increasing year by year, especially in recent years. Quantity and quality of the articles are becoming more and influential. In the global research, the USA is leading in this field, accounting for half above of the whole articles, followed by countries like Japan, Germany, and France and also some emerging nations like China, in the second place, and India. Subjects like nanoparticles, tumor marker, and drug delivery are the common research focus. So, with more and more scientists' interests and attention drawn to this field, it is likely to make major breakthroughs in the coming years.
Collapse
|
42
|
Affiliation(s)
- Bethany Powell Gray
- Department of Internal Medicine and The Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8807, United States
| | - Kathlynn C. Brown
- Department of Internal Medicine and The Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8807, United States
| |
Collapse
|
43
|
Alhasan AH, Patel PC, Choi CHJ, Mirkin CA. Exosome encased spherical nucleic acid gold nanoparticle conjugates as potent microRNA regulation agents. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2014; 10:186-92. [PMID: 24106176 PMCID: PMC3947239 DOI: 10.1002/smll.201302143] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Indexed: 05/25/2023]
Abstract
Exosomes are a class of naturally occurring nanomaterials that play crucial roles in the protection and transport of endogenous macromolecules, such as microRNA and mRNA, over long distances. Intense effort is underway to exploit the use of exosomes to deliver synthetic therapeutics. Herein, transmission electron microscopy is used to show that when spherical nucleic acid (SNA) constructs are endocytosed into PC-3 prostate cancer cells, a small fraction of them (<1%) can be naturally sorted into exosomes. The exosome-encased SNAs are secreted into the extracellular environment from which they can be isolated and selectively re-introduced into the cell type from which they were derived. In the context of anti-miR21 experiments, the exosome-encased SNAs knockdown miR-21 target by approximately 50%. Similar knockdown of miR-21 by free SNAs requires a ≈3000-fold higher concentration.
Collapse
Affiliation(s)
- Ali H. Alhasan
- Department of Chemistry and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208-3113, USA
- Interdepartmental Biological Sciences Program, Northwestern University, 2205 Tech Drive, Evanston, IL 60208-3113, USA
| | - Pinal C. Patel
- AuraSense Therapeutics, LLC, 8045 Lamon Avenue, Suite 410, Skokie, IL 60077
| | - Chung Hang J. Choi
- Department of Chemistry and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208-3113, USA
| | - Chad A. Mirkin
- Department of Chemistry and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208-3113, USA
| |
Collapse
|
44
|
Petrenko VA, Jayanna PK. Phage protein-targeted cancer nanomedicines. FEBS Lett 2013; 588:341-9. [PMID: 24269681 DOI: 10.1016/j.febslet.2013.11.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 11/10/2013] [Indexed: 12/17/2022]
Abstract
Nanoencapsulation of anticancer drugs improves their therapeutic indices by virtue of the enhanced permeation and retention effect which achieves passive targeting of nanoparticles in tumors. This effect can be significantly enhanced by active targeting of nanovehicles to tumors. Numerous ligands have been proposed and used in various studies with peptides being considered attractive alternatives to antibodies. This is further reinforced by the availability of peptide phage display libraries which offer an unlimited reservoir of target-specific probes. In particular landscape phages with multivalent display of target-specific peptides which enable the phage particle itself to become a nanoplatform creates a paradigm for high throughput selection of nanoprobes setting the stage for personalized cancer management. Despite its promise, this conjugate of combinatorial chemistry and nanotechnology has not made a significant clinical impact in cancer management due to a lack of using robust processes that facilitate scale-up and manufacturing. To this end we proposed the use of phage fusion protein as the navigating modules of novel targeted nanomedicine platforms which are described in this review.
Collapse
Affiliation(s)
- V A Petrenko
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, AL 36849, United States.
| | - P K Jayanna
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, AL 36849, United States
| |
Collapse
|
45
|
Mura S, Zouhiri F, Lerondel S, Maksimenko A, Mougin J, Gueutin C, Brambilla D, Caron J, Sliwinski E, Lepape A, Desmaele D, Couvreur P. Novel isoprenoyl nanoassembled prodrug for paclitaxel delivery. Bioconjug Chem 2013; 24:1840-9. [PMID: 24134705 DOI: 10.1021/bc400210x] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
A new paclitaxel (Ptx) prodrug was designed by coupling a single terpene unit (MIP) to the hydroxyl group in position 2' of the drug molecule. Using a squalene derivative of polyethylene glycol (SQ-PEG) as surface active agent, the resulting bioconjugate (PtxMIP) self-assembled in water leading to the formation of stable nanoparticles (PtxMIP_SQ-PEG NPs) with an impressively high drug loading (82%). In vivo, the anticancer activity of this novel Ptx nanoassembled prodrug was compared to the conventional Cremophor-containing formulation (Taxol) on a murine model of breast cancer lung metastasis induced by intravenous injection of 4T1 tumor cells, genetically modified to stably express firefly luciferase. Cell growth was assessed noninvasively by bioluminescence imaging (BLI) which enabled monitoring tumor metastatic burden in the same animals. PtxMIP_SQ-PEG nanoparticles slowed metastatic spread and were better tolerated than the Cremophor-containing formulation (i.e., free drug), thus demonstrating the potential of terpene-based nanoassembled prodrugs in the improvement of the therapeutic index of Ptx in balb/c mice.
Collapse
Affiliation(s)
- Simona Mura
- Université Paris-Sud , Faculté de Pharmacie, 5 rue Jean-Baptiste Clément, 92296 Châtenay-Malabry cedex, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Multifunctional nanoparticles for targeted delivery of immune activating and cancer therapeutic agents. J Control Release 2013; 172:1020-34. [PMID: 24140748 DOI: 10.1016/j.jconrel.2013.10.012] [Citation(s) in RCA: 147] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 10/08/2013] [Accepted: 10/09/2013] [Indexed: 01/20/2023]
Abstract
Nanoparticles (NPs) have been extensively investigated for applications in both experimental and clinical settings to improve delivery efficiency of therapeutic and diagnostic agents. Most recently, novel multifunctional nanoparticles have attracted much attention because of their ability to carry diverse functionalities to achieve effective synergistic therapeutic treatments. Multifunctional NPs have been designed to co-deliver multiple components, target the delivery of drugs by surface functionalization, and realize therapy and diagnosis simultaneously. In this review, various materials of diverse chemistries for fabricating multifunctional NPs with distinctive architectures are discussed and compared. Recent progress involving multifunctional NPs for immune activation, anticancer drug delivery, and synergistic theranostics is the focus of this review. Overall, this comprehensive review demonstrates that multifunctional NPs have distinctive properties that make them highly suitable for targeted therapeutic delivery in these areas.
Collapse
|
47
|
Qin L, Zhang F, Lu X, Wei X, Wang J, Fang X, Si D, Wang Y, Zhang C, Yang R, Liu C, Liang W. Polymeric micelles for enhanced lymphatic drug delivery to treat metastatic tumors. J Control Release 2013; 171:133-42. [DOI: 10.1016/j.jconrel.2013.07.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 06/11/2013] [Accepted: 07/07/2013] [Indexed: 10/26/2022]
|
48
|
Wang T, Hartner WC, Gillespie JW, Praveen KP, Yang S, Mei LA, Petrenko VA, Torchilin VP. Enhanced tumor delivery and antitumor activity in vivo of liposomal doxorubicin modified with MCF-7-specific phage fusion protein. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2013; 10:421-30. [PMID: 24028893 DOI: 10.1016/j.nano.2013.08.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 08/13/2013] [Accepted: 08/18/2013] [Indexed: 12/16/2022]
Abstract
UNLABELLED A novel strategy to improve the therapeutic index of chemotherapy has been developed by the integration of nanotechnology with phage technique. The objective of this study was to combine phage display, identifying tumor-targeting ligands, with a liposomal nanocarrier for targeted delivery of doxorubicin. Following the proof of concept in cell-based experiments, this study focused on in vivo assessment of antitumor activity and potential side-effects of phage fusion protein-modified liposomal doxorubicin. MCF-7-targeted phage-Doxil treatments led to greater tumor remission and faster onset of antitumor activity than the treatments with non-targeted formulations. The enhanced anticancer effect induced by the targeted phage-Doxil correlated with an improved tumor accumulation of doxorubicin. Tumor sections consistently revealed enhanced apoptosis, reduced proliferation activity and extensive necrosis. Phage-Doxil-treated mice did not show any sign of hepatotoxicity and maintained overall health. Therefore, MCF-7-targeted phage-Doxil seems to be an active and tolerable chemotherapy for breast cancer treatment. FROM THE CLINICAL EDITOR The authors of this study successfully combined phage display with a liposomal nanocarrier for targeted delivery of doxorubicin using MCF-7-targeted phage-Doxil nanocarriers in a rodent model. The method demonstrated improved efficiency and reduced hepatotoxicity, paving the way to future clinical trials addressing breast cancer.
Collapse
Affiliation(s)
- Tao Wang
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA, USA
| | - William C Hartner
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA, USA
| | - James W Gillespie
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, AL, USA
| | - Kulkarni P Praveen
- Center for Translational Imaging, Northeastern University, Boston, MA, USA
| | - Shenghong Yang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Leslie A Mei
- Department of Biology, College of Science, Northeastern University, Boston, MA, USA
| | - Valery A Petrenko
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, AL, USA
| | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA, USA.
| |
Collapse
|
49
|
Patricio BFDC, Albernaz MDS, Patricio BFDC, Albernaz MDS, Santos-Oliveira R. Development of nanoradiopharmaceuticals by labeling polymer nanoparticles with tc-99m. World J Nucl Med 2013; 12:24-6. [PMID: 23961252 PMCID: PMC3745629 DOI: 10.4103/1450-1147.113946] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Nanomedicine is considered as the future of modern medicine. Hence, serious global efforts are being made for the development of nanopharmaceuticals. Among all the nanopharmaceuticals developed so far, radiopharmaceuticals constitute only a very small portion, as noted in the published literature. The procedures for development of nanoradiopharmaceuticals are complex. In this paper we discuss the results of a research directed at developing nanoradiopharmaceuticals based on three different types of nanopharmaceuticals as alternative drug delivery systems.
Collapse
Affiliation(s)
- B F de Carvalho Patricio
- Laboratory of Nanoradiopharmaceuticals, Nuclear Engineering Institute, Rio de Janeiro, RJ, Brazil
| | | | | | | | | |
Collapse
|
50
|
Wu L, Fang S, Shi S, Deng J, Liu B, Cai L. Hybrid Polypeptide Micelles Loading Indocyanine Green for Tumor Imaging and Photothermal Effect Study. Biomacromolecules 2013; 14:3027-33. [DOI: 10.1021/bm400839b] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Lei Wu
- Guangdong Key Laboratory of Nanomedicine, Institute
of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced
Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Shengtao Fang
- Guangdong Key Laboratory of Nanomedicine, Institute
of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced
Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Shuai Shi
- Guangdong Key Laboratory of Nanomedicine, Institute
of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced
Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Jizhe Deng
- Guangdong Key Laboratory of Nanomedicine, Institute
of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced
Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Bin Liu
- Guangdong Key Laboratory of Nanomedicine, Institute
of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced
Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, Institute
of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced
Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China
| |
Collapse
|