1
|
Nasr Azadani M, Abed A, Mirzaei SA, Mahjoubin-Tehran M, Hamblin M, Rahimian N, Mirzaei H. Nanoparticles in Cancer Theranostics: Focus on Gliomas. BIONANOSCIENCE 2025; 15:129. [DOI: 10.1007/s12668-024-01752-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/27/2024] [Indexed: 01/05/2025]
|
2
|
Reilly RM, Georgiou CJ, Brown MK, Cai Z. Radiation nanomedicines for cancer treatment: a scientific journey and view of the landscape. EJNMMI Radiopharm Chem 2024; 9:37. [PMID: 38703297 PMCID: PMC11069497 DOI: 10.1186/s41181-024-00266-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/22/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Radiation nanomedicines are nanoparticles labeled with radionuclides that emit α- or β-particles or Auger electrons for cancer treatment. We describe here our 15 years scientific journey studying locally-administered radiation nanomedicines for cancer treatment. We further present a view of the radiation nanomedicine landscape by reviewing research reported by other groups. MAIN BODY Gold nanoparticles were studied initially for radiosensitization of breast cancer to X-radiation therapy. These nanoparticles were labeled with 111In to assess their biodistribution after intratumoural vs. intravenous injection. Intravenous injection was limited by high liver and spleen uptake and low tumour uptake, while intratumoural injection provided high tumour uptake but low normal tissue uptake. Further, [111In]In-labeled gold nanoparticles modified with trastuzumab and injected iintratumourally exhibited strong tumour growth inhibition in mice with subcutaneous HER2-positive human breast cancer xenografts. In subsequent studies, strong tumour growth inhibition in mice was achieved without normal tissue toxicity in mice with human breast cancer xenografts injected intratumourally with gold nanoparticles labeled with β-particle emitting 177Lu and modified with panitumumab or trastuzumab to specifically bind EGFR or HER2, respectively. A nanoparticle depot (nanodepot) was designed to incorporate and deliver radiolabeled gold nanoparticles to tumours using brachytherapy needle insertion techniques. Treatment of mice with s.c. 4T1 murine mammary carcinoma tumours with a nanodepot incorporating [90Y]Y-labeled gold nanoparticles inserted into one tumour arrested tumour growth and caused an abscopal growth-inhibitory effect on a distant second tumour. Convection-enhanced delivery of [177Lu]Lu-AuNPs to orthotopic human glioblastoma multiforme (GBM) tumours in mice arrested tumour growth without normal tissue toxicity. Other groups have explored radiation nanomedicines for cancer treatment in preclinical animal tumour xenograft models using gold nanoparticles, liposomes, block copolymer micelles, dendrimers, carbon nanotubes, cellulose nanocrystals or iron oxide nanoparticles. These nanoparticles were labeled with radionuclides emitting Auger electrons (111In, 99mTc, 125I, 103Pd, 193mPt, 195mPt), β-particles (177Lu, 186Re, 188Re, 90Y, 198Au, 131I) or α-particles (225Ac, 213Bi, 212Pb, 211At, 223Ra). These studies employed intravenous or intratumoural injection or convection enhanced delivery. Local administration of these radiation nanomedicines was most effective and minimized normal tissue toxicity. CONCLUSIONS Radiation nanomedicines have shown great promise for treating cancer in preclinical studies. Local intratumoural administration avoids sequestration by the liver and spleen and is most effective for treating tumours, while minimizing normal tissue toxicity.
Collapse
Affiliation(s)
- Raymond M Reilly
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada.
- Princess Margaret Cancer Centre, Toronto, ON, Canada.
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada.
- Joint Department of Medical Imaging, University Health Network, Toronto, ON, Canada.
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada.
| | | | - Madeline K Brown
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Zhongli Cai
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
3
|
Gandhi S, Shende P. Anti-CD64 Antibody-Conjugated PLGA Nanoparticles Containing Methotrexate and Gold for Theranostics Application in Rheumatoid Arthritis. AAPS PharmSciTech 2024; 25:22. [PMID: 38267687 DOI: 10.1208/s12249-024-02733-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 12/21/2023] [Indexed: 01/26/2024] Open
Abstract
Rheumatoid arthritis, an autoimmune disorder, exerts a considerable effect on quality of life. The inflammatory mechanism involved in rheumatoid arthritis is not clearly known, and therefore the need to develop effective medicines as well as new methods for early detection is a challenge. In this study, we developed PLGA nanoparticles containing gold and methotrexate in core and anti-CD64 antibody conjugated to nanoparticle surface via coupling process. The nanoparticles were examined for their surface morphology using SEM and TEM. The mean particle size, zeta potential, and PDI values of nanoparticles were 413.6 ± 2.89 nm, -10.12 ± 2.12 mV, and 0.23 ± 0.04, respectively, indicating good stability and particle homogeneity. In vitro drug release revealed a controlled release pattern with 93.44 ± 1.60% up to 72 h of release in the presence of pH 5.8, indicating the influence of pH and NIR on drug release. In vivo results on adjuvant-induced arthritis on Wistar rats indicated that animals receiving antibody-conjugated nanoparticles showed improvement in clinical indices and arthritic score as compared to non-conjugated nanoparticles and free drugs. This innovative drug delivery system will be an excellent strategy to maximize therapeutic effectiveness by limiting dosage-related side effects.
Collapse
Affiliation(s)
- Sahil Gandhi
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai, India
| | - Pravin Shende
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai, India.
| |
Collapse
|
4
|
Tincu (Iurciuc) CE, Andrițoiu CV, Popa M, Ochiuz L. Recent Advancements and Strategies for Overcoming the Blood-Brain Barrier Using Albumin-Based Drug Delivery Systems to Treat Brain Cancer, with a Focus on Glioblastoma. Polymers (Basel) 2023; 15:3969. [PMID: 37836018 PMCID: PMC10575401 DOI: 10.3390/polym15193969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/23/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive malignant tumor, and the most prevalent primary malignant tumor affecting the brain and central nervous system. Recent research indicates that the genetic profile of GBM makes it resistant to drugs and radiation. However, the main obstacle in treating GBM is transporting drugs through the blood-brain barrier (BBB). Albumin is a versatile biomaterial for the synthesis of nanoparticles. The efficiency of albumin-based delivery systems is determined by their ability to improve tumor targeting and accumulation. In this review, we will discuss the prevalence of human glioblastoma and the currently adopted treatment, as well as the structure and some essential functions of the BBB, to transport drugs through this barrier. We will also mention some aspects related to the blood-tumor brain barrier (BTBB) that lead to poor treatment efficacy. The properties and structure of serum albumin were highlighted, such as its role in targeting brain tumors, as well as the progress made until now regarding the techniques for obtaining albumin nanoparticles and their functionalization, in order to overcome the BBB and treat cancer, especially human glioblastoma. The albumin drug delivery nanosystems mentioned in this paper have improved properties and can overcome the BBB to target brain tumors.
Collapse
Affiliation(s)
- Camelia-Elena Tincu (Iurciuc)
- Department of Natural and Synthetic Polymers, “Cristofor Simionescu” Faculty of Chemical Engineering and Protection of the Environment, “Gheorghe Asachi” Technical University, 73, Prof. Dimitrie Mangeron Street, 700050 Iasi, Romania;
- Department of Pharmaceutical Technology, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16, University Street, 700115 Iasi, Romania;
| | - Călin Vasile Andrițoiu
- Apitherapy Medical Center, Balanesti, Nr. 336-337, 217036 Gorj, Romania;
- Specialization of Nutrition and Dietetics, Faculty of Pharmacy, Vasile Goldis Western University of Arad, Liviu Rebreanu Street, 86, 310045 Arad, Romania
| | - Marcel Popa
- Department of Natural and Synthetic Polymers, “Cristofor Simionescu” Faculty of Chemical Engineering and Protection of the Environment, “Gheorghe Asachi” Technical University, 73, Prof. Dimitrie Mangeron Street, 700050 Iasi, Romania;
- Faculty of Dental Medicine, “Apollonia” University of Iasi, 11, Pacurari Street, 700511 Iasi, Romania
- Academy of Romanian Scientists, 3 Ilfov Street, 050045 Bucharest, Romania
| | - Lăcrămioara Ochiuz
- Department of Pharmaceutical Technology, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16, University Street, 700115 Iasi, Romania;
| |
Collapse
|
5
|
Tan KF, In LLA, Vijayaraj Kumar P. Surface Functionalization of Gold Nanoparticles for Targeting the Tumor Microenvironment to Improve Antitumor Efficiency. ACS APPLIED BIO MATERIALS 2023; 6:2944-2981. [PMID: 37435615 DOI: 10.1021/acsabm.3c00202] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
Gold nanoparticles (AuNPs) have undergone significant research for their use in the treatment of cancer. Numerous researchers have established their potent antitumor properties, which have greatly impacted the treatment of cancer. AuNPs have been used in four primary anticancer treatment modalities, namely radiation, photothermal therapy, photodynamic therapy, and chemotherapy. However, the ability of AuNPs to destroy cancer is lacking and can even harm healthy cells without the right direction to transport them to the tumor microenvironment. Consequently, a suitable targeting technique is needed. Based on the distinct features of the human tumor microenvironment, this review discusses four different targeting strategies that target the four key features of the tumor microenvironment, including abnormal vasculature, overexpression of specific receptors, an acidic microenvironment, and a hypoxic microenvironment, to direct surface-functionalized AuNPs to the tumor microenvironment and increase antitumor efficacies. In addition, some current completed or ongoing clinical trials of AuNPs will also be discussed below to further reinforce the concept of using AuNPs in anticancer therapy.
Collapse
Affiliation(s)
- Kin Fai Tan
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras, Kuala Lumpur 56000, Malaysia
| | - Lionel Lian Aun In
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | - Palanirajan Vijayaraj Kumar
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
6
|
Ashique S, Anand K. Radiolabelled Extracellular Vesicles as Imaging Modalities for Precise Targeted Drug Delivery. Pharmaceutics 2023; 15:pharmaceutics15051426. [PMID: 37242668 DOI: 10.3390/pharmaceutics15051426] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/20/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Extracellular vesicles (ECVs) have been abandoned as bio-inspired drug delivery systems (DDS) in the biomedical field. ECVs have a natural ability to cross over extracellular and intracellular barriers, making them superior to manufactured nanoparticles. Additionally, they have the ability to move beneficial biomolecules among far-flung bodily cells. These advantages and the accomplishment of favorable in vivo results convincingly show the value of ECVs in medication delivery. The usage of ECVs is constantly being improved, as it might be difficult to develop a consistent biochemical strategy that is in line with their useful clinical therapeutic uses. Extracellular vesicles (ECVs) have the potential to enhance the therapy of diseases. Imaging technologies, particularly radiolabelled imaging, have been exploited for non-invasive tracking to better understand their in vivo activity.
Collapse
Affiliation(s)
- Sumel Ashique
- Department of Pharmaceutics, Bharat Institute of Technology (BIT), School of Pharmacy, Meerut 250103, India
| | - Krishnan Anand
- Department of Chemical Pathology, School of Pathology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| |
Collapse
|
7
|
Chen M, Shou Z, Jin X, Chen Y. Emerging strategies in nanotechnology to treat respiratory tract infections: realizing current trends for future clinical perspectives. Drug Deliv 2022; 29:2442-2458. [PMID: 35892224 PMCID: PMC9341380 DOI: 10.1080/10717544.2022.2089294] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A boom in respiratory tract infection cases has inflicted a socio-economic burden on the healthcare system worldwide, especially in developing countries. Limited alternative therapeutic options have posed a major threat to human health. Nanotechnology has brought an immense breakthrough in the pharmaceutical industry in a jiffy. The vast applications of nanotechnology ranging from early diagnosis to treatment strategies are employed for respiratory tract infections. The research avenues explored a multitude of nanosystems for effective drug delivery to the target site and combating the issues laid through multidrug resistance and protective niches of the bacteria. In this review a brief introduction to respiratory diseases and multifaceted barriers imposed by bacterial infections are enlightened. The manuscript reviewed different nanosystems, i.e. liposomes, solid lipid nanoparticles, polymeric nanoparticles, dendrimers, nanogels, and metallic (gold and silver) which enhanced bactericidal effects, prevented biofilm formation, improved mucus penetration, and site-specific delivery. Moreover, most of the nanotechnology-based recent research is in a preclinical and clinical experimental stage and safety assessment is still challenging.
Collapse
Affiliation(s)
- Minhua Chen
- Emergency & Intensive Care Unit Center, Department of Intensive Care Unit, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Zhangxuan Shou
- Department of Pharmacy, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xue Jin
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Yingjun Chen
- Department of Infectious Diseases, People's Hospital of Tiantai County, Taizhou, China
| |
Collapse
|
8
|
Zhao Y, Feng Y, Li J, Cui C, Wang A, Fang J, Zhang Y, Ye S, Mao Q, Wang X, Shi H. Endogenous ROS-Mediated Covalent Immobilization of Gold Nanoparticles in Mitochondria: A “Sharp Sword” in Tumor Radiotherapy. ACS Chem Biol 2022; 17:2355-2365. [DOI: 10.1021/acschembio.2c00475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Yan Zhao
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Yali Feng
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Jiachen Li
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Chaoxiang Cui
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Anna Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Jing Fang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Yuqi Zhang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Shuyue Ye
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Qiulian Mao
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Xiaoyan Wang
- Department of Ultrasound, Heping Hospital Affiliated to Changzhi Medical College, Changzhi 046000, P. R. China
| | - Haibin Shi
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| |
Collapse
|
9
|
Roy I, Krishnan S, Kabashin AV, Zavestovskaya IN, Prasad PN. Transforming Nuclear Medicine with Nanoradiopharmaceuticals. ACS NANO 2022; 16:5036-5061. [PMID: 35294165 DOI: 10.1021/acsnano.1c10550] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Nuclear medicine is expected to make major advances in cancer diagnosis and therapy; tumor-targeted radiopharmaceuticals preferentially eradicate tumors while causing minimal damage to healthy tissues. The current scope of nuclear medicine can be significantly expanded by integration with nanomedicine, which utilizes nanoparticles for cancer diagnosis and therapy by capitalizing on the increased surface area-to-volume ratio, the passive/active targeting ability and high loading capacity, the greater interaction cross section with biological tissues, the rich surface properties of nanomaterials, the facile decoration of nanomaterials with a plethora of functionalities, and the potential for multiplexing several functionalities within one construct. This review provides a comprehensive discussion of nuclear nanomedicine using tumor-targeted nanoparticles for cancer radiation therapy with either pre-embedded radionuclides or nonradioactive materials which can be extrinsically triggered using various external nuclear particle sources to produce in situ radioactivity. In addition, it describes the prospect of combining nuclear nanomedicine with other modalities to enable synergistically enhanced combination therapies. The review also discusses advances in the fabrication of radionuclides as well as describes laser ablation technologies for producing nanoradiopharmaceuticals, which combine the ease of production with exceptional purity and rapid biodegradability, along with additional imaging or therapeutic functionalities. From a practical standpoint, these attributes of nanoradiopharmaceuticals may provide distinct advantages in diagnostic/therapeutic sensitivity and specificity, imaging resolution, and scalability of turnkey platforms. Coupling image-guided targeted radiation therapy with the possibility of in situ activation of nanomaterials as well as combining with other therapeutic modalities using a multifunctional nanoplatform could herald an era of exciting technological and therapeutic advances to radically transform the landscape of nuclear medicine. The review concludes with a discussion of current challenges and presents the authors' views on future opportunities to stimulate further research in this rewarding field of high societal impact.
Collapse
Affiliation(s)
- Indrajit Roy
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Sunil Krishnan
- Department of Radiation Oncology, Mayo Clinic Florida, Jacksonville, Florida 32224, United States
| | - Andrei V Kabashin
- Aix Marseille University, CNRS, LP3, Campus de Luminy - Case 917, 13288 Marseille, France
- MEPhI, Institute of Engineering Physics for Biomedicine (PhysBio), 115409 Moscow, Russia
| | - Irina N Zavestovskaya
- MEPhI, Institute of Engineering Physics for Biomedicine (PhysBio), 115409 Moscow, Russia
- Nuclear Physics and Astrophysics Department, LPI of RAS, 119991 Moscow, Russia
| | - Paras N Prasad
- MEPhI, Institute of Engineering Physics for Biomedicine (PhysBio), 115409 Moscow, Russia
- Department of Chemistry and Institute for Lasers, Photonics, and Biophotonics, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| |
Collapse
|
10
|
Nguyen PV, Hervé-Aubert K, Chourpa I, Allard-Vannier E. Active targeting strategy in nanomedicines using anti-EGFR ligands - A promising approach for cancer therapy and diagnosis. Int J Pharm 2021; 609:121134. [PMID: 34571073 DOI: 10.1016/j.ijpharm.2021.121134] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/09/2021] [Accepted: 09/22/2021] [Indexed: 12/21/2022]
Abstract
As active targeting using nanomedicines establishes itself as a strategy of choice in cancer therapy, several target receptors or ligands overexpressed in cancer cells have been identified and exploited. Among them, the epidermal growth factor receptor (EGFR) has emerged as one of the most promising oncomarkers for active targeting nanomedicines due to its overexpression and its active involvement in a wide range of cancer types. Henceforth, many novel EGFR-targeted nanomedicines for cancer therapy have been developed, giving encouraging results both in vitro and in vivo. This review focuses on different applications of such medicines in oncotherapy. On an important note, the contribution of EGFR-targeting ligands to final therapy efficacy along with current challenges and possible solutions or alternatives are emphasized.
Collapse
Affiliation(s)
- Phuoc Vinh Nguyen
- EA6295 Nanomédicaments et Nanosondes, Université de Tours, Tours, France
| | - Katel Hervé-Aubert
- EA6295 Nanomédicaments et Nanosondes, Université de Tours, Tours, France
| | - Igor Chourpa
- EA6295 Nanomédicaments et Nanosondes, Université de Tours, Tours, France
| | | |
Collapse
|
11
|
Anik MI, Mahmud N, Al Masud A, Hasan M. Gold nanoparticles (GNPs) in biomedical and clinical applications: A review. NANO SELECT 2021. [DOI: 10.1002/nano.202100255] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Muzahidul I. Anik
- Department of Chemical Engineering University of Rhode Island South Kingstown Rhode Island USA
| | - Niaz Mahmud
- Department of Biomedical Engineering Military Institute of Science and Technology Dhaka Bangladesh
| | - Abdullah Al Masud
- Department of Chemical Engineering Bangladesh University of Engineering and Technology Dhaka Bangladesh
| | - Maruf Hasan
- Department of Biomedical Engineering Military Institute of Science and Technology Dhaka Bangladesh
| |
Collapse
|
12
|
Evaluation of the Targeting and Therapeutic Efficiency of Anti-EGFR Functionalised Nanoparticles in Head and Neck Cancer Cells for Use in NIR-II Optical Window. Pharmaceutics 2021; 13:pharmaceutics13101651. [PMID: 34683944 PMCID: PMC8537270 DOI: 10.3390/pharmaceutics13101651] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/29/2021] [Accepted: 10/04/2021] [Indexed: 11/16/2022] Open
Abstract
Gold nanoparticles have been indicated for use in a diagnostic and/or therapeutic role in several cancer types. The use of gold nanorods (AuNRs) with a surface plasmon resonance (SPR) in the second near-infrared II (NIR-II) optical window promises deeper anatomical penetration through increased maximum permissible exposure and lower optical attenuation. In this study, the targeting and therapeutic efficiency of anti-epidermal growth factor receptor (EGFR)-antibody-functionalised AuNRs with an SPR at 1064 nm was evaluated in vitro. Four cell lines, KYSE-30, CAL-27, Hep-G2 and MCF-7, which either over- or under-expressed EGFR, were used once confirmed by flow cytometry and immunofluorescence. Optical microscopy demonstrated a significant difference (p < 0.0001) between targeted AuNRs (tAuNRs) and untargeted AuNRs (uAuNRs) in all four cancer cell lines. This study demonstrated that anti-EGFR functionalisation significantly increased the association of tAuNRs with each EGFR-positive cancer cell. Considering this, the MTT assay showed that photothermal therapy (PTT) significantly increased cancer cell death (>97%) in head and neck cancer cell line CAL-27 using tAuNRs but not uAuNRs, apoptosis being the major mechanism of cell death. This successful targeting and therapeutic outcome highlight the future use of tAuNRs for molecular photoacoustic imaging or tumour treatment through plasmonic photothermal therapy.
Collapse
|
13
|
Daems N, Michiels C, Lucas S, Baatout S, Aerts A. Gold nanoparticles meet medical radionuclides. Nucl Med Biol 2021; 100-101:61-90. [PMID: 34237502 DOI: 10.1016/j.nucmedbio.2021.06.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 05/25/2021] [Accepted: 06/04/2021] [Indexed: 12/15/2022]
Abstract
Thanks to their unique optical and physicochemical properties, gold nanoparticles have gained increased interest as radiosensitizing, photothermal therapy and optical imaging agents to enhance the effectiveness of cancer detection and therapy. Furthermore, their ability to carry multiple medically relevant radionuclides broadens their use to nuclear medicine SPECT and PET imaging as well as targeted radionuclide therapy. In this review, we discuss the radiolabeling process of gold nanoparticles and their use in (multimodal) nuclear medicine imaging to better understand their specific distribution, uptake and retention in different in vivo cancer models. In addition, radiolabeled gold nanoparticles enable image-guided therapy is reviewed as well as the enhancement of targeted radionuclide therapy and nanobrachytherapy through an increased dose deposition and radiosensitization, as demonstrated by multiple Monte Carlo studies and experimental in vitro and in vivo studies.
Collapse
Affiliation(s)
- Noami Daems
- Radiobiology Research Unit, Interdisciplinary Biosciences, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Boeretang 200, 2400 Mol, Belgium.
| | - Carine Michiels
- Unité de Recherche en Biologie Cellulaire-NARILIS, University of Namur, Rue de Bruxelles 61, 5000 Namur, Belgium
| | - Stéphane Lucas
- Laboratory of Analysis by Nuclear Reaction (LARN)-NARILIS, University of Namur, Rue de Bruxelles 61, 5000 Namur, Belgium
| | - Sarah Baatout
- Radiobiology Research Unit, Interdisciplinary Biosciences, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Boeretang 200, 2400 Mol, Belgium
| | - An Aerts
- Radiobiology Research Unit, Interdisciplinary Biosciences, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Boeretang 200, 2400 Mol, Belgium
| |
Collapse
|
14
|
Shende P, Gandhi S. Current strategies of radiopharmaceuticals in theranostic applications. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102594] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
15
|
Abdelbaky SB, Ibrahim MT, Samy H, Mohamed M, Mohamed H, Mustafa M, Abdelaziz MM, Forrest ML, Khalil IA. Cancer immunotherapy from biology to nanomedicine. J Control Release 2021; 336:410-432. [PMID: 34171445 DOI: 10.1016/j.jconrel.2021.06.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/16/2021] [Accepted: 06/19/2021] [Indexed: 02/06/2023]
Abstract
With the significant drawbacks of conventional cancer chemotherapeutics, cancer immunotherapy has demonstrated the ability to eradicate cancer cells and circumvent multidrug resistance (MDR) with fewer side effects than traditional cytotoxic therapies. Various immunotherapeutic agents have been investigated for that purpose including checkpoint inhibitors, cytokines, monoclonal antibodies and cancer vaccines. All these agents aid immune cells to recognize and engage tumor cells by acting on tumor-specific pathways, antigens or cellular targets. However, immunotherapeutics are still associated with some concerns such as off-target side effects and poor pharmacokinetics. Nanomedicine may resolve some limitations of current immunotherapeutics such as localizing delivery, controlling release and enhancing the pharmacokinetic profile. Herein, we discuss recent advances of immunotherapeutic agents with respect to their development and biological mechanisms of action, along with the advantages that nanomedicine strategies lend to immunotherapeutics by possibly improving therapeutic outcomes and minimizing side effects.
Collapse
Affiliation(s)
- Salma B Abdelbaky
- University of Science and Technology, Zewail City, 6th of October City, Giza 12578, Egypt; Molecular, Cellular, and Developmental Biology, College of Arts and Sciences, The Ohio State University, Columbus, OH 43210, United States of America
| | - Mayar Tarek Ibrahim
- University of Science and Technology, Zewail City, 6th of October City, Giza 12578, Egypt; Department of Chemistry, Center for Scientific Computation, Center for Drug Discovery, Design, and Delivery (CD4), Southern Methodist University, Dallas, Texas 75275, United States of America
| | - Hebatallah Samy
- University of Science and Technology, Zewail City, 6th of October City, Giza 12578, Egypt; Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Menatalla Mohamed
- University of Science and Technology, Zewail City, 6th of October City, Giza 12578, Egypt; Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario L8S 4M1, Canada
| | - Hebatallah Mohamed
- University of Science and Technology, Zewail City, 6th of October City, Giza 12578, Egypt; Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario L8S 4M1, Canada
| | - Mahmoud Mustafa
- University of Science and Technology, Zewail City, 6th of October City, Giza 12578, Egypt
| | - Moustafa M Abdelaziz
- Department of Bioengineering, School of Engineering, The University of Kansas, Lawrence, KS 66045, USA
| | - M Laird Forrest
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS 66047, USA.
| | - Islam A Khalil
- Department of Pharmaceutics, College of Pharmacy and Drug Manufacturing, Misr University of Science and Technology (MUST), 6th of October, Giza 12582, Egypt.
| |
Collapse
|
16
|
Nanomedicines functionalized with anti-EGFR ligands for active targeting in cancer therapy: Biological strategy, design and quality control. Int J Pharm 2021; 605:120795. [PMID: 34119579 DOI: 10.1016/j.ijpharm.2021.120795] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/28/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023]
Abstract
Recently, active targeting using nanocarriers with biological ligands has emerged as a novel strategy for improving the delivery of therapeutic and/or imaging agents to tumor cells. The presence of active targeting moieties on the surface of nanomedicines has been shown to play an important role in enhancing their accumulation in tumoral cells and tissues versus healthy ones. This property not only helps to increase the therapeutic index but also to minimize possible side effects of the designed nanocarriers. Since the overexpression of epidermal growth factor receptors (EGFR) is a common occurrence linked to the progression of a broad variety of cancers, the potential application of anti-EGFR immunotherapy and EGFR-targeting ligands in active targeting nanomedicines is getting increasing attention. Henceforth, the EGFR-targeted nanomedicines were extensively studied in vitro and in vivo but exhibited both satisfactory and disappointing results, depending on used protocols. This review is designed to give an overview of a variety of EGFR-targeting ligands available for nanomedicines, how to conjugate them onto the surface of nanoparticles, and the main analytical methods to confirm this successful conjugation.
Collapse
|
17
|
The potential clinical applications of radionuclide labeled/doped gold-based nanomaterials. RADIATION MEDICINE AND PROTECTION 2020. [DOI: 10.1016/j.radmp.2020.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
18
|
EGFR targeting for cancer therapy: Pharmacology and immunoconjugates with drugs and nanoparticles. Int J Pharm 2020; 592:120082. [PMID: 33188892 DOI: 10.1016/j.ijpharm.2020.120082] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/21/2020] [Accepted: 11/08/2020] [Indexed: 12/11/2022]
Abstract
The epidermal growth factor receptor (EGFR) belongs to the tyrosine kinase receptors family and is present in the epithelial cell membrane. Its endogenous activation occurs through the binding of different endogenous ligands, including the epidermal growth factor (EGF), leading to signaling cascades able to maintain normal cellular functions. Although involved in the development and maintenance of tissues in normal conditions, when EGFR is overexpressed, it stimulates the growth and progression of tumors, resulting in angiogenesis, invasion and metastasis, through some main cascades such as Ras/Raf/MAPK, PIK-3/AKT, PLC-PKC and STAT. Besides, considering the limitations of conventional chemotherapy that result in high toxicity and low tumor specificity, EGFR is currently considered an important target. As a result, several monoclonal antibodies are currently approved for use in cancer treatment, such as cetuximab (CTX), panitumumab, nimotuzumab, necitumumab and others are in clinical trials. Aiming to combine the chemotherapeutic agent toxicity and specific targeting to EGFR overexpressing tumor tissues, two main strategies will be discussed in this review: antibody-drug conjugates (ADCs) and antibody-nanoparticle conjugates (ANCs). Briefly, ADCs consist of antibodies covalently linked through a spacer to the cytotoxic drug. Upon administration, binding to EGFR and endocytosis, ADCs suffer chemical and enzymatic reactions leading to the release and accumulation of the drug. Instead, ANCs consist of nanotechnology-based formulations, such as lipid, polymeric and inorganic nanoparticles able to protect the drug against inactivation, allowing controlled release and also passive accumulation in tumor tissues by the enhanced permeability and retention effect (EPR). Furthermore, ANCs undergo active targeting through EGFR receptor-mediated endocytosis, leading to the formation of lysosomes and drug release into the cytosol. Herein, we will present and discuss some important aspects regarding EGFR structure, its role on internal signaling pathways and downregulation aspects. Then, considering that EGFR is a potential therapeutic target for cancer therapy, the monoclonal antibodies able to target this receptor will be presented and discussed. Finally, ADCs and ANCs state of the art will be reviewed and recent studies and clinical progresses will be highlighted. To the best of our knowledge, this is the first review paper to address specifically the EGFR target and its application on ADCs and ANCs.
Collapse
|
19
|
Kukkar D, Kukkar P, Kumar V, Hong J, Kim KH, Deep A. Recent advances in nanoscale materials for antibody-based cancer theranostics. Biosens Bioelectron 2020; 173:112787. [PMID: 33190049 DOI: 10.1016/j.bios.2020.112787] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/08/2020] [Accepted: 10/30/2020] [Indexed: 02/07/2023]
Abstract
The quest for advanced management tools or options of various cancers has been on the rise to efficiently reduce their risks of mortality without the demerits of conventional treatments (e.g., undesirable side effects of the medications on non-target tissues, non-targeted distribution, slow clearance of the administered drugs, and the development of drug resistance over the duration of therapy). In this context, nanomaterials-antibody conjugates can offer numerous advantages in the development of cancer theranostics over conventional delivery systems (e.g., highly specific and enhanced biodistribution of the drug in targeted tissues, prolonged systemic circulation, low toxicity, and minimally invasive molecular imaging). This review comprehensively discusses and evaluates recent advances in the application of nanomaterial-antibody bioconjugates for cancer theranostics for the further advancement in the control of diverse cancerous diseases. Further, discussion is expanded to cover the various challenges and limitations associated with the design and development of nanomaterial-antibody conjugates applicable towards better management of cancer.
Collapse
Affiliation(s)
- Deepak Kukkar
- Department of Nanotechnology, Sri Guru Granth Sahib World University, Fatehgarh Sahib, Punjab, 140406, India
| | - Preeti Kukkar
- Department of Chemistry, Mata Gujri College, Fatehgarh Sahib, Punjab, 140406, India
| | - Vanish Kumar
- National Agri-Food Biotechnology Institute (NABI), S.A.S. Nagar, Punjab, 140306, India
| | - Jongki Hong
- College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul, 02447, Republic of Korea
| | - Ki-Hyun Kim
- Department of Civil and Environmental Engineering, Hanyang University, Seoul, 04763 Republic of Korea.
| | - Akash Deep
- Central Scientific Instruments Organization (CSIR-CSIO), Sector 30 C, Chandigarh, 160030, India.
| |
Collapse
|
20
|
Goddard ZR, Marín MJ, Russell DA, Searcey M. Active targeting of gold nanoparticles as cancer therapeutics. Chem Soc Rev 2020; 49:8774-8789. [PMID: 33089858 DOI: 10.1039/d0cs01121e] [Citation(s) in RCA: 157] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Gold nanoparticles (AuNPs) are of increasing interest for their unique properties and their biocompatability, minimal toxicity, multivalency and size tunability make them exciting drug carriers. The functionalisaton of AuNPs with targeting moieties allows for their selective delivery to cancers, with antibodies, proteins, peptides, aptamers, carbohydrates and small molecules all exploited. Here, we review the recent advances in targeted-AuNPs for the treatment of cancer, with a particular focus on these classes of targeting ligands. We highlight the benefits and potential drawbacks of each ligand class and propose directions in which the field could grow.
Collapse
Affiliation(s)
- Zoë Rachael Goddard
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.
| | | | | | | |
Collapse
|
21
|
Zhang L, Mazouzi Y, Salmain M, Liedberg B, Boujday S. Antibody-Gold Nanoparticle Bioconjugates for Biosensors: Synthesis, Characterization and Selected Applications. Biosens Bioelectron 2020; 165:112370. [DOI: 10.1016/j.bios.2020.112370] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 01/22/2023]
|
22
|
Karpuz M, Silindir-Gunay M, Ozer AY, Ozturk SC, Yanik H, Tuncel M, Aydin C, Esendagli G. Diagnostic and therapeutic evaluation of folate-targeted paclitaxel and vinorelbine encapsulating theranostic liposomes for non-small cell lung cancer. Eur J Pharm Sci 2020; 156:105576. [PMID: 32987115 DOI: 10.1016/j.ejps.2020.105576] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 09/14/2020] [Accepted: 09/22/2020] [Indexed: 12/22/2022]
Abstract
NSCLC is the most common type of lung cancer. However, non-specific contrast agents, radiopharmaceuticals, and treatment methods are insufficient in early diagnosis and eradication of all tumor tissue. Therefore, the formulation of a novel, targeted, specific theranostic agents possess critical importance. In our previous study, paclitaxel and vinorelbine encapsulating, Tc-99m radiolabeled, folate targeted, nanosized liposomes were formulated and found promising due to characterization properties, high cellular uptake, and cytotoxicity. In this study, in vivo therapeutic and diagnostic efficacy of liposomal formulations were tested by biodistribution study, evaluation of tumor growth inhibition, and histopathologic examination after in vitro assays on LLC1 cells. Both actively and passively targeted liposomal formulations exhibited high cellular uptake, and co-drug encapsulating liposomes showed a greater cytotoxicity profiles than free drug combination in LLC1 cells. By the results of biodistribution studies performed in NSCLC tumor-bearing C57BL/6 mice, the uptake of radiolabeled, actively folate targeted, co-drug encapsulating liposomal formulation was found to be higher in tumor tissue when compared to non-actively targeted one. Also, more effective treatment was achieved by using folate-targeted, co-drug encapsulating liposomal formulation when compared to free drugs combination according to changes in tumor size of mice. Furthermore, liposomal formulations showed lower toxicity compared to free drug combinations in the toxicity study considering body weight. Moreover, according to the histopathological study, folate targeted, co-drug encapsulating liposomes not only inhibited the tumor growth effectively but also restricted the lung metastasis entirely.
Collapse
Affiliation(s)
- Merve Karpuz
- Department of Radiopharmacy, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey; Department of Radiopharmacy, Faculty of Pharmacy, Izmir KatipCelebi University, Izmir, Turkey
| | - Mine Silindir-Gunay
- Department of Radiopharmacy, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - A Yekta Ozer
- Department of Radiopharmacy, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey.
| | - Suleyman Can Ozturk
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Hamdullah Yanik
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Murat Tuncel
- Department of Nuclear Medicine, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Cisel Aydin
- Department of Pathology, Faculty of Medicine, Koc University, Istanbul, Turkey
| | - Gunes Esendagli
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| |
Collapse
|
23
|
Chen CC, Chang DY, Li JJ, Chan HW, Chen JT, Chang CH, Liu RS, Chang CA, Chen CL, Wang HE. Investigation of biodistribution and tissue penetration of PEGylated gold nanostars and their application for photothermal cancer treatment in tumor-bearing mice. J Mater Chem B 2020; 8:65-77. [DOI: 10.1039/c9tb02194a] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PEGylated gold nanostars (pAuNSs) and their radioactive surrogate (111In–DTPA–pAuNS), with unique physiochemical properties, are thought to be a promising agent for image-guided photothermal therapy (PTT).
Collapse
Affiliation(s)
- Chao-Cheng Chen
- National Yang-Ming University
- Department of Biomedical Imaging and Radiological Sciences
- Taipei
- Taiwan
| | - Deng-Yuan Chang
- National Yang-Ming University
- Department of Biomedical Imaging and Radiological Sciences
- Taipei
- Taiwan
| | - Jia-Je Li
- National Yang-Ming University
- Department of Biomedical Imaging and Radiological Sciences
- Taipei
- Taiwan
| | - Hui-Wen Chan
- National Yang-Ming University
- Department of Biomedical Imaging and Radiological Sciences
- Taipei
- Taiwan
| | | | | | - Ren-Shyan Liu
- National Yang-Ming University
- Department of Biomedical Imaging and Radiological Sciences
- Taipei
- Taiwan
- National Yang-Ming University
| | - C. Allen Chang
- National Yang-Ming University
- Department of Biomedical Imaging and Radiological Sciences
- Taipei
- Taiwan
- National Yang-Ming University
| | - Chuan-Lin Chen
- National Yang-Ming University
- Department of Biomedical Imaging and Radiological Sciences
- Taipei
- Taiwan
| | - Hsin-Ell Wang
- National Yang-Ming University
- Department of Biomedical Imaging and Radiological Sciences
- Taipei
- Taiwan
| |
Collapse
|
24
|
Sharifi M, Attar F, Saboury AA, Akhtari K, Hooshmand N, Hasan A, El-Sayed MA, Falahati M. Plasmonic gold nanoparticles: Optical manipulation, imaging, drug delivery and therapy. J Control Release 2019; 311-312:170-189. [PMID: 31472191 DOI: 10.1016/j.jconrel.2019.08.032] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 08/25/2019] [Accepted: 08/26/2019] [Indexed: 12/20/2022]
Abstract
Over the past two decades, the development of plasmonic nanoparticle (NPs), especially gold (Au) NPs, is being pursued more seriously in the medical fields such as imaging, drug delivery, and theranostic systems. However, there is no comprehensive review on the effect of the physical and chemical parameters of AuNPs on their plasmonic properties as well as the use of these unique characteristic in medical activities such as imaging and therapeutics. Therefore, in this literature the surface plasmon resonance (SPR) modeling of AuNPs was accurately captured toward precision medicine. Indeed, we investigated the importance of plasmonic properties of AuNPs in optical manipulation, imaging, drug delivery, and photothermal therapy (PTT) of cancerous cells based on their physicochemical properties. Finally, some challenges regarding the commercialization of AuNPs in future medicine such as, cytotoxicity, lack of standards for medical applications, high cost, and time-consuming process were discussed.
Collapse
Affiliation(s)
- Majid Sharifi
- Department of Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Animal Science, Faculty of Agriculture, University of Tabriz, Tabriz, Iran
| | - Farnoosh Attar
- Department of Biology, Faculty of Food Industry & Agriculture, Standard Research Institute, Karaj, Iran
| | - Ali Akbar Saboury
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Keivan Akhtari
- Department of Physics, University of Kurdistan, Sanandaj, Iran
| | - Nasrin Hooshmand
- Laser Dynamics Laboratory, School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, United States
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center, Qatar University, Doha 2713, Qatar.
| | - Mostafa A El-Sayed
- Laser Dynamics Laboratory, School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, United States.
| | - Mojtaba Falahati
- Department of Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
25
|
Gold Nanoparticles and Nanorods in Nuclear Medicine: A Mini Review. APPLIED SCIENCES-BASEL 2019. [DOI: 10.3390/app9163232] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
In the last decade, many innovative nanodrugs have been developed, as well as many nanoradiocompounds that show amazing features in nuclear imaging and/or radiometabolic therapy. Their potential uses offer a wide range of possibilities. It can be possible to develop nondimensional systems of existing radiopharmaceuticals or build engineered systems that combine a nanoparticle with the radiopharmaceutical, a tracer, and a target molecule, and still develop selective nanodetection systems. This review focuses on recent advances regarding the use of gold nanoparticles and nanorods in nuclear medicine. The up-to-date advancements will be shown concerning preparations with special attention on the dimensions and functionalizations that are most used to attain an enhanced performance of gold engineered nanomaterials. Many ideas are offered regarding recent in vitro and in vivo studies. Finally, the recent clinical trials and applications are discussed.
Collapse
|
26
|
Wu CY, Lin JJ, Chang WY, Hsieh CY, Wu CC, Chen HS, Hsu HJ, Yang AS, Hsu MH, Kuo WY. Development of theranostic active-targeting boron-containing gold nanoparticles for boron neutron capture therapy (BNCT). Colloids Surf B Biointerfaces 2019; 183:110387. [PMID: 31394419 DOI: 10.1016/j.colsurfb.2019.110387] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/08/2019] [Accepted: 07/23/2019] [Indexed: 12/13/2022]
Abstract
Successful boron neutron capture therapy (BNCT) requires sufficient and specific delivery of boron atoms to malignant cells. Gold nanoparticles (AuNPs) have been used as a useful delivery system for selectively releasing cytotoxic payloads in the tumor. However, studies demonstrating the in vivo distribution or pharmacokinetics of boron-containing AuNPs via noninvasive imaging are lacking. This study aims to develop theranostic AuNP-boron cage assemblies (B-AuNPs) and evaluate its feasibility for BNCT. The commercial citrate-coated AuNPs were subjected to PEGylation, azide addition, and carborane modification on the surface. To further arm the AuNPs, we conjugated anti-HER2 antibody (61 IgG) with boron-containing PEGylated AuNPs to form 61-B-AuNPs. The diameter and radiolabeling efficiency of boron-containing AuNPs were determined by dynamic light scattering (DLS) and radio thin-layer chromatography (radio TLC), respectively. Noninvasive single-photon emission computed tomography (SPECT)/computed tomography (CT) imaging was performed to determine the pharmacokinetics of radioiodinated AuNPs in N87 gastric cancer xenografts, and the content of boron in tumor and muscle was assessed by inductively coupled plasma mass spectrometry (ICP-MS). After the 3-step modification, the diameter of B-AuNPs increased by ˜25 nm, and antibody conjugation did not affect the diameter of AuNPs. Radioactive iodine (I-123) was introduced in AuNPs by Click chemistry under copper catalysis. The radiolabeling efficiency of 123I-B-AuNPs and 123I-61-B-AuNPs was approximately 60 ± 5%. After purification, the radiochemical purity (RCP) of these NPs was greater than 90%. MicroSPECT/CT imaging showed that the tumor-to-muscle (T/M) ratio of 123I-B-AuNP-injected mice reached 1.91 ± 0.17 at 12 h post-injection, while that of 123I-61-B-AuNP-injected mice was 12.02 ± 0.94. However, the increased uptake of AuNPs by the thyroid was observed at 36 h after the administration of 123I-61-B-AuNPs, indicating antibody-mediated phagocytosis. The T/M ratio, assessed by ICP-MS, of B-AuNP- and 61-B-AuNP-injected mice was 4.91 ± 2.75 and 41.05 ± 11.15, respectively. We successfully developed detectable HER2-targeting boron-containing AuNPs with high RCP and an acceptable yield. Noninvasive imaging could be a valuable tool for the noninvasive determination of the pharmacokinetics of AuNPs and measurement of boron concentration in the tumor.
Collapse
Affiliation(s)
- Chun-Yi Wu
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan; Master Program for Biomedical Engineering, China Medical University, Taichung, Taiwan.
| | - Jia-Jia Lin
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan
| | - Wen-Yi Chang
- Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Cheng-Ying Hsieh
- Department of Chemistry, National Changhua University of Education, Changhua, Taiwan
| | - Chin-Ching Wu
- Department of Public Health, China Medical University, Taichung, Taiwan
| | - Hong-Sen Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Hung-Ju Hsu
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - An-Suei Yang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Ming-Hua Hsu
- Department of Chemistry, National Changhua University of Education, Changhua, Taiwan
| | - Wei-Ying Kuo
- Genomics Research Center, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
27
|
Thambiraj S, Shruthi S, Vijayalakshmi R, Ravi Shankaran D. Evaluation of cytotoxic activity of docetaxel loaded gold nanoparticles for lung cancer drug delivery. Cancer Treat Res Commun 2019; 21:100157. [PMID: 31310876 DOI: 10.1016/j.ctarc.2019.100157] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 07/02/2019] [Accepted: 07/09/2019] [Indexed: 11/19/2022]
Abstract
The effective use of the gold nanoparticle (AuNPs) conjugated drugs for targeted drug delivery applications is one of the most promising research areas in the field of cancer. Herein, we aimed to establish a nano drug conjugated with docetaxel as a possible therapy option. Gold nanoparticles were synthesized by chemical reduction method. This is followed by the conjugation with an anticancer drug, docetaxel (Dtx) by a non-covalent method and folic acid (FA) was conjugated by a covalent method. The physicochemical characteristics of the synthesized AuNPs, Dtx and FA were studied by various analytical techniques such as UV-vis spectroscopy (UV-vis), Fourier transform infrared spectroscopy (FTIR), X-ray diffraction (XRD), Raman spectroscopy, X-ray photoelectron spectroscopy (XPS), field emission scanning electron microscope (FE-SEM) high-resolution transmission electron microscope (HR-TEM) and energy dispersive X-ray spectroscopy (EDS). The surface morphology and microstructure of the synthesized AuNPs and gold conjugates (AuNPs-Dtx-FA) were examined by FESEM and HR-TEM. The average size of the spherical shaped AuNPs was observed in the range of 5-18 nm. XPS and EDS spectra were examined the oxidation state and chemical composition of the synthesized nanoparticles. The cytotoxicity of the synthesized AuNPs nano-conjugates was evaluated by in-vitro studies against lung cancer cell line (H520). The chemical reduction method followed here in the development of AuNPs is a simple and one-step process, which exhibits the excellent binding specificity, biocompatibility and feasible for the large scale up process of the AuNPs.
Collapse
Affiliation(s)
- S Thambiraj
- Nano-Bio Materials and Sensors Laboratory, National Centre for Nanoscience and Nanotechnology, University of Madras, Guindy Campus, Chennai 600025, India
| | - S Shruthi
- Department of Preventive Oncology, Cancer Institute (WIA), Adyar, Chennai 600020, India
| | - R Vijayalakshmi
- Department of Preventive Oncology, Cancer Institute (WIA), Adyar, Chennai 600020, India
| | - D Ravi Shankaran
- Nano-Bio Materials and Sensors Laboratory, National Centre for Nanoscience and Nanotechnology, University of Madras, Guindy Campus, Chennai 600025, India.
| |
Collapse
|
28
|
Gonciar D, Mocan T, Matea CT, Zdrehus C, Mosteanu O, Mocan L, Pop T. Nanotechnology in metastatic cancer treatment: Current Achievements and Future Research Trends. J Cancer 2019; 10:1358-1369. [PMID: 31031845 PMCID: PMC6485233 DOI: 10.7150/jca.28394] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 01/04/2019] [Indexed: 12/20/2022] Open
Abstract
The systemic spread of malignant cells from a primary site, a process termed metastasis represents a global challenge in cancer treatment. There is a real need to develop novel therapy strategies and nanomedicine may have remarkable and valuable contribution through specific and selective delivery of chemotherapeutic agents, through its intrinsic cytotoxic activity or through imaging applications, appealing also in the context of cancer personalized therapy. This review is focused on the applications of nanoparticles in the treatment of metastatic cancer, particularly on the possible effect on cell stabilization, growth inhibition, eventual interaction with adhesion molecules and antiangiogenic effect.
Collapse
Affiliation(s)
- Diana Gonciar
- Third Surgery Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy Cluj-Napoca, Romania.,Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology "Prof.Dr. Octavian Fodor" Cluj-Napoca , Romania
| | - Teodora Mocan
- Physiology Department, "Iuliu Hatieganu" University of Medicine and Pharmacy Cluj-Napoca, Romania.,Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology "Prof.Dr. Octavian Fodor" Cluj-Napoca , Romania
| | - Cristian Tudor Matea
- Third Surgery Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy Cluj-Napoca, Romania.,Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology "Prof.Dr. Octavian Fodor" Cluj-Napoca , Romania
| | - Claudiu Zdrehus
- Third Surgery Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy Cluj-Napoca, Romania.,Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology "Prof.Dr. Octavian Fodor" Cluj-Napoca , Romania
| | - Ofelia Mosteanu
- Third Surgery Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy Cluj-Napoca, Romania.,Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology "Prof.Dr. Octavian Fodor" Cluj-Napoca , Romania
| | - Lucian Mocan
- Third Surgery Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy Cluj-Napoca, Romania.,Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology "Prof.Dr. Octavian Fodor" Cluj-Napoca , Romania
| | - Teodora Pop
- Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology "Prof.Dr. Octavian Fodor" Cluj-Napoca , Romania
| |
Collapse
|
29
|
Li S, Bouchy S, Penninckx S, Marega R, Fichera O, Gallez B, Feron O, Martinive P, Heuskin AC, Michiels C, Lucas S. Antibody-functionalized gold nanoparticles as tumor-targeting radiosensitizers for proton therapy. Nanomedicine (Lond) 2019; 14:317-333. [PMID: 30675822 DOI: 10.2217/nnm-2018-0161] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
AIM This study aimed at developing antibody-functionalized gold nanoparticles (AuNPs) to selectively target cancer cells and probing their potential radiosensitizing effects under proton irradiation. MATERIALS & METHODS AuNPs were conjugated with cetuximab (Ctxb-AuNPs). Ctxb-AuNP uptake was evaluated by transmission electron microscopy and atomic absorption spectroscopy. Radioenhancing effect was assessed using conventional clonogenic assay. RESULTS & CONCLUSION Ctxb-AuNPs specifically bound to and accumulated in EGFR-overexpressing A431 cells, compared with EGFR-negative MDA-MB-453 cells. Ctxb-AuNPs enhanced the effect of proton irradiation in A431 cells but not in MDA-MB-453 cells. These data indicate, for the first time, that combining enhanced uptake by specific targeting and radioenhancing effect, using conjugated AuNPs, is a promising strategy to increase cell killing by protontherapy.
Collapse
Affiliation(s)
- Sha Li
- Research Center for the Physics of Matter & Radiation (PMR-LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, B-5000 Namur, Belgium
| | - Sandra Bouchy
- Unité de Recherche en Biologie Cellulaire (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, B-5000 Namur, Belgium
| | - Sebastien Penninckx
- Research Center for the Physics of Matter & Radiation (PMR-LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, B-5000 Namur, Belgium
| | - Riccardo Marega
- Research Center for the Physics of Matter & Radiation (PMR-LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, B-5000 Namur, Belgium
| | - Ornella Fichera
- Research Center for the Physics of Matter & Radiation (PMR-LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, B-5000 Namur, Belgium
| | - Bernard Gallez
- Biomedical Magnetic Resonance Group (REMA), Louvain Drug Research Institute, Université Catholique de Louvain, B-1200 Woluwé, Saint Lambert, Belgium
| | - Olivier Feron
- Pole of Pharmacology & Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCL (Université Catholique de Louvain), B-1200 Brussels, Belgium
| | - Philippe Martinive
- Department of Radiotherapy & Oncology, CHU & University of Liège, B-4000 Liège, Belgium
| | - Anne-Catherine Heuskin
- Research Center for the Physics of Matter & Radiation (PMR-LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, B-5000 Namur, Belgium
| | - Carine Michiels
- Unité de Recherche en Biologie Cellulaire (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, B-5000 Namur, Belgium
| | - Stéphane Lucas
- Research Center for the Physics of Matter & Radiation (PMR-LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, B-5000 Namur, Belgium
| |
Collapse
|
30
|
Patout M, Guisier F, Brune X, Bohn P, Romieu A, Sarafan-Vasseur N, Sesboüé R, Renard PY, Thiberville L, Salaün M. Real-time molecular optical micro-imaging of EGFR mutations using a fluorescent erlotinib based tracer. BMC Pulm Med 2019; 19:3. [PMID: 30612556 PMCID: PMC6322267 DOI: 10.1186/s12890-018-0760-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 12/06/2018] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND EGFR mutations are routinely explored in lung adenocarcinoma by sequencing tumoral DNA. The aim of this study was to evaluate a fluorescent-labelled erlotinib based theranostic agent for the molecular imaging of mutated EGFR tumours in vitro and ex vivo using a mice xenograft model and fibred confocal fluorescence microscopy (FCFM). METHODS The fluorescent tracer was synthesized in our laboratory by addition of fluorescein to an erlotinib molecule. Three human adenocarcinoma cell lines with mutated EGFR (HCC827, H1975 and H1650) and one with wild-type EGFR (A549) were xenografted on 35 Nude mice. MTT viability assay was performed after exposure to our tracer. In vitro imaging was performed at 1 μM tracer solution, and ex vivo imaging was performed on fresh tumours excised from mice and exposed to a 1 μM tracer solution in PBS for 1 h. Real-time molecular imaging was performed using FCFM and median fluorescence intensity (MFI) was recorded for each experiment. RESULTS MTT viability assay confirmed that addition of fluorescein to erlotinib did not suppress the cytotoxic of erlotinib on tumoral cells. In vitro FCFM imaging showed that our tracer was able to distinguish cell lines with mutated EGFR from those lines with wild-type EGFR (p < 0.001). Ex vivo FCFM imaging of xenografts with mutated EGFR had a significantly higher MFI than wild-type (p < 0.001). At a cut-off value of 354 Arbitrary Units, MFI of our tracer had a sensitivity of 100% and a specificity of 96.3% for identifying mutated EGFR tumours. CONCLUSION Real time molecular imaging using fluorescent erlotinib is able to identify ex vivo tumours with EGFR mutations.
Collapse
Affiliation(s)
- Maxime Patout
- Rouen University Hospital, Clinique Pneumologique & CIC INSERM U 1404, F-76000, Rouen, France. .,Normandie University, UNIROUEN, LITIS, Quant.I.F - EA 4108, F-76000, Rouen, France. .,Service de Pneumologie, Oncologie Thoracique, Soins Intensifs Respiratoires, CHU de Rouen, 1 rue de Germont, 76031, Rouen Cedex, France.
| | - Florian Guisier
- Rouen University Hospital, Clinique Pneumologique & CIC INSERM U 1404, F-76000, Rouen, France.,Normandie University, UNIROUEN, LITIS, Quant.I.F - EA 4108, F-76000, Rouen, France
| | - Xavier Brune
- Normandie University, COBRA, UMR 6014 & FR 3038; CNRS, F-76000, Rouen, France
| | - Pierre Bohn
- Rouen University Hospital, Clinique Pneumologique & CIC INSERM U 1404, F-76000, Rouen, France
| | - Anthony Romieu
- Normandie University, COBRA, UMR 6014 & FR 3038; CNRS, F-76000, Rouen, France.,Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR 6302, CNRS, University, Bourgogne Franche-Comté, 21078, Dijon, France
| | - Nasrin Sarafan-Vasseur
- Génétique du cancer et des maladies neuropsychiatriques, Normandie University, UNIROUEN, INSERM, F-76000, Rouen, France
| | - Richard Sesboüé
- Génétique du cancer et des maladies neuropsychiatriques, Normandie University, UNIROUEN, INSERM, F-76000, Rouen, France
| | - Pierre-Yves Renard
- Normandie University, COBRA, UMR 6014 & FR 3038; CNRS, F-76000, Rouen, France
| | - Luc Thiberville
- Rouen University Hospital, Clinique Pneumologique & CIC INSERM U 1404, F-76000, Rouen, France.,Normandie University, UNIROUEN, LITIS, Quant.I.F - EA 4108, F-76000, Rouen, France
| | - Mathieu Salaün
- Rouen University Hospital, Clinique Pneumologique & CIC INSERM U 1404, F-76000, Rouen, France.,Normandie University, UNIROUEN, LITIS, Quant.I.F - EA 4108, F-76000, Rouen, France
| |
Collapse
|
31
|
Wang QY, Li HM, Dong ZP, Li BX, Huo M, Lu T, Wang Y. Peptide-mediated cationic micelles drug-delivery system applied on a VEGFR3-overexpressed tumor. J Mater Chem B 2019; 7:1076-1086. [DOI: 10.1039/c8tb02255k] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Copolymers as a kind of drug delivery carrier always lack targeting efficiency.
Collapse
Affiliation(s)
- Qi Y. Wang
- Key Laboratory of Biomedical Functional Materials
- School of Sciences
- China
- Pharmaceutical University
- Nanjing 211198
| | - Hong M. Li
- Key Laboratory of Biomedical Functional Materials
- School of Sciences
- China
- Pharmaceutical University
- Nanjing 211198
| | - Zhi P. Dong
- Key Laboratory of Biomedical Functional Materials
- School of Sciences
- China
- Pharmaceutical University
- Nanjing 211198
| | - Bing X. Li
- Key Laboratory of Biomedical Functional Materials
- School of Sciences
- China
- Pharmaceutical University
- Nanjing 211198
| | - Ming Huo
- Key Laboratory of Biomedical Functional Materials
- School of Sciences
- China
- Pharmaceutical University
- Nanjing 211198
| | - Tao Lu
- Key Laboratory of Biomedical Functional Materials
- School of Sciences
- China
- Pharmaceutical University
- Nanjing 211198
| | - Yue Wang
- Key Laboratory of Biomedical Functional Materials
- School of Sciences
- China
- Pharmaceutical University
- Nanjing 211198
| |
Collapse
|
32
|
Kleynhans J, Grobler AF, Ebenhan T, Sathekge MM, Zeevaart JR. Radiopharmaceutical enhancement by drug delivery systems: A review. J Control Release 2018; 287:177-193. [DOI: 10.1016/j.jconrel.2018.08.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/02/2018] [Accepted: 08/03/2018] [Indexed: 12/17/2022]
|
33
|
Li J, Yang Y, Zhang X, Zhang B, Chang H, Wei W. Resonance Rayleigh scattering assay for EGFR using antibody immobilized gold nanoparticles. LUMINESCENCE 2018; 33:1326-1332. [PMID: 30264501 DOI: 10.1002/bio.3550] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/12/2018] [Accepted: 08/20/2018] [Indexed: 12/17/2022]
Abstract
A highly selectivity determination of epidermal growth factor receptor (EGFR) has been described in the article. Antibody immobilized cysteamine (Cys) functionalized gold nanoparticles (AuNP) were proposed as immunosensors, and resonance Rayleigh scattering (RRS) was used for detection. First, Cys stabilized AuNPs (Cys-AuNP) were prepared by the reduction of chloroauric acid with sodium borohydride in the presence of Cys. Further, anti-EGFR antibody (Cetuximab, C225) was covalently linked to the Cys-AuNP by carbodiimide-mediated amidation protocol to yield the C225-AuNP immunoprobe. The prepared conjugations were characterized by ultraviolet-visible (UV-vis) spectroscopy, transmission electron microscopy (TEM), Fourier transform infrared (FT-IR) and sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE). Based on the specific binding of C225 to EGFR, an RRS method was established to determine the concentration of EGFR. Under the optimal conditions, the concentration of EGFR was related to the intensity of RRS in the range 30-130 ng ml-1 with a low detection limit of 4.0 ng ml-1 . Meanwhile, the proposed immunosensor exhibited excellent selectivity and anti-interference property. The method was applied to the determination of EGFR in human serum and cancer cell lysate samples with satisfactory results.
Collapse
Affiliation(s)
- Junbo Li
- Department of Chemistry and Chemical Engineering, Taiyuan University of Technology, Taiyuan, P.R. China.,Pharmaceutical Department, Changzhi Medical College, Changzhi, P.R. China
| | - Yanqiang Yang
- Changzhi Entry-exit Inspection and Quarantine Bureau, Changzhi, P.R. China
| | - Xinxin Zhang
- Changzhi Entry-exit Inspection and Quarantine Bureau, Changzhi, P.R. China
| | - Bing Zhang
- Department of Chemistry and Chemical Engineering, Taiyuan University of Technology, Taiyuan, P.R. China
| | - Honghong Chang
- Department of Chemistry and Chemical Engineering, Taiyuan University of Technology, Taiyuan, P.R. China
| | - Wenlong Wei
- Department of Chemistry and Chemical Engineering, Taiyuan University of Technology, Taiyuan, P.R. China
| |
Collapse
|
34
|
Laprise-Pelletier M, Simão T, Fortin MA. Gold Nanoparticles in Radiotherapy and Recent Progress in Nanobrachytherapy. Adv Healthc Mater 2018; 7:e1701460. [PMID: 29726118 DOI: 10.1002/adhm.201701460] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/07/2018] [Indexed: 12/29/2022]
Abstract
Over the last few decades, gold nanoparticles (GNPs) have emerged as "radiosensitizers" in oncology. Radiosensitizers are additives that can enhance the effects of radiation on biological tissues treated with radiotherapy. The interaction of photons with GNPs leads to the emission of low-energy and short-range secondary electrons, which in turn increase the dose deposited in tissues. In this context, GNPs are the subject of intensive theoretical and experimental studies aiming at optimizing the parameters leading to greater dose enhancement and highest therapeutic effect. This review describes the main mechanisms occurring between photons and GNPs that lead to dose enhancement. The outcome of theoretical simulations of the interactions between GNPs and photons is presented. Finally, the findings of the most recent in vivo studies about interactions between GNPs and photon sources (e.g., external beams, brachytherapy sources, and molecules labeled with radioisotopes) are described. The advantages and challenges inherent to each of these approaches are discussed. Future directions, providing new guidelines for the successful translation of GNPs into clinical applications, are also highlighted.
Collapse
Affiliation(s)
- Myriam Laprise-Pelletier
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval (CR-CHU de Québec); Axe Médecine Régénératrice; Québec G1L 3L5 QC Canada
- Department of Mining; Metallurgy and Materials Engineering; Université Laval; Québec G1V 0A6 QC Canada
- Centre de Recherche sur les Matériaux Avancés (CERMA); Université Laval; Québec G1V 0A6 QC Canada
| | - Teresa Simão
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval (CR-CHU de Québec); Axe Médecine Régénératrice; Québec G1L 3L5 QC Canada
- Department of Mining; Metallurgy and Materials Engineering; Université Laval; Québec G1V 0A6 QC Canada
- Centre de Recherche sur les Matériaux Avancés (CERMA); Université Laval; Québec G1V 0A6 QC Canada
| | - Marc-André Fortin
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval (CR-CHU de Québec); Axe Médecine Régénératrice; Québec G1L 3L5 QC Canada
- Department of Mining; Metallurgy and Materials Engineering; Université Laval; Québec G1V 0A6 QC Canada
- Centre de Recherche sur les Matériaux Avancés (CERMA); Université Laval; Québec G1V 0A6 QC Canada
| |
Collapse
|
35
|
Bhattacharya P, Shetake NG, Pandey BN, Kumar A. Receptor tyrosine kinase signaling in cancer radiotherapy and its targeting for tumor radiosensitization. Int J Radiat Biol 2018; 94:628-644. [DOI: 10.1080/09553002.2018.1478160] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Poushali Bhattacharya
- Radiation Signaling and Cancer Biology Section, Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Neena G. Shetake
- Radiation Signaling and Cancer Biology Section, Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Badri N. Pandey
- Radiation Signaling and Cancer Biology Section, Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Amit Kumar
- Radiation Signaling and Cancer Biology Section, Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| |
Collapse
|
36
|
Synthesis and Bioevaluation of Iodine-131 Directly Labeled Cyclic RGD-PEGylated Gold Nanorods for Tumor-Targeted Imaging. CONTRAST MEDIA & MOLECULAR IMAGING 2017; 2017:6081724. [PMID: 29434531 PMCID: PMC5757100 DOI: 10.1155/2017/6081724] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 12/04/2017] [Indexed: 01/09/2023]
Abstract
Introduction Radiolabeled gold nanoparticles play an important role in biomedical application. The aim of this study was to prepare iodine-131 (131I)-labeled gold nanorods (GNRs) conjugated with cyclic RGD and evaluate its biological characteristics for targeted imaging of integrin αvβ3-expressing tumors. Methods HS-PEG(5000)-COOH molecules were applied to replace CTAB covering the surface of bare GNRs for better biocompatibility, and c(RGDfK) peptides were conjugated onto the carboxyl terminal of GNR-PEG-COOH via EDC/NHS coupling reactions. The nanoconjugate was characterized, and 131I was directly tagged on the surface of GNRs via AuI bonds for SPECT/CT imaging. We preliminarily studied the characteristics of the probe and its feasibility for tumor-targeting SPECT/CT imaging. Results The [131I]GNR-PEG-cRGD probe was prepared in a simple and rapid manner and was stable in both PBS and fetal bovine serum. It targeted selectively and could be taken up by tumor cells mainly via integrin αvβ3-receptor-mediated endocytosis. In vivo imaging, biodistribution, and autoradiography results showed evident tumor uptake in integrin αvβ3-expressing tumors. Conclusions These promising results showed that this smart nanoprobe can be used for angiogenesis-targeted SPECT/CT imaging. Furthermore, the nanoprobe possesses a remarkable capacity for highly efficient photothermal conversion in the near-infrared region, suggesting its potential as a multifunctional theranostic agent.
Collapse
|
37
|
Li C, Tan J, Chang J, Li W, Liu Z, Li N, Ji Y. Radioiodine-labeled anti-epidermal growth factor receptor binding bovine serum albumin-polycaprolactone for targeting imaging of glioblastoma. Oncol Rep 2017; 38:2919-2926. [DOI: 10.3892/or.2017.5937] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 08/02/2017] [Indexed: 11/06/2022] Open
|
38
|
Same S, Aghanejad A, Akbari Nakhjavani S, Barar J, Omidi Y. Radiolabeled theranostics: magnetic and gold nanoparticles. BIOIMPACTS 2016; 6:169-181. [PMID: 27853680 PMCID: PMC5108989 DOI: 10.15171/bi.2016.23] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 09/21/2016] [Accepted: 09/27/2016] [Indexed: 01/08/2023]
Abstract
![]()
Introduction: Growing advances in nanotechnology have facilitated the applications of newly emerged nanomaterials in the field of biomedical/pharmaceutical sciences. Following this trend, the multifunctional nanoparticles (NPs) play a significant role in development of advanced drug delivery systems (DDSs) such as diapeutics/theranostics used for simultaneous diagnosis and therapy. Multifunctional radiolabeled NPs with capability of detecting, visualizing and destroying diseased cells with least side effects have been considered as an emerging filed in presentation of the best choice in solving the therapeutic problems. Functionalized magnetic and gold NPs (MNPs and GNPs, respectively) have produced the potential of nanoparticles as sensitive multifunctional probes for molecular imaging, photothermal therapy and drug delivery and targeting.
Methods: In this study, we review the most recent works on the improvement of various techniques for development of radiolabeled magnetic and gold nanoprobes, and discuss the methods for targeted imaging and therapies.
Results: The receptor-specific radiopharmaceuticals have been developed to localized radiotherapy in disease sites. Application of advanced multimodal imaging methods and related modality imaging agents labeled with various radioisotopes (e.g., 125I, 111In, 64Cu, 68Ga, 99mTc) and MNPs/GNPs have significant effects on treatment and prognosis of cancer therapy. In addition, the surface modification with biocompatible polymer such as polyethylene glycol (PEG) have resulted in development of stealth NPs that can evade the opsonization and immune clearance. These long-circulating agents can be decorated with homing agents as well as radioisotopes for targeted imaging and therapy purposes.
Conclusion: The modified MNPs or GNPs have wide applications in concurrent diagnosis and therapy of various malignancies. Once armed with radioisotopes, these nanosystems (NSs) can be exploited for combined multimodality imaging with photothermal/photodynamic therapy while delivering the loaded drugs or genes to the targeted cells/tissues. These NSs will be a game changer in combating various cancers.
Collapse
Affiliation(s)
- Saeideh Same
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sattar Akbari Nakhjavani
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran ; Department of Molecular Medicine, School of Advanced Technologies in Medicine, International Campus, Tehran University of Medical Sciences, Tehran, Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
39
|
Chakravarty R, Hong H, Cai W. Image-Guided Drug Delivery with Single-Photon Emission Computed Tomography: A Review of Literature. Curr Drug Targets 2016; 16:592-609. [PMID: 25182469 DOI: 10.2174/1389450115666140902125657] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 08/24/2014] [Accepted: 08/26/2014] [Indexed: 12/18/2022]
Abstract
Tremendous resources are being invested all over the world for prevention, diagnosis, and treatment of various types of cancer. Successful cancer management depends on accurate diagnosis of the disease along with precise therapeutic protocol. The conventional systemic drug delivery approaches generally cannot completely remove the competent cancer cells without surpassing the toxicity limits to normal tissues. Therefore, development of efficient drug delivery systems holds prime importance in medicine and healthcare. Also, molecular imaging can play an increasingly important and revolutionizing role in disease management. Synergistic use of molecular imaging and targeted drug delivery approaches provides unique opportunities in a relatively new area called 'image-guided drug delivery' (IGDD). Single-photon emission computed tomography (SPECT) is the most widely used nuclear imaging modality in clinical context and is increasingly being used to guide targeted therapeutics. The innovations in material science have fueled the development of efficient drug carriers based on, polymers, liposomes, micelles, dendrimers, microparticles, nanoparticles, etc. Efficient utilization of these drug carriers along with SPECT imaging technology have the potential to transform patient care by personalizing therapy to the individual patient, lessening the invasiveness of conventional treatment procedures and rapidly monitoring the therapeutic efficacy. SPECT-IGDD is not only effective for the treatment of cancer but might also find utility in the management of several other diseases. Herein, we provide a concise overview of the latest advances in SPECT-IGDD procedures and discuss the challenges and opportunities for advancement of the field.
Collapse
Affiliation(s)
- Rubel Chakravarty
- Isotope Production and Applications Division, Bhabha Atomic Research Centre, Mumbai 400 085, India.
| | | | | |
Collapse
|
40
|
Li W, Ji YH, Li CX, Liu ZY, Li N, Fang L, Chang J, Tan J. Evaluation of therapeutic effectiveness of 131I-antiEGFR-BSA-PCL in a mouse model of colorectal cancer. World J Gastroenterol 2016; 22:3758-3768. [PMID: 27076760 PMCID: PMC4814738 DOI: 10.3748/wjg.v22.i14.3758] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 11/02/2015] [Accepted: 01/18/2016] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the biological effects of internal irradiation, and the therapeutic effectiveness was assessed of 131I-labeled anti-epidermal growth factor receptor (EGFR) liposomes, derived from cetuximab, when used as a tumor-targeting carrier in a colorectal cancer mouse model.
METHODS: We described the liposomes and characterized their EGFR-targeted binding and cellular uptake in EGFR-overexpressing LS180 colorectal cancer cells. After intra-tumor injections of 74 MBq (740 MBq/mL) 131I-antiEGFR-BSA-PCL, we investigated the biological effects of internal irradiation and the therapeutic efficacy of 131I-antiEGFR-BSA-PCL on colorectal cancer in a male BALB/c mouse model. Tumor size, body weight, histopathology, and SPECT imaging were monitored for 33 d post-therapy.
RESULTS: The rapid radioiodine uptake of 131I-antiEGFR-BSA-PCL and 131I-BSA-PCL reached maximum levels at 4 h after incubation, and the 131I uptake of 131I-antiEGFR-BSA-PCL was higher than that of 131I-BSA-PCL in vitro. The 131I tissue distribution assay revealed that 131I-antiEGFR-BSA-PCL was markedly taken up by the tumor. Furthermore, a tissue distribution assay revealed that 131I-antiEGFR-BSA-PCL was markedly taken up by the tumor and reached its maximal uptake value of 21.0 ± 1.01 %ID/g (%ID/g is the percentage injected dose per gram of tissue) at 72 h following therapy; the drug concentration in the tumor was higher than that in the liver, heart, colon, or spleen. Tumor size measurements showed that tumor development was significantly inhibited by treatments with 131I-antiEGFR-BSA-PCL and 131I-BSA-PCL. The volume of tumor increased, and treatment rate with 131I-antiEGFR-BSA-PCL was 124% ± 7%, lower than that with 131I-BSA-PCL (127% ± 9%), 131I (143% ± 7%), and normal saline (146% ± 10%). The percentage losses in original body weights were 39% ± 3%, 41% ± 4%, 49% ± 5%, and 55% ± 13%, respectively. The best survival and cure rates were obtained in the group treated with 131I-antiEGFR-BSA-PCL. The animals injected with 131I-antiEGFR-BSA-PCL and 131I-BSA-PCL showed more uniform focused liposome distribution within the tumor area.
CONCLUSION: This study demonstrated the potential beneficial application of 131I-antiEGFR-BSA-PCL for treating colorectal cancer. 131I-antiEGFR-BSA-PCL suppressed the development of xenografted colorectal cancer in nude mice, thereby providing a novel candidate for receptor-mediated targeted radiotherapy.
Collapse
|
41
|
Li W, Liu Z, Li C, Li N, Fang L, Chang J, Tan J. Radionuclide therapy using ¹³¹I-labeled anti-epidermal growth factor receptor-targeted nanoparticles suppresses cancer cell growth caused by EGFR overexpression. J Cancer Res Clin Oncol 2016; 142:619-32. [PMID: 26573511 DOI: 10.1007/s00432-015-2067-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 10/23/2015] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Anti-epidermal growth factor receptor (EGFR)-targeted nanoparticles can be used to deliver a therapeutic and imaging agent to EGFR-overexpressing tumor cells. (131)I-labeled anti-EGFR nanoparticles derived from cetuximab were used as a tumor-targeting vehicle in radionuclide therapy. METHODS This paper describes the construction of the anti-EGFR nanoparticle EGFR-BSA-PCL. This nanoparticle was characterized for EGFR-targeted binding and cellular uptake in EGFR-overexpressing cancer cells by using flow cytometry and confocal microscopy. Anti-EGFR and non-targeted nanoparticles were labeled with (131)I using the chloramine-T method. Analyses of cytotoxicity and targeted cell killing with (131)I were performed using the MTT assay. The time-dependent cellular uptake of (131)I-labeled anti-EGFR nanoparticles proved the slow-release effects of nanoparticles. A radioiodine therapy study was also performed in mice. RESULTS The EGFR-targeted nanoparticle EGFR-BSA-PCL and the non-targeted nanoparticle BSA-PCL were constructed; the effective diameters were approximately 100 nm. The results from flow cytometry and confocal microscopy revealed significant uptake of EGFR-BSA-PCL in EGFR-overexpressing tumor cells. Compared with EGFR-BSA-PCL, BSA-PCL could also bind to cells, but tumor cell retention was minimal and weak. In MTT assays, the EGFR-targeted radioactive nanoparticle (131)I-EGFR-BSA-PCL showed greater cytotoxicity and targeted cell killing than the non-targeted nanoparticle (131)I-BSA-PCL. The radioiodine uptake of both (131)I-labeled nanoparticles, (131)I-EGFR-BSA-PCL and (131)I-BSA-PCL, was rapid and reached maximal levels 4 h after incubation, but the (131)I uptake of (131)I-EGFR-BSA-PCL was higher than that of (131)I-BSA-PCL. On day 15, the average tumor volumes of the (131)I-EGFR-BSA-PCL and (131)I-BSA-PCL groups showed a slow growth relationship compared with that of the control group. CONCLUSION The EGFR-targeted nanoparticle EGFR-BSA-PCL demonstrated superior cellular binding and uptake compared with those of the control BSA-PCL. The EGFR-targeted radioactive nanoparticle (131)I-EGFR-BSA-PCL exhibited favorable intracellular retention of (131)I. Radionuclide therapy using (131)I-EGFR-BSA-PCL, which showed excellent targeted cell killing, suppressed cancer cell growth caused by EGFR overexpression.
Collapse
Affiliation(s)
- Wei Li
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052, People's Republic of China
| | - Zhongyun Liu
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, 264003, Shandong, People's Republic of China
| | - Chengxia Li
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052, People's Republic of China
| | - Ning Li
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052, People's Republic of China
| | - Lei Fang
- Institute of Nanobiotechnology, School of Materials Science and Engineering, Tianjin Key Laboratory of Composites and Functional Materials, Tianjin University, Tianjin, 300072, People's Republic of China
| | - Jin Chang
- Institute of Nanobiotechnology, School of Materials Science and Engineering, Tianjin Key Laboratory of Composites and Functional Materials, Tianjin University, Tianjin, 300072, People's Republic of China
| | - Jian Tan
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052, People's Republic of China.
| |
Collapse
|
42
|
The Tumor Microenvironment as a Barrier to Cancer Nanotherapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 936:165-190. [PMID: 27739048 DOI: 10.1007/978-3-319-42023-3_9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Although extensive research effort and resources have been dedicated to the development of nanotherapeutics to treat cancer, few formulations have reached clinical application. A major reason is that the large number of parameters available to tune nanotherapy characteristics coupled with the variability in tumor tissue precludes evaluation of complex interactions through experimentation alone. In order to optimize the nanotechnology design and gain further insight into these phenomena, mathematical modeling and computational simulation have been applied to complement empirical work. In this chapter, we discuss modeling work related to nanotherapy and the tumor microenvironment. We first summarize the biology underlying the dysregulated tumor microenvironment, followed by a description of major nano-scale parameters. We then present an overview of the mathematical modeling of cancer nanotherapy, including evaluation of nanotherapy in multi-dimensional tumor tissue, coupling of nanotherapy with vascular flow, modeling of nanotherapy in combination with in vivo imaging, modeling of nanoparticle transport based on in vitro data, modeling of vasculature-bound nanoparticles, evaluation of nanotherapy using pharmacokinetic modeling, and modeling of nano-based hyperthermia. We conclude that an even tighter interdisciplinary effort between biological, material, and physical scientists is needed in order to eventually overcome the tumor microenvironment barrier to successful nanotherapy.
Collapse
|
43
|
Zhu J, Zhao L, Cheng Y, Xiong Z, Tang Y, Shen M, Zhao J, Shi X. Radionuclide (131)I-labeled multifunctional dendrimers for targeted SPECT imaging and radiotherapy of tumors. NANOSCALE 2015; 7:18169-18178. [PMID: 26477402 DOI: 10.1039/c5nr05585g] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
We report the synthesis, characterization, and utilization of radioactive (131)I-labeled multifunctional dendrimers for targeted single-photon emission computed tomography (SPECT) imaging and radiotherapy of tumors. In this study, amine-terminated poly(amidoamine) dendrimers of generation 5 (G5·NH2) were sequentially modified with 3-(4'-hydroxyphenyl)propionic acid-OSu (HPAO) and folic acid (FA) linked with polyethylene glycol (PEG), followed by acetylation modification of the dendrimer remaining surface amines and labeling of radioactive iodine-131 ((131)I). The generated multifunctional (131)I-G5·NHAc-HPAO-PEG-FA dendrimers were characterized via different methods. We show that prior to (131)I labeling, the G5·NHAc-HPAO-PEG-FA dendrimers conjugated with approximately 9.4 HPAO moieties per dendrimer are noncytotoxic at a concentration up to 20 μM and are able to target cancer cells overexpressing FA receptors (FAR), thanks to the modified FA ligands. In the presence of a phenol group, radioactive (131)I is able to be efficiently labeled onto the dendrimer platform with good stability and high radiochemical purity, and render the platform with an ability for targeted SPECT imaging and radiotherapy of an FAR-overexpressing xenografted tumor model in vivo. The designed strategy to use the facile dendrimer nanotechnology may be extended to develop various radioactive theranostic nanoplatforms for targeted SPECT imaging and radiotherapy of different types of cancer.
Collapse
Affiliation(s)
- Jingyi Zhu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, People's Republic of China
| | - Lingzhou Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200080, People's Republic of China.
| | - Yongjun Cheng
- Department of Nuclear Medicine, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200080, People's Republic of China.
| | - Zhijuan Xiong
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, People's Republic of China.
| | - Yueqin Tang
- Experiment Center, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200080, People's Republic of China
| | - Mingwu Shen
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, People's Republic of China.
| | - Jinhua Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200080, People's Republic of China.
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, People's Republic of China and College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, People's Republic of China.
| |
Collapse
|
44
|
Transdermal treatment of the surgical and burned wound skin via phytochemical-capped gold nanoparticles. Colloids Surf B Biointerfaces 2015; 135:166-174. [DOI: 10.1016/j.colsurfb.2015.07.058] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 07/01/2015] [Accepted: 07/21/2015] [Indexed: 12/24/2022]
|
45
|
England CG, Gobin AM, Frieboes HB. Evaluation of uptake and distribution of gold nanoparticles in solid tumors. EUROPEAN PHYSICAL JOURNAL PLUS 2015; 130:231. [PMID: 27014559 PMCID: PMC4800753 DOI: 10.1140/epjp/i2015-15231-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Although nanotherapeutics offer a targeted and potentially less toxic alternative to systemic chemotherapy in cancer treatment, nanotherapeutic transport is typically hindered by abnormal characteristics of tumor tissue. Once nanoparticles targeted to tumor cells arrive in the circulation of tumor vasculature, they must extravasate from irregular vessels and diffuse through the tissue to ideally reach all malignant cells in cytotoxic concentrations. The enhanced permeability and retention effect can be leveraged to promote extravasation of appropriately sized particles from tumor vasculature; however, therapeutic success remains elusive partly due to inadequate intra-tumoral transport promoting heterogeneous nanoparticle uptake and distribution. Irregular tumor vasculature not only hinders particle transport but also sustains hypoxic tissue kregions with quiescent cells, which may be unaffected by cycle-dependent chemotherapeutics released from nanoparticles and thus regrow tumor tissue following nanotherapy. Furthermore, a large proportion of systemically injected nanoparticles may become sequestered by the reticuloendothelial system, resulting in overall diminished efficacy. We review recent work evaluating the uptake and distribution of gold nanoparticles in pre-clinical tumor models, with the goal to help improve nanotherapy outcomes. We also examine the potential role of novel layered gold nanoparticles designed to address some of these critical issues, assessing their uptake and transport in cancerous tissue.
Collapse
Affiliation(s)
- Christopher G England
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY 40292, USA; James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40292, USA
| | - André M Gobin
- Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA
| | - Hermann B Frieboes
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY 40292, USA; James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40292, USA; Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA
| |
Collapse
|
46
|
Abstract
Theranostics is a promising field that combines therapeutics and diagnostics into single multifunctional formulations. This field is driven by advancements in nanoparticle systems capable of providing the necessary functionalities. By utilizing these powerful nanomedicines, the concept of personalized medicine can be realized by tailoring treatment strategies to the individual. This review gives a brief overview of the components of a theranostic system and the challenges that designing truly multifunctional nanoparticles present. Considerations when choosing a class of nanoparticle include the size, shape, charge, and surface chemistry, while classes of nanoparticles discussed are polymers, liposomes, dendrimers, and polymeric micelles. Targeting to disease states can be achieved either through passive or active targeting which uses specific ligands to target receptors that are overexpressed in tumors and common targeting elements are presented. To image the interactions with disease states, contrast agents are included in the nanoparticle formulation. Imaging options include optical imaging techniques, computed tomography, nuclear based, and magnetic resonance imaging. The interplay between all of these components needs to be carefully considered when designing a theranostic system.
Collapse
|
47
|
Trouiller AJ, Hebié S, El Bahhaj F, Napporn TW, Bertrand P. Chemistry for oncotheranostic gold nanoparticles. Eur J Med Chem 2015; 99:92-112. [PMID: 26057706 DOI: 10.1016/j.ejmech.2015.05.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 05/13/2015] [Accepted: 05/14/2015] [Indexed: 12/18/2022]
Abstract
This review presents in a comprehensive ways the chemical methods used to functionalize gold nanoparticles with focus on anti-cancer applications. The review covers the parameters required for the synthesis gold nanoparticles with defined shapes and sizes, method for targeted delivery in tumours, and selected examples of anti-cancers compounds delivered with gold nanoparticles. A short survey of bioassays for oncology based on gold nanoparticles is also presented.
Collapse
Affiliation(s)
- Anne Juliette Trouiller
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers, Equipe Synthèse Organique, 4 rue Michel Brunet, B28, 86073 Poitiers, France
| | - Seydou Hebié
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers, Equipe SAMCat, 4 rue Michel Brunet, B27, 86073 Poitiers, France
| | - Fatima El Bahhaj
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers, Equipe Synthèse Organique, 4 rue Michel Brunet, B28, 86073 Poitiers, France
| | - Teko W Napporn
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers, Equipe SAMCat, 4 rue Michel Brunet, B27, 86073 Poitiers, France
| | - Philippe Bertrand
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers, Equipe Synthèse Organique, 4 rue Michel Brunet, B28, 86073 Poitiers, France.
| |
Collapse
|
48
|
Liu J, Liang Y, Liu T, Li D, Yang X. Anti-EGFR-Conjugated Hollow Gold Nanospheres Enhance Radiocytotoxic Targeting of Cervical Cancer at Megavoltage Radiation Energies. NANOSCALE RESEARCH LETTERS 2015; 10:218. [PMID: 25995714 PMCID: PMC4437992 DOI: 10.1186/s11671-015-0923-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 05/04/2015] [Indexed: 05/29/2023]
Abstract
The study aimed to confirm that anti-epidermal growth factor receptor (EGFR) monoclonal antibody-conjugated hollow gold nanospheres (anti-EGFR/HGNs) can be selectively uptaken by cervical cancer cells and induce its apoptosis when combined with radiotherapy, as a result enhancing radiosensitivity of cervical cancer cells. HGNs with a mean diameter of 54.6 ± 7.11 nm and wall thickness of 5.01 ± 2.23 nm were viewed by transmission electron microscopy (TEM). Cell uptake was assayed by inductively coupled plasma atomic emission spectroscopy (ICP-AES). The cytotoxicity on HeLa cells, which were used in our experiment, was assessed by CCK-8 assay. Cell cycle and apoptosis were examined by an Annexin V-FITC/propidium iodide (PI) kit with flow cytometry (FCM). The expression of several critical apoptosis-related proteins, including Bcl-2, Bax, Bad, and active caspase 3, was tested by western blot analysis. Cells treated by anti-EGFR/HGNs showed an obvious increase in nanoparticle uptake compared to naked HGNs. Anti-EGFR/HGNs combined with radiation resulted in a significant growth inhibition, compared with radiation combined with naked HGNs. Anti-EGFR/HGNs remarkably increased the ratio of HeLa cells in the G2/M phase and induced more apoptosis by an obvious deregulation of Bcl-2 and upregulation of Bax, Bad, and caspase 3 when combined with radiation. Therefore, anti-EGFR/HGNs can increase the targeted uptake of HGNs by HeLa cells and enhance radiocytotoxic targeting of cervical cancer at megavoltage radiation energies.
Collapse
Affiliation(s)
- Jiao Liu
- />Department of Gynecology and Obstetrics, Qilu Hospital, Shandong University, Wenhuaxilu No. 107, Jinan, 250012 Shandong Province China
| | - Ying Liang
- />Department of Gynecology and Obstetrics, Qilu Hospital, Shandong University, Wenhuaxilu No. 107, Jinan, 250012 Shandong Province China
| | - Ting Liu
- />Department of Gynecology and Obstetrics, Qilu Hospital, Shandong University, Wenhuaxilu No. 107, Jinan, 250012 Shandong Province China
| | - Dengke Li
- />Department of General Surgery, Qilu Hospital, Shandong University, Wenhuaxilu No. 107, Jinan, 250012 Shandong Province China
| | - Xingsheng Yang
- />Department of Gynecology and Obstetrics, Qilu Hospital, Shandong University, Wenhuaxilu No. 107, Jinan, 250012 Shandong Province China
| |
Collapse
|
49
|
Multimodal Imaging and Theranostic Application of Disease-Directed Agents. TOPICS IN MEDICINAL CHEMISTRY 2015. [DOI: 10.1007/7355_2015_91] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
50
|
Qian Y, Qiu M, Wu Q, Tian Y, Zhang Y, Gu N, Li S, Xu L, Yin R. Enhanced cytotoxic activity of cetuximab in EGFR-positive lung cancer by conjugating with gold nanoparticles. Sci Rep 2014; 4:7490. [PMID: 25502402 PMCID: PMC4265789 DOI: 10.1038/srep07490] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 11/28/2014] [Indexed: 11/20/2022] Open
Abstract
Cetuximab (C225) is a unique agent, targeting epidermal growth factor receptor (EGFR)-positive cancer. However, the therapeutic effect of C225 in EGFR high-expressing non-small cell lung cancer (NSCLC) remains poor. Here, we report that conjugation of C225 with gold nanoparticles (AuNPs) enhances the cytotoxicity of C225 in NSCLC both in vitro and in vivo. The NSCLC cell lines A549 (EGFRhigh) and H1299 (EGFRlow) were employed to investigate different responses to C225, IgG-AuNPs and C225-AuNPs. The antitumor properties of C225-AuNPs were explored in vivo by establishing a tumor xenograft model in nude mice. Overall, the therapeutic effect of C225-AuNPs was more pronounced in EGFRhigh A549 cells compared with EGFRlow H1299 cells. The cytotoxic effect of C225-AuNPs in A549 cells increased in a dose-dependent manner. C225-AuNPs significantly suppressed A549 cell proliferation and migration capacity and accelerated apoptosis compared with C225, and this effect was probably due to enhanced EGFR endocytosis and the subsequent suppression of downstream signaling pathway. Finally in the tumor xenograft of nude mice, treatment with C225-AuNPs also led to a significant reduction in tumor weight and volume with low toxicity. Our findings suggest that C225-AuNPs conjugate has promising potential for targeted therapy of EGFR positive NSCLC patients.
Collapse
Affiliation(s)
- Yichun Qian
- 1] Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Cancer Institute of Jiangsu Province, Nanjing, P.R. China 210009 [2] The First Clinical College, Nanjing Medical University, Nanjing, P.R. China 210029
| | - Mantang Qiu
- 1] Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Cancer Institute of Jiangsu Province, Nanjing, P.R. China 210009 [2] The First Clinical College, Nanjing Medical University, Nanjing, P.R. China 210029
| | - Qingquan Wu
- Department of Thoracic Surgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, P. R. China 223300
| | - Yanyan Tian
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, P.R. China 210096
| | - Yu Zhang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, P.R. China 210096
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, P.R. China 210096
| | - Suyi Li
- Department of Oncology, Southeast University Affiliated Zhongda Hospital, Nanjing, P.R. China 210009
| | - Lin Xu
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Cancer Institute of Jiangsu Province, Nanjing, P.R. China 210009
| | - Rong Yin
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Cancer Institute of Jiangsu Province, Nanjing, P.R. China 210009
| |
Collapse
|