1
|
Tunçel A, Maschauer S, Prante O, Yurt F. In Vitro Assessment of 177Lu-Labeled Trastuzumab-Targeted Mesoporous Carbon@Silica Nanostructure for the Treatment of HER2-Positive Breast Cancer. Pharmaceuticals (Basel) 2024; 17:732. [PMID: 38931400 PMCID: PMC11206869 DOI: 10.3390/ph17060732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 05/27/2024] [Accepted: 06/01/2024] [Indexed: 06/28/2024] Open
Abstract
This study assessed the effectiveness of a trastuzumab-targeted 177Lu-labeled mesoporous Carbon@Silica nanostructure (DOTA@TRA/MC@Si) for HER2-positive breast cancer treatment, focusing on its uptake, internalization, and efflux in breast cancer cells. The synthesized PEI-MC@Si nanocomposite was reacted with DOTA-NHS-ester, confirmed by the Arsenazo(III) assay. Following this, TRA was conjugated to the DOTA@PEI-MC@Si for targeting. DOTA@PEI-MC@Si and DOTA@TRA/MC@Si nanocomposites were labeled with 177Lu, and their efficacy was evaluated through in vitro radiolabeling experiments. According to the results, the DOTA@TRA/MC@Si nanocomposite was successfully labeled with 177Lu, yielding a radiochemical yield of 93.0 ± 2.4%. In vitro studies revealed a higher uptake of the [177Lu]Lu-DOTA@TRA/MC@Si nanocomposite in HER2-positive SK-BR-3 cells (44.0 ± 4.6% after 24 h) compared to MDA-MB-231 cells (21.0 ± 2.3%). The IC50 values for TRA-dependent uptake in the SK-BR-3 and BT-474 cells were 0.9 µM and 1.3 µM, respectively, indicating affinity toward HER-2 receptor-expressing cells. The lipophilic distribution coefficients of the radiolabeled nanocomposites were determined to be 1.7 ± 0.3 for [177Lu]Lu-DOTA@TRA/MC@Si and 1.5 ± 0.2 for [177Lu]Lu-DOTA@PEI-MC@Si, suggesting sufficient passive transport through the cell membrane and increased accumulation in target tissues. The [177Lu]Lu-DOTA@TRA/MC@Si nanocomposite showed an uptake into HER2-positive cell lines, marking a valuable step toward the development of a nanoparticle-based therapeutic agent for an improved treatment strategy for HER2-positive breast cancer.
Collapse
Affiliation(s)
- Ayça Tunçel
- Department of Nuclear Applications, Institute of Nuclear Science, Ege University, Bornova 35100, Turkey;
| | - Simone Maschauer
- Department of Nuclear Medicine, Molecular Imaging and Radiochemistry, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Ulmenweg 18, D-91054 Erlangen, Germany;
| | - Olaf Prante
- Department of Nuclear Medicine, Molecular Imaging and Radiochemistry, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Ulmenweg 18, D-91054 Erlangen, Germany;
| | - Fatma Yurt
- Department of Nuclear Applications, Institute of Nuclear Science, Ege University, Bornova 35100, Turkey;
| |
Collapse
|
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
|
Huynh M, Vinck R, Gibert B, Gasser G. Strategies for the Nuclear Delivery of Metal Complexes to Cancer Cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311437. [PMID: 38174785 DOI: 10.1002/adma.202311437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/20/2023] [Indexed: 01/05/2024]
Abstract
The nucleus is an essential organelle for the function of cells. It holds most of the genetic material and plays a crucial role in the regulation of cell growth and proliferation. Since many antitumoral therapies target nucleic acids to induce cell death, tumor-specific nuclear drug delivery could potentiate therapeutic effects and prevent potential off-target side effects on healthy tissue. Due to their great structural variety, good biocompatibility, and unique physico-chemical properties, organometallic complexes and other metal-based compounds have sparked great interest as promising anticancer agents. In this review, strategies for specific nuclear delivery of metal complexes are summarized and discussed to highlight crucial parameters to consider for the design of new metal complexes as anticancer drug candidates. Moreover, the existing opportunities and challenges of tumor-specific, nucleus-targeting metal complexes are emphasized to outline some new perspectives and help in the design of new cancer treatments.
Collapse
Affiliation(s)
- Marie Huynh
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry of Life and Health Sciences, Laboratory for Inorganic Chemistry, Paris, F-75005, France
- Gastroenterology and technologies for Health, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS5286, Université Lyon 1, Lyon, 69008, France
| | - Robin Vinck
- Orano, 125 avenue de Paris, Châtillon, 92320, France
| | - Benjamin Gibert
- Gastroenterology and technologies for Health, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS5286, Université Lyon 1, Lyon, 69008, France
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry of Life and Health Sciences, Laboratory for Inorganic Chemistry, Paris, F-75005, France
| |
Collapse
|
4
|
Davarci G, Wängler C, Eberhardt K, Geppert C, Schirrmacher R, Freudenberg R, Pretze M, Wängler B. Radiosynthesis of Stable 198Au-Nanoparticles by Neutron Activation of α vβ 3-Specific AuNPs for Therapy of Tumor Angiogenesis. Pharmaceuticals (Basel) 2023; 16:1670. [PMID: 38139797 PMCID: PMC10747377 DOI: 10.3390/ph16121670] [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: 11/06/2023] [Revised: 11/24/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
This paper reports on the development of stable tumor-specific gold nanoparticles (AuNPs) activated by neutron irradiation as a therapeutic option for the treatment of cancer with high tumor angiogenesis. The AuNPs were designed with different mono- or dithiol-ligands and decorated with different amounts of Arg-Gly-Asp (RGD) peptides as a tumor-targeting vector for αvβ3 integrin, which is overexpressed in tissues with high tumor angiogenesis. The AuNPs were evaluated for avidity in vitro and showed favorable properties with respect to tumor cell accumulation. Furthermore, the therapeutic properties of the [198Au]AuNPs were evaluated in vitro on U87MG cells in terms of cell survival, suggesting that these [198Au]AuNPs are a useful basis for future therapeutic concepts.
Collapse
Affiliation(s)
- Güllü Davarci
- Molecular Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, 68167 Mannheim, Germany;
| | - Carmen Wängler
- Biomedical Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, 68167 Mannheim, Germany;
- Mannheim Institute for Intelligent Systems in Medicine MIISM, Medical Faculty Mannheim of Heidelberg University, 68167 Mannheim, Germany
| | - Klaus Eberhardt
- Research Reactor TRIGA Mainz, Institute for Nuclear Chemistry, Johannes-Gutenberg-Universität Mainz, 55128 Mainz, Germany; (K.E.); (C.G.)
| | - Christopher Geppert
- Research Reactor TRIGA Mainz, Institute for Nuclear Chemistry, Johannes-Gutenberg-Universität Mainz, 55128 Mainz, Germany; (K.E.); (C.G.)
| | - Ralf Schirrmacher
- Department of Oncology, Division of Oncological Imaging, University of Alberta, Edmonton, AB T6G 2R3, Canada;
| | - Robert Freudenberg
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany;
| | - Marc Pretze
- Molecular Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, 68167 Mannheim, Germany;
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany;
| | - Björn Wängler
- Molecular Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, 68167 Mannheim, Germany;
| |
Collapse
|
5
|
Bentivoglio V, Nayak P, Varani M, Lauri C, Signore A. Methods for Radiolabeling Nanoparticles (Part 3): Therapeutic Use. Biomolecules 2023; 13:1241. [PMID: 37627307 PMCID: PMC10452659 DOI: 10.3390/biom13081241] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/04/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Following previously published systematic reviews on the diagnostic use of nanoparticles (NPs), in this manuscript, we report published methods for radiolabeling nanoparticles with therapeutic alpha-emitting, beta-emitting, or Auger's electron-emitting isotopes. After analyzing 234 papers, we found that different methods were used with the same isotope and the same type of nanoparticle. The most common type of nanoparticles used are the PLGA and PAMAM nanoparticles, and the most commonly used therapeutic isotope is 177Lu. Regarding labeling methods, the direct encapsulation of the isotope resulted in the most reliable and reproducible technique. Radiolabeled nanoparticles show promising results in metastatic breast and lung cancer, although this field of research needs more clinical studies, mainly on the comparison of nanoparticles with chemotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Alberto Signore
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Faculty of Medicine and Psychology, “Sapienza” University of Rome, 00185 Rome, Italy; (V.B.); (P.N.); (M.V.); (C.L.)
| |
Collapse
|
6
|
Ahmadi M, Emzhik M, Mosayebnia M. Nanoparticles labeled with gamma-emitting radioisotopes: an attractive approach for in vivo tracking using SPECT imaging. Drug Deliv Transl Res 2023; 13:1546-1583. [PMID: 36811810 DOI: 10.1007/s13346-023-01291-1] [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] [Accepted: 01/03/2023] [Indexed: 02/24/2023]
Abstract
Providing accurate molecular imaging of the body and biological process is critical for diagnosing disease and personalizing treatment with the minimum side effects. Recently, diagnostic radiopharmaceuticals have gained more attention in precise molecular imaging due to their high sensitivity and appropriate tissue penetration depth. The fate of these radiopharmaceuticals throughout the body can be traced using nuclear imaging systems, including single-photon emission computed tomography (SPECT) and positron emission tomography (PET) modalities. In this regard, nanoparticles are attractive platforms for delivering radionuclides into targets because they can directly interfere with the cell membranes and subcellular organelles. Moreover, applying radiolabeled nanomaterials can decrease their toxicity concerns because radiopharmaceuticals are usually administrated at low doses. Therefore, incorporating gamma-emitting radionuclides into nanomaterials can provide imaging probes with valuable additional properties compared to the other carriers. Herein, we aim to review (1) the gamma-emitting radionuclides used for labeling different nanomaterials, (2) the approaches and conditions adopted for their radiolabeling, and (3) their application. This study can help researchers to compare different radiolabeling methods in terms of stability and efficiency and choose the best way for each nanosystem.
Collapse
Affiliation(s)
- Mahnaz Ahmadi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marjan Emzhik
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mona Mosayebnia
- Department of Pharmaceutical Chemistry and Radiopharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Niayesh Junction, Vali-E-Asr Ave, Tehran, 14155-6153, Iran.
| |
Collapse
|
7
|
Lodato M, Plaisance V, Pawlowski V, Kwapich M, Barras A, Buissart E, Dalle S, Szunerits S, Vicogne J, Boukherroub R, Abderrahmani A. Venom Peptides, Polyphenols and Alkaloids: Are They the Next Antidiabetics That Will Preserve β-Cell Mass and Function in Type 2 Diabetes? Cells 2023; 12:cells12060940. [PMID: 36980281 PMCID: PMC10047094 DOI: 10.3390/cells12060940] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/09/2023] [Accepted: 03/17/2023] [Indexed: 03/22/2023] Open
Abstract
Improvement of insulin secretion by pancreatic β-cells and preservation of their mass are the current challenges that future antidiabetic drugs should meet for achieving efficient and long-term glycemic control in patients with type 2 diabetes (T2D). The successful development of glucagon-like peptide 1 (GLP-1) analogues, derived from the saliva of a lizard from the Helodermatidae family, has provided the proof of concept that antidiabetic drugs directly targeting pancreatic β-cells can emerge from venomous animals. The literature reporting on the antidiabetic effects of medicinal plants suggests that they contain some promising active substances such as polyphenols and alkaloids, which could be active as insulin secretagogues and β-cell protectors. In this review, we discuss the potential of several polyphenols, alkaloids and venom peptides from snake, frogs, scorpions and cone snails. These molecules could contribute to the development of new efficient antidiabetic medicines targeting β-cells, which would tackle the progression of the disease.
Collapse
Affiliation(s)
- Michele Lodato
- University Lille, CNRS, Centrale Lille, University Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
| | - Valérie Plaisance
- University Lille, CNRS, Centrale Lille, University Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
| | - Valérie Pawlowski
- University Lille, CNRS, Centrale Lille, University Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
| | - Maxime Kwapich
- University Lille, CNRS, Centrale Lille, University Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
- Service de Diabétologie et d’Endocrinologie, CH Dunkerque, 59385 Dunkirk, France
| | - Alexandre Barras
- University Lille, CNRS, Centrale Lille, University Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
| | - Emeline Buissart
- University Lille, CNRS, Centrale Lille, University Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
| | - Stéphane Dalle
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier, France
| | - Sabine Szunerits
- University Lille, CNRS, Centrale Lille, University Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
| | - Jérôme Vicogne
- University Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Rabah Boukherroub
- University Lille, CNRS, Centrale Lille, University Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
| | - Amar Abderrahmani
- University Lille, CNRS, Centrale Lille, University Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
- Correspondence: ; Tel.: +33-362531704
| |
Collapse
|
8
|
Nanostrategies for Therapeutic and Diagnostic Targeting of Gastrin-Releasing Peptide Receptor. Int J Mol Sci 2023; 24:ijms24043455. [PMID: 36834867 PMCID: PMC9958678 DOI: 10.3390/ijms24043455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/04/2023] [Accepted: 02/05/2023] [Indexed: 02/11/2023] Open
Abstract
Advances in nanomedicine bring the attention of researchers to the molecular targets that can play a major role in the development of novel therapeutic and diagnostic modalities for cancer management. The choice of a proper molecular target can decide the efficacy of the treatment and endorse the personalized medicine approach. Gastrin-releasing peptide receptor (GRPR) is a G-protein-coupled membrane receptor, well known to be overexpressed in numerous malignancies including pancreatic, prostate, breast, lung, colon, cervical, and gastrointestinal cancers. Therefore, many research groups express a deep interest in targeting GRPR with their nanoformulations. A broad spectrum of the GRPR ligands has been described in the literature, which allows tuning of the properties of the final formulation, particularly in the field of the ligand affinity to the receptor and internalization possibilities. Hereby, the recent advances in the field of applications of various nanoplatforms that are able to reach the GRPR-expressing cells are reviewed.
Collapse
|
9
|
Mattern A, Habermann S, Zegke M, Wickleder MS, Alberto R. High-Yield 99mTc Labeling of Gold Nanoparticles Carrying Atropine and Adrenaline. Bioconjug Chem 2022; 33:1741-1749. [PMID: 35973128 DOI: 10.1021/acs.bioconjchem.2c00351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
This work focuses on the synthesis, purification, and analytical characterization of novel multifunctional Au NPs radiolabeled with 99mTc. These mixed-ligand shell Au NPs represent pharmacologically relevant samples for potential application in theragnostics. A ligand using a plain linker with a rather long chain consisting of 10 CH2 groups and a thiol moiety along with the PADA chelator has been used for both the attachment to the Au NP surface and for the 99mTc(CO)3+ complexation. We have combined this with our approach of stabilizing Au NP without any PEG or other stabilizing groups. Thus, monoligand shell Au NPs were radiolabeled by different strategies (prelabeling and postlabeling). Additionally, pharmacologically relevant Au NPs were synthesized carrying both a biofunctionalization with either atropine or adrenaline and the 99mTc radiolabel. All samples were obtained in very good yields (up to 80% of the total activity loaded onto the column) and completely/particularly purified using desalting columns. Detailed analytical characterization of the Au NPs before and after radiolabeling has proven the NPs' robustness throughout the process. Their intact functionalization, shape, and stability was confirmed by transmission electron microscopy (TEM), ultraviolet/visible (UV/vis) spectroscopy, dynamic light scattering (DLS), and infrared (IR) spectroscopy. The presented strategy represents a versatile building block system that can be adapted to a variety of bioactive molecules and may be of high relevance for theragnostic applications.
Collapse
Affiliation(s)
- Annabelle Mattern
- Institute of Inorganic Chemistry, University of Cologne, Greinstraße 6, 50939 Cologne, Germany
| | - Sebastian Habermann
- Institute of Inorganic Chemistry, University of Cologne, Greinstraße 6, 50939 Cologne, Germany
| | - Markus Zegke
- Institute of Inorganic Chemistry, University of Cologne, Greinstraße 6, 50939 Cologne, Germany
| | | | - Roger Alberto
- Department of Chemistry, University of Zurich, Winterthurer Strasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
10
|
IAEA Contribution to Nanosized Targeted Radiopharmaceuticals for Drug Delivery. Pharmaceutics 2022; 14:pharmaceutics14051060. [PMID: 35631646 PMCID: PMC9146346 DOI: 10.3390/pharmaceutics14051060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/23/2022] [Accepted: 05/09/2022] [Indexed: 11/17/2022] Open
Abstract
The rapidly growing interest in the application of nanoscience in the future design of radiopharmaceuticals and the development of nanosized radiopharmaceuticals in the late 2000′s, resulted in the creation of a Coordinated Research Project (CRP) by the International Atomic Energy Agency (IAEA) in 2014. This CRP entitled ‘Nanosized delivery systems for radiopharmaceuticals’ involved a team of expert scientist from various member states. This team of scientists worked on a number of cutting-edge areas of nanoscience with a focus on developing well-defined, highly effective and site-specific delivery systems of radiopharmaceuticals. Specifically, focus areas of various teams of scientists comprised of the development of nanoparticles (NPs) based on metals, polymers, and gels, and their conjugation/encapsulation or decoration with various tumor avid ligands such as peptides, folates, and small molecule phytochemicals. The research and development efforts also comprised of developing optimum radiolabeling methods of various nano vectors using diagnostic and therapeutic radionuclides including Tc-99m, Ga-68, Lu-177 and Au-198. Concerted efforts of teams of scientists within this CRP has resulted in the development of various protocols and guidelines on delivery systems of nanoradiopharmaceuticals, training of numerous graduate students/post-doctoral fellows and publications in peer reviewed journals while establishing numerous productive scientific networks in various participating member states. Some of the innovative nanoconstructs were chosen for further preclinical applications—all aimed at ultimate clinical translation for treating human cancer patients. This review article summarizes outcomes of this major international scientific endeavor.
Collapse
|
11
|
Longoria-García S, Sánchez-Domínguez CN, Gallardo-Blanco H. Recent applications of cell-penetrating peptide guidance of nanosystems in breast and prostate cancer (Review). Oncol Lett 2022; 23:103. [PMID: 35154434 PMCID: PMC8822396 DOI: 10.3892/ol.2022.13223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 01/17/2022] [Indexed: 12/24/2022] Open
Abstract
Cell-penetrating peptides (CPPs) are small peptides from natural sources or designed from other protein sequences that can penetrate cell membranes. This property has been used in biomedicine to add them to biomolecules to improve their capacity for cell internalization and as a guidance tool for specific cell types. CPPs have been shown to enhance cellular uptake in vitro and in vivo, improving the efficacy of anticancer drugs such as doxorubicin and paclitaxel, while also limiting their cytotoxic effects on healthy cells and tissues. The current study reviews the internalization and major therapeutic results achieved from the functionalization of nanosystems with CPPs for guidance into breast and prostate cancer cells in vitro and in vivo. In addition, the practical results obtained are specifically discussed for use as a starting point for scientists looking to begin research in this field.
Collapse
Affiliation(s)
- Samuel Longoria-García
- Department of Biochemistry and Molecular Medicine, School of Medicine, Autonomous University of Nuevo Leon, Monterrey, Nuevo León 64460, Mexico
| | - Celia Nohemi Sánchez-Domínguez
- Department of Biochemistry and Molecular Medicine, School of Medicine, Autonomous University of Nuevo Leon, Monterrey, Nuevo León 64460, Mexico
| | - Hugo Gallardo-Blanco
- Department of Genetics, University Hospital ‘José Eleuterio González’, Autonomous University of Nuevo Leon, Monterrey, Nuevo León 64460, Mexico
| |
Collapse
|
12
|
Recent Progress in Technetium-99m-Labeled Nanoparticles for Molecular Imaging and Cancer Therapy. NANOMATERIALS 2021; 11:nano11113022. [PMID: 34835786 PMCID: PMC8618883 DOI: 10.3390/nano11113022] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/05/2021] [Accepted: 11/06/2021] [Indexed: 12/12/2022]
Abstract
Nanotechnology has played a tremendous role in molecular imaging and cancer therapy. Over the last decade, scientists have worked exceptionally to translate nanomedicine into clinical practice. However, although several nanoparticle-based drugs are now clinically available, there is still a vast difference between preclinical products and clinically approved drugs. An efficient translation of preclinical results to clinical settings requires several critical studies, including a detailed, highly sensitive, pharmacokinetics and biodistribution study, and selective and efficient drug delivery to the target organ or tissue. In this context, technetium-99m (99mTc)-based radiolabeling of nanoparticles allows easy, economical, non-invasive, and whole-body in vivo tracking by the sensitive clinical imaging technique single-photon emission computed tomography (SPECT). Hence, a critical analysis of the radiolabeling strategies of potential drug delivery and therapeutic systems used to monitor results and therapeutic outcomes at the preclinical and clinical levels remains indispensable to provide maximum benefit to the patient. This review discusses up-to-date 99mTc radiolabeling strategies of a variety of important inorganic and organic nanoparticles and their application to preclinical imaging studies.
Collapse
|
13
|
Apostolopoulou A, Chiotellis A, Salvanou EA, Makrypidi K, Tsoukalas C, Kapiris F, Paravatou-Petsotas M, Papadopoulos M, Pirmettis IC, Koźmiński P, Bouziotis P. Synthesis and In Vitro Evaluation of Gold Nanoparticles Functionalized with Thiol Ligands for Robust Radiolabeling with 99mTc. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:2406. [PMID: 34578721 PMCID: PMC8471789 DOI: 10.3390/nano11092406] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 12/12/2022]
Abstract
Radiolabeled gold nanoparticles (AuNPs) have been widely used for cancer diagnosis and therapy over recent decades. In this study, we focused on the development and in vitro evaluation of four new Au nanoconjugates radiolabeled with technetium-99m (99mTc) via thiol-bearing ligands attached to the NP surface. More specifically, AuNPs of two different sizes (2 nm and 20 nm, referred to as Au(2) and Au(20), respectively) were functionalized with two bifunctional thiol ligands (referred to as L1H and L2H). The shape, size, and morphology of both bare and ligand-bearing AuNPs were characterized by transmission electron microscopy (TEM) and dynamic light scattering (DLS) techniques. In vitro cytotoxicity was assessed in 4T1 murine mammary cancer cells. The AuNPs were successfully radiolabeled with 99mTc-carbonyls at high radiochemical purity (>95%) and showed excellent in vitro stability in competition studies with cysteine and histidine. Moreover, lipophilicity studies were performed in order to determine the lipophilicity of the radiolabeled conjugates, while a hemolysis assay was performed to investigate the biocompatibility of the bare and functionalized AuNPs. We have shown that the functionalized AuNPs developed in this study lead to stable radiolabeled nanoconstructs with the potential to be applied in multimodality imaging or for in vivo tracking of drug-carrying AuNPs.
Collapse
Affiliation(s)
- Adamantia Apostolopoulou
- National Center for Scientific Research “Demokritos”, Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, Agia Paraskevi, 15341 Athens, Greece; (A.A.); (A.C.); (E.-A.S.); (K.M.); (C.T.); (F.K.); (M.P.-P.); (M.P.); (I.C.P.)
- Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis Zographou, 15771 Athens, Greece
| | - Aristeidis Chiotellis
- National Center for Scientific Research “Demokritos”, Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, Agia Paraskevi, 15341 Athens, Greece; (A.A.); (A.C.); (E.-A.S.); (K.M.); (C.T.); (F.K.); (M.P.-P.); (M.P.); (I.C.P.)
| | - Evangelia-Alexandra Salvanou
- National Center for Scientific Research “Demokritos”, Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, Agia Paraskevi, 15341 Athens, Greece; (A.A.); (A.C.); (E.-A.S.); (K.M.); (C.T.); (F.K.); (M.P.-P.); (M.P.); (I.C.P.)
| | - Konstantina Makrypidi
- National Center for Scientific Research “Demokritos”, Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, Agia Paraskevi, 15341 Athens, Greece; (A.A.); (A.C.); (E.-A.S.); (K.M.); (C.T.); (F.K.); (M.P.-P.); (M.P.); (I.C.P.)
| | - Charalampos Tsoukalas
- National Center for Scientific Research “Demokritos”, Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, Agia Paraskevi, 15341 Athens, Greece; (A.A.); (A.C.); (E.-A.S.); (K.M.); (C.T.); (F.K.); (M.P.-P.); (M.P.); (I.C.P.)
| | - Fotis Kapiris
- National Center for Scientific Research “Demokritos”, Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, Agia Paraskevi, 15341 Athens, Greece; (A.A.); (A.C.); (E.-A.S.); (K.M.); (C.T.); (F.K.); (M.P.-P.); (M.P.); (I.C.P.)
| | - Maria Paravatou-Petsotas
- National Center for Scientific Research “Demokritos”, Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, Agia Paraskevi, 15341 Athens, Greece; (A.A.); (A.C.); (E.-A.S.); (K.M.); (C.T.); (F.K.); (M.P.-P.); (M.P.); (I.C.P.)
| | - Minas Papadopoulos
- National Center for Scientific Research “Demokritos”, Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, Agia Paraskevi, 15341 Athens, Greece; (A.A.); (A.C.); (E.-A.S.); (K.M.); (C.T.); (F.K.); (M.P.-P.); (M.P.); (I.C.P.)
| | - Ioannis C. Pirmettis
- National Center for Scientific Research “Demokritos”, Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, Agia Paraskevi, 15341 Athens, Greece; (A.A.); (A.C.); (E.-A.S.); (K.M.); (C.T.); (F.K.); (M.P.-P.); (M.P.); (I.C.P.)
| | - Przemysław Koźmiński
- Centre of Radiochemistry and Nuclear Chemistry, Institute of Nuclear Chemistry and Technology, Dorodna 16 Str., 03-195 Warsaw, Poland;
| | - Penelope Bouziotis
- National Center for Scientific Research “Demokritos”, Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, Agia Paraskevi, 15341 Athens, Greece; (A.A.); (A.C.); (E.-A.S.); (K.M.); (C.T.); (F.K.); (M.P.-P.); (M.P.); (I.C.P.)
| |
Collapse
|
14
|
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
|
15
|
Zhang K, Sun Y, Wu S, Zhou M, Zhang X, Zhou R, Zhang T, Gao Y, Chen T, Chen Y, Yao X, Watanabe Y, Tian M, Zhang H. Systematic imaging in medicine: a comprehensive review. Eur J Nucl Med Mol Imaging 2021; 48:1736-1758. [PMID: 33210241 DOI: 10.1007/s00259-020-05107-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 11/08/2020] [Indexed: 01/05/2023]
Abstract
Systematic imaging can be broadly defined as the systematic identification and characterization of biological processes at multiple scales and levels. In contrast to "classical" diagnostic imaging, systematic imaging emphasizes on detecting the overall abnormalities including molecular, functional, and structural alterations occurring during disease course in a systematic manner, rather than just one aspect in a partial manner. Concomitant efforts including improvement of imaging instruments, development of novel imaging agents, and advancement of artificial intelligence are warranted for achievement of systematic imaging. It is undeniable that scientists and radiologists will play a predominant role in directing this burgeoning field. This article introduces several recent developments in imaging modalities and nanoparticles-based imaging agents, and discusses how systematic imaging can be achieved. In the near future, systematic imaging which combines multiple imaging modalities with multimodal imaging agents will pave a new avenue for comprehensive characterization of diseases, successful achievement of image-guided therapy, precise evaluation of therapeutic effects, and rapid development of novel pharmaceuticals, with the final goal of improving human health-related outcomes.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Yujie Sun
- State Key Laboratory of Membrane Biology, Biodynamic Optical Imaging Center, School of Life Sciences, Peking University, Beijing, China
| | - Shuang Wu
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Min Zhou
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
- Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaohui Zhang
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Rui Zhou
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Tingting Zhang
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Yuanxue Gao
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Ting Chen
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Yao Chen
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Xin Yao
- Department of Gastroenterology, The First Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yasuyoshi Watanabe
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan.
| | - Mei Tian
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.
| | - Hong Zhang
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China.
- The College of Biomedical Engineering and Instrument Science of Zhejiang University, Hangzhou, China.
| |
Collapse
|
16
|
Prognostic and Theranostic Applications of Positron Emission Tomography for a Personalized Approach to Metastatic Castration-Resistant Prostate Cancer. Int J Mol Sci 2021; 22:ijms22063036. [PMID: 33809749 PMCID: PMC8002334 DOI: 10.3390/ijms22063036] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 01/25/2023] Open
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) represents a condition of progressive disease in spite of androgen deprivation therapy (ADT), with a broad spectrum of manifestations ranging from no symptoms to severe debilitation due to bone or visceral metastatization. The management of mCRPC has been profoundly modified by introducing novel therapeutic tools such as antiandrogen drugs (i.e., abiraterone acetate and enzalutamide), immunotherapy through sipuleucel-T, and targeted alpha therapy (TAT). This variety of approaches calls for unmet need of biomarkers suitable for patients’ pre-treatment selection and prognostic stratification. In this scenario, imaging with positron emission computed tomography (PET/CT) presents great and still unexplored potential to detect specific molecular and metabolic signatures, some of whom, such as the prostate specific membrane antigen (PSMA), can also be exploited as therapeutic targets, thus combining diagnosis and therapy in the so-called “theranostic” approach. In this review, we performed a web-based and desktop literature research to investigate the prognostic and theranostic potential of several PET imaging probes, such as 18F-FDG, 18F-choline and 68Ga-PSMA-11, also covering the emerging tracers still in a pre-clinical phase (e.g., PARP-inhibitors’ analogs and the radioligands binding to gastrin releasing peptide receptors/GRPR), highlighting their potential for defining personalized care pathways in mCRPC.
Collapse
|
17
|
de Araújo JTC, Duarte JL, Di Filippo LD, Araújo VHS, Carvalho GC, Chorilli M. Nanosystem functionalization strategies for prostate cancer treatment: a review. J Drug Target 2021; 29:808-821. [PMID: 33645369 DOI: 10.1080/1061186x.2021.1892121] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Prostate cancer (PC) has a high morbidity and mortality rate worldwide, and the current clinical guidelines can vary depending on the stage of the disease. Drug delivery nanosystems (DDNs) can improve biopharmaceutical properties of encapsulated anti-cancer drugs by modulating their release kinetics, improving physicochemical stability and reducing toxicity. DDN can also enhance the ability of specific targeting through surface modification by coupling ligands (antibodies, nucleic acids, peptides, aptamer, proteins), thus favouring the cell internalisation process by endocytosis. The purposes of this review are to describe the limitations in the treatment of PC, explore different functionalization such as polymeric, lipid and inorganic nanosystems aimed at the treatment of PC, and demonstrate the improvement of this modification for an active target, as alternative and promising candidates for new therapies.
Collapse
Affiliation(s)
| | - Jonatas Lobato Duarte
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Leonardo Delello Di Filippo
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Victor Hugo Sousa Araújo
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Gabriela Corrêa Carvalho
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Marlus Chorilli
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| |
Collapse
|
18
|
Rádis-Baptista G. Cell-Penetrating Peptides Derived from Animal Venoms and Toxins. Toxins (Basel) 2021; 13:147. [PMID: 33671927 PMCID: PMC7919042 DOI: 10.3390/toxins13020147] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/31/2021] [Accepted: 02/09/2021] [Indexed: 12/18/2022] Open
Abstract
Cell-penetrating peptides (CPPs) comprise a class of short polypeptides that possess the ability to selectively interact with the cytoplasmic membrane of certain cell types, translocate across plasma membranes and accumulate in the cell cytoplasm, organelles (e.g., the nucleus and mitochondria) and other subcellular compartments. CPPs are either of natural origin or de novo designed and synthesized from segments and patches of larger proteins or designed by algorithms. With such intrinsic properties, along with membrane permeation, translocation and cellular uptake properties, CPPs can intracellularly convey diverse substances and nanomaterials, such as hydrophilic organic compounds and drugs, macromolecules (nucleic acids and proteins), nanoparticles (nanocrystals and polyplexes), metals and radionuclides, which can be covalently attached via CPP N- and C-terminals or through preparation of CPP complexes. A cumulative number of studies on animal toxins, primarily isolated from the venom of arthropods and snakes, have revealed the cell-penetrating activities of venom peptides and toxins, which can be harnessed for application in biomedicine and pharmaceutical biotechnology. In this review, I aimed to collate examples of peptides from animal venoms and toxic secretions that possess the ability to penetrate diverse types of cells. These venom CPPs have been chemically or structurally modified to enhance cell selectivity, bioavailability and a range of target applications. Herein, examples are listed and discussed, including cysteine-stabilized and linear, α-helical peptides, with cationic and amphipathic character, from the venom of insects (e.g., melittin, anoplin, mastoparans), arachnids (latarcin, lycosin, chlorotoxin, maurocalcine/imperatoxin homologs and wasabi receptor toxin), fish (pardaxins), amphibian (bombesin) and snakes (crotamine and cathelicidins).
Collapse
Affiliation(s)
- Gandhi Rádis-Baptista
- Laboratory of Biochemistry and Biotechnology, Institute for Marine Sciences, Federal University of Ceara, Fortaleza 60165-081, Brazil
| |
Collapse
|
19
|
Pellico J, Gawne PJ, T M de Rosales R. Radiolabelling of nanomaterials for medical imaging and therapy. Chem Soc Rev 2021; 50:3355-3423. [PMID: 33491714 DOI: 10.1039/d0cs00384k] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nanomaterials offer unique physical, chemical and biological properties of interest for medical imaging and therapy. Over the last two decades, there has been an increasing effort to translate nanomaterial-based medicinal products (so-called nanomedicines) into clinical practice and, although multiple nanoparticle-based formulations are clinically available, there is still a disparity between the number of pre-clinical products and those that reach clinical approval. To facilitate the efficient clinical translation of nanomedicinal-drugs, it is important to study their whole-body biodistribution and pharmacokinetics from the early stages of their development. Integrating this knowledge with that of their therapeutic profile and/or toxicity should provide a powerful combination to efficiently inform nanomedicine trials and allow early selection of the most promising candidates. In this context, radiolabelling nanomaterials allows whole-body and non-invasive in vivo tracking by the sensitive clinical imaging techniques positron emission tomography (PET), and single photon emission computed tomography (SPECT). Furthermore, certain radionuclides with specific nuclear emissions can elicit therapeutic effects by themselves, leading to radionuclide-based therapy. To ensure robust information during the development of nanomaterials for PET/SPECT imaging and/or radionuclide therapy, selection of the most appropriate radiolabelling method and knowledge of its limitations are critical. Different radiolabelling strategies are available depending on the type of material, the radionuclide and/or the final application. In this review we describe the different radiolabelling strategies currently available, with a critical vision over their advantages and disadvantages. The final aim is to review the most relevant and up-to-date knowledge available in this field, and support the efficient clinical translation of future nanomedicinal products for in vivo imaging and/or therapy.
Collapse
Affiliation(s)
- Juan Pellico
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, UK.
| | | | | |
Collapse
|
20
|
Zhang Y, Wang Y, Meng L, Huang Q, Zhu Y, Cui W, Cheng Y, Liu R. Targeted micelles with chemotherapeutics and gene drugs to inhibit the G1/S and G2/M mitotic cycle of prostate cancer. J Nanobiotechnology 2021; 19:17. [PMID: 33422073 PMCID: PMC7796562 DOI: 10.1186/s12951-020-00756-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 12/15/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Chemotherapy and gene therapy are used in clinical practice for the treatment of castration-resistant prostate cancer. However, the poor efficiency of drug delivery and serious systemic side effects remain an obstacle to wider application of these drugs. Herein, we report newly designed PEO-PCL micelles that were self-assembled and modified by spermine ligand, DCL ligand and TAT peptide to carry docetaxel and anti-nucleostemin siRNA. RESULTS The particle size of the micelles was 42 nm, the zeta potential increased from - 12.8 to 15 mV after grafting with spermine, and the optimal N/P ratio was 25:1. Cellular MTT experiments suggested that introduction of the DCL ligand resulted in high toxicity toward PSMA-positive cells and that the TAT peptide enhanced the effect. The expression of nucleostemin was significantly suppressed in vitro and in vivo, and the tumour-inhibition experiment showed that the dual-drug delivery system suppressed CRPC tumour proliferation. CONCLUSIONS This targeted drug delivery system inhibited the G1/S and G2/M mitotic cycle via synergistic interaction of chemotherapeutics and gene drugs.
Collapse
Affiliation(s)
- Yiran Zhang
- Tianjin Institute of Urology & Department of Urology, The Second Hospital of Tianjin Medical University, 23 Pingjiang Road, Hexi District, Tianjin, 300211, People's Republic of China.,Department of Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, Shanghai, 200233, People's Republic of China.,Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, People's Republic of China
| | - Yanming Wang
- Tianjin Key Laboratory of Molecular Drug Research, College of Pharmacy, Nankai University College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China
| | - Li Meng
- Tianjin Key Laboratory of Molecular Drug Research, College of Pharmacy, Nankai University College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China
| | - Qingqing Huang
- Tianjin Key Laboratory of Molecular Drug Research, College of Pharmacy, Nankai University College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China
| | - Yueqi Zhu
- Department of Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, Shanghai, 200233, People's Republic of China
| | - Wenguo Cui
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, People's Republic of China.
| | - Yingsheng Cheng
- Department of Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, Shanghai, 200233, People's Republic of China.
| | - Ranlu Liu
- Tianjin Institute of Urology & Department of Urology, The Second Hospital of Tianjin Medical University, 23 Pingjiang Road, Hexi District, Tianjin, 300211, People's Republic of China.
| |
Collapse
|
21
|
α vβ 3-Specific Gold Nanoparticles for Fluorescence Imaging of Tumor Angiogenesis. NANOMATERIALS 2021; 11:nano11010138. [PMID: 33430079 PMCID: PMC7827626 DOI: 10.3390/nano11010138] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/22/2020] [Accepted: 12/30/2020] [Indexed: 11/17/2022]
Abstract
This paper reports on the development of tumor-specific gold nanoparticles (AuNPs) as theranostic tools intended for target accumulation and the detection of tumor angiogenesis via optical imaging (OI) before therapy is performed, being initiated via an external X-ray irradiation source. The AuNPs were decorated with a near-infrared dye, and RGD peptides as the tumor targeting vector for αvβ3-integrin, which is overexpressed in tissue with high tumor angiogenesis. The AuNPs were evaluated in an optical imaging setting in vitro and in vivo exhibiting favorable diagnostic properties with regards to tumor cell accumulation, biodistribution, and clearance. Furthermore, the therapeutic properties of the AuNPs were evaluated in vitro on pUC19 DNA and on A431 cells concerning acute and long-term toxicity, indicating that these AuNPs could be useful as radiosensitizers in therapeutic concepts in the future.
Collapse
|
22
|
Moody TW, Lee L, Ramos-Alvarez I, Iordanskaia T, Mantey SA, Jensen RT. Bombesin Receptor Family Activation and CNS/Neural Tumors: Review of Evidence Supporting Possible Role for Novel Targeted Therapy. Front Endocrinol (Lausanne) 2021; 12:728088. [PMID: 34539578 PMCID: PMC8441013 DOI: 10.3389/fendo.2021.728088] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022] Open
Abstract
G-protein-coupled receptors (GPCRs) are increasingly being considered as possible therapeutic targets in cancers. Activation of GPCR on tumors can have prominent growth effects, and GPCRs are frequently over-/ectopically expressed on tumors and thus can be used for targeted therapy. CNS/neural tumors are receiving increasing attention using this approach. Gliomas are the most frequent primary malignant brain/CNS tumor with glioblastoma having a 10-year survival <1%; neuroblastomas are the most common extracranial solid tumor in children with long-term survival<40%, and medulloblastomas are less common, but one subgroup has a 5-year survival <60%. Thus, there is an increased need for more effective treatments of these tumors. The Bombesin-receptor family (BnRs) is one of the GPCRs that are most frequently over/ectopically expressed by common tumors and is receiving particular attention as a possible therapeutic target in several tumors, particularly in prostate, breast, and lung cancer. We review in this paper evidence suggesting why a similar approach in some CNS/neural tumors (gliomas, neuroblastomas, medulloblastomas) should also be considered.
Collapse
Affiliation(s)
- Terry W. Moody
- Department of Health and Human Services, National Cancer Institute, Center for Cancer Training, Office of the Director, Bethesda, MD, United States
| | - Lingaku Lee
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
- Department of Gastroenterology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Irene Ramos-Alvarez
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Tatiana Iordanskaia
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Samuel A. Mantey
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Robert T. Jensen
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Robert T. Jensen,
| |
Collapse
|
23
|
Cell-penetrating peptides in oncologic pharmacotherapy: A review. Pharmacol Res 2020; 162:105231. [PMID: 33027717 DOI: 10.1016/j.phrs.2020.105231] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/23/2020] [Accepted: 09/30/2020] [Indexed: 01/10/2023]
Abstract
Cancer is the second leading cause of death in the world and its treatment is extremely challenging, mainly due to its complexity. Cell-Penetrating Peptides (CPPs) are peptides that can transport into the cell a wide variety of biologically active conjugates (or cargoes), and are, therefore, promising in the treatment and in the diagnosis of several types of cancer. Some notable examples are TAT and Penetratin, capable of penetrating the central nervous system (CNS) and, therefore, acting in cancers of this system, such as Glioblastoma Multiforme (GBM). These above-mentioned peptides, conjugated with traditional chemotherapeutic such as Doxorubicin (DOX) and Paclitaxel (PTX), have also been shown to induce apoptosis of breast and liver cancer cells, as well as in lung cancer cells, respectively. In other cancers, such as esophageal cancer, the attachment of Magainin 2 (MG2) to Bombesin (MG2B), another CPP, led to pronounced anticancer effects. Other examples are CopA3, that selectively decreased the viability of gastric cancer cells, and the CPP p28. Furthermore, in preclinical tests, the anti-tumor efficacy of this peptide was evaluated on human breast cancer, prostate cancer, ovarian cancer, and melanoma cells in vitro, leading to high expression of p53 and promoting cell cycle arrest. Despite the numerous in vitro and in vivo studies with promising results, and the increasing number of clinical trials using CPPs, few treatments reach the expected clinical efficacy. Usually, their clinical application is limited by its poor aqueous solubility, immunogenicity issues and dose-limiting toxicity. This review describes the most recent advances and innovations in the use of CPPs in several types of cancer, highlighting their crucial importance for various purposes, from therapeutic to diagnosis. Further clinical trials with these peptides are warranted to examine its effects on various types of cancer.
Collapse
|
24
|
Tooyserkani R, Lipiński W, Willemsen B, Löwik DWPM. Activation of cell-penetrating peptide fragments by disulfide formation. Amino Acids 2020; 52:1161-1168. [PMID: 32737661 PMCID: PMC7497323 DOI: 10.1007/s00726-020-02880-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/26/2020] [Indexed: 11/30/2022]
Abstract
Three cell-penetrating peptides (CPPs), Tat, Pep-3 and penetratin, were split into two parts and each fragment was terminated with a cysteine residue, to allow disulfide bridge formation, as well as a fluorescent label, for visualization and quantitative analysis. After disulfide formation between two complementary CPP fragments, cellular uptake of the resulting conjugates was observed. As confirmed by in vitro experiments, the conjugated peptides showed uptake activity comparable to the native CPP sequences, while the truncated peptides were hardly active. Until now, this split CPP strategy has only been demonstrated for oligo-arginine CPPs, but here we demonstrate that it is also applicable to other cell-penetrating peptides. This wider applicability may help in the design of new activatable cell-penetrating peptides for, e.g., targeted drug delivery.
Collapse
Affiliation(s)
- Raheleh Tooyserkani
- Radboud University Nijmegen, Institute for Molecules and Materials, Bio-Organic Chemistry, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Wojciech Lipiński
- Radboud University Nijmegen, Institute for Molecules and Materials, Bio-Organic Chemistry, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Bob Willemsen
- Radboud University Nijmegen, Institute for Molecules and Materials, Bio-Organic Chemistry, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Dennis W P M Löwik
- Radboud University Nijmegen, Institute for Molecules and Materials, Bio-Organic Chemistry, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands.
| |
Collapse
|
25
|
Yari H, Gali H, Awasthi V. Nanoparticles for Targeting of Prostate Cancer. Curr Pharm Des 2020; 26:5393-5413. [PMID: 32693761 DOI: 10.2174/1381612826666200721001500] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 05/27/2020] [Indexed: 11/22/2022]
Abstract
Prostate cancer (PCa) is the leading cause of death by cancer in men. Because of the drastic decline in the survival rate of PCa patients with advanced/metastatic disease, early diagnosis of disease and therapy without toxic side effects is crucial. Chemotherapy is widely used to control the progression of PCa at the later stages; however, it is associated with off-target toxicities and severe adverse effects due to the lack of specificity. Delivery of therapeutic or diagnostic agents by using targeted nanoparticles is a promising strategy to enhance accuracy and sensitivity of diagnosis of PCa and to increase efficacy and specificity of therapeutic agents. Numerous efforts have been made in past decades to create nanoparticles with different architectural bases for specific delivery payloads to prostate tumors. Major PCa associated cell membrane protein markers identified as targets for such purposes include folate receptor, sigma receptors, transferrin receptor, gastrin-releasing peptide receptor, urokinase plasminogen activator receptor, and prostate specific membrane antigen. Among these markers, prostate specific membrane antigen has emerged as an extremely specific and sensitive targetable marker for designing targeted nanoparticle-based delivery systems for PCa. In this article, we review contemporary advances in design, specificity, and efficacy of nanoparticles functionalized against PCa. Whenever feasible, both diagnostic as well as therapeutic applications are discussed.
Collapse
Affiliation(s)
- Hooman Yari
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Hariprasad Gali
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Vibhudutta Awasthi
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| |
Collapse
|
26
|
Datta P, Ray S. Nanoparticulate formulations of radiopharmaceuticals: Strategy to improve targeting and biodistribution properties. J Labelled Comp Radiopharm 2020; 63:333-355. [PMID: 32220029 DOI: 10.1002/jlcr.3839] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/17/2020] [Accepted: 03/08/2020] [Indexed: 02/06/2023]
Abstract
Application of nanotechnology principles in drug delivery has created opportunities for treatment of several diseases. Nanotechnology offers the advantage of overcoming the adverse biopharmaceutics or pharmacokinetic properties of drug molecules, to be determined by the transport properties of the particles themselves. Through the manipulation of size, shape, charge, and type of nanoparticle delivery system, variety of distribution profiles may be obtained. However, there still exists greater need to derive and standardize definitive structure property relationships for the distribution profiles of the delivery system. When applied to radiopharmaceuticals, the delivery systems assume greater significance. For the safety and efficacy of both diagnostics and therapeutic radiopharmaceuticals, selective localization in target tissue is even more important. At the same time, the synthesis and fabrication reactions of radiolabelled nanoparticles need to be completed in much shorter time. Moreover, the extensive understanding of the several interesting optical and magnetic properties of materials in nanoscale provides for achieving multiple objectives in nuclear medicine. This review discusses the various nanoparticle systems, which are applied for radionuclides and analyses the important bottlenecks that are required to be overcome for their more widespread clinical adaptation.
Collapse
Affiliation(s)
- Pallab Datta
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology Shibpur, Howrah, India
| | | |
Collapse
|
27
|
Salvanou EA, Stellas D, Tsoukalas C, Mavroidi B, Paravatou-Petsotas M, Kalogeropoulos N, Xanthopoulos S, Denat F, Laurent G, Bazzi R, Roux S, Bouziotis P. A Proof-of-Concept Study on the Therapeutic Potential of Au Nanoparticles Radiolabeled with the Alpha-Emitter Actinium-225. Pharmaceutics 2020; 12:pharmaceutics12020188. [PMID: 32098286 PMCID: PMC7076693 DOI: 10.3390/pharmaceutics12020188] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 01/20/2020] [Accepted: 02/19/2020] [Indexed: 01/01/2023] Open
Abstract
Actinium-225 (225Ac) is receiving increased attention for its application in targeted radionuclide therapy, due to the short range of its emitted alpha particles in conjunction with their high linear energy transfer, which lead to the eradication of tumor cells while sparing neighboring healthy tissue. The objective of our study was the evaluation of a gold nanoparticle radiolabeled with 225Ac as an injectable radiopharmaceutical form of brachytherapy for local radiation treatment of cancer. Au@TADOTAGA was radiolabeled with 225Ac at pH 5.6 (30 min at 70 °C), and in vitro stability was evaluated. In vitro cytotoxicity was assessed in U-87 MG cancer cells, and in vivo biodistribution was performed by intravenous and intratumoral administration of [225Ac]225Ac-Au@TADOTAGA in U-87 MG tumor-bearing mice. A preliminary study to assess therapeutic efficacy of the intratumorally-injected radio-nanomedicine was performed over a period of 22 days, while the necrotic effect on tumors was evaluated by a histopathology study. We have shown that [225Ac]225Ac-Au@TADOTAGA resulted in the retardation of tumor growth after its intratumoral injection in U87MG tumor-bearing mice, even though very low activities were injected per mouse. This gold nanoparticle radiopharmaceutical could be applied as an unconventional brachytherapy in injectable form for local radiation treatment of cancer.
Collapse
Affiliation(s)
- Evangelia-Alexandra Salvanou
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Center for Scientific Research “Demokritos”, 15341 Athens, Greece; (E.-A.S.); (C.T.); (M.P.-P.); (S.X.)
| | - Dimitris Stellas
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Charalampos Tsoukalas
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Center for Scientific Research “Demokritos”, 15341 Athens, Greece; (E.-A.S.); (C.T.); (M.P.-P.); (S.X.)
| | - Barbara Mavroidi
- Institute of Biosciences and Applications, National Center for Scientific Research “Demokritos”, 15341 Athens, Greece;
| | - Maria Paravatou-Petsotas
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Center for Scientific Research “Demokritos”, 15341 Athens, Greece; (E.-A.S.); (C.T.); (M.P.-P.); (S.X.)
| | | | - Stavros Xanthopoulos
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Center for Scientific Research “Demokritos”, 15341 Athens, Greece; (E.-A.S.); (C.T.); (M.P.-P.); (S.X.)
| | - Franck Denat
- ICMUB, UMR 6302 CNRS-UB, Université Bourgogne Franche-Comté, 21000 Dijon, France;
| | - Gautier Laurent
- Institut UTINAM, UMR 6213 CNRS-UBFC, Université Bourgogne Franche-Comté, 25030 Besançon, France; (G.L.); (R.B.); (S.R.)
| | - Rana Bazzi
- Institut UTINAM, UMR 6213 CNRS-UBFC, Université Bourgogne Franche-Comté, 25030 Besançon, France; (G.L.); (R.B.); (S.R.)
| | - Stephane Roux
- Institut UTINAM, UMR 6213 CNRS-UBFC, Université Bourgogne Franche-Comté, 25030 Besançon, France; (G.L.); (R.B.); (S.R.)
| | - Penelope Bouziotis
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Center for Scientific Research “Demokritos”, 15341 Athens, Greece; (E.-A.S.); (C.T.); (M.P.-P.); (S.X.)
- Correspondence: ; Tel.: +30-21-0650-3687
| |
Collapse
|
28
|
Majerova P, Hanes J, Olesova D, Sinsky J, Pilipcinec E, Kovac A. Novel Blood-Brain Barrier Shuttle Peptides Discovered through the Phage Display Method. Molecules 2020; 25:molecules25040874. [PMID: 32079185 PMCID: PMC7070575 DOI: 10.3390/molecules25040874] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/13/2020] [Accepted: 02/14/2020] [Indexed: 01/12/2023] Open
Abstract
Delivery of therapeutic agents into the brain is a major challenge in central nervous system drug development. The blood–brain barrier (BBB) prevents access of biotherapeutics to their targets in the central nervous system and, therefore, prohibits the effective treatment of many neurological disorders. To find blood–brain barrier shuttle peptides that could target therapeutics to the brain, we applied a phage display technology on a primary endothelial rat cellular model. Two identified peptides from a 12 mer phage library, GLHTSATNLYLH and VAARTGEIYVPW, were selected and their permeability was validated using the in vitro BBB model. The permeability of peptides through the BBB was measured by ultra-performance liquid chromatography-tandem mass spectrometry coupled to a triple-quadrupole mass spectrometer (UHPLC-MS/MS). We showed higher permeability for both peptides compared to N–C reversed-sequence peptides through in vitro BBB: for peptide GLHTSATNLYLH 3.3 × 10−7 cm/s and for peptide VAARTGEIYVPW 1.5 × 10−6 cm/s. The results indicate that the peptides identified by the in vitro phage display technology could serve as transporters for the administration of biopharmaceuticals into the brain. Our results also demonstrated the importance of proper BBB model for the discovery of shuttle peptides through phage display libraries.
Collapse
Affiliation(s)
- Petra Majerova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia; (P.M.); (J.H.); (D.O.); (J.S.)
| | - Jozef Hanes
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia; (P.M.); (J.H.); (D.O.); (J.S.)
| | - Dominika Olesova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia; (P.M.); (J.H.); (D.O.); (J.S.)
| | - Jakub Sinsky
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia; (P.M.); (J.H.); (D.O.); (J.S.)
| | - Emil Pilipcinec
- Department of Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04181 Kosice, Slovakia;
| | - Andrej Kovac
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia; (P.M.); (J.H.); (D.O.); (J.S.)
- Department of Pharmacology and Toxicology, The University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04181 Kosice, Slovakia
- Correspondence: ; Tel.: +421-254788100
| |
Collapse
|
29
|
Ferro-Flores G. Targeted Nanomedicines: In the Right Route Towards Improved Therapies. CURRENT CANCER THERAPY REVIEWS 2020. [DOI: 10.2174/1573394715666181224144500] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Guillermina Ferro-Flores
- Laboratorio Nacional de Investigacion y Desarrollo de Radiofarmacos CONACytT Instituto Nacional de Investigaciones Nucleares La Marquesa, 52750, Mexico
| |
Collapse
|
30
|
Silva F, Paulo A, Pallier A, Même S, Tóth É, Gano L, Marques F, Geraldes CF, Castro MMC, Cardoso AM, Jurado AS, López-Larrubia P, Lacerda S, Cabral Campello MP. Dual Imaging Gold Nanoplatforms for Targeted Radiotheranostics. MATERIALS 2020; 13:ma13030513. [PMID: 31978954 PMCID: PMC7040626 DOI: 10.3390/ma13030513] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/08/2020] [Accepted: 01/20/2020] [Indexed: 02/07/2023]
Abstract
Gold nanoparticles (AuNPs) are interesting for the design of new cancer theranostic tools, mainly due to their biocompatibility, easy molecular vectorization, and good biological half-life. Herein, we report a gold nanoparticle platform as a bimodal imaging probe, capable of coordinating Gd3+ for Magnetic Resonance Imaging (MRI) and 67Ga3+ for Single Photon Emission Computed Tomography (SPECT) imaging. Our AuNPs carry a bombesin analogue with affinity towards the gastrin releasing peptide receptor (GRPr), overexpressed in a variety of human cancer cells, namely PC3 prostate cancer cells. The potential of these multimodal imaging nanoconstructs was thoroughly investigated by the assessment of their magnetic properties, in vitro cellular uptake, biodistribution, and radiosensitisation assays. The relaxometric properties predict a potential T1- and T2- MRI application. The promising in vitro cellular uptake of 67Ga/Gd-based bombesin containing particles was confirmed through biodistribution studies in tumor bearing mice, indicating their integrity and ability to target the GRPr. Radiosensitization studies revealed the therapeutic potential of the nanoparticles. Moreover, the DOTA chelating unit moiety versatility gives a high theranostic potential through the coordination of other therapeutically interesting radiometals. Altogether, our nanoparticles are interesting nanomaterial for theranostic application and as bimodal T1- and T2- MRI / SPECT imaging probes.
Collapse
Affiliation(s)
- Francisco Silva
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Campus Tecnológico e Nuclear, Estrada Nacional 10, Km 139.7, 2695-066 Bobadela LRS, Portugal; (F.S.); (A.P.); (L.G.); (F.M.)
- Departamento de Engenharia e Ciências Nucleares (DECN), Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal
| | - António Paulo
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Campus Tecnológico e Nuclear, Estrada Nacional 10, Km 139.7, 2695-066 Bobadela LRS, Portugal; (F.S.); (A.P.); (L.G.); (F.M.)
- Departamento de Engenharia e Ciências Nucleares (DECN), Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal
| | - Agnès Pallier
- Centre de Biophysique Moléculaire, CNRS, UPR 4301, Université d’Orléans, Rue Charles Sadron, 45071 Orléans CEDEX 2, France; (A.P.); (S.M.)
| | - Sandra Même
- Centre de Biophysique Moléculaire, CNRS, UPR 4301, Université d’Orléans, Rue Charles Sadron, 45071 Orléans CEDEX 2, France; (A.P.); (S.M.)
| | - Éva Tóth
- Centre de Biophysique Moléculaire, CNRS, UPR 4301, Université d’Orléans, Rue Charles Sadron, 45071 Orléans CEDEX 2, France; (A.P.); (S.M.)
| | - Lurdes Gano
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Campus Tecnológico e Nuclear, Estrada Nacional 10, Km 139.7, 2695-066 Bobadela LRS, Portugal; (F.S.); (A.P.); (L.G.); (F.M.)
- Departamento de Engenharia e Ciências Nucleares (DECN), Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal
| | - Fernanda Marques
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Campus Tecnológico e Nuclear, Estrada Nacional 10, Km 139.7, 2695-066 Bobadela LRS, Portugal; (F.S.); (A.P.); (L.G.); (F.M.)
- Departamento de Engenharia e Ciências Nucleares (DECN), Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal
| | - Carlos F.G.C. Geraldes
- Department of Life Sciences, Faculty of Science and TechnologyUniversity of Coimbra, Calçada Martim de Freitas, 3000-393 Coimbra, Portugal (A.S.J.)
- Coimbra Chemistry Center, University of Coimbra, 3004-535 Coimbra, Portugal
- CIBIT/ICNAS Instituto de Ciências Nucleares Aplicadas à Saúde. Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - M. Margarida C.A. Castro
- Department of Life Sciences, Faculty of Science and TechnologyUniversity of Coimbra, Calçada Martim de Freitas, 3000-393 Coimbra, Portugal (A.S.J.)
- Coimbra Chemistry Center, University of Coimbra, 3004-535 Coimbra, Portugal
| | - Ana M. Cardoso
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal;
- Institute for Interdisciplinary Research of the University of Coimbra, 3030-789 Coimbra, Portugal
| | - Amália S. Jurado
- Department of Life Sciences, Faculty of Science and TechnologyUniversity of Coimbra, Calçada Martim de Freitas, 3000-393 Coimbra, Portugal (A.S.J.)
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal;
| | - Pilar López-Larrubia
- Instituto de Investigaciones Biomédicas “Alberto Sols” CSIC/UAM, c/ Arturo Duperier 4, 28029 Madrid, Spain;
| | - Sara Lacerda
- Centre de Biophysique Moléculaire, CNRS, UPR 4301, Université d’Orléans, Rue Charles Sadron, 45071 Orléans CEDEX 2, France; (A.P.); (S.M.)
- Correspondence: (M.P.C.C.); (S.L.)
| | - Maria Paula Cabral Campello
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Campus Tecnológico e Nuclear, Estrada Nacional 10, Km 139.7, 2695-066 Bobadela LRS, Portugal; (F.S.); (A.P.); (L.G.); (F.M.)
- Departamento de Engenharia e Ciências Nucleares (DECN), Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal
- Correspondence: (M.P.C.C.); (S.L.)
| |
Collapse
|
31
|
Lahooti A, Shanehsazzadeh S, Laurent S. Preliminary studies of 68Ga-NODA-USPION-BBN as a dual-modality contrast agent for use in positron emission tomography/magnetic resonance imaging. NANOTECHNOLOGY 2020; 31:015102. [PMID: 31519003 DOI: 10.1088/1361-6528/ab4446] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The aim of this study was to propose a new dual-modality nanoprobe for positron emission tomography/magnetic resonance imaging (PET/MRI) for the early diagnosis of breast cancer. For synthesis of the nanoprobe, polyethylene glycol-coated ultra-small superparamagnetic iron-oxide nanoparticles (USPION) armed with NODA-GA chelate and grafted with bombesin (BBN) were radiolabeled with 68Ga. After characterization, in vitro studies to evaluate the cell binding affinity of the nanoprobe were done by performing Perl's Prussian blue cell staining and MRI imaging. Finally, for in vivo studies, magnetic resonance images were taken in SCID mice bearing breast cancer tumor pre- and post-injection, and a multimodal nanoScan PET/computed tomography was used to perform preclinical imaging of the radiolabeled nanoparticles. Afterwards, a biodistribution study was done on sacrificed mice. The results showed that the highest r1 and r2 values were measured for USPIONs at 20 and 60 MHz, respectively. From the in vitro studies, the optical density of the cells after incubation increased with the increase of the iron concentration and the duration of incubation. However, the T2 values decreased when the iron concentration increased. Furthermore, from in vivo studies, the T2 and signal intensity decreased during the elapsed time post-injection in the tumor area. In this study, the in vitro studies showed that the affinity of cancer cells to nanoprobe increases meaningfully after conjugation with BBN, and also by increasing the duration of incubation and the iron concentration. Meanwhile, the in vivo results confirmed that the blood clearance of the nanoprobe happened during the first 120 min post-injection of the radiolabeled nanoprobe and also confirmed the targeting ability of that to a gastrin-releasing peptide receptor positive tumor.
Collapse
Affiliation(s)
- Afsaneh Lahooti
- NMR and Molecular Imaging Laboratory, Department of General, Organic and Biomedical Chemistry, University of Mons, 23 Place du Parc, B-7000, Mons, Belgium
| | | | | |
Collapse
|
32
|
Pérez-Medina C, Teunissen AJ, Kluza E, Mulder WJ, van der Meel R. Nuclear imaging approaches facilitating nanomedicine translation. Adv Drug Deliv Rev 2020; 154-155:123-141. [PMID: 32721459 DOI: 10.1016/j.addr.2020.07.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/08/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023]
Abstract
Nanomedicine approaches can effectively modulate the biodistribution and bioavailability of therapeutic agents, improving their therapeutic index. However, despite the ever-increasing amount of literature reporting on preclinical nanomedicine, the number of nanotherapeutics receiving FDA approval remains relatively low. Several barriers exist that hamper the effective preclinical evaluation and clinical translation of nanotherapeutics. Key barriers include insufficient understanding of nanomedicines' in vivo behavior, inadequate translation from murine models to larger animals, and a lack of patient stratification strategies. Integrating quantitative non-invasive imaging techniques in nanomedicine development offers attractive possibilities to address these issues. Among the available imaging techniques, nuclear imaging by positron emission tomography (PET) and single-photon emission computed tomography (SPECT) are highly attractive in this context owing to their quantitative nature and uncontested sensitivity. In basic and translational research, nuclear imaging techniques can provide critical quantitative information about pharmacokinetic parameters, biodistribution profiles or target site accumulation of nanocarriers and their associated payload. During clinical evaluation, nuclear imaging can be used to select patients amenable to nanomedicine treatment. Here, we review how nuclear imaging-based approaches are increasingly being integrated into nanomedicine development and discuss future developments that will accelerate their clinical translation.
Collapse
|
33
|
Delivery systems exploiting natural cell transport processes of macromolecules for intracellular targeting of Auger electron emitters. Nucl Med Biol 2019; 80-81:45-56. [PMID: 31810828 DOI: 10.1016/j.nucmedbio.2019.11.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/24/2019] [Accepted: 11/25/2019] [Indexed: 12/17/2022]
Abstract
The presence of Auger electrons (AE) among the decay products of a number of radionuclides makes these radionuclides an attractive means for treating cancer because these short-range electrons can cause significant damage in the immediate vicinity of the decomposition site. Moreover, the extreme locality of the effect provides a potential for selective eradication of cancer cells with minimal damage to adjacent normal cells provided that the delivery of the AE emitter to the most vulnerable parts of the cell can be achieved. Few cellular compartments have been regarded as the desired target site for AE emitters, with the cell nucleus generally recognized as the preferred site for AE decay due to the extreme sensitivity of nuclear DNA to direct damage by radiation of high linear energy transfer. Thus, the advantages of AE emitters for cancer therapy are most likely to be realized by their selective delivery into the nucleus of the malignant cells. To achieve this goal, delivery systems must combine a challenging complex of properties that not only provide cancer cell preferential recognition but also cell entry followed by transport into the cell nucleus. A promising strategy for achieving this is the recruitment of natural cell transport processes of macromolecules, involved in each of the aforementioned steps. To date, a number of constructs exploiting intracellular transport systems have been proposed for AE emitter delivery to the nucleus of a targeted cell. An example of such a multifunctional vehicle that provides smart step-by-step delivery is the so-called modular nanotransporter, which accomplishes selective recognition, binding, internalization, and endosomal escape followed by nuclear import of the delivered radionuclide. The current review will focus on delivery systems utilizing various intracellular transport pathways and their combinations in order to provide efficient targeting of AE to the cancer cell nucleus.
Collapse
|
34
|
Pooja D, Gunukula A, Gupta N, Adams DJ, Kulhari H. Bombesin receptors as potential targets for anticancer drug delivery and imaging. Int J Biochem Cell Biol 2019; 114:105567. [DOI: 10.1016/j.biocel.2019.105567] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 07/02/2019] [Accepted: 07/05/2019] [Indexed: 12/24/2022]
|
35
|
Gibbens-Bandala B, Morales-Avila E, Ferro-Flores G, Santos-Cuevas C, Meléndez-Alafort L, Trujillo-Nolasco M, Ocampo-García B. 177Lu-Bombesin-PLGA (paclitaxel): A targeted controlled-release nanomedicine for bimodal therapy of breast cancer. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 105:110043. [PMID: 31546458 DOI: 10.1016/j.msec.2019.110043] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 06/26/2019] [Accepted: 07/31/2019] [Indexed: 12/28/2022]
Abstract
The gastrin-releasing peptide receptor (GRPr) is overexpressed in >75% of breast cancers. 177Lu-Bombesin (177Lu-BN) has demonstrated the ability to target GRPr and facilitate efficient delivery of therapeutic radiation doses to malignant cells. Poly(d,l‑lactide‑co‑glycolide) acid (PLGA) nanoparticles can work as smart drug controlled-release systems activated through pH changes. Considering that paclitaxel (PTX) is a first-line drug for cancer treatment, this work aimed to synthesize and chemically characterize a novel polymeric PTX-loaded nanosystem with grafted 177Lu-BN and to evaluate its performance as a targeted controlled-release nanomedicine for concomitant radiotherapy and chemotherapy of breast cancer. PLGA(PTX) nanoparticles were synthesized using the single emulsification-solvent evaporation method with PVA as a stabilizer in the presence of PTX. Thereafter, the activation of PLGA carboxylic groups for BN attachment through the Lys1-amine group was performed. Results of the chemical characterization by FT-IR, DLS, HPLC and SEM/TEM demonstrated the successful synthesis of BN-PLGA(PTX) with a hydrodynamic diameter of 163.54 ± 33.25 nm. The entrapment efficiency of paclitaxel was 92.8 ± 3.6%. The nanosystem showed an adequate controlled release of the anticancer drug, which increased significantly due to the pH change from neutral (pH = 7.4) to acidic conditions (pH = 5.3). After labeling with 177Lu and purification by ultrafiltration, 177Lu-BN-PLGA(PTX) was obtained with a radiochemical purity of 99 ± 1%. In vitro and in vivo studies using MDA-MB-231 breast cancer cells (GRPr-positive) demonstrated a 177Lu-BN-PLGA(PTX) specific uptake and a significantly higher cytotoxic effect for the radiolabeled nanosystem than the unlabeled BN-PLGA(PTX) nanoparticles. Using a pulmonary micrometastasis MDA-MB-231 model, the added value of 177Lu-BN-PLGA(PTX) for tumor imaging was confirmed. The 177Lu-BN-PLGA(PTX) nanomedicine is suitable as a targeted paclitaxel delivery system with concomitant radiotherapeutic effect for the treatment of GRPr-positive breast cancer.
Collapse
Affiliation(s)
- Brenda Gibbens-Bandala
- Departamento de Materiales Radiactivos, Instituto Nacional de Investigaciones Nucleares, Carretera México-Toluca S/N, Ocoyoacac, Estado de México 52750, Mexico; Facultad de Química, Universidad Autónoma del Estado de México, Paseo Tollocan S/N, Toluca, Estado de México 50180, Mexico
| | - Enrique Morales-Avila
- Facultad de Química, Universidad Autónoma del Estado de México, Paseo Tollocan S/N, Toluca, Estado de México 50180, Mexico
| | - Guillermina Ferro-Flores
- Departamento de Materiales Radiactivos, Instituto Nacional de Investigaciones Nucleares, Carretera México-Toluca S/N, Ocoyoacac, Estado de México 52750, Mexico
| | - Clara Santos-Cuevas
- Departamento de Materiales Radiactivos, Instituto Nacional de Investigaciones Nucleares, Carretera México-Toluca S/N, Ocoyoacac, Estado de México 52750, Mexico
| | | | - Maydelid Trujillo-Nolasco
- Departamento de Materiales Radiactivos, Instituto Nacional de Investigaciones Nucleares, Carretera México-Toluca S/N, Ocoyoacac, Estado de México 52750, Mexico; Facultad de Química, Universidad Autónoma del Estado de México, Paseo Tollocan S/N, Toluca, Estado de México 50180, Mexico
| | - Blanca Ocampo-García
- Departamento de Materiales Radiactivos, Instituto Nacional de Investigaciones Nucleares, Carretera México-Toluca S/N, Ocoyoacac, Estado de México 52750, Mexico.
| |
Collapse
|
36
|
Synthesis and preclinical evaluation of the 177Lu-DOTA-PSMA(inhibitor)-Lys3-bombesin heterodimer designed as a radiotheranostic probe for prostate cancer. Nucl Med Commun 2019; 40:278-286. [PMID: 30763290 DOI: 10.1097/mnm.0000000000000966] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKROUND Human tumors show intrinsic heterogeneity and changes in phenotype during disease progression, which implies different expression levels of cell surface receptors. The research on new heterodimeric lutetium-177 (Lu)-radiopharmaceuticals interacting with two different targets on tumor cells is a strategy for improvement of radiotheranostic performance. This study aimed to synthesize and characterize the Lu-DOTA-PSMA(inhibitor)-Lys-bombesin (Lu-DOTA-iPSMA-Lys-BN) heterodimer and to evaluate its potential to target prostate-specific membrane antigen (PSMA) and gastrin-releasing peptide receptor (GRPr) overexpressed in prostate cancer. METHODS The heterodimeric conjugate was synthesized and characterized by infrarred, mass, and H-NMR spectroscopies. The ligand was labeled with Lu and the radiochemical purity was assessed by radio-high-performance liquid chromatography. PSMA/GRPr affinity and the heterobivalent effect on cell viability were evaluated in LNCaP and PC3 prostate cancer cell lines. The biodistribution profile (3 and 96 h) was assessed in athymic mice with induced prostate tumors. Using pulmonary LNCaP (PSMA-positive) and PC3 (GRPr-negative) micrometastasis models, the influence of heterobivalency and affinity on tumor uptake was quantified (micro-SPECT/CT). RESULTS Lu-iPSMA-BN (radiochemical purity>98%) showed specific recognition for PSMA and GRPr (IC50=5.62 and 3.49 nmol/l, respectively) with a significant decrease in cell viability (10.15% of cell viability in LNCaP and 40.10% in PC3 at 48 h), as well as high LNCaP and PC3 tumor uptake (5.21 and 3.21% ID/g at 96 h, respectively). Micro-SPECT/CT imaging showed the heterodimer ability to target the tumors (SUVmax of 1.93±0.30 and 1.76±0.10 in LNCaP and PC3, respectively), possibly influenced by the heterobivalent effect. Lu-DOTA-iPSMA-Lys-BN showed suitable affinity for PSMA and GRPr. CONCLUSION The results warrant further preclinical studies to establish the Lu-radiotracer theranostic efficacy.
Collapse
|
37
|
Ottemann BM, Helmink AJ, Zhang W, Mukadam I, Woldstad C, Hilaire JR, Liu Y, McMillan JM, Edagwa BJ, Mosley RL, Garrison JC, Kevadiya BD, Gendelman HE. Bioimaging predictors of rilpivirine biodistribution and antiretroviral activities. Biomaterials 2018; 185:174-193. [PMID: 30245386 PMCID: PMC6556898 DOI: 10.1016/j.biomaterials.2018.09.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 09/05/2018] [Accepted: 09/11/2018] [Indexed: 12/22/2022]
Abstract
Antiretroviral therapy (ART) has changed the outcome of human immunodeficiency virus type one (HIV-1) infection from certain death to a life free of disease co-morbidities. However, infected people must remain on life-long daily ART. ART reduces but fails to eliminate the viral reservoir. In order to improve upon current treatment regimens, our laboratory created long acting slow effective release (LASER) ART nanoformulated prodrugs from native medicines. LASER ART enables antiretroviral drugs (ARVs) to better reach target sites of HIV-1 infection while, at the same time, improve ART's half-life and potency. However, novel ARV design has been slowed by prolonged pharmacokinetic testing requirements. To such ends, tri-modal theranostic nanoparticles were created with single-photon emission computed tomography (SPECT/CT), magnetic resonance imaging (MRI) and fluorescence capabilities to predict LASER ART biodistribution. The created theranostic ARV probes were then employed to monitor drug tissue distribution and potency. Intrinsically 111Indium (111In) radiolabeled, europium doped cobalt-ferrite particles and rilpivirine were encased in a polycaprolactone core surrounded by a lipid shell (111InEuCF-RPV). Particle cell and tissue distribution, and antiretroviral activities were sustained in macrophage tissue depots. 111InEuCF-PCL/RPV particles injected into mice demonstrated co-registration of MRI and SPECT/CT tissue signals with RPV and cobalt. Cell and animal particle biodistribution paralleled ARV activities. We posit that particle selection can predict RPV distribution and potency facilitated by multifunctional theranostic nanoparticles.
Collapse
Affiliation(s)
- Brendan M Ottemann
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Austin J Helmink
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Wenting Zhang
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Insiya Mukadam
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - James R Hilaire
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yutong Liu
- Department of Radiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - JoEllyn M McMillan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Benson J Edagwa
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - R Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jered C Garrison
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bhavesh D Kevadiya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA; Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
38
|
Farzin L, Sheibani S, Moassesi ME, Shamsipur M. An overview of nanoscale radionuclides and radiolabeled nanomaterials commonly used for nuclear molecular imaging and therapeutic functions. J Biomed Mater Res A 2018; 107:251-285. [PMID: 30358098 DOI: 10.1002/jbm.a.36550] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 08/08/2018] [Accepted: 09/03/2018] [Indexed: 02/06/2023]
Abstract
Recent advances in the field of nanotechnology applications in nuclear medicine offer the promise of better diagnostic and therapeutic options. In recent years, increasing efforts have been focused on developing nanoconstructs that can be used as core platforms for attaching medical radionuclides with different strategies for the purposes of molecular imaging and targeted drug delivery. This review article presents an introduction to some commonly used nanomaterials with zero-dimensional, one-dimensional, two-dimensional, and three-dimensional structures, describes the various methods applied to radiolabeling of nanomaterials, and provides illustrative examples of application of the nanoscale radionuclides or radiolabeled nanocarriers in nuclear nanomedicine. Especially, the passive and active nanotargeting delivery of radionuclides with illustrating examples for tumor imaging and therapy was reviewed and summarized. The accurate and early diagnosis of cancer can lead to increased survival rates for different types of this disease. Although, the conventional single-modality diagnostic methods such as positron emission tomography/single photon emission computed tomography or MRI used for such purposes are powerful means; most of these are limited by sensitivity or resolution. By integrating complementary signal reporters into a single nanoparticulate contrast agent, multimodal molecular imaging can be performed as scalable images with high sensitivity, resolution, and specificity. The advent of radiolabeled nanocarriers or radioisotope-loaded nanomaterials with magnetic, plasmonic, or fluorescent properties has stimulated growing interest in the developing multimodality imaging probes. These new developments in nuclear nanomedicine are expected to introduce a paradigm shift in multimodal molecular imaging and thereby opening up an era of new diagnostic medical imaging agents. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 251-285, 2019.
Collapse
Affiliation(s)
- Leila Farzin
- Radiation Application Research School, Nuclear Science and Technology Research Institute, Tehran, Iran
| | - Shahab Sheibani
- Radiation Application Research School, Nuclear Science and Technology Research Institute, Tehran, Iran
| | - Mohammad Esmaeil Moassesi
- Radiation Application Research School, Nuclear Science and Technology Research Institute, Tehran, Iran
| | | |
Collapse
|
39
|
González-Ruíz A, Ferro-Flores G, Jiménez-Mancilla N, Escudero-Castellanos A, Ocampo-García B, Luna-Gutiérrez M, Santos-Cuevas C, Morales-Avila E, Isaac-Olivé K. In vitro and in vivo synergistic effect of radiotherapy and plasmonic photothermal therapy on the viability of cancer cells using 177Lu–Au-NLS-RGD-Aptamer nanoparticles under laser irradiation. J Radioanal Nucl Chem 2018. [DOI: 10.1007/s10967-018-6266-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
40
|
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
|
41
|
Mendoza-Nava H, Ferro-Flores G, Ramírez FDM, Ocampo-García B, Santos-Cuevas C, Azorín-Vega E, Jiménez-Mancilla N, Luna-Gutiérrez M, Isaac-Olivé K. Fluorescent, Plasmonic, and Radiotherapeutic Properties of the 177Lu-Dendrimer-AuNP-Folate-Bombesin Nanoprobe Located Inside Cancer Cells. Mol Imaging 2018; 16:1536012117704768. [PMID: 28654384 PMCID: PMC5469519 DOI: 10.1177/1536012117704768] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The integration of fluorescence and plasmonic properties into one molecule is of importance in developing multifunctional imaging and therapy nanoprobes. The aim of this research was to evaluate the fluorescent properties and the plasmonic–photothermal, therapeutic, and radiotherapeutic potential of 177Lu–dendrimer conjugated to folate and bombesin with gold nanoparticles in the dendritic cavity (177Lu–DenAuNP–folate–bombesin) when it is internalized in T47D breast cancer cells. The intense near-Infrared (NIR) fluorescence emitted at 825 nm from the conjugate inside cells corroborated the usefulness of DenAuNP–folate–bombesin for optical imaging. After laser irradiation, the presence of the nanosystem in cells caused a significant increase in the temperature of the medium (46.8°C, compared to 39.1°C without DenAuNP–folate–bombesin, P < 0.05), resulting in a significant decrease in cell viability (down to 16.51% ± 1.52%) due to the 177Lu–DenAuNP–folate–bombesin plasmonic properties. After treatment with 177Lu–DenAuNP–folate–bombesin, the T47D cell viability decreased 90% because of the radiation-absorbed dose (63.16 ± 4.20 Gy) delivered inside the cells. The 177Lu–DenAuNP–folate–bombesin nanoprobe internalized in cancer cells exhibited properties suitable for optical imaging, plasmonic–photothermal therapy, and targeted radiotherapy.
Collapse
Affiliation(s)
- Héctor Mendoza-Nava
- 1 Departamento de Materiales Radiactivos, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac, Estado de México, Mexico.,2 Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Mexico
| | - Guillermina Ferro-Flores
- 1 Departamento de Materiales Radiactivos, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac, Estado de México, Mexico
| | - Flor de María Ramírez
- 3 Departamento de Química, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac, Estado de México, Mexico
| | - Blanca Ocampo-García
- 1 Departamento de Materiales Radiactivos, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac, Estado de México, Mexico
| | - Clara Santos-Cuevas
- 1 Departamento de Materiales Radiactivos, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac, Estado de México, Mexico
| | - Erika Azorín-Vega
- 1 Departamento de Materiales Radiactivos, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac, Estado de México, Mexico
| | | | - Myrna Luna-Gutiérrez
- 1 Departamento de Materiales Radiactivos, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac, Estado de México, Mexico
| | - Keila Isaac-Olivé
- 2 Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Mexico
| |
Collapse
|
42
|
Hernández B, Coïc YM, López-Tobar E, Sanchez-Cortes S, Baron B, Pflüger F, Kruglik SG, Cohen R, Ghomi M. Dynamical Behavior of Somatostatin-14 and Its Cyclic Analogues as Analyzed in Bulk and on Plasmonic Silver Nanoparticles. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2018; 112:81-121. [PMID: 29680244 DOI: 10.1016/bs.apcsb.2018.01.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Primarily known as the inhibitor of growth hormone release, the role of somatostatin in many other inhibiting activities upon binding to its five G-protein-coupled receptors has been elucidated. Because of the short half-life of somatostatin, a number of synthetic analogues were elaborated for this peptide hormone. Herein, after recalling the main somatostatin therapeutic interests, we present the dynamical behavior of somatostatin-14 and its two currently used synthetic cyclic analogues, octreotide and pasireotide. Physical techniques, such as fluorescence, UV-visible absorption, circular dichroism, Raman spectroscopy, surface-enhanced Raman spectroscopy, and transmission electron microscopy, were jointly used in order to get information on the solution structural features, as well as on the anchoring sites of the three peptides on silver colloids. While somatostatin-14 adopts a rather unordered chain within the submillimolar concentration range, its cyclic analogues were revealed to be ordered, i.e., stabilized either in a type-II' β-turn (octreotide) or in a face-to-face γ-turn/type-I β-turn (pasireotide) structure. Nevertheless, a progressive structuring trend was observed in somatostatin-14 upon increasing concentration to the millimolar range. Because of their cationic character, the three peptides have revealed their capability to bind onto negatively charged silver nanoparticles. The high affinity of the peptides toward metallic particles seems to be extremely promising for the elaboration of somatostatin-based functionalized plasmonic nanoparticles that can be used in diagnosis, drug delivery, and therapy.
Collapse
Affiliation(s)
- Belén Hernández
- Laboratoire Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UMR 7369, Université de Reims, Faculté des Sciences, Reims Cedex 2, France; Groupe de Biophysique Moléculaire, Sorbonne Paris Cité, Université Paris 13, UFR Santé-Médecine-Biologie Humaine, Bobigny, France
| | - Yves-Marie Coïc
- Institut Pasteur, Unité de Chimie des Biomolécules, UMR 3523, Paris Cedex 15, France
| | | | | | - Bruno Baron
- Institut Pasteur, Plate-Forme de Biophysique de Macromolécules et de leurs Interactions, Paris Cedex 15, France
| | - Fernando Pflüger
- Groupe de Biophysique Moléculaire, Sorbonne Paris Cité, Université Paris 13, UFR Santé-Médecine-Biologie Humaine, Bobigny, France
| | - Sergei G Kruglik
- Laboratoire Jean-Perrin, Sorbonne Université, UPMC Univ. Paris 06, CNRS UMR 8237, Paris, France
| | - Régis Cohen
- Service d'Endocrinologie, Centre Hospitalier de Saint-Denis, Saint-Denis, France
| | - Mahmoud Ghomi
- Laboratoire Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UMR 7369, Université de Reims, Faculté des Sciences, Reims Cedex 2, France; Groupe de Biophysique Moléculaire, Sorbonne Paris Cité, Université Paris 13, UFR Santé-Médecine-Biologie Humaine, Bobigny, France.
| |
Collapse
|
43
|
Simpson EJ, Gobbo P, Bononi FC, Murrell E, Workentin MS, Luyt LG. Bombesin-functionalized water-soluble gold nanoparticles for targeting prostate cancer. ACTA ACUST UNITED AC 2017. [DOI: 10.1002/jin2.33] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Emily J. Simpson
- Western University Chemistry; 1151 Richmond Street London Ontario N6A 5B7 Canada
| | - Pierangelo Gobbo
- Western University Chemistry; 1151 Richmond Street London Ontario N6A 5B7 Canada
| | - Fernanda C. Bononi
- Western University Chemistry; 1151 Richmond Street London Ontario N6A 5B7 Canada
| | - Emily Murrell
- Western University Chemistry; 1151 Richmond Street London Ontario N6A 5B7 Canada
| | - Mark S. Workentin
- Western University Chemistry; 1151 Richmond Street London Ontario N6A 5B7 Canada
| | - Leonard G. Luyt
- Western University Chemistry; 1151 Richmond Street London Ontario N6A 5B7 Canada
- London Regional Cancer Program; Western University Oncology; 790 Commissioners Rd. E London Ontario N6A 4L6 Canada
| |
Collapse
|
44
|
Multimodal molecular 3D imaging for the tumoral volumetric distribution assessment of folate-based biosensors. Med Biol Eng Comput 2017; 56:1135-1148. [PMID: 29192382 DOI: 10.1007/s11517-017-1755-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 11/20/2017] [Indexed: 12/21/2022]
Abstract
The aim of this study was to characterize the in vivo volumetric distribution of three folate-based biosensors by different imaging modalities (X-ray, fluorescence, Cerenkov luminescence, and radioisotopic imaging) through the development of a tridimensional image reconstruction algorithm. The preclinical and multimodal Xtreme imaging system, with a Multimodal Animal Rotation System (MARS), was used to acquire bidimensional images, which were processed to obtain the tridimensional reconstruction. Images of mice at different times (biosensor distribution) were simultaneously obtained from the four imaging modalities. The filtered back projection and inverse Radon transformation were used as main image-processing techniques. The algorithm developed in Matlab was able to calculate the volumetric profiles of 99mTc-Folate-Bombesin (radioisotopic image), 177Lu-Folate-Bombesin (Cerenkov image), and FolateRSense™ 680 (fluorescence image) in tumors and kidneys of mice, and no significant differences were detected in the volumetric quantifications among measurement techniques. The imaging tridimensional reconstruction algorithm can be easily extrapolated to different 2D acquisition-type images. This characteristic flexibility of the algorithm developed in this study is a remarkable advantage in comparison to similar reconstruction methods.
Collapse
|
45
|
González-Ruíz A, Ferro-Flores G, Azorín-Vega E, Ocampo-García B, Ramírez FDM, Santos-Cuevas C, De León-Rodríguez L, Isaac-Olivé K, Luna-Gutiérrez M, Morales-Ávila E. Synthesis and in vitro evaluation of an antiangiogenic cancer-specific dual-targeting 177Lu-Au-nanoradiopharmaceutical. J Radioanal Nucl Chem 2017. [DOI: 10.1007/s10967-017-5465-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
46
|
Papagiannopoulou D. Technetium-99m radiochemistry for pharmaceutical applications. J Labelled Comp Radiopharm 2017; 60:502-520. [PMID: 28618064 DOI: 10.1002/jlcr.3531] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 06/08/2017] [Accepted: 06/08/2017] [Indexed: 12/14/2022]
Abstract
Technetium-99m (99m Tc) is a widely used radionuclide, and the development of 99m Tc imaging agents continues to be in demand. This overview discusses basic principles of 99m Tc radiopharmaceutical preparation and design and focuses on the 99m Tc radiochemistry relevant to its pharmaceutical applications. The 99m Tc complexes are described based on the most typical examples in each category, keeping up with the state-of-the-art in the field. In addition, the main current strategies to develop targeted 99m Tc radiopharmaceuticals are summarized.
Collapse
Affiliation(s)
- Dionysia Papagiannopoulou
- Department of Pharmaceutical Chemistry, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
47
|
Elgqvist J. Nanoparticles as Theranostic Vehicles in Experimental and Clinical Applications-Focus on Prostate and Breast Cancer. Int J Mol Sci 2017; 18:E1102. [PMID: 28531102 PMCID: PMC5455010 DOI: 10.3390/ijms18051102] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 05/13/2017] [Accepted: 05/15/2017] [Indexed: 12/27/2022] Open
Abstract
Prostate and breast cancer are the second most and most commonly diagnosed cancer in men and women worldwide, respectively. The American Cancer Society estimates that during 2016 in the USA around 430,000 individuals were diagnosed with one of these two types of cancers, and approximately 15% of them will die from the disease. In Europe, the rate of incidences and deaths are similar to those in the USA. Several different more or less successful diagnostic and therapeutic approaches have been developed and evaluated in order to tackle this issue and thereby decrease the death rates. By using nanoparticles as vehicles carrying both diagnostic and therapeutic molecular entities, individualized targeted theranostic nanomedicine has emerged as a promising option to increase the sensitivity and the specificity during diagnosis, as well as the likelihood of survival or prolonged survival after therapy. This article presents and discusses important and promising different kinds of nanoparticles, as well as imaging and therapy options, suitable for theranostic applications. The presentation of different nanoparticles and theranostic applications is quite general, but there is a special focus on prostate cancer. Some references and aspects regarding breast cancer are however also presented and discussed. Finally, the prostate cancer case is presented in more detail regarding diagnosis, staging, recurrence, metastases, and treatment options available today, followed by possible ways to move forward applying theranostics for both prostate and breast cancer based on promising experiments performed until today.
Collapse
Affiliation(s)
- Jörgen Elgqvist
- Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden.
- Department of Physics, University of Gothenburg, 412 96 Gothenburg, Sweden.
| |
Collapse
|
48
|
Lai P, Lechtman E, Mashouf S, Pignol JP, Reilly RM. Depot system for controlled release of gold nanoparticles with precise intratumoral placement by permanent brachytherapy seed implantation (PSI) techniques. Int J Pharm 2016; 515:729-739. [DOI: 10.1016/j.ijpharm.2016.11.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 10/28/2016] [Accepted: 11/01/2016] [Indexed: 12/29/2022]
|
49
|
Jin CS, Overchuk M, Cui L, Wilson BC, Bristow RG, Chen J, Zheng G. Nanoparticle-Enabled Selective Destruction of Prostate Tumor Using MRI-Guided Focal Photothermal Therapy. Prostate 2016; 76:1169-81. [PMID: 27198587 DOI: 10.1002/pros.23203] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 04/27/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND The Magnetic Resonance Imaging (MRI)-guided focal laser therapy has shown early promise in Phase 1 trial treating low/intermediate-risk localized prostate cancer (PCa), but the lack of tumor selectivity and low efficiency of heat generation remain as drawbacks of agent-free laser therapy. Intrinsic multifunctional porphyrin-nanoparticles (porphysomes) have been exploited to treat localized PCa by MRI-guided focal photothermal therapy (PTT) with significantly improved efficiency and tumor selectivity over prior methods of PTT, providing an effective and safe alternative to active surveillance or radical therapy. METHODS The tumor accumulation of porphysomes chelated with copper-64 was determined and compared with the clinic standard (18) F-FDG in an orthotropic PCa mouse model by positron emission tomography (PET) imaging, providing quantitative assessment for PTT dosimetry. The PTT was conducted with MRI-guided light delivery and monitored by MR thermometry, mimicking the clinical protocol. The efficacy of treatment and adverse effects to surround tissues were evaluated by histology analysis and tumor growth in survival study via MRI. RESULTS Porphysomes showed superior tumor-to-prostate selectivity over (18) F-FDG (6:1 vs. 0.36:1). MR thermometry detected tumor temperature increased to ≥55°C within 2 min (671 nm at 500 mW), but minimal increase in surrounding tissues. Porphysome enabled effective PTT eradication of tumor without damaging adjacent organs in orthotropic PCa mouse model. CONCLUSIONS Porphysome-enabled MRI-guided focal PTT could be an effective and safe approach to treat PCa at low risk of progression, thus addressing the significant unmet clinical needs and benefiting an ever-growing number of patients who may be over-treated and risk unnecessary side effects from radical therapies. Prostate 76:1169-1181, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Cheng S Jin
- Princess Margaret Cancer Center, UHN, Toronto, Canada
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, University of Toronto, Toronto, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Marta Overchuk
- Princess Margaret Cancer Center, UHN, Toronto, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Liyang Cui
- Princess Margaret Cancer Center, UHN, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Medical Isotopes Research Center, Peking University, Beijing, China
| | - Brian C Wilson
- Princess Margaret Cancer Center, UHN, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Robert G Bristow
- Princess Margaret Cancer Center, UHN, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Juan Chen
- Princess Margaret Cancer Center, UHN, Toronto, Canada
| | - Gang Zheng
- Princess Margaret Cancer Center, UHN, Toronto, Canada
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, University of Toronto, Toronto, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| |
Collapse
|
50
|
Reissig F, Mamat C, Steinbach J, Pietzsch HJ, Freudenberg R, Navarro-Retamal C, Caballero J, Kotzerke J, Wunderlich G. Direct and Auger Electron-Induced, Single- and Double-Strand Breaks on Plasmid DNA Caused by 99mTc-Labeled Pyrene Derivatives and the Effect of Bonding Distance. PLoS One 2016; 11:e0161973. [PMID: 27583677 PMCID: PMC5008623 DOI: 10.1371/journal.pone.0161973] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 08/15/2016] [Indexed: 11/29/2022] Open
Abstract
It is evident that 99mTc causes radical-mediated DNA damage due to Auger electrons, which were emitted simultaneously with the known γ-emission of 99mTc. We have synthesized a series of new 99mTc-labeled pyrene derivatives with varied distances between the pyrene moiety and the radionuclide. The pyrene motif is a common DNA intercalator and allowed us to test the influence of the radionuclide distance on damages of the DNA helix. In general, pUC 19 plasmid DNA enables the investigation of the unprotected interactions between the radiotracers and DNA that results in single-strand breaks (SSB) or double-strand breaks (DSB). The resulting DNA fragments were separated by gel electrophoresis and quantified by fluorescent staining. Direct DNA damage and radical-induced indirect DNA damage by radiolysis products of water were evaluated in the presence or absence of the radical scavenger DMSO. We demonstrated that Auger electrons directly induced both SSB and DSB in high efficiency when 99mTc was tightly bound to the plasmid DNA and this damage could not be completely prevented by DMSO, a free radical scavenger. For the first time, we were able to minimize this effect by increasing the carbon chain lengths between the pyrene moiety and the 99mTc nuclide. However, a critical distance between the 99mTc atom and the DNA helix could not be determined due to the significantly lowered DSB generation resulting from the interaction which is dependent on the type of the 99mTc binding motif. The effect of variable DNA damage caused by the different chain length between the pyrene residue and the Tc-core as well as the possible conformations of the applied Tc-complexes was supplemented with molecular dynamics (MD) calculations. The effectiveness of the DNA-binding 99mTc-labeled pyrene derivatives was demonstrated by comparison to non-DNA-binding 99mTcO4–, since nearly all DNA damage caused by 99mTcO4– was prevented by incubating with DMSO.
Collapse
Affiliation(s)
- Falco Reissig
- University Hospital/ Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Department of Nuclear Medicine, Dresden, Germany
- * E-mail: (GW); (FR)
| | - Constantin Mamat
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Joerg Steinbach
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Hans-Juergen Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Robert Freudenberg
- University Hospital/ Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Department of Nuclear Medicine, Dresden, Germany
| | - Carlos Navarro-Retamal
- Centro de Bioinformática y Simulación Molecular, Facultad de Ingeniería, Universidad de Talca, 2 Norte 685, Casilla 721, Talca, Chile
| | - Julio Caballero
- Centro de Bioinformática y Simulación Molecular, Facultad de Ingeniería, Universidad de Talca, 2 Norte 685, Casilla 721, Talca, Chile
| | - Joerg Kotzerke
- University Hospital/ Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Department of Nuclear Medicine, Dresden, Germany
| | - Gerd Wunderlich
- University Hospital/ Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Department of Nuclear Medicine, Dresden, Germany
- * E-mail: (GW); (FR)
| |
Collapse
|