51
|
Patil Y, Shmeeda H, Amitay Y, Ohana P, Kumar S, Gabizon A. Targeting of folate-conjugated liposomes with co-entrapped drugs to prostate cancer cells via prostate-specific membrane antigen (PSMA). NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:1407-1416. [DOI: 10.1016/j.nano.2018.04.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 02/23/2018] [Accepted: 04/10/2018] [Indexed: 01/23/2023]
|
52
|
Kouwenberg J, de Pooter J, Wolterbeek H, Denkova A, Bos A. Alpha radiation dosimetry using Fluorescent Nuclear Track Detectors. RADIAT MEAS 2018. [DOI: 10.1016/j.radmeas.2018.04.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
53
|
Pruszynski M, D'Huyvetter M, Bruchertseifer F, Morgenstern A, Lahoutte T. Evaluation of an Anti-HER2 Nanobody Labeled with 225Ac for Targeted α-Particle Therapy of Cancer. Mol Pharm 2018; 15:1457-1466. [PMID: 29502411 DOI: 10.1021/acs.molpharmaceut.7b00985] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Human epidermal growth factor receptor type 2 (HER2) is overexpressed in numerous carcinomas. Nanobodies (Nbs) are the smallest antibody-derived fragments with beneficial characteristics for molecular imaging and radionuclide therapy. Therefore, HER2-targeting nanobodies could offer a valuable platform for radioimmunotherapy, especially when labeled with α-particle emitters, which provide highly lethal and localized radiation to targeted cells with minimal exposure to surrounding healthy tissues. In this study, the anti-HER2 2Rs15d-nanobody was conjugated with 2-(4-isothiocyanatobenzyl)-1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid ( p-SCN-Bn-DOTA) and radiolabeled with an α-emitter 225Ac with a high yield (>90%) and a radiochemical purity above 95%. The 225Ac-DOTA-Nb binding affinity was 4.12 ± 0.47 nM with an immunoreactive fraction above 80%. Binding to low HER2-expressing MDA-MB-231 cells was negligible, whereas HER2-overexpressing SKOV-3 cells could be blocked with an excess of unlabeled nanobody, confirming the specificity of binding. Noncompeting binding to HER2 was observed in the presence of an excess of trastuzumab. The cell-associated fraction of 225Ac-DOTA-Nb was 34.72 ± 16.66% over 24 h. In vitro, the radioconjugate was toxic in an HER2-mediated and dose-dependent manner, resulting in IC50 values of 10.2 and 322.1 kBq/mL for 225Ac-DOTA-Nb and the 225Ac-DOTA control, respectively, on SKOV-3 cells, and 282.2 kBq/mL for 225Ac-DOTA-Nb on MDA-MB-231 cells. Ex vivo biodistribution studies, performed in mice bearing subcutaneous HER2-overexpressing and low HER2-expressing tumors, showed a fast uptake in SKOV-3 tumors compared to MDA-MB-231 (4.01 ± 1.58% ID/g vs 0.49 ± 0.20% ID/g after 2 h), resulting also in high tumor-to-normal tissue ratios. In addition, coinjection of 225Ac-DOTA-Nb with Gelofusine reduced kidney retention by 70%. This study shows that 225Ac-DOTA-Nb is a promising new radioconjugate for targeted α-particle therapy and supports its further development.
Collapse
Affiliation(s)
| | - Matthias D'Huyvetter
- In Vivo Cellular and Molecular Imaging Laboratory , Vrije Universiteit Brussel , Brussels , Belgium
| | - Frank Bruchertseifer
- European Commission, Joint Research Centre , Department for Nuclear Safety and Security , Karlsruhe , Germany
| | - Alfred Morgenstern
- European Commission, Joint Research Centre , Department for Nuclear Safety and Security , Karlsruhe , Germany
| | - Tony Lahoutte
- In Vivo Cellular and Molecular Imaging Laboratory , Vrije Universiteit Brussel , Brussels , Belgium.,Nuclear Medicine Department , UZ Brussel , Brussels , Belgium
| |
Collapse
|
54
|
Kouwenberg JJM, Kremers GJ, Slotman JA, Wolterbeek HT, Houtsmuller AB, Denkova AG, Bos AJJ. Alpha particle spectroscopy using FNTD and SIM super-resolution microscopy. J Microsc 2018; 270:326-334. [PMID: 29393521 DOI: 10.1111/jmi.12686] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/22/2017] [Accepted: 01/12/2018] [Indexed: 11/26/2022]
Abstract
Structured illumination microscopy (SIM) for the imaging of alpha particle tracks in fluorescent nuclear track detectors (FNTD) was evaluated and compared to confocal laser scanning microscopy (CLSM). FNTDs were irradiated with an external alpha source and imaged using both methodologies. SIM imaging resulted in improved resolution, without increase in scan time. Alpha particle energy estimation based on the track length, direction and intensity produced results in good agreement with the expected alpha particle energy distribution. A pronounced difference was seen in the spatial scattering of alpha particles in the detectors, where SIM showed an almost 50% reduction compared to CLSM. The improved resolution of SIM allows for more detailed studies of the tracks induced by ionising particles. The combination of SIM and FNTDs for alpha radiation paves the way for affordable and fast alpha spectroscopy and dosimetry.
Collapse
Affiliation(s)
- J J M Kouwenberg
- Radiation, Science & Technology, Technische Universiteit Delft Faculteit Technische Natuurwetenschappen, Mekelweg 15, Delft, the Netherlands
| | - G J Kremers
- Erasmus Optical Imaging Centre, Erasmus MC, 's-Gravendijkwal 230, Rotterdam, the Netherlands
| | - J A Slotman
- Erasmus Optical Imaging Centre, Erasmus MC, 's-Gravendijkwal 230, Rotterdam, the Netherlands
| | - H T Wolterbeek
- Radiation, Science & Technology, Technische Universiteit Delft Faculteit Technische Natuurwetenschappen, Mekelweg 15, Delft, the Netherlands
| | - A B Houtsmuller
- Erasmus Optical Imaging Centre, Erasmus MC, 's-Gravendijkwal 230, Rotterdam, the Netherlands
| | - A G Denkova
- Radiation, Science & Technology, Technische Universiteit Delft Faculteit Technische Natuurwetenschappen, Mekelweg 15, Delft, the Netherlands
| | - A J J Bos
- Radiation, Science & Technology, Technische Universiteit Delft Faculteit Technische Natuurwetenschappen, Mekelweg 15, Delft, the Netherlands
| |
Collapse
|
55
|
Kwon YD, Chung HJ, Lee SJ, Lee SH, Jeong BH, Kim HK. Synthesis of novel multivalent fluorescent inhibitors with high affinity to prostate cancer and their biological evaluation. Bioorg Med Chem Lett 2018; 28:572-576. [DOI: 10.1016/j.bmcl.2018.01.047] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 01/17/2018] [Accepted: 01/23/2018] [Indexed: 12/27/2022]
|
56
|
Nedrow JR, Josefsson A, Park S, Bäck T, Hobbs RF, Brayton C, Bruchertseifer F, Morgenstern A, Sgouros G. Pharmacokinetics, microscale distribution, and dosimetry of alpha-emitter-labeled anti-PD-L1 antibodies in an immune competent transgenic breast cancer model. EJNMMI Res 2017; 7:57. [PMID: 28721684 PMCID: PMC5515722 DOI: 10.1186/s13550-017-0303-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/05/2017] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Studies combining immune checkpoint inhibitors with external beam radiation have shown a therapeutic advantage over each modality alone. The purpose of these works is to evaluate the potential of targeted delivery of high LET radiation to the tumor microenvironment via an immune checkpoint inhibitor. METHODS The impact of protein concentration on the distribution of 111In-DTPA-anti-PD-L1-BC, an 111In-antibody conjugate targeted to PD-L1, was evaluated in an immunocompetent mouse model of breast cancer. 225Ac-DOTA-anti-PD-L1-BC was evaluated by both macroscale (ex vivo biodistribution) and microscale (alpha-camera images at a protein concentration determined by the 111In data. RESULTS The evaluation of 111In-DTPA-anti-PD-L1-BC at 1, 3, and 10 mg/kg highlighted the impact of protein concentration on the distribution of the labeled antibody, particularly in the blood, spleen, thymus, and tumor. Alpha-camera images for the microscale distribution of 225Ac-DOTA-anti-PD-L1-BC showed a uniform distribution in the liver while highly non-uniform distributions were obtained in the thymus, spleen, kidney, and tumor. At an antibody dose of 3 mg/kg, the liver was dose-limiting with an absorbed dose of 738 mGy/kBq; based upon blood activity concentration measurements, the marrow absorbed dose was 29 mGy/kBq. CONCLUSIONS These studies demonstrate that 225Ac-DOTA-anti-PD-L1-BC is capable of delivering high LET radiation to PD-L1 tumors. The use of a surrogate SPECT agent, 111In-DTPA-anti-PD-L1-BC, is beneficial in optimizing the dose delivered to the tumor sites. Furthermore, an accounting of the microscale distribution of the antibody in preclinical studies was essential to the proper interpretation of organ absorbed doses and their likely relation to biologic effect.
Collapse
Affiliation(s)
- Jessie R Nedrow
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, CRBII 4M.61, 1550 Orleans Street, Baltimore, MD, 21231, USA
| | - Anders Josefsson
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, CRBII 4M.61, 1550 Orleans Street, Baltimore, MD, 21231, USA
| | - Sunju Park
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, CRBII 4M.61, 1550 Orleans Street, Baltimore, MD, 21231, USA
| | - Tom Bäck
- The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Robert F Hobbs
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cory Brayton
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Frank Bruchertseifer
- European Commission Joint Research Centre, Directorate for Nuclear Safety and Security, Karlsruhe, Germany
| | - Alfred Morgenstern
- European Commission Joint Research Centre, Directorate for Nuclear Safety and Security, Karlsruhe, Germany
| | - George Sgouros
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, CRBII 4M.61, 1550 Orleans Street, Baltimore, MD, 21231, USA.
| |
Collapse
|
57
|
Xie F, Peng F. Anti-Prostate Cancer Activity of 8-Hydroxyquinoline-2-Carboxaldehyde–Thiosemicarbazide Copper Complexes by Fluorescent Microscopic Imaging. J Fluoresc 2017; 27:1937-1941. [PMID: 28653241 DOI: 10.1007/s10895-017-2133-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/15/2017] [Indexed: 10/19/2022]
|
58
|
Sumera, Anwar A, Ovais M, Khan A, Raza A. Docetaxel‐loaded solid lipid nanoparticles: a novel drug delivery system. IET Nanobiotechnol 2017; 11:621-629. [PMCID: PMC8676273 DOI: 10.1049/iet-nbt.2017.0001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 03/18/2017] [Accepted: 03/30/2017] [Indexed: 07/29/2023] Open
Abstract
Over the past few years, taxanes have emerged as a new class of anticancer drugs. Docetaxel (DTX) the prototype of this class has been approved for the treatment of broad range of cancers. However, to date the commercial preparation of DTX (Taxotere®) is accompanying adverse side effects, intolerance, and poor solubility, which can be overcome by encapsulating them using solid lipid nanoparticles (SLNs). SLNs represent versatile delivery system of drugs with newer forms such as polymer–solid lipid hybrid, surface modified and long circulating nanoparticles bringing forth improved prospects for cancer chemotherapy. In this review, the authors have discussed the current uses of various SLNs formulations of DTX with key emphasis on controlled and site‐specific drug delivery along with enhanced antitumour activity elucidated via in vitro and in vivo studies. Furthermore, the review article highlights few approaches that can be used in combination with existing DTX‐loaded SLNs to supplement DTX drug delivery.
Collapse
Affiliation(s)
- Sumera
- Department of PharmacyUniversity of SwabiSwabi23430Pakistan
| | - Amania Anwar
- Atta‐ur‐Rehman School of Applied BiosciencesNational University of Science and TechnologyIslamabad44000Pakistan
| | - Muhammad Ovais
- Department of BiotechnologyFaculty of Biological SciencesQuaid‐i‐Azam UniversityIslamabad45320Pakistan
| | - Abad Khan
- Department of PharmacyUniversity of SwabiSwabi23430Pakistan
| | - Abida Raza
- Nanotheragnostics LabNational Institute of Lasers and OptronicsPakistan Atomic Energy CommissionIslamabad44000Pakistan
| |
Collapse
|
59
|
Alpha-particle radiotherapy: For large solid tumors diffusion trumps targeting. Biomaterials 2017; 130:67-75. [DOI: 10.1016/j.biomaterials.2017.03.035] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 03/20/2017] [Accepted: 03/23/2017] [Indexed: 12/29/2022]
|
60
|
de Almeida CEB, Alves LN, Rocha HF, Cabral-Neto JB, Missailidis S. Aptamer delivery of siRNA, radiopharmaceutics and chemotherapy agents in cancer. Int J Pharm 2017; 525:334-342. [PMID: 28373101 DOI: 10.1016/j.ijpharm.2017.03.086] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 03/29/2017] [Accepted: 03/31/2017] [Indexed: 01/09/2023]
Abstract
Aptamers are oligonucleotide reagents with high affinity and specificity, which among other therapeutic and diagnostic applications have the capability of acting as delivery agents. Thus, aptamers are capable of carrying small molecules, nanoparticles, radiopharmaceuticals or fluorescent agents as well as nucleic acid therapeutics specifically to their target cells. In most cases, the molecules may possess interesting therapeutic properties, but their lack of specificity for a particular cell type, or ability to internalise in such a cell, hinders their clinical development, or cause unwanted side effects. Thus, chemotherapy or radiotherapy agents, famous for their side effects, can be coupled to aptamers for specific delivery. Equally, siRNA have great therapeutic potential and specificity, but one of their shortcomings remain the delivery and internalisation into cells. Various methodologies have been proposed to date, including aptamers, to resolve this problem. Therapeutic or imaging reagents benefit from the adaptability and ease of chemical manipulation of aptamers, their high affinity for the specific marker of a cell type, and their internalisation ability via cell mediated endocytosis. In this review paper, we explore the potential of the aptamers as delivery agents and offer an update on current status and latest advancements.
Collapse
Affiliation(s)
- Carlos E B de Almeida
- Laboratório de Radiobiologia, Divisão de Física Médica, Instituto de Radioproteção e Dosimetria, Comissão Nacional de Energia Nuclear, Av. Salvador Allende S/N., Rio de Janeiro, RJ, CEP 22783-127, Brazil
| | - Lais Nascimento Alves
- Laboratório de Radiobiologia, Divisão de Física Médica, Instituto de Radioproteção e Dosimetria, Comissão Nacional de Energia Nuclear, Av. Salvador Allende S/N., Rio de Janeiro, RJ, CEP 22783-127, Brazil
| | - Henrique F Rocha
- Laboratório de Anticorpos Monoclonais, Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), Fundação Oswaldo Cruz, Av. Brasil, 4365-Manguinhos, Rio de Janeiro, RJ, CEP 21040-900, Brazil
| | - Januário Bispo Cabral-Neto
- Laboratório de Radiobiologia, Divisão de Física Médica, Instituto de Radioproteção e Dosimetria, Comissão Nacional de Energia Nuclear, Av. Salvador Allende S/N., Rio de Janeiro, RJ, CEP 22783-127, Brazil; Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Brg. Trompowski-Cidade Universitária, Rio de Janeiro, RJ, CEP 21044-020, Brazil
| | - Sotiris Missailidis
- Laboratório de Anticorpos Monoclonais, Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), Fundação Oswaldo Cruz, Av. Brasil, 4365-Manguinhos, Rio de Janeiro, RJ, CEP 21040-900, Brazil.
| |
Collapse
|
61
|
Pant K, Sedláček O, Nadar RA, Hrubý M, Stephan H. Radiolabelled Polymeric Materials for Imaging and Treatment of Cancer: Quo Vadis? Adv Healthc Mater 2017; 6. [PMID: 28218487 DOI: 10.1002/adhm.201601115] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/24/2016] [Indexed: 12/15/2022]
Abstract
Owing to their tunable blood circulation time and suitable plasma stability, polymer-based nanomaterials hold a great potential for designing and utilising multifunctional nanocarriers for efficient imaging and effective treatment of cancer. When tagged with appropriate radionuclides, they may allow for specific detection (diagnosis) as well as the destruction of tumours (therapy) or even customization of materials, aiming to both diagnosis and therapy (theranostic approach). This review provides an overview of recent developments of radiolabelled polymeric nanomaterials (natural and synthetic polymers) for molecular imaging of cancer, specifically, applying nuclear techniques such as positron emission tomography (PET) and single-photon emission computed tomography (SPECT). Different approaches to radiolabel polymers are evaluated from the methodical radiochemical point of view. This includes new bifunctional chelating agents (BFCAs) for radiometals as well as novel labelling methods. Special emphasis is given to eligible strategies employed to evade the mononuclear phagocytic system (MPS) in view of efficient targeting. The discussion encompasses promising strategies currently employed as well as emerging possibilities in radionuclide-based cancer therapy. Key issues involved in the clinical translation of radiolabelled polymers and future scopes of this intriguing research field are also discussed.
Collapse
Affiliation(s)
- Kritee Pant
- Helmholtz-Zentrum Dresden-Rossendorf; Institute of Radiopharmaceutical Cancer Research; Bautzner Landstraße 400 01328 Dresden Germany
| | - Ondřej Sedláček
- Institute of Macromolecular Chemistry; The Academy of Sciences of the Czech Republic; Heyrovského námeˇstí 2 16206 Prague 6 Czech Republic
| | - Robin A. Nadar
- Helmholtz-Zentrum Dresden-Rossendorf; Institute of Radiopharmaceutical Cancer Research; Bautzner Landstraße 400 01328 Dresden Germany
| | - Martin Hrubý
- Institute of Macromolecular Chemistry; The Academy of Sciences of the Czech Republic; Heyrovského námeˇstí 2 16206 Prague 6 Czech Republic
| | - Holger Stephan
- Helmholtz-Zentrum Dresden-Rossendorf; Institute of Radiopharmaceutical Cancer Research; Bautzner Landstraße 400 01328 Dresden Germany
| |
Collapse
|
62
|
Nonnekens J, Chatalic KL, Molkenboer-Kuenen JD, Beerens CE, Bruchertseifer F, Morgenstern A, Veldhoven-Zweistra J, Schottelius M, Wester HJ, van Gent DC, van Weerden WM, Boerman OC, de Jong M, Heskamp S. 213Bi-Labeled Prostate-Specific Membrane Antigen-Targeting Agents Induce DNA Double-Strand Breaks in Prostate Cancer Xenografts. Cancer Biother Radiopharm 2017; 32:67-73. [DOI: 10.1089/cbr.2016.2155] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- Julie Nonnekens
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands
| | - Kristell L.S. Chatalic
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Urology, Erasmus MC, Rotterdam, The Netherlands
| | | | | | - Frank Bruchertseifer
- European Commission, Joint Research Centre, Directorate for Nuclear Safety and Security, Karlsruhe, Germany
| | - Alfred Morgenstern
- European Commission, Joint Research Centre, Directorate for Nuclear Safety and Security, Karlsruhe, Germany
| | | | - Margret Schottelius
- Pharmaceutical Radiochemistry, Technische Universität München, Garching, Germany
| | - Hans-Jürgen Wester
- Pharmaceutical Radiochemistry, Technische Universität München, Garching, Germany
| | - Dik C. van Gent
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands
| | | | - Otto C. Boerman
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marion de Jong
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Sandra Heskamp
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
63
|
Goins B, Bao A, Phillips WT. Techniques for Loading Technetium-99m and Rhenium-186/188 Radionuclides into Preformed Liposomes for Diagnostic Imaging and Radionuclide Therapy. Methods Mol Biol 2017; 1522:155-178. [PMID: 27837538 DOI: 10.1007/978-1-4939-6591-5_13] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Liposomes can serve as carriers of radionuclides for diagnostic imaging and therapeutic applications. Herein, procedures are outlined for radiolabeling liposomes with the gamma-emitting radionuclide, technetium-99m (99mTc), for noninvasive detection of disease and for monitoring the pharmacokinetics and biodistribution of liposomal drugs, and/or with therapeutic beta-emitting radionuclides, rhenium-186/188 (186/188Re), for radionuclide therapy. These efficient and practical liposome radiolabeling methods use a post-labeling mechanism to load 99mTc or 186/188Re into preformed liposomes prepared in advance of the labeling procedure. For all liposome radiolabeling methods described, a lipophilic chelator is used to transport 99mTc or 186/188Re across the lipid bilayer of the preformed liposomes. Once within the liposome interior, the pre-encapsulated glutathione or ammonium sulfate (pH) gradient provides for stable entrapment of the 99mTc and 186/188Re within the liposomes. In the first method, 99mTc is transported across the lipid bilayer by the lipophilic chelator, hexamethylpropyleneamine oxime (HMPAO) and 99mTc-HMPAO becomes trapped by interaction with the pre-encapsulated glutathione within the liposomes. In the second method, 99mTc or 186/188Re is transported across the lipid bilayer by the lipophilic chelator, N,N-bis(2-mercaptoethyl)-N',N'-diethylethylenediamine (BMEDA), and 99mTc-BMEDA or 186/188Re-BMEDA becomes trapped by interaction with pre-encapsulated glutathione within the liposomes. In the third method, an ammonium sulfate (pH) gradient loading technique is employed using liposomes with an extraliposomal pH of 7.4 and an interior pH of 5.1. BMEDA, which is lipophilic at pH 7.4, serves as a lipophilic chelator for 99mTc or 186/188Re to transport the radionuclides across the lipid bilayer. Once within the more acidic liposome interior, 99mTc/186/188Re-BMEDA complex becomes protonated and more hydrophilic, which results in stable entrapment of the 99mTc/186/188Re-BMEDA complex within the liposomes. Since many commercially available liposomal drugs use an ammonium sulfate (pH) gradient for drug loading, these liposomal drugs can be directly radiolabeled with 99mTc-BMEDA for noninvasive monitoring of tissue distribution during treatment or with 186/188Re-BMEDA for combination chemo-radionuclide therapy.
Collapse
Affiliation(s)
- Beth Goins
- Department of Radiology, MSC 7800, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Ande Bao
- Department of Radiation Oncology, School of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, 11100 Euclid Avenue, Cleveland, OH, 44106, USA
| | - William T Phillips
- Department of Radiology, MSC 7800, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA.
| |
Collapse
|
64
|
Evans JC, Malhotra M, Cryan JF, O'Driscoll CM. The therapeutic and diagnostic potential of the prostate specific membrane antigen/glutamate carboxypeptidase II (PSMA/GCPII) in cancer and neurological disease. Br J Pharmacol 2016; 173:3041-3079. [PMID: 27526115 PMCID: PMC5056232 DOI: 10.1111/bph.13576] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Revised: 07/08/2016] [Accepted: 07/25/2016] [Indexed: 12/11/2022] Open
Abstract
Prostate specific membrane antigen (PSMA) otherwise known as glutamate carboxypeptidase II (GCPII) is a membrane bound protein that is highly expressed in prostate cancer and in the neovasculature of a wide variety of tumours including glioblastomas, breast and bladder cancers. This protein is also involved in a variety of neurological diseases including schizophrenia and ALS. In recent years, there has been a surge in the development of both diagnostics and therapeutics that take advantage of the expression and activity of PSMA/GCPII. These include gene therapy, immunotherapy, chemotherapy and radiotherapy. In this review, we discuss the biological roles that PSMA/GCPII plays, both in normal and diseased tissues, and the current therapies exploiting its activity that are at the preclinical stage. We conclude by giving an expert opinion on the future direction of PSMA/GCPII based therapies and diagnostics and hurdles that need to be overcome to make them effective and viable.
Collapse
Affiliation(s)
- James C Evans
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - Meenakshi Malhotra
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | | |
Collapse
|
65
|
Mangala LS, Wang H, Jiang D, Wu SY, Somasunderam A, Volk DE, Lokesh GLR, Li X, Pradeep S, Yang X, Haemmerle M, Rodriguez-Aguayo C, Nagaraja AS, Rupaimoole R, Bayraktar E, Bayraktar R, Li L, Tanaka T, Hu W, Ivan C, Gharpure KM, McGuire MH, Thiviyanathan V, Zhang X, Maiti SN, Bulayeva N, Choi HJ, Dorniak PL, Cooper LJ, Rosenblatt KP, Lopez-Berestein G, Gorenstein DG, Sood AK. Improving vascular maturation using noncoding RNAs increases antitumor effect of chemotherapy. JCI Insight 2016; 1:e87754. [PMID: 27777972 PMCID: PMC5070952 DOI: 10.1172/jci.insight.87754] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 09/13/2016] [Indexed: 12/17/2022] Open
Abstract
Current antiangiogenesis therapy relies on inhibiting newly developed immature tumor blood vessels and starving tumor cells. This strategy has shown transient and modest efficacy. Here, we report a better approach to target cancer-associated endothelial cells (ECs), reverse permeability and leakiness of tumor blood vessels, and improve delivery of chemotherapeutic agents to the tumor. First, we identified deregulated microRNAs (miRs) from patient-derived cancer-associated ECs. Silencing these miRs led to decreased vascular permeability and increased maturation of blood vessels. Next, we screened a thioaptamer (TA) library to identify TAs selective for tumor-associated ECs. An annexin A2-targeted TA was identified and used for delivery of miR106b-5p and miR30c-5p inhibitors, resulting in vascular maturation and antitumor effects without inducing hypoxia. These findings could have implications for improving vascular-targeted therapy.
Collapse
Affiliation(s)
- Lingegowda S. Mangala
- Department of Gynecologic Oncology and Reproductive Medicine
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hongyu Wang
- Institute of Molecular Medicine
- Department of Nanomedicine and Biomedical Engineering, The University of Texas Health Science Center, Houston, Texas, USA
| | - Dahai Jiang
- Department of Gynecologic Oncology and Reproductive Medicine
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sherry Y. Wu
- Department of Gynecologic Oncology and Reproductive Medicine
| | - Anoma Somasunderam
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, USA
| | - David E. Volk
- Institute of Molecular Medicine
- Department of Nanomedicine and Biomedical Engineering, The University of Texas Health Science Center, Houston, Texas, USA
| | | | - Xin Li
- Institute of Molecular Medicine
| | - Sunila Pradeep
- Department of Gynecologic Oncology and Reproductive Medicine
| | | | | | - Cristian Rodriguez-Aguayo
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | - Emine Bayraktar
- Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Recep Bayraktar
- Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Li Li
- Institute of Molecular Medicine
| | - Takemi Tanaka
- Biomedical Research Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Wei Hu
- Department of Gynecologic Oncology and Reproductive Medicine
| | - Cristina Ivan
- Department of Gynecologic Oncology and Reproductive Medicine
| | | | | | - Varatharasa Thiviyanathan
- Institute of Molecular Medicine
- Department of Nanomedicine and Biomedical Engineering, The University of Texas Health Science Center, Houston, Texas, USA
| | - Xinna Zhang
- Department of Gynecologic Oncology and Reproductive Medicine
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sourindra N. Maiti
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Hyun-Jin Choi
- Department of Gynecologic Oncology and Reproductive Medicine
| | | | - Laurence J.N. Cooper
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Gabriel Lopez-Berestein
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - David G. Gorenstein
- Institute of Molecular Medicine
- Department of Nanomedicine and Biomedical Engineering, The University of Texas Health Science Center, Houston, Texas, USA
- AM Biotechnologies, Houston, Texas, USA
| | - Anil K. Sood
- Department of Gynecologic Oncology and Reproductive Medicine
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
66
|
Thakur PS, Khan AM, Talegaonkar S, Ahmad FJ, Iqbal Z. Hurdles in selection process of nanodelivery systems for multidrug-resistant cancer. J Cancer Res Clin Oncol 2016; 142:2073-106. [PMID: 27116692 DOI: 10.1007/s00432-016-2167-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 04/14/2016] [Indexed: 10/21/2022]
Abstract
PURPOSE Most of the nanomedicines for treatment of multidrug-resistant cancer do not reach Phase III trials and many are terminated or withdrawn or are in an indeterminate state since long without any study results being presented. Extensive perusal of nanomedicine development research revealed that one of the critical aspects influencing clinical outcomes and which requires diligent scrutiny is selection process of nanodelivery system. METHODS Research papers and articles published on development of nanodelivery systems for treatment of multidrug-resistant cancer were analyzed. Observations and conclusions noted by these researchers which might shed some light on poor clinical performance of nanocarriers were collated and summarized under observation section. Further research articles were studied to find possible solutions which may be applied to these particular problems for resolving them. The inferences of these findings were composed in Result section. RESULT Plausible solutions for the observed obstacles were noted as examples of novel formulations that can yield the following: better in vivo imaging, precise targeting and dosing of a specific site and specific cell type in a particular cancer, modulation of tumor surroundings, intonation of systemic effects and high reproducibility. CONCLUSION The angle of approach to the development of best nanosystem for a specific type of tumor needs to be spun around. Some of these changes can be brought about by individual scientists, some need to be established by collated efforts of scientists globally and some await advent of better technologies. Regardless of the stratagem, it can be said decisively that the schematics of development phase need rethinking.
Collapse
Affiliation(s)
- P S Thakur
- Department of Pharmaceutics, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India
| | - A M Khan
- Department of Pharmaceutics, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India
| | - S Talegaonkar
- Department of Pharmaceutics, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India
| | - F J Ahmad
- Department of Pharmaceutics, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India
| | - Z Iqbal
- Department of Pharmaceutics, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India.
| |
Collapse
|
67
|
Mokhtarzadeh A, Tabarzad M, Ranjbari J, de la Guardia M, Hejazi M, Ramezani M. Aptamers as smart ligands for nano-carriers targeting. Trends Analyt Chem 2016. [DOI: 10.1016/j.trac.2016.06.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
68
|
Sempkowski M, Zhu C, Menzenski MZ, Kevrekidis IG, Bruchertseifer F, Morgenstern A, Sofou S. Sticky Patches on Lipid Nanoparticles Enable the Selective Targeting and Killing of Untargetable Cancer Cells. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2016; 32:8329-8338. [PMID: 27468779 DOI: 10.1021/acs.langmuir.6b01464] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Effective targeting by uniformly functionalized nanoparticles is limited to cancer cells expressing at least two copies of targeted receptors per nanoparticle footprint (approximately ≥2 × 10(5) receptor copies per cell); such a receptor density supports the required multivalent interaction between the neighboring receptors and the ligands from a single nanoparticle. To enable selective targeting below this receptor density, ligands on the surface of lipid vesicles were displayed in clusters that were designed to form at the acidic pH of the tumor interstitium. Vesicles with clustered HER2-targeting peptides within such sticky patches (sticky vesicles) were compared to uniformly functionalized vesicles. On HER2-negative breast cancer cells MDA-MB-231 and MCF7 {expressing (8.3 ± 0.8) × 10(4) and (5.4 ± 0.9) × 10(4) HER2 copies per cell, respectively}, only the sticky vesicles exhibited detectable specific targeting (KD ≈ 49-69 nM); dissociation (0.005-0.009 min(-1)) and endocytosis rates (0.024-0.026 min(-1)) were independent of HER2 expression for these cells. MDA-MB-231 and MCF7 were killed only by sticky vesicles encapsulating doxorubicin (32-40% viability) or α-particle emitter (225)Ac (39-58% viability) and were not affected by uniformly functionalized vesicles (>80% viability). Toxicities on cardiomyocytes and normal breast cells (expressing HER2 at considerably lower but not insignificant levels) were not observed, suggesting the potential of tunable clustered ligand display for the selective killing of cancer cells with low receptor densities.
Collapse
Affiliation(s)
| | | | | | - Ioannis G Kevrekidis
- Department of Chemical and Biological Engineering, Program in Applied and Computational Mathematics, Princeton University , A319 Engineering Quad, Princeton, New Jersey 08544, United States
| | - Frank Bruchertseifer
- European Commission, Joint Research Centre, Institute for Transuranium Elements , P.O. Box 2340, D-76125 Karlsruhe, Germany
| | - Alfred Morgenstern
- European Commission, Joint Research Centre, Institute for Transuranium Elements , P.O. Box 2340, D-76125 Karlsruhe, Germany
| | | |
Collapse
|
69
|
Biodistribution of [68Ga]PSMA-HBED-CC in Patients with Prostate Cancer: Characterization of Uptake in Normal Organs and Tumour Lesions. Mol Imaging Biol 2016; 18:428-36. [DOI: 10.1007/s11307-016-0945-x] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
70
|
Li W, Liu Z, Li C, Li N, Fang L, Chang J, Tan J. Radionuclide therapy using ¹³¹I-labeled anti-epidermal growth factor receptor-targeted nanoparticles suppresses cancer cell growth caused by EGFR overexpression. J Cancer Res Clin Oncol 2016; 142:619-32. [PMID: 26573511 DOI: 10.1007/s00432-015-2067-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 10/23/2015] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Anti-epidermal growth factor receptor (EGFR)-targeted nanoparticles can be used to deliver a therapeutic and imaging agent to EGFR-overexpressing tumor cells. (131)I-labeled anti-EGFR nanoparticles derived from cetuximab were used as a tumor-targeting vehicle in radionuclide therapy. METHODS This paper describes the construction of the anti-EGFR nanoparticle EGFR-BSA-PCL. This nanoparticle was characterized for EGFR-targeted binding and cellular uptake in EGFR-overexpressing cancer cells by using flow cytometry and confocal microscopy. Anti-EGFR and non-targeted nanoparticles were labeled with (131)I using the chloramine-T method. Analyses of cytotoxicity and targeted cell killing with (131)I were performed using the MTT assay. The time-dependent cellular uptake of (131)I-labeled anti-EGFR nanoparticles proved the slow-release effects of nanoparticles. A radioiodine therapy study was also performed in mice. RESULTS The EGFR-targeted nanoparticle EGFR-BSA-PCL and the non-targeted nanoparticle BSA-PCL were constructed; the effective diameters were approximately 100 nm. The results from flow cytometry and confocal microscopy revealed significant uptake of EGFR-BSA-PCL in EGFR-overexpressing tumor cells. Compared with EGFR-BSA-PCL, BSA-PCL could also bind to cells, but tumor cell retention was minimal and weak. In MTT assays, the EGFR-targeted radioactive nanoparticle (131)I-EGFR-BSA-PCL showed greater cytotoxicity and targeted cell killing than the non-targeted nanoparticle (131)I-BSA-PCL. The radioiodine uptake of both (131)I-labeled nanoparticles, (131)I-EGFR-BSA-PCL and (131)I-BSA-PCL, was rapid and reached maximal levels 4 h after incubation, but the (131)I uptake of (131)I-EGFR-BSA-PCL was higher than that of (131)I-BSA-PCL. On day 15, the average tumor volumes of the (131)I-EGFR-BSA-PCL and (131)I-BSA-PCL groups showed a slow growth relationship compared with that of the control group. CONCLUSION The EGFR-targeted nanoparticle EGFR-BSA-PCL demonstrated superior cellular binding and uptake compared with those of the control BSA-PCL. The EGFR-targeted radioactive nanoparticle (131)I-EGFR-BSA-PCL exhibited favorable intracellular retention of (131)I. Radionuclide therapy using (131)I-EGFR-BSA-PCL, which showed excellent targeted cell killing, suppressed cancer cell growth caused by EGFR overexpression.
Collapse
Affiliation(s)
- Wei Li
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052, People's Republic of China
| | - Zhongyun Liu
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, 264003, Shandong, People's Republic of China
| | - Chengxia Li
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052, People's Republic of China
| | - Ning Li
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052, People's Republic of China
| | - Lei Fang
- Institute of Nanobiotechnology, School of Materials Science and Engineering, Tianjin Key Laboratory of Composites and Functional Materials, Tianjin University, Tianjin, 300072, People's Republic of China
| | - Jin Chang
- Institute of Nanobiotechnology, School of Materials Science and Engineering, Tianjin Key Laboratory of Composites and Functional Materials, Tianjin University, Tianjin, 300072, People's Republic of China
| | - Jian Tan
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052, People's Republic of China.
| |
Collapse
|
71
|
Dougherty CA, Cai W, Hong H. Applications of aptamers in targeted imaging: state of the art. Curr Top Med Chem 2016; 15:1138-52. [PMID: 25866268 DOI: 10.2174/1568026615666150413153400] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 02/13/2015] [Accepted: 02/14/2015] [Indexed: 01/23/2023]
Abstract
Aptamers are single-stranded oligonucleotides with high affinity and specificity to the target molecules or cells, thus they can serve as an important category of molecular targeting ligand. Since their discovery, aptamers have been rapidly translated into clinical practice. The strong target affinity/selectivity, cost-effectivity, chemical versatility and safety of aptamers are superior to traditional peptides- or proteins-based ligands which make them unique choices for molecular imaging. Therefore, aptamers are considered to be extremely useful to guide various imaging contrast agents to the target tissues or cells for optical, magnetic resonance, nuclear, computed tomography, ultrasound and multimodality imaging. This review aims to provide an overview of aptamers' advantages as targeting ligands and their application in targeted imaging. Further research in synthesis of new types of aptamers and their conjugation with new categories of contrast agents is required to develop clinically translatable aptamer-based imaging agents which will eventually result in improved patient care.
Collapse
Affiliation(s)
| | - Weibo Cai
- Department of Radiology, University of Wisconsin - Madison, Wisconsin 53705-2275, United States.
| | | |
Collapse
|
72
|
Wibmer AG, Burger IA, Sala E, Hricak H, Weber WA, Vargas HA. Molecular Imaging of Prostate Cancer. Radiographics 2015; 36:142-59. [PMID: 26587888 DOI: 10.1148/rg.2016150059] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Prostate cancer is the most common noncutaneous malignancy among men in the Western world. The natural history and clinical course of prostate cancer are markedly diverse, ranging from small indolent intraprostatic lesions to highly aggressive disseminated disease. An understanding of this biologic heterogeneity is considered a necessary requisite in the quest for the adoption of precise and personalized management strategies. Molecular imaging offers the potential for noninvasive assessment of the biologic interactions underpinning prostate carcinogenesis. Currently, numerous molecular imaging probes are in clinical use or undergoing preclinical or clinical evaluation. These probes can be divided into those that image increased cell metabolism, those that target prostate cancer-specific membrane proteins and receptor molecules, and those that bind to the bone matrix adjacent to metastases to bone. The increased metabolism and vascular changes in prostate cancer cells can be evaluated with radiolabeled analogs of choline, acetate, glucose, amino acids, and nucleotides. The androgen receptor, prostate-specific membrane antigen, and gastrin-releasing peptide receptor (ie, bombesin) are overexpressed in prostate cancer and can be targeted by specific radiolabeled imaging probes. Because metastatic prostate cancer cells induce osteoblastic signaling pathways of adjacent bone tissue, bone-seeking radiotracers are sensitive tools for the detection of metastases to bone. Knowledge about the underlying biologic processes responsible for the phenotypes associated with the different stages of prostate cancer allows an appropriate choice of methods and helps avoid pitfalls.
Collapse
Affiliation(s)
- Andreas G Wibmer
- From the Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065 (A.G.W., E.S., H.H., W.A.W., H.A.V.); and Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (I.A.B.)
| | - Irene A Burger
- From the Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065 (A.G.W., E.S., H.H., W.A.W., H.A.V.); and Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (I.A.B.)
| | - Evis Sala
- From the Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065 (A.G.W., E.S., H.H., W.A.W., H.A.V.); and Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (I.A.B.)
| | - Hedvig Hricak
- From the Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065 (A.G.W., E.S., H.H., W.A.W., H.A.V.); and Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (I.A.B.)
| | - Wolfgang A Weber
- From the Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065 (A.G.W., E.S., H.H., W.A.W., H.A.V.); and Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (I.A.B.)
| | - Hebert Alberto Vargas
- From the Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065 (A.G.W., E.S., H.H., W.A.W., H.A.V.); and Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (I.A.B.)
| |
Collapse
|
73
|
Zhu C, Bandekar A, Sempkowski M, Banerjee SR, Pomper MG, Bruchertseifer F, Morgenstern A, Sofou S. Nanoconjugation of PSMA-Targeting Ligands Enhances Perinuclear Localization and Improves Efficacy of Delivered Alpha-Particle Emitters against Tumor Endothelial Analogues. Mol Cancer Ther 2015; 15:106-113. [PMID: 26586724 DOI: 10.1158/1535-7163.mct-15-0207] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 10/16/2015] [Indexed: 11/16/2022]
Abstract
This study aims to evaluate the effect on killing efficacy of the intracellular trafficking patterns of α-particle emitters by using different radionuclide carriers in the setting of targeted antivascular α-radiotherapy. Nanocarriers (lipid vesicles) targeted to the prostate-specific membrane antigen (PSMA), which is unique to human neovasculature for a variety of solid tumors, were loaded with the α-particle generator actinium-225 and were compared with a PSMA-targeted radiolabeled antibody. Actinium-225 emits a total of four α-particles per decay, providing highly lethal and localized irradiation of targeted cells with minimal exposure to surrounding healthy tissues. Lipid vesicles were derivatized with two types of PSMA-targeting ligands: a fully human PSMA antibody (mAb) and a urea-based, low-molecular-weight agent. Target selectivity and extent of internalization were evaluated on monolayers of human endothelial cells (HUVEC) induced to express PSMA in static incubation conditions and in a flow field. Both types of radiolabeled PSMA-targeted vesicles exhibit similar killing efficacy, which is greater than the efficacy of the radiolabeled control mAb when compared on the basis of delivered radioactivity per cell. Fluorescence confocal microscopy demonstrates that targeted vesicles localize closer to the nucleus, unlike antibodies which localize near the plasma membrane. In addition, targeted vesicles cause larger numbers of dsDNAs per nucleus of treated cells compared with the radiolabeled mAb. These findings demonstrate that radionuclide carriers, such as PSMA-targeted lipid-nanocarriers, which localize close to the nucleus, increase the probability of α-particle trajectories crossing the nuclei, and, therefore, enhance the killing efficacy of α-particle emitters.
Collapse
Affiliation(s)
- Charles Zhu
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854
| | - Amey Bandekar
- Department of Chemical and Biochemical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854
| | - Michelle Sempkowski
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854
| | - Sangeeta Ray Banerjee
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical School, Baltimore, MD 21287
| | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical School, Baltimore, MD 21287
| | - Frank Bruchertseifer
- European Commission, Joint Research Centre, Institute for Transuranium Elements, P.O. Box 2340, D-76125 Karlsruhe, Germany
| | - Alfred Morgenstern
- European Commission, Joint Research Centre, Institute for Transuranium Elements, P.O. Box 2340, D-76125 Karlsruhe, Germany
| | - Stavroula Sofou
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854
- Department of Chemical and Biochemical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854
| |
Collapse
|
74
|
Gijs M, Aerts A, Impens N, Baatout S, Luxen A. Aptamers as radiopharmaceuticals for nuclear imaging and therapy. Nucl Med Biol 2015; 43:253-71. [PMID: 26746572 DOI: 10.1016/j.nucmedbio.2015.09.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 08/24/2015] [Accepted: 09/10/2015] [Indexed: 12/27/2022]
Abstract
Today, radiopharmaceuticals belong to the standard instrumentation of nuclear medicine, both in the context of diagnosis and therapy. The majority of radiopharmaceuticals consist of targeting biomolecules which are designed to interact with a disease-related molecular target. A plethora of targeting biomolecules of radiopharmaceuticals exists, including antibodies, antibody fragments, proteins, peptides and nucleic acids. Nucleic acids have some significant advantages relative to proteinaceous biomolecules in terms of size, production, modifications, possible targets and immunogenicity. In particular, aptamers (non-coding, synthetic, single-stranded DNA or RNA oligonucleotides) are of interest because they can bind a molecular target with high affinity and specificity. At present, few aptamers have been investigated preclinically for imaging and therapeutic applications. In this review, we describe the use of aptamers as targeting biomolecules of radiopharmaceuticals. We also discuss the chemical modifications which are needed to turn aptamers into valuable (radio-)pharmaceuticals, as well as the different radiolabeling strategies that can be used to radiolabel oligonucleotides and, in particular, aptamers.
Collapse
Affiliation(s)
- Marlies Gijs
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK·CEN), Mol, Belgium; Cyclotron Research Centre, University of Liège, Liège, Belgium
| | - An Aerts
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK·CEN), Mol, Belgium
| | - Nathalie Impens
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK·CEN), Mol, Belgium
| | - Sarah Baatout
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK·CEN), Mol, Belgium
| | - André Luxen
- Cyclotron Research Centre, University of Liège, Liège, Belgium.
| |
Collapse
|
75
|
Zechmann CM. Imaging for Prostate Cancer. CURRENT RADIOLOGY REPORTS 2015. [DOI: 10.1007/s40134-015-0107-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
76
|
Seeta Rama Raju G, Benton L, Pavitra E, Yu JS. Multifunctional nanoparticles: recent progress in cancer therapeutics. Chem Commun (Camb) 2015; 51:13248-59. [DOI: 10.1039/c5cc04643b] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In recent times, several biocompatible nanomaterials with different morphologies and compositions, such as metals, metal oxides, and polymers, have been employed as multi-functional biomaterials to target cancer cells.
Collapse
Affiliation(s)
- G. Seeta Rama Raju
- Department of Electronics and Radio Engineering
- Optoelectronics and Nanodevices Laboratory
- Kyung Hee University
- Yongin-si
- Republic of Korea
| | - Leah Benton
- Department of Biology
- Emory University
- Atlanta
- USA
| | - E. Pavitra
- Department of Electronics and Radio Engineering
- Optoelectronics and Nanodevices Laboratory
- Kyung Hee University
- Yongin-si
- Republic of Korea
| | - Jae Su Yu
- Department of Electronics and Radio Engineering
- Optoelectronics and Nanodevices Laboratory
- Kyung Hee University
- Yongin-si
- Republic of Korea
| |
Collapse
|
77
|
Tykvart J, Navrátil V, Sedlák F, Corey E, Colombatti M, Fracasso G, Koukolík F, Bařinka C, Sácha P, Konvalinka J. Comparative analysis of monoclonal antibodies against prostate-specific membrane antigen (PSMA). Prostate 2014; 74:1674-90. [PMID: 25262926 DOI: 10.1002/pros.22887] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 08/05/2014] [Indexed: 12/26/2022]
Abstract
BACKGROUND Prostate-specific membrane antigen (PSMA), also known as glutamate carboxypeptidase II (GCPII), is generally recognized as a diagnostic and therapeutic cancer antigen and a molecular address for targeted imaging and drug delivery studies. Due to its significance in cancer research, numerous monoclonal antibodies (mAbs) against GCPII have been described and marketed in the past decades. Unfortunately, some of these mAbs are poorly characterized, which might lead to their inappropriate use and misinterpretation of the acquired results. METHODS We collected the 13 most frequently used mAbs against GCPII and quantitatively characterized their binding to GCPII by enzyme-linked immunosorbent assay (ELISA) and surface plasmon resonance (SPR). Using a peptide library, we mapped epitopes recognized by a given mAb. Finally, we assessed the applicability of these mAbs to routine experimental setups, including Western blotting, immunohistochemistry, and flow cytometry. RESULTS ELISA and SPR analyses revealed that mAbs J591, J415, D2B, 107-1A4, GCP-05, and 2G7 bind preferentially to GCPII in native form, while mAbs YPSMA-1, YPSMA-2, GCP-02, GCP-04, and 3E6 bind solely to denatured GCPII. mAbs 24.4E6 and 7E11-C5.3 recognize both forms of GCPII. Additionally, we determined that GCP-02 and 3E6 cross-react with mouse GCPII, while GCP-04 recognizes GCPII and GCPIII proteins from both human and mouse. CONCLUSION This comparative analysis provides the first detailed quantitative characterization of the most commonly used mAbs against GCPII and can serve as a guideline for the scientific community to use them in a proper and efficient way.
Collapse
Affiliation(s)
- J Tykvart
- Gilead Sciences and IOCB Research Centre, Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Czech Republic; Department of Biochemistry, Faculty of Natural Science, Charles University, Czech Republic
| | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
|
79
|
Baek SE, Lee KH, Park YS, Oh DK, Oh S, Kim KS, Kim DE. RNA aptamer-conjugated liposome as an efficient anticancer drug delivery vehicle targeting cancer cells in vivo. J Control Release 2014; 196:234-42. [PMID: 25450401 DOI: 10.1016/j.jconrel.2014.10.018] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 10/08/2014] [Accepted: 10/16/2014] [Indexed: 02/06/2023]
Abstract
To minimize the systemic toxicity prevalent to chemotherapeutics, we designed a novel anticancer drug-encapsulating liposome conjugated with an RNA aptamer specific to the prostate specific membrane antigen (PSMA), which is expressed on the surface of prostate cancer cells. The RNA aptamer-conjugated liposome, termed an aptamosome, was prepared by the post-insertion method, in which RNA aptamer-conjugated micelles were inserted into a liposome. These nanosized (90-100 nm) aptamer-conjugated liposomes specifically bind to LNCaP prostate epithelial cells that express PSMA and thus cause the nanoparticles to have significantly enhanced in vitro cellular binding and uptake as compared with nontargeted nanoparticles that lack the PSMA aptamer. Aptamosomes encapsulated with the anticancer drug doxorubicin (Dox) were significantly more toxic to the targeted LNCaP cells than to nontargeted cancer cells. Dox-encapsulating aptamosomes administered to LNCaP xenograft nude mice were selectively retained in tumor tissue. We also demonstrated in vivo anticancer efficacy of the Dox-encapsulating PSMA-aptamosomes on tumor size regression in LNCaP xenograft mice. We suggest that the encapsulation of toxic chemicals with aptamer-conjugated liposomes will enable the use of these bioconjugates in clinical practice with fewer side effects.
Collapse
Affiliation(s)
- Si Eun Baek
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 143-701, Republic of Korea
| | - Kwang Hyun Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 143-701, Republic of Korea
| | - Yong Serk Park
- Department of Biomedical Laboratory Science, Yonsei University, Wonju 220-710, Republic of Korea
| | - Deok-Kun Oh
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 143-701, Republic of Korea
| | - Sangtaek Oh
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul 136-702, Republic of Korea
| | - Keun-Sik Kim
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 302-718, Republic of Korea.
| | - Dong-Eun Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 143-701, Republic of Korea.
| |
Collapse
|
80
|
Zhu J, Huang H, Dong S, Ge L, Zhang Y. Progress in aptamer-mediated drug delivery vehicles for cancer targeting and its implications in addressing chemotherapeutic challenges. Theranostics 2014; 4:931-44. [PMID: 25057317 PMCID: PMC4107293 DOI: 10.7150/thno.9663] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 06/23/2014] [Indexed: 12/28/2022] Open
Abstract
Aptamers are novel oligonucleotides with flexible three-dimensional configurations that recognize and bind to their cognate targets, including tumor surface receptors, in a high-affinity and highly specific manner. Because of their unique intrinsic properties, a variety of aptamer-mediated nanovehicles have been developed to directionally transport anti-cancer drugs to tumor sites to minimize systemic cytotoxicity and to enhance permeation by these tumoricidal agents. Despite advances in the selection and synthesis of aptamers and in the conjugation and self-assembly of nanotechnologies, current chemotherapy and drug delivery systems face great challenges. These challenges are due to the limitations of aptamers and vehicles and because of complicated tumor mechanisms, including heterogeneity, anti-cancer drug resistance, and hypoxia-induced aberrances. In this review, we will summarize current approaches utilizing tumor surface hallmarks and aptamers and their roles and mechanisms in therapeutic nanovehicles targeting tumors. Delivery forms include nanoparticles, nanotubes, nanogels, aptamer-drug conjugates, and novel molecular trains. Moreover, the obstacles posed by the aforementioned issues will be highlighted, and possible solutions will be acknowledged. Furthermore, future perspectives will be presented, including cutting-edge integration with RNA interference nanotechnology and personalized chemotherapy, which will facilitate innovative approaches to aptamer-based therapeutics.
Collapse
|