1
|
Mazzocco C, Genevois C, Li Q, Doudnikoff E, Dutheil N, Leste-Lasserre T, Arotcarena ML, Bezard E. In vivo bioluminescence imaging of the intracerebral fibroin-controlled AAV-α-synuclein diffusion for monitoring the central nervous system and peripheral expression. Sci Rep 2024; 14:9710. [PMID: 38678103 PMCID: PMC11055870 DOI: 10.1038/s41598-024-60613-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/25/2024] [Indexed: 04/29/2024] Open
Abstract
Among the several animal models of α-synucleinopathies, the well-known viral vector-mediated delivery of wild-type or mutated (A53T) α-synuclein requires new tools to increase the lesion in mice and follow up in vivo expression. To this end, we developed a bioluminescent expression reporter of the human A53T-α-synuclein gene using the NanoLuc system into an AAV2/9, embedded or not in a fibroin solution to stabilise its expression in space and time. We first verified the expression of the fused protein in vitro on transfected cells by bioluminescence and Western blotting. Next, two groups of C57Bl6Jr mice were unilaterally injected with the AAV-NanoLuc-human-A53T-α-synuclein above the substantia nigra combined (or not) with fibroin. We first show that the in vivo cerebral bioluminescence signal was more intense in the presence of fibroin. Using immunohistochemistry, we find that the human-A53T-α-synuclein protein is more restricted to the ipsilateral side with an overall greater magnitude of the lesion when fibroin was added. However, we also detected a bioluminescence signal in peripheral organs in both conditions, confirmed by the presence of viral DNA corresponding to the injected AAV in the liver using qPCR.
Collapse
Affiliation(s)
- Claire Mazzocco
- Institut des Maladies Neurodégénératives, UMR 5293, Univ. de Bordeaux, 33000, Bordeaux, France
- Institut des Maladies Neurodégénératives, UMR 5293, CNRS, 33000, Bordeaux, France
| | - Coralie Genevois
- VIVOPTIC-TBM-Core Univ Bordeaux, UAR 3427, 33000, Bordeaux, France
| | - Qin Li
- Motac Neuroscience, Manchester, M15 6WE, UK
| | - Evelyne Doudnikoff
- Institut des Maladies Neurodégénératives, UMR 5293, Univ. de Bordeaux, 33000, Bordeaux, France
- Institut des Maladies Neurodégénératives, UMR 5293, CNRS, 33000, Bordeaux, France
| | - Nathalie Dutheil
- Institut des Maladies Neurodégénératives, UMR 5293, Univ. de Bordeaux, 33000, Bordeaux, France
- Institut des Maladies Neurodégénératives, UMR 5293, CNRS, 33000, Bordeaux, France
| | | | - Marie-Laure Arotcarena
- Institut des Maladies Neurodégénératives, UMR 5293, Univ. de Bordeaux, 33000, Bordeaux, France
- Institut des Maladies Neurodégénératives, UMR 5293, CNRS, 33000, Bordeaux, France
| | - Erwan Bezard
- Institut des Maladies Neurodégénératives, UMR 5293, Univ. de Bordeaux, 33000, Bordeaux, France.
- Institut des Maladies Neurodégénératives, UMR 5293, CNRS, 33000, Bordeaux, France.
- Motac Neuroscience, Manchester, M15 6WE, UK.
| |
Collapse
|
2
|
Jiao J, Zhang J, Wen W, Qin W, Chen X. Prostate-specific membrane antigen-targeted surgery in prostate cancer: Accurate identification, real-time diagnosis, and precise resection. Theranostics 2024; 14:2736-2756. [PMID: 38773975 PMCID: PMC11103491 DOI: 10.7150/thno.95039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 04/11/2024] [Indexed: 05/24/2024] Open
Abstract
Radical prostatectomy (RP) combined with pelvic lymph node dissection (PLND) is the first step in multimodal treatment of prostate cancer (PCa) without distant metastases. For a long time, the surgical resection range has been highly dependent on the surgeon's visualization and experience with preoperative imaging. With the rapid development of prostate-specific membrane antigen positron emission tomography and single-photon emission computed tomography (PSMA-PET and PSMA-SPECT), PSMA-targeted surgery has been introduced for a more accurate pathological diagnosis and complete resection of positive surgical margins (PSMs) and micro-lymph node metastases (LNMs). We reviewed PSMA-targeted surgeries, including PSMA-PET-guided prostatic biopsy (PSMA-TB), PSMA-targeted radio-guided surgery (PSMA-RGS), PSMA-targeted fluorescence-guided surgery (PSMA-FGS), and multi-modality/multi-targeted PSMA-targeted surgery. We also discuss the strengths and challenges of PSMA-targeted surgery, and propose that PSMA-targeted surgery could be a great addition to existing surgery protocols, thereby improving the accuracy and convenience of surgery for primary and recurrent PCa in the near future.
Collapse
Affiliation(s)
- Jianhua Jiao
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
- Innovation Center for Tumor Immunocytology Therapy Technology, Xijing Innovation Research Institute, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jingjing Zhang
- Departments of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Theranostics Center of Excellenece, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
| | - Weihong Wen
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Weijun Qin
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
- Innovation Center for Tumor Immunocytology Therapy Technology, Xijing Innovation Research Institute, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, 138673, Singapore, Singapore
- Theranostics Center of Excellenece, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
| |
Collapse
|
3
|
Boutin J, Genevois C, Couillaud F, Lamrissi-Garcia I, Guyonnet-Duperat V, Bibeyran A, Lalanne M, Amintas S, Moranvillier I, Richard E, Blouin JM, Dabernat S, Moreau-Gaudry F, Bedel A. CRISPR editing to mimic porphyria combined with light: A new preclinical approach for prostate cancer. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200772. [PMID: 38596305 PMCID: PMC10899051 DOI: 10.1016/j.omton.2024.200772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/15/2024] [Accepted: 02/06/2024] [Indexed: 04/11/2024]
Abstract
Thanks to its very high genome-editing efficiency, CRISPR-Cas9 technology could be a promising anticancer weapon. Clinical trials using CRISPR-Cas9 nuclease to ex vivo edit and alter immune cells are ongoing. However, to date, this strategy still has not been applied in clinical practice to directly target cancer cells. Targeting a canonical metabolic pathway essential to good functioning of cells without potential escape would represent an attractive strategy. We propose to mimic a genetic metabolic disorder in cancer cells to weaken cancer cells, independent of their genomic abnormalities. Mutations affecting the heme biosynthesis pathway are responsible for porphyria, and most of them are characterized by an accumulation of toxic photoreactive porphyrins. This study aimed to mimic porphyria by using CRISPR-Cas9 to inactivate UROS, leading to porphyrin accumulation in a prostate cancer model. Prostate cancer is the leading cancer in men and has a high mortality rate despite therapeutic progress, with a primary tumor accessible to light. By combining light with gene therapy, we obtained high efficiency in vitro and in vivo, with considerable improvement in the survival of mice. Finally, we achieved the preclinical proof-of-principle of performing cancer CRISPR gene therapy.
Collapse
Affiliation(s)
- Julian Boutin
- University of Bordeaux, INSERM, UMR 1312, Bordeaux Institute of Oncology, 146 Rue Léo Saignat, 33076 Bordeaux, France
- CHU de Bordeaux, Biochemistry Laboratory, 33000 Bordeaux, France
| | - Coralie Genevois
- University of Bordeaux, INSERM, UMR 1312, Bordeaux Institute of Oncology, 146 Rue Léo Saignat, 33076 Bordeaux, France
- Vivoptic Platform INSERM US 005—CNRS UAR 3427-TBM-Core, Bordeaux University, 33000 Bordeaux, France
| | - Franck Couillaud
- University of Bordeaux, INSERM, UMR 1312, Bordeaux Institute of Oncology, 146 Rue Léo Saignat, 33076 Bordeaux, France
- Vivoptic Platform INSERM US 005—CNRS UAR 3427-TBM-Core, Bordeaux University, 33000 Bordeaux, France
| | - Isabelle Lamrissi-Garcia
- University of Bordeaux, INSERM, UMR 1312, Bordeaux Institute of Oncology, 146 Rue Léo Saignat, 33076 Bordeaux, France
| | - Veronique Guyonnet-Duperat
- University of Bordeaux, INSERM, UMR 1312, Bordeaux Institute of Oncology, 146 Rue Léo Saignat, 33076 Bordeaux, France
- Vect’UB, Vectorology Platform, INSERM US 005—CNRS UAR 3427-TBM-Core, Bordeaux University, 33000 Bordeaux, France
| | - Alice Bibeyran
- University of Bordeaux, INSERM, UMR 1312, Bordeaux Institute of Oncology, 146 Rue Léo Saignat, 33076 Bordeaux, France
- Vect’UB, Vectorology Platform, INSERM US 005—CNRS UAR 3427-TBM-Core, Bordeaux University, 33000 Bordeaux, France
| | - Magalie Lalanne
- University of Bordeaux, INSERM, UMR 1312, Bordeaux Institute of Oncology, 146 Rue Léo Saignat, 33076 Bordeaux, France
| | - Samuel Amintas
- University of Bordeaux, INSERM, UMR 1312, Bordeaux Institute of Oncology, 146 Rue Léo Saignat, 33076 Bordeaux, France
- CHU de Bordeaux, Tumor Biology and Tumor Bank Laboratory, 33000 Bordeaux, France
| | - Isabelle Moranvillier
- University of Bordeaux, INSERM, UMR 1312, Bordeaux Institute of Oncology, 146 Rue Léo Saignat, 33076 Bordeaux, France
| | - Emmanuel Richard
- University of Bordeaux, INSERM, UMR 1312, Bordeaux Institute of Oncology, 146 Rue Léo Saignat, 33076 Bordeaux, France
- CHU de Bordeaux, Biochemistry Laboratory, 33000 Bordeaux, France
| | - Jean-Marc Blouin
- University of Bordeaux, INSERM, UMR 1312, Bordeaux Institute of Oncology, 146 Rue Léo Saignat, 33076 Bordeaux, France
- CHU de Bordeaux, Biochemistry Laboratory, 33000 Bordeaux, France
| | - Sandrine Dabernat
- University of Bordeaux, INSERM, UMR 1312, Bordeaux Institute of Oncology, 146 Rue Léo Saignat, 33076 Bordeaux, France
- CHU de Bordeaux, Biochemistry Laboratory, 33000 Bordeaux, France
| | - François Moreau-Gaudry
- University of Bordeaux, INSERM, UMR 1312, Bordeaux Institute of Oncology, 146 Rue Léo Saignat, 33076 Bordeaux, France
- CHU de Bordeaux, Biochemistry Laboratory, 33000 Bordeaux, France
| | - Aurélie Bedel
- University of Bordeaux, INSERM, UMR 1312, Bordeaux Institute of Oncology, 146 Rue Léo Saignat, 33076 Bordeaux, France
- CHU de Bordeaux, Biochemistry Laboratory, 33000 Bordeaux, France
| |
Collapse
|
4
|
Abusalem M, Martiniova L, Soebianto S, DePalatis L, Ravizzini G. Current Status of Radiolabeled Monoclonal Antibodies Targeting PSMA for Imaging and Therapy. Cancers (Basel) 2023; 15:4537. [PMID: 37760506 PMCID: PMC10526399 DOI: 10.3390/cancers15184537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/21/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Prostate cancer (PCa) is one of the most prevalent cancer diagnoses among men in the United States and in several other developed countries. The prostate specific membrane antigen (PSMA) has been recognized as a promising molecular target in PCa, which has led to the development of specific radionuclide-based tracers for imaging and radiopharmaceuticals for PSMA targeted therapy. These compounds range from small molecule ligands to monoclonal antibodies (mAbs). Monoclonal antibodies play a crucial role in targeting cancer cell-specific antigens with a high degree of specificity while minimizing side effects to normal cells. The same mAb can often be labeled in different ways, such as with radionuclides suitable for imaging with Positron Emission Tomography (β+ positrons), Gamma Camera Scintigraphy (γ photons), or radiotherapy (β- electrons, α-emitters, or Auger electrons). Accordingly, the use of radionuclide-based PSMA-targeting compounds in molecular imaging and therapeutic applications has significantly grown in recent years. In this article, we will highlight the latest developments and prospects of radiolabeled mAbs that target PSMA for the detection and treatment of prostate cancer.
Collapse
Affiliation(s)
- Mohammed Abusalem
- Department of Nuclear Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lucia Martiniova
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sarita Soebianto
- Department of Nuclear Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Louis DePalatis
- BioDevelopment Solutions, LLC, 226 Becker Circle, Johnstown, CO 80534, USA
| | - Gregory Ravizzini
- Department of Nuclear Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
5
|
Muñoz-López P, Ribas-Aparicio RM, Becerra-Báez EI, Fraga-Pérez K, Flores-Martínez LF, Mateos-Chávez AA, Luria-Pérez R. Single-Chain Fragment Variable: Recent Progress in Cancer Diagnosis and Therapy. Cancers (Basel) 2022; 14:cancers14174206. [PMID: 36077739 PMCID: PMC9455005 DOI: 10.3390/cancers14174206] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/25/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Recombinant antibody fragments have shown remarkable potential as diagnostic and therapeutic tools in the fight against cancer. The single-chain fragment variable (scFv) that contains the complete antigen-binding domains of a whole antibody, has several advantages such as a high specificity and affinity for antigens, a low immunogenicity, and the proven ability to penetrate tumor tissues and diffuse. This review provides an overview of the current studies on the principle, generation, and applications of scFvs, particularly in the diagnosis and therapy of cancer, and underscores their potential use in clinical trials. Abstract Cancer remains a public health problem worldwide. Although conventional therapies have led to some excellent outcomes, some patients fail to respond to treatment, they have few therapeutic alternatives and a poor survival prognosis. Several strategies have been proposed to overcome this issue. The most recent approach is immunotherapy, particularly the use of recombinant antibodies and their derivatives, such as the single-chain fragment variable (scFv) containing the complete antigen-binding domains of a whole antibody that successfully targets tumor cells. This review describes the recent progress made with scFvs as a cancer diagnostic and therapeutic tool, with an emphasis on preclinical approaches and their potential use in clinical trials.
Collapse
Affiliation(s)
- Paola Muñoz-López
- Unit of Investigative Research on Hemato-Oncological Diseases, Hospital Infantil de México Federico Gómez, Doctor Márquez 162, Mexico City 06720, Mexico
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Prolongación de Carpio y Plan de Ayala S/N, Mexico City 11340, Mexico
| | - Rosa María Ribas-Aparicio
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Prolongación de Carpio y Plan de Ayala S/N, Mexico City 11340, Mexico
| | - Elayne Irene Becerra-Báez
- Unit of Investigative Research on Hemato-Oncological Diseases, Hospital Infantil de México Federico Gómez, Doctor Márquez 162, Mexico City 06720, Mexico
- Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Prolongación de Carpio y Plan de Ayala S/N, Mexico City 11340, Mexico
| | - Karla Fraga-Pérez
- Unit of Investigative Research on Hemato-Oncological Diseases, Hospital Infantil de México Federico Gómez, Doctor Márquez 162, Mexico City 06720, Mexico
| | - Luis Fernando Flores-Martínez
- Unit of Investigative Research on Hemato-Oncological Diseases, Hospital Infantil de México Federico Gómez, Doctor Márquez 162, Mexico City 06720, Mexico
| | - Armando Alfredo Mateos-Chávez
- Unit of Investigative Research on Hemato-Oncological Diseases, Hospital Infantil de México Federico Gómez, Doctor Márquez 162, Mexico City 06720, Mexico
| | - Rosendo Luria-Pérez
- Unit of Investigative Research on Hemato-Oncological Diseases, Hospital Infantil de México Federico Gómez, Doctor Márquez 162, Mexico City 06720, Mexico
- Correspondence: ; Tel.: +52-(55)-5228-9917 (ext. 4401)
| |
Collapse
|
6
|
Wheeler TT, Cao P, Ghouri MD, Ji T, Nie G, Zhao Y. Nanotechnological strategies for prostate cancer imaging and diagnosis. Sci China Chem 2022. [DOI: 10.1007/s11426-022-1271-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
7
|
Near-Infrared Fluorescence Imaging of EGFR-Overexpressing Tumors in the Mouse Xenograft Model Using scFv-IRDye800CW and Cetuximab-IRDye800CW. Mol Imaging 2022; 2022:9589820. [PMID: 35517713 PMCID: PMC9042373 DOI: 10.1155/2022/9589820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 03/21/2022] [Indexed: 12/18/2022] Open
Abstract
EGFR (epidermal growth factor receptor) is overexpressed in a variety of human cancers (including squamous cell carcinoma of head and neck, colon cancer, and some breast cancers) and therefore is regarded as an ideal target for cancer therapy or imaging purposes. In the current study, we produced a scFv-based near-infrared probe (called cet.Hum.scFv-IRDye-800CW) and evaluated its ability in recognizing and imaging of EGFR-overexpressing tumors in a mouse model. Like the molecular probe consisting of its parental antibody (cetuximab, an FDA-approved monoclonal antibody) and IRD800CW, cet.Hum.scFv-IRDye-800CW was able to recognize EGFR-overexpressing tumors in mice. cet.Hum.scFv-IRDye-800CW was found to be superior to the cetuximab-based probe in imaging of mouse tumors. The tumor-to-background ratio and blood clearance rate were higher when cet.Hum.scFv-IRDye-800CW was used as an imaging probe.
Collapse
|
8
|
Buckle T, van Willigen DM, Welling MM, van Leeuwen FW. Pre-clinical development of fluorescent tracers and translation towards clinical application. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00045-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
9
|
Zhao J, Xu J, Yang T, Yu X, Cheng C, Zhang T, Ren Z, Li N, Yang F, Li G. Expression, purification and characterisation of a human anti-CDK4 single-chain variable fragment antibody. BMC Biotechnol 2021; 21:71. [PMID: 34930213 PMCID: PMC8690526 DOI: 10.1186/s12896-021-00729-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 12/08/2021] [Indexed: 12/05/2022] Open
Abstract
Background Cyclin-dependent kinase 4 (CDK4) when hyperactivated drives development and maintenance of most tumour types, thus prompting its use as an essential cancer treatment target and a diagnostic tool. Target-binding molecules, such as single-chain variable fragment (scFv) antibodies, hold tremendous potential for use in a wide range of cancer diagnostic and therapeutic applications. Results A human anti-CDK4 scFv antibody (AK2) derived from a human phage display library was expressed in soluble form in Escherichia coli and shown to be secreted into the culture supernatant. Next, soluble AK2 within culture supernatant was successfully purified using affinity chromatography then was shown, using enzyme-linked immunosorbent assays, to bind to recombinant human CDK4 with high affinity and specificity. Further analyses of AK2 interactions with intracellular components demonstrated that AK2 recognised and interacted specifically with endogenous CDK4 and thus could be useful for detection of CDK4 within tumour cells. Conclusions A novel anti-CDK4 scFv antibody that can recognise and interact specifically with recombinant human CDK4 and endogenous CDK4 in tumour cells was expressed and purified successfully. These results suggest that the anti-CDK4 scFv antibody may serve as a new and promising tool for achieving CDK4-targeted diagnosis, prognosis and treatment of numerous types of cancers. Supplementary Information The online version contains supplementary material available at 10.1186/s12896-021-00729-z.
Collapse
Affiliation(s)
- Jialiang Zhao
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Jingjing Xu
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Tianbin Yang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Xinze Yu
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Cheng Cheng
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Tong Zhang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Ze Ren
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Na Li
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Fang Yang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China.
| | - Guiying Li
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China.
| |
Collapse
|
10
|
Validity of Anti-PSMA ScFvD2B as a Theranostic Tool: A Narrative-Focused Review. Biomedicines 2021; 9:biomedicines9121870. [PMID: 34944686 PMCID: PMC8698710 DOI: 10.3390/biomedicines9121870] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/03/2021] [Accepted: 12/07/2021] [Indexed: 11/17/2022] Open
Abstract
Prostate cancer (PCa) is the second leading cause of cancer among men, and its diagnosis and adequate staging are fundamental. Among the biomarkers identified in recent years for PCa management, prostate-specific-membrane-antigen (PSMA), physiologically expressed at a low level on healthy prostate and in other normal tissues and highly overexpressed in PCa, represents a reliable marker ideal for imaging and therapy. The development of anti-PSMA antibodies, such as D2B, demonstrated slow clearance of intact antibodies compared with fragments resulting in low tumor-to-blood ratios; however, the modular structural and functional nature of antibodies allowed the generation of smaller fragments, such as scFvs. In this review of the anti-PSMA antibody fragment scFvD2B, we combined further characterization of its biomolecular and tissue cross-reactivity characteristics with a comprehensive summary of what has already been performed in preclinical models to evaluate imaging and therapeutic activities. A molecular dynamics study was performed, and ScFvD2B occupied a limited conformational space, characterized by low-energy conformational basins, confirming the high stability of the protein structure. In the cross-reactivity study, the weak/absent immunoreactivity in non-tumor tissues was comparable to the PSMA expression reported in the literature. Biodistribution studies and therapeutic treatments were conducted in different animal models obtained by subcutaneous or locoregional injection of PSMA-positive-versus-negative xenografts. The maximum tumor uptake was observed for 123I(SPECT), 124I(PET), and optical imaging, which avoids kidney accumulation (compared with radiometals) and leads to an optimal tumor-to-kidney and tumor-to-background ratios. Regarding its possible use in therapy, experimental data suggested a strong and specific antitumor activity, in vitro and in vivo, obtained using CAR-T or NK-92/CAR cells expressing scFvD2B. Based on presented/reviewed data, we consider that scFvD2B, due to its versatility and robustness, seems to: (i) overcome some problems observed in other studied scFvs, very often relatively unstable and prone to form aggregates; (ii) have sufficient tumor-to-background ratios for targeting and imaging PSMA-expressing cancer; (iii) significantly redirect immune killing cells to PSMA-positive tumors when inserted in second-generation CAR-T or NK-92/CAR cells. These data suggest that our product can be considered the right reagent to fill the gap that still exists in PCa diagnosis and treatment.
Collapse
|
11
|
Meléndez-Alafort L, Ferro-Flores G, Santos-Cuevas C, Ocampo-García B, Turato S, Fracasso G, Bolzati C, Rosato A, De Nardo L. Preclinical dosimetric studies of 177 Lu-scFvD2B and comparison with 177 Lu-PSMA-617 and 177 Lu-iPSMA endoradiotherapeutic agents. Med Phys 2021; 48:4064-4074. [PMID: 33966284 DOI: 10.1002/mp.14936] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 04/19/2021] [Accepted: 04/30/2021] [Indexed: 11/08/2022] Open
Abstract
PURPOSE Internal dosimetry has become a very important tool to evaluate the risks and benefits of new endoradiotherapeutic agents. Nowadays, some of the most successful targeted radionuclide therapy (TRT) agents are 177 Lu-DOTA conjugates based on low molecular weight (LMW) Glu-ureido PSMA inhibitors. It has, however, been demonstrated that the DOTA chelating moiety reduces the internalization of the LMW-PSMA agent and its radiation dose to the tumor. Previously, we reported that 177 Lu-scFvD2B, an antibody-based construct, demonstrated statistically significant higher cell uptake and internalization in LNCaP prostate cancer (PCa) cells (PSMA-positive) when compared to the LMW-PSMA agents, 177 Lu-PSMA-617 and 177 Lu-iPSMA, two of the endoradiotherapeutic agents which currently are the most used in PCa therapy. The aim of this study is to estimate the preclinical 177 Lu-scFvD2B organ and tumor-absorbed doses, and to compare the values with those of 177 Lu-PSMA-617 and 177 Lu-iPSMA. METHODS 177 Lu-scFvD2B, 177 Lu-PSMA-617, and 177 Lu-iPSMA were prepared and their radiochemical purity determined. Biodistribution studies of each radiopharmaceutical were then carried out in healthy mice to define the main source organs (SO) and to calculate the number of disintegrations in each source organs per unit of administered activity (NSO ). Absorbed dose in the main organs were then calculated for each 177 Lu-conjugate by means of OLINDA/EXM 2.1.1 software, using the calculated NSO for both the adult male and the mouse phantoms as program inputs. Images of mice bearing micropulmonary tumors injected with 177 Lu-conjugates were also obtained. Tumor standardized uptake values (SUV) for the different conjugates, obtained from the 3D SPECT image reconstruction of these mice, were used as the number of disintegrations in a tumor site per unit of administered activity (NT ). The tumor-absorbed dose was calculated using the published electron dose S-values for sphere models with diameters ranging from 10 µm to 10 mm and considering a uniform activity distribution and tumor density equivalent to water density. RESULTS All 177 Lu-labeled agents were obtained in high yield (98%). Dosimetric studies carried out using mouse phantoms demonstrated that organ absorbed doses of 177 Lu-scFvD2B were from 1.4 to 2.3 times higher than those for 177 Lu-iPSMA and from 1.5 to 2.6 times higher than those for 177 Lu-PSMA-617. However, the 177 Lu-scFvD2B values of tumor-absorbed doses for all investigated tumor sizes were from 2.8 to 3.0 times greater than those calculated for 177 Lu-iPSMA and 177 Lu-PSMA-617, respectively. Moreover, 177 Lu-scFvD2B showed the highest tumor/kidney ratio when compared to those reported for 177 Lu-albumin conjugates. CONCLUSIONS In this preclinical study, we demonstrated the potential of 177 Lu-scFvD2B as a therapeutic agent for PSMA-expressing tumors, due to its higher tumor-absorbed dose when compared with 177 Lu-LMW agents.
Collapse
Affiliation(s)
| | - Guillermina Ferro-Flores
- Laboratorio Nacional de Investigación y Desarrollo de Radiofármacos-CONACyT, Instituto Nacional de Investigaciones Nucleares, Carretera México-Toluca S/N, La Marquesa, Ocoyoacac,, 52750, Estado de México, México
| | - Clara Santos-Cuevas
- Laboratorio Nacional de Investigación y Desarrollo de Radiofármacos-CONACyT, Instituto Nacional de Investigaciones Nucleares, Carretera México-Toluca S/N, La Marquesa, Ocoyoacac,, 52750, Estado de México, México
| | - Blanca Ocampo-García
- Laboratorio Nacional de Investigación y Desarrollo de Radiofármacos-CONACyT, Instituto Nacional de Investigaciones Nucleares, Carretera México-Toluca S/N, La Marquesa, Ocoyoacac,, 52750, Estado de México, México
| | - Sofia Turato
- Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35138, Padua, Italy
| | - Giulio Fracasso
- Department of Medicine, University of Verona, Piaz. Le L.A. Scuro 10, 37134, Verona, Italy
| | - Cristina Bolzati
- Institute of Condensed Matter Chemistry and Energy Technologies, ICMATE-CNR, Corso Stati Uniti 4, 35127, Padua, Italy
| | - Antonio Rosato
- Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35138, Padua, Italy.,Department of Surgery, Oncology and Gastroenterology, University of Padua, Via Gattamelata 64, 35138, Padua, Italy
| | - Laura De Nardo
- Department of Physics and Astronomy, University of Padua, Via Marzolo 8, 35131, Padua, Italy.,INFN, Sezione di Padova, Via Marzolo 8, 35131, Padua, Italy
| |
Collapse
|
12
|
Unique Benefits of Tumor-Specific Nanobodies for Fluorescence Guided Surgery. Biomolecules 2021; 11:biom11020311. [PMID: 33670740 PMCID: PMC7921980 DOI: 10.3390/biom11020311] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/11/2021] [Accepted: 02/14/2021] [Indexed: 12/11/2022] Open
Abstract
Tumor-specific fluorescence labeling is promising for real-time visualization of solid malignancies during surgery. There are a number of technologies to confer tumor-specific fluorescence. Antibodies have traditionally been used due to their versatility in modifications; however, their large size hampers efficient fluorophore delivery. Nanobodies are a novel class of molecules, derived from camelid heavy-chain only antibodies, that have shown promise for tumor-specific fluorescence labeling. Nanobodies are ten times smaller than standard antibodies, while maintaining antigen-binding capacity and have advantageous features, including rapidity of tumor labeling, that are reviewed in the present report. The present report reviews special considerations needed in developing nanobody probes, the status of current literature on the use of nanobody probes in fluorescence guided surgery, and potential challenges to be addressed for clinical translation.
Collapse
|
13
|
Engineered Fragments of the PSMA-Specific 5D3 Antibody and Their Functional Characterization. Int J Mol Sci 2020; 21:ijms21186672. [PMID: 32932591 PMCID: PMC7555429 DOI: 10.3390/ijms21186672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 11/29/2022] Open
Abstract
Prostate-Specific Membrane Antigen (PSMA) is an established biomarker for the imaging and experimental therapy of prostate cancer (PCa), as it is strongly upregulated in high-grade primary, androgen-independent, and metastatic lesions. Here, we report on the development and functional characterization of recombinant single-chain Fv (scFv) and Fab fragments derived from the 5D3 PSMA-specific monoclonal antibody (mAb). These fragments were engineered, heterologously expressed in insect S2 cells, and purified to homogeneity with yields up to 20 mg/L. In vitro assays including ELISA, immunofluorescence and flow cytometry, revealed that the fragments retain the nanomolar affinity and single target specificity of the parent 5D3 antibody. Importantly, using a murine xenograft model of PCa, we verified the suitability of fluorescently labeled fragments for in vivo imaging of PSMA-positive tumors and compared their pharmacokinetics and tissue distribution to the parent mAb. Collectively, our data provide an experimental basis for the further development of 5D3 recombinant fragments for future clinical use.
Collapse
|
14
|
Sheetz T, Mills J, Tessari A, Pawlikowski M, Braddom AE, Posid T, Zynger DL, James C, Embrione V, Parbhoo K, Foray C, Coppola V, Croce CM, Palmieri D. NCL Inhibition Exerts Antineoplastic Effects against Prostate Cancer Cells by Modulating Oncogenic MicroRNAs. Cancers (Basel) 2020; 12:E1861. [PMID: 32664322 PMCID: PMC7408652 DOI: 10.3390/cancers12071861] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/05/2020] [Accepted: 07/06/2020] [Indexed: 11/16/2022] Open
Abstract
Prostate cancer (PCa) is the most frequently diagnosed cancer in men and second most common cause of cancer-related deaths in the United States. Androgen deprivation therapy (ADT) is only temporarily effective for advanced-stage PCa, as the disease inevitably progresses to castration-resistant prostate cancer (CRPC). The protein nucleolin (NCL) is overexpressed in several types of human tumors where it is also mislocalized to the cell surface. We previously reported the identification of a single-chain fragment variable (scFv) immuno-agent that is able to bind NCL on the surface of breast cancer cells and inhibit proliferation both in vitro and in vivo. In the present study, we evaluated whether NCL could be a valid therapeutic target for PCa, utilizing DU145, PC3 (CRPC), and LNCaP (androgen-sensitive) cell lines. First, we interrogated the publicly available databases and noted that higher NCL mRNA levels are associated with higher Gleason Scores as well as with recurrent and metastatic tumors. Then, using our anti-NCL scFv, we demonstrated that NCL is expressed on the surface of all three tested cell lines and that NCL inhibition results in reduced proliferation and migration. We also measured the inhibitory effect of NCL targeting on the biogenesis of oncogenic microRNAs such as miR-21, -221 and -222, which was cell context dependent. Taken together, our data provide evidence that NCL targeting inhibits the key hallmarks of malignancy in PCa cells and may provide a novel therapeutic option for patients with advanced-stage PCa.
Collapse
Affiliation(s)
- Tyler Sheetz
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (T.S.); (J.M.); (A.T.); (M.P.); (A.E.B.); (V.E.); (K.P.); (C.F.); (V.C.); (C.M.C.)
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- Department of Urology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA;
| | - Joseph Mills
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (T.S.); (J.M.); (A.T.); (M.P.); (A.E.B.); (V.E.); (K.P.); (C.F.); (V.C.); (C.M.C.)
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Anna Tessari
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (T.S.); (J.M.); (A.T.); (M.P.); (A.E.B.); (V.E.); (K.P.); (C.F.); (V.C.); (C.M.C.)
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Megan Pawlikowski
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (T.S.); (J.M.); (A.T.); (M.P.); (A.E.B.); (V.E.); (K.P.); (C.F.); (V.C.); (C.M.C.)
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Ashley E. Braddom
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (T.S.); (J.M.); (A.T.); (M.P.); (A.E.B.); (V.E.); (K.P.); (C.F.); (V.C.); (C.M.C.)
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Tasha Posid
- Department of Urology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA;
| | - Debra L. Zynger
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA;
| | - Cindy James
- Mass Spectroscopy and Proteomics Facility, The Ohio State University, Columbus, OH 43210, USA;
| | - Valerio Embrione
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (T.S.); (J.M.); (A.T.); (M.P.); (A.E.B.); (V.E.); (K.P.); (C.F.); (V.C.); (C.M.C.)
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Kareesma Parbhoo
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (T.S.); (J.M.); (A.T.); (M.P.); (A.E.B.); (V.E.); (K.P.); (C.F.); (V.C.); (C.M.C.)
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Claudia Foray
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (T.S.); (J.M.); (A.T.); (M.P.); (A.E.B.); (V.E.); (K.P.); (C.F.); (V.C.); (C.M.C.)
| | - Vincenzo Coppola
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (T.S.); (J.M.); (A.T.); (M.P.); (A.E.B.); (V.E.); (K.P.); (C.F.); (V.C.); (C.M.C.)
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Carlo M. Croce
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (T.S.); (J.M.); (A.T.); (M.P.); (A.E.B.); (V.E.); (K.P.); (C.F.); (V.C.); (C.M.C.)
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Dario Palmieri
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (T.S.); (J.M.); (A.T.); (M.P.); (A.E.B.); (V.E.); (K.P.); (C.F.); (V.C.); (C.M.C.)
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
15
|
Quicker, deeper and stronger imaging: A review of tumor-targeted, near-infrared fluorescent dyes for fluorescence guided surgery in the preclinical and clinical stages. Eur J Pharm Biopharm 2020; 152:123-143. [PMID: 32437752 DOI: 10.1016/j.ejpb.2020.05.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 05/03/2020] [Accepted: 05/03/2020] [Indexed: 12/12/2022]
Abstract
Cancer is a public health problem and the main cause of human mortality and morbidity worldwide. Complete removal of tumors and metastatic lymph nodes in surgery is significantly beneficial for the prognosis of patients. Tumor-targeted, near-infrared fluorescent (NIRF) imaging is an emerging field of real-time intraoperative cancer imaging based on tumor-targeted NIRF dyes. Targeted NIRF dyes contain NIRF fluorophores and specific binding ligands such as antibodies, peptides and small molecules. The present article reviews recently updated tumor-targeted NIRF dyes for the molecular imaging of malignant tumors in the preclinical stage and clinical trials. The strengths and challenges of NIRF agents with tumor-targeting ability are also summarized. Smaller ligands, near infrared II dyes, dual-modality dyes and activatable dyes may contribute to quicker, deeper, stronger imaging in the nearest future. In this review, we highlighted tumor-targeted NIRF dyes for fluorescence-guided surgery and their potential clinical translation.
Collapse
|
16
|
Prévot G, Bsaibess T, Daniel J, Genevois C, Clermont G, Sasaki I, Marais S, Couillaud F, Crauste-Manciet S, Blanchard-Desce M. Multimodal optical contrast agents as new tools for monitoring and tuning nanoemulsion internalisation into cancer cells. From live cell imaging to in vivo imaging of tumours. NANOSCALE ADVANCES 2020; 2:1590-1602. [PMID: 36132308 PMCID: PMC9416932 DOI: 10.1039/c9na00710e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 02/19/2020] [Indexed: 05/28/2023]
Abstract
Tailor-made NIR emitting dyes were designed as multimodal optical probes. These asymmetric amphiphilic compounds show combined intense absorption in the visible region, NIR fluorescence emission, high two-photon absorption in the NIR (with the maximum located around 1000 nm) as well as large Stokes' shift values and second-harmonic generation ability. Thanks to their structure, high loading into nanoemulsions (NEs) could be achieved leading to very high one- and two-photon brightness. These dyes were demonstrated to act as multimodal contrast agents able to generate different optical modalities of interest for bioimaging. Indeed, the uptake and carrier behaviour of the dye-loaded NEs into cancer cells could be monitored by simultaneous two-photon fluorescence and second-harmonic generation optical imaging. Multimodal imaging provided deep insight into the mechanism and kinetics of dye internalisation. Quite interestingly, the nature of the dyes was also found to influence both the kinetics of endocytosis and the internalisation pathways in glioblastoma cancer cells. By modulating the charge distribution within the dyes, the NEs can be tuned to escape lysosomes and enter the mitochondria. Moreover, surface functionalization with PEG macromolecules was realized to yield stealth NIRF-NEs which could be used for in vivo NIRF imaging of subcutaneous tumours in mice.
Collapse
Affiliation(s)
- Geoffrey Prévot
- Univ. Bordeaux, ARNA Laboratory, Team ChemBioPharm, U1212 INSERM - UMR 5320 CNRS 146 Rue Léo Saignat 33076 Bordeaux Cedex France
| | - Talia Bsaibess
- Univ. Bordeaux, Institut des Sciences Moléculaires (CNRS UMR 5255) 33405 Talence France
| | - Jonathan Daniel
- Univ. Bordeaux, Institut des Sciences Moléculaires (CNRS UMR 5255) 33405 Talence France
| | - Coralie Genevois
- Univ. Bordeaux, Molecular Imaging and Innovative Therapies (IMOTION), EA7435 Bordeaux 33000 France
| | - Guillaume Clermont
- Univ. Bordeaux, Institut des Sciences Moléculaires (CNRS UMR 5255) 33405 Talence France
| | - Isabelle Sasaki
- Univ. Bordeaux, Institut des Sciences Moléculaires (CNRS UMR 5255) 33405 Talence France
| | - Sebastien Marais
- Bordeaux Imaging Center, UMS 3420 CNRS - Univ. Bordeaux, US4 Inserm 33000 Bordeaux France
| | - Franck Couillaud
- Univ. Bordeaux, Molecular Imaging and Innovative Therapies (IMOTION), EA7435 Bordeaux 33000 France
| | - Sylvie Crauste-Manciet
- Univ. Bordeaux, ARNA Laboratory, Team ChemBioPharm, U1212 INSERM - UMR 5320 CNRS 146 Rue Léo Saignat 33076 Bordeaux Cedex France
- Pharmaceutical Technology Department, Bordeaux University Hospital Bordeaux France
| | | |
Collapse
|
17
|
Assessment of an scFv Antibody Fragment Against ELTD1 in a G55 Glioblastoma Xenograft Model. Transl Oncol 2020; 13:100737. [PMID: 32208341 PMCID: PMC7090355 DOI: 10.1016/j.tranon.2019.12.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/11/2019] [Accepted: 12/12/2019] [Indexed: 01/27/2023] Open
Abstract
Glioblastoma (GBM), the most common primary brain tumor found in adults, is extremely aggressive. These high-grade gliomas, which are very diffuse, highly vascular, and invasive, undergo unregulated vascular angiogenesis. Despite available treatments, the median survival for patients is dismal. ELTD1 (EGF, latrophilin, and 7 transmembrane domain containing protein 1) is an angiogenic biomarker highly expressed in human high-grade gliomas. Recent studies have demonstrated that the blood-brain barrier, as well as the blood-tumor barrier, is not equally disrupted in GBM patients. This study therefore aimed to optimize an antibody treatment against ELTD1 using a smaller scFv fragment of a monoclonal antibody that binds against the external region of ELTD1 in a G55 glioma xenograft glioma preclinical model. Morphological magnetic resonance imaging (MRI) was used to determine tumor volumes and quantify perfusion rates. We also assessed percent survival following tumor postdetection. Tumor tissue was also assessed to confirm and quantify the presence of the ELTD1 scFv molecular targeted MRI probe, as well as microvessel density and Notch1 levels. In addition, we used molecular-targeted MRI to localize our antibodies in vivo. This approach showed that our scFv antibody attached-molecular MRI probe was effective in targeting and localizing diffuse tumor regions. Through this analysis, we determined that our anti-ELTD1 scFv antibody treatments were successful in increasing survival, decreasing tumor volumes, and normalizing vascular perfusion and Notch1 levels within tumor regions. This study demonstrates that our scFv fragment antibody against ELTD1 may be useful and potential antiangiogenic treatments against GBM.
Collapse
|
18
|
Long M, Lin F, Wang X, Chen X, Liu L, Zhang H, Dong K. Adenovirus-mediated anti-AEG-1 ScFv expression driven by stathmin promoter inhibits tumor growth in cervical cancer. Cancer Cell Int 2020; 20:79. [PMID: 32190003 PMCID: PMC7068931 DOI: 10.1186/s12935-020-1159-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 02/29/2020] [Indexed: 12/14/2022] Open
Abstract
Background Astrocyte-elevated gene-1 (AEG-1) is over-expressed in many cancer cells and has multiple key functions in tumor initiation and progression. Currently, targeted-AEG-1 siRNA is one of the most common techniques to down-regulate AEG-1 expression, but the lack of tumor specificity and available delivery system make it difficult to enter clinical trials. Methods In this study, we creatively developed an adenovirus-mediated anti-AEG-1 single-chain antibody fragment (ScFv) expression system driven by a tumor specific promoter, and experimented with it in human cervical carcinoma cells to investigate the effect on tumor’s proliferation and apoptosis. Results The results showed that of HeLa and SiHa cells treated with this recombinant anti-AEG-1 ScFv adenovirus not only inhibited cell growth, but induced apoptosis both in vitro and in vivo. Furthermore, we also observed that the expressions of several apoptosis-related genes like Akt 1 and c-Myc decreased, while NF-κB (p65) and cleaved caspase 3 increased on protein levels in vivo. Conclusion We concluded that stathmin promoter-driving anti-AEG-1 ScFv adenoviral system may be a breakthrough for its dual-specificity, and serve as an adjuvant tumor specific therapy method in the treatment for human cervical cancers.
Collapse
Affiliation(s)
- Min Long
- Department of Medical Laboratory, Tangdu Hospital, Airforce Military Medical University, Xinsi Road, Xi'an, 710038 Shaanxi China
| | - Fang Lin
- Department of Medical Laboratory, Tangdu Hospital, Airforce Military Medical University, Xinsi Road, Xi'an, 710038 Shaanxi China
| | - Xi Wang
- Department of Medical Laboratory, Tangdu Hospital, Airforce Military Medical University, Xinsi Road, Xi'an, 710038 Shaanxi China
| | - Xi Chen
- Department of Medical Laboratory, Tangdu Hospital, Airforce Military Medical University, Xinsi Road, Xi'an, 710038 Shaanxi China
| | - Li Liu
- Department of Medical Laboratory, Tangdu Hospital, Airforce Military Medical University, Xinsi Road, Xi'an, 710038 Shaanxi China
| | - Huizhong Zhang
- Department of Medical Laboratory, Tangdu Hospital, Airforce Military Medical University, Xinsi Road, Xi'an, 710038 Shaanxi China
| | - Ke Dong
- Department of Medical Laboratory, Tangdu Hospital, Airforce Military Medical University, Xinsi Road, Xi'an, 710038 Shaanxi China
| |
Collapse
|
19
|
Zhao X, Ning Q, Mo Z, Tang S. A promising cancer diagnosis and treatment strategy: targeted cancer therapy and imaging based on antibody fragment. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 47:3621-3630. [PMID: 31468992 DOI: 10.1080/21691401.2019.1657875] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
With the arrival of the precision medicine and personalized treatment era, targeted therapy that improves efficacy and reduces side effects has become the mainstream approach of cancer treatment. Antibody fragments that further enhance penetration and retain the most critical antigen-specific binding functions are considered the focus of research targeting cancer imaging and therapy. Thanks to the superior penetration and rapid blood clearance of antibody fragments, antibody fragment-based imaging agents enable efficient and sensitive imaging of tumour sites. In tumour-targeted therapy, antibody fragments can directly inhibit tumour proliferation and growth, serve as an ideal carrier for delivery of anti-tumour drugs, or manipulate the immune system to eliminate tumour cells. In this review, the excellent physicochemical properties and the basic structure of antibody fragments are expressly depicted depicted, the progress of antibody fragments in cancer therapy and imaging are thoroughly summarized, and the future development of antibody fragments is predicted.
Collapse
Affiliation(s)
- Xuhong Zhao
- Learning Key Laboratory for Pharmacoproteomics of Hunan Province, Institute of Pharmacy and Pharmacology, University of South China , Hengyang , China.,Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, Hunan University of Medicine , Huaihua , China
| | - Qian Ning
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, Hunan University of Medicine , Huaihua , China
| | - Zhongcheng Mo
- Department of Histology and Embryology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China , Hengyang , China
| | - Shengsong Tang
- Learning Key Laboratory for Pharmacoproteomics of Hunan Province, Institute of Pharmacy and Pharmacology, University of South China , Hengyang , China.,Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, Hunan University of Medicine , Huaihua , China
| |
Collapse
|
20
|
Debie P, Hernot S. Emerging Fluorescent Molecular Tracers to Guide Intra-Operative Surgical Decision-Making. Front Pharmacol 2019; 10:510. [PMID: 31139085 PMCID: PMC6527780 DOI: 10.3389/fphar.2019.00510] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 04/24/2019] [Indexed: 12/26/2022] Open
Abstract
Fluorescence imaging is an emerging technology that can provide real-time information about the operating field during cancer surgery. Non-specific fluorescent agents, used for the assessment of blood flow and sentinel lymph node detection, have so far dominated this field. However, over the last decade, several clinical studies have demonstrated the great potential of targeted fluorescent tracers to visualize tumor lesions in a more specific way. This has led to an exponential growth in the development of novel molecular fluorescent contrast agents. In this review, the design of fluorescent molecular tracers will be discussed, with particular attention for agents and approaches that are of interest for clinical translation.
Collapse
Affiliation(s)
| | - Sophie Hernot
- Laboratory for in vivo Cellular and Molecular Imaging (ICMI-BEFY/MIMA), Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
21
|
Diao W, Cai H, Chen L, Jin X, Liao X, Jia Z. Recent Advances in Prostate-Specific Membrane Antigen-Based Radiopharmaceuticals. Curr Top Med Chem 2019; 19:33-56. [PMID: 30706785 DOI: 10.2174/1568026619666190201100739] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 12/13/2018] [Accepted: 12/15/2018] [Indexed: 02/05/2023]
Abstract
BACKGROUND Prostate cancer (PCa) is the most common sex-related malignancy with high mortality in men worldwide. Prostate-specific membrane antigen (PSMA) is overexpressed on the surface of most prostate tumor cells and considered a valuable target for both diagnosis and therapy of prostate cancer. A series of radiolabeled agents have been developed based on the featured PSMA ligands in the previous decade and have demonstrated promising outcomes in clinical research of primary and recurrent PCa. Furthermore, the inspiring response and safety of lutetium-177-PSMA-617 (177Lu-PSMA-617) radiotherapy represent the potential for expanded therapeutic options for metastatic castration-resistant PCa. Retrospective cohort studies have revealed that radiolabeled PSMA agents are the mainstays of the current success, especially in detecting prostate cancer with metastasis and biochemical recurrence. OBJECTIVE This review is intended to present a comprehensive overview of the current literature on PSMA ligand-based agents for both radionuclide imaging and therapeutic approaches, with a focus on those that have been clinically adopted. CONCLUSION PSMA-based diagnosis and therapy hold great promise for improving the clinical management of prostate cancer.
Collapse
Affiliation(s)
- Wei Diao
- Department of Nuclear Medicine, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Huawei Cai
- Department of Nuclear Medicine, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Lihong Chen
- Department of Biochemistry & Molecular Biology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Xi Jin
- Institute of Urology, Department of Urology, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Xinyang Liao
- Institute of Urology, Department of Urology, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Zhiyun Jia
- Department of Nuclear Medicine, West China Hospital, Sichuan University, 610041, Chengdu, China
| |
Collapse
|
22
|
Olson MT, Ly QP, Mohs AM. Fluorescence Guidance in Surgical Oncology: Challenges, Opportunities, and Translation. Mol Imaging Biol 2019; 21:200-218. [PMID: 29942988 PMCID: PMC6724738 DOI: 10.1007/s11307-018-1239-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Surgical resection continues to function as the primary treatment option for most solid tumors. However, the detection of cancerous tissue remains predominantly subjective and reliant on the expertise of the surgeon. Surgery that is guided by fluorescence imaging has shown clinical relevance as a new approach to detecting the primary tumor, tumor margins, and metastatic lymph nodes. It is a technique to reduce recurrence and increase the possibility of a curative resection. While significant progress has been made in developing this emerging technology as a tool to assist the surgeon, further improvements are still necessary. Refining imaging agents and tumor targeting strategies to be a precise and reliable surgical strategy is essential in order to translate this technology into patient care settings. This review seeks to provide a comprehensive update on the most recent progress of fluorescence-guided surgery and its translation into the clinic. By highlighting the current status and recent developments of fluorescence image-guided surgery in the field of surgical oncology, we aim to offer insight into the challenges and opportunities that require further investigation.
Collapse
Affiliation(s)
- Madeline T Olson
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Quan P Ly
- Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Aaron M Mohs
- Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 5-12315 Scott Research Tower, Omaha, NE, 68198, USA.
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
23
|
Fogaça RL, Alvarenga LM, Woiski TD, Becker-Finco A, Teixeira KN, Silva SK, de Moraes RN, Noronha LD, Noiray M, de Figueiredo BC, Billiald P, Moura JD. Biomolecular engineering of antidehydroepiandrosterone antibodies: a new perspective in cancer diagnosis and treatment using single-chain antibody variable fragment. Nanomedicine (Lond) 2019; 14:689-705. [PMID: 30691340 DOI: 10.2217/nnm-2018-0230] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Aim: To develop a monoclonal antibody against dehydroepiandrosterone (DHEA) and miniaturize it, generating a single-chain antibody variable fragment (scFv) against DHEA as an adrenocortical carcinoma (ACC) marker. Material & methods: DHEA conjugated to keyhole limpet hemocyanin was used as an immunogen to obtain anti-DHEA hybridomas. Variable fragments were cloned from hybridoma 5B7 total RNA, and used to detect DHEA in normal adrenal tissue and ACC cells. Results: IgM monoclonal antibody was highly specific, and the recombinant scFv preserved parental antibody characteristics, allowing tissue localization of DHEA. Conclusion: Undefined small lesions are challenges for clinicians and impact clinical adrenocortical tumor management. Generating an anti-DHEA scFv facilitates development of imaging tests for early diagnosis of pediatric ACC.
Collapse
Affiliation(s)
- Rafaela L Fogaça
- Pós-graduação em Engenharia de Bioprocessos e Biotecnologia, Curitiba, Brazil
- Departamento de Patologia Básica – UFPR, Curitiba, Brazil
| | - Larissa M Alvarenga
- Pós-graduação em Engenharia de Bioprocessos e Biotecnologia, Curitiba, Brazil
- Pós graduação em Microbiologia, Parasitologia e Patologia, Curitiba, Brazil
- Departamento de Patologia Básica – UFPR, Curitiba, Brazil
| | - Thiago D Woiski
- Instituto Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, Brazil
| | | | | | - Sabrina K Silva
- Pós graduação em Microbiologia, Parasitologia e Patologia, Curitiba, Brazil
- Departamento de Patologia Básica – UFPR, Curitiba, Brazil
| | | | - Lúcia de Noronha
- Pós graduação em Microbiologia, Parasitologia e Patologia, Curitiba, Brazil
- Departamento de Medicina, PUCPR, Curitiba, Brazil
| | - Magali Noiray
- Université Paris-Sud, Faculté de Pharmacie, INSERM UMR S1193 & UMS IPSIT, Châtenay-Malabry, France
| | | | - Philippe Billiald
- Université Paris-Sud, Faculté de Pharmacie, INSERM UMR S1193 & UMS IPSIT, Châtenay-Malabry, France
- Muséum National d'Histoire Naturelle, CNRS UMR 7245, Paris, France
| | - Juliana de Moura
- Pós-graduação em Engenharia de Bioprocessos e Biotecnologia, Curitiba, Brazil
- Pós graduação em Microbiologia, Parasitologia e Patologia, Curitiba, Brazil
- Departamento de Patologia Básica – UFPR, Curitiba, Brazil
| |
Collapse
|
24
|
Fu R, Carroll L, Yahioglu G, Aboagye EO, Miller PW. Antibody Fragment and Affibody ImmunoPET Imaging Agents: Radiolabelling Strategies and Applications. ChemMedChem 2018; 13:2466-2478. [PMID: 30246488 PMCID: PMC6587488 DOI: 10.1002/cmdc.201800624] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Indexed: 12/12/2022]
Abstract
Antibodies have long been recognised as potent vectors for carrying diagnostic medical radionuclides, contrast agents and optical probes to diseased tissue for imaging. The area of ImmunoPET combines the use of positron emission tomography (PET) imaging with antibodies to improve the diagnosis, staging and monitoring of diseases. Recent developments in antibody engineering and PET radiochemistry have led to a new wave of experimental ImmunoPET imaging agents that are based on a range of antibody fragments and affibodies. In contrast to full antibodies, engineered affibody proteins and antibody fragments such as minibodies, diabodies, single-chain variable region fragments (scFvs), and nanobodies are much smaller but retain the essential specificities and affinities of full antibodies in addition to more desirable pharmacokinetics for imaging. Herein, recent key developments in the PET radiolabelling strategies of antibody fragments and related affibody molecules are highlighted, along with the main PET imaging applications of overexpressed antigen-associated tumours and immune cells.
Collapse
Affiliation(s)
- Ruisi Fu
- Department of ChemistryImperial College LondonExhibition RoadSouth Kensington, LondonSW7 2AZUK
- Comprehensive Cancer Imaging Centre, Department of Surgery and CancerImperial College London, Hammersmith CampusDu Cane RoadLondonW12 0NNUK
| | - Laurence Carroll
- Comprehensive Cancer Imaging Centre, Department of Surgery and CancerImperial College London, Hammersmith CampusDu Cane RoadLondonW12 0NNUK
| | - Gokhan Yahioglu
- Department of ChemistryImperial College LondonExhibition RoadSouth Kensington, LondonSW7 2AZUK
- Antikor Biopharma Ltd.StevenageSG1 2FXUK
| | - Eric O. Aboagye
- Comprehensive Cancer Imaging Centre, Department of Surgery and CancerImperial College London, Hammersmith CampusDu Cane RoadLondonW12 0NNUK
| | - Philip W. Miller
- Department of ChemistryImperial College LondonExhibition RoadSouth Kensington, LondonSW7 2AZUK
| |
Collapse
|
25
|
VCAM-1 + macrophages guide the homing of HSPCs to a vascular niche. Nature 2018; 564:119-124. [PMID: 30455424 DOI: 10.1038/s41586-018-0709-7] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 09/14/2018] [Indexed: 12/13/2022]
Abstract
Haematopoietic stem and progenitor cells (HSPCs) give rise to all blood lineages that support the entire lifespan of vertebrates1. After HSPCs emerge from endothelial cells within the developing dorsal aorta, homing allows the nascent cells to anchor in their niches for further expansion and differentiation2-5. Unique niche microenvironments, composed of various blood vessels as units of microcirculation and other niche components such as stromal cells, regulate this process6-9. However, the detailed architecture of the microenvironment and the mechanism for the regulation of HSPC homing remain unclear. Here, using advanced live imaging and a cell-labelling system, we perform high-resolution analyses of the HSPC homing in caudal haematopoietic tissue of zebrafish (equivalent to the fetal liver in mammals), and reveal the role of the vascular architecture in the regulation of HSPC retention. We identify a VCAM-1+ macrophage-like niche cell population that patrols the inner surface of the venous plexus, interacts with HSPCs in an ITGA4-dependent manner, and directs HSPC retention. These cells, named 'usher cells', together with caudal venous capillaries and plexus, define retention hotspots within the homing microenvironment. Thus, the study provides insights into the mechanism of HSPC homing and reveals the essential role of a VCAM-1+ macrophage population with patrolling behaviour in HSPC retention.
Collapse
|
26
|
Generation and characterization of novel recombinant anti-hERG1 scFv antibodies for cancer molecular imaging. Oncotarget 2018; 9:34972-34989. [PMID: 30405887 PMCID: PMC6201861 DOI: 10.18632/oncotarget.26200] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 09/21/2018] [Indexed: 01/12/2023] Open
Abstract
Modern molecular imaging techniques have greatly improved tumor detection and post-treatment follow-up of cancer patients. In this context, antibody-based imaging is rapidly becoming the gold standard, since it combines the unique specificity of antibodies with the sensitivity of the different imaging technologies. The aim of this study was to generate and characterize antibodies in single chain Fragment variable (scFv) format directed to an emerging cancer biomarker, the human ether-à-go-go-related gene-1 (hERG1) potassium channel, and to obtain a proof of concept for their potential use for in vivo molecular imaging. The anti-hERG1scFv was generated from a full length monoclonal antibody and then mutagenized, substituting a Phenylalanine residue in the third framework of the VH domain with a Cysteine residue. The resulting scFv-hERG1-Cys showed much higher stability and protein yield, increased affinity and more advantageous binding kinetics, compared to the “native” anti-hERG1scFv. The scFv-hERG1-Cys was hence chosen and characterized: it showed a good binding to the native hERG1 antigen expressed on cells, was stable in serum and displayed a fast pharmacokinetic profile once injected intravenously in nude mice. The calculated half-life was 3.1 hours and no general toxicity or cardiac toxic effects were detected. Finally, the in vivo distribution of an Alexa Fluor 750 conjugated scFv-hERG1-Cys was evaluated both in healthy and tumor-bearing nude mice, showing a good tumor-to-organ ratio, ideal for visualizing hERG1-expressing tumor masses in vivo. In conclusion, the scFv-hERG1-Cys possesses features which make it a suitable tool for application in cancer molecular imaging.
Collapse
|
27
|
Pesapane F, Czarniecki M, Suter MB, Turkbey B, Villeirs G. Imaging of distant metastases of prostate cancer. Med Oncol 2018; 35:148. [DOI: 10.1007/s12032-018-1208-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 09/06/2018] [Indexed: 02/06/2023]
|
28
|
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]
|
29
|
Alibakhshi A, Abarghooi Kahaki F, Ahangarzadeh S, Yaghoobi H, Yarian F, Arezumand R, Ranjbari J, Mokhtarzadeh A, de la Guardia M. Targeted cancer therapy through antibody fragments-decorated nanomedicines. J Control Release 2017; 268:323-334. [DOI: 10.1016/j.jconrel.2017.10.036] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 10/22/2017] [Accepted: 10/24/2017] [Indexed: 01/10/2023]
|
30
|
Thoeny HC, Barbieri S, Froehlich JM, Turkbey B, Choyke PL. Functional and Targeted Lymph Node Imaging in Prostate Cancer: Current Status and Future Challenges. Radiology 2017; 285:728-743. [PMID: 29155624 DOI: 10.1148/radiol.2017161517] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Patients with prostate cancer who have regional lymph node (LN) metastases face an increased risk of death from disease and are therefore treated aggressively. Surgical LN dissection is the established method of staging regional nodes; however, this invasive technique carries substantial morbidities and a noninvasive imaging method is needed to reduce or eliminate the need for extended pelvic LN dissections (ePLND). Conventional computed tomography and magnetic resonance (MR) imaging have proven insensitive and nonspecific because both use nodal size criteria, which is notoriously inaccurate. Novel imaging techniques such as functional MR imaging by using diffusion-weighted MR imaging, MR lymphography with iron oxide particles, and targeted positron emission tomography imaging are currently under development and appear to improve LN staging of prostate cancer. Although progress is being made in staging nodes with imaging, it has not reached the point of replacing ePLND. In this review, the strengths and limitations of these new functional and targeted LN imaging techniques for prostate cancer are discussed. © RSNA, 2017.
Collapse
Affiliation(s)
- Harriet C Thoeny
- From the Department of Diagnostic, Pediatric, and Interventional Radiology, Inselspital University Hospital, Bern, Switzerland (H.C.T., S.B., J.M.F.); and Molecular Imaging Program, National Cancer Institute, National Institutes of Health, 10 Center Dr, Room B3B69F, Bethesda, MD 20892 (B.T., P.L.C.)
| | - Sebastiano Barbieri
- From the Department of Diagnostic, Pediatric, and Interventional Radiology, Inselspital University Hospital, Bern, Switzerland (H.C.T., S.B., J.M.F.); and Molecular Imaging Program, National Cancer Institute, National Institutes of Health, 10 Center Dr, Room B3B69F, Bethesda, MD 20892 (B.T., P.L.C.)
| | - Johannes M Froehlich
- From the Department of Diagnostic, Pediatric, and Interventional Radiology, Inselspital University Hospital, Bern, Switzerland (H.C.T., S.B., J.M.F.); and Molecular Imaging Program, National Cancer Institute, National Institutes of Health, 10 Center Dr, Room B3B69F, Bethesda, MD 20892 (B.T., P.L.C.)
| | - Baris Turkbey
- From the Department of Diagnostic, Pediatric, and Interventional Radiology, Inselspital University Hospital, Bern, Switzerland (H.C.T., S.B., J.M.F.); and Molecular Imaging Program, National Cancer Institute, National Institutes of Health, 10 Center Dr, Room B3B69F, Bethesda, MD 20892 (B.T., P.L.C.)
| | - Peter L Choyke
- From the Department of Diagnostic, Pediatric, and Interventional Radiology, Inselspital University Hospital, Bern, Switzerland (H.C.T., S.B., J.M.F.); and Molecular Imaging Program, National Cancer Institute, National Institutes of Health, 10 Center Dr, Room B3B69F, Bethesda, MD 20892 (B.T., P.L.C.)
| |
Collapse
|
31
|
Haylock AK, Nilvebrant J, Mortensen A, Velikyan I, Nestor M, Falk R. Generation and evaluation of antibody agents for molecular imaging of CD44v6-expressing cancers. Oncotarget 2017; 8:65152-65170. [PMID: 29029420 PMCID: PMC5630320 DOI: 10.18632/oncotarget.17996] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 04/26/2017] [Indexed: 11/25/2022] Open
Abstract
AIM The aim of this study was to generate and characterize scFv antibodies directed to human CD44v6, as well as to radiolabel and evaluate top candidates in vitro and in vivo for their potential use in CD44v6-targeted molecular imaging in cancer patients. MATERIALS AND METHODS Phage display selections were used to isolate CD44v6-specific scFvs. A chain shuffling strategy was employed for affinity maturation based on a set of CD44v6-specific first-generation clones. Two second-generation scFv clones were then chosen for labeling with 111In or 125I and assessed for CD44v6-specific binding on cultured tumor cells. In vivo uptake and distribution was evaluated in tumor-bearing mice using a dual tumor model. Finally, a proof-of-concept small animal PET-CT study was performed on one of the candidates labeled with 124I. RESULTS Two affinity-matured clones, CD44v6-scFv-A11 and CD44v6-scFv-H12, displayed promising binding kinetics. Seven out of eight radiolabeled conjugates demonstrated CD44v6-specific binding. In vivo studies on selected candidates demonstrated very advantageous tumor-to-organ ratios, in particular for iodinated conjugates, where 125I-labeled scFvs exhibited favorable kinetics and tumor-to-blood ratios above five already at 24 hours p.i.. The small animal PET-CT study using 124I-labeled CD44v6-scFv-H12 was in line with the biodistribution data, clearly visualizing the high CD44v6-expressing tumor. CONCLUSION The single chain fragments, CD44v6-scFv-A11 and CD44v6-scFv-H12 specifically bind to CD44v6, and the radiolabeled counterparts provide high tumor-to-blood ratios and fast clearance from organs and blood. We conclude that radioiodinated CD44v6-scFv-A11 and CD44v6-scFv-H12 possess features highly suitable for stringent molecular imaging.
Collapse
Affiliation(s)
- Anna-Karin Haylock
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Johan Nilvebrant
- Division of Protein Technology, School of Biotechnology, Royal Institute of Technology, Stockholm, Sweden
| | - Anja Mortensen
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Irina Velikyan
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Marika Nestor
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ronny Falk
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
32
|
Bedford R, Tiede C, Hughes R, Curd A, McPherson MJ, Peckham M, Tomlinson DC. Alternative reagents to antibodies in imaging applications. Biophys Rev 2017; 9:299-308. [PMID: 28752365 PMCID: PMC5578921 DOI: 10.1007/s12551-017-0278-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 07/06/2017] [Indexed: 12/21/2022] Open
Abstract
Antibodies have been indispensable tools in molecular biology, biochemistry and medical research. However, a number of issues surrounding validation, specificity and batch variation of commercially available antibodies have prompted research groups to develop novel non-antibody binding reagents. The ability to select highly specific monoclonal non-antibody binding proteins without the need for animals, the ease of production and the ability to site-directly label has enabled a wide variety of applications to be tested, including imaging. In this review, we discuss the success of a number of non-antibody reagents in imaging applications, including the recently reported Affimer.
Collapse
Affiliation(s)
- R Bedford
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, UK
| | - C Tiede
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, UK
| | - R Hughes
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, UK
| | - A Curd
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, UK
| | - M J McPherson
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, UK
| | - Michelle Peckham
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, UK.
| | - Darren C Tomlinson
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, UK.
| |
Collapse
|
33
|
Adumeau L, Genevois C, Roudier L, Schatz C, Couillaud F, Mornet S. Impact of surface grafting density of PEG macromolecules on dually fluorescent silica nanoparticles used for the in vivo imaging of subcutaneous tumors. Biochim Biophys Acta Gen Subj 2017; 1861:1587-1596. [PMID: 28179102 DOI: 10.1016/j.bbagen.2017.01.036] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/30/2017] [Accepted: 01/31/2017] [Indexed: 01/18/2023]
Abstract
BACKGROUND In the context of systematically administered nanomedicines, the physicochemistry of NP surfaces must be controlled as a prerequisite to improve blood circulation time, and passive and active targeting. In particular, there is a real need to develop NP stealth and labelling for both in vivo and microscopic fluorescence imaging in a mice model. METHODS We have synthesized NIR/red dually fluorescent silica nanoparticles of 19nm covalently covered by a PEG layer of different grafting density in the brush conformational regime by using a reductive amination reaction. These particles were characterized by TEM, DRIFT, DLS, TGA, ζ potential measurements, UV-vis and fluorescence spectroscopy. Prostate tumors were generated in mice by subcutaneous injection of RM1-CMV-Fluc cells. Tumor growth was monitored by BLI after a D-luciferin injection. Four samples of PEGylated fluorescent NPs were individually intravenously injected into 6 mice (N=6, total 24 mice). Nanoparticle distribution was investigated using in vivo fluorescence reflectance imaging (FRI) over 48h and microscopy imaging was employed to localize the NPs within tumors in vitro. RESULTS Fluorescent NP accumulation, due to the enhanced permeability and retention (EPR) effect, increases gradually as a function of increased PEG surface grafting density with a huge difference observed for the highest density grafting. For the highest grafting density, a blood circulation time of up to 24h was observed with a strong reduction in uptake by the liver. In vivo experimental results suggest that the biodistribution of NPs is very sensitive to slight variations in surface grafting density when the NPs present a high curvature radius. CONCLUSION This study underlines the need to compensate a high curvature radius with a PEG-saturated NP surface to improve blood circulation and accumulation within tumors through the EPR effect. Dually fluorescent NPs PEGylated to saturation display physical properties useful for assessing the susceptibility of tumors to the EPR effect. GENERAL SIGNIFICANCE Control of the physicochemical features of nanoparticle surfaces to improve blood circulation times and monitoring of the EPR effect. This article is part of a Special Issue entitled "Recent Advances in Bionanomaterials" Guest Editor: Dr. Marie-Louise Saboungi and Dr. Samuel D. Bader.
Collapse
Affiliation(s)
- Laurent Adumeau
- CNRS, Univ. Bordeaux, ICMCB, UPR 9048, F-33600 Pessac, France
| | - Coralie Genevois
- Univ. Bordeaux, EA 7435 IMOTION - Imagerie moléculaire et thérapies innovantes en oncologie, 33706 Bordeaux, France
| | - Lydia Roudier
- CNRS, Univ. Bordeaux, ICMCB, UPR 9048, F-33600 Pessac, France
| | | | - Franck Couillaud
- Univ. Bordeaux, EA 7435 IMOTION - Imagerie moléculaire et thérapies innovantes en oncologie, 33706 Bordeaux, France.
| | - Stéphane Mornet
- CNRS, Univ. Bordeaux, ICMCB, UPR 9048, F-33600 Pessac, France.
| |
Collapse
|