1
|
Sarlak S, Pagès G, Luciano F. Enhancing radiotherapy techniques for Triple-Negative breast cancer treatment. Cancer Treat Rev 2025; 136:102939. [PMID: 40286498 DOI: 10.1016/j.ctrv.2025.102939] [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: 01/31/2025] [Revised: 03/22/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025]
Abstract
Breast cancer is the most prevalent cancer among women worldwide, with various subtypes that require distinct treatment approaches. Among these, Triple-Negative Breast Bancer (TNBC) is recognized as the most aggressive form, often associated with poor prognosis due to its lack of targeted therapeutic options. This review specifically focuses on Radiotherapy (RT) as a treatment modality for TNBC, evaluating recent advancements and ongoing challenges, particularly the issue of radioresistance. RT remains an essential part in the management of breast cancer, including TNBC. Over the years, multiple improvements have been made to enhance RT effectiveness and minimize resistance. The introduction of advanced techniques such as Stereotactic Body Radiation Therapy (SBRT) and Stereotactic Radiosurgery (SRS) has significantly improved precision and reduced toxicity. More recently, proton radiation therapy, a novel RT modality, has been introduced, offering enhanced dose distribution and reducing damage to surrounding healthy tissues. Despite these technological advancements, a subset of TNBC patients continues to exhibit resistance to RT, leading to recurrence and poor treatment outcomes. To overcome radioresistance, there is an increasing interest in combining RT with targeted therapeutic agents that sensitize cancer cells to radiation. Radiosensitizing drugs have been explored to enhance the efficacy of RT by making cancer cells more susceptible to radiation-induced damage. Potential candidates include DNA damage repair inhibitors, immune checkpoint inhibitors, and small-molecule targeted therapies that interfere with key survival pathways in TNBC cells. In conclusion, while RT remains a crucial modality for TNBC treatment, radioresistance remains a significant challenge. Future research should focus on optimizing RT techniques while integrating radiosensitizing agents to improve treatment efficacy. By combining RT with targeted drug therapy, a more effective and personalized treatment approach can be developed, ultimately improving patient outcomes and reducing recurrence rates in TNBC.
Collapse
Affiliation(s)
- Saharnaz Sarlak
- Cote d'Azur University (UCA), Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR 7284, CNRS UMR 7284; INSERM U1081, Centre Antoine Lacassagne, France.
| | - Gilles Pagès
- Cote d'Azur University (UCA), Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR 7284, CNRS UMR 7284; INSERM U1081, Centre Antoine Lacassagne, France.
| | - Frédéric Luciano
- Cote d'Azur University (UCA), Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR 7284, CNRS UMR 7284; INSERM U1081, Centre Antoine Lacassagne, France.
| |
Collapse
|
2
|
N'Guessan É, Raes F, Ahmadi M, Bacot S, Dumas L, Leenhardt J, Debiossat M, André C, Lenormand JL, Ghezzi C, Fagret D, Lombardi C, Broisat A. Enhancing theranostic potential of anti-mesothelin sdAb through site-specific labeling at a unique conserved lysine by molecular engineering. EJNMMI Radiopharm Chem 2025; 10:19. [PMID: 40293556 PMCID: PMC12037457 DOI: 10.1186/s41181-025-00340-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Mesothelin is a 40 kDa glycoprotein overexpressed in several cancers, including triple-negative breast cancer (TNBC). The anti-mesothelin single-domain antibody (sdAb, or nanobody) A1 can serve as a radio-theranostic agent, but random DOTA conjugation on lysines yields heterogeneous products. RESULTS We reengineered A1-His by directed mutagenesis to produce four single-lysine variants (A1K1-His, A1K2-His, A1K3-His, and A1K4-His). Each was site-specifically conjugated with p-SCN-Bn-DOTA, radiolabeled with 68Ga, and evaluated by PET imaging in mice bearing HCC70 TNBC xenografts, followed by ex vivo biodistribution at 1 h post-injection. All mutants were successfully produced and site-specifically conjugated. A1K1-His showed lower conjugation efficiency and increased liver/spleen retention, whereas A1K3-His exhibited reduced stability. A1K2-His and A1K4-His performed best overall. Removing the His-tag and administering gelofusin further lowered renal uptake. Notably, A1K2 displayed tumor-to-kidney and tumor-to-liver ratios 2.4 and 1.9 times higher, respectively, than A1K4 (p < 0.01). CONCLUSIONS For the first time, site-specific DOTA conjugation using sdAb derivatives containing a single lysine was achieved, avoiding the production of mixed final compounds. These findings identify 68Ga-DOTA-A1K2 as the leading candidate for mesothelin-expressing tumor imaging with minimal off-target uptake. Ongoing studies will assess its therapeutic utility with 177Lu-DOTA-A1K2. Since these four lysines are conserved in many sdAbs, this strategy may be broadly applicable for site-specific sdAb labeling.
Collapse
Affiliation(s)
| | - Florian Raes
- Univ. Grenoble Alpes, INSERM U1039, LRB, Grenoble, France
| | - Mitra Ahmadi
- Univ. Grenoble Alpes, INSERM U1039, LRB, Grenoble, France
| | - Sandrine Bacot
- Univ. Grenoble Alpes, INSERM U1039, LRB, Grenoble, France
| | - Laurent Dumas
- Univ. Grenoble Alpes, INSERM U1039, LRB, Grenoble, France
| | - Julien Leenhardt
- Univ. Grenoble Alpes, INSERM U1039, LRB, Grenoble, France
- Department of Nuclear Medicine, Univ. Grenoble Alpes, CHU Grenoble Alpes, Grenoble, France
| | | | - Clémence André
- Univ. Grenoble Alpes, INSERM U1039, LRB, Grenoble, France
| | | | | | - Daniel Fagret
- Univ. Grenoble Alpes, INSERM U1039, LRB, Grenoble, France
| | - Charlotte Lombardi
- Univ. Grenoble Alpes, INSERM U1039, LRB, Grenoble, France
- Univ. Grenoble Alpes, CNRS U5525, TIMC-Tree, La Tronche, France
| | - Alexis Broisat
- Univ. Grenoble Alpes, INSERM U1039, LRB, Grenoble, France.
| |
Collapse
|
3
|
Benloucif A, Meyer D, Balasse L, Goubard A, Danner L, Bouhlel A, Castellano R, Guillet B, Chames P, Kerfelec B. Rapid nanobody-based imaging of mesothelin expressing malignancies compatible with blocking therapeutic antibodies. Front Immunol 2023; 14:1200652. [PMID: 37388728 PMCID: PMC10303918 DOI: 10.3389/fimmu.2023.1200652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/25/2023] [Indexed: 07/01/2023] Open
Abstract
Introduction Mesothelin (MSLN) is overexpressed in a wide variety of cancers with few therapeutic options and has recently emerged as an attractive target for cancer therapy, with a large number of approaches currently under preclinical and clinical investigation. In this respect, developing mesothelin specific tracers as molecular companion tools for predicting patient eligibility, monitoring then response to mesothelin-targeting therapies, and tracking the evolution of the disease or for real-time visualisation of tumours during surgery is of growing importance. Methods We generated by phage display a nanobody (Nb S1) and used enzymatic approaches were used to site-directed conjugate Nb S1 with either ATTO 647N fluorochrome or NODAGA chelator for fluorescence and positron emission tomography imaging (PET) respectively. Results We demonstrated that Nb S1 displays a high apparent affinity and specificity for human mesothelin and demonstrated that the binding, although located in the membrane distal domain of mesothelin, is not impeded by the presence of MUC16, the only known ligand of mesothelin, nor by the therapeutic antibody amatuximab. In vivo experiments showed that both ATTO 647N and [68Ga]Ga-NODAGA-S1 rapidly and specifically accumulated in mesothelin positive tumours compared to mesothelin negative tumours or irrelevant Nb with a high tumour/background ratio. The ex vivo biodistribution profile analysis also confirmed a significantly higher uptake of Nb S1 in MSLN-positive tumours than in MSLNlow tumours. Conclusion We demonstrated for the first time the use of an anti-MSLN nanobody as PET radiotracer for same day imaging of MSLN+ tumours, targeting an epitope compatible with the monitoring of amatuximab-based therapies and current SS1-derived-drug conjugates.
Collapse
Affiliation(s)
- Abdennour Benloucif
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Damien Meyer
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Laure Balasse
- Aix Marseille Univ, CNRS, Centre Européen de Recherche en Imagerie Medicale (CERIMED), Marseille, France
- Aix-marseille University, INSERM, INRAE, Centre de recherche en Cardiovasculaire et Nutrition (C2VN), Marseille, France
| | - Armelle Goubard
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, TrGET Preclinical Platform, Marseille, France
| | - Lucile Danner
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Ahlem Bouhlel
- Aix Marseille Univ, CNRS, Centre Européen de Recherche en Imagerie Medicale (CERIMED), Marseille, France
- Aix-marseille University, INSERM, INRAE, Centre de recherche en Cardiovasculaire et Nutrition (C2VN), Marseille, France
| | - Rémy Castellano
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, TrGET Preclinical Platform, Marseille, France
| | - Benjamin Guillet
- Aix Marseille Univ, CNRS, Centre Européen de Recherche en Imagerie Medicale (CERIMED), Marseille, France
- Aix-marseille University, INSERM, INRAE, Centre de recherche en Cardiovasculaire et Nutrition (C2VN), Marseille, France
| | - Patrick Chames
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Brigitte Kerfelec
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| |
Collapse
|
4
|
Küppers J, Kürpig S, Bundschuh RA, Essler M, Lütje S. Radiolabeling Strategies of Nanobodies for Imaging Applications. Diagnostics (Basel) 2021; 11:1530. [PMID: 34573872 PMCID: PMC8471529 DOI: 10.3390/diagnostics11091530] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/30/2021] [Accepted: 08/20/2021] [Indexed: 02/06/2023] Open
Abstract
Nanobodies are small recombinant antigen-binding fragments derived from camelid heavy-chain only antibodies. Due to their compact structure, pharmacokinetics of nanobodies are favorable compared to full-size antibodies, allowing rapid accumulation to their targets after intravenous administration, while unbound molecules are quickly cleared from the circulation. In consequence, high signal-to-background ratios can be achieved, rendering radiolabeled nanobodies high-potential candidates for imaging applications in oncology, immunology and specific diseases, for instance in the cardiovascular system. In this review, a comprehensive overview of central aspects of nanobody functionalization and radiolabeling strategies is provided.
Collapse
Affiliation(s)
- Jim Küppers
- Department of Nuclear Medicine, University Hospital Bonn, 53127 Bonn, Germany; (S.K.); (R.A.B.); (M.E.); (S.L.)
| | | | | | | | | |
Collapse
|
5
|
Montemagno C, Cassim S, De Leiris N, Durivault J, Faraggi M, Pagès G. Pancreatic Ductal Adenocarcinoma: The Dawn of the Era of Nuclear Medicine? Int J Mol Sci 2021; 22:6413. [PMID: 34203923 PMCID: PMC8232627 DOI: 10.3390/ijms22126413] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), accounting for 90-95% of all pancreatic tumors, is a highly devastating disease associated with poor prognosis. The lack of accurate diagnostic tests and failure of conventional therapies contribute to this pejorative issue. Over the last decade, the advent of theranostics in nuclear medicine has opened great opportunities for the diagnosis and treatment of several solid tumors. Several radiotracers dedicated to PDAC imaging or internal vectorized radiotherapy have been developed and some of them are currently under clinical consideration. The functional information provided by Positron Emission Tomography (PET) or Single Photon Emission Computed Tomography (SPECT) could indeed provide an additive diagnostic value and thus help in the selection of patients for targeted therapies. Moreover, the therapeutic potential of β-- and α-emitter-radiolabeled agents could also overcome the resistance to conventional therapies. This review summarizes the current knowledge concerning the recent developments in the nuclear medicine field for the management of PDAC patients.
Collapse
Affiliation(s)
- Christopher Montemagno
- Département de Biologie Médicale, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (S.C.); (J.D.); (G.P.)
- Institute for Research on Cancer and Aging of Nice, Centre Antoine Lacassagne, CNRS UMR 7284 and IN-SERM U1081, Université Cote d’Azur, 06200 Nice, France
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur—Centre Scientifique de Monaco, 98000 Monaco, Monaco
| | - Shamir Cassim
- Département de Biologie Médicale, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (S.C.); (J.D.); (G.P.)
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur—Centre Scientifique de Monaco, 98000 Monaco, Monaco
| | - Nicolas De Leiris
- Nuclear Medicine Department, Grenoble-Alpes University Hospital, 38000 Grenoble, France;
- Laboratoire Radiopharmaceutiques Biocliniques, Univ. Grenoble Alpes, INSERM, CHU Grenoble Alpes, 38000 Grenoble, France
| | - Jérôme Durivault
- Département de Biologie Médicale, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (S.C.); (J.D.); (G.P.)
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur—Centre Scientifique de Monaco, 98000 Monaco, Monaco
| | - Marc Faraggi
- Centre Hospitalier Princesse Grace, Nuclear Medicine Department, 98000 Monaco, Monaco;
| | - Gilles Pagès
- Département de Biologie Médicale, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (S.C.); (J.D.); (G.P.)
- Institute for Research on Cancer and Aging of Nice, Centre Antoine Lacassagne, CNRS UMR 7284 and IN-SERM U1081, Université Cote d’Azur, 06200 Nice, France
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur—Centre Scientifique de Monaco, 98000 Monaco, Monaco
| |
Collapse
|
6
|
Noninvasive Classification of Human Triple Negative Breast Cancer by PET Imaging with GRP78-Targeted Molecular Probe [ 68Ga]DOTA-VAP. Mol Imaging Biol 2021; 22:772-779. [PMID: 31452065 DOI: 10.1007/s11307-019-01416-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
PURPOSE There is currently no effective noninvasive method for accurate molecular typing of triple negative breast cancer (TNBC) except needle biopsy. Glucoregulated Protein 78 (GRP78) is overexpressed in TNBC cells and tumors which closely related to the invasion, metastasis, and drug resistance of cancer. Meanwhile, it has been verified that VAP peptide bind specifically to GRP78 in vitro and in vivo. In this study, we constructed a GRP78-targeted molecular probe Ga-68-radiolabeled DOTA-VAP conjugate ([68Ga]DOTA-VAP) based on VAP peptide, and evaluated its potential to distinguish TNBC from non-TNBC tumors. PROCEDURES DOTA-VAP was synthesized and then radiolabeled with Ga-68 to obtain [68Ga]DOTA-VAP. The expression of GRP78 in TNBC MDA-MB-231 and non-TNBC MCF-7 cells was validated by Western Blot, and cell binding or uptake experiments with both [68Ga]DOTA-VAP and 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) were also performed. Biodistribution analysis and positron emission tomography (PET) imaging of [68Ga]DOTA-VAP were carried out in subcutaneous MDA-MB-231 and MCF-7 human breast cancer tumor models with [18F]FDG PET imaging as comparison. RESULTS [68Ga]DOTA-VAP was prepared with high radiochemical purity which showed excellent stability in vitro. The MDA-MB-231 tumors were clearly visualized by [68Ga]DOTA-VAP PET imaging with a low background, except for the relatively high liver uptake. Cells and tumors of MDA-MB-231 could be distinguished from MCF-7 by [68Ga]DOTA-VAP instead of [18F]FDG. Biodistribution results were consistent with the imaging results. The blocking study with excess cold peptide showed significantly reduced tumor uptake, which indicated the specificity of [68Ga]DOTA-VAP targeting MDA-MB-231 tumors in vivo. CONCLUSIONS GRP78-targeted PET imaging with [68Ga]DOTA-VAP provided an effective approach for the noninvasive accurate classification of TNBC from other breast cancer subtypes comparing with [18F]FDG. GRP78 may be a potential target for the diagnosis and treatment of TNBC. For clinical transformation, efforts should be made to overcome deficiencies of [68Ga]DOTA-VAP such as relative high uptake in normal tissues.
Collapse
|
7
|
Berland L, Kim L, Abousaway O, Mines A, Mishra S, Clark L, Hofman P, Rashidian M. Nanobodies for Medical Imaging: About Ready for Prime Time? Biomolecules 2021; 11:637. [PMID: 33925941 PMCID: PMC8146371 DOI: 10.3390/biom11050637] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 12/13/2022] Open
Abstract
Recent advances in medical treatments have been revolutionary in shaping the management and treatment landscape of patients, notably cancer patients. Over the last decade, patients with diverse forms of locally advanced or metastatic cancer, such as melanoma, lung cancers, and many blood-borne malignancies, have seen their life expectancies increasing significantly. Notwithstanding these encouraging results, the present-day struggle with these treatments concerns patients who remain largely unresponsive, as well as those who experience severely toxic side effects. Gaining deeper insight into the cellular and molecular mechanisms underlying these variable responses will bring us closer to developing more effective therapeutics. To assess these mechanisms, non-invasive imaging techniques provide valuable whole-body information with precise targeting. An example of such is immuno-PET (Positron Emission Tomography), which employs radiolabeled antibodies to detect specific molecules of interest. Nanobodies, as the smallest derived antibody fragments, boast ideal characteristics for this purpose and have thus been used extensively in preclinical models and, more recently, in clinical early-stage studies as well. Their merit stems from their high affinity and specificity towards a target, among other factors. Furthermore, their small size (~14 kDa) allows them to easily disperse through the bloodstream and reach tissues in a reliable and uniform manner. In this review, we will discuss the powerful imaging potential of nanobodies, primarily through the lens of imaging malignant tumors but also touching upon their capability to image a broader variety of nonmalignant diseases.
Collapse
Affiliation(s)
- Léa Berland
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (L.B.); (L.K.); (O.A.); (A.M.); (S.M.); (L.C.)
- Université Côte d’Azur, CNRS, INSERM, IRCAN, 06100 Nice, France;
| | - Lauren Kim
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (L.B.); (L.K.); (O.A.); (A.M.); (S.M.); (L.C.)
- Department of Chemistry and Bioengineering, Harvard University, Cambridge, MA 02138, USA
| | - Omar Abousaway
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (L.B.); (L.K.); (O.A.); (A.M.); (S.M.); (L.C.)
| | - Andrea Mines
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (L.B.); (L.K.); (O.A.); (A.M.); (S.M.); (L.C.)
| | - Shruti Mishra
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (L.B.); (L.K.); (O.A.); (A.M.); (S.M.); (L.C.)
| | - Louise Clark
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (L.B.); (L.K.); (O.A.); (A.M.); (S.M.); (L.C.)
| | - Paul Hofman
- Université Côte d’Azur, CNRS, INSERM, IRCAN, 06100 Nice, France;
- Laboratory of Clinical and Experimental Pathology, FHU OncoAge, Nice Center Hospital, 06100 Nice, France
| | - Mohammad Rashidian
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (L.B.); (L.K.); (O.A.); (A.M.); (S.M.); (L.C.)
- Department of Radiology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
8
|
Harmand TJ, Islam A, Pishesha N, Ploegh HL. Nanobodies as in vivo, non-invasive, imaging agents. RSC Chem Biol 2021; 2:685-701. [PMID: 34212147 PMCID: PMC8190910 DOI: 10.1039/d1cb00023c] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/02/2021] [Indexed: 12/12/2022] Open
Abstract
In vivo imaging has become in recent years an incredible tool to study biological events and has found critical applications in diagnostic medicine. Although a lot of efforts and applications have been achieved using monoclonal antibodies, other types of delivery agents are being developed. Among them, VHHs, antigen binding fragments derived from camelid heavy chain-only antibodies, also known as nanobodies, have particularly attracted attention. Indeed, their stability, fast clearance, good tissue penetration, high solubility, simple cloning and recombinant production make them attractive targeting agents for imaging modalities such as PET, SPECT or Infra-Red. In this review, we discuss the pioneering work that has been carried out using VHHs and summarize the recent developments that have been made using nanobodies for in vivo, non-invasive, imaging.
Collapse
Affiliation(s)
- Thibault J Harmand
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School Boston MA USA
| | - Ashraful Islam
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School Boston MA USA
- Department of Clinical Medicine, UiT The Arctic University of Norway Tromso Norway
| | - Novalia Pishesha
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School Boston MA USA
- Society of Fellows, Harvard University Cambridge MA USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard Cambridge MA USA
| | - Hidde L Ploegh
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School Boston MA USA
| |
Collapse
|
9
|
Tu Y, Liu Z, Wang F, Liu P, Tao J, Li C, Han Z, Li Z, Ma Y, Gu Y. AT1R-Specific Ligand Angiotensin II as a Novel SPECT Agent for Hepatocellular Carcinoma Diagnosis. ACS Sens 2020; 5:4072-4080. [PMID: 33232127 DOI: 10.1021/acssensors.0c02180] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Hepatocellular carcinoma (HCC) is characterized by a high mortality and early diagnosis and treatment are critically needed. Ang II type 1 receptor (AT1R) has recently emerged as a potential molecular target for cancer diagnosis and intervention. Here, we labeled angiotensin II (Ang II), an AT1R ligand that is overexpressed in various solid cancers, with the near-infrared fluorescent dye, MPA, and radionuclide technetium-99m, and evaluated its capacity for HCC detection. These analyses were done in vitro using HepG2 (AT1R-positive) and BxPC3 (AT1R-negative) cell lines, and in vivo using a subcutaneous and orthotopic xenograft mouse model by fluorescence and SPECT imaging. Both Ang II-PEG4-MPA- and [99mTc]Tc-HYNIC-PEG4-Ang II-bound AT1R exhibited a high affinity in vitro and [99mTc]Tc-HYNIC-PEG4-Ang II displayed an acceptable level of in vitro stability in rat plasma and whole blood. In vivo imaging revealed excellent specific tumor-targeting in HepG2 mouse xenografts models. In vitro and in vivo competition experiments revealed specific Ang II-PEG4-MPA and [99mTc]Tc-HYNIC-PEG4-Ang II uptake by HepG2 cells and tumors. Altogether, AT1R-positive tumors were successfully detected via fluorescence and SPECT imaging using Ang II-PEG4-MPA and [99mTc]Tc-HYNIC-PEG4-Ang II, respectively. Given their superior targeting capacity, Ang II-PEG4-MPA and [99mTc]Tc-HYNIC-PEG4-Ang II are promising tools for HCC detection and warrant clinical translation.
Collapse
Affiliation(s)
- Yuanbiao Tu
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 24 Tongjia Lane, Gulou District, Nanjing 210009, China
- Jiangzhong Cancer Research Center, Jiangxi University of Traditional Chinese Medicine, No. 1688 Meiling Road, Wanli District, Nanchang 330004, China
| | - Zicun Liu
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| | - Fang Wang
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| | - Peifei Liu
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| | - Ji Tao
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| | - Changsheng Li
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| | - Zhihao Han
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| | - Zhaolun Li
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| | - Yi Ma
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| | - Yueqing Gu
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| |
Collapse
|
10
|
Abstract
Antibodies and antibody fragments have found wide application for therapeutic and diagnostic purposes. Single-domain antibody fragments, also known as 'heavy-chain variable domains' or 'nanobodies', are a recent addition to the toolbox. Discovered some 30 years ago, nanobodies are the smallest antibody-derived fragments that retain antigen-binding properties. Their small size, stability, specificity, affinity and ease of manufacture make them appealing for use as imaging agents in the laboratory and the clinic. With the recent surge in immunotherapeutics and the success of cancer immunotherapy, it is important to be able to image immune responses and cancer biomarkers non-invasively to allocate resources and guide the best possible treatment of patients with cancer. This article reviews recent advances in the application of nanobodies as cancer imaging agents. While much work has been done in preclinical models, first-in-human applications are beginning to show the value of nanobodies as imaging agents.
Collapse
Affiliation(s)
- M. Rashidian
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - H. Ploegh
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
11
|
Wang X, Wang F, Han J, Yang Z, Zhu H, Yang G. Construction and Preclinical Evaluation of a 124/131I-Labeled Radiotracer for the Detection of Mesothelin-Overexpressing Cancer. Mol Pharm 2020; 17:1875-1883. [PMID: 32356995 DOI: 10.1021/acs.molpharmaceut.9b01281] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Mesothelin is a molecular biomarker of many types of solid cancers, which may represent a highly promising new target in the development of cancer-targeted diagnostic agents. A human anti-mesothelin antibody with a low molecular weight, ET210sc, was applied; this antibody has potent affinity and can penetrate tissue quickly and stably without causing immunoreactions. We developed a new 124/131I-labeled radiotracer of ET210sc. The 124/131I-labeled ET210sc radiotracer showed excellent radiochemical quality (with over 99% radiolabeling yield, 0.07 GBq/μmol specific activity) and remarkable stability in phosphate-buffered saline (>95% at 3 days). The radiotracer retained its potent affinity (dissociation constant, Kd = 0.101 nM). The radiotracer specifically bound to mesothelin-positive cells in vitro. Interestingly, the radiotracer exhibited significant positive-to-negative tumor uptake ratios (1.5:1) 3 days postinjection. The estimated absorbed doses of each organ (e.g., 0.704 mGy/MBq for the rectum; 0.341 mGy/MBq for the spleen) met the medical safety standards for further clinical applications. Our findings provide an initial proof of concept for the potential use of 124/131I-labeled ET210sc radiotracers to detect mesothelin-overexpressing cancer. 124I-ET210sc is proposed to be an ideal imaging agent for further clinical applications.
Collapse
Affiliation(s)
- Xudong Wang
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China
| | - Feng Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jintao Han
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China
| | - Zhi Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Hua Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Gen Yang
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China
| |
Collapse
|
12
|
99mTc-A1 as a Novel Imaging Agent Targeting Mesothelin-Expressing Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2019; 11:cancers11101531. [PMID: 31658755 PMCID: PMC6827014 DOI: 10.3390/cancers11101531] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/08/2019] [Accepted: 10/08/2019] [Indexed: 12/24/2022] Open
Abstract
Mesothelin is a membrane-associated protein overexpressed in pancreatic ductal adenocarcinoma (PDAC). Some mesothelin-targeted therapies are in clinical development but the identification of patients eligible for such therapies is still challenging. The objective of this study was to perform the imaging of mesothelin in mice models of PDAC with a technetium-labeled anti-mesothelin single-domain antibody (99mTc-A1). Methods: The Cancer Genomic Atlas (TCGA) database was used to determine the prognostic role of mesothelin in PDAC. 99mTc-A1 was evaluated both in vitro in PDAC cells (SW1990 and AsPC-1) and in vivo in an experimental model of mesothelin-expressing PDAC (AsPC-1) in mice. Results: TCGA analysis showed that PDAC patients with high mesothelin expression had a shorter overall survival (P = 0.00066). The binding of 99mTc-A1 was 2.1-fold greater in high-mesothelin-expressing AsPC-1 cells when compared to moderate-mesothelin-expressing SW1990 cells (p < 0.05). In vivo, the 99mTc-A1 uptake was 3.5-fold higher in AsPC-1-derived tumors as compared to a technetium-labeled irrelevant antibody (99mTc-Ctl) (p < 0.01). Conclusions: 99mTc-A1 accurately allows imaging of mesothelin-expressing experimental PDAC tumors. Our experiments paved the way for the development of a companion test for mesothelin-targeted therapies.
Collapse
|
13
|
Liu Z, Ren F, Zhang H, Yuan Q, Jiang Z, Liu H, Sun Q, Li Z. Boosting often overlooked long wavelength emissions of rare-earth nanoparticles for NIR-II fluorescence imaging of orthotopic glioblastoma. Biomaterials 2019; 219:119364. [PMID: 31352311 DOI: 10.1016/j.biomaterials.2019.119364] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 07/13/2019] [Accepted: 07/14/2019] [Indexed: 01/06/2023]
Abstract
Rare-earth nanoparticles (RE NPs) with narrow long wavelength emissions have been recently investigated for their potential application for fluorescence imaging in the second near-infrared window (NIR-II). Previously these RE NPs have a very limited application in the diagnosis and treatment of deep-seated tumors such as brain tumors, due to their weak fluorescence in the range of 1300-1700 nm. Herein, we report a significant enhancement of more than 10 times regular emission of NaNdF4 nanoparticles at 1340 nm wavelength by coating them with an inert layer of NaLuF4, followed by sensitizing with a near-infrared dye (IR-808). We deliver these highly bright nanoparticles into the brain by using focused ultrasound to temporarily open the blood-brain barrier (BBB), and then detect the orthotopic glioblastoma by fluorescence imaging at 1340 nm. The images obtained from long wavelength fluorescence (i.e. 1340 nm) exhibited better resolution and contrast compared to the short wavelength fluorescence (i.e. 1060 nm). Our study not only provides insights for enhancing often overlooked emissions of rare-earth nanoparticles for NIR-II fluorescence imaging of deep-seated tumors, but also demonstrates great potential of focused ultrasound based technology in delivering nanotheranostic agents.
Collapse
Affiliation(s)
- Zheng Liu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, PR China
| | - Feng Ren
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, PR China
| | - Hao Zhang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, PR China
| | - Qiang Yuan
- The Second Affiliated Hospital of Soochow University, Suzhou, 215004, PR China
| | - Zhilin Jiang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, PR China
| | - Hanghang Liu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, PR China
| | - Qiao Sun
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, PR China
| | - Zhen Li
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, PR China.
| |
Collapse
|
14
|
Debie P, Devoogdt N, Hernot S. Targeted Nanobody-Based Molecular Tracers for Nuclear Imaging and Image-Guided Surgery. Antibodies (Basel) 2019; 8:E12. [PMID: 31544818 PMCID: PMC6640687 DOI: 10.3390/antib8010012] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/29/2018] [Accepted: 01/07/2019] [Indexed: 12/11/2022] Open
Abstract
Molecular imaging is paving the way towards noninvasive detection, staging, and treatment follow-up of diseases such as cancer and inflammation-related conditions. Monoclonal antibodies have long been one of the staples of molecular imaging tracer design, although their long blood circulation and high nonspecific background limits their applicability. Nanobodies, unique antibody-binding fragments derived from camelid heavy-chain antibodies, have excellent properties for molecular imaging as they are able to specifically find their target early after injection, with little to no nonspecific background. Nanobody-based tracers using either nuclear or fluorescent labels have been heavily investigated preclinically and are currently making their way into the clinic. In this review, we will discuss different important factors in nanobody-tracer design, as well as the current state of the art regarding their application for nuclear and fluorescent imaging purposes. Furthermore, we will discuss how nanobodies can also be exploited for molecular therapy applications such as targeted radionuclide therapy and photodynamic therapy.
Collapse
Affiliation(s)
- Pieterjan Debie
- Laboratory for in vivo Cellular and Molecular Imaging, ICMI-BEFY/MIMA, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Nick Devoogdt
- Laboratory for in vivo Cellular and Molecular Imaging, ICMI-BEFY/MIMA, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Sophie Hernot
- Laboratory for in vivo Cellular and Molecular Imaging, ICMI-BEFY/MIMA, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| |
Collapse
|
15
|
Montemagno C, Pagès G. Cholesterol content of serum lipoprotein fractions in children maintained on chronic hemodialysis. Cancers (Basel) 1981; 12:cancers12040821. [PMID: 32235331 PMCID: PMC7226533 DOI: 10.3390/cancers12040821] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/03/2020] [Accepted: 03/05/2020] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the most common malignancy in women throughout the world. Metastatic dissemination to vital organs is the leading cause of breast cancer-related deaths. The treatment of metastases is mainly based on the primary tumor characteristics. However, breast cancer metastases exhibit high heterogeneity leading to different prognosis and therapeutic responses. Getting access to phenotype of metastases would allow better management of patients. The advent of theranostics in nuclear medicine has opened new opportunities for the diagnosis and treatment of cancer patients. The aim of this review is to provide an overview of current knowledge and future directions in nuclear medicine for therapeutic management of metastatic breast cancer patients.
Collapse
Affiliation(s)
- Christopher Montemagno
- Département de Biologie Médicale, Centre Scientifique de Monaco, 98000 Monaco, Monaco;
- Institute for Research on Cancer and Aging of Nice, Centre Antoine Lacassagne, CNRS UMR 7284 and INSERM U1081, University Côte d’Azur, 06200 Nice, France
- Correspondence: ; Tel.: +377-97-77-44-10
| | - Gilles Pagès
- Département de Biologie Médicale, Centre Scientifique de Monaco, 98000 Monaco, Monaco;
- Institute for Research on Cancer and Aging of Nice, Centre Antoine Lacassagne, CNRS UMR 7284 and INSERM U1081, University Côte d’Azur, 06200 Nice, France
| |
Collapse
|