1
|
Pandey S, Kaur G, Rana N, Chopra S, Rather I, Kumar R, Laroiya I, Chadha VD, Satz S, Stabin MG, Mittal BR, Shukla J. Advancing Cancer Theranostics Through Integrin αVβ3-Targeted Peptidomimetic IAC: From Bench to Bedside. Cancer Biother Radiopharm 2024; 39:632-643. [PMID: 38977419 DOI: 10.1089/cbr.2023.0140] [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] [Indexed: 07/10/2024] Open
Abstract
Introduction: The expression of alpha-five beta-three (αVβ3) integrins is upregulated in various malignancies undergoing angiogenesis. The development of integrin antagonists as diagnostic probes makes the αVβ3 integrin a suitable candidate for targeting tumor angiogenesis. The goal of this study was to optimize the radiolabeling and evaluate the potential of conjugated integrin antagonist carbamate (IAC), a peptidomimetic, as a theranostic radiopharmaceutical for targeting tumor angiogenesis. Methodology: Radiolabeling of DOTAGA [2,2',2"-{10-(2,6-dioxotetrahydro-2H-pyran-3-yl)-1,4,7,10-tetraazacyclododecane-1,4,7-triyl} triacetic-acid]-IAC with [68Ga]Ga, [177Lu]Lu, and [225Ac]Ac was optimized. The binding affinity (Kd) of DOTAGA-IAC for the αVβ3 receptor and cancer cell lines was quantified. The biodistribution studies were conducted in healthy Wistar rats. Dosimetry analysis was performed on [177Lu]Lu-DOTAGA-IAC distribution data. A pilot study of [68Ga]Ga-DOTAGA-IAC and [18F]FDG Positron Emission Tomography (PET/CT) imaging was performed in five patients with histopathologically confirmed breast cancer. PET/CT findings were compared between [68Ga]Ga-DOTAGA-IAC and [18F]FDG in these patients. Results: Radiopharmaceuticals were prepared with high radiochemical purity (>99.9%). Kd and Bmax measurements were 15.02 nM and 417 fmol for αVβ3 receptor protein: 115.7 nM and 295.3 fmol for C6 glioma cells. Biodistribution studies in rats suggested the excretion via kidneys and partially through the hepatobiliary route. The effective dose of [177Lu]Lu-DOTAGA-IAC was found to be 0.17 mSv/MBq. The dynamic study in patients revealed the optimal imaging time to be 30-35 mins postadministration. Out of the cohort, [68Ga]Ga-DOTAGA-IAC detected the primary lesions in all five patients with a mean standard uptake value (SUVmax) of 3.94 ± 0.58 compared with [18F]FDG (SUVmax 13.8 ± 6.53). Conclusion: The study demonstrates that DOTAGA-IAC exhibits strong binding to αVβ3 integrin, positioning it as a promising PET agent for assessing primary and metastatic cancers. The outcomes from the pilot study suggest the potential of [68Ga]Ga-DOTAGA-IAC PET/CT in breast carcinoma diagnosis. While recognizing the theranostic potential of DOTAGA-IAC for αVβ3 integrin-expressing tumors, further clinical investigations are warranted to comprehensively assess therapeutic efficacy.
Collapse
Affiliation(s)
- Somit Pandey
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India
| | - Gurvinder Kaur
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India
| | - Nivedita Rana
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India
| | - Sejal Chopra
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India
| | - Imran Rather
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research (P.G.I.M.E.R), Chandigarh, India
| | - Rajender Kumar
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India
| | - Ishita Laroiya
- Department of Surgery, Post Graduate Institute of Medical Education & Research (P.G.I.M.E.R), Chandigarh, India
| | - Vijayta D Chadha
- Center for Nuclear Medicine, Panjab University, Chandigarh, India
| | - Stanley Satz
- Advanced Innovative Partners, Inc., Miami, Florida, USA
| | | | - Bhagwant Rai Mittal
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India
| | - Jaya Shukla
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India
| |
Collapse
|
2
|
Hofman MS, Tran B, Feldman DR, Pokorska-Bocci A, Pichereau S, Wessen J, Haskali MB, Sparks RB, Vlasyuk O, Galetic I. First-in-Human Safety, Imaging, and Dosimetry of a Carbonic Anhydrase IX-Targeting Peptide, [ 68Ga]Ga-DPI-4452, in Patients with Clear Cell Renal Cell Carcinoma. J Nucl Med 2024; 65:jnumed.123.267175. [PMID: 38388517 PMCID: PMC11064824 DOI: 10.2967/jnumed.123.267175] [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: 12/04/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
[68Ga]Ga-DPI-4452, a first-in-class carbonic anhydrase IX-binding radiolabeled peptide, is the imaging agent of a theranostic pair with [177Lu]Lu-DPI-4452, developed for selecting and treating patients with carbonic anhydrase IX-expressing tumors. Here, [68Ga]Ga-DPI-4452 imaging characteristics, dosimetry, pharmacokinetics, and safety were assessed in 3 patients with clear cell renal cell carcinoma. Methods: After [68Ga]Ga-DPI-4452 administration, patients underwent serial full-body PET/CT imaging. Blood and urine were sampled. Safety was monitored for 7 d after injection. Results: Tumor uptake was observed at all time points (15 min to 4 h). Across 36 lesions, the SUVmax at 1 h after administration ranged from 6.8 to 211.6 (mean, 64.6 [SD, 54.8]). The kidneys, liver, and bone marrow demonstrated low activity. [68Ga]Ga-DPI-4452 was rapidly eliminated from blood and urine. No clinically significant toxicity was observed. Conclusion: [68Ga]Ga-DPI-4452 showed exceptional tumor uptake in patients with clear cell renal cell carcinoma, with very high tumor-to-background ratios and no significant adverse events, suggesting potential diagnostic and patient selection applications.
Collapse
Affiliation(s)
- Michael S Hofman
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia;
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Ben Tran
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Darren R Feldman
- Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, New York
| | | | | | | | - Mohammad B Haskali
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | | | - Olena Vlasyuk
- Debiopharm International SA, Lausanne, Switzerland; and
| | - Ivana Galetic
- Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, New York
| |
Collapse
|
3
|
Zhu T, Hsu JC, Guo J, Chen W, Cai W, Wang K. Radionuclide-based theranostics - a promising strategy for lung cancer. Eur J Nucl Med Mol Imaging 2023; 50:2353-2374. [PMID: 36929181 PMCID: PMC10272099 DOI: 10.1007/s00259-023-06174-8] [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: 12/15/2022] [Accepted: 02/25/2023] [Indexed: 03/18/2023]
Abstract
PURPOSE This review aims to provide a comprehensive overview of the latest literature on personalized lung cancer management using different ligands and radionuclide-based tumor-targeting agents. BACKGROUND Lung cancer is the leading cause of cancer-related deaths worldwide. Due to the heterogeneity of lung cancer, advances in precision medicine may enhance the disease management landscape. More recently, theranostics using the same molecule labeled with two different radionuclides for imaging and treatment has emerged as a promising strategy for systemic cancer management. In radionuclide-based theranostics, the target, ligand, and radionuclide should all be carefully considered to achieve an accurate diagnosis and optimal therapeutic effects for lung cancer. METHODS We summarize the latest radiotracers and radioligand therapeutic agents used in diagnosing and treating lung cancer. In addition, we discuss the potential clinical applications and limitations associated with target-dependent radiotracers as well as therapeutic radionuclides. Finally, we provide our views on the perspectives for future development in this field. CONCLUSIONS Radionuclide-based theranostics show great potential in tailored medical care. We expect that this review can provide an understanding of the latest advances in radionuclide therapy for lung cancer and promote the application of radioligand theranostics in personalized medicine.
Collapse
Affiliation(s)
- Tianxing Zhu
- Department of Respiratory Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China
- Lingang Laboratory, Shanghai, 200031, China
| | - Jessica C Hsu
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Jingpei Guo
- Department of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, Guangdong, China
| | - Weiyu Chen
- Department of Respiratory Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China.
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China.
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| | - Kai Wang
- Department of Respiratory Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China.
| |
Collapse
|
4
|
Chen Y, Guo ZN, He RQ, Huang ZG, Luo JY, Tang W, Huang SN, Chen G. How has the field of metastatic breast cancer in bones evolved over the past 22 years? J Bone Oncol 2023; 40:100480. [PMID: 37251089 PMCID: PMC10209145 DOI: 10.1016/j.jbo.2023.100480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/19/2023] [Accepted: 04/08/2023] [Indexed: 05/31/2023] Open
Abstract
Background Although knowledge on metastatic breast cancer in bones (MBCB) has increased rapidly over the past 22 years, a comprehensive and objective bibliometric analysis is still lacking. Materials and methods We used R, VOSviewer, and Citespace software to conduct a bibliometric analysis of 5,497 papers on MBCB from the Web of Science Core Collection (WOSCC) using author, institution, country/region, citation, and keyword indicators. Results A general strong sense of scholarly collaboration was noted in the MBCB field at the author, research institution, and country/region levels. We discovered some outstanding authors and highly productive institutions, but with less collaboration with other academic groups. Unbalanced and uncoordinated developments were observed among countries/regions in the field of MBCB research. We also found that by using various indicators and applying different analysis methods to them, we were able to broadly identify primary clinical practices, relevant clinical experiments, and directions for bioinformatics regarding MBCB, changes over the past 22 years, and current challenges in the field. The development of knowledge on MBCB is progressing greatly; however, MBCB is still incurable. Conclusion This study is the first to use bibliometrics to provide an overall analysis of the scientific output of MBCB studies. Palliative therapies for MBCB are mostly in a mature state. However, research on the molecular mechanisms and immune response to tumors related to the development of treatments to cure MBCB remains relatively immature. Therefore, further research should be undertaken in this area.
Collapse
Affiliation(s)
- Yi Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road Nanning, Guangxi Zhuang Autonomous Region 530021, PR China
| | - Zhen-Ning Guo
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road Nanning, Guangxi Zhuang Autonomous Region 530021, PR China
| | - Rong-Quan He
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road Nanning, Guangxi Zhuang Autonomous Region 530021, PR China
| | - Zhi-Guang Huang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road Nanning, Guangxi Zhuang Autonomous Region 530021, PR China
| | - Jia-Yuan Luo
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road Nanning, Guangxi Zhuang Autonomous Region 530021, PR China
| | - Wei Tang
- Department of Breast Surgery, Guangxi Medical University Cancer Hospital, No. 71 Hedi Rd, Nanning, Guangxi Zhuang Autonomous Region, 530021, PR China
| | - Su-Ning Huang
- Department of Radiotherapy, Guangxi Medical University Cancer Hospital, No. 71 Hedi Rd, Nanning, Guangxi Zhuang Autonomous Region, 530021, PR China
| | - Gang Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road Nanning, Guangxi Zhuang Autonomous Region 530021, PR China
| |
Collapse
|
5
|
Gomari MM, Abkhiz S, Pour TG, Lotfi E, Rostami N, Monfared FN, Ghobari B, Mosavi M, Alipour B, Dokholyan NV. Peptidomimetics in cancer targeting. Mol Med 2022; 28:146. [PMID: 36476230 PMCID: PMC9730693 DOI: 10.1186/s10020-022-00577-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 11/16/2022] [Indexed: 12/12/2022] Open
Abstract
The low efficiency of treatment strategies is one of the main obstacles to developing cancer inhibitors. Up to now, various classes of therapeutics have been developed to inhibit cancer progression. Peptides due to their small size and easy production compared to proteins are highly regarded in designing cancer vaccines and oncogenic pathway inhibitors. Although peptides seem to be a suitable therapeutic option, their short lifespan, instability, and low binding affinity for their target have not been widely applicable against malignant tumors. Given the peptides' disadvantages, a new class of agents called peptidomimetic has been introduced. With advances in physical chemistry and biochemistry, as well as increased knowledge about biomolecule structures, it is now possible to chemically modify peptides to develop efficient peptidomimetics. In recent years, numerous studies have been performed to the evaluation of the effectiveness of peptidomimetics in inhibiting metastasis, angiogenesis, and cancerous cell growth. Here, we offer a comprehensive review of designed peptidomimetics to diagnose and treat cancer.
Collapse
Affiliation(s)
- Mohammad Mahmoudi Gomari
- grid.411746.10000 0004 4911 7066Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shadi Abkhiz
- grid.411746.10000 0004 4911 7066Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Taha Ghantab Pour
- grid.411746.10000 0004 4911 7066Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ehsan Lotfi
- grid.411746.10000 0004 4911 7066Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Neda Rostami
- grid.411425.70000 0004 0417 7516Department of Chemical Engineering, Faculty of Engineering, Arak University, Arak, Iran
| | - Fatemeh Nafe Monfared
- grid.411705.60000 0001 0166 0922Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Ghobari
- grid.412831.d0000 0001 1172 3536Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mona Mosavi
- grid.411746.10000 0004 4911 7066Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Behruz Alipour
- grid.411705.60000 0001 0166 0922Medical Biotechnology Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nikolay V. Dokholyan
- grid.240473.60000 0004 0543 9901Department of Pharmacology, Penn State College of Medicine, Hershey, PA USA ,grid.240473.60000 0004 0543 9901Department of Biochemistry & Molecular Biology, Penn State College of Medicine, Hershey, PA USA
| |
Collapse
|
6
|
Novruzov E, Mori Y, Antke C, Dabir M, Schmitt D, Kratochwil C, Koerber SA, Haberkorn U, Giesel FL. A Role of Non-FDG Tracers in Lung Cancer? Semin Nucl Med 2022; 52:720-733. [PMID: 35803770 DOI: 10.1053/j.semnuclmed.2022.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 11/11/2022]
Abstract
Since the introduction of PET/CT hybrid imaging about two decades ago the landscape of oncological imaging has fundamentally changed, opening a new era of molecular imaging with emphasis on functional characterization of biological processes such as metabolism, cellular proliferation, hypoxia, apoptosis, angiogenesis and immune response. The most commonly assessed functional hallmark of cancer is the increased metabolism in tumor cells due to well-known Warburg effect, because of which FDG has been the most employed radiotracer, the so-called pan-cancer agent, in oncological imaging. However, several limitations such as low specificity and low sensitivity for several histopathological forms of lung cancer as well as high background uptake in the normal tissue of FDG imaging lead to numerous serious pitfalls. This restricts its utilization and diagnostic value in lung cancer imaging, even though this is currently considered to be the method of choice in pulmonary cancer imaging. Accurate initial tumor staging and therapy response monitoring with respect to the TNM criteria plays a crucial role in therapy planning and management in patients with lung cancer. To this end, many efforts have been made for decades to develop novel PET radiopharmaceuticals with innovative approaches that go beyond the assessment of increased glycolytic activity alone. Radiopharmaceuticals targeting DNA synthesis, amino acid metabolism, angiogenesis, or hypoxia have been extensively studied, leading to the emergence of indications for specific clinical questions or as a complementary imaging tool alongside existing conventional or FDG imaging. Nevertheless, despite some initial encouraging results, these tracers couldn't gain a widespread use and acceptance in clinical routine. However, given its mechanism of action and some initial pilot studies regarding lung cancer imaging, FAPI has emerged as a very promising alternative tool that could provide superior or comparable diagnostic performance to FDG imaging in lung cancer entities. Thus, in this review article, we summarized the current PET radiopharmaceuticals, different imaging approaches and discussed the potential benefits and clinical applications of these agents in lung cancer imaging.
Collapse
Affiliation(s)
- Emil Novruzov
- Department of Nuclear Medicine, Medical Faculty, Heinrich-Heine-University, University Hospital Dusseldorf, Dusseldorf, Germany
| | - Yuriko Mori
- Department of Nuclear Medicine, Medical Faculty, Heinrich-Heine-University, University Hospital Dusseldorf, Dusseldorf, Germany
| | - Christina Antke
- Department of Nuclear Medicine, Medical Faculty, Heinrich-Heine-University, University Hospital Dusseldorf, Dusseldorf, Germany
| | - Mardjan Dabir
- Department of Nuclear Medicine, Medical Faculty, Heinrich-Heine-University, University Hospital Dusseldorf, Dusseldorf, Germany
| | - Dominik Schmitt
- Department of Nuclear Medicine, Medical Faculty, Heinrich-Heine-University, University Hospital Dusseldorf, Dusseldorf, Germany
| | - Clemens Kratochwil
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan A Koerber
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Uwe Haberkorn
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Frederik L Giesel
- Department of Nuclear Medicine, Medical Faculty, Heinrich-Heine-University, University Hospital Dusseldorf, Dusseldorf, Germany.
| |
Collapse
|
7
|
van der Heide CD, Dalm SU. Radionuclide imaging and therapy directed towards the tumor microenvironment: a multi-cancer approach for personalized medicine. Eur J Nucl Med Mol Imaging 2022; 49:4616-4641. [PMID: 35788730 PMCID: PMC9606105 DOI: 10.1007/s00259-022-05870-1] [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: 04/12/2022] [Accepted: 06/09/2022] [Indexed: 12/19/2022]
Abstract
Targeted radionuclide theranostics is becoming more and more prominent in clinical oncology. Currently, most nuclear medicine compounds researched for cancer theranostics are directed towards targets expressed in only a small subset of cancer types, limiting clinical applicability. The identification of cancer-specific targets that are (more) universally expressed will allow more cancer patients to benefit from these personalized nuclear medicine–based interventions. A tumor is not merely a collection of cancer cells, it also comprises supporting stromal cells embedded in an altered extracellular matrix (ECM), together forming the tumor microenvironment (TME). Since the TME is less genetically unstable than cancer cells, and TME phenotypes can be shared between cancer types, it offers targets that are more universally expressed. The TME is characterized by the presence of altered processes such as hypoxia, acidity, and increased metabolism. Next to the ECM, the TME consists of cancer-associated fibroblasts (CAFs), macrophages, endothelial cells forming the neo-vasculature, immune cells, and cancer-associated adipocytes (CAAs). Radioligands directed at the altered processes, the ECM, and the cellular components of the TME have been developed and evaluated in preclinical and clinical studies for targeted radionuclide imaging and/or therapy. In this review, we provide an overview of the TME targets and their corresponding radioligands. In addition, we discuss what developments are needed to further explore the TME as a target for radionuclide theranostics, with the hopes of stimulating the development of novel TME radioligands with multi-cancer, or in some cases even pan-cancer, application.
Collapse
Affiliation(s)
| | - Simone U Dalm
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
8
|
Balma M, Liberini V, Racca M, Laudicella R, Bauckneht M, Buschiazzo A, Nicolotti DG, Peano S, Bianchi A, Albano G, Quartuccio N, Abgral R, Morbelli SD, D'Alessandria C, Terreno E, Huellner MW, Papaleo A, Deandreis D. Non-conventional and Investigational PET Radiotracers for Breast Cancer: A Systematic Review. Front Med (Lausanne) 2022; 9:881551. [PMID: 35492341 PMCID: PMC9039137 DOI: 10.3389/fmed.2022.881551] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/22/2022] [Indexed: 02/05/2023] Open
Abstract
Breast cancer is one of the most common malignancies in women, with high morbidity and mortality rates. In breast cancer, the use of novel radiopharmaceuticals in nuclear medicine can improve the accuracy of diagnosis and staging, refine surveillance strategies and accuracy in choosing personalized treatment approaches, including radioligand therapy. Nuclear medicine thus shows great promise for improving the quality of life of breast cancer patients by allowing non-invasive assessment of the diverse and complex biological processes underlying the development of breast cancer and its evolution under therapy. This review aims to describe molecular probes currently in clinical use as well as those under investigation holding great promise for personalized medicine and precision oncology in breast cancer.
Collapse
Affiliation(s)
- Michele Balma
- Nuclear Medicine Department, S. Croce e Carle Hospital, Cuneo, Italy
- *Correspondence: Michele Balma
| | - Virginia Liberini
- Nuclear Medicine Department, S. Croce e Carle Hospital, Cuneo, Italy
- Division of Nuclear Medicine, Department of Medical Science, University of Turin, Turin, Italy
| | - Manuela Racca
- Nuclear Medicine Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Riccardo Laudicella
- Department of Biomedical and Dental Sciences and of Morpho-Functional Imaging, Nuclear Medicine Unit, University of Messina, Messina, Italy
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Nuclear Medicine Unit, Fondazione Istituto G. Giglio, Cefalù, Italy
| | - Matteo Bauckneht
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Health Science (DISSAL), University of Genoa, Genoa, Italy
| | - Ambra Buschiazzo
- Nuclear Medicine Department, S. Croce e Carle Hospital, Cuneo, Italy
| | | | - Simona Peano
- Nuclear Medicine Department, S. Croce e Carle Hospital, Cuneo, Italy
| | - Andrea Bianchi
- Nuclear Medicine Department, S. Croce e Carle Hospital, Cuneo, Italy
| | - Giovanni Albano
- Nuclear Medicine Unit, Fondazione Istituto G. Giglio, Cefalù, Italy
| | - Natale Quartuccio
- Nuclear Medicine Unit, A.R.N.A.S. Civico di Cristina and Benfratelli Hospitals, Palermo, Italy
| | - Ronan Abgral
- Department of Nuclear Medicine, University Hospital of Brest, Brest, France
| | - Silvia Daniela Morbelli
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Health Science (DISSAL), University of Genoa, Genoa, Italy
| | | | - Enzo Terreno
- Department of Molecular Biotechnology and Health Sciences, Molecular & Preclinical Imaging Centers, University of Turin, Turin, Italy
| | - Martin William Huellner
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Alberto Papaleo
- Nuclear Medicine Department, S. Croce e Carle Hospital, Cuneo, Italy
| | - Désirée Deandreis
- Division of Nuclear Medicine, Department of Medical Science, University of Turin, Turin, Italy
| |
Collapse
|
9
|
Vatsa R, Shukla J, Kumar S, Chakraboarty S, Dash A, Singh G, Mittal BR. Effect of Macro-Cyclic Bifunctional Chelators DOTA and NODAGA on Radiolabeling and In Vivo Biodistribution of Ga-68 Cyclic RGD Dimer. Cancer Biother Radiopharm 2019; 34:427-435. [DOI: 10.1089/cbr.2019.2811] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Rakhee Vatsa
- Department of Nuclear Medicine and PET, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Jaya Shukla
- Department of Nuclear Medicine and PET, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Sunil Kumar
- Department of Nuclear Medicine and PET, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | | | - Ashutosh Dash
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Gurpreet Singh
- Department of General Surgery, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Bhagwant Rai Mittal
- Department of Nuclear Medicine and PET, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
10
|
Mukherjee A, Paul M, Mukherjee S. Recent Progress in the Theranostics Application of Nanomedicine in Lung Cancer. Cancers (Basel) 2019; 11:cancers11050597. [PMID: 31035440 PMCID: PMC6562381 DOI: 10.3390/cancers11050597] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 04/27/2019] [Accepted: 04/27/2019] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is one of the leading causes of cancer-related death worldwide. Non-small cell lung cancer (NSCLC) causes around 80% to 90% of deaths. The lack of an early diagnosis and inefficiency in conventional therapies causes poor prognosis and overall survival of lung cancer patients. Recent progress in nanomedicine has encouraged the development of an alternative theranostics strategy using nanotechnology. The interesting physico-chemical properties in the nanoscale have generated immense advantages for nanoparticulate systems for the early detection and active delivery of drugs for a better theranostics strategy for lung cancer. This present review provides a detailed overview of the recent progress in the theranostics application of nanoparticles including liposomes, polymeric, metal and bio-nanoparticles. Further, we summarize the advantages and disadvantages of each approach considering the improvement for the lung cancer theranostics.
Collapse
Affiliation(s)
- Anubhab Mukherjee
- Department of Translational Neurosciences and Neurotherapeutics, John Wayne Cancer Institute, Providence Saint John's Health Center, 2200 Santa Monica Boulevard, Santa Monica, CA 90404, USA.
| | - Manash Paul
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, The University of California, Los Angeles (UCLA) Factor Bldg. 10-240, 621 Charles E. Young Dr., Los Angeles, CA 90095, USA.
| | - Sudip Mukherjee
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77005, USA.
| |
Collapse
|
11
|
Khalid U, Vi C, Henri J, Macdonald J, Eu P, Mandarano G, Shigdar S. Radiolabelled Aptamers for Theranostic Treatment of Cancer. Pharmaceuticals (Basel) 2018; 12:ph12010002. [PMID: 30586898 PMCID: PMC6469178 DOI: 10.3390/ph12010002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 11/30/2018] [Accepted: 12/17/2018] [Indexed: 12/18/2022] Open
Abstract
Cancer has a high incidence and mortality rate worldwide, which continues to grow as millions of people are diagnosed annually. Metastatic disease caused by cancer is largely responsible for the mortality rates, thus early detection of metastatic tumours can improve prognosis. However, a large number of patients will also present with micrometastasis tumours which are often missed, as conventional medical imaging modalities are unable to detect micrometastases due to the lack of specificity and sensitivity. Recent advances in radiochemistry and the development of nucleic acid based targeting molecules, have led to the development of novel agents for use in cancer diagnostics. Monoclonal antibodies may also be used, however, they have inherent issues, such as toxicity, cost, unspecified binding and their clinical use can be controversial. Aptamers are a class of single-stranded RNA or DNA ligands with high specificity, binding affinity and selectivity for a target, which makes them promising for molecular biomarker imaging. Aptamers are presented as being a superior choice over antibodies because of high binding affinity and pH stability, amongst other factors. A number of aptamers directed to cancer cell markers (breast, lung, colon, glioblastoma, melanoma) have been radiolabelled and characterised to date. Further work is ongoing to develop these for clinical applications.
Collapse
Affiliation(s)
- Umair Khalid
- School of Medicine Deakin University, Geelong, Victoria 3128, Australia.
| | - Chris Vi
- School of Medicine Deakin University, Geelong, Victoria 3128, Australia.
| | - Justin Henri
- School of Medicine Deakin University, Geelong, Victoria 3128, Australia.
| | - Joanna Macdonald
- School of Medicine Deakin University, Geelong, Victoria 3128, Australia.
| | - Peter Eu
- School of Medicine Deakin University, Geelong, Victoria 3128, Australia.
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia.
| | - Giovanni Mandarano
- School of Medicine Deakin University, Geelong, Victoria 3128, Australia.
| | - Sarah Shigdar
- School of Medicine Deakin University, Geelong, Victoria 3128, Australia.
- Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria 3128, Australia.
| |
Collapse
|
12
|
Buckle T, van Willigen DM, Spa SJ, Hensbergen AW, van der Wal S, de Korne CM, Welling MM, van der Poel HG, Hardwick JCH, van Leeuwen FWB. Tracers for Fluorescence-Guided Surgery: How Elongation of the Polymethine Chain in Cyanine Dyes Alters the Pharmacokinetics of a Dual-Modality c[RGDyK] Tracer. J Nucl Med 2018; 59:986-992. [PMID: 29449447 DOI: 10.2967/jnumed.117.205575] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 01/12/2018] [Indexed: 01/07/2023] Open
Abstract
The potential of receptor-mediated fluorescence-based image-guided surgery tracers is generally linked to the near-infrared emission profile and good-manufacturing-production availability of fluorescent dyes. Surprisingly, little is known about the critical interaction between the structural composition of the dyes and the pharmacokinetics of the tracers. In this study, a dual-modality tracer design was used to systematically and quantitatively evaluate the influence of elongation of the polymethine chain in a fluorescent cyanine dye on the imaging potential of a targeted tracer. Methods: As a model system, the integrin marker αvβ3 was targeted using arginylglycylaspartisc acid [RGD]-based vectors functionalized with a 111In-diethylenetriaminepentaacetic acid (DTPA) chelate and a fluorescent dye: (Cy3-(SO3)methyl-COOH [emission wavelength (λem), 580 nm], Cy5-(SO3)methyl-COOH [λem, 680 nm], or Cy7-(SO3)methyl-COOH [λem, 780 nm]). Tracers were analyzed for differences in photophysical properties, serum protein binding, chemical or optical stability, and signal penetration through tissue. Receptor affinities were evaluated using saturation and competition experiments. In vivo biodistribution (SPECT imaging and percentage injected dose per gram of tissue) was assessed in tumor-bearing mice and complemented with in vivo and ex vivo fluorescence images obtained using a clinical-grade multispectral fluorescence laparoscope. Results: Two carbon-atom-step variations in the polymethine chain of the fluorescent cyanine dyes were shown to significantly influence the chemical and photophysical characteristics (e.g., stability, brightness, and tissue penetration) of the hybrid RGD tracers. DTPA-Cy5-(SO3)methyl-COOH-c[RGDyK] structurally outperformed its Cy3 and Cy7 derivatives. Radioactivity-based evaluation of in vivo tracer pharmacokinetics yielded the lowest nonspecific uptake and highest tumor-to-background ratio for DTPA-Cy5-(SO3)methyl-COOH-c[RGDyK] (13.2 ± 1.7), with the Cy3 and Cy7 analogs trailing at respective tumor-to-background ratios of 5.7 ± 0.7 and 4.7 ± 0.7. Fluorescence-based assessment of tumor visibility revealed a similar trend. Conclusion: These findings underline that variations in the polymethine chain lengths of cyanine dyes have a profound influence on the photophysical properties, stability, and in vivo targeting capabilities of fluorescent imaging tracers. In a direct comparison, the intermediate-length dye (Cy5) yielded a superior c[RGDyK] tracer, compared with the shorter (Cy3) and longer (Cy7) analogs.
Collapse
Affiliation(s)
- Tessa Buckle
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.,Division of Molecular Pathology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Danny M van Willigen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Silvia J Spa
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Albertus W Hensbergen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Steffen van der Wal
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Clarize M de Korne
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mick M Welling
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Henk G van der Poel
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands; and
| | - James C H Hardwick
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Fijs W B van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands .,Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands; and
| |
Collapse
|
13
|
Prospective of 68Ga Radionuclide Contribution to the Development of Imaging Agents for Infection and Inflammation. CONTRAST MEDIA & MOLECULAR IMAGING 2018. [PMID: 29531507 PMCID: PMC5817300 DOI: 10.1155/2018/9713691] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
During the last decade, the utilization of 68Ga for the development of imaging agents has increased considerably with the leading position in the oncology. The imaging of infection and inflammation is lagging despite strong unmet medical needs. This review presents the potential routes for the development of 68Ga-based agents for the imaging and quantification of infection and inflammation in various diseases and connection of the diagnosis to the treatment for the individualized patient management.
Collapse
|
14
|
αvβ3 and α5β1 integrin-specific ligands: From tumor angiogenesis inhibitors to vascularization promoters in regenerative medicine? Biotechnol Adv 2017; 36:208-227. [PMID: 29155160 DOI: 10.1016/j.biotechadv.2017.11.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/07/2017] [Accepted: 11/13/2017] [Indexed: 12/30/2022]
Abstract
Integrins are cell adhesion receptors predominantly important during normal and tumor angiogenesis. A sequence present on several extracellular matrix proteins composed of Arg-Gly-Asp (RGD) has attracted attention due to its role in cell adhesion mediated by integrins. The development of ligands that can bind to integrins involved in tumor angiogenesis and brake disease progression has resulted in new investigational drug entities reaching the clinical trial phase in humans. The use of integrin-specific ligands can be useful for the vascularization of regenerative medicine constructs, which remains a major limitation for translation into clinical practice. In order to enhance vascularization, immobilization of integrin-specific RGD peptidomimetics within constructs is a recommended approach, due to their high specificity and selectivity towards certain desired integrins. This review endeavours to address the potential of peptidomimetic-coated biomaterials as vascular network promoters for regenerative medicine purposes. Clinical studies involving molecules tracking active integrins in cancer angiogenesis and reasons for their failure are also addressed.
Collapse
|
15
|
Arrieta O, Garcia-Perez FO, Michel-Tello D, Ramírez-Tirado LA, Pitalua-Cortes Q, Cruz-Rico G, Macedo-Pérez EO, Cardona AF, Garza-Salazar JDL. Response Assessment of 68Ga-DOTA-E-[c(RGDfK)] 2 PET/CT in Lung Adenocarcinoma Patients Treated with Nintedanib Plus Docetaxel. J Nucl Med 2017; 59:403-409. [PMID: 28818987 DOI: 10.2967/jnumed.117.192393] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 07/13/2017] [Indexed: 12/22/2022] Open
Abstract
Nintedanib is an oral angiokinase inhibitor used as second-line treatment for non-small cell lung cancer. New radiotracers, such as 68Ga-DOTA-E-[c(RGDfK)]2, that target αvβ3 integrin might have an impact as a noninvasive method for assessing angiogenesis inhibitors. Methods: From July 2011 through October 2015, 38 patients received second-line nintedanib plus docetaxel. All patients underwent PET/CT with 68Ga-DOTA-E-[c(RGDfK)]2 radiotracer and blood-sample tests to quantify angiogenesis factors (fibroblast growth factor, vascular endothelial growth factor, and platelet-derived growth factor AB) before and after completing 2 therapy cycles. Results: Of the 38 patients, 31 had available baseline and follow-up PET/CT. Baseline lung tumor volume addressed with 68Ga-DOTA-E-[c(RGDfK)]2 PET/CT correlated with serum vascular endothelial growth factor levels, whereas baseline lung/liver SUVmax index correlated with platelet-derived growth factor AB. After treatment, the overall response rate and disease control rate were 7.9% and 47.3%, respectively. A greater decrease in lung tumor volume (-37.2% vs. -27.6%) was associated with a better disease control rate in patients (P = 0.005). Median progression-free survival was 3.7 mo. Nonsmokers and patients with a higher baseline lung tumor volume were more likely to have a higher progression-free survival (6.4 vs. 3.74 [P = 0.023] and 6.4 vs. 2.1 [P = 0.003], respectively). Overall survival was not reached. Patients with a greater decrease in lung SUVmax (not reached vs. 7.1 mo; P = 0.016) and a greater decrease in the lung/spleen SUVmax index (not reached vs. 7.1; P = 0.043) were more likely to have a longer overall survival. Conclusion:68Ga-DOTA-E-[c(RGDfK)]2 PET/CT is a potentially useful tool for assessing responses to angiogenesis inhibitors. Further analysis and novel studies are warranted to identify patients who might benefit from this therapy.
Collapse
Affiliation(s)
- Oscar Arrieta
- Thoracic Oncology Unit, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Francisco O Garcia-Perez
- Department of Nuclear Medicine and Molecular Imagenology, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - David Michel-Tello
- Thoracic Oncology Unit, Instituto Nacional de Cancerología, Mexico City, Mexico
| | | | - Quetzali Pitalua-Cortes
- Department of Nuclear Medicine and Molecular Imagenology, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Graciela Cruz-Rico
- Laboratory of Personalized Medicine, Instituto Nacional de Cancerología, Mexico City, Mexico
| | | | - Andrés F Cardona
- Clinical and Translational Oncology Group, Clínica del Country, Bogotá, Colombia; and.,Foundation for Clinical and Applied Cancer Research, Bogotá, Colombia
| | | |
Collapse
|
16
|
Abstract
Somatostatin receptor PET/CT using (68)Ga-labeled somatostatin analogs, is a mainstay for the evaluation of the somatostatin receptor status in neuroendocrine neoplasms. In addition, the assessment of glucose metabolism by (18)F-FDG PET/CT at diagnosis can overcome probable shortcomings of histopathologic grading. This offers a systematic theranostic approach for the management of neuroendocrine neoplasms, that is, patient selection for the appropriate treatment-surgery, somatostatin analogs, peptide receptor radionuclide therapy, targeted therapies like everolimus and sunitinib, or chemotherapy-and also for therapy response monitoring. Novel targets, for example, the chemokine receptor CXCR4 in higher-grade tumors and glucagon like peptide-1 receptor in insulinomas, appear promising for imaging. Scandium-44 and Copper-64, especially on account of their longer half-life (for pretherapeutic dosimetry) and cyclotron production (which favors mass production), might be the potential alternatives to (68)Ga for PET/CT imaging. The future of molecular imaging lies in Radiomics, that is, qualitative and quantitative characterization of tumor phenotypes in correlation with tumor genomics and proteomics, for a personalized cancer management.
Collapse
Affiliation(s)
- Harshad R Kulkarni
- THERANOSTICS Center for Molecular Radiotherapy and Molecular Imaging, ENETS Center of Excellence, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Aviral Singh
- THERANOSTICS Center for Molecular Radiotherapy and Molecular Imaging, ENETS Center of Excellence, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Richard P Baum
- THERANOSTICS Center for Molecular Radiotherapy and Molecular Imaging, ENETS Center of Excellence, Zentralklinik Bad Berka, Bad Berka, Germany.
| |
Collapse
|
17
|
Bubenshchikov VB, Maruk AY, Bruskin AB, Kodina GE. Preparation and properties of 68Ga complexes with RGD peptide derivatives. RADIOCHEMISTRY 2016. [DOI: 10.1134/s1066362216050106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
18
|
Kenny L. The Use of Novel PET Tracers to Image Breast Cancer Biologic Processes Such as Proliferation, DNA Damage and Repair, and Angiogenesis. J Nucl Med 2016; 57 Suppl 1:89S-95S. [PMID: 26834108 DOI: 10.2967/jnumed.115.157958] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The balance between proliferation and cell death is pivotal to breast tumor growth. Because of a combination of environmental and genetic factors leading to activation of oncogenes or inactivation of tumor suppressor genes, these processes become deregulated in cancer. PET imaging of proliferation, angiogenesis, and DNA damage and repair offers the opportunity to monitor therapeutic efficacy to detect changes in tumor biology that may precede physical size reduction and simultaneously allows the study of intratumoral and intertumoral heterogeneity.This review examines recent developments in breast cancer imaging using novel probes. The probes discussed here are not licensed for routine use and are at various stages of development ranging from preclinical development (e.g., the DNA repair marker γH2AX) to clinical validation in larger studies (such as the proliferation probe 3'-deoxy-3'-(18)F-fluorothymidine [(18)F-FLT]). In breast cancer, most studies have focused on proliferation imaging mainly based on (18)F-labeled thymidine analogs. Initial studies have been promising; however, the results of larger validation studies are necessary before being incorporated into routine clinical use. Although there are distinct advantages in using process-specific probes, properties such as metabolism need careful consideration, because high background uptake in the liver due to glucuronidation in the case of (18)F-FLT may limit utility for imaging of liver metastases.Targeting angiogenesis has had some success in tumors such as renal cell carcinoma; however, angiogenesis inhibitors have not been particularly successful in the clinical treatment of breast cancer. This could be potentially attributed to patient selection due to the lack of validated predictive and responsive biomarkers; the quest for a successful noninvasive biomarker for angiogenesis could solve this challenge. Finally, we look at cell death including apoptosis and DNA damage and repair probes, the most well-studied example being (18)F-annexin V; more recently, probes that target caspase endoproteases have been developed and are undergoing early clinical validation studies.Further clinical studies including analysis of test-retest variability are essential to determine sensitivity and future utility of these probes in breast cancer.
Collapse
Affiliation(s)
- Laura Kenny
- Department of Surgery and Cancer, Comprehensive Cancer Imaging Center, Imperial College London, London, United Kingdom
| |
Collapse
|
19
|
Radiolabeling of DOTA-like conjugated peptides with generator-produced (68)Ga and using NaCl-based cationic elution method. Nat Protoc 2016; 11:1057-66. [PMID: 27172166 PMCID: PMC5506837 DOI: 10.1038/nprot.2016.060] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Gallium-68 (68Ga) is a generator-produced radionuclide with a short half-life (t½ = 68 min) that is particularly well suited for molecular imaging by positron emission tomography (PET). Methods have been developed to synthesize 68Ga-labeled imaging agents possessing certain drawbacks, such as longer synthesis time because of a required final purification step, the use of organic solvents or concentrated hydrochloric acid (HCl). In our manuscript, we provide a detailed protocol for the use of an advantageous sodium chloride (NaCl)-based method for radiolabeling of chelator-modified peptides for molecular imaging. By working in a lead-shielded hot-cell system, 68Ga3+ of the generator eluate is trapped on a cation exchanger cartridge (100 mg, ∼8 mm long and 5 mm diameter) and then eluted with acidified 5 M NaCl solution directly into a sodium acetate-buffered solution containing a DOTA (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid) or DOTA-like chelator-modified peptide. The main advantages of this procedure are the high efficiency and the absence of organic solvents. It can be applied to a variety of peptides, which are stable in 1 M NaCl solution at a pH value of 3–4 during reaction. After labeling, neutralization, sterile filtration and quality control (instant thin-layer chromatography (iTLC), HPLC and pH), the radiopharmaceutical can be directly administered to patients, without determination of organic solvents, which reduces the overall synthesis-to-release time. This procedure has been adapted easily to automated synthesis modules, which leads to a rapid preparation of 68Ga radiopharmaceuticals (12–16 min).
Collapse
|