1
|
Howell HJ, McGale JP, Choucair A, Shirini D, Aide N, Postow MA, Wang L, Tordjman M, Lopci E, Lecler A, Champiat S, Chen DL, Deandreis D, Dercle L. Artificial Intelligence for Drug Discovery: An Update and Future Prospects. Semin Nucl Med 2025; 55:406-422. [PMID: 39966029 DOI: 10.1053/j.semnuclmed.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/20/2025]
Abstract
Artificial intelligence (AI) has become a pivotal tool for medical image analysis, significantly enhancing drug discovery through improved diagnostics, staging, prognostication, and response assessment. At a high level, AI-driven image analysis enables the quantification and synthesis of previously qualitative imaging characteristics, facilitating the identification of novel disease-specific biomarkers, patient risk stratification, prognostication, and adverse event prediction. In addition, AI can assist in response assessment by capturing changes in imaging "phenotype" over time, allowing for optimized treatment plans based on real-time analysis. Integrating this emerging technology into drug discovery pipelines has the potential to accelerate the identification and development of new pharmaceuticals by assisting in target identification and patient selection, as well as reducing the incidence, and therefore cost, of failed trials through high-throughput, reproducible, and data-driven insights. Continued progress in AI applications will shape the future of medical imaging, ultimately fostering more efficient, accurate, and tailored drug discovery processes. Herein, we offer a comprehensive overview of how AI enhances medical imaging to inform drug development and therapeutic strategies.
Collapse
Affiliation(s)
- Harrison J Howell
- Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Jeremy P McGale
- Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | | | - Dorsa Shirini
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nicolas Aide
- Centre Havrais d'Imagerie Nucléaire, Octeville, France
| | - Michael A Postow
- Department of Medicine, Memorial Sloan Kettering and Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Lucy Wang
- School of Medicine, New York Medical College, Valhalla, NY
| | - Mickael Tordjman
- Department of Radiology, Biomedical Engineering & Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Egesta Lopci
- Nuclear Medicine Unit, IRCCS-Humanitas Research Hospital, Rozzano, Italy
| | - Augustin Lecler
- Department of Neuroradiology, Foundation Adolphe de Rothschild Hospital, Université Paris Cité, Paris, France
| | - Stéphane Champiat
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Delphine L Chen
- Department of Radiology, University of Washington, Seattle, WA
| | | | - Laurent Dercle
- Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY.
| |
Collapse
|
2
|
Tao W, Zhang J, Meng X, Han X, Wang Q, Lin Y, Cheng L, Liu M, Da D, Zhang H, Fan J, Zhang L, Liu S, Li S, Gao F, Ren Y. Development and clinical evaluation of [ 68Ga]Ga-NODAGA-ADAPT6 as a novel HER2-targeted PET radiotracer for breast cancer imaging and treatment monitoring. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07286-z. [PMID: 40257612 DOI: 10.1007/s00259-025-07286-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: 02/08/2025] [Accepted: 04/10/2025] [Indexed: 04/22/2025]
Abstract
PURPOSE Accurate assessment of human epidermal growth factor receptor type 2 (HER2) expression is crucial for diagnosis, treatment planning, and monitoring of breast cancer patients. A 68Ga-labeled tracer based on the albumin-binding domain-derived affinity protein 6 (ADAPT6) was developed to evaluate HER2 expression in breast cancer. METHODS The gene encoding ADAPT6 was modified with N-terminal (GHEHEHEDANS) and C-terminal (GSSC) extensions to enhance its functionality. The precursor was synthesized, purified, and characterized, followed by radiolabeling with 68Ga to produce [68Ga]Ga-NODAGA-ADAPT6. In vivo metabolism and biodistribution studies were performed in HCC1954 (HER2-positive) and MDA-MB-468 (HER2-negative) tumor-bearing mice. Additionally, with ethical approval and informed consent, 22 breast cancer patients underwent [68Ga]Ga-NODAGA-ADAPT6 PET imaging to assess HER2 expression in primary and metastatic lesions. RESULTS The tracer was prepared with a radiochemical purity exceeding 99% and demonstrated high stability in vivo. Micro-PET/CT imaging revealed significant accumulation of the radiotracer in HCC1954 tumors, which was markedly reduced after HER2 blockade with trastuzumab. In contrast, MDA-MB-468 tumors showed minimal uptake. In the clinical study, [68Ga]Ga-NODAGA-ADAPT6 PET images displayed varying levels of radiotracer uptake in primary and metastatic lesions, which correlated well with the HER2 expression status determined by pathological analysis. CONCLUSION [68Ga]Ga-NODAGA-ADAPT6 exhibited excellent pharmacokinetic properties and high specificity for HER2-expressing lesions in PET imaging. These findings highlight its potential as a promising tool for distinguishing different levels of HER2 expression in breast cancer, aiding in personalized treatment strategies.
Collapse
Affiliation(s)
- Weijing Tao
- Department of Nuclear Medicine, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huai'an, 223300, Jiangsu, China.
| | - Jinglin Zhang
- Key Laboratory for Experimental Teratology of the Ministry of Education and Research Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Xin Meng
- Department of Breast and Thyroid Surgery, The Huai'an Clinical College of Xuzhou Medical University, Huai'an, 223300, Jiangsu, China
| | - Xuedong Han
- Department of Breast and Thyroid Surgery, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huai'an, Jiangsu, 223300, China
| | - Qiuhu Wang
- Department of Nuclear Medicine, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huai'an, 223300, Jiangsu, China
| | - Yixiang Lin
- Key Laboratory for Experimental Teratology of the Ministry of Education and Research Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Luyi Cheng
- Department of Nuclear Medicine, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huai'an, 223300, Jiangsu, China
| | - Minmin Liu
- Department of Breast and Thyroid Surgery, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huai'an, Jiangsu, 223300, China
| | - Dongzhu Da
- Department of Breast and Thyroid Surgery, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huai'an, Jiangsu, 223300, China
| | - Huai Zhang
- Department of Nuclear Medicine, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huai'an, 223300, Jiangsu, China
| | - Junfu Fan
- Department of Nuclear Medicine, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huai'an, 223300, Jiangsu, China
| | - Lianmei Zhang
- Department of Pathology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huai'an, 223300, Jiangsu, China
| | - Shuangyue Liu
- Department of Breast and Thyroid Surgery, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huai'an, Jiangsu, 223300, China
| | - Shuo Li
- Department of Breast and Thyroid Surgery, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huai'an, Jiangsu, 223300, China
| | - Feng Gao
- Key Laboratory for Experimental Teratology of the Ministry of Education and Research Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, Shandong, China.
| | - Yi Ren
- Department of Breast and Thyroid Surgery, The Huai'an Clinical College of Xuzhou Medical University, Huai'an, 223300, Jiangsu, China.
- Department of Breast and Thyroid Surgery, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huai'an, Jiangsu, 223300, China.
| |
Collapse
|
3
|
Géraud A, Gougis P, de Nonneville A, Beaufils M, Bertucci F, Billon E, Brisou G, Gravis G, Greillier L, Guerin M, Mezni E, Mitry E, Noel R, Pignon J, Sabatier R, Seguin L, Spano JP, Vicier C, Viret F, Goncalves A, Ciccolini J. Pharmacology and pharmacokinetics of antibody-drug conjugates, where do we stand? Cancer Treat Rev 2025; 135:102922. [PMID: 40157134 DOI: 10.1016/j.ctrv.2025.102922] [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: 01/24/2025] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 04/01/2025]
Abstract
Antibody-drug conjugates (ADCs) are a rising therapeutic class in oncology and hematology, with eleven drugs approved by the US Food and Drug Administration as of January 2025. These "magic bullets" have a complex structure, including a monoclonal antibody, a linker, attachment sites, and a payload usually disrupting microtubules, targeting DNA, or inhibiting topoisomerase 1. By targeting specific tumor antigens, they are expected to be exquisitely effective in releasing "supertoxic" payloads inside tumor cells after intracellular trafficking. Additionally, they may exert a bystander effect, wherein the released payloads act on neighboring cells, amplifying their therapeutic impact regardless of target expression. ADCs have been game-changing drugs to treat tumors with once dismal prognoses or with previously considered unactionable targets, such as HER2-low or triple-negative breast cancer. To what extent there is room for personalized medicine to improve the toxicity/efficacy ratio remains unknown. However, there are inherent issues related to the complexity of the pharmacokinetics of ADCs and their assessments: efficacy or toxicity may be influenced by the clearance of the intact ADC, the circulating payload, or the payload-linker complex. Deciphering these multifaceted exposure-outcomes relationships for both efficacy and safety endpoints, is critical for advancing precision medicine and enabling personalized dosing strategies. To improve future developments and broaden their therapeutic scope, several strategies can be developed, including developing adequate combinations with other treatment classes (cytotoxic agents, immune-checkpoint inhibitors, oral molecular-targeted therapies). In this review, we will discuss the PK/PD aspects of ADCs and their dosing to improve their use in current and future indications.
Collapse
Affiliation(s)
- Arthur Géraud
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France; COMPO Team, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm U1068, Aix Marseille University, 13009 Marseille, France.
| | - Paul Gougis
- Department of Medical Oncology, Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris (AP-HP), 75013 Paris, France; Sorbonne Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Assistance Publique - Hôpitaux de Paris (AP-HP), Clinical Investigation Center (CIC-1901), Department of Pharmacology, Pitié-Salpêtrière Hospital, Paris, France; Residual Tumor & Response to Treatment Laboratory, RT2Lab, INSERM, U932 Immunity and Cancer, Institut Curie, France
| | - Alexandre de Nonneville
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Mathilde Beaufils
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - François Bertucci
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Emilien Billon
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Gabriel Brisou
- Department of Hematology, Institut Paoli-Calmettes, CRCM, Aix-Marseille University,13009 Marseille, France
| | - Gwenaelle Gravis
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Laurent Greillier
- COMPO Team, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm U1068, Aix Marseille University, 13009 Marseille, France; Department of Multidisciplinary Oncology and Therapeutic Innovations, Assistance Publique-Hôpitaux de Marseille (AP-HM), Aix Marseille University (AMU), 13015 Marseille, France
| | - Mathilde Guerin
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Essia Mezni
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Emmanuel Mitry
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Robin Noel
- Department of Hematology, Institut Paoli-Calmettes, CRCM, Aix-Marseille University,13009 Marseille, France
| | - Joséphine Pignon
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Renaud Sabatier
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Lorène Seguin
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Jean-Philippe Spano
- Department of Medical Oncology, Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris (AP-HP), 75013 Paris, France
| | - Cécile Vicier
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Frederic Viret
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Anthony Goncalves
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Joseph Ciccolini
- COMPO Team, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm U1068, Aix Marseille University, 13009 Marseille, France; Biogenopole, La timone University Hospital of Marseille, Assistance Publique-Hôpitaux de Marseille (AP-HM), 13005 Marseille, France
| |
Collapse
|
4
|
Wang K, Lin X, Lv X, Xie M, Wu J, Wu JJ, Luo Y. Nanozyme-based aptasensors for the detection of tumor biomarkers. J Biol Eng 2025; 19:13. [PMID: 39920818 PMCID: PMC11806818 DOI: 10.1186/s13036-025-00485-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025] Open
Abstract
A nanozyme-based aptasensor combines the unique properties of nanozymes with the specificity of aptamers for the detection of various biomolecules. Nanozymes are nanomaterials that possess enzyme-like properties, demonstrating substantial potential for enhancing the sensing capabilities of biosensors. In recent years, the incorporation of nanozymes into biosensors has opened new avenues for the detection of tumor biomarkers. The unique attributes of nanozymes and aptamers lead to biosensors characterized by high sensitivity, specificity, reproducibility and accuracy in analytical performance. This article reviews the research progress of nanozyme-based aptasensors in tumor biomarker detection over the past decade. We categorize these sensors based on their sensing modes and target types, and examine the properties and applications of the nanozymes employed in these devices, providing a thorough discussion of the strengths and weaknesses associated with each sensor type. Finally, the review highlights the strengths and challenges associated with nanozyme-based biosensors and envisions future developments and applications in this field. The objective is to provide insights for improving biosensor performance in tumor biomarker detection, thereby contributing to advancements in precision cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Ke Wang
- Key Laboratory of Optoelectronic Technology and Systems of Ministry of Education of China, Chongqing University, Chongqing, 400044, China
| | - Xiaogang Lin
- Key Laboratory of Optoelectronic Technology and Systems of Ministry of Education of China, Chongqing University, Chongqing, 400044, China.
| | - Xiao Lv
- Key Laboratory of Optoelectronic Technology and Systems of Ministry of Education of China, Chongqing University, Chongqing, 400044, China
| | - Mingna Xie
- Key Laboratory of Optoelectronic Technology and Systems of Ministry of Education of China, Chongqing University, Chongqing, 400044, China
| | - Jinyu Wu
- Key Laboratory of Optoelectronic Technology and Systems of Ministry of Education of China, Chongqing University, Chongqing, 400044, China
| | - Jie Jayne Wu
- Department of Electrical Engineering and Computer Science, The University of Tennessee, Knoxville, TN, 37996, USA.
| | - Yang Luo
- Chongqing General Hospital, Chongqing University, Chongqing, 401147, China
| |
Collapse
|
5
|
Cook GJR, Alberts IL, Wagner T, Fischer BM, Nazir MS, Lilburn D. The impact of long axial field of view (LAFOV) PET on oncologic imaging. Eur J Radiol 2025; 183:111873. [PMID: 39647272 PMCID: PMC11904125 DOI: 10.1016/j.ejrad.2024.111873] [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: 08/30/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/10/2024]
Abstract
The development of long axial field of view (LAFOV) positron emission tomography coupled with computed tomography (PET/CT) scanners might be considered the biggest step forward in PET imaging since it became a mainstream clinical modality. Despite increased capital and maintenance costs and data storage requirements, the improvement in image quality, significantly faster acquisition times and lower radiopharmaceutical administered activities, allow a high quality and more efficient clinical service. This step change in technology overcomes some of the limitations of standard short axial field of view scanners. It allows simultaneous imaging of all body systems, and with the ability to obtain high temporal resolution data, it increases potential research applications, particularly in multisystem disease or for dosimetry measurements of novel radiopharmaceuticals. The improvements in sensitivity and signal-to-noise facilitates the use of tracers with long half-lives and low administered activity (e.g. [89Zr]-labelled monoclonal antibodies) or very short half-lives (e.g. [82Rb]), opening up applications that hitherto have been challenging. It is early in the evolution of LAFOV PET/CT and the advantages these systems offer have still to be fully realised in providing additional impact in clinical practice. In this article we describe the potential advantages of LAFOV PET technology and some of the clinical and research applications where it has been applied as well as some of the future developments that may enhance the modality further.
Collapse
Affiliation(s)
- Gary J R Cook
- King's College London & Guy's and St Thomas' PET Centre, School of Biomedical Engineering and Imaging Sciences, King's College, London SE1 7EH, UK.
| | - Ian L Alberts
- Molecular Imaging and Therapy, BC Cancer Agency, Vancouver, BC, Canada; Department of Radiology, University of British Columbia, Vancouver, BC, Canada.
| | - Thomas Wagner
- Department of Nuclear Medicine, Royal Free London NHS Trust, London NW3 1TX, UK.
| | - B Malene Fischer
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital-Rigshospitalet, 2100 Copenhagen, Denmark.
| | - Muhummad Sohaib Nazir
- Department of Cardiovascular Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK; Cardio-Oncology Centre of Excellence, Royal Brompton Hospital, Guy's and St Thomas' NHS Foundation Trust, London, UK.
| | - David Lilburn
- King's College London & Guy's and St Thomas' PET Centre, School of Biomedical Engineering and Imaging Sciences, King's College, London SE1 7EH, UK.
| |
Collapse
|
6
|
Movahed F, Navaei O, Taghlidi S, Nurzadeh M, Gharaati ME, Rabiei M. Radiolabeled HER2-targeted molecular probes in breast cancer imaging: current knowledge and future prospective. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03691-7. [PMID: 39751821 DOI: 10.1007/s00210-024-03691-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 11/27/2024] [Indexed: 01/04/2025]
Abstract
Breast cancer is the most frequent non-dermatologic malignancy in women. Breast cancer is characterized by the expression of the human epidermal growth factor receptor type 2 (HER2), and the presence or lack of estrogen receptor (ER) and progesterone receptor (PR) expression. HER2 overexpression is reported in about 20 to 25% of breast cancer patients, which is usually linked to cancer progression, metastases, and poor survival. Immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) are the gold standards for determining HER2 status, even though IHC has largely focused on quantifying HER2+ status versus "other" HER2 status (including variants with low or no expression). Recent findings regarding the beneficial therapeutic effects of anti-HER2 monoclonal antibodies (mAb) in HER2low metastatic patients lead to changes in the classic definition of advanced breast cancer, and methods for precise assessment of HER2 status are being developed. As a result, various radiolabeled HER-targeted mAbs and antibody fragments have been designed to avoid repeated biopsies with potential bias due to tumor heterogeneity, including single-chain variable fragment (scFv), F(ab')2, affibody, and nanobody. These small targeting radiotracers displayed favorable biodistributions, clearance, and stability, allowing for higher image quality, shorter circulation half-life, and lower immunogenicity. This study aimed to comprehensively review the application of radiolabeled anti-HER2 antibody fragments in breast cancer in vivo imaging and provide a better understanding of targeted HER2 quantification.
Collapse
Affiliation(s)
- Fatemeh Movahed
- Department of Gynecology and Obstetrics, Yas Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ouldouz Navaei
- Department of Biotechnology, Università Milano-Bicocca Milano, Milan, Italy
| | - Shiva Taghlidi
- Medicine and Surgery, Università Degli Studi Di Milano-Bicocca, Milan, Italy
| | - Maryam Nurzadeh
- Department of Fetomaternal, Faculty of Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| | - Maryam Eslami Gharaati
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Rabiei
- Obstetrics and Gynecology Department, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Hou X, Kong X, Yao Y, Liu S, Ren Y, Hu M, Wang Z, Zhu H, Yang Z. Next Generation of Solid Target Radionuclide Antibody Conjugates for Tumor Immuno-Therapy. J Labelled Comp Radiopharm 2024; 67:396-409. [PMID: 39480113 DOI: 10.1002/jlcr.4124] [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: 04/26/2024] [Revised: 08/07/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024]
Abstract
Immune checkpoint therapy has emerged as an effective treatment option for various types of cancers. Key immune checkpoint molecules, such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein 1 (PD-1), and lymphocyte activation gene 3 (LAG-3), have become pivotal targets in cancer immunotherapy. Antibodies designed to inhibit these molecules have demonstrated significant clinical efficacy. Nevertheless, the ability to monitor changes in the immune status of tumors and predict treatment response remains limited. Conventional methods, such as assessing lymphocytes in peripheral blood or conducting tumor biopsies, are inadequate for providing real-time, spatial information about T-cell distributions within heterogeneous tumors. Positron emission tomography (PET) using T-cell specific probes represents a promising and noninvasive approach to monitor both systemic and intratumoral immune changes during treatment. This technique holds substantial clinical significance and potential utility. In this paper, we review the applications of PET probes that target immune cells in molecular imaging.
Collapse
Affiliation(s)
- Xingguo Hou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Peking University Cancer Hospital & Institute, Beijing, China
- Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
| | - Xiangxing Kong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yuan Yao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Peking University Cancer Hospital & Institute, Beijing, China
| | - Song Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ya'nan Ren
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Peking University Cancer Hospital & Institute, Beijing, China
- Guizhou University School of Medicine, Guiyang, Guizhou, China
| | - Muye Hu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zilei Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hua Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Peking University Cancer Hospital & Institute, Beijing, China
- Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
| | - Zhi Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Peking University Cancer Hospital & Institute, Beijing, China
- Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
| |
Collapse
|
8
|
Tian R, Kong J, He Y, Xu G, Chen T, Han J. Radiosynthesis and preclinical evaluations of [ 18F]AlF-RESCA-5F7 as a novel molecular probe for HER2 tumor imaging. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2024; 14:175-181. [PMID: 39027646 PMCID: PMC11253080 DOI: 10.62347/bvpk1360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/15/2024] [Indexed: 07/20/2024]
Abstract
HER2 overexpression is associated with various tumor types and prompted the development of targeted therapies. Previously, iso-[211At]SGMAB-5F7 was developed as a HER2-targeted alpha therapy agent, demonstrating promising therapeutic efficacy in the preclinical stage. Aiming for an 18F-labeled tracer for companion diagnostics in clinical translation, we employed the Al18F-RESCA strategy in our current work and investigated whether [18F]AlF-RESCA-5F7 could visualize HER2 expression in vivo. [18F]AlF-RESCA-5F7 was attained with high radiochemical purity (> 99%) and molar activity in the range of 16.5 ± 8.8 GBq/μmol (n = 8). Compared to previously reported radiotracers that contained 5F7 as the HER2-targeting carrier and fluorine-18 as the positron-emitting isotope, the radiosynthesis was simplified to one single step within 30 min. The dissociation constant of [18F]AlF-RESCA-5F7 was determined as 3.3 nM via saturation binding assay using SKOV3 ovarian carcinoma cells. Tumor uptake of the novel tracer in Balb/c nude mice bearing SKOV3 xenografts was 4.69 ± 1.51, 3.34 ± 0.82 and 3.77 ± 0.99 %ID/g at 1, 2, and 4 h post-injection. Even though high retention of radioactivity was seen in the kidneys, micro-PET/CT imaging of [18F]AlF-RESCA-5F7 delineated the tumor up to 4 h post-injection with minimal activity in the gallbladder, intestines, and bone. This study suggests that [18F]AlF-RESCA-5F7 is a promising HER2 PET radiotracer with an eased radiolabeling method. Whether [18F]AlF-RESCA-5F7 could work as a companion diagnostic agent to assist in patient stratification and treatment monitoring of iso-[211At]SGMAB-5F7 warrants further investigation.
Collapse
Affiliation(s)
- Ruhua Tian
- Department of Physiology, School of Basic Medicine, Guizhou Medical UniversityGuiyang 550009, Guizhou, China
| | - Jinping Kong
- Department of Physiology, School of Basic Medicine, Guizhou Medical UniversityGuiyang 550009, Guizhou, China
| | - Yingfang He
- Institute of Radiation Medicine, Fudan UniversityNo. 2094 Xietu Road, Shanghai 200032, China
| | - Guoqiang Xu
- Department of Physiology, School of Basic Medicine, Guizhou Medical UniversityGuiyang 550009, Guizhou, China
| | - Tengxiang Chen
- Department of Physiology, School of Basic Medicine, Guizhou Medical UniversityGuiyang 550009, Guizhou, China
| | - Junbin Han
- Department of Physiology, School of Basic Medicine, Guizhou Medical UniversityGuiyang 550009, Guizhou, China
- Institute of Radiation Medicine, Fudan UniversityNo. 2094 Xietu Road, Shanghai 200032, China
| |
Collapse
|
9
|
Li T, You Q, Zhang S, Li R, Xie S, Li D, Ai S, Yang R, Guo H. Performance of 18F-FDG PET/MRI and its parameters in staging and neoadjuvant therapy response evaluation in bladder cancer. iScience 2024; 27:109657. [PMID: 38689640 PMCID: PMC11059538 DOI: 10.1016/j.isci.2024.109657] [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: 09/07/2023] [Revised: 11/19/2023] [Accepted: 04/01/2024] [Indexed: 05/02/2024] Open
Abstract
18F-FDG PET/MRI shows potential efficacy in the diagnosis of bladder cancer (BLCA). However, the performance of 18F-FDG PET/MRI in staging and neoadjuvant therapy (NAT) response evaluation for BLCA patients remains elusive. Here, we conduct this study to evaluate the performance of 18F-FDG PET/MRI and its derived parameters for tumor staging and NAT response prediction in BLCA. Forty BLCA patients were retrospectively enrolled to evaluate the performance of 18F-FDG PET/MRI in staging and NAT response prediction in BLCA. The feasibility of using 18F-FDG PET/MRI-related parameters for tumor staging and NAT response evaluation was also analyzed. In conclusion, 18F-FDG PET/MRI is found to show good performance in the BLCA staging and NAT response prediction. Moreover, ΔSUVmean is an efficacious candidate parameter for NAT response prediction. This study highlights that 18F-FDG PET/MRI is a promising imaging approach in the clinical diagnosis and treatment for BLCA.
Collapse
Affiliation(s)
- Tianhang Li
- Department of Urology, Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China
- Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China
| | - Qinqin You
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Shiwei Zhang
- Department of Urology, Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Rushuai Li
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Shangxun Xie
- Department of Urology, Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Danyan Li
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Medical School of Nanjing University, Nanjing, China
| | - Shuyue Ai
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Rong Yang
- Department of Urology, Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Hongqian Guo
- Department of Urology, Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
10
|
Valenza C, Guidi L, Battaiotto E, Trapani D, Sartore Bianchi A, Siena S, Curigliano G. Targeting HER2 heterogeneity in breast and gastrointestinal cancers. Trends Cancer 2024; 10:113-123. [PMID: 38008666 DOI: 10.1016/j.trecan.2023.11.001] [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: 09/18/2023] [Revised: 10/29/2023] [Accepted: 11/01/2023] [Indexed: 11/28/2023]
Abstract
About 20% of breast and gastric cancers and 3% of colorectal carcinomas overexpress the human epidermal growth factor receptor 2 (HER2) and are sensitive to HER2-directed agents. The expression of HER2 may differ within the same tumoral lesion (spatial intralesional heterogeneity), from different tumor locations (spatial interlesional heterogeneity), and throughout treatments (temporal heterogeneity). Spatial and temporal heterogeneity may impact on response and resistance to HER2-targeting agents and its prevalence and predictive role changes across HER2-overexpressing solid tumors. Therefore, the definition and the characterization of HER2 heterogeneity pose many challenges and its implementation as a reproducible predictive biomarker would help in guiding treatment modulation.
Collapse
Affiliation(s)
- Carmine Valenza
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Lorenzo Guidi
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Elena Battaiotto
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Dario Trapani
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Andrea Sartore Bianchi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Salvatore Siena
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| |
Collapse
|
11
|
Malhaire C. Radiomics in 18F-FDG PET/CT predicts HER2 status in breast cancer with equivocal immunohistochemistry. Eur J Radiol 2024; 170:111238. [PMID: 38043379 DOI: 10.1016/j.ejrad.2023.111238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 11/25/2023] [Indexed: 12/05/2023]
Affiliation(s)
- Caroline Malhaire
- Institut Curie, Department of Medical Imaging, PSL Research University, 26 rue d'Ulm, 75005 Paris, France; Institut Curie, Research Center, U1288-LITO, Inserm, Paris-Saclay University, 91401 Orsay, France.
| |
Collapse
|
12
|
Seban RD. Reply to: HER2-Low Breast Cancer Can Be Visualized by HER2 PET. J Nucl Med 2023; 64:1841-1842. [PMID: 37709533 DOI: 10.2967/jnumed.123.266490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 08/08/2023] [Accepted: 08/22/2023] [Indexed: 09/16/2023] Open
|