1
|
Liu X, Chen Y, Zhang F, Qiu F, Xu X, Zhang J, He S, Ding D, Tan W, Song S. Preclinical Evaluation of 68Ga-Labeled SL1 Aptamer for c-Met Targeted PET Imaging. Mol Pharm 2025; 22:1615-1623. [PMID: 39930702 DOI: 10.1021/acs.molpharmaceut.4c01344] [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: 03/04/2025]
Abstract
Tyrosine protein kinase c-Met, encoded by the Met gene, is a membrane-associated receptor tyrosine kinase that is often aberrantly expressed in a wide range of tumors. The development of imaging probes specifically targeting c-Met is critical for improving cancer diagnostics. In this study, we successfully designed and fabricated an aptamer molecular imaging probe ([68Ga]Ga-NOTA-SL1) with high radiochemical purity (RCP), good stability in vitro, and high affinity for c-Met expressed tumors. As shown by the micro-PET/CT scanning, [68Ga]Ga-NOTA-SL1 efficiently imaged tumor models with varying c-Met expression. The quantitative analysis of micro-PET/CT showed tumor uptake of [68Ga]Ga-NOTA-SL1 in the HCC827 tumor models (30 min, 2.93 ± 0.64%ID/g; 60 min, 2.03 ± 0.67%ID/g; 90 min, 1.63 ± 0.61%ID/g), PC-9 tumor models (30 min, 2.1 ± 0.72%ID/g; 60 min, 1.7 ± 0.56%ID/g; 90 min, 1.33 ± 0.38%ID/g), and HCT116 tumor models (30 min, 1.4 ± 0.17%ID/g; 60 min, 1.23 ± 0.15%ID/g; 90 min, 0.97 ± 0.21%ID/g). The results of immunohistochemistry (IHC) further confirmed the targeting ability of [68Ga]Ga-NOTA-SL1 to c-Met from a molecular pathological perspective. The probe effectively imaged c-Met-positive tumors and demonstrated a favorable metabolism profile and targeting performance in non-small cell lung cancer (NSCLC) or colorectal cancer tumor models. Consequently, this probe shows promise as an imaging agent capable of providing valuable diagnostic insights into tumors with aberrant c-Met expression.
Collapse
Affiliation(s)
- Xuwei Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College and Center for Biomedical Imaging, Fudan University, Shanghai 200032, P. R. China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, P. R. China
- Key Laboratory of Nuclear Physics and Ion-Beam Application (MOE), Fudan University, Shanghai 200433, China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai 201315, China
- College of Chemistry and Materials Science, Shanghai Normal University, Shanghai 200233, P. R. China
| | - Yamei Chen
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Fengsheng Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College and Center for Biomedical Imaging, Fudan University, Shanghai 200032, P. R. China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, P. R. China
- Key Laboratory of Nuclear Physics and Ion-Beam Application (MOE), Fudan University, Shanghai 200433, China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai 201315, China
| | - Fengshuang Qiu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College and Center for Biomedical Imaging, Fudan University, Shanghai 200032, P. R. China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, P. R. China
- Key Laboratory of Nuclear Physics and Ion-Beam Application (MOE), Fudan University, Shanghai 200433, China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai 201315, China
| | - Xiaoping Xu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College and Center for Biomedical Imaging, Fudan University, Shanghai 200032, P. R. China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, P. R. China
- Key Laboratory of Nuclear Physics and Ion-Beam Application (MOE), Fudan University, Shanghai 200433, China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai 201315, China
| | - Jianping Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College and Center for Biomedical Imaging, Fudan University, Shanghai 200032, P. R. China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, P. R. China
- Key Laboratory of Nuclear Physics and Ion-Beam Application (MOE), Fudan University, Shanghai 200433, China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai 201315, China
| | - Simin He
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College and Center for Biomedical Imaging, Fudan University, Shanghai 200032, P. R. China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, P. R. China
- Key Laboratory of Nuclear Physics and Ion-Beam Application (MOE), Fudan University, Shanghai 200433, China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai 201315, China
| | - Ding Ding
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Weihong Tan
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College and Center for Biomedical Imaging, Fudan University, Shanghai 200032, P. R. China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, P. R. China
- Key Laboratory of Nuclear Physics and Ion-Beam Application (MOE), Fudan University, Shanghai 200433, China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai 201315, China
| |
Collapse
|
2
|
Minne RL, Luo NY, Mork CM, Wopat MR, Esbona K, Javeri S, Nickel KP, Hernandez R, LeBeau AM, Kimple RJ, Baschnagel AM. Evaluation of a Novel MET-Targeting Camelid-Derived Antibody in Head and Neck Cancer. Mol Pharm 2024; 21:6376-6384. [PMID: 39513517 PMCID: PMC11987585 DOI: 10.1021/acs.molpharmaceut.4c00938] [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] [Indexed: 11/15/2024]
Abstract
In head and neck squamous cell carcinoma (HNSCC), the mesenchymal epithelial transition (MET) receptor drives cancer growth, proliferation, and metastasis. MET is known to be overexpressed in HNSCC and, therefore, is an appealing therapeutic target. In this study, we evaluated MET expression in patients with HNSCC and investigated the potential imaging application of a novel MET-binding single-domain camelid antibody using positron emission tomography/computed tomography (PET/CT) in a preclinical MET-expressing HNSCC model. Multiplex immunostaining for MET protein performed on a tissue microarray from 203 patients with HNSCC found 86% of patients to have MET expression, with 14% having high expression and 53% having low MET expression. Using The Cancer Genome Atlas (TCGA) database, high MET RNA expression was associated with worse progression-free survival and overall survival in patients with HPV-negative HSNCC. Utilizing flow cytometry and immunofluorescence, our novel camelid antibody fused to a human IgG Fc chain (1E7-Fc) showed high binding affinity and specificity to high MET-expressing Detroit 562 cells but not to low MET-expressing HNSCC cells. The efficacy and biodistribution of [89Zr]Zr-1E7-Fc as a PET imaging agent was then investigated in a MET-expressing head and neck xenograft model. [89Zr]Zr-1E7-Fc rapidly localized and showed high tumor uptake in Detroit 562 xenografts (8.4% ID/g at 72 h postinjection), with rapid clearance from the circulatory system (2.7 tumor-to-blood radioactivity ratio at 72 h postinjection). Our preclinical data suggest that the camelid antibody 1E7-Fc could be a potential theranostic agent for HNSCC. Further investigations are warranted to confirm these findings in patients and to evaluate 1E7-Fc as an imaging agent and platform to deliver radionuclide or drug therapy to MET-driven cancers.
Collapse
Affiliation(s)
- Rachel L Minne
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Natalie Y Luo
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Caroline M Mork
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Madalynn R Wopat
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Karla Esbona
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Saahil Javeri
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Kwangok P Nickel
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Reinier Hernandez
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Aaron M LeBeau
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Randall J Kimple
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Andrew M Baschnagel
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| |
Collapse
|
3
|
Nakamoto Y, Inui Y, Hotta M, Wakabayashi H, Hanaoka H. Recent advancements in new tracers from first-in-human studies. Ann Nucl Med 2024; 38:877-883. [PMID: 39325320 PMCID: PMC11489164 DOI: 10.1007/s12149-024-01979-5] [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: 08/16/2024] [Accepted: 09/04/2024] [Indexed: 09/27/2024]
Abstract
Recent advancements in the development of positron emission tomography (PET) tracers have significantly enhanced our ability to image neuroinflammatory processes and neurotransmitter systems, which are vital for understanding and treating neurodegenerative and psychiatric disorders. Similarly, innovative tracers in oncology provide detailed images of the metabolic and molecular characteristics of tumors, which are crucial for tailoring targeted therapies and monitoring responses, including radiotherapy. Notable advancements include programmed death ligand 1 (PD-L1)-targeting agents for lung cancer, prostate-specific membrane antigen-based tracers for prostate cancer, chemokine receptor-targeting agents for hematological malignancies, human epidermal growth factor receptor 2 (HER2)-targeting tracers for various cancers, Claudin 18 based tracers for epithelial tumors, glutamine tracers for colorectal cancer, and ascorbic acid analogs for assessing cancer metabolism and therapy efficacy. Additionally, novel tracers have been developed for non-neurological and non-oncological applications, including adrenal imaging, amyloidosis, and human immunodeficiency virus (HIV) infection. This overview focuses on the newly developed tracers, particularly those used in neurology and oncology.
Collapse
Affiliation(s)
- Yuji Nakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan.
| | - Yoshitaka Inui
- Department of Radiology, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 470-1192, Japan
| | - Masatoshi Hotta
- Department of Radiology and Nuclear Medicine, National Center for Global Health and Medicine, 1-21-1 Toyama Shinjuku-Ku, Tokyo, 162-8655, Japan
| | - Hiroshi Wakabayashi
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Hirofumi Hanaoka
- Division of Fundamental Technology Development, Near InfraRed Photo-ImmunoTherapy Research Institute at Kansai Medical University, 2-5-1, Shin-Machi, Hirakata, 573-1010, Japan
| |
Collapse
|
4
|
Collamati F, Morganti S, van Oosterom MN, Campana L, Ceci F, Luzzago S, Mancini-Terracciano C, Mirabelli R, Musi G, Nicolanti F, Orsi I, van Leeuwen FWB, Faccini R. First-in-human validation of a DROP-IN β-probe for robotic radioguided surgery: defining optimal signal-to-background discrimination algorithm. Eur J Nucl Med Mol Imaging 2024; 51:3098-3108. [PMID: 38376805 PMCID: PMC11300660 DOI: 10.1007/s00259-024-06653-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/07/2024] [Indexed: 02/21/2024]
Abstract
PURPOSE In radioguided surgery (RGS), radiopharmaceuticals are used to generate preoperative roadmaps (e.g., PET/CT) and to facilitate intraoperative tracing of tracer avid lesions. Within RGS, there is a push toward the use of receptor-targeted radiopharmaceuticals, a trend that also has to align with the surgical move toward minimal invasive robotic surgery. Building on our initial ex vivo evaluation, this study investigates the clinical translation of a DROP-IN β probe in robotic PSMA-guided prostate cancer surgery. METHODS A clinical-grade DROP-IN β probe was developed to support the detection of PET radioisotopes (e.g., 68 Ga). The prototype was evaluated in 7 primary prostate cancer patients, having at least 1 lymph node metastases visible on PSMA-PET. Patients were scheduled for radical prostatectomy combined with extended pelvic lymph node dissection. At the beginning of surgery, patients were injected with 1.1 MBq/kg of [68Ga]Ga-PSMA. The β probe was used to trace PSMA-expressing lymph nodes in vivo. To support intraoperative decision-making, a statistical software algorithm was defined and optimized on this dataset to help the surgeon discriminate between probe signals coming from tumors and healthy tissue. RESULTS The DROP-IN β probe helped provide the surgeon with autonomous and highly maneuverable tracer detection. A total of 66 samples (i.e., lymph node specimens) were analyzed in vivo, of which 31 (47%) were found to be malignant. After optimization of the signal cutoff algorithm, we found a probe detection rate of 78% of the PSMA-PET-positive samples, a sensitivity of 76%, and a specificity of 93%, as compared to pathologic evaluation. CONCLUSION This study shows the first-in-human use of a DROP-IN β probe, supporting the integration of β radio guidance and robotic surgery. The achieved competitive sensitivity and specificity help open the world of robotic RGS to a whole new range of radiopharmaceuticals.
Collapse
Affiliation(s)
| | - Silvio Morganti
- National Institute of Nuclear Physics (INFN), Section of Rome, Rome, Italy
| | - Matthias N van Oosterom
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Lorenzo Campana
- National Institute of Nuclear Physics (INFN), Section of Rome, Rome, Italy
- Department of Scienze di Base e Applicate per l'Ingegneria (SBAI), Sapienza University of Rome, Rome, Italy
| | - Francesco Ceci
- Division of Nuclear Medicine, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology-Oncology, University of Milan, Milan, Italy
| | - Stefano Luzzago
- Department of Oncology and Hematology-Oncology, University of Milan, Milan, Italy
- Department of Urology, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Carlo Mancini-Terracciano
- National Institute of Nuclear Physics (INFN), Section of Rome, Rome, Italy
- Department of Physics, Sapienza University of Rome, Rome, Italy
| | - Riccardo Mirabelli
- National Institute of Nuclear Physics (INFN), Section of Rome, Rome, Italy.
- Department of Scienze di Base e Applicate per l'Ingegneria (SBAI), Sapienza University of Rome, Rome, Italy.
| | - Gennaro Musi
- Department of Oncology and Hematology-Oncology, University of Milan, Milan, Italy
- Department of Urology, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Francesca Nicolanti
- National Institute of Nuclear Physics (INFN), Section of Rome, Rome, Italy
- Department of Physics, Sapienza University of Rome, Rome, Italy
| | - Ilaria Orsi
- National Institute of Nuclear Physics (INFN), Section of Rome, Rome, Italy
- Department of Physics, Sapienza University of Rome, Rome, Italy
| | - Fijs W B van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Riccardo Faccini
- National Institute of Nuclear Physics (INFN), Section of Rome, Rome, Italy
- Department of Physics, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
5
|
Luo NY, Minne RL, Gallant JP, Gunaratne GS, West JL, Javeri S, Robertson AJ, Lake EW, Engle JW, Mixdorf JC, Aluicio-Sarduy E, Nickel KP, Hernandez R, Kimple RJ, Baschnagel AM, LeBeau AM. Development of an Engineered Single-Domain Antibody for Targeting MET in Non-Small Cell Lung Cancer. Bioconjug Chem 2024; 35:389-399. [PMID: 38470611 PMCID: PMC12060584 DOI: 10.1021/acs.bioconjchem.4c00019] [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] [Indexed: 03/14/2024]
Abstract
The Mesenchymal Epithelial Transition (MET) receptor tyrosine kinase is upregulated or mutated in 5% of non-small-cell lung cancer (NSCLC) patients and overexpressed in multiple other cancers. We sought to develop a novel single-domain camelid antibody with high affinity for MET that could be used to deliver conjugated payloads to MET expressing cancers. From a naïve camelid variable-heavy-heavy (VHH) domain phage display library, we identified a VHH clone termed 1E7 that displayed high affinity for human MET and was cross-reactive with MET across multiple species. When expressed as a bivalent human Fc fusion protein, 1E7-Fc was found to selectively bind to EBC-1 (MET amplified) and UW-Lung 21 (MET exon 14 mutated) cell lines by flow cytometry and immunofluorescence imaging. Next, we investigated the ability of [89Zr]Zr-1E7-Fc to detect MET expression in vivo by PET/CT imaging. [89Zr]Zr-1E7-Fc demonstrated rapid localization and high tumor uptake in both xenografts with a %ID/g of 6.4 and 5.8 for EBC-1 and UW-Lung 21 at 24 h, respectively. At the 24 h time point, clearance from secondary and nontarget tissues was also observed. Altogether, our data suggest that 1E7-Fc represents a platform technology that can be employed to potentially both image and treat MET-altered NSCLC.
Collapse
Affiliation(s)
- Natalie Y. Luo
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Rachel L. Minne
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Joseph P. Gallant
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- Molecular and Cellular Pharmacology Program, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Gihan S Gunaratne
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Jayden L. West
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- Molecular and Cellular Pharmacology Program, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Saahil Javeri
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Austin J. Robertson
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- Molecular and Cellular Pharmacology Program, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Eric W. Lake
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Jonathan W. Engle
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Jason C. Mixdorf
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Eduardo Aluicio-Sarduy
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Kwang P. Nickel
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Reinier Hernandez
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Randall J. Kimple
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Andrew M. Baschnagel
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Aaron M. LeBeau
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| |
Collapse
|
6
|
Bu L, Ma X, Ji A, Geng K, Feng H, Li L, Zhang A, Cheng Z. Development of a novel 18F-labeled small molecule probe for PET imaging of mesenchymal epithelial transition receptor expression. Eur J Nucl Med Mol Imaging 2024; 51:656-668. [PMID: 37940685 DOI: 10.1007/s00259-023-06495-8] [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: 08/08/2023] [Accepted: 10/26/2023] [Indexed: 11/10/2023]
Abstract
The mesenchymal epithelial transition factor (c-Met) is frequently overexpressed in numerous cancers and has served as a validated anticancer target. Inter- and intra-tumor heterogeneity of c-Met, however, challenges the use of anti-MET therapies, highlighting an urgent need to develop an alternative tool for visualizing whole-body c-Met expression quantitatively and noninvasively. Here we firstly reported an 18F labeled, small-molecule quinine compound-based PET probe, 1-(4-(5-amino-7-(trifluoromethyl) quinolin-3-yl) piperazin-1-yl)-2-(fluoro-[18F]) propan-1-one, herein referred as [18F]-AZC. METHODS [18F]-AZC was synthesized via a one-step substitution reaction and characterized by radiochemistry methods. [18F]-AZC specificity and affinity toward c-Met were assessed by cell uptake assay, with or without cold compound [19F]-AZC or commercial c-Met inhibitor blocking. MicroPET/CT imaging and biodistribution studies were conducted in subcutaneous murine xenografts of glioma. Additionally, [18F]-AZC was then further evaluated in orthotopic glioma xenografts, by microPET/CT imaging accompanied with MRI and autoradiography for co-registration of the tumor. Immunofluorescence staining was also carried out to qualitatively evaluate the c-Met expression in tumor tissue, co-localizes with H&E staining. RESULTS This probe shows easy radiosynthesis, high stability in vitro and in vivo, high targeting affinity, and favorable lipophilicity and brain transport coefficient. [18F]-AZC demonstrates excellent tumor imaging properties in vivo and can delineate c-Met positive glioma specifically at 1 h after intravenous injection of the probe. Moreover, favorable correlation was observed between the [18F]-AZC accumulation and the amount of c-Met expression in tumor. CONCLUSION This novel imaging probe could be applied as a valuable tool for management of anti-c-Met therapies in patients in the future.
Collapse
Affiliation(s)
- Lihong Bu
- PET-CT/MRI Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Stanford University, Stanford, CA, 94305-5484, USA
| | - Xiaowei Ma
- PET-CT Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Aiyan Ji
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 201203, China
- Department of Pharmacy, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Kaijun Geng
- National Key Laboratory of Innovative Immunotherapy, Shanghai Frontiers Science Center for Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hongyan Feng
- PET-CT/MRI Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Li Li
- PET-CT/MRI Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ao Zhang
- National Key Laboratory of Innovative Immunotherapy, Shanghai Frontiers Science Center for Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Stanford University, Stanford, CA, 94305-5484, USA.
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 201203, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai rim Advanced Research Institute for Drug Discovery, Yantai, 264117, Shandong, China.
| |
Collapse
|
7
|
Rusu T, Delion M, Pirot C, Blin A, Rodenas A, Talbot JN, Veran N, Portal C, Montravers F, Cadranel J, Prignon A. Fully automated radiolabeling of [ 68Ga]Ga-EMP100 targeting c-MET for PET-CT clinical imaging. EJNMMI Radiopharm Chem 2023; 8:30. [PMID: 37843660 PMCID: PMC10579204 DOI: 10.1186/s41181-023-00213-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/03/2023] [Indexed: 10/17/2023] Open
Abstract
BACKGROUND c-MET is a transmembrane receptor involved in many biological processes and contributes to cell proliferation and migration during cancer invasion process. Its expression is measured by immunehistochemistry on tissue biopsy in clinic, although this technique has its limitations. PET-CT could allow in vivo mapping of lesions expressing c-MET, providing whole-body detection. A number of radiopharmaceuticals are under development for this purpose but are not yet in routine clinical use. EMP100 is a cyclic oligopeptide bound to a DOTA chelator, with nanomolar affinity for c-MET. The aim of this project was to develop an automated method for radiolabelling the radiopharmaceutical [68Ga]Ga-EMP100. RESULTS The main results showed an optimal pH range between 3.25 and 3.75 for the complexation reaction and a stabilisation of the temperature at 90 °C, resulting in an almost complete incorporation of gallium-68 after 10 min of heating. In these experiments, 90 µg of EMP-100 peptide were initially used and then lower amounts (30, 50, 75 µg) were explored to determine the minimum required for sufficient synthesis yield. Radiolysis impurities were identified by radio-HPLC and ascorbic acid and ethanol were used to improve the purity of the compound. Three batches of [68Ga]Ga-EMP100 were then prepared according to the optimised parameters and all met the established specifications. Finally, the stability of [68Ga]Ga-EMP100 was assessed at room temperature over 3 h with satisfactory results in terms of appearance, pH, radiochemical purity and sterility. CONCLUSIONS For the automated synthesis of [68Ga]Ga-EMP100, the parameters of pH, temperature, precursor peptide content and the use of adjuvants for impurity management were efficiently optimised, resulting in the production of three compliant and stable batches according to the principles of good manufacturing practice. [68Ga]Ga-EMP100 was successfully synthesised and is now available for clinical development in PET-CT imaging.
Collapse
Affiliation(s)
- Timofei Rusu
- THERANOSCAN Clinical Research Group Sorbonne University, Tenon Hospital AP-HP, Paris, France.
- Positron Molecular Imaging Laboratory (LIMP) UMS28 Small Animal Phenotyping, Sorbonne University, Paris, France.
- Nuclear Medicine Imaging Department and Radiopharmacy, Tenon Hospital AP-HP, Paris, France.
- Radiopharmacist - Hôpital Tenon Assistance Publique - Hôpitaux de Paris, Paris, France.
| | - Matthieu Delion
- Nuclear Medicine Imaging Department and Radiopharmacy, Tenon Hospital AP-HP, Paris, France
| | - Charlotte Pirot
- Nuclear Medicine Imaging Department and Radiopharmacy, Tenon Hospital AP-HP, Paris, France
| | - Amaury Blin
- Nuclear Medicine Imaging Department and Radiopharmacy, Tenon Hospital AP-HP, Paris, France
| | - Anita Rodenas
- THERANOSCAN Clinical Research Group Sorbonne University, Tenon Hospital AP-HP, Paris, France
| | - Jean-Noël Talbot
- Institut National des Sciences et Techniques Nucléaires (INSTN), Saclay, France
| | - Nicolas Veran
- CHRU de Nancy Pôle Pharmacie : Centre Hospitalier Régional Universitaire de Nancy Pôle Pharmacie, Nancy, France
| | | | - Françoise Montravers
- Nuclear Medicine Imaging Department and Radiopharmacy, Tenon Hospital AP-HP, Paris, France
| | - Jacques Cadranel
- THERANOSCAN Clinical Research Group Sorbonne University, Tenon Hospital AP-HP, Paris, France
- Service de Pneumologie et Oncologie Thoracique, APHP - Hôpital Tenon and Sorbonne Université, Paris, France
| | - Aurélie Prignon
- THERANOSCAN Clinical Research Group Sorbonne University, Tenon Hospital AP-HP, Paris, France
- Positron Molecular Imaging Laboratory (LIMP) UMS28 Small Animal Phenotyping, Sorbonne University, Paris, France
| |
Collapse
|
8
|
Huang W, Peng Y, Zhang Y, Qiu Y, Liu Y, Wang A, Kang L. Multimodality imaging of Xp11.2 translocation/TFE3 gene fusion associated with renal cell carcinoma: a case report. Front Med (Lausanne) 2023; 10:1266630. [PMID: 37795411 PMCID: PMC10546202 DOI: 10.3389/fmed.2023.1266630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 08/31/2023] [Indexed: 10/06/2023] Open
Abstract
Background Xp11.2 translocation/TFE3 gene fusion associated with renal cell carcinoma (Xp11.2 RCC) exhibits unique biological characteristics and is associated with an increased incidence of tumor thrombosis, lymph node metastasis, and advanced disease stages. Multimodality imaging, including US, contrast-enhanced CT, multi-parametric MRI, and 18F-FDG PET/CT plays a crucial role in the preoperative diagnosis and differentiation of renal tumors. Case report A 15-year-old female presented with lumbar pain worsened, and developed persistent painless hematuria. The CT attenuation values of the scan without contrast, corticomedullary phase, nephrographic phase, and delayed phases were 35 HU, 83 HU, 82 HU, and 75 HU, respectively. The solid component of the mass displayed heterogeneous marked enhancement. Furthermore, MRU indicated that the lesion involved the cortical medulla and infringed on the renal sinus fat. The lesion appeared isosignal in T1WI, slightly low signal in T2WI, and slightly high signal in DWI. The degree of enhancement in the three phases of enhancement scan was lower than that in the renal parenchyma, and hemorrhage and necrosis were observed within the internal part of the lesion. To further clarify the staging, the patient underwent 18F-FDG PET/CT. PET/CT images showed multiple irregular occupancies in the right kidney with unclear borders, showing a heterogeneous increase in 18F-FDG uptake, with SUVmax values ranging from 2.3 to 5.2 in the routine imaging phase (60 min post-injection), compared to SUVmax values ranging from 2.8 to 6.9 in the delayed imaging phase (160 min post-injection). Additionally, multiple enlarged and fused lymph nodes were observed in the medial part of the right kidney and the retroperitoneum, exhibiting a heterogeneous increase in 18F-FDG uptake, with SUVmax values ranging from 4.1 to 8.7 in the routine imaging phase, compared to SUVmax values ranging from 4.4 to 9.1 in the delayed imaging phase. The postoperative pathology, immunohistochemistry, and molecular analysis of histiocytes were consistent with a diagnosis of Xp11.2 RCC. One month after surgery, enhanced-CT examination of the patient revealed lung metastasis, peritoneal metastasis, and multiple lymph node metastases throughout the body, with an overall survival of 16 months. Conclusion Xp11.2 RCC exhibits unique biological characteristics and is associated with an increased incidence of tumor thrombosis, lymph node metastasis, and advanced disease stages. Long-term follow-up is essential to monitor the likelihood of recurrence and metastasis. 18F-FDG PET/CT examination can comprehensively visualize the lesion's location and extent, providing a basis for clinical tumor staging and aiding in treatment monitoring and follow-up. To address the limitations of FDG, the utilization of specific tracers designed for RCC or tracers that are not excreted via the urinary system would be ideal. Further advancements in molecular imaging technologies and the development of novel tracers hold great promise in advancing the diagnosis and management of RCC, ultimately contributing to better patient outcomes and overall disease management.
Collapse
Affiliation(s)
- Wenpeng Huang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - Yushuo Peng
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - Yongbai Zhang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - Yongkang Qiu
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - Yi Liu
- Department of Urology, Peking University First Hospital, Beijing, China
- Institute of Urology, Peking University, Beijing, China
- National Urological Cancer Center, Beijing, China
| | - Aixiang Wang
- Department of Urology, Peking University First Hospital, Beijing, China
- Institute of Urology, Peking University, Beijing, China
- National Urological Cancer Center, Beijing, China
| | - Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| |
Collapse
|
9
|
Teh JH, Amgheib A, Fu R, Barnes C, Abrahams J, Ashek A, Wang N, Yang Z, Mansoorudeen M, Long NJ, Aboagye EO. Evaluation of [ 18F]AlF-EMP-105 for Molecular Imaging of C-Met. Pharmaceutics 2023; 15:1915. [PMID: 37514101 PMCID: PMC10383791 DOI: 10.3390/pharmaceutics15071915] [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: 05/16/2023] [Revised: 06/29/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
C-Met is a receptor tyrosine kinase that is overexpressed in a range of different cancer types, and has been identified as a potential biomarker for cancer imaging and therapy. Previously, a 68Ga-labelled peptide, [68Ga]Ga-EMP-100, has shown promise for imaging c-Met in renal cell carcinoma in humans. Herein, we report the synthesis and preliminary biological evaluation of an [18F]AlF-labelled analogue, [18F]AlF-EMP-105, for c-Met imaging by positron emission tomography. EMP-105 was radiolabelled using the aluminium-[18F]fluoride method with 46 ± 2% RCY and >95% RCP in 35-40 min. In vitro evaluation showed that [18F]AlF-EMP-105 has a high specificity for c-Met-expressing cells. Radioactive metabolite analysis at 5 and 30 min post-injection revealed that [18F]AlF-EMP-105 has good blood stability, but undergoes transformation-transchelation, defluorination or demetallation-in the liver and kidneys. PET imaging in non-tumour-bearing mice showed high radioactive accumulation in the kidneys, bladder and urine, demonstrating that the tracer is cleared predominantly as [18F]fluoride by the renal system. With its high specificity for c-Met expressing cells, [18F]AlF-EMP-105 shows promise as a potential diagnostic tool for imaging cancer.
Collapse
Affiliation(s)
- Jin Hui Teh
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, UK
| | - Ala Amgheib
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London W12 0NN, UK
| | - Ruisi Fu
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London W12 0NN, UK
| | - Chris Barnes
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London W12 0NN, UK
| | - Joel Abrahams
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London W12 0NN, UK
| | - Ali Ashek
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London W12 0NN, UK
| | - Ning Wang
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London W12 0NN, UK
| | - Zixuan Yang
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London W12 0NN, UK
| | - Muneera Mansoorudeen
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London W12 0NN, UK
| | - Nicholas J Long
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, UK
| | - Eric O Aboagye
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London W12 0NN, UK
| |
Collapse
|
10
|
Flörsch B, Taugner J, Käsmann L, Kenndoff S, Guggenberger J, Tufman A, Reinmuth N, Duell T, Belka C, Eze C, Manapov F. Treatment patterns and prognosis of patients with inoperable stage III NSCLC after completion of concurrent chemoradiotherapy ± immune checkpoint inhibition: a decade-long single-center historical analysis. J Cancer Res Clin Oncol 2023; 149:3267-3276. [PMID: 35915184 PMCID: PMC10314870 DOI: 10.1007/s00432-022-04174-z] [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: 06/04/2022] [Accepted: 06/25/2022] [Indexed: 11/30/2022]
Abstract
PURPOSE To investigate the impact of treatment time and patterns in inoperable stage III non-small cell lung cancer (NSCLC) following concurrent chemoradiotherapy (cCRT) ± immune checkpoint inhibitors (ICIs). METHODS Patients were stratified by treatment year: A (2011-2014), B (2015-2017) and C (2018-2020). Tumor- and treatment-related characteristics regarding locoregional recurrence-free survival (LRRFS), progression-free survival (PFS) and overall survival (OS) were investigated. RESULTS One hundred and thirty-six consecutive patients were analyzed. All patients completed thoracic radiotherapy (TRT) to a total dose ≥ 60.0 Gy; 36 (26%) patients received ICI. Median PFS in subgroups A, B and C was 8.0, 8.2 and 26.3 months (p = 0.007). Median OS was 19.9 months, 23.4 months and not reached (NR), respectively. In group C, median LRRFS and PFS were 27.2 vs. NR; and 14.2 vs. 26.3 months in patients treated with and without ICI. On multivariate analysis planning target volume (PTV) ≥ 700 cc was a negative prognosticator of LRRFS (HR 2.194; p = 0.001), PFS (HR 1.522; p = 0.042) and OS (HR 2.883; p = 0.001); ICI was a predictor of LRRFS (HR 0.497; p = 0.062), PFS (HR 0.571; p = 0.071) and OS (HR 0.447; p = 0.1). In the non-ICI cohort, multivariate analyses revealed PTV ≥ 700 cc (p = 0.047) and a maximum standardized uptake value (SUVmax) ≥ 13.75 (p = 0.012) were predictors of PFS; PTV ≥ 700 cc (p = 0.017), SUVmax ≥ 13.75 (p = 0.002) and a total lung V20 ≥ 30% (V20 ≥ 30) (p < 0.05) were predictors of OS. CONCLUSIONS Patients treated after 2018 had improved survival regardless of ICI use. Implementation of ICI resulted in further significant increase of all tested survival endpoints. PTV ≥ 700 cc and ICI were only prognosticators for LRRFS, PFS and OS in the analyzed cohort.
Collapse
Affiliation(s)
- Benedikt Flörsch
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
| | - Julian Taugner
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
| | - Lukas Käsmann
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany.
- Member of the German Center for Lung Research (DZL), Comprehensive Pneumology Center Munich (CPC-M), Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.
| | - Saskia Kenndoff
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
| | - Julian Guggenberger
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
| | - Amanda Tufman
- Division of Respiratory Medicine and Thoracic Oncology, Department of Internal Medicine V, Thoracic Oncology Centre Munich, LMU Munich, Munich, Germany
| | - Niels Reinmuth
- Asklepios Kliniken GmbH, Asklepios Fachkliniken Munich, Gauting, Germany
| | - Thomas Duell
- Asklepios Kliniken GmbH, Asklepios Fachkliniken Munich, Gauting, Germany
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
- Member of the German Center for Lung Research (DZL), Comprehensive Pneumology Center Munich (CPC-M), Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Chukwuka Eze
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
| | - Farkhad Manapov
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
- Member of the German Center for Lung Research (DZL), Comprehensive Pneumology Center Munich (CPC-M), Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| |
Collapse
|
11
|
Machiraju D, Hassel JC. Targeting the cMET pathway to enhance immunotherapeutic approaches for mUM patients. Front Oncol 2023; 12:1068029. [PMID: 36761417 PMCID: PMC9902905 DOI: 10.3389/fonc.2022.1068029] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/28/2022] [Indexed: 01/25/2023] Open
Abstract
The liver is the most preferential initial site of metastasis for uveal melanoma (mUM), and this preference is associated with rapid mortality in mUM patients. Despite the significant clinical benefits of Immune checkpoint inhibitors (ICIs) in metastatic cutaneous melanoma patients, ICIs have shown little to no benefit in mUM patients. A potential reason for this inefficiency of ICI could be partly devoted to the involvement of the liver itself, thanks to its rich source of growth factors and immunosuppressive microenvironment. Uveal melanoma cells show increased expression of a transmembrane protein called cMET, which is known as the sole receptor for the Hepatocyte growth factor (HGF). Hyperactivation of cMET by HGF contributes to mUM development, and the liver, being the major source of HGF, may partially explain the metastasis of uveal melanoma cells to the liver. In addition, cMET/HGF signaling has also been shown to mediate resistance to ICI treatment, directly and indirectly, involving tumor and immune cell populations. Therefore, targeting the cMET/HGF interaction may enhance the efficacy of immunotherapeutic regimes for mUM patients. Hence in this minireview, we will discuss the rationale for combining cMET inhibitors/antibodies with leading immune checkpoint inhibitors for treating mUM. We will also briefly highlight the challenges and opportunities in targeting cMET in mUM.
Collapse
|
12
|
Unterrainer LM, Todica A, Beyer L, Brendel M, Holzgreve A, Kauffmann-Guerrero D, Unterrainer M, Bartenstein P, Tufman A. 68Ga-EMP-100 PET/CT-a novel method for non-invasive assessment of c-MET expression in non-small cell lung cancer. Eur J Nucl Med Mol Imaging 2023; 50:628-629. [PMID: 36253641 PMCID: PMC9816193 DOI: 10.1007/s00259-022-05995-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/06/2022] [Indexed: 01/11/2023]
Affiliation(s)
- Lena M Unterrainer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
| | - Andrei Todica
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- DIE RADIOLOGIE, Munich, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Adrien Holzgreve
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Diego Kauffmann-Guerrero
- Division of Respiratory Medicine and Thoracic Oncology, Department of Internal Medicine V, Thoracic Oncology Center Munich, University Hospital, LMU Munich, Munich, Germany
| | - Marcus Unterrainer
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Amanda Tufman
- Division of Respiratory Medicine and Thoracic Oncology, Department of Internal Medicine V, Thoracic Oncology Center Munich, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
13
|
Boekestijn I, van Oosterom MN, Dell'Oglio P, van Velden FHP, Pool M, Maurer T, Rietbergen DDD, Buckle T, van Leeuwen FWB. The current status and future prospects for molecular imaging-guided precision surgery. Cancer Imaging 2022; 22:48. [PMID: 36068619 PMCID: PMC9446692 DOI: 10.1186/s40644-022-00482-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 08/21/2022] [Indexed: 01/19/2023] Open
Abstract
Molecular imaging technologies are increasingly used to diagnose, monitor, and guide treatment of i.e., cancer. In this review, the current status and future prospects of the use of molecular imaging as an instrument to help realize precision surgery is addressed with focus on the main components that form the conceptual basis of intraoperative molecular imaging. Paramount for successful interventions is the relevance and accessibility of surgical targets. In addition, selection of the correct combination of imaging agents and modalities is critical to visualize both microscopic and bulk disease sites with high affinity and specificity. In this context developments within engineering/imaging physics continue to drive the growth of image-guided surgery. Particularly important herein is enhancement of sensitivity through improved contrast and spatial resolution, features that are critical if sites of cancer involvement are not to be overlooked during surgery. By facilitating the connection between surgical planning and surgical execution, digital surgery technologies such as computer-aided visualization nicely complement these technologies. The complexity of image guidance, combined with the plurality of technologies that are becoming available, also drives the need for evaluation mechanisms that can objectively score the impact that technologies exert on the performance of healthcare professionals and outcome improvement for patients.
Collapse
Affiliation(s)
- Imke Boekestijn
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Matthias N van Oosterom
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Paolo Dell'Oglio
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Urology, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Floris H P van Velden
- Medical Physics, Department of Radiology , Leiden University Medical Center, Leiden, the Netherlands
| | - Martin Pool
- Department of Clinical Farmacy and Toxicology, Leiden University Medical Center, Leiden, the Netherlands
| | - Tobias Maurer
- Martini-Klinik Prostate Cancer Centre Hamburg, Hamburg, Germany
| | - Daphne D D Rietbergen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Tessa Buckle
- Interventional Molecular Imaging Laboratory, Department of Radiology, 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.
| |
Collapse
|
14
|
Floresta G, Abbate V. Recent progress in the imaging of c-Met aberrant cancers with positron emission tomography. Med Res Rev 2022; 42:1588-1606. [PMID: 35292998 PMCID: PMC9314990 DOI: 10.1002/med.21885] [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: 07/09/2021] [Revised: 01/31/2022] [Accepted: 02/22/2022] [Indexed: 11/06/2022]
Abstract
Tyrosine-protein kinase Met-also known as c-Met or HGFR-is a membrane receptor protein with associated tyrosine kinase activity physiologically stimulated by its natural ligand, the hepatocyte growth factor (HGF), and is involved in different ways in cancer progression and tumourigenesis. Targeting c-Met with pharmaceuticals has been preclinically proved to have significant benefits for cancer treatment. Recently, evaluating the protein status during and before c-Met targeted therapy has been shown of relevant importance by different studies, demonstrating that there is a correlation between the status (e.g., aberrant activation and overexpression) of the HGFR with therapy response and clinical prognosis. Currently, clinical imaging based on positron emission tomography (PET) appears as one of the most promising tools for the in vivo real-time scanning of irregular alterations of the tyrosine-protein kinase Met and for the diagnosis of c-Met related cancers. In this study, we review the recent progress in the imaging of c-Met aberrant cancers with PET. Particular attention is directed on the development of PET probes with a range of different sizes (HGF, antibodies, anticalines, peptides, and small molecules), and radiolabeled with different radionuclides. The goal of this review is to report all the preclinical imaging studies based on PET imaging reported until now for in vivo diagnosis of c-Met in oncology to support the design of novel and more effective PET probes for in vivo evaluation of c-Met.
Collapse
Affiliation(s)
- Giuseppe Floresta
- Department of Analytical, Environmental and Forensic Sciences, Institute of Pharmaceutical Sciences, King's College London, London, UK
| | - Vincenzo Abbate
- Department of Analytical, Environmental and Forensic Sciences, Institute of Pharmaceutical Sciences, King's College London, London, UK
| |
Collapse
|
15
|
Urso L, Castello A, Rocca GC, Lancia F, Panareo S, Cittanti C, Uccelli L, Florimonte L, Castellani M, Ippolito C, Frassoldati A, Bartolomei M. Role of PSMA-ligands imaging in Renal Cell Carcinoma management: current status and future perspectives. J Cancer Res Clin Oncol 2022; 148:1299-1311. [PMID: 35217902 PMCID: PMC9114025 DOI: 10.1007/s00432-022-03958-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 02/14/2022] [Indexed: 12/17/2022]
Abstract
Background Renal masses detection is continually increasing worldwide, with Renal Cell Carcinoma (RCC) accounting for approximately 90% of all renal cancers and remaining one of the most aggressive urological malignancies. Despite improvements in cancer management, accurate diagnosis and treatment strategy of RCC by computed tomography (CT) and magnetic resonance imaging (MRI) are still challenging. Prostate-Specific Membrane Antigen (PSMA) is known to be highly expressed on the endothelial cells of the neovasculature of several solid tumors other than prostate cancer, including RCC. In this context, recent preliminary studies reported a promising role for positron emission tomography (PET)/CT with radiolabeled molecules targeting PSMA, in alternative to fluorodeoxyglucose (FDG) in RCC patients. Purpose The aim of our review is to provide an updated overview of current evidences and major limitations regarding the use of PSMA PET/CT in RCC. Methods A literature search, up to 31 December 2021, was performed using the following electronic databases: PubMed, SCOPUS, Web of Science, and Google Scholar. Results The findings of this review suggest that PSMA PET/CT could represent a valid imaging option for diagnosis, staging, and therapy response evaluation in RCC, particularly in clear cell RCC. Conclusions Further studies are needed for this “relatively” new imaging modality to consolidate its indications, timing, and practical procedures.
Collapse
Affiliation(s)
- Luca Urso
- Department of Translational Medicine, University of Ferrara, Via Aldo Moro 8, 44124, Ferrara, Italy.,Nuclear Medicine Unit, Oncological Medical and Specialists Department, University Hospital of Ferrara, Ferrara, Italy
| | - Angelo Castello
- Department of Nuclear Medicine, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Federica Lancia
- Oncological Medical and Specialists Department, Oncology Unit, University Hospital of Ferrara, Ferrara, Italy
| | - Stefano Panareo
- Nuclear Medicine Unit, Oncology and Haematology Department, University Hospital of Modena, Modena, Italy
| | - Corrado Cittanti
- Department of Translational Medicine, University of Ferrara, Via Aldo Moro 8, 44124, Ferrara, Italy. .,Nuclear Medicine Unit, Oncological Medical and Specialists Department, University Hospital of Ferrara, Ferrara, Italy.
| | - Licia Uccelli
- Department of Translational Medicine, University of Ferrara, Via Aldo Moro 8, 44124, Ferrara, Italy.,Nuclear Medicine Unit, Oncological Medical and Specialists Department, University Hospital of Ferrara, Ferrara, Italy
| | - Luigia Florimonte
- Department of Nuclear Medicine, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Massimo Castellani
- Department of Nuclear Medicine, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Carmelo Ippolito
- Urology Unit, Surgical Department, University Hospital of Ferrara, Ferrara, Italy
| | - Antonio Frassoldati
- Oncological Medical and Specialists Department, Oncology Unit, University Hospital of Ferrara, Ferrara, Italy
| | - Mirco Bartolomei
- Nuclear Medicine Unit, Oncological Medical and Specialists Department, University Hospital of Ferrara, Ferrara, Italy
| |
Collapse
|