1
|
Zhou X, Yan S, Ma X, Zhu H, Liu B, Yang X, Jia B, Yang Z, Wu N, Li N. Efficacy of radiolabelled PD-L1-targeted nanobody in predicting and evaluating the combined immunotherapy and chemotherapy for resectable non-small cell lung cancer. Eur J Nucl Med Mol Imaging 2025; 52:2343-2354. [PMID: 39912938 DOI: 10.1007/s00259-025-07115-3] [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: 10/31/2024] [Accepted: 01/24/2025] [Indexed: 02/07/2025]
Abstract
BACKGROUND This study aimed to assess the predictive and evaluative value of PD-L1 targeted 68Ga-THP-APN09 PET/CT in the neoadjuvant immunotherapy combined with chemotherapy for resectable non-small cell lung cancer (NSCLC), and to explore its potential in indicating immunotherapy-related adverse effects (irAEs). METHODS Fifty patients with resectable NSCLC enrolled in this prospective study underwent baseline 68Ga-THP-APN09 PET/CT and 18F-FDG PET/CT, with follow-up 18F-FDG PET/CT conducted, additionally, 36 patients received follow-up 68Ga-THP-APN09 PET/CT. Surgery was performed following 2-4 cycles of toripalimab combined with chemotherapy if R0 resection was feasible. The major pathologic response (MPR) state of the post-operative specimen and the adverse effects following combined therapy were documented. The correlation between PD-L1 expression and baseline 68Ga-THP-APN09 PET/CT uptake was determined. The predictive and evaluative efficacies of baseline and follow-up 68Ga-THP-APN09 PET/CT, along with 18F-FDG PET/CT, in determining MPR, were compared. RESULTS The SUVmax values of baseline 68Ga-THP-APN09 PET/CT were significantly higher in patients exhibiting high-positive PD-L1 expression compared to those with low-positive and negative expression (P = 0.001). And the SUVmax values of baseline 68Ga-THP-APN09 PET/CT in the response group, as determined by 18F-FDG PET/CT evaluation, were significantly higher than those in the non-response group (3.4 vs. 2.4, P < 0.001). Totally, 41 patients underwent surgery, of which 27 achieved MPR, while 14 did not. The SUVmax in baseline 68Ga-THP-APN09 PET/CT demonstrated statistical significance between the MPR and non-MPR groups, with area under the ROC curve (AUC) of 0.88 (95%CI: 0.77-0.99) in identifying MPR. However, the SUVmax in baseline 18F-FDG PET/CT failed to demonstrated significant predictive power, with AUC values of 0.68 (95%CI: 0.50-0.86, P = 0.076). While the SUVmax in follow-up 68Ga-THP-APN09 and 18F-FDG PET/CT, along with their change rate (ΔSUVmax%), demonstrated good predictive efficacy in identifying MPR, with AUC values of 0.81 (0.64-0.98), 0.91 (0.82-1.00), 0.93 (0.84-1.00), and 0.96 (0.89-1.00), respectively. Furthermore, the follow-up 68Ga-THP-APN09 PET/CT could remarkedly indicate the potential for thyroiditis. CONCLUSION Baseline 68Ga-THP-APN09 PET/CT alone could predict efficacy and assist in patient screening for immunotherapy combined chemotherapy in resectable NSCLC, and the follow-up 68Ga-THP-APN09 PET/CT and their change rates could aid in therapy evaluation. Additionally, follow-up 68Ga-THP-APN09 PET/CT could be utilized to monitor the immunotherapy-related thyroiditis during the therapy. TRIAL REGISTRATION NCT05156515, registered 8 December 2021, https://register. CLINICALTRIALS gov/prs/app/action/SelectProtocol?sid=S000BMSI%26;selectaction=Edit%26;uid=U000503E%26;ts=2%26;cx=zeghuw .
Collapse
Affiliation(s)
- Xin Zhou
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd, Beijing, 100142, China
| | - Shi Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Xiaopan Ma
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd, Beijing, 100142, China
- Department of Nuclear Medicine, Affiliated Hospital of Hubei, Xiangyang Central Hospital, University of Arts and Science, Xiangyang, 441138, China
| | - Hua Zhu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd, Beijing, 100142, China
| | - Bing Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Xin Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Bing Jia
- Medical Isotopes Research Center, Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, No.38 Xueyuan Rd, Beijing, 100191, China.
| | - Zhi Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd, Beijing, 100142, China.
| | - Nan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China.
| | - Nan Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd, Beijing, 100142, China.
| |
Collapse
|
2
|
Hughes DJ, Chand G, Johnson J, Tegala R, Bailey D, Adamson K, Edmonds S, Meszaros LK, Moore AEB, Manickavasagar T, Ndagire S, Gennatas S, Georgiou A, Ghosh S, Josephs D, Karapanagiotou E, McLean E, Ting HH, Spicer J, Goh V, Cook GJR. PD-L1 imaging with [ 99mTc]NM-01 SPECT/CT is associated with metabolic response to pembrolizumab with/without chemotherapy in advanced lung cancer. Br J Cancer 2025; 132:913-921. [PMID: 40188291 DOI: 10.1038/s41416-025-02991-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 02/15/2025] [Accepted: 03/18/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Programmed death-ligand 1 (PD-L1) immunohistochemistry is a predictive biomarker for anti-PD-(L)1 therapy in non-small cell lung cancer (NSCLC). It is not a reliable predictor of clinical benefit with non-invasive imaging providing a potential solution. We present the PECan study, the aim of which to assess the relationship of [99mTc]-labeled anti-PD-L1 single-domain antibody (NM-01) single-photon emission computed tomography (SPECT)/CT with metabolic response to anti-PD-(L)1. METHODS PD-L1 tumour proportion score (TPS) measured using SP263 assay. [99mTc]NM-01 SPECT/CT and [18F]FDG PET/CT performed before and 9-weeks following pembrolizumab with/without chemotherapy in patients with advanced NSCLC. Tumor (T) to blood pool (BP) maximum region of interest (ROImax) measurements performed in primary and metastatic lesions using SPECT/CT images. RESULTS Fifteen patients were included (median age 63 years, 9 male). Intertumoural heterogeneity evident in 10(67%) patients. Mean [99mTc]NM-01 T:BP demonstrated moderate correlation with PD-L1 TPS (r = 0.45, p < 0.05). Depth of [18F]FDG PET/CT metabolic response at 9-weeks (n = 13), correlated strongly with baseline [99mTc]NM-01 T:BP (r = -0.73, p < 0.05), but only moderately with PD-L1 TPS (r = -0.46, p = 0.06). CONCLUSION [99mTc]NM-01 SPECT/CT allows non-invasive quantification of PD-L1 in primary tumour and metastases in NSCLC. [99mTc]NM-01 uptake moderately correlates with PD-L1 immunohistochemistry, determines heterogeneity, and is associated with early metabolic response to anti-PD-1 pembrolizumab. CLINICAL TRIALS REGISTRATION PD-L1 Expression in Cancer (PECan) study (NCT04436406), registered 18 June 2020 https://clinicaltrials.gov/ct2/show/NCT04436406.
Collapse
Affiliation(s)
- Daniel Johnathan Hughes
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
- King's College London & Guy's and St. Thomas' PET Centre, St Thomas' Hospital, London, UK
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Gitasha Chand
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
- Nanomab Technology (UK) Limited, Borehamwood, Hertfordshire, UK
| | - Jessica Johnson
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | - Ronan Tegala
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | - Damion Bailey
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | - Kathryn Adamson
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | - Scott Edmonds
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | | | - Amelia Elizabeth Broomfield Moore
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
| | - Thubeena Manickavasagar
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Department of Radiology, Guy's and St. Thomas' NHS Foundation Trust, St Thomas' Hospital, London, UK
| | - Susan Ndagire
- King's Health Partners Cancer Biobank, Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, Great Maze Pond, London, UK
| | - Spyridon Gennatas
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Alexandros Georgiou
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, Great Maze Pond, London, UK
| | - Sharmistha Ghosh
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Debra Josephs
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, Great Maze Pond, London, UK
| | - Eleni Karapanagiotou
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, Great Maze Pond, London, UK
| | - Emma McLean
- Department of Histopathology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Hong Hoi Ting
- Nanomab Technology (UK) Limited, Borehamwood, Hertfordshire, UK
| | - James Spicer
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, Great Maze Pond, London, UK
| | - Vicky Goh
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
- Department of Radiology, Guy's and St. Thomas' NHS Foundation Trust, St Thomas' Hospital, London, UK
| | - Gary J R Cook
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK.
- King's College London & Guy's and St. Thomas' PET Centre, St Thomas' Hospital, London, UK.
| |
Collapse
|
3
|
Wu Y, Wang H, Gu Y, Zhang Y, Li G, Huang Y, Cao M, Chen X, Guan Y, Xu D, Wei W, Xie F. Head-to-head comparison of peptide-based and nanobody-based radiotracers in detecting PD-L1 expression in non-small cell lung cancer. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07316-w. [PMID: 40338301 DOI: 10.1007/s00259-025-07316-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/24/2025] [Indexed: 05/09/2025]
Abstract
BACKGROUND Immunotherapy based on programmed cell death protein receptor 1 and its ligand (PD-1/PD-L1) has become an important method for treating non-small cell lung cancer (NSCLC). Peptide-based and nanobody-based PET tracers offer potential advantages in PD-L1 detection, yet their comparative tumor uptake and biodistribution remain unclear. This study aimed to evaluate and compare [68Ga]Ga-DOTA-WL12 (a peptide-based tracer) and [68Ga]Ga-NOTA-RW102 (a nanobody-based tracer) in assessing PD-L1 expression in primary and metastatic NSCLC, providing insights for future radiotracer design and theranostic applications. METHODS Ten patients diagnosed with NSCLC underwent [68Ga]Ga-DOTA-WL12 and [68Ga]Ga-NOTA-RW102 PET/CT scans, with four of these patients also receiving [18F]FDG PET/CT scans. The tracer uptakes, quantified by maximum standardized uptake value (SUVmax), mean standardized uptake value (SUVmean), and target-to-background ratio (TBR), were compared between [68Ga]Ga-DOTA-WL12 and [68Ga]Ga-NOTA-RW102 PET/CT. RESULTS DOTA-WL12 and NOTA-RW102 exhibited favorable binding affinities with PD-L1, with equilibrium dissociation constant (KD) values of 0.2 nM and 0.0047 nM, respectively. Subsequent human studies revealed significant variations (P < 0.05) in the uptake of [68Ga]Ga-DOTA-WL12 and [68Ga]Ga-NOTA-RW102 across the liver (SUVmean: 20.43 ± 4.26 vs. 6.12 ± 1.36, p = 0.015), kidney (SUVmean: 2.40 ± 0.34 vs. 22.37 ± 2.88, P = 0.015), spleen (SUVmean: 2.44 ± 0.67 vs. 18.49 ± 3.90, P = 0.015), and lung background (SUVmean: 0.18 ± 0.12 vs. 1.09 ± 0.29, P = 0.015). Meanwhile, we found that the correlation between SUVmax and PD-L1 TPS was significantly stronger with [68Ga]Ga-DOTA-WL12 compared to [68Ga]Ga-NOTA-RW102 (P < 0.0001, r = 0.9471 vs. P = 0.0241, r = 0.5235). CONCLUSION The uptake of peptide-based [68Ga]Ga-DOTA-WL12 was more strongly correlated with PD-L1 TPS in primary and metastatic tumor lesions compared to [68Ga]Ga-NOTA-RW102. They also displayed different distribution, suggesting that peptide-based and nanobody-based radiotracers may have different clinical implications, particularly in radiotherapy.
Collapse
Affiliation(s)
- Yanfei Wu
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Hao Wang
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Yue Gu
- Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, China
| | - You Zhang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Guanglei Li
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuan Huang
- Rehabilitation Department, Huashan Hospital, Fudan University, Shanghai, China
| | - Min Cao
- Department of Thoracic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xiaofeng Chen
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yihui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Dong Xu
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China.
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
4
|
Zhang J, Song Z, Zhang Y, Zhang C, Xue Q, Zhang G, Tan F. Recent advances in biomarkers for predicting the efficacy of immunotherapy in non-small cell lung cancer. Front Immunol 2025; 16:1554871. [PMID: 40406096 PMCID: PMC12095235 DOI: 10.3389/fimmu.2025.1554871] [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: 01/03/2025] [Accepted: 04/18/2025] [Indexed: 05/26/2025] Open
Abstract
Lung cancer continues to be the primary cause of cancer-related deaths globally, with non-small cell lung cancer (NSCLC) accounting for approximately 85% of all instances. Recently, immune checkpoint inhibitors (ICIs) have transformed the treatment approach for NSCLC, however, only a subset of patients experiences significant benefits. Therefore, identifying reliable biomarkers to forecast the efficacy of ICIs is crucial for ensuring the safety and effectiveness of treatments, becoming a major focus of current research efforts. This review highlights the recent advances in predictive biomarkers for the efficacy of ICIs in the treatment of NSCLC, including PD-L1 expression, tertiary lymphoid structures (TLS), tumor-infiltrating lymphocytes (TILs), tumor genomic alterations, transcriptional signatures, circulating biomarkers, and the microbiome. Furthermore, it underscores the pivotal roles of liquid biopsy, sequencing technologies, and digital pathology in biomarker discovery. Special attention is given to the predictive value of TLS, circulating biomarkers, and transcriptional signatures. The review concludes that the integration of multiple biomarkers holds promise for achieving more accurate efficacy predictions and optimizing personalized immunotherapy strategies. By providing a comprehensive overview of the current progress, this review offers valuable insights into biomarker-based precision medicine for NSCLC and outlines future research directions.
Collapse
Affiliation(s)
- Jiacheng Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zehao Song
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuanjie Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chentong Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guochao Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
5
|
Peeters E, van Genugten EAJ, Heskamp S, de Vries IJM, van Herpen C, Koenen HJPM, Kneilling M, van der Post RS, van Dop WA, Westdorp H, Aarntzen E. Exploring molecular imaging to investigate immune checkpoint inhibitor-related toxicity. J Immunother Cancer 2025; 13:e011009. [PMID: 40341021 PMCID: PMC12060888 DOI: 10.1136/jitc-2024-011009] [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: 12/09/2024] [Accepted: 04/18/2025] [Indexed: 05/10/2025] Open
Abstract
Immune checkpoint inhibitors (ICI) boost the endogenous anticancer immunity, evoking long-lasting anticancer responses in a subset of patients with solid tumors. Simultaneously, ICI are also associated with serious toxicities, impacting treatment duration and the quality of life. The proposed processes underlying ICI-related toxicity include T-cell activation and recruitment to non-tumor tissues, involvement of other immune cells and fibroblasts and the host' microbiome composition. However, the exact mechanisms of these processes remain incompletely understood, hindering clinicians' ability to predict and identify ICI-related toxicity in the early stages of treatment. Molecular imaging may play a role as a non-invasive biomarker, providing a tool to study ICI-related toxicity. This review discusses the applications of molecular imaging to answer questions regarding the mechanisms, detection, and prediction of ICI-related toxicity. Potential targets and the current state of development of suitable imaging techniques are discussed.
Collapse
Affiliation(s)
- Eva Peeters
- Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Sandra Heskamp
- Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carla van Herpen
- Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hans J P M Koenen
- Laboratory of Medical Immunology, Radboud University Medical Center, Nijmegen, Gelderland, The Netherlands
| | - Manfred Kneilling
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University of Tübingen, Tubingen, Baden-Württemberg, Germany
- Department of Dermatology, University of Tübingen, Tubingen, Baden-Württemberg, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls Universität Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Rachel S van der Post
- Department of Pathology, Radboud University Medical Center, Nijmegen, Gelderland, The Netherlands
| | - Willemijn A van Dop
- Department of Gastroenterology, Radboud University Medical Center, Nijmegen, Gelderland, The Netherlands
| | - Harm Westdorp
- Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Erik Aarntzen
- Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Nuclear Medicine, Eberhard Karls Universität Tübingen, Tübingen, Baden-Württemberg, Germany
- Department of Nuclear Medicine and Molecular Imaging, University Medical Centre Groningen, Groningen, Groningen, The Netherlands
| |
Collapse
|
6
|
Huang Y, Zheng D, Li C, Pi X, Wang S, Li Z, Li Y, Liang Y. Synthesis and preclinical evaluation of an Al 18F radio-fluorinated bivalent PD-L1 nanobody. Eur J Med Chem 2025; 289:117487. [PMID: 40085976 DOI: 10.1016/j.ejmech.2025.117487] [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: 01/22/2025] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Immunotherapy targeting the programmed death 1/programmed death ligand 1 (PD-1/PD-L1) pathway has achieved remarkable clinical success, but there is a shortage of effective approaches for screening suitable patients. Recently developed PD-L1 nanobody probes have limitations, including limited availability of radionuclides, short tumor retention times, and accumulation in non-target organs. To enhance tumor retention and improve tumor-to-normal tissue contrast, we herein report the synthesis and preclinical evaluation of two Al18F-labeled bivalent PD-L1 nanobody probes ([18F]TzTCO-BINb109 and [18F]RESCA-BINb109). Preliminary results indicated that [18F]TzTCO-BINb109 had a greater affinity for PD-L1 and better stability than [18F]RESCA-BINb109. Micro-PET/CT revealed that [18F]TzTCO-BINb109 uptake in A549-PDL1 tumors peaked at 240 min post-injection (3.19 ± 0.49 %ID/g) and demonstrated sustained retention without in vivo defluorination. In contrast, [18F]RESCA-BINb109 exhibited shorter tumor retention (at 60 and 240 min, 2.08 ± 0.22 and 1.37 ± 0.26 %ID/g, respectively) and significant defluorination in vivo. Ex vivo biodistribution studies revealed that the tumor uptake of [18F]TzTCO-BINb109 was consistent with the PET results, with the highest uptake by A549-PDL1 tumor cells (3.43 ± 0.94 %ID/g) compared with H1975 (0.93 ± 0.18 %ID/g) and A549 (0.68 ± 0.12 %ID/g) cells observed at 240 min post-injection. Compared with the previously reported monomeric PD-L1-targeting nanobody probe, [68Ga]NOTA-Nb109, [18F]TzTCO-BINb109 demonstrated enhanced tumor uptake, prolonged retention, and superior tumor-to-normal tissue contrast, contributing to higher imaging quality. These results confirmed that the bivalent PD-L1 nanobody radioligand, [18F]TzTCO-BINb109, was a promising diagnostic probe for PD-L1 detection, efficacy evaluation, and prescription optimization of immune checkpoint inhibitor therapies.
Collapse
Affiliation(s)
- Yong Huang
- Department of Nuclear Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - Dongye Zheng
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China
| | - Chengze Li
- Department of Nuclear Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - Xixuan Pi
- Department of Traditional Chinese Medicine, Shenzhen Futian District Maternity & Child Healthcare Hospital, Shenzhen, 518000, China
| | - Senlin Wang
- Department of Nuclear Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - Zhongjing Li
- Department of Nuclear Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - Yiluo Li
- Department of Nuclear Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - Ying Liang
- Department of Nuclear Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China.
| |
Collapse
|
7
|
Yang W, Ma SC, Fang Z, Liu Y, Zhang X, Wang F, Wang C, Wang Y, Wang X, Chen W, Luo H, Yang L, Zhang S, Zeng B, Liu Z, Ou Q, Cai J, Yeung SCJ, Cheng C. TP53-centric ctDNA complements PET/CT for non-invasive assessment of pathological complete response and survival after neoadjuvant immunochemotherapy in esophageal squamous cell carcinoma: a prospective cohort study. Int J Surg 2025; 111:3256-3268. [PMID: 40146232 PMCID: PMC12165533 DOI: 10.1097/js9.0000000000002341] [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: 01/08/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025]
Abstract
BACKGROUND Accurate assessment of pathologic complete response (pCR) after neoadjuvant immunochemotherapy (NICT) is crucial to implement active surveillance or tailor therapeutic strategies for esophageal squamous cell carcinoma (ESCC), while reliable non-invasive methods for pCR prediction are lacking. We aimed to evaluate the potential of integrating circulating tumor DNA (ctDNA) and PET/CT for predicting pCR to NICT for ESCC. METHODS A total of 123 eligible patients were enrolled, including 68 patients from our prospective clinical trial (ChiCTR2000028900) and a real-world study (NCT04822103) that formed the discovery cohort, as well as 55 patients from another clinical trial (ChiCTR2100051763) comprising the validation cohort. Blood samples for ctDNA sequencing and PET/CT metrics were collected before and after NICT. RESULTS The ctDNA status and PET/CT parameters at the post-NICT stage rather than the pre-NICT stage significantly differentiated pCR from non-pCR patients. ctDNA and PET/CT synergistically enhanced the prediction of pCR from perspectives of sensitivity and specificity, respectively. The model integrating ctDNA concentration and mean standardized uptake value (SUVmean) demonstrated area under curves (AUCs) of 0.860 in the discovery cohort and 0.798 in the validation cohort for pCR prediction and stratified patients into high- and low-risk groups with differential survival prospects. The key gene modules converged on TP53 as the core mutation for pCR prediction, among which those located in the exon regions contributed the most to its predictive capacity. The model constructed based on TP53 mutation and SUVmean differentiated pCR from non-pCR with comparable performance to the model based on PET/CT and the overall ctDNA concentration. CONCLUSION The combination of post-treatment TP53 -centric ctDNA and PET/CT synergistically enhances the prediction of pCR following NICT in ESCC patients, indicating the potential to inform clinical decision-making for these patients.
Collapse
Affiliation(s)
- Weixiong Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Si-Cong Ma
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zengli Fang
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yao Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xin Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Fang Wang
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chenxuan Wang
- Nanjing Geneseeq Technology Inc, Nanjing, Jiangsu, China
| | - Yuze Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaoyan Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenfang Chen
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hui Luo
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lingling Yang
- Nanjing Geneseeq Technology Inc, Nanjing, Jiangsu, China
| | - Shuishen Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Bo Zeng
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhenguo Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qiuxiang Ou
- Nanjing Geneseeq Technology Inc, Nanjing, Jiangsu, China
| | - Junchao Cai
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Sai-Ching Jim Yeung
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chao Cheng
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
8
|
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
|
9
|
Liu J, Cheng P, Xu C, Pu K. Molecular probes for in vivo optical imaging of immune cells. Nat Biomed Eng 2025; 9:618-637. [PMID: 39984703 DOI: 10.1038/s41551-024-01275-7] [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: 05/23/2023] [Accepted: 09/23/2024] [Indexed: 02/23/2025]
Abstract
Advancing the understanding of the various roles and components of the immune system requires sophisticated methods and technology for the detection of immune cells in their natural states. Recent advancements in the development of molecular probes for optical imaging have paved the way for non-invasive visualization and real-time monitoring of immune responses and functions. Here we discuss recent progress in the development of molecular probes for the selective imaging of specific immune cells. We emphasize the design principles of the probes and their comparative performance when using various optical modalities across disease contexts. We highlight molecular probes for imaging tumour-infiltrating immune cells, and their applications in drug screening and in the prediction of therapeutic outcomes of cancer immunotherapies. We also discuss the use of these probes in visualizing immune cells in atherosclerosis, lung inflammation, allograft rejection and other immune-related conditions, and the translational opportunities and challenges of using optical molecular probes for further understanding of the immune system and disease diagnosis and prognosis.
Collapse
Affiliation(s)
- Jing Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Penghui Cheng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Cheng Xu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
10
|
Zhou X, Yan S, Li D, Zhu H, Liu B, Liu S, Zhao W, Yang Z, Wu N, Li N. Radiolabelled anti-PD-L1 peptide PET/CT in predicting the efficacy of neoadjuvant immunotherapy combined with chemotherapy in resectable non-small cell lung cancer. Ann Nucl Med 2025; 39:364-372. [PMID: 39673015 DOI: 10.1007/s12149-024-02009-0] [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: 06/26/2024] [Accepted: 12/02/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND This study aimed to evaluate the predictive value of baseline PD-L1 targeted peptide 68Ga-NOTA-WL12 PET/CT in neoadjuvant immunotherapy combined with chemotherapy of resectable NSCLC. METHODS Patients with resectable NSCLC (n = 20) enrolled in this prospective study received baseline paired 68Ga-NOTA-WL12 PET/CT and 18F-FDG PET/CT. After 2-4 cycles of toripalimab plus nab-paclitaxel and cisplatin, surgery was performed if R0 resection was available. The major pathologic response (MPR) state of the post-operative specimen was recorded. The imaging parameters of the 68Ga-NOTA-WL12 PET/CT, 18F-FDG PET/CT and CT between the MPR and non-MPR groups and their predictive efficacy of MPR were compared. RESULTS Among 20 patients, 17 patients underwent surgery, 10 achieved an MPR and 7 did not. The SUVmax and tumour-to-blood pool (TBR) of baseline 68Ga-NOTA-WL12 in the MPR group were higher than those in the non-MPR group, and the difference in TBR was statistically significant. The ΔSULpeak% of 18F-FDG exhibited differences between the MPR and non-MPR groups with no significance. Baseline 18F-FDG PET/CT parameters and ΔD% failed to differentiate the two groups. The areas under the ROC curves of SUVmax, TBR in 68Ga-NOTA-WL12 PET/CT, ΔD% and ΔSULpeak% in 18F-FDG PET/CT were 0.76, 0.79, 0.71 and 0.80, respectively, in predicting MPR. CONCLUSION Baseline 68Ga-NOTA-WL12 PET/CT has a potential to predict the pathological response of neoadjuvant immunotherapy combined with chemotherapy in patients with resectable NSCLC, whose efficacy is comparable to that of therapy evaluations employing baseline and follow-up CT and 18F-FDG PET/CT examinations. TRIAL REGISTRATION NCT04304066, registered 13 November 2020, https://register. CLINICALTRIALS gov/prs/app/action/SelectProtocol?sid=S000AEI9&selectaction=Edit&uid=U000503E&ts=2&cx=-awajet .
Collapse
Affiliation(s)
- Xin Zhou
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Shi Yan
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Dan Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Hua Zhu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Bing Liu
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Shiwei Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Wei Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Zhi Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China.
| | - Nan Wu
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China.
| | - Nan Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China.
| |
Collapse
|
11
|
Liang Y, Xie M, Zang X, Zhang X, Xue X. Evaluation of ImmunoPET in the efficacy and prognosis of immunotherapy for lung cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189289. [PMID: 39999945 DOI: 10.1016/j.bbcan.2025.189289] [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: 11/28/2024] [Revised: 02/12/2025] [Accepted: 02/16/2025] [Indexed: 02/27/2025]
Abstract
Advances in immune oncology have established immunotherapy as the first-line standard treatment for lung cancer; however, its efficacy remains limited to a subset of patients. Developing predictive biomarkers within the tumor microenvironment (TME) to assess the efficacy and prognosis of immunotherapy can enhance drug development and treatment strategies. Immuno-positron emission tomography (ImmunoPET) non-invasively visualizes the biological distribution of key targets in the TME using highly specific, radiolabeled tracers. PET imaging of the TME can serve as a reliable biomarker for predicting and monitoring responses to immune therapy, complementing existing immunohistochemical techniques. This review will focus on the development of ImmunoPET biomarkers, as well as the application of corresponding tracers and radionuclides in lung cancer. We will focus on available clinical tracers and those under development, outlining each TME target and its clinical validation for tumor immunotherapy efficacy and prognosis, while discussing the latest advances that may enhance ImmunoPET in future.
Collapse
Affiliation(s)
- Yiran Liang
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Mei Xie
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Xuefeng Zang
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Xin Zhang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong 261000, China
| | - Xinying Xue
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China.
| |
Collapse
|
12
|
Nie H, Peng L, Yang T, Chen J, Xie C, Xue L, Zhang D, Wu R, Zhang X, Zha Z. 68Ga-Labeled Peptide for Noninvasive Quantifying Tumor Exposure of PD-L1 Therapeutics. ACS OMEGA 2025; 10:12495-12504. [PMID: 40191352 PMCID: PMC11966317 DOI: 10.1021/acsomega.4c11396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/02/2025] [Accepted: 03/10/2025] [Indexed: 04/09/2025]
Abstract
PURPOSE Targeting the programmed death protein 1/programmed death-ligand 1 (PD-1/PD-L1) immune checkpoint blockade therapy plays a critical role in cancer therapy. However, not all patients benefit from this approach, with PD-L1 expression levels being a significant contributing factor. Positron emission tomography (PET) imaging of PD-L1 offers a noninvasive, whole-body, and dynamic assessment of its expression. This study aims to develop a novel peptide-based PD-L1 tracer, [68Ga]HF12, to quantitatively evaluate PD-L1 expression in tumors, thereby offering clinical guidance. METHODS HF12 was successfully synthesized and radiolabeled with 68Ga to yield [68Ga]HF12. In vitro binding assays confirmed the specific binding affinity of HF12 for PD-L1 using CHO-hPD-L1 and CHO cell lines. Subsequent in vivo positron emission tomography (PET) imaging and biodistribution studies assessed [68Ga]HF12 for monitoring PD-L1 expression levels in tumor-bearing mice, including those subjected to immunotherapy. Furthermore, PD-L1 expression in tumor tissues was evaluated by using autoradiography, Western blotting, and immunohistochemical (IHC) analysis. RESULTS The synthesis of [68Ga]HF12 was successfully achieved with a radiochemical purity and yield exceeding 95%. Cellular uptake studies indicated that [68Ga]HF12 demonstrated both high specificity and significant uptake in PD-L1-positive CHO-hPD-L1 cells. Micro-PET imaging and biodistribution studies revealed that [68Ga]HF12 was preferentially accumulated in CHO-hPD-L1 tumors compared to PD-L1-negative CHO tumors. Treatment with Atezolizumab resulted in a significant reduction in [68Ga]HF12 uptake in CHO-hPD-L1 tumors relative to pretreatment levels, whereas no significant changes were observed in the phosphate-buffered saline (PBS) control group. Subsequent biodistribution studies, along with Western blotting and immunohistochemical analyses, confirmed that PD-L1 expression levels in tumors were reduced following immunotherapy, consistent with the results obtained from PET imaging. CONCLUSIONS [68Ga]HF12 was successfully synthesized as a radiotracer for noninvasive quantitative PET imaging of PD-L1 expression levels. This radiotracer exhibited the potential to quantify PD-L1 expression across various tumors, thereby facilitating the prediction of patient response to anti-PD-1 and anti-PD-L1 immunotherapies and monitoring therapeutic efficacy.
Collapse
Affiliation(s)
- Hui Nie
- Department
of Nuclear Medicine, The First Affiliated
Hospital of Sun Yat-sen University, 58# Zhongshan Er Road, Guangzhou 510080, Guangdong, China
| | - Lei Peng
- Department
of Ultrasound Medicine, The First People’s
Hospital of FoShan, 81#
Lingnan Avenue North, Foshan 528000, Guangdong, China
| | - Tianhong Yang
- Department
of Nuclear Medicine, The First Affiliated
Hospital of Sun Yat-sen University, 58# Zhongshan Er Road, Guangzhou 510080, Guangdong, China
| | - Junyu Chen
- College
of Environmental Science and Engineering, North China Electric Power University, Beijing 102206, China
| | - Chengde Xie
- College
of Environmental Science and Engineering, North China Electric Power University, Beijing 102206, China
| | - Lingyu Xue
- Department
of Nuclear Medicine, The First Affiliated
Hospital of Sun Yat-sen University, 58# Zhongshan Er Road, Guangzhou 510080, Guangdong, China
| | - Dake Zhang
- Department
of Nuclear Medicine, The First Affiliated
Hospital of Sun Yat-sen University, 58# Zhongshan Er Road, Guangzhou 510080, Guangdong, China
| | - Renbo Wu
- Department
of Nuclear Medicine, The First Affiliated
Hospital of Sun Yat-sen University, 58# Zhongshan Er Road, Guangzhou 510080, Guangdong, China
| | - Xiangsong Zhang
- Department
of Nuclear Medicine, The First Affiliated
Hospital of Sun Yat-sen University, 58# Zhongshan Er Road, Guangzhou 510080, Guangdong, China
| | - Zhihao Zha
- Department
of Nuclear Medicine, The First Affiliated
Hospital of Sun Yat-sen University, 58# Zhongshan Er Road, Guangzhou 510080, Guangdong, China
| |
Collapse
|
13
|
Chen Y, Hu Y, Li A, Zhang G, Guo D, Yao X, Zeng B, Tang G, Jiang B, Jiang L. Preclinical and first‑in‑human evaluation of [ 68Ga]Ga-DOTA-PEG 2-Asp 2-PDL1P PET imaging to assess tumor PD-L1 expression. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07173-7. [PMID: 40029371 DOI: 10.1007/s00259-025-07173-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 02/17/2025] [Indexed: 03/05/2025]
Abstract
PURPOSE PD-L1 PET imaging can provide a non-invasively and real-time assessment of PD-L1 expression status at tumor sites. This study aimed to evaluate the targeting efficacy and biodistribution of a novel peptide-based PD-L1 PET agent, [68Ga]Ga-DOTA-PEG2-Asp2-PDL1P, in preclinical studies and human participants. METHODS [68Ga]Ga-DOTA-PEG2-Asp2-PDL1P was synthesized and the probe stability was analyzed in vitro and in vivo. Cellular uptake of the probe was evaluated using tumor cell lines with different PD-L1 expression levels. Small animal PET imaging and semi-quantitative studies were conducted in PC3, H1975 and A549 tumor-bearing mice models, with tumor PD-L1 expression confirmed through immunofluorescence and immunohistochemistry. Furthermore, [68Ga]Ga-DOTA-PEG2-Asp2-PDL1P PET imaging was performed in 1 healthy volunteer and 14 lung cancer patients to assess biodistribution and PD-L1 expression at tumor sites. RESULTS [68Ga]Ga-DOTA-PEG2-Asp2-PDL1P exhibited a radiochemical purity of > 99% and had good stability both in vitro and in vivo. In vitro cellular uptake and in vivo small animal PET imaging revealed the probe binding to PD-L1 with high affinity and specificity, consistent with the results of immunofluorescence and immunohistochemistry. In the clinical study involving 15 participants, [68Ga]Ga-DOTA-PEG2-Asp2-PDL1P was proven safe with demonstrating low uptake in normal organs and physiologically excreting via the urinary system. Lung cancer patients with high PD-L1 expression (TPS 70-90%) exhibited higher tumor uptake and tumor-to-background ratios than those with negative or low PD-L1 expression (TPS < 1-10%), with SUVmax of 1.89-2.27 vs. 0.87-1.01, tumor-to-lung ratios of 4.73-7.68 vs. 1.61-2.35, and tumor-to-muscle ratios of 6.73-12.61 vs. 4.35-5.61. CONCLUSION [68Ga]Ga-DOTA-PEG2-Asp2-PDL1P showed promising as a PET agent to assess tumor PD-L1 expression in preclinical and first-in-human studies, offering a non-invasive, real-time and accurate tool to address clinical challenges in predicting and assessing the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Yang Chen
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yinting Hu
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ao Li
- Department of Pulmonary Surgery, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Guojin Zhang
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Danyi Guo
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xinchao Yao
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Baozhen Zeng
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ganghua Tang
- Nanfang PET Center, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China.
| | - Benyuan Jiang
- Department of Pulmonary Surgery, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
- Department of Pulmonary Surgery, Guangdong Provincial People's Hospital, Guangdong Lung Cancer Institute, Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510080, China.
| | - Lei Jiang
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital, Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510080, China.
| |
Collapse
|
14
|
Vinjamuri S, Pant V. Demystifying the Role of Immuno PET-CT in Non-Small Cell Lung Cancer: Clinical Value and Research Trends. Semin Nucl Med 2025; 55:212-220. [PMID: 40016063 DOI: 10.1053/j.semnuclmed.2025.02.004] [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: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 03/01/2025]
Abstract
The management of Lung cancer, especially non-small cell lung cancer has undergone a paradigm shift recently with the advent of new treatment approaches including focused radiotherapy as well as evolution of a newer class of immunotherapy agents. Treatment efficacy and survival rates have improved and it is now even more important that patients are selected for appropriate interventions on the basis of a comprehensive assessment including a range of imaging as well as in-vitro tests such as immunohistochemistry. A new class of tracers targeting programmed cell death such as PD1 and PDL1 (broadly classed as Immuno PET) are being increasingly used in the molecular characterisation of patients deemed resistant to standard treatment approaches and being considered for additional interventions such as immunotherapy. In this review, we review the latest evidence in the field and propose a summary of clinical usefulness and provide a review of the research trends in this exciting and evolving field.
Collapse
Affiliation(s)
- Sobhan Vinjamuri
- Department of Nuclear Medicine, Royal Liverpool University Hospital, Liverpool, UK.
| | - Vineet Pant
- Department of Nuclear Medicine, Royal Liverpool University Hospital, Liverpool, UK
| |
Collapse
|
15
|
Richlitzki C, Manapov F, Holzgreve A, Rabe M, Werner RA, Belka C, Unterrainer M, Eze C. Advances of PET/CT in Target Delineation of Lung Cancer Before Radiation Therapy. Semin Nucl Med 2025; 55:190-201. [PMID: 40064578 DOI: 10.1053/j.semnuclmed.2025.02.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025]
Abstract
In the clinical management of lung cancer, radiotherapy remains a cornerstone of multimodal treatment strategies, often used alongside surgery or in combination with systemic therapies such as chemotherapy, tyrosine kinase inhibitors, and immune checkpoint inhibitors. While conventional imaging modalities like computed tomography (CT) and magnetic resonance imaging (MRI) continue to play a central role in staging, response assessment, and radiotherapy planning, advanced imaging techniques, particularly [18F]FDG PET/CT, are being increasingly integrated into routine clinical practice. These advanced techniques address the limitations of standard imaging by providing insight into molecular and metabolic tumor characteristics, enabling precise tumor visualization, accurate target volume delineation, and early treatment response assessment. This review examines the role of radiotherapy in the multidisciplinary management of lung cancer, detailing current concepts of morphological and functional imaging for staging and treatment planning. It also highlights the growing importance of PET-based radiotherapy planning, emphasizing its contributions to target volume definition and predictive value for treatment outcomes. Recent methodological advances, including the integration of artificial intelligence (AI), radiomics, technical innovations, and novel PET ligands, are discussed, highlighting their potential to improve the precision, efficacy, and personalization of lung cancer radiotherapy planning.
Collapse
Affiliation(s)
- Cedric Richlitzki
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Farkhad Manapov
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Adrien Holzgreve
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany; Ahmanson Translational Theranostics Division, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Moritz Rabe
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Rudolf Alexander Werner
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany; The Russell H Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins School of Medicine, Baltimore, MD
| | - Claus Belka
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany; German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany; Comprehensive Pneumology Center Munich, Member of the German Center for Lung Research, Munich, Germany; Bavarian Cancer Research Center, Munich, Germany
| | - Marcus Unterrainer
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany; Die Radiologie, Munich, Germany
| | - Chukwuka Eze
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
16
|
Ayeni A, Evbuomwan O, Vangu MDTW. The Role of [ 18F]FDG PET/CT in Monitoring of Therapy Response in Lung Cancer. Semin Nucl Med 2025; 55:175-189. [PMID: 40021362 DOI: 10.1053/j.semnuclmed.2025.02.002] [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: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 03/03/2025]
Abstract
Lung cancer remains a leading cause of cancer deaths worldwide, with an all stage 5-year relative survival rate of less than 30%. Multiple treatment strategies are available and continue to evolve, with therapy primarily tailored to the type and stage of the disease. Accurate monitoring of therapy response is crucial for optimizing treatment outcomes. PET/CT imaging with [18F]FDG has become the standard of care across various phases of lung cancer management due to its ability to assess metabolic activity. This review underscores the pivotal role of [18F]FDG PET/CT in evaluating therapy response in lung cancer, particularly in non-small cell lung cancer (NSCLC). It examines conventional response criteria and their adaptations in the era of immunotherapy, highlighting the value of integrating metabolic imaging with established criteria to improve treatment assessment and guide clinical decisions. The potential of non-[18F]FDG PET tracers targeting diverse biological pathways to provide deeper insights into tumor biology, therapy response and predictive outcomes is also explored. Additionally, the emerging role of radiomics in enhancing treatment efficacy assessment and improving patient management is briefly highlighted. Despite the challenges in the routine clinical application of various metabolic response criteria, [18F]FDG PET/CT remains a crucial tool in monitoring therapy response in lung cancer. Ongoing advancements in therapeutic strategies, radiopharmaceuticals, and imaging techniques continue to drive progress in lung cancer management, promising improved patient outcomes.
Collapse
Affiliation(s)
- Akinwale Ayeni
- Division of Nuclear Medicine, Department of Radiation Sciences, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, South Africa; Nuclear Medicine, Klerksdorp/Tshepong Hospital Complex, Klerksdorp, North West Province, South Africa; Division of Nuclear Medicine, Department of Radiation Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| | - Osayande Evbuomwan
- Department of Nuclear Medicine, Faculty of Health Sciences, University of The Free State, Bloemfontein, South Africa
| | - Mboyo-Di-Tamba Willy Vangu
- Division of Nuclear Medicine, Department of Radiation Sciences, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
17
|
Ma X, Hu B, Zhou X, Wang L, Chen H, Xie F, Zhu H, Jia B, Yang Z. Development and First-in-Human evaluation of a Site-Specific [ 18F]-Labeled PD-L1 nanobody PET radiotracer for noninvasive imaging in NSCLC. Bioorg Chem 2025; 156:108222. [PMID: 39889552 DOI: 10.1016/j.bioorg.2025.108222] [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: 12/24/2024] [Revised: 01/13/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Immunohistochemistry (IHC) for PD-L1 detection is limited by its invasiveness and heterogeneity of tumors. To address these challenges, a new PD-L1-targeted nanobody-based immune-PET radiotracer [18F]AlF-APN09 was developed using the site-specific radiolabeling method with the complexing agent (Mal-RESCA) under mild conditions. [18F]AlF-APN09 was prepared at room temperature (pH 4.6-4.8) within 20 min with satisfactory radiochemical yields (45.8 ± 4.48 %, non-decay corrected), high radiochemical purity (>98 %) and moderate apparent molar activity (15-35 GBq/μmol), and remained stable in both PBS and 5 % HSA after 4 h (>90 %). Cell uptake studies indicated variable levels of surface PD-L1 expression in the following order: A549PD-L1 > H1975 > A549. In micro-PET/CT imaging, A549PD-L1 and H1975 tumors were distinctly visualized in a 6.0:1 and 3.2:1 ratios over PD-L1-negative A549 tumors in vivo. Ex vivo biodistribution studies showed tumor uptake values of 6.47 ± 1.06 %ID/g (A549PD-L1) and 2.27 ± 0.19 %ID/g (H1975), significantly higher than 0.90 ± 0.28 %ID/g in A549 tumors. The estimated effective radiation dose in humans was 8.65E-03 mSv/MBq, lower than that of conventional [18F]FDG. First-in-human imaging was conducted on a single resectable non-small cell lung cancer (NSCLC) subject without any adverse reactions. The radiotracer exhibited renal excretion with minimal hepatobiliary clearance. Tumor uptake reached SUVmax 4.20 at 2 h post-injection, demonstrating high contrast and rapid clearance. After PD-1 inhibitor immunotherapy and chemotherapy, the subject showed a therapeutic response and postoperative pathological specimens confirmed a major pathological response (MPR). These results suggest that we have successfully developed a new PD-L1-targeted nanobody PET tracer using the site-specific labeling method with the complexing agent (Mal-RESCA) within 20 min under mild conditions and [18F]AlF-APN09 is a promising noninvasive PET radiotracer for visualizing PD-L1 expression in tumors, offering rapid tumor targeting, excellent signal-to-noise ratios, and favorable clearance properties.
Collapse
Affiliation(s)
- Xiaopan Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142 China; Department of Nuclear Medicine, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441138 China
| | - Biao Hu
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191 China
| | - Xin Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142 China
| | - Lei Wang
- Department of Laboratory Medicine, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441138 China
| | - Hui Chen
- Department of Nuclear Medicine, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441138 China
| | - Fei Xie
- Department of Nuclear Medicine, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441138 China
| | - Hua Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142 China.
| | - Bing Jia
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191 China.
| | - Zhi Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142 China.
| |
Collapse
|
18
|
Lau D, Elliott T. Imaging antigen processing and presentation in cancer. IMMUNOTHERAPY ADVANCES 2025; 5:ltaf002. [PMID: 40265075 PMCID: PMC12012451 DOI: 10.1093/immadv/ltaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 02/04/2025] [Indexed: 04/24/2025] Open
Abstract
Introduction Antigen processing and presentation are vital processes of the adaptive immunity. These processes involve a series of intracellular and extracellular events, including the enzymology within cells during antigen processing, the loading and presentation of antigenic peptides on major histocompatibility complexes, the recruitment of T cells, their interaction with antigen-presenting cells, and the expression of adhesion, co-stimulatory and co-inhibitory molecules at the T cell immunological synapse. These events collectively fine-tune and sustain antigen recognition and T cell function. Dysregulation of this machinery can profoundly impact the efficacy of cancer immunotherapy. Imaging technologies have emerged as powerful tools for elucidating the mechanisms underlying antigen processing and presentation. By providing complementary perspectives into the cellular and molecular interactions at play, imaging has significantly enhanced our understanding of these complex immunological events in cancer. Such insights can improve the monitoring of immunotherapy responses, facilitate the identification of effective treatments, and aid in predicting patient outcomes. Methods This review explores the role of imaging in studying antigen processing and presentation in the context of cancer. Conclusion It highlights key considerations for developing imaging tools and biomarkers to detect components of these pathways. Additionally, it examines the strengths and limitations of various imaging approaches and discusses their potential for clinical translation.
Collapse
Affiliation(s)
- Doreen Lau
- Centre for Inflammation Research and Translational Medicine, Department of Life Sciences, Division of Biosciences, Brunel University London, London, United Kingdom
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Tim Elliott
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
19
|
Ning YD, Song YX, He YQ, Li H, Liu SY. Discordant Responses Between Imaging Examination and Surgical Pathology of Head and Heck Squamous Cell Carcinoma After Neoadjuvant Immunotherapy Combined With Chemotherapy. World J Oncol 2025; 16:59-69. [PMID: 39850520 PMCID: PMC11750755 DOI: 10.14740/wjon1973] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/21/2024] [Indexed: 01/25/2025] Open
Abstract
Background We here investigated the value of imaging examination in evaluating tumor remission-based surgery in patients with head and neck squamous cell carcinoma (HNSCC), who had undergone neoadjuvant immunotherapy combined with chemotherapy (NICC). Methods HNSCC patients who underwent NICC and surgery from May 2021 to September 2023 were retrospectively analyzed. All patients had to undergo imaging examination evaluation, including enhanced computed tomography (CT) and enhanced magnetic resonance (MR) imaging before and after NICC. Data related to clinical parameters, complete response of the primary site (PrCR), complete response of the primary site and the lymph node (PLCR), complete response of the lymph node (LCR), and tumor response (TR), were gathered. The paired Chi-square test and t-test were conducted to analyze the differences in responses between imaging examination and pathology. Binary logistic regression was applied to analyze the relevant clinical factors of differences in responses. Results In total, data of 41 patients were included in this study. Significant discordant responses were observed between enhanced CT, magnetic resonance imaging (MRI), and pathology in PrCR (4.9%, 7.3% vs. 41.5%), LCR (12.2%, 7.3% vs. 53.7%), PLCR (0%, 0% vs. 31.7%), and TR (severe 29.3%,17.1% vs. 25.61%) (P < 0.05). Patients with hypopharyngeal cancer (odds ratio (OR): 7.04), oral cancer (OR: 3.64), higher neutrophil to lymphocyte ratio (NLR) (OR: 2.05), and earlier T stage (OR: 0.71) exhibited a larger response difference between enhanced CT and pathology. Patients with younger age (OR: 0.79) hypopharyngeal cancer (OR: 22.81), oral cancer (OR: 2.65), higher NLR (OR: 19.47), and earlier T stage (OR: 0.29) exhibited a larger response difference between enhanced MR and pathology. Conclusions Discordant responses were noted between the imaging examination and surgical pathology of HNSCC after NICC. Hypopharyngeal cancer, higher NLR, and earlier T stage may predict a higher response difference.
Collapse
Affiliation(s)
- Yu Dong Ning
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- These authors contributed equally to this study
| | - Yi Xuan Song
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- These authors contributed equally to this study
| | - Yu Qin He
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Han Li
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shao Yan Liu
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
20
|
Zhou H, Bao G, Zhao J, Zhu X. Nuclear Molecular Imaging for Evaluating T Cell Exhaustion. Mol Pharm 2025; 22:103-112. [PMID: 39586059 DOI: 10.1021/acs.molpharmaceut.4c00970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
T cells are indispensable for the therapeutic efficacy of cancer immunotherapies, including immune checkpoint blockade. However, prolonged antigen exposure also drives T cells into exhaustion, which is characterized by upregulated inhibitory molecules, impaired effector functions, reduced cytotoxicity, altered metabolism, etc. Noninvasive monitoring of T cell exhaustion allows a timely identification of cancer patients that are most likely to benefit from immunotherapies. In this Review, we briefly explain the biological cascades underlying the modulation of inhibitory molecules, present a concise update on the nuclear molecular imaging tracers of T cell exhaustion, and then discuss the potential opportunities for future development.
Collapse
Affiliation(s)
- Huimin Zhou
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Guangfa Bao
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Department of Nuclear Medicine, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, China
| | - Jun Zhao
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Department of Anatomy, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiaohua Zhu
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| |
Collapse
|
21
|
Tao J, Wang F, Zeng Z, Zhou W, Wang Z, He C, Zhu J, Zhao C, Zhu H. Novel Peptide-Based 68Ga-Labeled Radiotracer for Preclinical Studies of TIM3 Expression. Mol Pharm 2025; 22:270-283. [PMID: 39513616 DOI: 10.1021/acs.molpharmaceut.4c00884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
T-cell immunoglobulin and mucin domain-3 (TIM3) is an immune checkpoint that plays a negative regulatory role in the immune response. TIM3-targeted drugs inhibit this negative regulation, thereby modulating the level of immune response activation. In the previous investigation, several peptides targeting TIM3 were identified through screening from a phage peptide library. In this research, three peptides were selected to construct the radioactive molecular probes according to the characteristic that targeting TIM3 drugs would lead to the increase of interferon-γ (IFN-γ) secretion. Molecular docking was performed to assess the binding properties of the selected peptides with the TIM3 protein. To further enhance the targeting properties, one of the peptides with a higher-affinity peptide was structurally modified. Then, 68Ga was used to construct the peptide probe 68Ga-DOTA-peptide by labeling the six peptides with 68Ga riboprobes, and the binding affinity and specificity were assessed using TIM3 overexpressing cell line A549TIM3 and the parental A549 cells. In addition, in Micro-PET/CT imaging, transfected model mice were dynamically imaged for 30 min after injection of 3.7-7.4 MBq 68Ga-DOTA-peptides via the tail vein. Meanwhile, the same dose of molecular probes was injected in the MC38 model (colorectal cancer in mice) and the CCRCC (clear cell renal cell carcinoma) xenografted model, followed by static scans at 15, 30, and 60 min postinjection. Finally, immunohistochemical (IHC) staining was performed to assess TIM3 expression in the dissected tumor tissues. The molecular docking results showed that the binding energy of P26 to TIM3 protein was -6.5 kcal/mol, which was lower than that of P24 to TIM3 protein, -3.6 kcal/mol, indicating that the affinity of P26 peptide to TIM3 protein was higher than that of P24 and P20 peptide. After structural modification of the P26 peptide, P26NH2, r-NH2, and P26X2 were obtained, and the above peptides were successfully constructed into six targeting TIM3 peptide probes by 68Ga labeling. Cellular uptake experiments demonstrated that 68Ga-DOTA-P26, 68Ga-DOTA-P26NH2, and 68Ga-DOTA-r-NH2 showed significantly higher uptake in A549TIM3 cells than in A549 cells and could be blocked by the unlabeled peptide. Micro-PET imaging experiments showed that the uptake of each probe in the A549TIM3 model tumor tissue was significantly higher than that in the A549 model tumor tissue, and a comparison of the tumor-to-cardiac uptake ratios of each group showed that the 68Ga-DOTA-P26 had a better tumor-to-cardiac uptake ratio in the A549TIM3 model than several other molecular probes, and in the MC38 model, similar results were obtained, with the difference that the 68Ga-DOTA-P26NH2 had the highest tumor-to-cardiac uptake ratio in the CCRCC model. Finally, validation by IHC showed that A549TIM3, MC38, and CCRCC tumor tissues had varying degrees of TIM3 expression. Upon comparison of ex vivo and in vivo studies, one of them, the 68Ga-DOTA-P26 probe, demonstrated significant target specificity for TIM3. These results suggest that studying peptide probes targeting TIM3 will promote the process of TIM3-targeted drug research and is expected to guide the application of TIM3 immune checkpoint drugs in immunotherapy.
Collapse
Affiliation(s)
- Jinping Tao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Fei Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ziqing Zeng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Wenyuan Zhou
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zilei Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Chengxue He
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jinyu Zhu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Chuanke Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Hua Zhu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
- School of Medicine, Guizhou University, Guiyang 550025, China
- Department of Chemistry, Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
22
|
Wu L, Wei D, Chen W, Wu C, Lu Z, Li S, Liu W. Comprehensive Potential of Artificial Intelligence for Predicting PD-L1 Expression and EGFR Mutations in Lung Cancer: A Systematic Review and Meta-Analysis. J Comput Assist Tomogr 2025; 49:101-112. [PMID: 39143665 DOI: 10.1097/rct.0000000000001644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
OBJECTIVE To evaluate the methodological quality and the predictive performance of artificial intelligence (AI) for predicting programmed death ligand 1 (PD-L1) expression and epidermal growth factor receptors (EGFR) mutations in lung cancer (LC) based on systematic review and meta-analysis. METHODS AI studies based on PET/CT, CT, PET, and immunohistochemistry (IHC)-whole-slide image (WSI) were included to predict PD-L1 expression or EGFR mutations in LC. The modified Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) tool was used to evaluate the methodological quality. A comprehensive meta-analysis was conducted to analyze the overall area under the curve (AUC). The Cochrane diagnostic test and I2 statistics were used to assess the heterogeneity of the meta-analysis. RESULTS A total of 45 AI studies were included, of which 10 were used to predict PD-L1 expression and 35 were used to predict EGFR mutations. Based on the analysis using the QUADAS-2 tool, 37 studies achieved a high-quality score of 7. In the meta-analysis of PD-L1 expression levels, the overall AUCs for PET/CT, CT, and IHC-WSI were 0.80 (95% confidence interval [CI], 0.77-0.84), 0.74 (95% CI, 0.69-0.77), and 0.95 (95% CI, 0.93-0.97), respectively. For EGFR mutation status, the overall AUCs for PET/CT, CT, and PET were 0.85 (95% CI, 0.81-0.88), 0.83 (95% CI, 0.80-0.86), and 0.75 (95% CI, 0.71-0.79), respectively. The Cochrane Diagnostic Test revealed an I2 value exceeding 50%, indicating substantial heterogeneity in the PD-L1 and EGFR meta-analyses. When AI was combined with clinicopathological features, the enhancement in predicting PD-L1 expression was not substantial, whereas the prediction of EGFR mutations showed improvement compared to the CT and PET models, albeit not significantly so compared to the PET/CT models. CONCLUSIONS The overall performance of AI in predicting PD-L1 expression and EGFR mutations in LC has promising clinical implications.
Collapse
Affiliation(s)
- Linyong Wu
- From the Department of Medical Ultrasound, Maoming People's Hospital, Maoming
| | - Dayou Wei
- From the Department of Medical Ultrasound, Maoming People's Hospital, Maoming
| | - Wubiao Chen
- Radiology Imaging Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, PR China
| | - Chaojun Wu
- From the Department of Medical Ultrasound, Maoming People's Hospital, Maoming
| | - Zhendong Lu
- Radiology Imaging Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, PR China
| | - Songhua Li
- From the Department of Medical Ultrasound, Maoming People's Hospital, Maoming
| | - Wenci Liu
- Radiology Imaging Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, PR China
| |
Collapse
|
23
|
Kummar S, Razak AA, Laurie S, Glatt DM, Kell S, Diep AN, Schmidt M, Hom C, German C, Shringarpure SS, Majeed SR, Rasco D. First-in-Human Study of 23ME-00610, an Antagonistic Antibody for Genetically Validated CD200R1 Immune Checkpoint, in Participants with Advanced Solid Malignancies. CANCER RESEARCH COMMUNICATIONS 2025; 5:94-105. [PMID: 39651931 PMCID: PMC11734590 DOI: 10.1158/2767-9764.crc-24-0568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 01/16/2025]
Abstract
PURPOSE In this phase 1 portion of a first-in-human phase 1/2a study (NCT05199272), 23ME-00610 was evaluated in participants with advanced solid malignancies to determine its safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD). Exploratory biomarkers were evaluated to examine potential correlates of efficacy and safety. PATIENTS AND METHODS Eligible participants (≥18 years) were administered 23ME-00610 intravenously every 3 weeks (Q3W) using an accelerated titration design followed by a traditional 3 + 3 design, with an initial dose level of 2 mg. RESULTS Twenty-eight participants were enrolled across seven cohorts and received a median of four cycles of 23ME-00610. No treatment-related serious adverse events (AE) were observed, and the maximum tolerated dose was not reached. Overall, the PK of 23ME-00610 was linear and dose proportional for doses ≥60 mg, with a median terminal half-life of 13 days at 1,400 mg. Peripheral saturation of CD200R1 was observed for doses ≥60 mg. Immune-related AEs, including rash, pruritus, and hypothyroidism, were predicted by phenome-wide association studies and observed for doses ≥60 mg. A confirmed partial response was observed in a participant with well-differentiated pancreatic neuroendocrine cancer whose tumor was among those with the highest tumor CD200 expression. CONCLUSIONS 23ME-00610 has mild-to-moderate on-target AEs and PK/PD consistent with tumor target saturation and dosing every 3 weeks. The trend for clinical benefit in participants with tumor CD200 expression suggests that 23ME-00610 inhibits CD200R1 signaling and may reverse CD200-mediated immune evasion. Based on PK/PD, safety, and preliminary antitumor activity, 1,400 mg Q3W was selected as the dose for further study. SIGNIFICANCE Genome-wide association studies (GWAS) of the 23andMe genetic database identified CD200R1 as a promising therapeutic target for cancer. This phase 1 study of 23ME-00610, a CD200R1 antagonist IgG1, showed acceptable safety and tolerability, PK supporting Q3W dosing, and PD and preliminary clinical activity supporting an initial recommended phase 2 dose of 1,400 mg.
Collapse
Affiliation(s)
- Shivaani Kummar
- Knight Cancer Institute, Oregon Health Sciences University, Portland, Oregon
| | | | | | | | - Sariah Kell
- 23andMe Therapeutics, South San Francisco, California
| | - Anh N. Diep
- 23andMe Therapeutics, South San Francisco, California
| | - Maike Schmidt
- 23andMe Therapeutics, South San Francisco, California
| | - Clifford Hom
- 23andMe Therapeutics, South San Francisco, California
| | | | | | | | - Drew Rasco
- The START Center for Cancer Care, San Antonio, Texas
| |
Collapse
|
24
|
Lee G, Moon SH, Kim JH, Jeong DY, Choi J, Choi JY, Lee HY. Multimodal Imaging Approach for Tumor Treatment Response Evaluation in the Era of Immunotherapy. Invest Radiol 2025; 60:11-26. [PMID: 39018248 DOI: 10.1097/rli.0000000000001096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/19/2024]
Abstract
ABSTRACT Immunotherapy is likely the most remarkable advancement in lung cancer treatment during the past decade. Although immunotherapy provides substantial benefits, their therapeutic responses differ from those of conventional chemotherapy and targeted therapy, and some patients present unique immunotherapy response patterns that cannot be judged under the current measurement standards. Therefore, the response monitoring of immunotherapy can be challenging, such as the differentiation between real response and pseudo-response. This review outlines the various tumor response patterns to immunotherapy and discusses methods for quantifying computed tomography (CT) and 18 F-fluorodeoxyglucose positron emission tomography (PET) in the field of lung cancer. Emerging technologies in magnetic resonance imaging (MRI) and non-FDG PET tracers are also explored. With immunotherapy responses, the role for imaging is essential in both anatomical radiological responses (CT/MRI) and molecular changes (PET imaging). Multiple aspects must be considered when assessing treatment responses using CT and PET. Finally, we introduce multimodal approaches that integrate imaging and nonimaging data, and we discuss future directions for the assessment and prediction of lung cancer responses to immunotherapy.
Collapse
Affiliation(s)
- Geewon Lee
- From the Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (G.L., D.Y.J., J.C., H.Y.L.); Department of Radiology and Medical Research Institute, Pusan National University Hospital, Pusan National University School of Medicine, Busan, South Korea (G.L.); Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (S.H.M., J.Y.C.); Industrial Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea (J.H.K.); Department of Digital Health, SAIHST, Sungkyunkwan University, Seoul, South Korea (J.C.); and Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea (H.Y.L.)
| | | | | | | | | | | | | |
Collapse
|
25
|
Wang J, Serafini A, Kuker R, Ayubcha C, Cohen G, Nadel H, McKinney A, Alavi A, Yu JQ. The State-of-the-Art PET Tracers in Glioblastoma and High-grade Gliomas and Implications for Theranostics. PET Clin 2025; 20:147-164. [PMID: 39482219 DOI: 10.1016/j.cpet.2024.09.009] [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: 11/03/2024]
Abstract
MR imaging is currently the main imaging modality used for the diagnosis and post therapeutic assessment of glioblastomas. Recently, several innovative PET radioactive tracers have been investigated for the evaluation of glioblastomas (GBM). These radiotracers target several biochemical and pathophysiological processes seen in tumors. These include glucose metabolism, DNA synthesis and cell proliferation, amino acid transport, cell membrane biosynthesis, specific membrane antigens such as prostatic specific membrane antigens, fibroblast activation protein inhibitor, translocator protein and hypoxia sensing agents, and antibodies targeting specific cell receptor antigen. This review aims to discuss the clinical value of these PET radiopharmaceuticals in the evaluation and treatment of GBMs.
Collapse
Affiliation(s)
- Jiaqiong Wang
- Division of Nuclear Medicine, Department of Radiology, Temple University Health System, Fox Chase Cancer Center, Philadelphia, PA 19140, USA.
| | - Aldo Serafini
- Division of Nuclear Medicine, Department of Radiology, University of Miami Miller School of Medicine, Jackson Memorial Hospital, Miami, FL, USA
| | - Russ Kuker
- Division of Nuclear Medicine, Department of Radiology, University of Miami Miller School of Medicine, Jackson Memorial Hospital, Miami, FL, USA
| | - Cyrus Ayubcha
- Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Gary Cohen
- Department of Radiology, Temple University Health System, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Helen Nadel
- Department of Radiology, Lucile Packard Children's Hospital at Stanford, Stanford University School of Medicine, Stanford, CA, USA
| | - Alexander McKinney
- Department of Radiology, University of Miami Miller School of Medicine, Jackson Memorial Hospital, Miami, FL, USA
| | - Abass Alavi
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jian Q Yu
- Division of Nuclear Medicine, Department of Radiology, Fox Chase Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
26
|
Dimitrakopoulou-Strauss A, Pan L, Sachpekidis C. Non-[ 18F]FDG PET-Radiopharmaceuticals in Oncology. Pharmaceuticals (Basel) 2024; 17:1641. [PMID: 39770483 PMCID: PMC11677833 DOI: 10.3390/ph17121641] [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: 11/18/2024] [Revised: 11/26/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Molecular imaging is a growing field, driven by technological advances, such as the improvement of PET-CT scanners through the introduction of digital detectors and scanners with an extended field of view, resulting in much higher sensitivity and a variety of new specific radiopharmaceuticals that allow the visualization of specific molecular pathways and even theragnostic approaches. In oncology, the development of dedicated tracers is crucial for personalized therapeutic approaches. Novel peptides allow the visualization of many different targets, such as PD-1 and PD-L1 expression, chemokine expression, HER expression, T-cell imaging, microenvironmental imaging, such as FAP imaging, and many more. In this article, we review recent advances in the development of non-[18F]FDG PET radiopharmaceuticals and their current clinical applications in oncology, as well as some future aspects.
Collapse
Affiliation(s)
- Antonia Dimitrakopoulou-Strauss
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | | | | |
Collapse
|
27
|
Teles L, Tolboom N, Plasschaert SL, Poot AJ, Braat AJ, van Noesel MM. Potential of non-FDG PET radiotracers for paediatric patients with solid tumours. EJC PAEDIATRIC ONCOLOGY 2024; 4:100203. [DOI: 10.1016/j.ejcped.2024.100203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
28
|
Malicki S, Czarna A, Żyła E, Pucelik B, Gałan W, Chruścicka B, Kamińska M, Sochaj-Gregorczyk A, Magiera-Mularz K, Wang J, Winiarski M, Benedyk-Machaczka M, Kozieł J, Dubin G, Mydel P. Development of selective ssDNA micro-probe for PD1 detection as a novel strategy for cancer imaging. Sci Rep 2024; 14:28652. [PMID: 39562585 PMCID: PMC11576874 DOI: 10.1038/s41598-024-74891-7] [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: 03/10/2023] [Accepted: 09/30/2024] [Indexed: 11/21/2024] Open
Abstract
Programmed death receptor 1, PD1, modulates the function of immune cells by providing inhibitory signals and constitutes the marker of immune exhaustion. Monitoring the level of PD1 promises a useful diagnostic approach in autoimmune diseases and cancer. Here we describe the development of an ssDNA aptamer-based molecular probe capable of specific recognition of human PD1 receptor. The aptamer was selected using SELEX, its sequence was further optimized, and the affinity and specificity were determined in biochemical assays. The aptamer was converted into a fluorescent probe and its potential in molecular imaging was demonstrated in a culture of human cells overexpressing PD1 and murine pancreatic organoids / immune cells mixed co-culture model. We conclude that the provided aptamers are suitable probes for imaging of PD1 expressing immune cells even in complex cellular models and may find future utility as diagnostic tools.
Collapse
Affiliation(s)
- Stanisław Malicki
- Laboratory of Proteolysis and Post-translational Modification of Proteins, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, Krakow, 30-387, Poland.
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland.
| | - Anna Czarna
- Protein Crystallography Research, Group Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, Krakow, 30-387, Poland
| | - Edyta Żyła
- Protein Crystallography Research, Group Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, Krakow, 30-387, Poland
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland
| | - Barbara Pucelik
- Protein Crystallography Research, Group Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, Krakow, 30-387, Poland
- 5Łukasiewicz Research Network, Krakow Institute of Technology, ul. Zakopiańska 73, Kraków, 30-418, Poland
| | - Wojciech Gałan
- Department of Computational Biophysics and Bioinformatics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland
| | - Barbara Chruścicka
- Laboratory of Proteolysis and Post-translational Modification of Proteins, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, Krakow, 30-387, Poland
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland
| | - Marta Kamińska
- Broegelmann Research Laboratory, University of Bergen, Haukeland universitetssykehus Laboratoriebygget, Bergen, 5009, Norway
| | - Alicja Sochaj-Gregorczyk
- Laboratory of Proteolysis and Post-translational Modification of Proteins, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, Krakow, 30-387, Poland
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland
| | - Katarzyna Magiera-Mularz
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, Krakow, 30-387, Poland
- Laboratory of protein NMR, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, Krakow, 30- 387, Poland
| | - Jun Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, People's Republic of China
| | - Marek Winiarski
- 2nd Department of General Surgery, Faculty of Medicine, Jagiellonian University Medical College, Kraków, 31-008, Poland
| | - Małgorzata Benedyk-Machaczka
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland
| | - Joanna Kozieł
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland
| | - Grzegorz Dubin
- Protein Crystallography Research, Group Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, Krakow, 30-387, Poland.
| | - Piotr Mydel
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland.
- Broegelmann Research Laboratory, University of Bergen, Haukeland universitetssykehus Laboratoriebygget, Bergen, 5009, Norway.
| |
Collapse
|
29
|
Wu Q, Shao H, Zhai W, Huang G, Liu J, Calais J, Wei W. Molecular imaging of renal cell carcinomas: ready for prime time. Nat Rev Urol 2024:10.1038/s41585-024-00962-z. [PMID: 39543358 DOI: 10.1038/s41585-024-00962-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2024] [Indexed: 11/17/2024]
Abstract
The clinical diagnosis of renal cell carcinoma (RCC) is constantly evolving. Diagnostic imaging of RCC relying on enhanced computed tomography (CT) and magnetic resonance imaging (MRI) is commonly used for renal mass characterization and assessment of tumour thrombosis, whereas pathology is the gold standard for establishing diagnosis. However, molecular imaging is rapidly improving the clinical management of RCC, particularly clear-cell RCC. Molecular imaging aids in the non-invasive visualization and characterization of specific biomarkers such as carbonic anhydrase IX and CD70 within the tumours, which help to assess tumour heterogeneity and status. Target-specific molecular imaging of RCCs will substantially improve the diagnostic landscape of RCC and will further facilitate clinical decision-making regarding initial staging and re-staging, monitoring of recurrence and metastasis, patient stratification and selection, and the prediction and evaluation of treatment responses.
Collapse
Affiliation(s)
- Qianyun Wu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hongda Shao
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Zhai
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Jeremie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA.
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
30
|
Dev ID, Puranik AD, Singh B, Prasad V. Current and Future Perspectives of PDL1 PET and SPECT Imaging. Semin Nucl Med 2024; 54:966-975. [PMID: 39510854 DOI: 10.1053/j.semnuclmed.2024.09.008] [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: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 11/15/2024]
Abstract
Programmed Death 1 (PD1) and Programmed Death Ligand (PDL1) play a crucial role in tumor microenvironment by helping cancer cells evade innate immunity. Numerous inhibitor anticancer drugs targeting this interplay have been used in clinical practice and many more are in preclinical stage. These drugs have shown promising results in achieving good response and long-term clinical benefit, is routinely performed to identify patients who may benefit. However, there are major challenges associated with these immunohistochemistry tests which have opened the space for noninvasive imaging modalities using PD1 and PDL1 inhibitors labeled with either PET or SPECT radionuclides. These radiopharmaceuticals, although primarily developed for the field of immunotherapy, have great potential in expanding and optimizing the combination of radiopharmaceutical therapies with PD1-PDL1 targeting anticancer drugs. This review elaborates currently available PET and SPECT radiopharmaceuticals targeting PD1-PDL1 axis. It also explores the potential future role of newer targets which are being developed and tested in various preclinical studies.
Collapse
Affiliation(s)
- Indraja D Dev
- Assistant Professor, Department of Nuclear Medicine and Molecular Imaging, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Center, Homi Bhabha National Institute, Mumbai, India
| | - Ameya D Puranik
- Professor, Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, India
| | - Baljinder Singh
- Professor, Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Vikas Prasad
- Director of Clinical Theranostics, Division of Nuclear Medicine, Mallinckrodt Institute of Radiology, Washington, WA.
| |
Collapse
|
31
|
Zhao Y, Yin X, Zhou M, Rao W, Ji X, Wang X, Xiao X, Hu S. Noninvasive Monitoring of Programmed Death-Ligand 2 Expression with Positron Emission Tomography using 68Ga-labeled Peptide Antagonist in Preclinical and Exploratory Human Studies. RESEARCH (WASHINGTON, D.C.) 2024; 7:0523. [PMID: 39494220 PMCID: PMC11528066 DOI: 10.34133/research.0523] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 10/14/2024] [Accepted: 10/14/2024] [Indexed: 11/05/2024]
Abstract
While the expression of programmed death ligand-1 (PD-L1) is associated with response to immune therapy, PD-L1-negative patients may still benefit from immune treatment. Programmed death ligand-2 (PD-L2), another crucial immune checkpoint molecule interacting with PD-1, correlates with the efficacy of various tumor immune therapies. This study investigates the expression of PD-L2 in non-small cell lung cancer (NSCLC) patients following anti-PD-1 therapy and its predictive value for clinical survival outcomes. Additionally, we explore the noninvasive, real-time, and dynamic quantitative analysis potential of PD-L2 positron emission tomography (PET) imaging in transplanted tumors. We utilized [68Ga]Ga-labeled peptide HN11-1 for PD-L2 PET imaging. The results indicate a higher response rate to anti-PD-1 therapy in patients positive for both PD-L1 and PD-L2, with PD-L2 status independently predicting progression-free survival (PFS) with pembrolizumab treatment. Furthermore, [68Ga]Ga-HN11-1 PET imaging demonstrates specificity in assessing PD-L2 status. Overall, we confirm the correlation between high PD-L2 expression and favorable PFS in NSCLC patients post anti-PD-1 therapy and highlight the promising potential of [68Ga]Ga-HN11-1 as a specific tracer for PD-L2 in preclinical and initial human trials.
Collapse
Affiliation(s)
- Yajie Zhao
- Department of Nuclear Medicine, Xiangya Hospital,
Central South University, Changsha 410008, China
| | - Xiaoqin Yin
- Department of Nuclear Medicine, Xiangya Hospital,
Central South University, Changsha 410008, China
| | - Ming Zhou
- Department of Nuclear Medicine, Xiangya Hospital,
Central South University, Changsha 410008, China
| | - Wanqian Rao
- Department of Nuclear Medicine, Xiangya Hospital,
Central South University, Changsha 410008, China
| | - Xuan Ji
- Department of Periodontology,
Suzhou Stomatological Hospital, Suzhou, Jiangsu 215026, China
| | - Xiaobo Wang
- Department of Nuclear Medicine and State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xijing Hospital,
Fourth Military Medical University, Xi’an 710032, China
| | - XiaoXiong Xiao
- Department of Thoracic Surgery, Xiangya Hospital,
Central South University, Changsha 410008, China
| | - Shuo Hu
- Department of Nuclear Medicine, Xiangya Hospital,
Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha 410008, China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital,
Central South University, Changsha 410008, China
| |
Collapse
|
32
|
Kim SY, Chung HW, So Y, Lee MH, Lee EJ. Recent Updates of PET in Lymphoma: FDG and Beyond. Biomedicines 2024; 12:2485. [PMID: 39595051 PMCID: PMC11592097 DOI: 10.3390/biomedicines12112485] [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: 09/07/2024] [Revised: 10/12/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Lymphoma is one of the most common cancers worldwide, categorized into Hodgkin lymphoma and non-Hodgkin lymphoma. 18F-fluorodeoxyglucose positron emission tomography (FDG PET) has become an essential imaging tool for evaluating patients with lymphoma in terms of initial diagnosis, staging, prognosis, and treatment response assessment. Recent advancements in imaging technology and methodologies, along with the development of artificial intelligence, have revolutionized the evaluation of complex imaging data, enhancing the diagnostic and predictive power of PET in lymphoma. However, FDG is not cancer-specific, but it primarily reflects glucose metabolism, which has prompted the investigation of alternative PET tracers to address this limitation. Novel PET radiotracers, such as fibroblast activation protein inhibitors targeting the tumor microenvironment, have recently shown promising results in evaluating various malignancies compared to FDG PET. Furthermore, with the rapid advancements in immunotherapy and the favorable imaging properties of 89Zr, immunoPET has emerged as a promising modality, offering insights into the functional and molecular status of the immune system. ImmunoPET can also facilitate the development of new antibody therapeutics and radioimmunotherapy by providing pharmacokinetic and pharmacodynamic data. This review provides comprehensive insights into the current clinical applications of FDG PET in lymphoma, while also exploring novel PET imaging radiotracers beyond FDG, discussing their mechanisms of action and potential impact on patient management.
Collapse
Affiliation(s)
- Sung-Yong Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Republic of Korea; (S.-Y.K.); (M.H.L.)
| | - Hyun Woo Chung
- Department of Nuclear Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Republic of Korea;
- Research Institute of Medical Science, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Republic of Korea
| | - Young So
- Department of Nuclear Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Republic of Korea;
| | - Mark Hong Lee
- Division of Hemato-Oncology, Department of Internal Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Republic of Korea; (S.-Y.K.); (M.H.L.)
| | - Eun Jeong Lee
- Department of Nuclear Medicine, Seoul Medical Center, 156 Sinnae-ro, Jungnang-gu, Seoul 02053, Republic of Korea;
| |
Collapse
|
33
|
Enke JS, Bundschuh RA, Claus R, Lapa C. New PET Tracers for Lymphoma. PET Clin 2024; 19:463-474. [PMID: 38969567 DOI: 10.1016/j.cpet.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2024]
Abstract
While functional imaging with [18F]Fluoro-deoxy-glucose positron emission tomography (PET)/computed tomography is a well-established imaging modality in most lymphoma entities, novel tracers addressing cell surface receptors, tumor biology, and the microenvironment are being developed. Especially, with the emergence of immuno-PET targeting surface markers of lymphoma cells, a new imaging modality of immunotherapies is evolving, which might especially aid in relapsed and refractory disease stages. This review highlights different new PET tracers in indolent and aggressive lymphoma subtypes and summarizes the current state of immuno-PET imaging in lymphoma.
Collapse
Affiliation(s)
- Johanna S Enke
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Stenglinstr. 2, 86156 Augsburg, Germany.
| | - Ralph A Bundschuh
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Stenglinstr. 2, 86156 Augsburg, Germany
| | - Rainer Claus
- Hematology and Oncology, Faculty of Medicine, University of Augsburg, Stenglinstr. 2, 86156 Augsburg, Germany; Pathology, Faculty of Medicine, University of Augsburg, Stenglinstr. 2, 86156 Augsburg, Germany
| | - Constantin Lapa
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Stenglinstr. 2, 86156 Augsburg, Germany
| |
Collapse
|
34
|
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
|
35
|
Tian Q, Jia JY, Qin C, Zhou H, Zhou SY, Qin YH, Wu YY, Shi J, Duan SF, Feng F. Prediction of programmed death-1 expression status in non-small cell lung cancer based on intratumoural and peritumoral computed tomography (CT) radiomics nomogram. Clin Radiol 2024; 79:e1089-e1100. [PMID: 38876960 DOI: 10.1016/j.crad.2024.05.008] [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: 10/11/2023] [Revised: 03/25/2024] [Accepted: 05/10/2024] [Indexed: 06/16/2024]
Abstract
AIMS This study aimed to predict the expression of programmed death-1 (PD-1) in non-small cell lung cancer (NSCLC) using intratumoral and peritumoral computed tomography (CT) radiomics nomogram. MATERIALS AND METHODS Two hundred patients pathologically diagnosed with NSCLC from two hospitals were retrospectively analyzed. Of these, 159 NSCLC patients from our hospital were randomly divided into a training cohort (n=96) and an internal validation cohort (n=63) at a ratio of 6:4, while 41 NSCLC patients from another medical institution served as the external validation cohort. The radiomic features of the gross tumor volume (GTV) and peritumoral volume (PTV) were extracted from the CT images. Optimal radiomics features were selected using least absolute shrinkage and selection operator regression analysis. Finally, a CT radiomics nomogram of clinically independent predictors combined with the best rad-score was constructed. RESULTS Compared with the 'GTV' and 'PTV' radiomics models, the combined 'GTV + PTV' radiomics model showed better predictive performance, and its area under the curve (AUC) values in the training, internal validation, and external validation cohorts were 0.90 (95% confidence interval [CI]: 0.83-0.97), 0.85 (95% CI: 0.74-0.96) and 0.78 (95% CI: 0.63-0.92). The nomogram constructed by the rad-score of the 'GTV + PTV' radiomics model combined with clinical independent predictors (prealbumin and monocyte) had the best performance, with AUC values in each cohort being 0.92 (95% CI: 0.85-0.98), 0.88 (95% CI: 0.78-0.97), and 0.80 (95% CI: 0.66-0.94), respectively. CONCLUSION The intratumoral and peritumoral CT radiomics nomogram may facilitate individualized prediction of PD-1 expression status in patients with NSCLC.
Collapse
Affiliation(s)
- Q Tian
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu 226361, PR China.
| | - J Y Jia
- Department of Medical Imaging Center, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huaian 223300, Jiangsu, PR China.
| | - C Qin
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu 226361, PR China.
| | - H Zhou
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu 226361, PR China.
| | - S-Y Zhou
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu 226361, PR China.
| | - Y H Qin
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu 226361, PR China.
| | - Y Y Wu
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu 226361, PR China.
| | - Jian Shi
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu 226361, PR China.
| | - S F Duan
- GE Healthcare China, Shanghai 210000, PR China.
| | - F Feng
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu 226361, PR China.
| |
Collapse
|
36
|
Bansal A, Lavoie RR, Lucien F, Kethamreddy M, Wootla B, Dong H, Park SS, Pandey MK. Synthesis and evaluation of anti-PD-L1-B11 antibody fragments for PET imaging of PD-L1 in breast cancer and melanoma tumor models. Sci Rep 2024; 14:19561. [PMID: 39174596 PMCID: PMC11341854 DOI: 10.1038/s41598-024-70385-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 08/16/2024] [Indexed: 08/24/2024] Open
Abstract
There is a critical need to non-invasively assess the PD-L1 expression in tumors as a predictive biomarker for determining the efficacy of anti-PD-1/PD-L1 immunotherapies. Non-invasive imaging modality like positron emission tomography (PET) can be a powerful tool to assess the PD-L1 expression in the whole body including multiple metastases as a patient selection criterion for the anti-PD-1/PD-L1 immunotherapy. In this study, we synthesized B11-nanobody, B11-scFv and B11-diabody fragments from the full-length anti-PD-L1 B11 IgG. Out of the three antibody fragments, B11-diabody showed higher nM affinity towards PD-L1 antigen as compared to B11-scFv and B11-nanobody. All three antibody fragments were successfully radiolabeled with 64Cu, a PET radioisotope. For radiolabeling, the antibody fragments were first conjugated with p-SCN-Bn-NOTA followed by chelation with 64Cu. All three radiolabeled antibody fragments were found to be stable in mouse and human sera for up to 24 h. Additionally, all three [64Cu]Cu-NOTA-B11-antibody fragments were evaluated in PD-L1 negative and human PD-L1 expressing cancer cells and subcutaneous tumor models. Based on the results, [64Cu]Cu-NOTA-B11-diabody has potential to be used as a PET imaging probe for assessing PD-L1 expression in tumors as early as 4 h post-injection, allowing faster assessment compared to the full length IgG based PET imaging probe.
Collapse
Affiliation(s)
- Aditya Bansal
- Division of Nuclear Medicine, Department of Radiology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Roxane R Lavoie
- Department of Urology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Fabrice Lucien
- Department of Urology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Manasa Kethamreddy
- Division of Nuclear Medicine, Department of Radiology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Bharath Wootla
- Office of Translation to Practice, Mayo Clinic, Rochester, MN, 55905, USA
| | - Haidong Dong
- Department of Urology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Sean S Park
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Mukesh K Pandey
- Division of Nuclear Medicine, Department of Radiology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
37
|
Garaulet G, Báez BB, Medrano G, Rivas-Sánchez M, Sánchez-Alonso D, Martinez-Torrecuadrada JL, Mulero F. Radioimmunotheragnosis in Cancer Research. Cancers (Basel) 2024; 16:2896. [PMID: 39199666 PMCID: PMC11352548 DOI: 10.3390/cancers16162896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/02/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
The combination of immunoPET-where an antibody (Ab) is labeled with an isotope for PET imaging-and radioimmunotherapy (RIT), using the same antibody with a therapeutic isotope, offers significant advantages in cancer management. ImmunoPET allows non-invasive imaging of antigen expression, which aids in patient selection for subsequent radioimmunotherapy. It also facilitates the assessment of tumor response to therapy, allowing for treatment adjustments if necessary. In addition, immunoPET provides critical pharmacokinetic data, including antibody biodistribution and clearance rates, which are essential for dosimetry calculations and treatment protocol optimization. There are still challenges to overcome. Identifying appropriate target antigens that are selectively expressed on cancer cells while minimally expressed on normal tissues remains a major hurdle to reduce off-target toxicity. In addition, it is critical to optimize the pharmacokinetics of radiolabeled antibodies to maximize tumor uptake and minimize normal tissue uptake, particularly in vital organs such as the liver and kidney. This approach offers the potential for targeted and personalized cancer therapy with reduced systemic toxicity by exploiting the specificity of monoclonal antibodies and the cytotoxic effects of radiation. However, further research is needed to address remaining challenges and to optimize these technologies for clinical use.
Collapse
Affiliation(s)
- Guillermo Garaulet
- Molecular Imaging Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (G.G.); (B.B.B.); (G.M.)
| | - Bárbara Beatriz Báez
- Molecular Imaging Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (G.G.); (B.B.B.); (G.M.)
| | - Guillermo Medrano
- Molecular Imaging Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (G.G.); (B.B.B.); (G.M.)
| | - María Rivas-Sánchez
- Protein Production Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (M.R.-S.); (D.S.-A.)
| | - David Sánchez-Alonso
- Protein Production Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (M.R.-S.); (D.S.-A.)
| | | | - Francisca Mulero
- Molecular Imaging Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (G.G.); (B.B.B.); (G.M.)
| |
Collapse
|
38
|
Hatanaka T, Saito N, Kakizaki S, Hiraoka A, Tada T, Kariyama K, Tani J, Takaguchi K, Itobayashi E, Ishikawa T, Toyoda H, Kawata K, Naganuma A, Yata Y, Ohama H, Matono T, Tada F, Nouso K, Morishita A, Tsutsui A, Nagano T, Nakamura S, Kumada T. Factors Affecting an Increase in Spleen Volume and Association of Spleen Volume Variation with the Clinical Outcomes of Atezolizumab and Bevacizumab Treatment for Hepatocellular Carcinoma: A Retrospective Analysis. Oncology 2024; 103:94-106. [PMID: 39163847 DOI: 10.1159/000541002] [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: 03/01/2024] [Accepted: 08/13/2024] [Indexed: 08/22/2024]
Abstract
INTRODUCTION Gastrointestinal varices rupture is considered to be prone to occur during atezolizumab and bevacizumab (Atez/Bev) treatment. This study aimed to investigate predictive factors affecting the increase in spleen volume (SpV) and the association of SpV variation with the clinical outcomes of Atez/Bev. METHODS A total of 164 HCC patients were included in this retrospective multicenter study. We measured SpV based on CT scans obtained before treatment and at evaluations. We used the inverse probability of treatment weight to address the imbalance between patient characteristics. RESULTS The median pretreatment SpV was 184 (130-257) cm3 and the median SpV variation was 27 (9-60) cm3. An increase in the SpV was observed in 140 patients (85.4%). Age <74 years (p = 0.03), mALBI grade 2b or 3 (p = 0.03), and pretreatment SpV ≥184 cm3 (p < 0.001) were significantly associated with increased SpV. There were no significant differences in progression-free survival (PFS) or overall survival (OS) between patients with SpV variation <25 cm3 and those with SpV variation ≥25 cm3 in the crude (p = 0.3 and 0.7) and IPTW-weighted cohorts (p = 0.08 and 0.8, respectively). Regarding pretreatment SpV, there were no significant differences in PFS or OS between patients with and without pretreatment spleen enlargement in the crude (both p = 0.3) and IPTW-weighted cohort (p = 0.6 and 0.3, respectively). CONCLUSION Caution is warranted to detect the aggravation of portal hypertension when administering Atez/Bev to young patients or patients with an impaired liver function or pretreatment spleen enlargement. The impact of spleen modulation by Atez/Bev appears to be limited on clinical efficacy.
Collapse
Affiliation(s)
- Takeshi Hatanaka
- Department of Gastroenterology, Gunma Saiseikai Maebashi Hospital, Maebashi, Japan
| | - Naoto Saito
- Department of Gastroenterology, Gunma Saiseikai Maebashi Hospital, Maebashi, Japan
| | - Satoru Kakizaki
- Department of Clinical Research, National Hospital Organization Takasaki General Medical Center, Takasaki, Japan
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Atsushi Hiraoka
- Gastroenterology Center, Ehime Prefectural Central Hospital, Matsuyama, Japan
| | - Toshifumi Tada
- Department of Internal Medicine, Japanese Red Cross Himeji Hospital, Himeji, Japan
| | - Kazuya Kariyama
- Department of Gastroenterology, Okayama City Hospital, Okayama, Japan
| | - Joji Tani
- Department of Gastroenterology and Neurology, Kagawa University, Takamatsu, Japan
| | - Koichi Takaguchi
- Department of Hepatology, Kagawa Prefectural Central Hospital, Takamatsu, Japan
| | - Ei Itobayashi
- Department of Gastroenterology, Asahi General Hospital, Asahi, Japan
| | - Toru Ishikawa
- Department of Gastroenterology, Saiseikai Niigata Hospital, Niigata, Japan
| | - Hidenori Toyoda
- Department of Gastroenterology and Hepatology, Ogaki Municipal Hospital, Ogaki, Japan
| | - Kazuhito Kawata
- Hepatology Division, Department of Internal Medicine II, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Atsushi Naganuma
- Department of Gastroenterology, National Hospital Organization Takasaki General Medical Center, Takasaki, Japan
| | - Yutaka Yata
- Department of Gastroenterology, Hanwa Memorial Hospital, Osaka, Japan
| | - Hideko Ohama
- Gastroenterology Center, Ehime Prefectural Central Hospital, Matsuyama, Japan
- Department of Gastroenterology, Takarazuka City Hospital, Takarazuka, Japan
| | - Tomomitsu Matono
- Department of Gastroenterology, Hyogo Prefectural Harima-Himeji General Medical Center, Himeji, Japan
| | - Fujimasa Tada
- Gastroenterology Center, Ehime Prefectural Central Hospital, Matsuyama, Japan
| | - Kazuhiro Nouso
- Department of Gastroenterology, Okayama City Hospital, Okayama, Japan
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Kagawa University, Takamatsu, Japan
| | - Akemi Tsutsui
- Department of Hepatology, Kagawa Prefectural Central Hospital, Takamatsu, Japan
| | - Takuya Nagano
- Department of Hepatology, Kagawa Prefectural Central Hospital, Takamatsu, Japan
| | - Shinichiro Nakamura
- Department of Internal Medicine, Japanese Red Cross Himeji Hospital, Himeji, Japan
| | - Takashi Kumada
- Department of Nursing, Gifu Kyoritsu University, Ogaki, Japan
| |
Collapse
|
39
|
Pezzana S, Blaess S, Kortendieck J, Hemmer N, Tako B, Pietura C, Ruoff L, Riel S, Schaller M, Gonzalez-Menendez I, Quintanilla-Martinez L, Mascioni A, Aivazian A, Wilson I, Maurer A, Pichler BJ, Kneilling M, Sonanini D. In-depth cross-validation of human and mouse CD4-specific minibodies for noninvasive PET imaging of CD4 + cells and response prediction to cancer immunotherapy. Theranostics 2024; 14:4582-4597. [PMID: 39239511 PMCID: PMC11373626 DOI: 10.7150/thno.95173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 07/11/2024] [Indexed: 09/07/2024] Open
Abstract
Increasing evidence emphasizes the pivotal role of CD4+ T cells in orchestrating cancer immunity. Noninvasive in vivo imaging of the temporal dynamics of CD4+ T cells and their distribution patterns might provide novel insights into their effector and regulator cell functions during cancer immunotherapy (CIT). Methods: We conducted a comparative analysis of 89Zr-labeled anti-mouse (m) and anti-human (h) CD4-targeting minibodies (Mbs) for in vivo positron emission tomography (PET)/magnetic resonance imaging (MRI) of CD4+ T cells in human xenografts, syngeneic tumor-bearing wild-type (WT), and human CD4+ knock-in (hCD4-KI) mouse models. Results: Both 89Zr-CD4-Mbs yielded high radiolabeling efficiencies of >90%, immunoreactivities of >70%, and specific in vitro binding to their target antigens. The specificity of in vivo targeting of 89Zr-hCD4-Mb was confirmed by PET/MRI, revealing ~4-fold greater 89Zr-hCD4-Mb uptake in subcutaneous hCD4+ hematopoietic peripheral blood acute lymphoblastic leukemia tumors (HPB-ALL) than in solid hCD4- diffuse histiocytic lymphomas (DHL) and 89Zr-mCD4-Mb uptake in hCD4+ HPB-ALL tumors. In a comparative cross-validation study in anti-programmed death ligand (αPD-L1)/anti-4-1BB-treated orthotopic PyMT mammary carcinoma-bearing hCD4-KI and WT mice, we detected 2- to 3-fold enhanced species-specific 89Zr-hCD4-Mb or 89Zr-mCD4-Mb uptake within CD4+ cell-enriched secondary lymphatic organs (lymph nodes and spleens). The 89Zr-hCD4-Mb uptake in the PyMT tumors was more pronounced in hCD4-KI mice compared to the WT control littermates. Most importantly, MC38 adenocarcinoma-bearing mice treated with a combination of αPD-L1 and anti-lymphocyte-activation gene 3 (αLag-3) antibodies exhibited ~1.4-fold higher 89Zr-mCD4-Mb uptake than mice that were not responsive to therapy or sham-treated mice. Conclusion: CD4 PET/MRI enabled monitoring of the CD4+ cell distribution in secondary lymphatic organs and the tumor microenvironment, capable of predicting sensitivity to CIT. Our imaging approach will provide deeper insights into the underlying molecular mechanisms of CD4-directed cancer immunotherapies in preclinical mouse models and is applicable for clinical translation.
Collapse
Affiliation(s)
- Stefania Pezzana
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tuebingen, University of Tuebingen, Tuebingen, Germany
| | - Simone Blaess
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tuebingen, University of Tuebingen, Tuebingen, Germany
| | - Jule Kortendieck
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tuebingen, University of Tuebingen, Tuebingen, Germany
| | - Nicole Hemmer
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tuebingen, University of Tuebingen, Tuebingen, Germany
| | - Bredi Tako
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tuebingen, University of Tuebingen, Tuebingen, Germany
- Department of Nuclear Medicine, University Hospital Tuebingen, Eberhard Karls University, Tuebingen, Germany
| | - Claudia Pietura
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tuebingen, University of Tuebingen, Tuebingen, Germany
| | - Lara Ruoff
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tuebingen, University of Tuebingen, Tuebingen, Germany
| | - Simon Riel
- Department of Dermatology, University Hospital Tuebingen, University of Tuebingen, Tuebingen, Germany
| | - Martin Schaller
- Department of Dermatology, University Hospital Tuebingen, University of Tuebingen, Tuebingen, Germany
| | - Irene Gonzalez-Menendez
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen, Germany
- Department of Pathology and Neuropathology, University Hospital Tuebingen, University of Tuebingen, Tuebingen, Germany
| | - Leticia Quintanilla-Martinez
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen, Germany
- Department of Pathology and Neuropathology, University Hospital Tuebingen, University of Tuebingen, Tuebingen, Germany
| | | | | | - Ian Wilson
- ImaginAb, Inglewood, United States of America
| | - Andreas Maurer
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tuebingen, University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen, Germany
| | - Bernd J. Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tuebingen, University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tuebingen, Tuebingen, Germany
| | - Manfred Kneilling
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tuebingen, University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen, Germany
- Department of Dermatology, University Hospital Tuebingen, University of Tuebingen, Tuebingen, Germany
| | - Dominik Sonanini
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tuebingen, University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen, Germany
- Department of Medical Oncology and Pneumology, University Hospital Tuebingen, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
40
|
Wondergem NE, Miedema IHC, van de Ven R, Zwezerijnen GJC, de Graaf P, Karagozoglu KH, Hendrickx JJ, Eerenstein SEJ, Bun RJ, Mulder DC, Voortman J, Boellaard R, Windhorst AD, Hagers JP, Peferoen LAN, de Gruijl TD, Bloemena E, Brakenhoff RH, Leemans CR, Menke-van der Houven van Oordt CW. Circulating T cell status and molecular imaging may predict clinical benefit of neoadjuvant PD-1 blockade in oral cancer. J Immunother Cancer 2024; 12:e009278. [PMID: 39038919 PMCID: PMC11268040 DOI: 10.1136/jitc-2024-009278] [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] [Accepted: 06/30/2024] [Indexed: 07/24/2024] Open
Abstract
BACKGROUND Addition of neoadjuvant immune checkpoint inhibition to standard-of-care interventions for locally advanced oral cancer could improve clinical outcome. METHODS In this study, 16 evaluable patients with stage III/IV oral cancer were treated with one dose of 480 mg nivolumab 3 weeks prior to surgery. Primary objectives were safety, feasibility, and suitability of programmed death receptor ligand-1 positron emission tomography (PD-L1 PET) as a biomarker for response. Imaging included 18F-BMS-986192 (PD-L1) PET and 18F-fluorodeoxyglucose (FDG) PET before and after nivolumab treatment. Secondary objectives included clinical and pathological response, and immune profiling of peripheral blood mononuclear cells (PBMCs) for response prediction. Baseline tumor biopsies and postnivolumab resection specimens were evaluated by histopathology. RESULTS Grade III or higher adverse events were not observed and treatment was not delayed in relation to nivolumab administration and other study procedures. Six patients (38%) had a pathological response, of whom three (19%) had a major (≥90%) pathological response (MPR). Tumor PD-L1 PET uptake (quantified using standard uptake value) was not statistically different in patients with or without MPR (median 5.3 vs 3.4). All major responders showed a significantly postnivolumab decreased signal on FDG PET. PBMC immune phenotyping showed higher levels of CD8+ T cell activation in MPR patients, evidenced by higher baseline expression levels of PD-1, TIGIT, IFNγ and lower levels of PD-L1. CONCLUSION Together these data support that neoadjuvant treatment of advanced-stage oral cancers with nivolumab was safe and induced an MPR in a promising 19% of patients. Response was associated with decreased FDG PET uptake as well as activation status of peripheral T cell populations.
Collapse
Affiliation(s)
- Niels E Wondergem
- Amsterdam UMC location Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Iris H C Miedema
- Amsterdam UMC location Vrije Universiteit Amsterdam, Medical Oncology, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Rieneke van de Ven
- Amsterdam UMC location Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, Netherlands
| | - Gerben J C Zwezerijnen
- Cancer Center Amsterdam, Imaging and Biomarkers, Cancer Centre Amsterdam, Amsterdam, The Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Pim de Graaf
- Cancer Center Amsterdam, Imaging and Biomarkers, Cancer Centre Amsterdam, Amsterdam, The Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, De Boelelaan 1117, Amsterdam, The Netherlands
| | - K Hakki Karagozoglu
- Amsterdam UMC and Academic Centre for Dentistry Amsterdam (ACTA), Oral and Maxillofacial Surgery/Oral Pathology, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Jan-Jaap Hendrickx
- Amsterdam UMC location Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Simone E J Eerenstein
- Amsterdam UMC location Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Rolf J Bun
- Oral and Maxillofacial Surgery, Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands
| | - Dorien C Mulder
- Oral and Maxillofacial Surgery, Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands
| | - Jens Voortman
- Amsterdam UMC location Vrije Universiteit Amsterdam, Medical Oncology, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Cancer Center Amsterdam, Imaging and Biomarkers, Cancer Centre Amsterdam, Amsterdam, The Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Albert D Windhorst
- Cancer Center Amsterdam, Imaging and Biomarkers, Cancer Centre Amsterdam, Amsterdam, The Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, De Boelelaan 1117, Amsterdam, The Netherlands
| | - J Pascal Hagers
- Amsterdam UMC location Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Laura A N Peferoen
- Amsterdam UMC location Vrije Universiteit Amsterdam, Pathology, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Tanja D de Gruijl
- Amsterdam UMC location Vrije Universiteit Amsterdam, Medical Oncology, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Elisabeth Bloemena
- Amsterdam UMC location Vrije Universiteit Amsterdam, Pathology, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Ruud H Brakenhoff
- Amsterdam UMC location Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - C René Leemans
- Amsterdam UMC location Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - C Willemien Menke-van der Houven van Oordt
- Amsterdam UMC location Vrije Universiteit Amsterdam, Medical Oncology, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Cancer Centre Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
41
|
Glazer SE, Kummar S, Mittra E. Illuminating immunotherapy response via precision T cell-targeted PET imaging. Front Med (Lausanne) 2024; 11:1233913. [PMID: 39104861 PMCID: PMC11298440 DOI: 10.3389/fmed.2024.1233913] [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: 06/03/2023] [Accepted: 06/20/2024] [Indexed: 08/07/2024] Open
Abstract
Traditionally, immunotherapy agent selection and treatment strategies are guided by biopsy-based histological information. However, biopsies are limited in that they are invasive, provide static information regarding the tumor immune microenvironment, and only sample a small part of one tumor site. The tumor microenvironment is dynamic and heterogenous. As a result, the immune milieu at one site may be distinct from other metastatic sites. These factors make identifying which patients are likely to respond to different immunotherapies and which harbor intrinsic resistance mechanisms difficult to identify based on a biopsy alone. As such, there is significant interest in alternative methodologies that better characterize the tumor immune microenvironment and monitor immunotherapy response. PET imaging potentially offers a non-invasive way to characterize the tumor immune microenvironment at the primary tumor and metastases and allow for longitudinal characterization. Herein, we review pre-clinically and clinically tested T cell-targeted PET radiopharmaceuticals, as T cells have been the dominant immunotherapy target, and their utility in both evaluating response to immunotherapy and in understanding the systemic immune response to treatment with immunotherapeutics.
Collapse
Affiliation(s)
- Sarah E. Glazer
- Division of Internal Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Shivaani Kummar
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Erik Mittra
- Division of Molecular Imaging and Therapy, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
42
|
Zhang X, Wang P, Shi G, Tang C, Xue H. AUNP-12 Near-Infrared Fluorescence Probes across NIR-I to NIR-II Enable In Vivo Detection of PD-1/PD-L1 Axis in the Tumor Microenvironment. Bioconjug Chem 2024; 35:1064-1074. [PMID: 38980173 PMCID: PMC11261610 DOI: 10.1021/acs.bioconjchem.4c00266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 06/18/2024] [Accepted: 06/21/2024] [Indexed: 07/10/2024]
Abstract
The innovative PD-1/PD-L1 pathway strategy is gaining significant traction in cancer therapeutics. However, fluctuating response rates of 20-40% to PD-1/PD-L1 inhibitors, coupled with the risk of hyperprogression after immunotherapy, underscore the need for accurate patient selection and the identification of more beneficiaries. Molecular imaging, specifically near-infrared (NIR) fluorescence imaging, is a valuable alternative for real-time, noninvasive visualization of dynamic PD-L1 expression in vivo. This research introduces AUNP-12, a novel PD-L1-targeting peptide antagonist conjugated with Cy5.5 and CH1055 for first (NIR-I) and second near-infrared (NIR-II) imaging. These probes have proven to be effective in mapping PD-L1 expression across various mouse tumor models, offering insights into tumor-immune interactions. This study highlights the potential of AUNP-12-Cy5.5 and AUNP-12-CH1055 for guiding clinical immunotherapy through precise patient stratification and dynamic monitoring, supporting the shift toward molecular imaging for personalized cancer care.
Collapse
Affiliation(s)
- Xinyu Zhang
- Department
of Radiology, State Key Laboratory of Complex Severe and Rare Diseases,
Peking Union Medical College Hospital, Peking
Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Ping Wang
- Department
of Cardiology, The Second Medical Center & National Clinical Research
Center for Geriatric Diseases, Chinese People’s
Liberation Army (PLA) General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Guangyuan Shi
- University
of Science and Technology of China, 96 Jinzhai Road, Hefei 230026, China
| | - Chu Tang
- Engineering
Research Center of Molecular and Neuro Imaging of Ministry of Education,
School of Life Science and Technology, Xidian
University, Xi’an 710126, China
| | - Huadan Xue
- Department
of Radiology, State Key Laboratory of Complex Severe and Rare Diseases,
Peking Union Medical College Hospital, Peking
Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| |
Collapse
|
43
|
Beer L, Hochmair M, Heilmann M, Hoda MA. ESR Bridges: imaging and treatment of lung cancer-a multidisciplinary view. Eur Radiol 2024; 34:4230-4232. [PMID: 38355985 DOI: 10.1007/s00330-023-10523-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 11/11/2023] [Accepted: 11/23/2023] [Indexed: 02/16/2024]
Affiliation(s)
- Lucian Beer
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria.
- Christian Doppler Laboratory for Machine Learning Driven Precision Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria.
| | - Maximilian Hochmair
- Department of Respiratory and Critical Care Medicine, Karl Landsteiner Institute of Lung Research and Pulmonary Oncology, Klinik Floridsdorf, Vienna, Austria
| | - Martin Heilmann
- Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
| | - Mir Ali Hoda
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
44
|
Miedema IHC, Pouw JEE, Kwakman A, Zwezerijnen GJC, Huisman MC, Timmer FEF, van de Ven R, de Gruijl TD, Hospers GAP, de Langen AJ, Menke-van der Houven van Oordt CW. Exploring the predictive potential of programmed death ligand 1 expression in healthy organs and lymph nodes as measured by 18F-BMS986-192 PET: pooled analysis of data from four solid tumor types. J Immunother Cancer 2024; 12:e008899. [PMID: 38886117 PMCID: PMC11184194 DOI: 10.1136/jitc-2024-008899] [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] [Accepted: 05/28/2024] [Indexed: 06/20/2024] Open
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) can elicit anticancer immune responses, but predictive biomarkers are needed. We measured programmed death ligand 1 (PD-L1) expression in organs and lymph nodes using 18F-BMS-986192 positron emission tomography (PET)-imaging and looked for correlations with response and immune-related adverse events. METHODS Four 18F-BMS-986192 PET studies in patients with melanoma, lung, pancreatic and oral cancer, receiving ICI treatment, were combined. Imaging data (organ standardized uptake value (SUV)mean, lymph node SUVmax) and clinical data (response to treatment and incidence of immune-related adverse events) were extracted. RESULTS Baseline PD-L1 uptake in the spleen was on average higher in non-responding patients than in responders (spleen SUVmean 16.1±4.4 vs 12.5±3.4, p=0.02). This effect was strongest in lung cancer, and not observed in oral cancer. In the oral cancer cohort, benign tumor-draining lymph nodes (TDLNs) had higher PD-L1 uptake (SUVmax 3.3 IQR 2.5-3.9) compared with non-TDLNs (SUVmax 1.8, IQR 1.4-2.8 p=0.04). Furthermore, in the same cohort non-responders showed an increase in PD-L1 uptake in benign TDLNs on-treatment with ICIs (+15%), while for responders the PD-L1 uptake decreased (-11%). PD-L1 uptake did not predict immune-related adverse events, though elevated thyroid uptake on-treatment correlated with pre-existing thyroid disease or toxicity. CONCLUSION PD-L1 PET uptake in the spleen is a potential negative predictor of response to ICIs. On-treatment with ICIs, PD-L1 uptake in benign TDLNs increases in non-responders, while it decreases in responders, potentially indicating a mechanism for resistance to ICIs in patients with oral cancer.
Collapse
Affiliation(s)
- Iris H C Miedema
- Department of Medical Oncology, Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Johanna E E Pouw
- Department of Medical Oncology, Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Anne Kwakman
- Department of Medical Oncology, Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Gerben J C Zwezerijnen
- Imaging and Biomarkers, Cancer Centre Amsterdam, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Marc C Huisman
- Imaging and Biomarkers, Cancer Centre Amsterdam, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Florentine E F Timmer
- Imaging and Biomarkers, Cancer Centre Amsterdam, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Rieneke van de Ven
- Otolaryngology / Head and Neck Surgery, Amsterdam UMC - Locatie VUMC, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Geke A P Hospers
- Medical Oncology, University Medical Centre Groningen, Groningen, The Netherlands
| | | | | |
Collapse
|
45
|
Ibrahim D, Simó C, Brown EL, Shmuel S, Panikar SS, Benton A, DeWeerd R, Dehdashti F, Park H, Pereira PMR. PD-L1 has a heterogeneous and dynamic expression in gastric cancer with implications for immunoPET. Front Immunol 2024; 15:1405485. [PMID: 38915392 PMCID: PMC11194338 DOI: 10.3389/fimmu.2024.1405485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/21/2024] [Indexed: 06/26/2024] Open
Abstract
Introduction This study aimed to investigate the dynamics of programmed death-ligand 1 (PD-L1) expression, spatial heterogeneity, and binding affinity of FDA-approved anti-PD-L1 antibodies (avelumab and atezolizumab) in gastric cancer. Additionally, we determined how PD-L1 glycosylation impacts antibody accumulation in gastric cancer cells. Methods Dynamic PD-L1 expression was examined in NCIN87 gastric cancer cells. Comparative binding studies of avelumab and atezolizumab were conducted in gastric cancer models, both in vitro and in vivo. Antibody uptake in tumors was visualized through positron emission tomography (PET) imaging. PD-L1 glycosylation status was determined via Western blot analyses before and after PNGase F treatment. Results Consistent findings revealed time-dependent PD-L1 induction in NCIN87 gastric cancer cells and spatial heterogeneity in tumors, as shown by PET imaging and immunofluorescence. Avelumab displayed superior binding affinity to NCIN87 cells compared to atezolizumab, confirmed by in vivo PET imaging and ex vivo biodistribution analyses. Notably, PD-L1 glycosylation at approximately 50 kDa was observed, with PNGase F treatment inducing a shift to 35 kDa in molecular weight. Tissue samples from patient-derived xenografts (PDXs) validated the presence of both glycosylated and deglycosylated PD-L1 (degPD-L1) forms in gastric cancer. Immunofluorescence microscopy and binding assays demonstrated enhanced avelumab binding post-deglycosylation. Discussion This study provides an understanding of dynamic and spatially heterogeneous PD-L1 expression in gastric cancer. Anti-PD-L1 immunoPET was able to visualize gastric tumors, and PD-L1 glycosylation has significant implications for antibody recognition. These insights contribute to demonstrating the complexities of PD-L1 in gastric cancer, holding relevance for refining PD-L1 imaging-based approaches.
Collapse
Affiliation(s)
- Dina Ibrahim
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Cristina Simó
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Emma L. Brown
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Shayla Shmuel
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Sandeep Surendra Panikar
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Alex Benton
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
- Cancer Biology Graduate Program, Washington University School of Medicine, St. Louis, MO, United States
| | - Rachel DeWeerd
- Cancer Biology Graduate Program, Washington University School of Medicine, St. Louis, MO, United States
| | - Farrokh Dehdashti
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Haeseong Park
- Gastrointestinal Cancer Center, Center for Cancer Therapeutic Innovation, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Patrícia M. R. Pereira
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
46
|
Badenhorst M, Windhorst AD, Beaino W. Navigating the landscape of PD-1/PD-L1 imaging tracers: from challenges to opportunities. Front Med (Lausanne) 2024; 11:1401515. [PMID: 38915766 PMCID: PMC11195831 DOI: 10.3389/fmed.2024.1401515] [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: 03/15/2024] [Accepted: 05/20/2024] [Indexed: 06/26/2024] Open
Abstract
Immunotherapy targeted to immune checkpoint inhibitors, such as the program cell death receptor (PD-1) and its ligand (PD-L1), has revolutionized cancer treatment. However, it is now well-known that PD-1/PD-L1 immunotherapy response is inconsistent among patients. The current challenge is to customize treatment regimens per patient, which could be possible if the PD-1/PD-L1 expression and dynamic landscape are known. With positron emission tomography (PET) imaging, it is possible to image these immune targets non-invasively and system-wide during therapy. A successful PET imaging tracer should meet specific criteria concerning target affinity, specificity, clearance rate and target-specific uptake, to name a few. The structural profile of such a tracer will define its properties and can be used to optimize tracers in development and design new ones. Currently, a range of PD-1/PD-L1-targeting PET tracers are available from different molecular categories that have shown impressive preclinical and clinical results, each with its own advantages and disadvantages. This review will provide an overview of current PET tracers targeting the PD-1/PD-L1 axis. Antibody, peptide, and antibody fragment tracers will be discussed with respect to their molecular characteristics and binding properties and ways to optimize them.
Collapse
Affiliation(s)
- Melinda Badenhorst
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, De Boelelaan, Amsterdam, Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, Netherlands
| | - Albert D. Windhorst
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, De Boelelaan, Amsterdam, Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, Netherlands
| | - Wissam Beaino
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, De Boelelaan, Amsterdam, Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, Netherlands
| |
Collapse
|
47
|
Sun Z, Xiao X, Liang S, Ma H, Sun Y, Zhao L, Wang C, Chang X, Zhao H, Guo H, Zhang Z. Consistency Analysis of Programmed Death Ligand 1 Expression in Non-Small Cell Lung Cancer Between Pleural Effusion and Matched Primary Lung Cancer Tissues by Immunohistochemical Double Staining. J Transl Med 2024; 104:102058. [PMID: 38626874 DOI: 10.1016/j.labinv.2024.102058] [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: 08/18/2023] [Revised: 02/14/2024] [Accepted: 04/05/2024] [Indexed: 05/12/2024] Open
Abstract
In clinical practice, programmed death ligand 1 (PD-L1) detection is prone to nonspecific staining due to the complex cellular composition of pleural effusion smears. In this study, diaminobenzidine (DAB) and 3-amino-9-ethylcarbazole (AEC) immunohistochemistry double staining was performed to investigate PD-L1 expression in tumor cells from malignant pleural effusion (MPE). MPE was considered as a metastasis in non-small cell lung cancer patients; thus, the heterogeneity between metastatic and primary lung cancer was revealed as well. Ninety paired specimens of MPE cell blocks and matched primary lung cancer tissues from non-small cell lung cancer patients were subjected to PD-L1 and thyroid transcription factor-1(TTF-1)/p63 immunohistochemistry double staining. Two experienced pathologists independently evaluated PD-L1 expression using 3 cutoffs (1%, 10%, and 50%). PD-L1 expression in MPE was strongly correlated with that in matched primary lung cancer tissues (R = 0.813; P < .001). Using a 4-tier scale (cutoffs: 1%, 10%, and 50%), the concordance was 71.1% (Cohen's κ = .534). Using a 2-tier scale, the concordance was 75.6% (1%, Cohen's κ = 0.53), 78.9% (10%, Cohen's κ = 0.574), and 95.6% (50%, Cohen's κ = 0.754). The rates of PD-L1 positivity in MPE (56.7%) were higher than that in lung tissues (32.2%). All 27 discordant cases had higher scores in MPE. The double-staining method provided superior identification of PD-L1-positive tumor cells on a background with nonspecific staining. In conclusion, PD-L1 expression was moderately concordant between metastatic MPE cell blocks and matched primary lung carcinoma tissues, with variability related to tumor heterogeneity. MPE should be considered to detect PD-L1 when histological specimens are unattainable, especially when PD-L1 expression is >50%. PD-L1 positivity rates were higher in MPE. Double staining can improve PD-L1 detection by reducing false-negative/positive results.
Collapse
Affiliation(s)
- Zihan Sun
- Cytopathology Section, Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoyue Xiao
- Cytopathology Section, Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuo Liang
- Cytopathology Section, Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haiyue Ma
- Cytopathology Section, Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue Sun
- Cytopathology Section, Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Linlin Zhao
- Cytopathology Section, Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cong Wang
- Cytopathology Section, Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinxiang Chang
- Cytopathology Section, Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huan Zhao
- Cytopathology Section, Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huiqin Guo
- Cytopathology Section, Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhihui Zhang
- Cytopathology Section, Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
48
|
Santos L, Moreira JN, Abrunhosa A, Gomes C. Brain metastasis: An insight into novel molecular targets for theranostic approaches. Crit Rev Oncol Hematol 2024; 198:104377. [PMID: 38710296 DOI: 10.1016/j.critrevonc.2024.104377] [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: 12/05/2023] [Revised: 04/11/2024] [Accepted: 04/25/2024] [Indexed: 05/08/2024] Open
Abstract
Brain metastases (BrM) are common malignant lesions in the central nervous system, and pose a significant threat in advanced-stage malignancies due to delayed diagnosis and limited therapeutic options. Their distinct genomic profiles underscore the need for molecular profiling to tailor effective treatments. Recent advances in cancer biology have uncovered molecular drivers underlying tumor initiation, progression, and metastasis. This, coupled with the advances in molecular imaging technology and radiotracer synthesis, has paved the way for the development of innovative radiopharmaceuticals with enhanced specificity and affinity for BrM specific targets. Despite the challenges posed by the blood-brain barrier to effective drug delivery, several radiolabeled compounds have shown promise in detecting and targeting BrM. This manuscript provides an overview of the recent advances in molecular biomarkers used in nuclear imaging and targeted radionuclide therapy in both clinical and preclinical settings. Additionally, it explores potential theranostic applications addressing the unique challenges posed by BrM.
Collapse
Affiliation(s)
- Liliana Santos
- Institute for Nuclear Sciences Applied to Health (ICNAS) and Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra 3000-548, Portugal; Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra 3000-548, Portugal
| | - João Nuno Moreira
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra 3004-504, Portugal; Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, Coimbra 3000-548, Portugal
| | - Antero Abrunhosa
- Institute for Nuclear Sciences Applied to Health (ICNAS) and Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra 3000-548, Portugal
| | - Célia Gomes
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra 3000-548, Portugal; Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, Coimbra 3000-548, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra 3000-075, Portugal.
| |
Collapse
|
49
|
Eertink JJ, Bahce I, Waterton JC, Huisman MC, Boellaard R, Wunder A, Thiele A, Menke-van der Houven van Oordt CW. The development process of 'fit-for-purpose' imaging biomarkers to characterize the tumor microenvironment. Front Med (Lausanne) 2024; 11:1347267. [PMID: 38818386 PMCID: PMC11138661 DOI: 10.3389/fmed.2024.1347267] [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: 11/30/2023] [Accepted: 04/24/2024] [Indexed: 06/01/2024] Open
Abstract
Immune-based treatment approaches are successfully used for the treatment of patients with cancer. While such therapies can be highly effective, many patients fail to benefit. To provide optimal therapy choices and to predict treatment responses, reliable biomarkers for the assessment of immune features in patients with cancer are of significant importance. Biomarkers (BM) that enable a comprehensive and repeatable assessment of the tumor microenvironment (TME), the lymphoid system, and the dynamics induced by drug treatment can fill this gap. Medical imaging, notably positron emission tomography (PET) and magnetic resonance imaging (MRI), providing whole-body imaging BMs, might deliver such BMs. However, those imaging BMs must be well characterized as being 'fit for purpose' for the intended use. This review provides an overview of the key steps involved in the development of 'fit-for-purpose' imaging BMs applicable in drug development, with a specific focus on pharmacodynamic biomarkers for assessing the TME and its modulation by immunotherapy. The importance of the qualification of imaging BMs according to their context of use (COU) as defined by the Food and Drug Administration (FDA) and National Institutes of Health Biomarkers, EndpointS, and other Tools (BEST) glossary is highlighted. We elaborate on how an imaging BM qualification for a specific COU can be achieved.
Collapse
Affiliation(s)
- Jakoba J. Eertink
- Department of Medical Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Idris Bahce
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, Netherlands
- Department of Pulmonary Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - John C. Waterton
- Centre for Imaging Sciences, University of Manchester, Manchester, United Kingdom
| | - Marc C. Huisman
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Ronald Boellaard
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Andreas Wunder
- Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach and der Riss, Germany
| | - Andrea Thiele
- Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach and der Riss, Germany
| | - Catharina W. Menke-van der Houven van Oordt
- Department of Medical Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
50
|
Tang S, Fan T, Wang X, Yu C, Zhang C, Zhou Y. Cancer Immunotherapy and Medical Imaging Research Trends from 2003 to 2023: A Bibliometric Analysis. J Multidiscip Healthc 2024; 17:2105-2120. [PMID: 38736544 PMCID: PMC11086400 DOI: 10.2147/jmdh.s457367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/16/2024] [Indexed: 05/14/2024] Open
Abstract
Purpose With the rapid development of immunotherapy, cancer treatment has entered a new phase. Medical imaging, as a primary diagnostic method, is closely related to cancer immunotherapy. However, until now, there has been no systematic bibliometric analysis of the state of this field. Therefore, the main purpose of this article is to clarify the past research trajectory, summarize current research hotspots, reveal dynamic scientific developments, and explore future research directions. Patients and Methods A comprehensive search was conducted on the Web of Science Core Collection (WoSCC) database to identify publications related to immunotherapy specifically for the medical imaging of carcinoma. The search spanned the period from the year 2003 to 2023. Several analytical tools were employed. These included CiteSpace (6.2.4), and the Microsoft Office Excel (2016). Results By searching the database, a total of 704 English articles published between 2003 and 2023 were obtained. We have observed a rapid increase in the number of publications since 2018. The two most active countries are the United States (n=265) and China (n=170). Pittock, Sean J and Abu-sbeih, Hamzah are very concerned about the relationship between cancer immunotherapy and medical images and have published more academic papers (n = 5; n = 4). Among the top 10 co-cited authors, Topalian Sl (n=43) cited ranked first, followed by Graus F (n=40) cited. According to clustering, timeline, and burst word analysis, the results show that the current research focus is on "MRI", "deep learning", "tumor microenvironment" and so on. Conclusion Medical imaging and cancer immunotherapy are hot topics. The United States is the country with the most publications and the greatest influence in this field, followed by China. "MRI", "PET/PET-CT", "deep learning", "immune-related adverse events" and "tumor microenvironment" are currently hot research topics and potential targets.
Collapse
Affiliation(s)
- Shuli Tang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150010, People’s Republic of China
| | - Tiantian Fan
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150010, People’s Republic of China
| | - Xinxin Wang
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150010, People’s Republic of China
| | - Can Yu
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150010, People’s Republic of China
| | - Chunhui Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150010, People’s Republic of China
| | - Yang Zhou
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150010, People’s Republic of China
| |
Collapse
|