1
|
Guan J, Gong X, Zeng H, Zhang W, Qin Q, Gou H, Liu X, Song B. Gastrointestinal tumor personalized immunotherapy: an integrated analysis from molecular genetics to imaging biomarkers. Therap Adv Gastroenterol 2025; 18:17562848251333527. [PMID: 40297204 PMCID: PMC12035075 DOI: 10.1177/17562848251333527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
The immunotherapy landscape for gastrointestinal (GI) tumors is rapidly evolving. There is an urgent need for reliable biomarkers capable of predicting treatment outcomes to optimize therapeutic strategies and enhance patient prognosis. This review presents a comprehensive overview of biomarkers associated with the immunotherapy response of GI tumors, covering advances in molecular genetics, histopathological markers, and imaging. Key molecular biomarkers, such as microsatellite instability, tumor mutational burden, and programmed death-ligand 1 expression, remain critical for identifying patients likely to benefit from immune checkpoint inhibitors. The significance of tumor-infiltrating lymphocytes, notably the CD8+ T cell to regulatory T cell ratio, as a predictor of immunotherapy response is explored. In addition, advanced imaging techniques, including computed tomography (CT), magnetic resonance imaging, and positron emission tomography-CT, facilitate the noninvasive evaluation of tumor biology and therapeutic response. By bridging molecular and imaging data, this integrated strategy enhances precision in patient selection, treatment monitoring, and adaptive therapy design. Future studies should aim to validate these biomarkers in larger, multicenter cohorts and focus on clinical translation to advance precision medicine in GI oncology.
Collapse
Affiliation(s)
- Jian Guan
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiology, Sichuan Provincial Corps Hospital, Chinese People’s Armed Police Forces, Leshan, China
| | - Xiaoling Gong
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Hanjiang Zeng
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Zhang
- Department of Radiology, Sichuan Provincial Corps Hospital, Chinese People’s Armed Police Forces, Leshan, China
| | - Qing Qin
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hongfeng Gou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xijiao Liu
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu 610041, China
- Department of Radiology, Sanya People’s Hospital, Sanya, Hainan, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu 610041, China
- Department of Radiology, Sanya People’s Hospital, Sanya, Hainan, China
| |
Collapse
|
2
|
Jia F, Jing S, Guo J. m6A regulator-mediated methylation modifications define the immune infiltration characteristics of the tumor microenvironment in prostate adenocarcinoma. Sci Rep 2024; 14:29047. [PMID: 39580517 PMCID: PMC11585623 DOI: 10.1038/s41598-024-77688-w] [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: 08/05/2024] [Accepted: 10/24/2024] [Indexed: 11/25/2024] Open
Abstract
Prostate adenocarcinoma (PRAD) persists as the predominant non-cutaneous malignancy diagnosed in males, which is a primary contributor to cancer-related mortality globally. It is reported that the progression of prostate adenocarcinoma is associated with various factors, including genetics, age, obesity, etc. Contemporary research indicates that epigenetic inheritance is a leading factor in the initiation and progression of cancer. RNA methylation modification is the most prevalent form of RNA modification, with N6-methyladenosine (m6A) representing the most common modification on mRNA and lncRNAs. However, the biological mechanisms underpinning this association in prostate adenocarcinoma and its correlation with patients' prognostic survival outcomes remain elusive. Our study elucidates the roles of the tumor microenvironment (TME) and genetic mutations during the initiation and progression of prostate adenocarcinoma. Additionally, we stratify prostate adenocarcinoma into distinct subtypes based on m6A scoring. This approach enhances our comprehension of the functional role of m6A in the development of prostate adenocarcinoma, offering novel insights into the clinical strategies and understanding the biological significance between prostate adenocarcinoma and m6A modification.
Collapse
Affiliation(s)
- Fajing Jia
- Department of General Medical, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences,Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shaoze Jing
- Department of Orthopedics, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jianjin Guo
- Department of General Medical, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences,Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China.
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
3
|
Wu C, Wen F, Lin F, Zeng Y, Lin X, Hu X, Zhang X, Zhang X, Wang X. Predictive performance of [ 18F]F-fibroblast activation protein inhibitor (FAPI)-42 positron emission tomography/computed tomography (PET/CT) in evaluating response of recurrent or metastatic gastrointestinal stromal tumors: complementary or alternative to [ 18F]fluorodeoxyglucose (FDG) PET/CT? Quant Imaging Med Surg 2024; 14:5333-5345. [PMID: 39144061 PMCID: PMC11320500 DOI: 10.21037/qims-24-192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 05/22/2024] [Indexed: 08/16/2024]
Abstract
Background Accurately and promptly predicting the response of gastrointestinal stromal tumors (GISTs) to targeted therapy is essential for optimizing treatment strategies. However, some fractions of recurrent or metastatic GISTs present as non-FDG-avid lesions, limiting the value of [18F]fluorodeoxyglucose positron emission tomography/computed tomography ([18F]FDG PET/CT) in treatment evaluation. This study evaluated the efficacy of [18F]F-fibroblast activation protein inhibitor (FAPI)-42 [18F]FAPI-42) PET/CT for assessing the treatment response in recurrent or metastatic GISTs, in comparison to [18F]FDG PET/CT and explores a model integrating PET/CT imaging and clinical parameters to optimize the clinical use of these diagnostic tools. Methods Our retrospective analysis included 27 patients with recurrent or metastatic GISTs who underwent [18F]FAPI-42 PET/CT and [18F]FDG PET/CT at baseline before switching targeted therapy. Treatment response status was divided into a progression group (PG) and a non-progression group (NPG) based on the Response Criteria in Solid Tumors (RECIST) 1.1, according to the contrast-enhanced computed tomography (CT) scan at six months. [18F]FAPI-42 and [18F]FDG PET/CT parameters including the mean standardized uptake value (SUVmean), the standard uptake value corrected for lean body mass (SULpeak), the maximum standardized uptake value (SUVmax), tumor-to-blood pool SUV ratio (TBR), tumor-to-liver SUV ratio (TLR), metabolic tumor volume (MTV)/FAPI-positive tumor volume (GTV-FAPI), total lesion glycolysis (TLG)/FAPI-positive total lesion accumulation (TLF) were correlated with the response status to identify indicative of treatment response. The predictive performance of them was quantified by generating receiver operating characteristic curves (ROC), calibration curves, and cross-validation. Results A total of 110 lesions were identified in 27 patients. Compared with PG, NPG was associated with lower levels of TBR and SUVmean in FDG PET/CT (TBR-FDG, SUVmean-FDG; P=0.033 and P=0.038, respectively), with higher SULpeak and TLF in FAPI PET/CT (SULpeak-FAPI, TLF-FAPI; P=0.10 and P=0.049, respectively). The predictive power of a composite-parameter model, including TBR-FDG, SULpeak-FAPI, gene mutation, and type of targeted therapy [area under the curve (AUC) =0.865], was superior to the few-parameter models incorporating TBR-FDG (AUC =0.637, P<0.001), SULpeak-FAPI (AUC =0.665, P<0.001) or both (AUC =0.721, P<0.001). Conclusions Both [18F]FAPI-42 PET/CT and [18F]FDG PET/CT have value in predicting the treatment response of recurrent or metastatic GISTs. And [18F]FAPI-42 PET/CT offers synergistic value when used in combination with [18F]FDG PET/CT. Notably, the nomogram generated from the model incorporating [18F]FAPI-42 PET/CT, [18F]FDG PET/CT parameters, gene mutation, and type of targeted therapy could yield more precise predictions of the response of recurrent metastatic GISTs.
Collapse
Affiliation(s)
- Chunhui Wu
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Fang Wen
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Fangzeng Lin
- Department of Radiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yu Zeng
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaojie Lin
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xin Hu
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiangsong Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xinhua Zhang
- Center of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoyan Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
4
|
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
|
5
|
Janani M, Poorkhani A, Amiriani T, Donyadideh G, Ahmadi F, Jorjanisorkhankalateh Y, Beheshti-Nia F, Kalaei Z, Roudbaraki M, Soltani M, Khori V, Alizadeh AM. Association of future cancer metastases with fibroblast activation protein-α: a systematic review and meta-analysis. Front Oncol 2024; 14:1339050. [PMID: 38751814 PMCID: PMC11094201 DOI: 10.3389/fonc.2024.1339050] [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: 11/15/2023] [Accepted: 04/04/2024] [Indexed: 05/18/2024] Open
Abstract
Introduction Fibroblast activation protein-α (FAP-α) is a vital surface marker of cancer-associated fibroblasts, and its high expression is associated with a higher tumor grade and metastasis. A systematic review and a meta-analysis were performed to associate future metastasis with FAP-α expression in cancer. Methods In our meta-analysis, relevant studies published before 20 February 2024 were systematically searched through online databases that included PubMed, Scopus, and Web of Science. The association between FAP-α expression and metastasis, including distant metastasis, lymph node metastasis, blood vessel invasion, vascular invasion, and neural invasion, was evaluated. A pooled odds ratio (OR) with 95% confidence intervals (CI) was reported as the measure of association. Results A total of 28meta-analysis. The random-effects model for five parameters showed that a high FAP-α expression was associated with blood vessel invasion (OR: 3.04, 95% CI: 1.54-5.99, I 2 = 63%, P = 0.001), lymphovascular invasion (OR: 3.56, 95% CI: 2.14-5.93, I 2 = 0.00%, P < 0.001), lymph node metastasis (OR: 2.73, 95% CI: 1.96-3.81, I 2 = 65%, P < 0.001), and distant metastasis (OR: 2.59; 95% CI: 1.16-5.79, I 2 = 81%, P < 0.001). However, our analysis showed no statistically significant association between high FAP-α expression and neural invasion (OR: 1.57, 95% CI: 0.84-2.93, I 2 = 38%, P = 0.161). Conclusions This meta-analysis indicated that cancer cells with a high FAP-α expression have a higher risk of metastasis than those with a low FAP-α expression. These findings support the potential importance of FAP-α as a biomarker for cancer metastasis prediction.
Collapse
Affiliation(s)
- Majid Janani
- Breast Disease Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirhoushang Poorkhani
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Taghi Amiriani
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ghazaleh Donyadideh
- Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farahnazsadat Ahmadi
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | | | - Fereshteh Beheshti-Nia
- Department of Epidemiology and Biostatistics, School of Public Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Kalaei
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Morad Roudbaraki
- Laboratory of Cell Physiology, Inserm U1003, University of Lille, Villeneuve d’Ascq, France
| | - Mahsa Soltani
- Breast Disease Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Khori
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ali Mohammad Alizadeh
- Breast Disease Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Bentestuen M, Nalliah S, Stolberg MMK, Zacho HD. How to Perform FAPI PET? An Expedited Systematic Review Providing a Recommendation for FAPI PET Imaging With Different FAPI Tracers. Semin Nucl Med 2024; 54:345-355. [PMID: 38052711 DOI: 10.1053/j.semnuclmed.2023.11.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 11/13/2023] [Indexed: 12/07/2023]
Abstract
This expedited systematic review aims to provide the first overview of the different Fibroblast activation protein inhibitor (FAPI) PET scan procedures in the literature and discuss how to efficiently obtain optimal FAPI PET images based on the best available evidence. The PubMed, Embase, Cochrane Library, and Web of Science databases were systematically searched in April 2023. Peer-reviewed cohort studies published in English and used FAPI tracers were included. Articles were excluded if critical scan procedure information was missing, or the article was not retrievable from a university library within 30 days. Data were grouped according to the FAPI tracer applied. Meta-analysis with proper statistics was deemed not feasible based on a pilot study. A total of 946 records were identified. After screening, 159 studies were included. [68Ga]Ga-FAPI-04 was applied in 98 studies (61%), followed by [68Ga]Ga-FAPI-46 in 19 studies (12%). Most studies did not report specific patient preparation. A mean/median administered activity of 80-200 MBq was most common; however, wide ranges were seen in [68Ga]Ga-FAPI-04 PET studies (56-370 MBq). An injection-to-scan-time of 60 minutes was dominant for all FAPI PET studies. A possible trend toward shorter injection-to-scan times was observed for [68Ga]Ga-FAPI-46. Three studies evaluated [68Ga]Ga-FAPI-46 PET acquisition at multiple time points in more than 593 cancer lesions, all yielding equivalent tumor detection at 10 minutes vs later time points despite slightly lower tumor-to-background Ratios. Despite the wide ranges, most institutions administer an average of 80-200 MBq [68Ga]Ga-FAPI-04/46 and scan patients at 60 minutes postinjection. For [68Ga]Ga-FAPI-46, the present evidence consistently supports the feasibility of image acquisition earlier than 30 minutes. Currently, data on the optimal FAPI PET scan procedure are limited, and more studies are encouraged. The current review can serve as a temporary guideline for institutions planning FAPI PET studies.
Collapse
Affiliation(s)
- Morten Bentestuen
- Department of Nuclear Medicine and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| | - Surenth Nalliah
- Department of Radiology, Aalborg University Hospital, Aalborg, Denmark
| | - Marie M K Stolberg
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Helle D Zacho
- Department of Nuclear Medicine and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
7
|
Rizzo A, Miceli A, Racca M, Bauckneht M, Morbelli S, Albano D, Dondi F, Bertagna F, Galizia D, Muoio B, Annunziata S, Treglia G. Diagnostic Accuracy of [ 68Ga]Ga Labeled Fibroblast-Activation Protein Inhibitors in Detecting Head and Neck Cancer Lesions Using Positron Emission Tomography: A Systematic Review and a Meta-Analysis. Pharmaceuticals (Basel) 2023; 16:1664. [PMID: 38139791 PMCID: PMC10748043 DOI: 10.3390/ph16121664] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 11/26/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
Several studies have examined the use of positron emission tomography (PET) using [68Ga]Ga-radiolabeled fibroblast-activation protein inhibitors (FAPi) across multiple subtypes of head and neck cancer (HNC). The purpose of the present study was to evaluate the diagnostic accuracy of a newly developed molecular imaging approach in the context of HNC through a comprehensive review and meta-analysis. A thorough literature review was conducted to identify scholarly articles about the diagnostic effectiveness of FAP-targeted PET imaging. The present study incorporates original publications assessing the efficacy of this innovative molecular imaging test in both newly diagnosed and previously treated HNC patients. This systematic review examined eleven investigations, of which nine were deemed suitable for inclusion in the subsequent meta-analysis. The quantitative synthesis yielded a pooled detection rate of 99% for primary HNC lesions. Additionally, on a per patient-based analysis, the pooled sensitivity and specificity for regional lymph node metastases were found to be 90% and 84%, respectively. The analysis revealed a statistical heterogeneity among the studies for the detection rate of primary HNC lesions. The quantitative findings presented in this study indicate a favorable diagnostic performance of FAP-targeted PET imaging in detecting primary HNC tumors. In contrast, discordant results concerning the diagnostic accuracy of lymph node metastases were found. However, further multicentric trials are required to validate the efficacy of FAP-targeted PET in this specific group of patients.
Collapse
Affiliation(s)
- Alessio Rizzo
- Department of Nuclear Medicine, Candiolo Cancer Institute, FPO–IRCCS, 10060 Turin, Italy; (A.R.); (M.R.)
| | - Alberto Miceli
- Nuclear Medicine Unit, Azienda Ospedaliera SS. Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy;
| | - Manuela Racca
- Department of Nuclear Medicine, Candiolo Cancer Institute, FPO–IRCCS, 10060 Turin, Italy; (A.R.); (M.R.)
| | - Matteo Bauckneht
- Division of Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, 16131 Genova, Italy; (M.B.); (S.M.)
- Department of Health Sciences (DISSAL), University of Genova, 16131 Genova, Italy
| | - Silvia Morbelli
- Division of Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, 16131 Genova, Italy; (M.B.); (S.M.)
- Department of Health Sciences (DISSAL), University of Genova, 16131 Genova, Italy
| | - Domenico Albano
- Division of Nuclear Medicine, Università degli Studi di Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (D.A.); (F.D.); (F.B.)
| | - Francesco Dondi
- Division of Nuclear Medicine, Università degli Studi di Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (D.A.); (F.D.); (F.B.)
| | - Francesco Bertagna
- Division of Nuclear Medicine, Università degli Studi di Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (D.A.); (F.D.); (F.B.)
| | - Danilo Galizia
- SC Oncologia Area Nord ASL CN1, 12038 Savigliano, Italy;
| | - Barbara Muoio
- Division of Medical Oncology, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, 6501 Bellinzona, Switzerland;
| | - Salvatore Annunziata
- Unità di Medicina Nucleare, GSTeP Radiopharmacy–TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy;
| | - Giorgio Treglia
- Clinic of Nuclear Medicine, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, 6501 Bellinzona, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| |
Collapse
|
8
|
Ding P, Liu P, Meng L, Zhao Q. Mechanisms and biomarkers of immune-related adverse events in gastric cancer. Eur J Med Res 2023; 28:492. [PMID: 37936161 PMCID: PMC10631148 DOI: 10.1186/s40001-023-01365-3] [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: 01/10/2023] [Accepted: 09/12/2023] [Indexed: 11/09/2023] Open
Abstract
Immune-checkpoint inhibitors (ICIs), different from traditional cancer treatment models, have shown unprecedented anti-tumor effects in the past decade, greatly improving the prognosis of many malignant tumors in clinical practice. At present, the most widely used ICIs in clinical immunotherapy for a variety of solid tumors are monoclonal antibodies against cytotoxic T lymphocyte antigen-4 (CTLA-4), programmed cell death protein 1 (PD-1) and their ligand PD-L1. However, tumor patients may induce immune-related adverse events (irAEs) while performing immunotherapy, and irAE is an obstacle to the prospect of ICI treatment. IrAE is a non-specific disease caused by immune system imbalance, which can occur in many tissues and organs. For example, skin, gastrointestinal tract, endocrine system and lung. Although the exact mechanism is not completely clear, related studies have shown that irAE may develop through many ways. Such as excessive activation of autoreactive T cells, excessive release of inflammatory cytokines, elevated levels of autoantibodies, and common antigens between tumors and normal tissues. Considering that the occurrence of severe IrAE not only causes irreversible damage to the patient's body, but also terminates immunotherapy due to immune intolerance. Therefore, accurate identification and screening of sensitive markers of irAE are the main beneficiaries of ICI treatment. Additionally, irAEs usually require specific management, the most common of which are steroids and immunomodulatory therapies. This review aims to summarize the current biomarkers for predicting irAE in gastric cancer and their possible mechanisms.
Collapse
Affiliation(s)
- Ping'an Ding
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
| | - Pengpeng Liu
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
| | - Lingjiao Meng
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China.
- Research Center of the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China.
| | - Qun Zhao
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China.
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China.
| |
Collapse
|
9
|
Arçay Öztürk A, Flamen P. FAP-targeted PET imaging in gastrointestinal malignancies: a comprehensive review. Cancer Imaging 2023; 23:79. [PMID: 37608378 PMCID: PMC10463504 DOI: 10.1186/s40644-023-00598-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 08/07/2023] [Indexed: 08/24/2023] Open
Abstract
F18-fluorodeoxyglucose positron emission tomography/computed tomography (FDG PET/CT) plays a crucial role in tumour diagnosis, staging, and therapy response evaluation of various cancer types and has been a standard imaging modality used in clinical oncology practice for many years. However, it has certain limitations in evaluating some particular gastrointestinal cancer types due to low FDG-avidity or interphering physiological background activity. Fibroblast activation protein (FAP), a protein of the tumour microenvironment, is overexpressed in a wide range of cancers which makes it an attractive target for both tumour imaging and therapy. Recently, FAP-targeted radiopharmaceuticals are widely used in clinical research and achieved great results in tumour imaging. Considering the limitations of FDG PET/CT and the lack of physiological FAP-targeted tracer uptake in liver and intestinal loops, gastrointestinal cancers are among the most promising indications of FAP-targeted imaging. Herein, we present a comprehensive review of FAP-targeted imaging in gastrointestinal cancers in order to clarify the current and potential future role of this class of molecules in gastrointestinal oncology.
Collapse
Affiliation(s)
- Ayça Arçay Öztürk
- Department of Nuclear Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.
| | - Patrick Flamen
- Department of Nuclear Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
10
|
Rizzo A, Racca M, Garrou F, Fenocchio E, Pellegrino L, Albano D, Dondi F, Bertagna F, Annunziata S, Treglia G. Diagnostic Performance of Positron Emission Tomography with Fibroblast-Activating Protein Inhibitors in Gastric Cancer: A Systematic Review and Meta-Analysis. Int J Mol Sci 2023; 24:10136. [PMID: 37373285 DOI: 10.3390/ijms241210136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Various papers have introduced the use of positron emission tomography (PET) with [68Ga]Ga-radiolabeled fibroblast-activation protein inhibitor (FAPi) radiopharmaceuticals in different subtypes of gastric cancer (GC). Our aim was to assess the diagnostic performance of this novel molecular imaging technique in GC with a systematic review and meta-analysis. A straightforward literature search of papers concerning the diagnostic performance of FAP-targeted PET imaging was performed. Original articles evaluating this novel molecular imaging examination in both newly diagnosed GC patients and GC patients with disease relapse were included. The systematic review included nine original studies, and eight of them were also eligible for meta-analysis. The quantitative synthesis provided pooled detection rates of 95% and 97% for the assessment of primary tumor and distant metastases, respectively, and a pooled sensitivity and specificity of 74% and 89%, respectively, for regional lymph node metastases. Significant statistical heterogeneity among the included studies was found only in the analysis of the primary tumor detection rate (I2 = 64%). Conclusions: Beyond the limitations of this systematic review and meta-analysis (i.e., all the included studies were conducted in Asia, and using [18F]FDG PET/CT as a comparator of the index test), the quantitative data provided demonstrate the promising diagnostic performance of FAP-targeted PET imaging in GC. Nevertheless, more prospective multicentric studies are needed to confirm the excellent performances of FAP-targeted PET in this cluster of patients.
Collapse
Affiliation(s)
- Alessio Rizzo
- Department of Nuclear Medicine, Candiolo Cancer Institute, FPO-IRCCS, 10060 Turin, Italy
| | - Manuela Racca
- Department of Nuclear Medicine, Candiolo Cancer Institute, FPO-IRCCS, 10060 Turin, Italy
| | - Federico Garrou
- Nuclear Medicine Unit, Department of Medical Sciences, AOU Città della Salute e della Scienza, University of Turin, 10126 Turin, Italy
| | - Elisabetta Fenocchio
- Department of Oncology, Candiolo Cancer Institute, FPO-IRCCS, 10060 Turin, Italy
| | - Luca Pellegrino
- Department of Surgery, Candiolo Cancer Institute, FPO-IRCCS, 10060 Turin, Italy
| | - Domenico Albano
- Division of Nuclear Medicine, Università degli Studi di Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Francesco Dondi
- Division of Nuclear Medicine, Università degli Studi di Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Francesco Bertagna
- Division of Nuclear Medicine, Università degli Studi di Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Salvatore Annunziata
- Unità di Medicina Nucleare, GSTeP Radiopharmacy-TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Giorgio Treglia
- Clinic of Nuclear Medicine, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, 6501 Bellinzona, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| |
Collapse
|
11
|
Borgonje PE, Andrews LM, Herder GJM, de Klerk JMH. Performance and Prospects of [ 68Ga]Ga-FAPI PET/CT Scans in Lung Cancer. Cancers (Basel) 2022; 14:cancers14225566. [PMID: 36428657 PMCID: PMC9688494 DOI: 10.3390/cancers14225566] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/05/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Fibroblast activation protein (FAP) could be a promising target for tumor imaging and therapy, as it is expressed in >90% of epithelial cancers. A high level of FAP-expression might be associated with worse prognosis in several cancer types, including lung cancer. FAPI binds this protein and allows for labelling to Gallium-68, as well as several therapeutic radiopharmaceuticals. As FAP is only expressed at insignificant levels in adult normal tissue, FAPI provides a highly specific tumor-marker for many epithelial cancers. In this review, current information on the use of [68Ga]Ga-FAPI PET/CT in lung cancer is presented. [68Ga]Ga-FAPI shows a high uptake (standardized uptake value = SUVmax) and tumor-to-background ratio (TBR) in primary lung cancer lesions, as well as in metastatic lesions of other tumor types located in the lung and in lung cancer metastases located throughout the body. Where a comparison was made to [18F]FDG PET/CT, [68Ga]Ga-FAPI showed a similar or higher SUVmax and TBR. In brain and bone metastases, [68Ga]Ga-FAPI PET/CT outperformed [18F]FDG PET/CT. In addition to this strong diagnostic performance, a possible prognostic value of [68Ga]Ga-FAPI PET/CT in lung cancer is proposed.
Collapse
Affiliation(s)
- Paula E. Borgonje
- Department of Clinical Pharmacy, Meander Medical Center, Maatweg 3, 3813 TZ Amersfoort, The Netherlands
| | - Louise M. Andrews
- Department of Clinical Pharmacy, Meander Medical Center, Maatweg 3, 3813 TZ Amersfoort, The Netherlands
| | - Gerarda J. M. Herder
- Department of Pulmonology, Meander Medical Center, Maatweg 3, 3813 TZ Amersfoort, The Netherlands
| | - John M. H. de Klerk
- Department of Nuclear Medicine, Meander Medical Center, Maatweg 3, 3813 TZ Amersfoort, The Netherlands
- Correspondence: ; Tel.: +31-33-850-5050
| |
Collapse
|
12
|
Zhao Y, Liu Y, Jia Y, Wang X, He J, Zhen S, Wang J, Liu L. Fibroblast activation protein in the tumor microenvironment predicts outcomes of PD-1 blockade therapy in advanced non-small cell lung cancer. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04250-4. [PMID: 35951090 DOI: 10.1007/s00432-022-04250-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/02/2022] [Indexed: 10/15/2022]
Abstract
PURPOSE The identification of robust predictive biomarkers of the response to programmed cell death-1 (PD-1) blockade remains a critical concern. Here, we investigated on fibroblast activation protein (FAP) as a microenvironment-derived biomarker of clinical outcomes of PD-1 blockade therapy, and the correlation between FAP expression and T cell infiltration in advanced non-small cell lung cancer (NSCLC). METHODS A total of 135 patients with advanced NSCLC who received PD-1 blockade therapy were retrospectively analyzed. The potential associations among FAP expression, CD3 + T cell and CD8 + T cell infiltration, and clinical outcomes of immunotherapy were validated by immunohistochemistry, bioinformatic analyses, and statistical measurements. RESULTS FAP was widely expressed in advanced NSCLC tissues. FAP was correlated with decreased density of CD8 + T cells (Spearman's rho - 0.32, p < 0.001) and immunosuppressive tumor microenvironment (TME) status. No correlations were detected between FAP and PD-L1 expression or with the density of CD3 + T cells. The patients with higher expression of FAP showed worse response rate (16.4% vs. 38.7%, p < 0.001) and worse progression-free survival (HR = 2.56, 95% CI 1.69-3.87, p < 0.001). In addition, FAP contributed to shortened overall survival in subgroups of the patients with squamous cell lung cancer (p = 0.020), PD-1 blockade monotherapy (p = 0.017), and first-line therapy (p = 0.028). CONCLUSION FAP is a potential predictive biomarker of resistance to PD-1 blockade. Further investigation is warranted to identify a strategy for targeting FAP to alleviate the immunosuppressive TME and broaden the clinical effectiveness of PD-1 blockade therapy.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China.,Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Yueping Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Yunlong Jia
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China
| | - Xiaoxiao Wang
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Jiankun He
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Shuman Zhen
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China
| | - Jiali Wang
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China
| | - Lihua Liu
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China. .,Cancer Research Institute of Hebei Province, Shijiazhuang, 050011, China. .,China International Cooperation Laboratory of Stem Cell Research, Hebei Medical University, Shijiazhuang, 050011, China.
| |
Collapse
|