1
|
Gan M, Liu N, Li W, Chen M, Bai Z, Liu D, Liu S. Metabolic targeting of regulatory T cells in oral squamous cell carcinoma: new horizons in immunotherapy. Mol Cancer 2024; 23:273. [PMID: 39696340 DOI: 10.1186/s12943-024-02193-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a prevalent oral malignancy, which poses significant health risks with a high mortality rate. Regulatory T cells (Tregs), characterized by their immunosuppressive capabilities, are intricately linked to OSCC progression and patient outcomes. The metabolic reprogramming of Tregs within the OSCC tumor microenvironment (TME) underpins their function, with key pathways such as the tryptophan-kynurenine-aryl hydrocarbon receptor, PI3K-Akt-mTOR and nucleotide metabolism significantly contributing to their suppressive activities. Targeting these metabolic pathways offers a novel therapeutic approach to reduce Treg-mediated immunosuppression and enhance anti-tumor responses. This review explores the metabolic dependencies and pathways that sustain Treg function in OSCC, highlighting key metabolic adaptations such as glycolysis, fatty acid oxidation, amino acid metabolism and PI3K-Akt-mTOR signaling pathway that enable Tregs to thrive in the challenging conditions of the TME. Additionally, the review discusses the influence of the oral microbiome on Treg metabolism and evaluates potential therapeutic strategies targeting these metabolic pathways. Despite the promising potential of these interventions, challenges such as selectivity, toxicity, tumor heterogeneity, and resistance mechanisms remain. The review concludes with perspectives on personalized medicine and integrative approaches, emphasizing the need for continued research to translate these findings into effective clinical applications for OSCC treatment.
Collapse
Affiliation(s)
- Menglai Gan
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China
| | - Nanshu Liu
- Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China
| | - Wenting Li
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China
| | - Mingwei Chen
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China
| | - Zhongyu Bai
- Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China
| | - Dongjuan Liu
- Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China.
| | - Sai Liu
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China.
| |
Collapse
|
2
|
Ramalingam S, Shantha S, Srinivasan CP, Priyathersini N, Muralitharan S, Sudhakar U, Thamizhchelvan H, Parvathi VD. Expression of mTOR, CD163, α-SMA, FOXp3 as survival predictors and its significance in patients with oral squamous cell carcinoma. BMC Oral Health 2024; 24:1487. [PMID: 39695576 DOI: 10.1186/s12903-024-05245-y] [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: 04/01/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Oral cancer; categorised under head and neck cancers (HNC) predominantly originates from squamous cells and is referred as oral squamous cell carcinoma (OSCC). Various factors (internal and external) causes OSCC. PI3K/AKT/mTOR pathways are known to be primarily mutated in HNC. mTOR remains as a key regulator for various physiological and developmental processes in normal and cancer cells. Cancer cells are surrounded by tumor microenvironment, majorly composed of immune cells. Cancer associated fibroblasts, macrophages and regulatory T cells controlled by mTOR, plays an important role in the progression of cancer. METHODS Two hundred sixty retrospective patient samples were collected along with their demographical and clinic-pathological data. Here, we have analysed expression of mTOR, α-SMA, CD163 and FOXp3 using immunohistochemistry and their survival outcomes were calculated using Kaplan-Meier statistical method. RESULTS Overexpression of CD163 and α-SMA was detected in samples of patients compared to mTOR and FOXp3. Their expression was compared with clinico-oncological parameters. We also observed two and three combinations of markers and its association with the prognosis of the cancer. The results suggest, higher the expression of all the four markers in combination correlated to poor prognosis of patients and vice-versa. CONCLUSION The study reveals that over expression of CD163 and α-SMA is strongly associated with disease outcome. The combinations of all the four marker expression profile will be emerging strategy towards prognosis and also to determine survival outcomes in patients. This is a pioneering observation of these combinations of markers in OSCC despite certain limitations.
Collapse
MESH Headings
- Humans
- Forkhead Transcription Factors/metabolism
- TOR Serine-Threonine Kinases/metabolism
- Mouth Neoplasms/pathology
- Mouth Neoplasms/metabolism
- Mouth Neoplasms/mortality
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Differentiation, Myelomonocytic/analysis
- Receptors, Cell Surface/metabolism
- Antigens, CD/metabolism
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/mortality
- Male
- Actins/metabolism
- Female
- Middle Aged
- Retrospective Studies
- Prognosis
- Aged
- Biomarkers, Tumor/metabolism
- Adult
- Survival Rate
Collapse
Affiliation(s)
- Suganya Ramalingam
- Department of Oral Pathology, Sri Ramachandra Dental College and Hospital, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, 600116, India
| | - Sivaramakrishnan Shantha
- Department of Oral Pathology, Sri Ramachandra Dental College and Hospital, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, 600116, India
| | - Chakravarthy Purushothaman Srinivasan
- Department of Oral Pathology, Sri Ramachandra Dental College and Hospital, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, 600116, India
| | - Nagarajan Priyathersini
- Department of Pathology, Sri Ramachandra Medical College and Research Institute, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, 600116, India
| | - Susruthan Muralitharan
- Department of Pathology, Indira Medical College and Hospitals, Pandur, 602001, Tamil Nadu, India
| | - Uma Sudhakar
- Department of Periodontics, Department of Dental Sciences, Tamil Nadu Dr. M.G.R. Medical University, Guindy, Chennai, 600032, India
| | - Harikrishnan Thamizhchelvan
- Department of Oral Pathology, Sri Ramachandra Dental College and Hospital, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, 600116, India.
| | - Venkatachalam Deepa Parvathi
- Department of Biomedical Sciences, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education & Research, Porur, Chennai, 600116, India.
| |
Collapse
|
3
|
Agarwal N, Jha AK. DNA hypermethylation of tumor suppressor genes among oral squamous cell carcinoma patients: a prominent diagnostic biomarker. Mol Biol Rep 2024; 52:44. [PMID: 39644423 DOI: 10.1007/s11033-024-10144-0] [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: 06/02/2024] [Accepted: 11/29/2024] [Indexed: 12/09/2024]
Abstract
Oral Squamous Cell Carcinoma is a globally revealing form of oral malignancy. Epigenetics, which studies genetic modifications in gene expression without altering the sequence of DNA, is crucial for understanding OSCC. Key epigenetic modifications such as histone modifications, DNA methylation, and microRNA regulation play significant roles in Oral carcinoma. Aberrant methylation of DNA of tumor suppressor genes which leads to their inactivation, promoting cancer development, and specific methylation patterns are emerging as biomarkers for early OSCC detection.Current treatments like surgery, radiotherapy, and chemotherapy often fall short, prompting research into epigenetic therapies. Agents like DNMT and HDAC inhibitors demonstrate the potential for reversing aberrant epigenetic patterns, perhaps reactivating silenced TSGs, and suppressing oncogenes. Despite early promise, the development of effective combination medicines and the identification of reliable biomarkers continue to present challenges.In OSCC, resistance to therapy is also influenced by epigenetic processes. Aberrant DNA methylation and changes in histone modifications impact genes involved in medication metabolism and the survival of cells. Enhancing treatment efficacy and overcoming medication resistance may be possible by recognizing and focusing on these processes. This review explores the interplay between epigenetic changes and OSCC, their role in the disease's initiation and progression, and their impact on diagnosis and treatment. It also discusses the potential of epigenetic drugs (epi-drugs) to improve diagnostic precision and treatment outcomes.
Collapse
Affiliation(s)
- Nistha Agarwal
- Department of Biotechnology, School of Biosciences and Technology, Galgotias University, Greater Noida, India
| | - Abhimanyu Kumar Jha
- Department of Biotechnology, School of Biosciences and Technology, Galgotias University, Greater Noida, India.
| |
Collapse
|
4
|
Essa AAM, Korayem MAM, Alghamdi MA, Alzahrani RA, Malibari NM, Kandil HE. Expression of CD68 and CD163 in malignant epithelial cells of oral squamous cell carcinoma: Phenotypic shift or mere cell fusion. Pathol Res Pract 2024; 263:155639. [PMID: 39383735 DOI: 10.1016/j.prp.2024.155639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 09/15/2024] [Accepted: 10/02/2024] [Indexed: 10/11/2024]
Abstract
BACKGROUND / PURPOSE The progression of epithelial to mesenchymal tissue (EMT) is a highly intricate process that facilitates the transformation of cancer cells, allowing them to changeover their characteristic epithelial properties to mesenchymal attributes. This notable change empowers the cells with enhanced mobility and the ability to migrate to distant locations. Furthermore, it is imperative to adopt the idea of macrophage tumor cell fusion to achieve comprehensive considerate of this phenomenon. Our primary objective was to conduct a thorough investigation of macrophage-restricted antigens expression, specifically CD68 and CD163, in malignant epithelial cells of oral cavity squamous cell carcinoma (OSCC) to elucidate aforementioned perceptions. MATERIALS AND METHODS CD68 and CD163 immunohistochemical expression were assessed in oral squamous cell carcinoma (OSCC), encompassing both the neoplastic cells and the tumor-associated macrophages (TAMs). RESULTS Both CD68 and CD163 antigens were revealed in OSCC malignant epithelial cells in a granular cell pattern, localized in membrane and cytoplasm of tumor cells respectively as well as in the infiltrating TAMs. CONCLUSION The macrophage antigens were not limited to the infiltrating tumor-associated macrophages (TAMs), but were also observed in a substantial proportion of OSCC malignant epithelial cells within the tumor parenchyma. This particular expression pattern may represent a subset of tumor cells that have undergone an epithelial to a mesenchymal phenotypic transition. In addition, fusion of macrophages with tumor cells cannot be excluded; both might be associated with increased metastatic activity of OSCC.
Collapse
Affiliation(s)
- Ahmed Abdelaziz Mohamed Essa
- Oral Pathology Department, Faculty of Dentistry, Tanta University, El-Gesh Street, Tanta, 31527, Egypt; Department of Biomedical Dental Sciences, Faculty of Dentistry, Al-Baha University, Al-Baha, 65511, Saudi Arabia.
| | | | - Mohammed A Alghamdi
- Otolaryngology Division, Surgery Department, Faculty of Medicine, Al-Baha University, Al-Baha, Saudi Arabia.
| | - Rajab A Alzahrani
- Otolaryngology Division, Surgery Department, Faculty of Medicine, Al-Baha University, Al-Baha, Saudi Arabia.
| | | | | |
Collapse
|
5
|
Meng Z, Li T, Li J, Ding S, Liu Y, Zhao G, Chen C, Zhao P, Zhou L. LncRNAPVT1 is Associated with Cancer-Associated Fibroblasts Proliferation Through Regulating TGF-βin Oral Squamous Cell Carcinoma. Immunol Invest 2024; 53:1250-1263. [PMID: 39189542 DOI: 10.1080/08820139.2024.2395874] [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: 08/28/2024]
Abstract
INTRODUCTION Human oral squamous cell carcinoma (OSCC) is the most common type of oral cancer and has a poor survival rate. Cell-cell communication between OSCC cells and cancer-associated fibroblasts (CAFs) plays important roles in OSCC progression. We previously demonstrated that CAFs promote OSCC cell migration and invasion. However, how OSCC cells influence CAFs proliferation is unknown. METHODS Knockdown of PVT1 was confirmed using lentivirus infection technique. CAFs in tissues were identified by staining the cells with α-SMA using immunohistochemical technique. CCK-8 assay was used to evaluate cell proliferation. The mRNA level of a gene was measured by qRT-PCR. Secreted TGF-β were detected using ELISA assay. RESULTS We found that knockdown of the long non-coding RNA (lncRNA) plasmacytoma variant translocation 1 (PVT1) was associated with a low density of CAFs in xenograft tumors in mice; further analysis revealed that PVT1 in OSCC cells induced CAF proliferation through transforming growth factor (TGF)-β. DISCUSSION Our results demonstrate that lncRNA PVT1 in tumor cells participates in CAF development in OSCC by regulating TGF-β. This study revealed a new mechanism by which PVT1 regulates OSCC progression and PVT1 is a potential therapeutic target in OSCC.
Collapse
Affiliation(s)
- Zhen Meng
- Biomedical Laboratory, Medical School of Liaocheng University, Liaocheng, Shandong Province, P.R. China
| | - Tongjuan Li
- Department of Stomatology, Anqiu Municipal Hospital, Weifang, Shandong Province, P.R. China
| | - Jun Li
- Precision Biomedical Laboratory of Liaocheng, Liaocheng People's Hospital, Medical School of Liaocheng University, Liaocheng, Shandong Province, P.R. China
| | - Shuxin Ding
- Department of Oral and Maxillofacial Surgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, P.R. China
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Weifang Medicial University, Weifang, Shandong Province, P.R. China
| | - Yujiao Liu
- Department of Oral and Maxillofacial Surgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, P.R. China
| | - Guoli Zhao
- Department of Pathology, Liaocheng Tumor Hospital, Liaocheng, Shandong Province, P.R. China
| | - Cheng Chen
- Department of Oral and Maxillofacial Surgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, P.R. China
| | - Peng Zhao
- Department of Oral and Maxillofacial Surgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, P.R. China
| | - Longxun Zhou
- Department of Oral and Maxillofacial Surgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, P.R. China
| |
Collapse
|
6
|
Huang X, Tian B, Ren Z, Zhang J, Yan W, Mo Y, Yuan J, Ma Y, Wang R, Liu R, Chen M, Yu J, Chen D. CD34 as a potential prognostic indicator for camrelizumab response in advanced non-small-cell lung cancer: insights from digital spatial profiling. Ther Adv Med Oncol 2024; 16:17588359241289671. [PMID: 39429466 PMCID: PMC11489950 DOI: 10.1177/17588359241289671] [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] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 09/19/2024] [Indexed: 10/22/2024] Open
Abstract
Background Given that only a small subset of patients with advanced non-small-cell lung cancer (aNSCLC) benefit from immune checkpoint inhibitors (ICIs), the effectiveness of ICIs is often compromised by the complex interplay within the tumor microenvironment (TME). Objectives To identify predictive biomarkers associated with ICI resistance at a multi-omics spatial level. Design A total of eight aNSCLC patients who received first-line anti-programmed cell death protein-1 (PD-1) monoclonal antibody camrelizumab at Shandong Cancer Hospital and Institute between 2021 and 2022 were included in the discovery cohort. An additional validation cohort of 45 samples from camrelizumab-treated aNSCLC patients was also enrolled. Methods NanoString GeoMx® digital spatial profiling was conducted at the transcriptomic and proteomic level within pan-cytokeratin (panCK+), CD45+, and CD68+ compartments. For validation, multiplex immunofluorescence (mIF) staining was performed. Results Distinct spatial expression patterns and levels of immune infiltration were observed between tumor and leukocyte compartments. Higher CD34 expression in the macrophage compartment correlated with poorer prognosis and response to camrelizumab (p < 0.05). mIF validation confirmed the association of elevated CD34 expression level with reduced progression-free survival (PFS; hazard ratio (HR) = 5.011, 95% confidence interval: 1.057-23.752, p = 0.042), outperforming traditional tumor markers in predictive accuracy. Conclusion Our findings identify CD34 as a novel spatial biomarker for anti-PD-1 therapy efficacy, potentially guiding the selection of aNSCLC patients who are more likely to benefit from ICI treatment. Trial registration ChiCTR2000040416.
Collapse
Affiliation(s)
- Xinyi Huang
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Baoqing Tian
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ziyuan Ren
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Cheeloo College of Medicine, Shandong University Cancer Center, Jinan, Shandong, China
| | - Jingxin Zhang
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Cheeloo College of Medicine, Shandong University Cancer Center, Jinan, Shandong, China
| | - Weiwei Yan
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Cheeloo College of Medicine, Shandong University Cancer Center, Jinan, Shandong, China
| | - You Mo
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jupeng Yuan
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yujiao Ma
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Cheeloo College of Medicine, Shandong University Cancer Center, Jinan, Shandong, China
| | - Ruiyang Wang
- Department of Oncology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Rufei Liu
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Minxin Chen
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jinming Yu
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250000, Shandong, China
| | - Dawei Chen
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250000, Shandong, China
| |
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: 4] [Impact Index Per Article: 4.0] [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
|