1
|
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 2024:00004424-990000000-00234. [PMID: 39018248 DOI: 10.1097/rli.0000000000001096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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 18F-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
|
2
|
Amrane K, Meur CL, Thuillier P, Berthou C, Uguen A, Deandreis D, Bourhis D, Bourbonne V, Abgral R. Review on radiomic analysis in 18F-fluorodeoxyglucose positron emission tomography for prediction of melanoma outcomes. Cancer Imaging 2024; 24:87. [PMID: 38970050 PMCID: PMC11225300 DOI: 10.1186/s40644-024-00732-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 06/24/2024] [Indexed: 07/07/2024] Open
Abstract
Over the past decade, several strategies have revolutionized the clinical management of patients with cutaneous melanoma (CM), including immunotherapy and targeted tyrosine kinase inhibitor (TKI)-based therapies. Indeed, immune checkpoint inhibitors (ICIs), alone or in combination, represent the standard of care for patients with advanced disease without an actionable mutation. Notably BRAF combined with MEK inhibitors represent the therapeutic standard for disease disclosing BRAF mutation. At the same time, FDG PET/CT has become part of the routine staging and evaluation of patients with cutaneous melanoma. There is growing interest in using FDG PET/CT measurements to predict response to ICI therapy and/or target therapy. While semiquantitative values such as standardized uptake value (SUV) are limited for predicting outcome, new measures including tumor metabolic volume, total lesion glycolysis and radiomics seem promising as potential imaging biomarkers for nuclear medicine. The aim of this review, prepared by an interdisciplinary group of experts, is to take stock of the current literature on radiomics approaches that could improve outcomes in CM.
Collapse
Affiliation(s)
- Karim Amrane
- Department of Oncology, Regional Hospital of Morlaix, Morlaix, 29600, France.
- Lymphocytes B et Autoimmunité, Inserm, UMR1227, Univ Brest, Inserm, LabEx IGO, Brest, France.
| | - Coline Le Meur
- Department of Radiotherapy, University Hospital of Brest, Brest, France
| | - Philippe Thuillier
- Department of Endocrinology, University Hospital of Brest, Brest, France
- UMR Inserm 1304 GETBO, University of Western Brittany, Brest, IFR 148, France
| | - Christian Berthou
- Lymphocytes B et Autoimmunité, Inserm, UMR1227, Univ Brest, Inserm, LabEx IGO, Brest, France
- Department of Hematology, University Hospital of Brest, Brest, France
| | - Arnaud Uguen
- Lymphocytes B et Autoimmunité, Inserm, UMR1227, Univ Brest, Inserm, LabEx IGO, Brest, France
- Department of Pathology, University Hospital of Brest, Brest, France
| | - Désirée Deandreis
- Department of Nuclear Medicine, Gustave Roussy Institute, University of Paris Saclay, Paris, France
| | - David Bourhis
- UMR Inserm 1304 GETBO, University of Western Brittany, Brest, IFR 148, France
- Department of Nuclear Medicine, University Hospital of Brest, Brest, France
| | - Vincent Bourbonne
- Department of Radiotherapy, University Hospital of Brest, Brest, France
- Inserm, UMR1101, LaTIM, University of Western Brittany, Brest, France
| | - Ronan Abgral
- UMR Inserm 1304 GETBO, University of Western Brittany, Brest, IFR 148, France
- Department of Nuclear Medicine, University Hospital of Brest, Brest, France
| |
Collapse
|
3
|
Daste A, Larroquette M, Gibson N, Lasserre M, Domblides C. Immunotherapy for head and neck squamous cell carcinoma: current status and perspectives. Immunotherapy 2024; 16:187-197. [PMID: 38126161 DOI: 10.2217/imt-2023-0174] [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: 12/23/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of several solid cancers, including head and neck squamous cell carcinoma (HNSCC). First approved for second-line settings, ICIs are now used for the first-line treatment of HNSCCs, mainly in combination with standard chemotherapy. This review focuses on the results of the main phase III studies evaluating ICIs in recurrent or metastatic HNSCCs. The efficacy and indications according to the PD-L1 status, the main predictive biomarker, are discussed. The results of trials assessing ICI efficacy for locally advanced disease, including the neoadjuvant setting are also discussed. Finally, therapeutic combinations that are potential treatments for HNSCCs, including ICIs and targeted therapies such as anti-EGFR agents, are presented.
Collapse
Affiliation(s)
- Amaury Daste
- Department of Medical Oncology, Hôpital Saint-André, CHU, Bordeaux, France
| | - Mathieu Larroquette
- Department of Medical Oncology, Hôpital Saint-André, CHU, Bordeaux, France
- IBGC, CNRS, UMR5095, University of Bordeaux, CNRS, IBGC, UMR 5095, Bordeaux, France
- Bordeaux University, 351 cours de la Libération CS10004 33405 Talence CEDEX, Bordeaux, France
| | - Nyere Gibson
- Department of Medical Oncology, Hôpital Saint-André, CHU, Bordeaux, France
- Bordeaux University, 351 cours de la Libération CS10004 33405 Talence CEDEX, Bordeaux, France
| | - Matthieu Lasserre
- Department of Medical Oncology, Hôpital Saint-André, CHU, Bordeaux, France
- Bordeaux University, 351 cours de la Libération CS10004 33405 Talence CEDEX, Bordeaux, France
| | - Charlotte Domblides
- Department of Medical Oncology, Hôpital Saint-André, CHU, Bordeaux, France
- Bordeaux University, 351 cours de la Libération CS10004 33405 Talence CEDEX, Bordeaux, France
- ImmunoConcEpt, CNRS UMR 5164, Bordeaux University, Bordeaux, 33076, France
| |
Collapse
|
4
|
Peisen F, Gerken A, Dahm I, Nikolaou K, Eigentler T, Amaral T, Moltz JH, Othman AE, Gatidis S. Pre-treatment 18F-FDG-PET/CT parameters as biomarkers for progression free survival, best overall response and overall survival in metastatic melanoma patients undergoing first-line immunotherapy. PLoS One 2024; 19:e0296253. [PMID: 38180971 PMCID: PMC10769042 DOI: 10.1371/journal.pone.0296253] [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: 10/04/2023] [Accepted: 12/08/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND Checkpoint inhibitors have drastically improved the therapy of patients with advanced melanoma. 18F-FDG-PET/CT parameters might act as biomarkers for response and survival and thus can identify patients that do not benefit from immunotherapy. However, little literature exists on the association of baseline 18F-FDG-PET/CT parameters with progression free survival (PFS), best overall response (BOR), and overall survival (OS). MATERIALS AND METHODS Using a whole tumor volume segmentation approach, we investigated in a retrospective registry study (n = 50) whether pre-treatment 18F-FDG-PET/CT parameters of three subgroups (tumor burden, tumor glucose uptake and non-tumoral hematopoietic tissue metabolism), can act as biomarkers for the primary endpoints PFS and BOR as well as for the secondary endpoint OS. RESULTS Compared to the sole use of clinical parameters, baseline 18F-FDG-PET/CT parameters did not significantly improve a Cox proportional-hazard model for PFS (C-index/AIC: 0.70/225.17 and 0.68/223.54, respectively; p = 0.14). A binomial logistic regression analysis for BOR was not statistically significant (χ2(15) = 16.44, p = 0.35), with a low amount of explained variance (Nagelkerke's R2 = 0.38). Mean FDG uptake of the spleen contributed significantly to a Cox proportional-hazard model for OS (HR 3.55, p = 0.04). CONCLUSIONS The present study could not confirm the capability of the pre-treatment 18F-FDG-PET/CT parameters tumor burden, tumor glucose uptake and non-tumoral hematopoietic tissue metabolism to act as biomarkers for PFS and BOR in metastatic melanoma patients receiving first-line immunotherapy. The documented potential of 18F-FDG uptake by immune-mediating tissues such as the spleen to act as a biomarker for OS has been reproduced.
Collapse
Affiliation(s)
- Felix Peisen
- Department of Diagnostic and Interventional Radiology, Eberhard Karls University, Tuebingen University Hospital, Tuebingen, Germany
| | | | - Isabel Dahm
- Department of Diagnostic and Interventional Radiology, Eberhard Karls University, Tuebingen University Hospital, Tuebingen, Germany
| | - Konstantin Nikolaou
- Department of Diagnostic and Interventional Radiology, Eberhard Karls University, Tuebingen University Hospital, Tuebingen, Germany
- Image-guided and Functionally Instructed Tumor Therapies (iFIT), The Cluster of Excellence (EXC 2180), Tuebingen, Germany
| | - Thomas Eigentler
- Center of Dermato-Oncology, Department of Dermatology, Eberhard Karls University, Tuebingen University Hospital, Tuebingen, Germany
- Department of Dermatology, Venereology and Allergology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humbolt-Universität zu Berlin, Berlin, Germany
| | - Teresa Amaral
- Center of Dermato-Oncology, Department of Dermatology, Eberhard Karls University, Tuebingen University Hospital, Tuebingen, Germany
| | | | - Ahmed E. Othman
- Department of Diagnostic and Interventional Radiology, Eberhard Karls University, Tuebingen University Hospital, Tuebingen, Germany
- Institute of Neuroradiology, Johannes Gutenberg University Hospital Mainz, Mainz, Germany
| | - Sergios Gatidis
- Department of Diagnostic and Interventional Radiology, Eberhard Karls University, Tuebingen University Hospital, Tuebingen, Germany
- Max Planck Institute for Intelligent Systems, Tuebingen, Germany
| |
Collapse
|
5
|
Yao S, Han Y, Yang M, Jin K, Lan H. Integration of liquid biopsy and immunotherapy: opening a new era in colorectal cancer treatment. Front Immunol 2023; 14:1292861. [PMID: 38077354 PMCID: PMC10702507 DOI: 10.3389/fimmu.2023.1292861] [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: 09/12/2023] [Accepted: 11/03/2023] [Indexed: 12/18/2023] Open
Abstract
Immunotherapy has revolutionized the conventional treatment approaches for colorectal cancer (CRC), offering new therapeutic prospects for patients. Liquid biopsy has shown significant potential in early screening, diagnosis, and postoperative monitoring by analyzing circulating tumor cells (CTC) and circulating tumor DNA (ctDNA). In the era of immunotherapy, liquid biopsy provides additional possibilities for guiding immune-based treatments. Emerging technologies such as mass spectrometry-based detection of neoantigens and flow cytometry-based T cell sorting offer new tools for liquid biopsy, aiming to optimize immune therapy strategies. The integration of liquid biopsy with immunotherapy holds promise for improving treatment outcomes in colorectal cancer patients, enabling breakthroughs in early diagnosis and treatment, and providing patients with more personalized, precise, and effective treatment strategies.
Collapse
Affiliation(s)
- Shiya Yao
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Yuejun Han
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Mengxiang Yang
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Ketao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Huanrong Lan
- Department of Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
6
|
Mayer KE, Gaa J, Wasserer S, Biedermann T, Persa OD. Whole-Body Imaging for the Primary Staging of Melanomas-A Single-Center Retrospective Study. Cancers (Basel) 2023; 15:5265. [PMID: 37958438 PMCID: PMC10648596 DOI: 10.3390/cancers15215265] [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: 09/28/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Melanoma staging at diagnosis predominantly depends on the tumor thickness. Sentinel lymph node biopsy (SLNB) is a common tool for primary staging. However, for tumors of >4 mm with ulceration, 3D whole-body imaging and, in particular, Fluor-18-Deoxyglucose positron emission tomography combined with computed tomography (18F-FDG-PET/CT), is recommended beforehand. This study aimed to investigate the real-world data of whole-body imaging for initial melanoma staging and its impact on the subsequent diagnostic and therapeutic procedures. METHODS In this retrospective single-center study, 94 patients receiving 18F-FDG-PET/CT and six patients with whole-body computed tomography (CT) scans were included. The clinical characteristics, imaging results, and histologic parameters of the primary tumors and metastases were analyzed. RESULTS Besides the patients with primary tumors characterized as pT4b (63%), the patients with pT4a tumors and pT3 tumors close to 4 mm in tumor thickness also received initial whole-body imaging. In 42.6% of the patients undergoing 18F-FDG-PET/CT, the imaging results led to a change in the diagnostic or therapeutic procedure following on from this. In 29% of cases, sentinel lymph node biopsy was no longer necessary. The sensitivity and specificity of 18F-FDG-PET/CT were 66.0% and 93.0%, respectively. CONCLUSION Whole-body imaging as a primary diagnostic tool is highly valuable and influences the subsequent diagnostic and therapeutic procedures in a considerable number of patients with a relatively high tumor thickness. It can help avoid the costs and invasiveness of redundant SLNB and simultaneously hasten the staging of patients at the time of diagnosis.
Collapse
Affiliation(s)
- Kristine E. Mayer
- Clinic and Policlinic for Dermatology and Allergology, Technical University Munich, 80802 Munich, Germany
| | - Jochen Gaa
- Institute for Diagnostic and Interventional Radiology, Technical University Munich, 81675 Munich, Germany
| | - Sophia Wasserer
- Clinic and Policlinic for Dermatology and Allergology, Technical University Munich, 80802 Munich, Germany
| | - Tilo Biedermann
- Clinic and Policlinic for Dermatology and Allergology, Technical University Munich, 80802 Munich, Germany
| | - Oana-Diana Persa
- Clinic and Policlinic for Dermatology and Allergology, Technical University Munich, 80802 Munich, Germany
| |
Collapse
|
7
|
Yu J, Lan L, Liu C, Zhu X. Improved prediction of prognosis and therapy response for lung adenocarcinoma after identification of DNA-directed RNA polymerase-associated lncRNAs. J Cancer Res Clin Oncol 2023; 149:12737-12754. [PMID: 37453971 DOI: 10.1007/s00432-023-05118-x] [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: 05/24/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND DNA-directed RNA polymerase (DDRP) related genes and long non-coding RNAs (lncRNAs) play an important role in the development of lung adenocarcinoma (LUAD), the leading cause of cancer-related death worldwide. Therefore, we aimed to construct a DDRP-associated lncRNA model to predict the prognosis of LUAD and to evaluate its sensitivity to immunotherapy and chemotherapy. METHODS To construct a predictive signature, we used univariate and multivariate Cox regression analyses, as well as the least absolute shrinkage and selection operator regression analysis. The prognostic model was verified by applying the ROC curve analysis, Kaplan-Meier analysis, GO/KEGG analysis, and a predictive nomogram. Eventually, immunotherapy and drug susceptibility were examined and stemness indices were analyzed. RESULTS 24 DDRP-associated lncRNAs were found as independent prognosis factors, which may be further developed as potential therapeutic vaccines for LUAD. The area under the ROC curve and the conformance index showed that the constructed model can predict the prognosis of LUAD patients. The predicted incidences of overall survival showed perfect conformance. And there were significant changes in immunological markers between the two risk subgroups in the model. Finally, an analysis of 50% maximum inhibitory concentration between the two risk subgroups showed that the high-risk subgroup was more sensitive to certain chemotherapy drugs. CONCLUSION We constructed a model that accurately predicts the outcomes of LUAD based on 24 DDRP-related lncRNAs and provided promising treatment options for the future.
Collapse
Affiliation(s)
- Jiaao Yu
- Clinical Laboratory, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
- Computational Systems Biology Lab (CSBL), Institute of Bioinformatics, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Liqiang Lan
- Department of Internal Medicine, Qingdao Sixth People's Hospital, Qingdao, China
| | - Caixin Liu
- Clinical Laboratory, The First Affiliated Hospital of Wannan Medical College, Wuhu, China.
| | - Xiao Zhu
- Computational Systems Biology Lab (CSBL), Institute of Bioinformatics, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.
- Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou Medical College, Hangzhou, China.
| |
Collapse
|
8
|
Sachpekidis C, Stein-Thoeringer CK, Kopp-Schneider A, Weru V, Dimitrakopoulou-Strauss A, Hassel JC. Can physiologic colonic [ 18F]FDG uptake in PET/CT imaging predict response to immunotherapy in metastatic melanoma? Eur J Nucl Med Mol Imaging 2023; 50:3709-3722. [PMID: 37452874 PMCID: PMC10547632 DOI: 10.1007/s00259-023-06327-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 06/27/2023] [Indexed: 07/18/2023]
Abstract
AIM The development of biomarkers that can reliably and early predict response to immune checkpoint inhibitors (ICIs) is crucial in melanoma. In recent years, the gut microbiome has emerged as an important regulator of immunotherapy response, which may, moreover, serve as a surrogate marker and prognosticator in oncological patients under immunotherapy. Aim of the present study is to investigate if physiologic colonic [18F]FDG uptake in PET/CT before start of ICIs correlates with clinical outcome of metastatic melanoma patients. The relation between [18F]FDG uptake in lymphoid cell-rich organs and long-term patient outcome is also assessed. METHODOLOGY One hundred nineteen stage IV melanoma patients scheduled for immunotherapy with ipilimumab, applied either as monotherapy or in combination with nivolumab, underwent baseline [18F]FDG PET/CT. PET/CT data analysis consisted of standardized uptake value (SUV), metabolic tumor volume (MTV), and total lesion glycolysis (TLG) calculations in the colon as well as measurements of the colon-to-liver SUV ratios (CLRmean, CLRmax). Visual grading of colon uptake based on a four-point scale was also performed. Moreover, the spleen-to-liver SUV ratios (SLRmean, SLRmax) and the bone marrow-to-liver SUV ratios (BLRmean, BLRmax) were calculated. We also measured serum lipopolysaccharide (LPS) levels as a marker for bacterial translocation and surrogate for mucosal defense homeostasis. The results were correlated with patients' best clinical response, progression-free survival (PFS), and overall survival (OS) as well as clinical signs of colitis. RESULTS Median follow-up [95%CI] from the beginning of immunotherapy was 64.6 months [61.0-68.6 months]. Best response to treatment was progressive disease (PD) for 60 patients, stable disease (SD) for 37 patients, partial response (PR) for 18 patients, and complete response (CR) for 4 patients. Kaplan-Meier curves demonstrated a trend for longer PFS and OS in patients with lower colonic SUV and CLR values; however, no statistical significance for these parameters as prognostic factors was demonstrated. On the other hand, patients showing disease control as best response to treatment (SD, PR, CR) had significantly lower colonic MTV and TLG than those showing PD. With regard to lymphoid cell-rich organs, significantly lower baseline SLRmax and BLRmax were observed in patients responding with disease control than progression to treatment. Furthermore, patients with lower SLRmax and BLRmax values had a significantly longer OS when dichotomized at their median. In multivariate analysis, PET parameters that were found to significantly adversely correlate with patient survival were colonic MTV for PFS, colonic TLG for PFS, and BLRmax for PFS and OS. CONCLUSIONS Physiologic colonic [18F]FDG uptake in PET/CT, as assessed by means of SUV, before start of ipilimumab-based treatment does not seem to independently predict patient survival of metastatic melanoma. On the other hand, volumetric PET parameters, such as MTV and TLG, derived from the normal gut may identify patients showing disease control to immunotherapy and significantly correlate with PFS. Moreover, the investigation of glucose metabolism in the spleen and the bone marrow may offer prognostic information.
Collapse
Affiliation(s)
- Christos Sachpekidis
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69210, Heidelberg, Germany.
| | - Christoph K Stein-Thoeringer
- Laboratory of Translational, Microbiome Science, Internal Medicine I, University Clinic Tuebingen, Tuebingen, Germany
| | | | - Vivienn Weru
- Department of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Antonia Dimitrakopoulou-Strauss
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69210, Heidelberg, Germany
| | - Jessica C Hassel
- Department of Dermatology and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
9
|
Sachpekidis C, Weru V, Kopp-Schneider A, Hassel JC, Dimitrakopoulou-Strauss A. The prognostic value of [ 18F]FDG PET/CT based response monitoring in metastatic melanoma patients undergoing immunotherapy: comparison of different metabolic criteria. Eur J Nucl Med Mol Imaging 2023; 50:2699-2714. [PMID: 37099131 PMCID: PMC10317882 DOI: 10.1007/s00259-023-06243-y] [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: 03/22/2023] [Accepted: 04/19/2023] [Indexed: 04/27/2023]
Abstract
PURPOSE To investigate the prognostic value of [18F]FDG PET/CT as part of response monitoring in metastatic melanoma patients treated with immune checkpoint inhibitors (ICIs). METHODS Sixty-seven patients underwent [18F]FDG PET/CT before start of treatment (baseline PET/CT), after two cycles (interim PET/CT) and after four cycles of ICIs administration (late PET/CT). Metabolic response evaluation was based on the conventional EORTC and PERCIST criteria, as well as the newly introduced, immunotherapy-modified PERCIMT, imPERCIST5 and iPERCIST criteria. Metabolic response to immunotherapy was classified according to four response groups (complete metabolic response [CMR], partial metabolic response [PMR], stable metabolic disease [SMD], progressive metabolic disease [PMD]), and further dichotomized by response rate (responders = [CMR] + [PMR] vs. non-responders = [PMD] + [SMD]), and disease control rate (disease control = [CMR] + [PMR] + [SMD] vs. [PMD]). The spleen-to-liver SUV ratios (SLRmean, SLRmax) and bone marrow-to-liver SUV ratios (BLRmean, BLRmax) were also calculated. The results of PET/CT were correlated with patients' overall survival (OS). RESULTS Median patient follow up [95% CI] was 61.5 months [45.3 - 66.7 months]. On interim PET/CT, the application of the novel PERCIMT demonstrated significantly longer survival for metabolic responders, while the rest criteria revealed no significant survival differences between the different response groups. Respectively on late PET/CT, both a trend for longer OS and significantly longer OS were observed in patients responding to ICIs with metabolic response and disease control after application of various criteria, both conventional and immunotherapy-modified. Moreover, patients with lower SLRmean values demonstrated significantly longer OS. CONCLUSION In patients with metastatic melanoma PET/CT-based response assessment after four ICIs cycles is significantly associated with OS after application of different metabolic criteria. The prognostic performance of the modality is also high after the first two ICIs cycles, especially with employment of novel criteria. In addition, investigation of spleen glucose metabolism may provide further prognostic information.
Collapse
Affiliation(s)
- Christos Sachpekidis
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69210 Heidelberg, Heidelberg, Germany.
| | - Vivienn Weru
- Department of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Jessica C Hassel
- Department of Dermatology and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Antonia Dimitrakopoulou-Strauss
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69210 Heidelberg, Heidelberg, Germany
| |
Collapse
|
10
|
Bianchi A, De Rimini ML, Sciuto R, Annovazzi A, Di Traglia S, Bauckneht M, Lanfranchi F, Morbelli S, Nappi AG, Ferrari C, Rubini G, Panareo S, Urso L, Bartolomei M, D'Arienzo D, Valente T, Rossetti V, Caroli P, Matteucci F, Aricò D, Bombaci M, Caponnetto D, Bertagna F, Albano D, Dondi F, Gusella S, Spimpolo A, Carriere C, Balma M, Buschiazzo A, Gallicchio R, Storto G, Ruffini L, Scarlattei M, Baldari G, Cervino AR, Cuppari L, Burei M, Trifirò G, Brugola E, Zanini CA, Alessi A, Fuoco V, Seregni E, Deandreis D, Liberini V, Moreci AM, Ialuna S, Pulizzi S, Evangelista L. Can Baseline [18F]FDG PET/CT Predict Response to Immunotherapy After 6 Months and Overall Survival in Patients with Lung Cancer or Malignant Melanoma? A Multicenter Retrospective Study. Cancer Biother Radiopharm 2023; 38:256-267. [PMID: 37098169 DOI: 10.1089/cbr.2022.0092] [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: 04/27/2023] Open
Abstract
Aim: To assess the role of baseline 18F-fluorodeoxyglucose ([18F]FDG)-positron emission tomography/computed tomography (PET/CT) in predicting response to immunotherapy after 6 months and overall survival (OS) in patients with lung cancer (LC) or malignant melanoma (MM). Methods: Data from a multicenter, retrospective study conducted between March and November 2021 were analyzed. Patients >18 years old with a confirmed diagnosis of LC or MM, who underwent a baseline [18F]FDG-PET/CT within 1-2 months before starting immunotherapy and had a follow-up of at least 12 months were included. PET scans were examined visually and semiquantitatively by physicians at peripheral centers. The metabolic tumor burden (number of lesions with [18F]FDG-uptake) and other parameters were recorded. Clinical response was assessed at 3 and 6 months after starting immunotherapy, and OS was calculated as the time elapsing between the PET scan and death or latest follow-up. Results: The study concerned 177 patients with LC and 101 with MM. Baseline PET/CT was positive in primary or local recurrent lesions in 78.5% and 9.9% of cases, in local/distant lymph nodes in 71.8% and 36.6%, in distant metastases in 58.8% and 84%, respectively, in LC and in MM patients. Among patients with LC, [18F]FDG-uptake in primary/recurrent lung lesions was more often associated with no clinical response to immunotherapy after 6 months than in cases without any tracer uptake. After a mean 21 months, 46.5% of patients with LC and 37.1% with MM had died. A significant correlation emerged between the site/number of [18F]FDG foci and death among patients with LC, but not among those with MM. Conclusions: In patients with LC who are candidates for immunotherapy, baseline [18F]FDG-PET/CT can help to predict response to this therapy after 6 months, and to identify those with a poor prognosis based on their metabolic parameters. For patients with MM, there was only a weak correlation between baseline PET/CT parameters, response to therapy, and survival.
Collapse
Affiliation(s)
- Andrea Bianchi
- Nuclear Medicine Unit, SC Medicina Nucleare, ASO S.Croce e Carle Cuneo, Cuneo, Italy
| | - Maria Luisa De Rimini
- Nuclear Medicine Unit, Department of Health Service, AORN Ospedali dei Colli, Naples, Italy
| | - Rosa Sciuto
- Nuclear Medicine Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Alessio Annovazzi
- Nuclear Medicine Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Silvia Di Traglia
- Nuclear Medicine Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Matteo Bauckneht
- Department of Health Sciences (DISSAL), University of Genova, Genova, Italy
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Francesco Lanfranchi
- Department of Health Sciences (DISSAL), University of Genova, Genova, Italy
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Silvia Morbelli
- Department of Health Sciences (DISSAL), University of Genova, Genova, Italy
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Anna Giulia Nappi
- Section of Nuclear Medicine, Interdisciplinary Department of Medicine, University of Bari "Aldo Moro," Bari, Italy
| | - Cristina Ferrari
- Section of Nuclear Medicine, Interdisciplinary Department of Medicine, University of Bari "Aldo Moro," Bari, Italy
| | - Giuseppe Rubini
- Section of Nuclear Medicine, Interdisciplinary Department of Medicine, University of Bari "Aldo Moro," Bari, Italy
| | - Stefano Panareo
- Nuclear Medicine Unit, Oncology and Haematology Department, University Hospital of Modena, Modena, Italy
| | - Luca Urso
- Nuclear Medicine Unit, Oncology and Specialistic Department, University Hospital of Ferrara, Ferrara, Italy
| | - Mirco Bartolomei
- Nuclear Medicine Unit, Oncology and Specialistic Department, University Hospital of Ferrara, Ferrara, Italy
| | - Davide D'Arienzo
- Nuclear Medicine Unit, Department of Health Service, AORN Ospedali dei Colli, Naples, Italy
| | - Tullio Valente
- U.O.C. Radiologia, Department of Servizi, AORN Ospedali dei Colli, Napoli, Italy
| | - Virginia Rossetti
- Nuclear Medicine Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori," Meldola, Italy
| | - Paola Caroli
- Nuclear Medicine Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori," Meldola, Italy
| | - Federica Matteucci
- Nuclear Medicine Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori," Meldola, Italy
| | - Demetrio Aricò
- Servizio di Medicina Nucleare, Humanitas Istituto Clinico Catanese, Misterbianco, Italy
| | - Michelangelo Bombaci
- Servizio di Medicina Nucleare, Humanitas Istituto Clinico Catanese, Misterbianco, Italy
| | - Domenica Caponnetto
- Servizio di Medicina Nucleare, Humanitas Istituto Clinico Catanese, Misterbianco, Italy
| | - Francesco Bertagna
- Nuclear Medicine Unit, University of Brescia and ASST Spedali Civili di Brescia, Brescia, Italy
| | - Domenico Albano
- Nuclear Medicine Unit, University of Brescia and ASST Spedali Civili di Brescia, Brescia, Italy
| | - Francesco Dondi
- Nuclear Medicine Unit, University of Brescia and ASST Spedali Civili di Brescia, Brescia, Italy
| | - Sara Gusella
- Nuclear Medicine Department, Central Hospital Bolzano (SABES-ASDAA), Bolzano-Bozen, Italy
| | - Alessandro Spimpolo
- Nuclear Medicine Department, Central Hospital Bolzano (SABES-ASDAA), Bolzano-Bozen, Italy
| | - Cinzia Carriere
- Dermatology Department, Central Hospital Bolzano (SABES-ASDAA), Bolzano-Bozen, Italy
| | - Michele Balma
- Nuclear Medicine Unit, SC Medicina Nucleare, ASO S.Croce e Carle Cuneo, Cuneo, Italy
| | - Ambra Buschiazzo
- Nuclear Medicine Unit, SC Medicina Nucleare, ASO S.Croce e Carle Cuneo, Cuneo, Italy
| | - Rosj Gallicchio
- Nuclear Medicine Unit, IRCCS CROB Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Giovanni Storto
- Nuclear Medicine Unit, IRCCS CROB Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Livia Ruffini
- Nuclear Medicine Division, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Maura Scarlattei
- Nuclear Medicine Division, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Giorgio Baldari
- Nuclear Medicine Division, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Anna Rita Cervino
- Nuclear Medicine Unit, Veneto Institute of Oncology IOV-IRCSS, Padua, Italy
| | - Lea Cuppari
- Nuclear Medicine Unit, Veneto Institute of Oncology IOV-IRCSS, Padua, Italy
| | - Marta Burei
- Nuclear Medicine Unit, Veneto Institute of Oncology IOV-IRCSS, Padua, Italy
| | - Giuseppe Trifirò
- Servizio di Medicina Nucleare ICS MAUGERI SPA SB-IRCCS, Pavia, Italy
| | | | - Carolina Arianna Zanini
- Department of Nuclear Medicine, Università Degli Studi di Milano, Milano Statale, Milan, Italy
| | - Alessandra Alessi
- Nuclear Medicine Division, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Valentina Fuoco
- Nuclear Medicine Division, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Ettore Seregni
- Nuclear Medicine Division, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Désirée Deandreis
- Nuclear Medicine Division, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Virginia Liberini
- Nuclear Medicine Unit, SC Medicina Nucleare, ASO S.Croce e Carle Cuneo, Cuneo, Italy
- Nuclear Medicine Division, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Antonino Maria Moreci
- Nuclear Medicine Unit, Az. Ospedaliera Ospedali Riuniti Villa Sofia-Cervello di Palermo, Palermo, Italy
| | - Salvatore Ialuna
- Nuclear Medicine Unit, Az. Ospedaliera Ospedali Riuniti Villa Sofia-Cervello di Palermo, Palermo, Italy
| | - Sabina Pulizzi
- Nuclear Medicine Unit, Az. Ospedaliera Ospedali Riuniti Villa Sofia-Cervello di Palermo, Palermo, Italy
| | - Laura Evangelista
- Nuclear Medicine Unit, Department of Medicine DIMED, University of Padua, Padua, Italy
| |
Collapse
|
11
|
Mangas Losada M, Romero Robles L, Mendoza Melero A, García Megías I, Villanueva Torres A, Garrastachu Zumarán P, Boulvard Chollet X, Lopci E, Ramírez Lasanta R, Delgado Bolton RC. [ 18F]FDG PET/CT in the Evaluation of Melanoma Patients Treated with Immunotherapy. Diagnostics (Basel) 2023; 13:978. [PMID: 36900122 PMCID: PMC10000458 DOI: 10.3390/diagnostics13050978] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/25/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Immunotherapy is based on manipulation of the immune system in order to act against tumour cells, with growing evidence especially in melanoma patients. The challenges faced by this new therapeutic tool are (i) finding valid evaluation criteria for response assessment; (ii) knowing and distinguishing between "atypical" response patterns; (iii) using PET biomarkers as predictive and response evaluation parameters and (iv) diagnosis and management of immunorelated adverse effects. This review is focused on melanoma patients analysing (a) the role of [18F]FDG PET/CT in the mentioned challenges; (b) the evidence of its efficacy. For this purpose, we performed a review of the literature, including original and review articles. In summary, although there are no clearly established or globally accepted criteria, modified response criteria are potentially appropriate for evaluation of immunotherapy benefit. In this context, [18F]FDG PET/CT biomarkers appear to be promising parameters in prediction and assessment of response to immunotherapy. Moreover, immunorelated adverse effects are recognized as predictors of early response to immunotherapy and may be associated with better prognosis and clinical benefit.
Collapse
Affiliation(s)
- María Mangas Losada
- Department of Diagnostic Imaging (Radiology) and Nuclear Medicine, University Hospital San Pedro and Centre for Biomedical Research of La Rioja (CIBIR), 26006 Logroño, Spain
| | - Leonardo Romero Robles
- Department of Diagnostic Imaging (Radiology) and Nuclear Medicine, University Hospital San Pedro and Centre for Biomedical Research of La Rioja (CIBIR), 26006 Logroño, Spain
| | - Alejandro Mendoza Melero
- Department of Diagnostic Imaging (Radiology) and Nuclear Medicine, University Hospital San Pedro and Centre for Biomedical Research of La Rioja (CIBIR), 26006 Logroño, Spain
| | - Irene García Megías
- Department of Diagnostic Imaging (Radiology) and Nuclear Medicine, University Hospital San Pedro and Centre for Biomedical Research of La Rioja (CIBIR), 26006 Logroño, Spain
| | - Amós Villanueva Torres
- Department of Diagnostic Imaging (Radiology) and Nuclear Medicine, University Hospital San Pedro and Centre for Biomedical Research of La Rioja (CIBIR), 26006 Logroño, Spain
| | - Puy Garrastachu Zumarán
- Department of Diagnostic Imaging (Radiology) and Nuclear Medicine, University Hospital San Pedro and Centre for Biomedical Research of La Rioja (CIBIR), 26006 Logroño, Spain
| | - Xavier Boulvard Chollet
- Department of Diagnostic Imaging (Radiology) and Nuclear Medicine, University Hospital San Pedro and Centre for Biomedical Research of La Rioja (CIBIR), 26006 Logroño, Spain
| | - Egesta Lopci
- Nuclear Medicine, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Rafael Ramírez Lasanta
- Department of Diagnostic Imaging (Radiology) and Nuclear Medicine, University Hospital San Pedro and Centre for Biomedical Research of La Rioja (CIBIR), 26006 Logroño, Spain
| | - Roberto C. Delgado Bolton
- Department of Diagnostic Imaging (Radiology) and Nuclear Medicine, University Hospital San Pedro and Centre for Biomedical Research of La Rioja (CIBIR), 26006 Logroño, Spain
| |
Collapse
|
12
|
The influence of metastatic patterns and tumor load on therapeutic efficacy of immunotherapy in patients with metastatic melanoma as determined by quantitative PET-parameters using [18F]-fluorodeoxyglucose PET/computed tomography. Melanoma Res 2023; 33:199-207. [PMID: 36866631 DOI: 10.1097/cmr.0000000000000883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
The introduction of immunotherapy was a revolution in the treatment of metastatic melanoma. Nevertheless, there are only few clinical parameters to predict response to immunotherapy. The purpose of this study was to identify metastatic patterns that can predict response by using noninvasive 18F-FDG PET/CT imaging. In 93 immunotherapy-treated patients, total metabolic tumor volume (MTV) was measured before and after treatment. The differences were compared to quantify therapy response. Patients were divided into seven subgroups regarding the affected organ systems. The results as well as clinical factors were evaluated in multivariate analyses. No subgroup of metastatic patterns had a significant difference in response rates, but with a trend towards poorer response regarding osseous and hepatic metastases. Osseous metastases presented with significant lower disease-specific survival (DSS) (P = 0.001). Sole lymph node metastases were the only subgroup with MTV reduction and with significant higher DSS (57.6 months; P = 0.033). Patients, who ever developed brain metastases, showed a high progression of MTV of 201 ml (P = 0.583) and poor DSS of 49.7 months (P = 0.077). Lower numbers of affected organs indicated significantly higher DSS (hazard ratio, 1.346; P = 0.006). Osseous metastases represented a negative predictive factor for response to immunotherapy and survival. Cerebral metastases, especially when nonresponsive to immunotherapy, predicted poor survival and high increase of MTV. A high number of affected organ systems was identified as a negative factor for response and survival. Patients with only lymph node metastases showed a better response and survival.
Collapse
|
13
|
Iravani A, Wallace R, Lo SN, Galligan A, Weppler AM, Hicks RJ, Sandhu S. FDG PET/CT Prognostic Markers in Patients with Advanced Melanoma Treated with Ipilimumab and Nivolumab. Radiology 2023; 307:e221180. [PMID: 36853183 DOI: 10.1148/radiol.221180] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
Background Despite improved response to combined ipilimumab and nivolumab (hereafter, IpiNivo) treatment for advanced melanoma, many patients exhibit primary or acquired resistance. This, combined with high risk of immune-related adverse events, makes identifying markers predictive of outcomes desirable. Purpose To investigate the prognostic value of fluorine 18 (18F) fluorodeoxyglucose (FDG) PET/CT parameters at baseline and as part of response monitoring in patients with advanced melanoma undergoing IpiNivo treatment. Materials and Methods This was a single-center retrospective study of adult patients with melanoma who received IpiNivo. Baseline FDG PET/CT parameters that included metabolic tumor volume (MTV), tumor stage, mutation status, Eastern Cooperative Oncology Group performance score, lactate dehydrogenase level, and treatment line were correlated with overall survival in univariable and multivariable Cox regression analyses. Treatment response as determined with FDG PET/CT was correlated with overall survival. Results In total, 122 patients (median age, 61 years [IQR, 51-69 years]; 89 men) were included; 78% (95 of 122) had an Eastern Cooperative Oncology Group score of 0, 52% (45 of 86) had an elevated lactate dehydrogenase level, 39% (48 of 122) had a metastatic stage of M1c and 45% (55 of 122) M1d, 45% (55 of 122) had BRAF V600E/K mutation, and the median MTV was 42 mL. Patients with a higher than median MTV at baseline FDG PET/CT had a lower 12-month survival rate compared with those with a lower than median MTV (43% [95% CI: 32, 58] vs 66% [95% CI: 55, 79], P < .001). In multivariable analysis, higher versus lower than median MTV, Eastern Cooperative Oncology Group performance scores of 1-2 versus 0, and subsequent versus first-line IpiNivo treatment were independently associated with overall survival (hazard ratio [HR]: 1.68 [95% CI: 1.02, 2.78], P = .04; 3.1 [95% CI: 1.8, 5.4], P < .001; and 11.2 [95% CI: 3.4, 37.1], P = .002, respectively). The 12-month overall survival rate was lower in patients with progressive disease than in those without progression (35% [95% CI: 24, 51] vs 90% [95% CI: 83, 99]; HR, 7.3 [95% CI: 3.9, 13.3]; P < .001). Conclusion Baseline fluorine 18 fluorodeoxyglucose PET/CT metabolic tumor volume was an independent prognostic marker in patients with advanced melanoma who received ipilimumab and nivolumab treatment. © RSNA, 2023 Supplemental material is available for this article.
Collapse
Affiliation(s)
- Amir Iravani
- From the Molecular Imaging and Therapeutic Nuclear Medicine (A.I.) and Department of Oncology (R.W., S.S.), Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology (A.I., S.S.) and St Vincent's Hospital Department of Medicine (A.G., R.J.H.), University of Melbourne, Melbourne, Australia; Department of Radiology, University of Washington, Seattle, Wash (A.I.); Melanoma Institute Australia, University of Sydney, North Sydney, Australia (S.N.L.); Faculty of Health and Medicine, University of Sydney, Sydney, Australia (S.N.L.); Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia (S.N.L.); Immunology and Diabetes Unit, St Vincent's Institute of Medical Research, Melbourne, Australia (A.G.); Department of Endocrinology and Diabetes, St Vincent's Hospital, Melbourne, Australia (A.G.); and Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada (A.M.W.)
| | - Roslyn Wallace
- From the Molecular Imaging and Therapeutic Nuclear Medicine (A.I.) and Department of Oncology (R.W., S.S.), Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology (A.I., S.S.) and St Vincent's Hospital Department of Medicine (A.G., R.J.H.), University of Melbourne, Melbourne, Australia; Department of Radiology, University of Washington, Seattle, Wash (A.I.); Melanoma Institute Australia, University of Sydney, North Sydney, Australia (S.N.L.); Faculty of Health and Medicine, University of Sydney, Sydney, Australia (S.N.L.); Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia (S.N.L.); Immunology and Diabetes Unit, St Vincent's Institute of Medical Research, Melbourne, Australia (A.G.); Department of Endocrinology and Diabetes, St Vincent's Hospital, Melbourne, Australia (A.G.); and Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada (A.M.W.)
| | - Serigne N Lo
- From the Molecular Imaging and Therapeutic Nuclear Medicine (A.I.) and Department of Oncology (R.W., S.S.), Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology (A.I., S.S.) and St Vincent's Hospital Department of Medicine (A.G., R.J.H.), University of Melbourne, Melbourne, Australia; Department of Radiology, University of Washington, Seattle, Wash (A.I.); Melanoma Institute Australia, University of Sydney, North Sydney, Australia (S.N.L.); Faculty of Health and Medicine, University of Sydney, Sydney, Australia (S.N.L.); Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia (S.N.L.); Immunology and Diabetes Unit, St Vincent's Institute of Medical Research, Melbourne, Australia (A.G.); Department of Endocrinology and Diabetes, St Vincent's Hospital, Melbourne, Australia (A.G.); and Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada (A.M.W.)
| | - Anna Galligan
- From the Molecular Imaging and Therapeutic Nuclear Medicine (A.I.) and Department of Oncology (R.W., S.S.), Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology (A.I., S.S.) and St Vincent's Hospital Department of Medicine (A.G., R.J.H.), University of Melbourne, Melbourne, Australia; Department of Radiology, University of Washington, Seattle, Wash (A.I.); Melanoma Institute Australia, University of Sydney, North Sydney, Australia (S.N.L.); Faculty of Health and Medicine, University of Sydney, Sydney, Australia (S.N.L.); Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia (S.N.L.); Immunology and Diabetes Unit, St Vincent's Institute of Medical Research, Melbourne, Australia (A.G.); Department of Endocrinology and Diabetes, St Vincent's Hospital, Melbourne, Australia (A.G.); and Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada (A.M.W.)
| | - Alison M Weppler
- From the Molecular Imaging and Therapeutic Nuclear Medicine (A.I.) and Department of Oncology (R.W., S.S.), Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology (A.I., S.S.) and St Vincent's Hospital Department of Medicine (A.G., R.J.H.), University of Melbourne, Melbourne, Australia; Department of Radiology, University of Washington, Seattle, Wash (A.I.); Melanoma Institute Australia, University of Sydney, North Sydney, Australia (S.N.L.); Faculty of Health and Medicine, University of Sydney, Sydney, Australia (S.N.L.); Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia (S.N.L.); Immunology and Diabetes Unit, St Vincent's Institute of Medical Research, Melbourne, Australia (A.G.); Department of Endocrinology and Diabetes, St Vincent's Hospital, Melbourne, Australia (A.G.); and Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada (A.M.W.)
| | - Rodney J Hicks
- From the Molecular Imaging and Therapeutic Nuclear Medicine (A.I.) and Department of Oncology (R.W., S.S.), Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology (A.I., S.S.) and St Vincent's Hospital Department of Medicine (A.G., R.J.H.), University of Melbourne, Melbourne, Australia; Department of Radiology, University of Washington, Seattle, Wash (A.I.); Melanoma Institute Australia, University of Sydney, North Sydney, Australia (S.N.L.); Faculty of Health and Medicine, University of Sydney, Sydney, Australia (S.N.L.); Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia (S.N.L.); Immunology and Diabetes Unit, St Vincent's Institute of Medical Research, Melbourne, Australia (A.G.); Department of Endocrinology and Diabetes, St Vincent's Hospital, Melbourne, Australia (A.G.); and Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada (A.M.W.)
| | - Shahneen Sandhu
- From the Molecular Imaging and Therapeutic Nuclear Medicine (A.I.) and Department of Oncology (R.W., S.S.), Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology (A.I., S.S.) and St Vincent's Hospital Department of Medicine (A.G., R.J.H.), University of Melbourne, Melbourne, Australia; Department of Radiology, University of Washington, Seattle, Wash (A.I.); Melanoma Institute Australia, University of Sydney, North Sydney, Australia (S.N.L.); Faculty of Health and Medicine, University of Sydney, Sydney, Australia (S.N.L.); Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia (S.N.L.); Immunology and Diabetes Unit, St Vincent's Institute of Medical Research, Melbourne, Australia (A.G.); Department of Endocrinology and Diabetes, St Vincent's Hospital, Melbourne, Australia (A.G.); and Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada (A.M.W.)
| |
Collapse
|
14
|
Lopci E, Castello A, Filippi L. Novelties from the Joint EANM/SNMMI/ANZSNM Guidelines on Immunotherapy. Cancer Biother Radiopharm 2023; 38:211-215. [PMID: 36730788 DOI: 10.1089/cbr.2022.0091] [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: 02/04/2023] Open
Abstract
In the past decade, the implementation of immunotherapy with checkpoint inhibitors has determined a major change in the management of oncological patients. The challenges associated to the new therapeutic regimen have promoted adapted criteria for response assessment to interpret imaging findings and atypical patterns of response. Parallel to the new morphological criteria, also 18fluoro-deoxyglucose positron emission/computed tomography imaging has required novel approaches and specific guidelines on how to perform, interpret, and report the scan in patients with solid tumors under immune checkpoint inhibitors therapy. A summary of the novelties related to the new joint international European Association of Nuclear Medicine (EANM)/Society of Nuclear Medicine and Molecular Imaging (SNMMI)/Australian and New Zealand Society of Nuclear Medicine (ANZSNM) guidelines on immunotherapy is provided herein to elucidate most critical aspects in image interpretation.
Collapse
Affiliation(s)
- Egesta Lopci
- Nuclear Medicine Unit, IRCCS-Humanitas Research Hospital, Rozzano, Italy
| | - Angelo Castello
- Nuclear Medicine Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca Filippi
- Department of Nuclear Medicine, Santa Maria Goretti Hospital, Latina, Italy
| |
Collapse
|
15
|
Dobre EG, Surcel M, Constantin C, Ilie MA, Caruntu A, Caruntu C, Neagu M. Skin Cancer Pathobiology at a Glance: A Focus on Imaging Techniques and Their Potential for Improved Diagnosis and Surveillance in Clinical Cohorts. Int J Mol Sci 2023; 24:1079. [PMID: 36674595 PMCID: PMC9866322 DOI: 10.3390/ijms24021079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/08/2023] Open
Abstract
Early diagnosis is essential for completely eradicating skin cancer and maximizing patients' clinical benefits. Emerging optical imaging modalities such as reflectance confocal microscopy (RCM), optical coherence tomography (OCT), magnetic resonance imaging (MRI), near-infrared (NIR) bioimaging, positron emission tomography (PET), and their combinations provide non-invasive imaging data that may help in the early detection of cutaneous tumors and surgical planning. Hence, they seem appropriate for observing dynamic processes such as blood flow, immune cell activation, and tumor energy metabolism, which may be relevant for disease evolution. This review discusses the latest technological and methodological advances in imaging techniques that may be applied for skin cancer detection and monitoring. In the first instance, we will describe the principle and prospective clinical applications of the most commonly used imaging techniques, highlighting the challenges and opportunities of their implementation in the clinical setting. We will also highlight how imaging techniques may complement the molecular and histological approaches in sharpening the non-invasive skin characterization, laying the ground for more personalized approaches in skin cancer patients.
Collapse
Affiliation(s)
- Elena-Georgiana Dobre
- Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095 Bucharest, Romania
| | - Mihaela Surcel
- Immunology Department, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania
| | - Carolina Constantin
- Immunology Department, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania
- Department of Pathology, Colentina University Hospital, 020125 Bucharest, Romania
| | | | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Monica Neagu
- Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095 Bucharest, Romania
- Immunology Department, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania
- Department of Pathology, Colentina University Hospital, 020125 Bucharest, Romania
| |
Collapse
|
16
|
Wang X, Yang X, Wang J, Dong C, Ding J, Wu M, Wang Y, Ding H, Zhang H, Sang X, Zhao H, Huo L. Metabolic Tumor Volume Measured by 18F-FDG PET/CT is Associated with the Survival of Unresectable Hepatocellular Carcinoma Treated with PD-1/PD-L1 Inhibitors Plus Molecular Targeted Agents. J Hepatocell Carcinoma 2023; 10:587-598. [PMID: 37063093 PMCID: PMC10094465 DOI: 10.2147/jhc.s401647] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 03/21/2023] [Indexed: 04/18/2023] Open
Abstract
Purpose The combination of PD-1/PD-L1 inhibitors and molecular targeted agents showed promising efficacy for unresectable hepatocellular carcinoma (uHCC). This study aimed to investigate the prognostic value of metabolic parameters from 18F-fluorodeoxyglucose positron emission tomography-computed tomography (18F-FDG PET/CT) in patients with uHCC underwent the combined therapies. Patients and Methods Patients with uHCC treated with a combination of immunotherapy and targeted therapy who underwent baseline 18F-FDG PET/CT between July 2018 and December 2021 were recruited retrospectively. The metabolic tumor volume (MTV), total lesion glycolysis (TLG), maximum standardized uptake values (SUVmax), and clinical and biological parameters were recorded. A multivariate prediction model was developed for overall survival (OS) using these parameters together with clinical prognostic factors. Results Seventy-seven patients were finally included. The median OS was 16.8 months. We found that a high MTV (≥39.65 cm3 as the median value) was significantly associated with OS (P<0.05). In multivariate analyses for OS, a high MTV, high Eastern Cooperative Oncology Group performance status (ECOG-PS, ≥1), Child-Pugh (B-C) grade, and the presence of bone metastasis were significantly associated with poor OS (HR 1.371, HR 3.73, HR 15.384, and HR 2.994, all P<0.05, respectively). A multivariate prognostic model including MTV and prognostic factors, such as ECOG-PS, Child-Pugh grade, and bone metastasis, further improved the identification of different OS subgroups. Conclusion High MTV is an adverse prognostic factor in patients with uHCC treated with a combination of immunotherapy and molecular targeted agents. Integrating PET/CT parameters with clinical prognostic factors could help to personalize immunotherapy.
Collapse
Affiliation(s)
- Xuezhu Wang
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Center for Rare Diseases Research, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Xu Yang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Jingnan Wang
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Center for Rare Diseases Research, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Chengyan Dong
- GE Healthcare China, Beijing, People’s Republic of China
| | - Jie Ding
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Center for Rare Diseases Research, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Meiqi Wu
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Center for Rare Diseases Research, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Yanyu Wang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Haiyan Ding
- Department of Biomedical Engineering, Tsinghua University, Beijing, People’s Republic of China
| | - Hui Zhang
- Department of Biomedical Engineering, Tsinghua University, Beijing, People’s Republic of China
| | - Xinting Sang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Haitao Zhao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- Correspondence: Li Huo; Haitao Zhao, #1 Shuaifuyuan, Dongcheng District, Beijing, People’s Republic of China, Tel +86 13910801986; +86 13901246374, Email ;
| | - Li Huo
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Center for Rare Diseases Research, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- Correspondence: Li Huo; Haitao Zhao, #1 Shuaifuyuan, Dongcheng District, Beijing, People’s Republic of China, Tel +86 13910801986; +86 13901246374, Email ;
| |
Collapse
|
17
|
Blanchet K, Sebille JC, Frenard C, Lecerf P, Khammari A, Carlier T, Bodet-Milin C, Dréno B. The predictive power of FDG-PET imaging with regard to immunotherapy in real-life conditions in advanced melanoma: An exploratory study. J Eur Acad Dermatol Venereol 2023; 37:e61-e62. [PMID: 35974443 DOI: 10.1111/jdv.18501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 07/27/2022] [Indexed: 12/15/2022]
Affiliation(s)
- Katleen Blanchet
- Department of Dermatology, CIC 1413, IT services, CHU Nantes, Nantes, France
| | | | - Cécile Frenard
- INSERM, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302, Nantes Université, Nantes, France
| | | | - Amir Khammari
- INSERM, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302, Nantes Université, Nantes, France
| | - Thomas Carlier
- Department of Nuclear Medicine, CHU Nantes, Nantes, France
| | | | - Brigitte Dréno
- INSERM, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302, Nantes Université, Nantes, France.,Nantes Université, Univ Angers, INSERM, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302, Nantes, France
| |
Collapse
|
18
|
Gao Y, Wu C, Chen X, Ma L, Zhang X, Chen J, Liao X, Liu M. PET/CT molecular imaging in the era of immune-checkpoint inhibitors therapy. Front Immunol 2022; 13:1049043. [PMID: 36341331 PMCID: PMC9630646 DOI: 10.3389/fimmu.2022.1049043] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 10/10/2022] [Indexed: 04/24/2024] Open
Abstract
Cancer immunotherapy, especially immune-checkpoint inhibitors (ICIs), has paved a new way for the treatment of many types of malignancies, particularly advanced-stage cancers. Accumulating evidence suggests that as a molecular imaging modality, positron emission tomography/computed tomography (PET/CT) can play a vital role in the management of ICIs therapy by using different molecular probes and metabolic parameters. In this review, we will provide a comprehensive overview of the clinical data to support the importance of 18F-fluorodeoxyglucose PET/CT (18F-FDG PET/CT) imaging in the treatment of ICIs, including the evaluation of the tumor microenvironment, discovery of immune-related adverse events, evaluation of therapeutic efficacy, and prediction of therapeutic prognosis. We also discuss perspectives on the development direction of 18F-FDG PET/CT imaging, with a particular emphasis on possible challenges in the future. In addition, we summarize the researches on novel PET molecular probes that are expected to potentially promote the precise application of ICIs.
Collapse
|
19
|
Kwon HR, Cho J, Park S, Lee SH, Ahn MJ, Choi JY, Lee KH, Jung HA, Moon SH. Metabolic parameters on baseline 18F-FDG PET/CT are potential predictive biomarkers for immunotherapy in patients with head and neck squamous cell carcinoma. Front Med (Lausanne) 2022; 9:896494. [PMID: 36226146 PMCID: PMC9548588 DOI: 10.3389/fmed.2022.896494] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 08/26/2022] [Indexed: 11/13/2022] Open
Abstract
PurposeWe evaluated baseline 18F-fluorodeoxyglucose (FDG) positron emission tomography/computed tomography (PET/CT) metabolic parameters for predicting prognosis in patients with head and neck squamous cell carcinoma (HNSCC) who were receiving immune checkpoint inhibitors (ICIs). In addition, we also investigated the relationships between immunohistochemical (IHC) biomarkers and metabolic parameters.Materials and methodsA total of 39 patients with HNSCC who underwent 18F-FDG PET/CT prior to ICI therapy between November 2015 and December 2020 were enrolled. PET parameters of tumor lesions included standardized uptake values, metabolic tumor volume (MTV), total lesion glycolysis (TLG), and spleen-to-liver ratio (SLR). Clinical variables, IHC markers, and derived neutrophil-to-lymphocyte ratio (dNLR) were also obtained. Analysis was performed using Cox proportional hazard model, Kaplan-Meier method with log-rank test, and Spearman's correlation.ResultsTotal MTV (TMTV), total TLG (TTLG), and a combined parameter consisting of TMTV and dNLR were significant predictors for progression-free survival (PFS) in univariable analysis (TMTV, p = 0.018; TTLG, p = 0.027; combined parameter, p = 0.021). Above all, the combined parameter was an independent prognostic factor for PFS in multivariable analysis. The group with low TMTV and low dNLR had longer PFS than the group with high TMTV and high dNLR (p = 0.036). SLR was the only significant predictor for overall survival (p = 0.019). Additionally, there was a negative correlation between programmed cell death-ligand 1 expression (one of the IHC markers) and MTV in subgroup analysis.ConclusionPET parameters on baseline 18F-FDG PET/CT were predictive biomarkers for prognosis in patients with HNSCC undergoing ICI therapy. With dNLR, more accurate prognostic prediction could be possible.
Collapse
Affiliation(s)
- Hye Ryeong Kwon
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Department of Nuclear Medicine, Ilsan Paik Hospital, Inje University College of Medicine, Goyang-si, South Korea
| | - Junhun Cho
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Sehhoon Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Se-Hoon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Joon Young Choi
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kyung-Han Lee
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hyun Ae Jung
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- *Correspondence: Hyun Ae Jung
| | - Seung Hwan Moon
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Seung Hwan Moon
| |
Collapse
|
20
|
Voglis S, Schaller V, Müller T, Gönel M, Winklhofer S, Mangana J, Dummer R, Serra C, Weller M, Regli L, Le Rhun E, Neidert MC. Maximal surgical tumour load reduction in immune-checkpoint inhibitor naïve patients with melanoma brain metastases correlates with prolonged survival. Eur J Cancer 2022; 175:158-168. [PMID: 36126476 DOI: 10.1016/j.ejca.2022.08.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/04/2022] [Accepted: 08/16/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Recent therapeutic advances in metastatic melanoma have led to improved overall survival (OS) rates, with consequently an increased incidence of brain metastases (BM). The role of BM resection in the era of targeted and immunotherapy should be reassessed. In the current study we analysed the role of residual intracranial tumour load in a cohort of melanoma BM patients. METHODS Retrospective single-centre analysis of a prospective registry of resected melanoma BM from 2013 to 2021. Correlations of residual tumour volume and outcome were determined with respect to patient, tumour and treatment regimens characteristics. RESULTS 121 individual patients (66% male, mean age 59.9 years) were identified and included in the study. Pre- and postoperative systemic treatments included BRAF/MEK inhibitors, as well as combination or monotherapy of immune-checkpoint inhibitors (ICIs). Median OS of the entire cohort was 20 months. Cox proportional-hazard analysis revealed postoperative anti-CTLA4+anti-PD-1 therapy (HR 0.07, p = .01) and postoperative residual intracranial tumour burden (HR 1.4, p = .027) as significant predictors for OS. Further analysis revealed that ICI-naïve patients with residual tumour volume ≤3.5 cm3 and postoperative ICI showed significantly prolonged OS compared to patients with residual volume >3.5 cm3 (p < .0001). Subgroup analysis of ICI-naïve patients showed steroid intake postoperatively to be negatively associated with OS, however residual tumour volume ≤3.5 cm3 remained independently correlated with superior OS (HR 0.14, p < .001). CONCLUSION Besides known predictive factors like postoperative ICI, a maximal intracranial tumour burden reduction seems to be beneficial, especially in ICI-naïve patients. This highlights the importance of local CNS control and the need to further investigating the role of initial surgical tumour load reduction in randomised clinical trials.
Collapse
Affiliation(s)
- Stefanos Voglis
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland.
| | - Valentina Schaller
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Timothy Müller
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Meltem Gönel
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Sebastian Winklhofer
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Joana Mangana
- Department of Dermatology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Carlo Serra
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Luca Regli
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Emilie Le Rhun
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland; Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Marian C Neidert
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland; Department of Neurosurgery, Cantonal Hospital St.Gallen, University of St.Gallen Medical School, St.Gallen, Switzerland
| |
Collapse
|
21
|
Abstract
MRI is a widely available clinical tool for cancer diagnosis and treatment monitoring. MRI provides excellent soft tissue imaging, using a wide range of contrast mechanisms, and can non-invasively detect tissue metabolites. These approaches can be used to distinguish cancer from normal tissues, to stratify tumor aggressiveness, and to identify changes within both the tumor and its microenvironment in response to therapy. In this review, the role of MRI in immunotherapy monitoring will be discussed and how it could be utilized in the future to address some of the unique clinical questions that arise from immunotherapy. For example, MRI could play a role in identifying pseudoprogression, mixed response, T cell infiltration, cell tracking, and some of the characteristic immune-related adverse events associated with these agents. The factors to be considered when developing MRI imaging biomarkers for immunotherapy will be reviewed. Finally, the advantages and limitations of each approach will be discussed, as well as the challenges for future clinical translation into routine clinical care. Given the increasing use of immunotherapy in a wide range of cancers and the ability of MRI to detect the microstructural and functional changes associated with successful response to immunotherapy, the technique has great potential for more widespread and routine use in the future for these applications.
Collapse
Affiliation(s)
- Doreen Lau
- Centre for Immuno-Oncology, University of Oxford, Oxford, UK
| | - Pippa G Corrie
- Department of Oncology, Addenbrooke's Hospital, Cambridge, UK
| | | |
Collapse
|
22
|
Küstner T, Vogel J, Hepp T, Forschner A, Pfannenberg C, Schmidt H, Schwenzer NF, Nikolaou K, la Fougère C, Seith F. Development of a Hybrid-Imaging-Based Prognostic Index for Metastasized-Melanoma Patients in Whole-Body 18F-FDG PET/CT and PET/MRI Data. Diagnostics (Basel) 2022; 12:2102. [PMID: 36140504 PMCID: PMC9498091 DOI: 10.3390/diagnostics12092102] [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: 07/14/2022] [Revised: 08/19/2022] [Accepted: 08/25/2022] [Indexed: 11/17/2022] Open
Abstract
Besides tremendous treatment success in advanced melanoma patients, the rapid development of oncologic treatment options comes with increasingly high costs and can cause severe life-threatening side effects. For this purpose, predictive baseline biomarkers are becoming increasingly important for risk stratification and personalized treatment planning. Thus, the aim of this pilot study was the development of a prognostic tool for the risk stratification of the treatment response and mortality based on PET/MRI and PET/CT, including a convolutional neural network (CNN) for metastasized-melanoma patients before systemic-treatment initiation. The evaluation was based on 37 patients (19 f, 62 ± 13 y/o) with unresectable metastasized melanomas who underwent whole-body 18F-FDG PET/MRI and PET/CT scans on the same day before the initiation of therapy with checkpoint inhibitors and/or BRAF/MEK inhibitors. The overall survival (OS), therapy response, metastatically involved organs, number of lesions, total lesion glycolysis, total metabolic tumor volume (TMTV), peak standardized uptake value (SULpeak), diameter (Dmlesion) and mean apparent diffusion coefficient (ADCmean) were assessed. For each marker, a Kaplan−Meier analysis and the statistical significance (Wilcoxon test, paired t-test and Bonferroni correction) were assessed. Patients were divided into high- and low-risk groups depending on the OS and treatment response. The CNN segmentation and prediction utilized multimodality imaging data for a complementary in-depth risk analysis per patient. The following parameters correlated with longer OS: a TMTV < 50 mL; no metastases in the brain, bone, liver, spleen or pleura; ≤4 affected organ regions; no metastases; a Dmlesion > 37 mm or SULpeak < 1.3; a range of the ADCmean < 600 mm2/s. However, none of the parameters correlated significantly with the stratification of the patients into the high- or low-risk groups. For the CNN, the sensitivity, specificity, PPV and accuracy were 92%, 96%, 92% and 95%, respectively. Imaging biomarkers such as the metastatic involvement of specific organs, a high tumor burden, the presence of at least one large lesion or a high range of intermetastatic diffusivity were negative predictors for the OS, but the identification of high-risk patients was not feasible with the handcrafted parameters. In contrast, the proposed CNN supplied risk stratification with high specificity and sensitivity.
Collapse
Affiliation(s)
- Thomas Küstner
- MIDAS.Lab, Department of Radiology, University Hospital of Tübingen, 72076 Tubingen, Germany
| | - Jonas Vogel
- Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, University Hospital Tübingen, 72076 Tubingen, Germany
| | - Tobias Hepp
- MIDAS.Lab, Department of Radiology, University Hospital of Tübingen, 72076 Tubingen, Germany
| | - Andrea Forschner
- Department of Dermatology, University Hospital of Tübingen, 72070 Tubingen, Germany
| | - Christina Pfannenberg
- Department of Radiology, Diagnostic and Interventional Radiology, University Hospital of Tübingen, 72076 Tubingen, Germany
| | - Holger Schmidt
- Faculty of Medicine, Eberhard-Karls-University Tübingen, 72076 Tubingen, Germany
- Siemens Healthineers, 91052 Erlangen, Germany
| | - Nina F. Schwenzer
- Faculty of Medicine, Eberhard-Karls-University Tübingen, 72076 Tubingen, Germany
| | - Konstantin Nikolaou
- Department of Radiology, Diagnostic and Interventional Radiology, University Hospital of Tübingen, 72076 Tubingen, Germany
- Cluster of Excellence iFIT (EXC 2180) Image-Guided and Functionally Instructed Tumor Therapies, Eberhard Karls University, 72076 Tubingen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Tübingen, 72076 Tubingen, Germany
| | - Christian la Fougère
- Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, University Hospital Tübingen, 72076 Tubingen, Germany
- Cluster of Excellence iFIT (EXC 2180) Image-Guided and Functionally Instructed Tumor Therapies, Eberhard Karls University, 72076 Tubingen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Tübingen, 72076 Tubingen, Germany
| | - Ferdinand Seith
- Department of Radiology, Diagnostic and Interventional Radiology, University Hospital of Tübingen, 72076 Tubingen, Germany
| |
Collapse
|
23
|
|
24
|
Hughes DJ, Subesinghe M, Taylor B, Bille A, Spicer J, Papa S, Goh V, Cook GJR. 18F FDG PET/CT and Novel Molecular Imaging for Directing Immunotherapy in Cancer. Radiology 2022; 304:246-264. [PMID: 35762888 DOI: 10.1148/radiol.212481] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Immunotherapy has transformed the treatment landscape of many cancers, with durable responses in disease previously associated with a poor prognosis. Patient selection remains a challenge, with predictive biomarkers an urgent unmet clinical need. Current predictive biomarkers, including programmed death-ligand 1 (PD-L1) (measured with immunohistochemistry), are imperfect. Promising biomarkers, including tumor mutation burden and tumor infiltrating lymphocyte density, fail to consistently predict response and have yet to translate to routine clinical practice. Heterogeneity of immune response within and between lesions presents a further challenge where fluorine 18 fluorodeoxyglucose PET/CT has a potential role in assessing response, stratifying treatment, and detecting and monitoring immune-related toxicities. Novel radiopharmaceuticals also present a unique opportunity to define the immune tumor microenvironment to better predict which patients may respond to therapy, for example by means of in vivo whole-body PD-L1 and CD8+ T cell expression imaging. In addition, longitudinal molecular imaging may help further define dynamic changes, particularly in cases of immunotherapy resistance, helping to direct a more personalized therapeutic approach. This review highlights current and emerging applications of molecular imaging to stratify, predict, and monitor molecular dynamics and treatment response in areas of clinical need.
Collapse
Affiliation(s)
- Daniel J Hughes
- From the Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, Westminster Bridge Road, 4th Floor, Lambeth Wing, London SE1 7EH, UK (D.J.H., M.S., V.G., G.J.R.C.); King's College London and Guy's and St Thomas' PET Centre, London, UK (D.J.H., M.S., G.J.R.C.); Comprehensive Cancer Centre (B.T., A.B.), Department of Thoracic Surgery (A.B.), and Department of Radiology (V.G.), Guy's and St Thomas' NHS Foundation Trust, London, UK; and School of Cancer and Pharmaceutical Sciences, King's College London, London, UK (J.S., S.P.)
| | - Manil Subesinghe
- From the Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, Westminster Bridge Road, 4th Floor, Lambeth Wing, London SE1 7EH, UK (D.J.H., M.S., V.G., G.J.R.C.); King's College London and Guy's and St Thomas' PET Centre, London, UK (D.J.H., M.S., G.J.R.C.); Comprehensive Cancer Centre (B.T., A.B.), Department of Thoracic Surgery (A.B.), and Department of Radiology (V.G.), Guy's and St Thomas' NHS Foundation Trust, London, UK; and School of Cancer and Pharmaceutical Sciences, King's College London, London, UK (J.S., S.P.)
| | - Benjamin Taylor
- From the Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, Westminster Bridge Road, 4th Floor, Lambeth Wing, London SE1 7EH, UK (D.J.H., M.S., V.G., G.J.R.C.); King's College London and Guy's and St Thomas' PET Centre, London, UK (D.J.H., M.S., G.J.R.C.); Comprehensive Cancer Centre (B.T., A.B.), Department of Thoracic Surgery (A.B.), and Department of Radiology (V.G.), Guy's and St Thomas' NHS Foundation Trust, London, UK; and School of Cancer and Pharmaceutical Sciences, King's College London, London, UK (J.S., S.P.)
| | - Andrea Bille
- From the Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, Westminster Bridge Road, 4th Floor, Lambeth Wing, London SE1 7EH, UK (D.J.H., M.S., V.G., G.J.R.C.); King's College London and Guy's and St Thomas' PET Centre, London, UK (D.J.H., M.S., G.J.R.C.); Comprehensive Cancer Centre (B.T., A.B.), Department of Thoracic Surgery (A.B.), and Department of Radiology (V.G.), Guy's and St Thomas' NHS Foundation Trust, London, UK; and School of Cancer and Pharmaceutical Sciences, King's College London, London, UK (J.S., S.P.)
| | - James Spicer
- From the Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, Westminster Bridge Road, 4th Floor, Lambeth Wing, London SE1 7EH, UK (D.J.H., M.S., V.G., G.J.R.C.); King's College London and Guy's and St Thomas' PET Centre, London, UK (D.J.H., M.S., G.J.R.C.); Comprehensive Cancer Centre (B.T., A.B.), Department of Thoracic Surgery (A.B.), and Department of Radiology (V.G.), Guy's and St Thomas' NHS Foundation Trust, London, UK; and School of Cancer and Pharmaceutical Sciences, King's College London, London, UK (J.S., S.P.)
| | - Sophie Papa
- From the Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, Westminster Bridge Road, 4th Floor, Lambeth Wing, London SE1 7EH, UK (D.J.H., M.S., V.G., G.J.R.C.); King's College London and Guy's and St Thomas' PET Centre, London, UK (D.J.H., M.S., G.J.R.C.); Comprehensive Cancer Centre (B.T., A.B.), Department of Thoracic Surgery (A.B.), and Department of Radiology (V.G.), Guy's and St Thomas' NHS Foundation Trust, London, UK; and School of Cancer and Pharmaceutical Sciences, King's College London, London, UK (J.S., S.P.)
| | - Vicky Goh
- From the Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, Westminster Bridge Road, 4th Floor, Lambeth Wing, London SE1 7EH, UK (D.J.H., M.S., V.G., G.J.R.C.); King's College London and Guy's and St Thomas' PET Centre, London, UK (D.J.H., M.S., G.J.R.C.); Comprehensive Cancer Centre (B.T., A.B.), Department of Thoracic Surgery (A.B.), and Department of Radiology (V.G.), Guy's and St Thomas' NHS Foundation Trust, London, UK; and School of Cancer and Pharmaceutical Sciences, King's College London, London, UK (J.S., S.P.)
| | - Gary J R Cook
- From the Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, Westminster Bridge Road, 4th Floor, Lambeth Wing, London SE1 7EH, UK (D.J.H., M.S., V.G., G.J.R.C.); King's College London and Guy's and St Thomas' PET Centre, London, UK (D.J.H., M.S., G.J.R.C.); Comprehensive Cancer Centre (B.T., A.B.), Department of Thoracic Surgery (A.B.), and Department of Radiology (V.G.), Guy's and St Thomas' NHS Foundation Trust, London, UK; and School of Cancer and Pharmaceutical Sciences, King's College London, London, UK (J.S., S.P.)
| |
Collapse
|
25
|
18FDG PET Assessment of Therapeutic Response in Patients with Advanced or Metastatic Melanoma Treated with First-Line Immune Checkpoint Inhibitors. Cancers (Basel) 2022; 14:cancers14133190. [PMID: 35804963 PMCID: PMC9264956 DOI: 10.3390/cancers14133190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 12/21/2022] Open
Abstract
Simple Summary In a retrospective study of patients with advanced or metastatic melanoma treated with first-line immune checkpoint inhibitors, we investigated the value of metabolic criteria, PERCIST 5 (criteria used for conventional chemotherapy), and imPERCIST5 (criteria adapted for immunotherapy therapeutic evaluation). Responding patients according to both criteria had better overall survival than that of not-responding patients, with a 2 years OS of 91% versus 39%, respectively. Combining different approaches to assess response could help improve the confidence in the test aiming at evaluating the response to immunotherapy. Abstract Background: Immune checkpoint inhibitors (ICI) are currently the first-line treatment for patients with metastatic melanoma. We investigated the value of positron emission tomography (PET) response criteria to assess the therapeutic response to first-line ICI in this clinical context and explore the potential contribution of total tumor metabolic volume (TMTV) analysis. Methods: We conducted a retrospective study in patients treated with first-line ICI for advanced or metastatic melanoma, with 18F-FDG PET/CT performed at baseline and 3 months after starting treatment. Patients’ metabolic response was classified according to PERCIST5 and imPERCIST 5 criteria. TMTV was recorded for each examination. Results: Twenty-nine patients were included. The median overall survival (OS) was 51.2 months (IQR 13.6—not reached), and the OS rate at 2 years was 58.6%. Patients classified as responders (complete and partial response) had a 90.9% 2-year OS rate versus 38.9% for non-responders (stable disease and progressive disease) (p = 0.03), for PERCIST5 and imPERCIST 5 criteria. The median change in metabolic volume was 9.8% (IQR −59–+140%). No significant correlation between OS and changes in TMTV was found. Conclusion: The evaluation of response to immunotherapy using metabolic imaging with PERCIST5 and imPERCIST5 was significantly associated with OS in patients with advanced or metastatic melanoma.
Collapse
|
26
|
Lopci E, Hicks RJ, Dimitrakopoulou-Strauss A, Dercle L, Iravani A, Seban RD, Sachpekidis C, Humbert O, Gheysens O, Glaudemans AWJM, Weber W, Wahl RL, Scott AM, Pandit-Taskar N, Aide N. Joint EANM/SNMMI/ANZSNM practice guidelines/procedure standards on recommended use of [ 18F]FDG PET/CT imaging during immunomodulatory treatments in patients with solid tumors version 1.0. Eur J Nucl Med Mol Imaging 2022; 49:2323-2341. [PMID: 35376991 PMCID: PMC9165250 DOI: 10.1007/s00259-022-05780-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/22/2022] [Indexed: 12/13/2022]
Abstract
PURPOSE The goal of this guideline/procedure standard is to assist nuclear medicine physicians, other nuclear medicine professionals, oncologists or other medical specialists for recommended use of [18F]FDG PET/CT in oncological patients undergoing immunotherapy, with special focus on response assessment in solid tumors. METHODS In a cooperative effort between the EANM, the SNMMI and the ANZSNM, clinical indications, recommended imaging procedures and reporting standards have been agreed upon and summarized in this joint guideline/procedure standard. CONCLUSIONS The field of immuno-oncology is rapidly evolving, and this guideline/procedure standard should not be seen as definitive, but rather as a guidance document standardizing the use and interpretation of [18F]FDG PET/CT during immunotherapy. Local variations to this guideline should be taken into consideration. PREAMBLE The European Association of Nuclear Medicine (EANM) is a professional non-profit medical association founded in 1985 to facilitate worldwide communication among individuals pursuing clinical and academic excellence in nuclear medicine. The Society of Nuclear Medicine and Molecular Imaging (SNMMI) is an international scientific and professional organization founded in 1954 to promote science, technology and practical application of nuclear medicine. The Australian and New Zealand Society of Nuclear Medicine (ANZSNM), founded in 1969, represents the major professional society fostering the technical and professional development of nuclear medicine practice across Australia and New Zealand. It promotes excellence in the nuclear medicine profession through education, research and a commitment to the highest professional standards. EANM, SNMMI and ANZSNM members are physicians, technologists, physicists and scientists specialized in the research and clinical practice of nuclear medicine. All three societies will periodically put forth new standards/guidelines for nuclear medicine practice to help advance the science of nuclear medicine and improve service to patients. Existing standards/guidelines will be reviewed for revision or renewal, as appropriate, on their fifth anniversary or sooner, if indicated. Each standard/guideline, representing a policy statement by the EANM/SNMMI/ANZSNM, has undergone a thorough consensus process, entailing extensive review. These societies recognize that the safe and effective use of diagnostic nuclear medicine imaging requires particular training and skills, as described in each document. These standards/guidelines are educational tools designed to assist practitioners in providing appropriate and effective nuclear medicine care for patients. These guidelines are consensus documents based on current knowledge. They are not intended to be inflexible rules or requirements of practice, nor should they be used to establish a legal standard of care. For these reasons and those set forth below, the EANM, SNMMI and ANZSNM caution against the use of these standards/guidelines in litigation in which the clinical decisions of a practitioner are called into question. The ultimate judgment regarding the propriety of any specific procedure or course of action must be made by medical professionals considering the unique circumstances of each case. Thus, there is no implication that an action differing from what is laid out in the guidelines/procedure standards, standing alone, is below standard of care. To the contrary, a conscientious practitioner may responsibly adopt a course of action different from that set forth in the standards/guidelines when, in the reasonable judgment of the practitioner, such course of action is indicated by the condition of the patient, limitations of available resources or advances in knowledge or technology subsequent to publication of the guidelines/procedure standards. The practice of medicine involves not only the science, but also the art of dealing with the prevention, diagnosis, alleviation and treatment of disease. The variety and complexity of human conditions make it impossible for general guidelines to consistently allow for an accurate diagnosis to be reached or a particular treatment response to be predicted. Therefore, it should be recognized that adherence to these standards/ guidelines will not ensure a successful outcome. All that should be expected is that practitioners follow a reasonable course of action, based on their level of training, current knowledge, clinical practice guidelines, available resources and the needs/context of the patient being treated. The sole purpose of these guidelines is to assist practitioners in achieving this objective. The present guideline/procedure standard was developed collaboratively by the EANM, the SNMMI and the ANZSNM, with the support of international experts in the field. They summarize also the views of the Oncology and Theranostics and the Inflammation and Infection Committees of the EANM, as well as the procedure standards committee of the SNMMI, and reflect recommendations for which the EANM and SNMMI cannot be held responsible. The recommendations should be taken into the context of good practice of nuclear medicine and do not substitute for national and international legal or regulatory provisions.
Collapse
Affiliation(s)
- E Lopci
- Nuclear Medicine Unit, IRCCS - Humanitas Research Hospital, Via Manzoni 56, 20089, Rozzano, Milano, Italy.
| | - R J Hicks
- The Department of Medicine, St Vincent's Medical School, the University of Melbourne, Melbourne, Australia
| | - A Dimitrakopoulou-Strauss
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69210, Heidelberg, Germany
| | - L Dercle
- Department of Radiology, New York Presbyterian, Columbia University Irving Medical Center, New York, NY, USA
| | - A Iravani
- Department of Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - R D Seban
- Department of Nuclear Medicine and Endocrine Oncology, Institut Curie, 92210, Saint-Cloud, France
- Laboratoire d'Imagerie Translationnelle en Oncologie, Inserm, Institut Curie, 91401, Orsay, France
| | - C Sachpekidis
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69210, Heidelberg, Germany
| | - O Humbert
- Department of Nuclear Medicine, Centre Antoine-Lacassagne, Université Côte d'Azur, Nice, France
- TIRO-UMR E 4320, Université Côte d'Azur, Nice, France
| | - O Gheysens
- Department of Nuclear Medicine, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - A W J M Glaudemans
- Nuclear Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - W Weber
- Department of Nuclear Medicine, Klinikum Rechts Der Isar, Technical University Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - R L Wahl
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - A M Scott
- Department of Molecular Imaging and Therapy, Austin Health, Studley Rd, Heidelberg, Victoria, 3084, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia
- Faculty of Medicine, University of Melbourne, Melbourne, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - N Pandit-Taskar
- Nuclear Medicine Service, Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave., New York, NY, 10021, USA
| | - N Aide
- Nuclear Medicine Department, University Hospital, Caen, France
- INSERM ANTICIPE, Normandie University, Caen, France
| |
Collapse
|
27
|
Xu B, Peng Z, An Y, Yan G, Yao X, Guan L, Sun M. Identification of Energy Metabolism-Related Gene Signatures From scRNA-Seq Data to Predict the Prognosis of Liver Cancer Patients. Front Cell Dev Biol 2022; 10:858336. [PMID: 35602603 PMCID: PMC9114438 DOI: 10.3389/fcell.2022.858336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/18/2022] [Indexed: 11/13/2022] Open
Abstract
The increasingly common usage of single-cell sequencing in cancer research enables analysis of tumor development mechanisms from a wider range of perspectives. Metabolic disorders are closely associated with liver cancer development. In recent years, liver cancer has been evaluated from different perspectives and classified into different subtypes to improve targeted treatment strategies. Here, we performed an analysis of liver cancer from the perspective of energy metabolism based on single-cell sequencing data. Single-cell and bulk sequencing data of liver cancer patients were obtained from GEO and TCGA/ICGC databases, respectively. Using the Seurat R package and protocols such as consensus clustering analysis, genes associated with energy metabolism in liver cancer were identified and validated. An energy metabolism-related score (EM score) was established based on five identified genes. Finally, the sensitivity of patients in different scoring groups to different chemotherapeutic agents and immune checkpoint inhibitors was analyzed. Tumor cells from liver cancer patients were found to divide into nine clusters, with cluster 4 having the highest energy metabolism score. Based on the marker genes of this cluster and TCGA database data, the five most stable key genes (ADH4, AKR1B10, CEBPZOS, ENO1, and FOXN2) were identified as energy metabolism-related genes in liver cancer. In addition, drug sensitivity analysis showed that patients in the low EM score group were more sensitive to immune checkpoint inhibitors and chemotherapeutic agents AICAR, metformin, and methotrexate.
Collapse
Affiliation(s)
- Boyang Xu
- Department of Gastroenterology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ziqi Peng
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yue An
- Department of Endoscopy, The First Hospital of China Medical University, Shenyang, China
| | - Guanyu Yan
- Department of Gastroenterology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xue Yao
- Department of Surgical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Lin Guan
- Department of Gastroenterology, The First Affiliated Hospital of China Medical University, Shenyang, China
- *Correspondence: Lin Guan, ; Mingjun Sun,
| | - Mingjun Sun
- Department of Gastroenterology, The First Affiliated Hospital of China Medical University, Shenyang, China
- *Correspondence: Lin Guan, ; Mingjun Sun,
| |
Collapse
|
28
|
Léger MA, Routy B, Juneau D. FDG PET/CT for Evaluation of Immunotherapy Response in Lung Cancer Patients. Semin Nucl Med 2022; 52:707-719. [DOI: 10.1053/j.semnuclmed.2022.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 11/11/2022]
|
29
|
Hlongwa KN, Mokoala KMG, Matsena-Zingoni Z, Vorster M, Sathekge MM. The Use of 18F-FDG PET/CT Metabolic Parameters in Predicting Overall Survival in Patients Undergoing Restaging for Malignant Melanoma. Diagnostics (Basel) 2022; 12:diagnostics12030595. [PMID: 35328148 PMCID: PMC8947629 DOI: 10.3390/diagnostics12030595] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/11/2022] [Accepted: 02/19/2022] [Indexed: 12/04/2022] Open
Abstract
Malignant melanoma is one of the more aggressive cancers in the skin, with an increasing incidence every year. Melanoma has a better prognosis if diagnosed early and survival tends to decrease once the disease has metastasized. Positron emission tomography (PET) with 2-[18F]fluoro-2-deoxy-D-glucose (18F-FDG) has been used extensively over the past two decades in staging and assessing responses to therapy in patients with melanoma. Metabolic PET parameters have been demonstrated to be independent prognostic factors for progression-free survival (PFS) and overall survival (OS) in different malignancies, melanoma included. In our study, we evaluated the metabolic parameters of 18F-FDG PET/CT (flourodeoxyglucose positron emission tomography/computed tomography) in predicting the overall survival in patients with malignant melanoma who presented for restaging. Metabolic PET parameters (maximum standardized uptake value (SUVmax), metabolic tumor volume (MTV) and total lesion glycolysis (TLG)) of the primary tumor, as well as whole-body MTV and TLG of the metastatic disease, were measured. Survival curves for OS were constructed and mortality rates were determined using the different PET variables. Forty-nine patients who presented for a PET/CT restaging in melanoma were included in this study. We found that non-survivors had significantly higher median MTV (11.86 cm3 vs. 5.68 cm3; p-value = 0.022), TLG (3125 vs. 14; p-value = 0.0357), whole-body MTV (53.9 cm3 vs. 14.4 cm3; p-value = 0.0076) and whole-body TLG (963.4 vs. 114.6; p-value = 0.0056). This demonstrated that high MTV and TLG values of the primary tumor and whole-body TLG as quantified by 18F-FDG PET/CT were prognostic factors for overall survival. The findings may potentially guide clinicians in decision making and identifying patients with a poorer prognosis.
Collapse
Affiliation(s)
- Khanyisile N Hlongwa
- Department of Nuclear Medicine, University of Pretoria and Steve Biko Academic Hospital, Pretoria 0001, South Africa
| | - Kgomotso M G Mokoala
- Department of Nuclear Medicine, University of Pretoria and Steve Biko Academic Hospital, Pretoria 0001, South Africa
| | - Zvifadzo Matsena-Zingoni
- Division of Epidemiology and Biostatistics, School of Public Health, University of Witwatersrand, Johannesburg 2193, South Africa
| | - Mariza Vorster
- Department of Nuclear Medicine, University of Pretoria and Steve Biko Academic Hospital, Pretoria 0001, South Africa
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0001, South Africa
| | - Mike M Sathekge
- Department of Nuclear Medicine, University of Pretoria and Steve Biko Academic Hospital, Pretoria 0001, South Africa
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0001, South Africa
| |
Collapse
|
30
|
Stadler JC, Belloum Y, Deitert B, Sementsov M, Heidrich I, Gebhardt C, Keller L, Pantel K. Current and Future Clinical Applications of ctDNA in Immuno-Oncology. Cancer Res 2022; 82:349-358. [PMID: 34815256 PMCID: PMC9397642 DOI: 10.1158/0008-5472.can-21-1718] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 10/06/2021] [Accepted: 11/09/2021] [Indexed: 01/07/2023]
Abstract
Testing peripheral blood for circulating tumor DNA (ctDNA) offers a minimally invasive opportunity to diagnose, characterize, and monitor the disease in individual cancer patients. ctDNA can reflect the actual tumor burden and specific genomic state of disease and thus might serve as a prognostic and predictive biomarker for immune checkpoint inhibitor (ICI) therapy. Recent studies in various cancer entities (e.g., melanoma, non-small cell lung cancer, colon cancer, and urothelial cancer) have shown that sequential ctDNA analyses allow for the identification of responders to ICI therapy, with a significant lead time to imaging. ctDNA assessment may also help distinguish pseudoprogression under ICI therapy from real progression. Developing dynamic changes in ctDNA concentrations as a potential surrogate endpoint of clinical efficacy in patients undergoing adjuvant immunotherapy is ongoing. Besides overall ctDNA burden, further ctDNA characterization can help uncover tumor-specific determinants (e.g., tumor mutational burden and microsatellite instability) of responses or resistance to immunotherapy. In future studies, standardized ctDNA assessments need to be included in interventional clinical trials across cancer entities to demonstrate the clinical utility of ctDNA as a biomarker for personalized cancer immunotherapy.
Collapse
Affiliation(s)
- Julia-Christina Stadler
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yassine Belloum
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Benjamin Deitert
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mark Sementsov
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Isabel Heidrich
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoffer Gebhardt
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Laura Keller
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Corresponding Authors: Klaus Pantel, Institute for Tumor Biologie, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, Hamburg, Hamburg, 20246, Germany. E-mail: ; and Laura Keller, E-mail:
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Corresponding Authors: Klaus Pantel, Institute for Tumor Biologie, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, Hamburg, Hamburg, 20246, Germany. E-mail: ; and Laura Keller, E-mail:
| |
Collapse
|
31
|
Alipour R, Iravani A, Hicks RJ. PET Imaging of Melanoma. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00123-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
32
|
Schweighofer-Zwink G, Manafi-Farid R, Kölblinger P, Hehenwarter L, Harsini S, Pirich C, Beheshti M. Prognostic value of 2-[ 18F]FDG PET-CT in metastatic melanoma patients receiving immunotherapy. Eur J Radiol 2021; 146:110107. [PMID: 34922117 DOI: 10.1016/j.ejrad.2021.110107] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/10/2021] [Accepted: 12/08/2021] [Indexed: 12/23/2022]
Abstract
PURPOSE The 2-fluorodeoxyglucose positron emission tomography/computed tomography (2-[18F]FDG PET/CT) is used for the evaluation of response to immunotherapy in malignant melanoma. Here, we evaluated the prognostic value of various metabolic parameters in baseline and different time points after therapy. METHODS In this retrospective study, 51 metastatic melanoma patients, who had received immunotherapy, were included. Patients with baseline and two follow-up 2-[18F]FDG PET/CT studies (3 and 6 months after therapy) were selected. Multiple metabolic parameters and tumor-to-background ratios (TBRs) were extracted and correlated with OS. RESULTS The 3- and 5-year OS rates were 49% and 43.1%, respectively. On baseline 2-[18F]FDG PET/CT, only standardized uptake value corrected for lean body mass (SULmax and SULpeak), as well as most of the TBRs were predictive for 3- and 5-year OS rates. Metabolic tumor volume (MTV), total lesion glycolysis (TLG), and most of the TBRs were predictive on both follow-up studies. Also, the changes in values of MTV, TLG and most of the TBRs from the baseline to the 3-month and 6- month follow-up studies were prognostic. On multivariate analysis, all of the most predictive parameters for OS were derived from the 3-month follow-up study. The ratio of TBRmean to the mediastinum was the best factor (cutoff value of 2.15, sensitivity of 88.5% and specificity of 68.0% for 3-year survival). CONCLUSION Metabolic parameters derived from 2-[18F]FDG PET/CT are valuable tools for the prediction of 3- and 5-year OS rates in metastatic melanoma patients undergoing immunotherapy. The 3-month follow-up 2-[18F]FDG PET/CT is of particular importance in this regard.
Collapse
Affiliation(s)
- Gregor Schweighofer-Zwink
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Reyhaneh Manafi-Farid
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical sciences, 1411713135 Tehran, Iran
| | - Peter Kölblinger
- Department of Dermatology, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Lukas Hehenwarter
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Sara Harsini
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical sciences, 1411713135 Tehran, Iran; Association of Nuclear Medicine and Molecular Imaging (ANMMI), Universal Scientific Education and Research Network (USERN), 1419733151 Tehran, Iran
| | - Christian Pirich
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Mohsen Beheshti
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria.
| |
Collapse
|
33
|
Hou J, Yang Y, Chen N, Chen D, Hu S. Prognostic Value of Volume-Based Parameters Measured by SSTR PET/CT in Neuroendocrine Tumors: A Systematic Review and Meta-Analysis. Front Med (Lausanne) 2021; 8:771912. [PMID: 34901087 PMCID: PMC8662524 DOI: 10.3389/fmed.2021.771912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/19/2021] [Indexed: 12/16/2022] Open
Abstract
Purpose: A meta-analysis was conducted to investigate the value of the volume parameters based on somatostatin receptor (SSTR)-positron emission tomography (PET) in predicting the prognosis in patients with neuroendocrine tumors (NETs). Material: PUBMED, EMBASE, Cochrane library, and Web of Knowledge were searched from January 1990 to May 2021 for studies evaluating prognostic value of volume-based parameters of SSTR PET/CT in NETs. The terms used were "volume," "positron emission tomography," "neuroendocrine tumors," and "somatostatin receptor." Pooled hazard ratio (HR) values were calculated to assess the correlations between volumetric parameters, including total tumor volume (TTV) and total-lesion SSTR expression (TL-SSTR), with progression-free survival (PFS) and overall survival (OS). Heterogeneity and subgroup analysis were performed. Funnel plots, Begg's and Egger's test were used to assess possible underlying publication bias. Results: Eight eligible studies involving 593 patients were included in the meta-analysis. In TTV, the pooled HRs of its prognostic value of PFS and OS were 2.24 (95% CI: 1.73-2.89; P < 0.00001) and 3.54 (95% CI, 1.77-7.09; P = 0.0004), respectively. In TL-SSTR, the pooled HR of the predictive value was 1.61 (95% CI, 0.48-5.44, P = 0.44) for PFS. Conclusion: High TTV was associated with a worse prognosis for PFS and OS in with patients NETs. The TTV of SSTR PET is a potential objective prognosis predictor.
Collapse
Affiliation(s)
- Jiale Hou
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Yi Yang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Na Chen
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Dengming Chen
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Shuo Hu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Biological Nanotechnology, Changsha, China.,National Clinical Research Center for Geriatric Disorders (XIANGYA), Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
34
|
Annovazzi A, Ferraresi V, Rea S, Russillo M, Renna D, Carpano S, Sciuto R. Prognostic value of total metabolic tumour volume and therapy-response assessment by [ 18F]FDG PET/CT in patients with metastatic melanoma treated with BRAF/MEK inhibitors. Eur Radiol 2021; 32:3398-3407. [PMID: 34779873 DOI: 10.1007/s00330-021-08355-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/02/2021] [Accepted: 09/24/2021] [Indexed: 12/31/2022]
Abstract
OBJECTIVES Target therapy with BRAF/MEK inhibitors in metastatic melanoma is characterised by a high response rate; however, acquired resistance to treatment develops in many cases. We aimed to investigate if baseline total metabolic tumour volume (TMTV) and therapy-response assessment by [18F]FDG PET/CT have a prognostic role on progression-free survival (PFS) and overall survival (OS) in patients with metastatic melanoma receiving BRAF ± MEK inhibitors. METHODS Fifty-seven patients who performed an [18F]FDG PET/CT at baseline and on treatment were retrospectively evaluated. A Cox proportional-hazard model was used to examine associations between OS and PFS with baseline clinical/PET parameters as well as for PET response. RESULTS According to EORTC criteria, 34 patients were classified as responders (partial/complete metabolic response [PMR/CMR]) and 23 as non-responders (progressive/stable metabolic disease [PMD/SMD]). Baseline characteristics associated with a shorter PFS were more than two metastatic organ sites and TMTV > 56 cm3; the latter was the only independent feature at multivariate analysis. Patients achieving a CMR were associated with a prolonged PFS compared with those with PMR (median PFS 42.9 vs 8.8 months; p = 0.009). Disease progression occurred in new-onset disease sites in 87.5% of CMR, 7.1% of PMR and 34.8% of PMD/SMD (p < 0.001). High baseline TMTV and lack of treatment response were independent prognostic factors for OS, stratifying patients in three different prognostic classes (median OS 6.7, 18.3 and 102.2 months, respectively). CONCLUSIONS Baseline TMTV and metabolic response may be useful prognostic indicators for PFS and OS in patients with advanced melanoma treated with BRAF/MEK inhibitors. KEY POINTS • In a retrospective cohort of 57 metastatic melanoma patients treated with BRAF/MEK inhibitors, a TMTV > 56 cm3 at baseline [18F]FDG PET/CT was significantly correlated with a shorter PFS and OS. • The combined use of baseline TMTV along with PET response during treatment allowed for the identification of three groups of patients with very different median OS.
Collapse
Affiliation(s)
- Alessio Annovazzi
- Nuclear Medicine Unit, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144, Rome, Italy.
| | - Virginia Ferraresi
- First Division of Medical Oncology, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
- Sarcomas and Rare Tumors Unit, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | - Sandra Rea
- Nuclear Medicine Unit, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144, Rome, Italy
| | - Michelangelo Russillo
- First Division of Medical Oncology, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | - Davide Renna
- First Division of Medical Oncology, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | - Silvia Carpano
- Second Division of Medical Oncology, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | - Rosa Sciuto
- Nuclear Medicine Unit, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144, Rome, Italy
| |
Collapse
|
35
|
Lopci E. Immunotherapy Monitoring with Immune Checkpoint Inhibitors Based on [ 18F]FDG PET/CT in Metastatic Melanomas and Lung Cancer. J Clin Med 2021; 10:jcm10215160. [PMID: 34768681 PMCID: PMC8584484 DOI: 10.3390/jcm10215160] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 12/15/2022] Open
Abstract
Immunotherapy with checkpoint inhibitors has prompted a major change not only in cancer treatment but also in medical imaging. In parallel with the implementation of new drugs modulating the immune system, new response criteria have been developed, aiming to overcome clinical drawbacks related to the new, unusual, patterns of response characterizing both solid tumors and lymphoma during the course of immunotherapy. The acknowledgement of pseudo-progression, hyper-progression, immune-dissociated response and so forth, has become mandatory for all imagers dealing with this clinical scenario. A long list of acronyms, i.e., irRC, iRECIST, irRECIST, imRECIST, PECRIT, PERCIMT, imPERCIST, iPERCIST, depicts the enormous effort made by radiology and nuclear medicine physicians in the last decade to optimize imaging parameters for better prediction of clinical benefit in immunotherapy regimens. Quite frequently, a combination of clinical-laboratory data with imaging findings has been tested, proving the ability to stratify patients into various risk groups. The next steps necessarily require a large scale validation of the most robust criteria, as well as the clinical implementation of immune-targeting tracers for immuno-PET or the exploitation of radiomics and artificial intelligence as complementary tools during the course of immunotherapy administration. For the present review article, a summary of PET/CT role for immunotherapy monitoring will be provided. By scrolling into various cancer types and applied response criteria, the reader will obtain necessary information for better understanding the potentials and limitations of the modality in the clinical setting.
Collapse
Affiliation(s)
- Egesta Lopci
- Nuclear Medicine Unit, IRCCS-Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, MI, Italy
| |
Collapse
|
36
|
Liberini V, Rubatto M, Mimmo R, Passera R, Ceci F, Fava P, Tonella L, Polverari G, Lesca A, Bellò M, Arena V, Ribero S, Quaglino P, Deandreis D. Predictive Value of Baseline [18F]FDG PET/CT for Response to Systemic Therapy in Patients with Advanced Melanoma. J Clin Med 2021; 10:jcm10214994. [PMID: 34768517 PMCID: PMC8584809 DOI: 10.3390/jcm10214994] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 12/24/2022] Open
Abstract
Background/Aim: To evaluate the association between baseline [18F]FDG-PET/CT tumor burden parameters and disease progression rate after first-line target therapy or immunotherapy in advanced melanoma patients. Materials and Methods: Forty four melanoma patients, who underwent [18F]FDG-PET/CT before first-line target therapy (28/44) or immunotherapy (16/44), were retrospectively analyzed. Whole-body and per-district metabolic tumor volume (MTV) and total lesion glycolysis (TLG) were calculated. Therapy response was assessed according to RECIST 1.1 on CT scan at 3 (early) and 12 (late) months. PET parameters were compared using the Mann–Whitney test. Optimal cut-offs for predicting progression were defined using the ROC curve. PFS and OS were studied using Kaplan–Meier analysis. Results: Median (IQR) MTVwb and TLGwb were 13.1 mL and 72.4, respectively. Non-responder patients were 38/44, 26/28 and 12/16 at early evaluation, and 33/44, 21/28 and 12/16 at late evaluation in the whole-cohort, target, and immunotherapy subgroup, respectively. At late evaluation, MTVbone and TLGbone were higher in non-responders compared to responder patients (all p < 0.037) in the whole-cohort and target subgroup and MTVwb and TLGwb (all p < 0.022) in target subgroup. No significant differences were found for the immunotherapy subgroup. No metabolic parameters were able to predict PFS. Controversially, MTVlfn, TLGlfn, MTVsoft + lfn, TLGsoft + lfn, MTVwb and TLGwb were significantly associated (all p < 0.05) with OS in both the whole-cohort and target therapy subgroup. Conclusions: Higher values of whole-body and bone metabolic parameters were correlated with poorer outcome, while higher values of whole-body, lymph node and soft tissue metabolic parameters were correlated with OS.
Collapse
Affiliation(s)
- Virginia Liberini
- Department of Medical Science, Division of Nuclear Medicine, University of Turin, 10126 Torino, Italy; (R.P.); (G.P.); (A.L.); (M.B.); (D.D.)
- Nuclear Medicine Department, S. Croce e Carle Hospital, 12100 Cuneo, Italy
- Correspondence:
| | - Marco Rubatto
- Department of Medical Sciences, Section of Dermatology, University of Turin, C.so Dogliotti, 10126 Torino, Italy; (M.R.); (P.F.); (L.T.); (S.R.); (P.Q.)
| | - Riccardo Mimmo
- Department of Medical Science, University of Turin, 10126 Torino, Italy;
| | - Roberto Passera
- Department of Medical Science, Division of Nuclear Medicine, University of Turin, 10126 Torino, Italy; (R.P.); (G.P.); (A.L.); (M.B.); (D.D.)
| | - Francesco Ceci
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy;
| | - Paolo Fava
- Department of Medical Sciences, Section of Dermatology, University of Turin, C.so Dogliotti, 10126 Torino, Italy; (M.R.); (P.F.); (L.T.); (S.R.); (P.Q.)
| | - Luca Tonella
- Department of Medical Sciences, Section of Dermatology, University of Turin, C.so Dogliotti, 10126 Torino, Italy; (M.R.); (P.F.); (L.T.); (S.R.); (P.Q.)
| | - Giulia Polverari
- Department of Medical Science, Division of Nuclear Medicine, University of Turin, 10126 Torino, Italy; (R.P.); (G.P.); (A.L.); (M.B.); (D.D.)
- PET Center, Affidea IRMET, 10135 Torino, Italy;
| | - Adriana Lesca
- Department of Medical Science, Division of Nuclear Medicine, University of Turin, 10126 Torino, Italy; (R.P.); (G.P.); (A.L.); (M.B.); (D.D.)
| | - Marilena Bellò
- Department of Medical Science, Division of Nuclear Medicine, University of Turin, 10126 Torino, Italy; (R.P.); (G.P.); (A.L.); (M.B.); (D.D.)
| | | | - Simone Ribero
- Department of Medical Sciences, Section of Dermatology, University of Turin, C.so Dogliotti, 10126 Torino, Italy; (M.R.); (P.F.); (L.T.); (S.R.); (P.Q.)
| | - Pietro Quaglino
- Department of Medical Sciences, Section of Dermatology, University of Turin, C.so Dogliotti, 10126 Torino, Italy; (M.R.); (P.F.); (L.T.); (S.R.); (P.Q.)
| | - Désirée Deandreis
- Department of Medical Science, Division of Nuclear Medicine, University of Turin, 10126 Torino, Italy; (R.P.); (G.P.); (A.L.); (M.B.); (D.D.)
| |
Collapse
|
37
|
Dall'Olio FG, Marabelle A, Caramella C, Garcia C, Aldea M, Chaput N, Robert C, Besse B. Tumour burden and efficacy of immune-checkpoint inhibitors. Nat Rev Clin Oncol 2021; 19:75-90. [PMID: 34642484 DOI: 10.1038/s41571-021-00564-3] [Citation(s) in RCA: 132] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2021] [Indexed: 01/07/2023]
Abstract
Accumulating evidence suggests that a high tumour burden has a negative effect on anticancer immunity. The concept of tumour burden, simply defined as the total amount of cancer in the body, in contrast to molecular tumour burden, is often poorly understood by the wider medical community; nonetheless, a possible role exists in defining the optimal treatment strategy for many patients. Historically, tumour burden has been assessed using imaging. In particular, CT scans have been used to evaluate both the number and size of metastases as well as the number of organs involved. These methods are now often complemented by metabolic tumour burden, measured using the more recently developed 2-deoxy-2-[18F]-fluoro-D-glucose (FDG)-PET/CT. Serum-based biomarkers, such as lactate dehydrogenase, can also reflect tumour burden and are often also correlated with a poor response to immune-checkpoint inhibitors. Other circulating markers (such as circulating free tumour DNA and/or circulating tumour cells) are also attracting research interest as surrogate markers of tumour burden. In this Review, we summarize evidence supporting the utility of tumour burden as a biomarker to guide the use of immune-checkpoint inhibitors. We also describe data and provide perspective on the various tools used for tumour burden assessment, with a particular emphasis on future therapeutic strategies that might address the issue of inferior outcomes among patients with cancer with a high tumour burden.
Collapse
Affiliation(s)
- Filippo G Dall'Olio
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France.,Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Specialized, Experimental and Diagnostic Medicine, University of Bologna, Bologna, Italy
| | - Aurélien Marabelle
- Drug Development Department, Gustave Roussy, Villejuif, France.,Faculty of Medicine, University Paris-Saclay, Kremlin Bicêtre, France.,Institut national de la santé et de la recherche médicale (INSERM), Gustave Roussy, Villejuif, France
| | - Caroline Caramella
- Department of Radiology, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Camilo Garcia
- Department of Nuclear Medicine and Endocrine Oncology, Institut Gustave Roussy and University Paris-Saclay, Villejuif, France
| | - Mihaela Aldea
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - Nathalie Chaput
- Laboratory of Immunomonitoring in Oncology, Gustave Roussy, Villejuif, France.,Faculty of Pharmacy, University Paris-Saclay, Chatenay-Malabry, France
| | - Caroline Robert
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France.,Faculty of Medicine, University Paris-Saclay, Kremlin Bicêtre, France.,Institut national de la santé et de la recherche médicale (INSERM), Gustave Roussy, Villejuif, France
| | - Benjamin Besse
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France. .,Faculty of Medicine, University Paris-Saclay, Kremlin Bicêtre, France.
| |
Collapse
|
38
|
Nakamoto R, Zaba LC, Liang T, Reddy SA, Davidzon G, Aparici CM, Nguyen J, Moradi F, Iagaru A, Franc BL. Prognostic Value of Bone Marrow Metabolism on Pretreatment 18F-FDG PET/CT in Patients with Metastatic Melanoma Treated with Anti-PD-1 Therapy. J Nucl Med 2021; 62:1380-1383. [PMID: 33547210 DOI: 10.2967/jnumed.120.254482] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 01/14/2021] [Indexed: 12/12/2022] Open
Abstract
Our purpose was to investigate the prognostic value of 18F-FDG PET/CT parameters in melanoma patients before beginning therapy with antibodies to the programmed cell death 1 receptor (anti-PD-1). Methods: Imaging parameters including SUVmax, metabolic tumor volume, and the ratio of bone marrow to liver SUVmean (BLR) were measured from baseline PET/CT in 92 patients before the start of anti-PD-1 therapy. The association with survival and imaging parameters combined with clinical factors was evaluated. Clinical and laboratory data were compared between the high-BLR group (>median) and the low-BLR group (≤median). Results: Multivariate analyses demonstrated that BLR was an independent prognostic factor for progression-free and overall survival (P = 0.017 and P = 0.011, respectively). The high-BLR group had higher white blood cell counts and neutrophil counts and a higher level of C-reactive protein than the low-BLR group (P < 0.05). Conclusion: Patients with a high BLR were associated with poor progression-free and overall survival, potentially explained by evidence of systemic inflammation known to be associated with immunosuppression.
Collapse
Affiliation(s)
- Ryusuke Nakamoto
- Department of Radiology, Stanford University, Stanford, California;
| | - Lisa C Zaba
- Department of Dermatology, Stanford University, Stanford, California; and
| | - Tie Liang
- Department of Radiology, Stanford University, Stanford, California
| | | | - Guido Davidzon
- Department of Radiology, Stanford University, Stanford, California
| | | | - Judy Nguyen
- Department of Radiology, Stanford University, Stanford, California
| | - Farshad Moradi
- Department of Radiology, Stanford University, Stanford, California
| | - Andrei Iagaru
- Department of Radiology, Stanford University, Stanford, California
| | | |
Collapse
|
39
|
Flaus A, Habouzit V, De Leiris N, Vuillez JP, Leccia MT, Perrot JL, Prevot N, Cachin F. FDG PET biomarkers for prediction of survival in metastatic melanoma prior to anti-PD1 immunotherapy. Sci Rep 2021; 11:18795. [PMID: 34552135 PMCID: PMC8458464 DOI: 10.1038/s41598-021-98310-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/31/2021] [Indexed: 12/22/2022] Open
Abstract
Our aim was to analyse whether biomarkers extracted from baseline 18F-FDG PET before anti-PD1 treatment contribute to prognostic survival information for early risk stratification in metastatic melanoma. Fifty-six patients, without prior systemic treatment, BRAF wild type, explored using 18F-FDG PET were included retrospectively. Our primary endpoint was overall survival (OS). Total metabolic tumoral volume (MTV) and forty-one IBSI compliant parameters were extracted from PET. Parameters associated with outcome were evaluated by a cox regression model and when significant helped build a prognostic score. Median follow-up was 22.1 months and 21 patients died. Total MTV and long zone emphasis (LZE) correlated with shorter OS and served to define three risk categories for the prognostic score. For low, intermediate and high risk groups, survival rates were respectively 91.1% (IC 95 80–1), 56.1% (IC 95 37.1–85) and 19% (IC 95 0.06–60.2) and hazard ratios were respectively 0.11 (IC 95 0.025–0.46), P = 0.0028, 1.2 (IC 95 0.48–2.8), P = 0.74 and 5.9 (IC 95 2.5–14), P < 0.0001. To conclude, a prognostic score based on total MTV and LZE separated metastatic melanoma patients in 3 categories with dramatically different outcomes. Innovative therapies should be tested in the group with the lowest prognosis score for future clinical trials.
Collapse
Affiliation(s)
- A Flaus
- Nuclear Medecine Department, Saint-Etienne University Hospital, University of Saint-Etienne, Saint-Etienne, France. .,Nuclear Medicine Department, East Group Hospital, Hospices Civils de Lyon, Lyon, France. .,Service de Medecine Nucléaire, Hôpital Nord, CHU de Saint-Etienne, 42 055, Saint-Etienne, Cedex 2, France.
| | - V Habouzit
- Nuclear Medecine Department, Saint-Etienne University Hospital, University of Saint-Etienne, Saint-Etienne, France
| | - N De Leiris
- Nuclear Medecine Department, CHU Grenoble Alpes, University Grenoble Alpes, Grenoble, France.,Laboratoire Radiopharmaceutiques Biocliniques, University Grenoble Alpes, INSERM, CHU Grenoble Alpes, 38000, Grenoble, France
| | - J P Vuillez
- Nuclear Medecine Department, CHU Grenoble Alpes, University Grenoble Alpes, Grenoble, France.,Laboratoire Radiopharmaceutiques Biocliniques, University Grenoble Alpes, INSERM, CHU Grenoble Alpes, 38000, Grenoble, France
| | - M T Leccia
- Dermatology Department, CHU Grenoble Alpes, University Grenoble Alpes, Grenoble, France
| | - J L Perrot
- Dermatology Department, Saint-Etienne University Hospital, University of Saint-Etienne, Saint-Etienne, France
| | - N Prevot
- Nuclear Medecine Department, Saint-Etienne University Hospital, University of Saint-Etienne, Saint-Etienne, France
| | - F Cachin
- Nuclear Medicine Department, Jean Perrin Cancer Center of Clermont-Ferrand, Clermont-Ferrand, France
| |
Collapse
|
40
|
Leung D, Bonacorsi S, Smith RA, Weber W, Hayes W. Molecular Imaging and the PD-L1 Pathway: From Bench to Clinic. Front Oncol 2021; 11:698425. [PMID: 34497758 PMCID: PMC8420047 DOI: 10.3389/fonc.2021.698425] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/22/2021] [Indexed: 01/24/2023] Open
Abstract
Programmed death-1 (PD-1) and programmed death ligand 1 (PD-L1) inhibitors target the important molecular interplay between PD-1 and PD-L1, a key pathway contributing to immune evasion in the tumor microenvironment (TME). Long-term clinical benefit has been observed in patients receiving PD-(L)1 inhibitors, alone and in combination with other treatments, across multiple tumor types. PD-L1 expression has been associated with response to immune checkpoint inhibitors, and treatment strategies are often guided by immunohistochemistry-based diagnostic tests assessing expression of PD-L1. However, challenges related to the implementation, interpretation, and clinical utility of PD-L1 diagnostic tests have led to an increasing number of preclinical and clinical studies exploring interrogation of the TME by real-time imaging of PD-(L)1 expression by positron emission tomography (PET). PET imaging utilizes radiolabeled molecules to non-invasively assess PD-(L)1 expression spatially and temporally. Several PD-(L)1 PET tracers have been tested in preclinical and clinical studies, with clinical trials in progress to assess their use in a number of cancer types. This review will showcase the development of PD-(L)1 PET tracers from preclinical studies through to clinical use, and will explore the opportunities in drug development and possible future clinical implementation.
Collapse
Affiliation(s)
- David Leung
- Translational Medicine, Bristol Myers Squibb, Princeton, NJ, United States
| | - Samuel Bonacorsi
- Translational Medicine, Bristol Myers Squibb, Princeton, NJ, United States
| | - Ralph Adam Smith
- Translational Medicine, Bristol Myers Squibb, Princeton, NJ, United States
| | - Wolfgang Weber
- Technische Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Wendy Hayes
- Translational Medicine, Bristol Myers Squibb, Princeton, NJ, United States
| |
Collapse
|
41
|
Fares J, Ulasov I, Timashev P, Lesniak MS. Emerging principles of brain immunology and immune checkpoint blockade in brain metastases. Brain 2021; 144:1046-1066. [PMID: 33893488 PMCID: PMC8105040 DOI: 10.1093/brain/awab012] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 12/12/2022] Open
Abstract
Brain metastases are the most common type of brain tumours, harbouring an immune microenvironment that can in principle be targeted via immunotherapy. Elucidating some of the immunological intricacies of brain metastases has opened a therapeutic window to explore the potential of immune checkpoint inhibitors in this globally lethal disease. Multiple lines of evidence suggest that tumour cells hijack the immune regulatory mechanisms in the brain for the benefit of their own survival and progression. Nonetheless, the role of the immune checkpoint in the complex interplays between cancers cells and T cells and in conferring resistance to therapy remains under investigation. Meanwhile, early phase trials with immune checkpoint inhibitors have reported clinical benefit in patients with brain metastases from melanoma and non-small cell lung cancer. In this review, we explore the workings of the immune system in the brain, the immunology of brain metastases, and the current status of immune checkpoint inhibitors in the treatment of brain metastases.
Collapse
Affiliation(s)
- Jawad Fares
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ilya Ulasov
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Maciej S Lesniak
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
42
|
Monaco L, Gemelli M, Gotuzzo I, Bauckneht M, Crivellaro C, Genova C, Cortinovis D, Zullo L, Ammoni LC, Bernasconi DP, Rossi G, Morbelli S, Guerra L. Metabolic Parameters as Biomarkers of Response to Immunotherapy and Prognosis in Non-Small Cell Lung Cancer (NSCLC): A Real World Experience. Cancers (Basel) 2021; 13:cancers13071634. [PMID: 33915801 PMCID: PMC8037395 DOI: 10.3390/cancers13071634] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/26/2021] [Accepted: 03/29/2021] [Indexed: 12/22/2022] Open
Abstract
Immune-checkpoint inhibitors (ICIs) have been proven to have great efficacy in non-small cell lung cancer (NSCLC) as single agents or in combination therapy, being capable to induce deep and durable remission. However, severe adverse events may occur and about 40% of patients do not benefit from the treatment. Predictive factors of response to ICIs are needed in order to customize treatment. The aim of this study is to evaluate the correlation between quantitative positron emission tomography (PET) parameters defined before starting ICI therapy and responses to treatment and patient outcome. We retrospectively analyzed 92 NSCLC patients treated with nivolumab, pembrolizumab or atezolizumab. Basal PET/computed tomography (CT) scan parameters (whole-body metabolic tumor volume-wMTV, total lesion glycolysis-wTLG, higher standardized uptake volume maximum and mean-SUVmax and SUVmean) were calculated for each patient and correlated with outcomes. Patients who achieved disease control (complete response + partial response + stable disease) had significantly lower MTV median values than patients who had not (progressive disease) (77 vs. 160.2, p = 0.039). Furthermore, patients with MTV and TLG values lower than the median values had improved OS compared to patients with higher MTV and TLG (p = 0.03 and 0.05, respectively). No relation was found between the other parameters and outcome. In conclusion, baseline metabolic tumor burden, measured with MTV, might be an independent predictor of treatment response to ICI and a prognostic biomarker in NSCLC patients.
Collapse
Affiliation(s)
- Lavinia Monaco
- School of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy; (L.M.); (L.G.)
| | - Maria Gemelli
- Medical Oncology, ASST Monza, San Gerardo Hospital, 20900 Monza, Italy; (M.G.); (D.C.)
| | - Irene Gotuzzo
- School of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy; (L.M.); (L.G.)
- Correspondence:
| | - Matteo Bauckneht
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.B.); (S.M.)
- Nuclear Medicine Unit, Department of Health Sciences, University of Genoa, 16132 Genoa, Italy
| | - Cinzia Crivellaro
- Nuclear Medicine, ASST Monza San Gerardo Hospital, 20900 Monza, Italy;
| | - Carlo Genova
- UOC Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
- Dipartimento di Medicina Interna e Specialità Mediche (DiMI), Facoltà di Medicina e Chirurgia, Università degli Studi di Genova, 16132 Genova, Italy
| | - Diego Cortinovis
- Medical Oncology, ASST Monza, San Gerardo Hospital, 20900 Monza, Italy; (M.G.); (D.C.)
| | - Lodovica Zullo
- UOC Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | | | - Davide Paolo Bernasconi
- Bicocca Biostatistics Bioinformatics and Bioimaging Center—B4, School of Medicine and Surgery, University Milano Bicocca, 20128 Milano, Italy;
| | - Giovanni Rossi
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy;
- UO Oncologia Medica, Ospedale Padre Antero Micone, 16153 Genova, Italy
| | - Silvia Morbelli
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.B.); (S.M.)
- Nuclear Medicine Unit, Department of Health Sciences, University of Genoa, 16132 Genoa, Italy
| | - Luca Guerra
- School of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy; (L.M.); (L.G.)
- Nuclear Medicine, ASST Monza San Gerardo Hospital, 20900 Monza, Italy;
| |
Collapse
|
43
|
Hindié E. Metastatic melanoma: can FDG-PET predict success of anti-PD-1 therapy and help determine when it can be discontinued? Eur J Nucl Med Mol Imaging 2021; 47:2227-2232. [PMID: 32322914 DOI: 10.1007/s00259-020-04826-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Elif Hindié
- Department of Nuclear Medicine, Bordeaux University and Hospitals, Bordeaux, France. .,Hôpital Haut-Lévêque, Avenue Magellan, 33604, Pessac, France.
| |
Collapse
|
44
|
Costa LB, Queiroz MA, Barbosa FG, Nunes RF, Zaniboni EC, Ruiz MM, Jardim D, Gomes Marin JF, Cerri GG, Buchpiguel CA. Reassessing Patterns of Response to Immunotherapy with PET: From Morphology to Metabolism. Radiographics 2020; 41:120-143. [PMID: 33275541 DOI: 10.1148/rg.2021200093] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cancer demands precise evaluation and accurate and timely assessment of response to treatment. Imaging must be performed early during therapy to allow adjustments to the course of treatment. For decades, cross-sectional imaging provided these answers, showing responses to the treatment through changes in tumor size. However, with the emergence of immune checkpoint inhibitors, complex immune response patterns were revealed that have quickly highlighted the limitations of this approach. Patterns of response beyond tumor size have been recognized and include cystic degeneration, necrosis, hemorrhage, and cavitation. Furthermore, new unique patterns of response have surfaced, like pseudoprogression and hyperprogression, while other patterns were shown to be deceptive, such as unconfirmed progressive disease. This evolution led to new therapeutic evaluation criteria adapted specifically for immunotherapy. Moreover, inflammatory adverse effects of the immune checkpoint blockade were identified, many of which were life threatening and requiring prompt intervention. Given complex concepts like tumor microenvironment and novel therapeutic modalities in the era of personalized medicine, increasingly sophisticated imaging techniques are required to address the intricate patterns of behavior of different neoplasms. Fluorine 18-fluorodeoxyglucose PET/CT has rapidly emerged as one such technique that spans both molecular biology and immunology. This imaging technique is potentially capable of identifying and tracking prognostic biomarkers owing to its combined use of anatomic and metabolic imaging, which enables it to characterize biologic processes in vivo. This tailored approach may provide whole-body quantification of the metabolic burden of disease, providing enhanced prediction of treatment response and improved detection of adverse events. ©RSNA, 2020.
Collapse
Affiliation(s)
- Larissa B Costa
- From the Departments of Radiology (L.B.C., M.A.Q., F.G.B., R.F.N., E.C.Z., M.M.R., J.F.G.M., G.G.C., C.A.B.) and Oncology (D.J.), Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, 01308-060 São Paulo, SP, Brazil; and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (M.A.Q., J.F.G.M., G.G.C., C.A.B.)
| | - Marcelo A Queiroz
- From the Departments of Radiology (L.B.C., M.A.Q., F.G.B., R.F.N., E.C.Z., M.M.R., J.F.G.M., G.G.C., C.A.B.) and Oncology (D.J.), Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, 01308-060 São Paulo, SP, Brazil; and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (M.A.Q., J.F.G.M., G.G.C., C.A.B.)
| | - Felipe G Barbosa
- From the Departments of Radiology (L.B.C., M.A.Q., F.G.B., R.F.N., E.C.Z., M.M.R., J.F.G.M., G.G.C., C.A.B.) and Oncology (D.J.), Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, 01308-060 São Paulo, SP, Brazil; and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (M.A.Q., J.F.G.M., G.G.C., C.A.B.)
| | - Rafael F Nunes
- From the Departments of Radiology (L.B.C., M.A.Q., F.G.B., R.F.N., E.C.Z., M.M.R., J.F.G.M., G.G.C., C.A.B.) and Oncology (D.J.), Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, 01308-060 São Paulo, SP, Brazil; and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (M.A.Q., J.F.G.M., G.G.C., C.A.B.)
| | - Elaine C Zaniboni
- From the Departments of Radiology (L.B.C., M.A.Q., F.G.B., R.F.N., E.C.Z., M.M.R., J.F.G.M., G.G.C., C.A.B.) and Oncology (D.J.), Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, 01308-060 São Paulo, SP, Brazil; and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (M.A.Q., J.F.G.M., G.G.C., C.A.B.)
| | - Mariana Mazo Ruiz
- From the Departments of Radiology (L.B.C., M.A.Q., F.G.B., R.F.N., E.C.Z., M.M.R., J.F.G.M., G.G.C., C.A.B.) and Oncology (D.J.), Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, 01308-060 São Paulo, SP, Brazil; and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (M.A.Q., J.F.G.M., G.G.C., C.A.B.)
| | - Denis Jardim
- From the Departments of Radiology (L.B.C., M.A.Q., F.G.B., R.F.N., E.C.Z., M.M.R., J.F.G.M., G.G.C., C.A.B.) and Oncology (D.J.), Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, 01308-060 São Paulo, SP, Brazil; and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (M.A.Q., J.F.G.M., G.G.C., C.A.B.)
| | - Jose Flavio Gomes Marin
- From the Departments of Radiology (L.B.C., M.A.Q., F.G.B., R.F.N., E.C.Z., M.M.R., J.F.G.M., G.G.C., C.A.B.) and Oncology (D.J.), Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, 01308-060 São Paulo, SP, Brazil; and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (M.A.Q., J.F.G.M., G.G.C., C.A.B.)
| | - Giovanni G Cerri
- From the Departments of Radiology (L.B.C., M.A.Q., F.G.B., R.F.N., E.C.Z., M.M.R., J.F.G.M., G.G.C., C.A.B.) and Oncology (D.J.), Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, 01308-060 São Paulo, SP, Brazil; and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (M.A.Q., J.F.G.M., G.G.C., C.A.B.)
| | - Carlos A Buchpiguel
- From the Departments of Radiology (L.B.C., M.A.Q., F.G.B., R.F.N., E.C.Z., M.M.R., J.F.G.M., G.G.C., C.A.B.) and Oncology (D.J.), Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, 01308-060 São Paulo, SP, Brazil; and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (M.A.Q., J.F.G.M., G.G.C., C.A.B.)
| |
Collapse
|
45
|
Cho SY, Huff DT, Jeraj R, Albertini MR. FDG PET/CT for Assessment of Immune Therapy: Opportunities and Understanding Pitfalls. Semin Nucl Med 2020; 50:518-531. [PMID: 33059821 PMCID: PMC8201415 DOI: 10.1053/j.semnuclmed.2020.06.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Immune checkpoint blockade has demonstrated the ability to modulate the immune system to produce durable responses in a wide range of cancers and has significantly impacted the standard of care. However, many cancer patients still do not respond to immune checkpoint blockade or have a limited duration of antitumor responses. Moreover, immune-related adverse events caused by immune checkpoint blockade can be severe and debilitating for some patients, limiting continuation of therapy and resulting in severe autoimmune conditions. Standard-of-care conventional anatomic imaging modalities and tumor response criteria have limitations to adequately assess tumor responses, especially early in the course of therapy, for risk-adapted clinical management to inform care of patients treated with immunotherapy. Molecular imaging with position emission tomography (PET) provides a noninvasive functional biomarker of tumor response, and of immune activation, for patients on immune-based therapies to help address these needs. 18F-FDG (FDG) PET/CT is readily available clinically and a number of studies have evaluated the use of this agent for assessment of prognosis, treatment response and immune activation for patients treated with immune checkpoint blockade. In this review paper, we discuss the current oncologic applications and imaging needs of cancer immunotherapy, recent studies applying FDG PET/CT for tumor response assessment, and evaluation of immune-related adverse events for improving clinical management. We largely focus on metastatic melanoma; however, we generalize where applicable to immunotherapy in other tumor types. We also briefly discuss PET imaging and quantitation as well as emerging non-FDG PET imaging radiotracers for cancer immunotherapy imaging.
Collapse
Affiliation(s)
- Steve Y Cho
- University of Wisconsin Carbone Cancer Center, Madison, WI; Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI.
| | - Daniel T Huff
- University of Wisconsin Carbone Cancer Center, Madison, WI; Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Robert Jeraj
- University of Wisconsin Carbone Cancer Center, Madison, WI; Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI; Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Mark R Albertini
- University of Wisconsin Carbone Cancer Center, Madison, WI; Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI; Medical Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
| |
Collapse
|
46
|
Lang D, Wahl G, Poier N, Graf S, Kiesl D, Lamprecht B, Gabriel M. Impact of PET/CT for Assessing Response to Immunotherapy-A Clinical Perspective. J Clin Med 2020; 9:jcm9113483. [PMID: 33126715 PMCID: PMC7694130 DOI: 10.3390/jcm9113483] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/18/2020] [Accepted: 10/22/2020] [Indexed: 12/25/2022] Open
Abstract
Cancer immunotherapy using immune-checkpoint inhibitors (ICI) has revolutionized the therapeutic landscape of various malignancies like non-small-cell lung cancer or melanoma. Pre-therapy response prediction and assessment during ICI treatment is challenging due to the lack of reliable biomarkers and the possibility of atypical radiological response patterns. Positron emission tomography/computed tomography (PET/CT) enables the visualization and quantification of metabolic lesion activity additional to conventional CT imaging. Various biomarkers derived from PET/CT have been reported as predictors for response to ICI and may aid to overcome the challenges clinicians currently face in the management of ICI-treated patients. In this narrative review, experts in nuclear medicine, thoracic oncology, dermatooncology, hemato- and internal oncology, urological and head/neck tumors performed literature reviews in their respective field and a joint discussion on the use of PET/CT in the context of ICI treatment. The aims were to give a clinical overview on present standards and evidence, to identify current challenges and fields of research and to enable an outlook to future developments and their possible implications. Multiple promising studies concerning ICI response assessment or prediction using biomarkers derived from PET/CT alone or as composite biomarkers have been identified for various malignancies and disease stages. Of interest, additional major incentives in the field may evolve from novel tracers specifically targeting immune-checkpoint molecules which could allow not only response assessment and prognosis, but also visualization of histological tumor cell properties like programmed death-ligand (PD-L1) expression in vivo. Despite the broad range of existing literature on PET/CT-derived biomarkers in ICI therapy, implications for daily clinical practice remain elusive. High-quality prospective data are urgently warranted to determine whether patients benefit from the application of PET/CT in terms of prognosis. At the moment, the lack of such evidence as well as the absence of standardized imaging methods and biomarkers still precludes PET/CT imaging to be included in the relevant clinical practice guidelines.
Collapse
Affiliation(s)
- David Lang
- Department of Pulmonology Johannes Kepler University Hospital Linz, Krankenhausstrasse 9, 4020 Linz, Austria; (D.L.); (B.L.)
| | - Gerald Wahl
- Department of Dermatology and Venerology, Johannes Kepler University Hospital Linz, Krankenhausstrasse 9, 4020 Linz, Austria;
| | - Nikolaus Poier
- Department of Otorhinolaryngology, Head and Neck Surgery, Johannes Kepler University Hospital Linz, Krankenhausstrasse 9, 4020 Linz, Austria;
| | - Sebastian Graf
- Department of Urology and Andrology, Johannes Kepler University Hospital Linz Krankenhausstrasse 9, 4020 Linz, Austria;
| | - David Kiesl
- University Clinic of Hematology and Internal Oncology Johannes Kepler University Hospital Linz, Krankenhausstrasse 9, 4020 Linz, Austria;
| | - Bernd Lamprecht
- Department of Pulmonology Johannes Kepler University Hospital Linz, Krankenhausstrasse 9, 4020 Linz, Austria; (D.L.); (B.L.)
| | - Michael Gabriel
- Institute of Nuclear Medicine and Endocrinology, Johannes Kepler University Hospital Linz, Krankenhausstrasse 9, 4020 Linz, Austria
- Correspondence: ; Tel.: +43-5-7680-83-6166; Fax: +43-5-7680-83-6165
| |
Collapse
|
47
|
van Sluis J, de Heer EC, Boellaard M, Jalving M, Brouwers AH, Boellaard R. Clinically feasible semi-automatic workflows for measuring metabolically active tumour volume in metastatic melanoma. Eur J Nucl Med Mol Imaging 2020; 48:1498-1510. [PMID: 33099667 PMCID: PMC8113298 DOI: 10.1007/s00259-020-05068-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 10/12/2020] [Indexed: 12/19/2022]
Abstract
Purpose Metabolically active tumour volume (MATV) is a potential quantitative positron emission tomography (PET) imaging biomarker in melanoma. Accumulating data indicate that low MATV may predict increased chance of response to immunotherapy and overall survival. However, metastatic melanoma can present with numerous (small) tumour lesions, making manual tumour segmentation time-consuming. The aim of this study was to evaluate multiple semi-automatic segmentation workflows to determine reliability and reproducibility of MATV measurements in patients with metastatic melanoma. Methods An existing cohort of 64 adult patients with histologically proven metastatic melanoma was used in this study. 18F-FDG PET/CT diagnostic baseline images were acquired using a European Association of Nuclear Medicine (EANM) Research Limited–accredited Siemens Biograph mCT PET/CT system (Siemens Healthineers, Knoxville, USA). PET data were analysed using manual, gradient-based segmentation and five different semi-automatic methods: three direct PET image–derived delineations (41MAX, A50P and SUV40) and two based on a majority-vote approach (MV2 and MV3), without and with (suffix ‘+’) manual lesion addition. Correlation between the different segmentation methods and their respective associations with overall survival was assessed. Results Correlation between the MATVs derived by the manual segmentation and semi-automated tumour segmentations ranged from R2 = 0.41 for A50P to R2 = 0.85 for SUV40+ and MV2+, respectively. Manual MATV segmentation did not differ significantly from the semi-automatic methods SUV40 (∆MATV mean ± SD 0.08 ± 0.60 mL, P = 0.303), SUV40+ (∆MATV − 0.10 ± 0.51 mL, P = 0.126), MV2+ (∆MATV − 0.09 ± 0.62 mL, P = 0.252) and MV3+ (∆MATV − 0.03 ± 0.55 mL, P = 0.615). Log-rank tests showed statistically significant overall survival differences between above and below median MATV patients for all segmentation methods with areas under the ROC curves of 0.806 for manual segmentation and between 0.756 [41MAX] and 0.807 [MV3+] for semi-automatic segmentations. Conclusions Simple and fast semi-automated FDG PET segmentation workflows yield accurate and reproducible MATV measurements that correlate well with manual segmentation in metastatic melanoma. The most readily applicable and user-friendly SUV40 method allows feasible MATV measurement in prospective multicentre studies required for validation of this potential PET imaging biomarker for clinical use. Electronic supplementary material The online version of this article (10.1007/s00259-020-05068-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Joyce van Sluis
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Ellen C de Heer
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Mayke Boellaard
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Mathilde Jalving
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Adrienne H Brouwers
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands. .,Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Cancer Center Amsterdam UMC, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
48
|
Wang W, Gao Z, Wang L, Li J, Yu J, Han S, Meng X. Application and Prospects of Molecular Imaging in Immunotherapy. Cancer Manag Res 2020; 12:9389-9403. [PMID: 33061627 PMCID: PMC7533904 DOI: 10.2147/cmar.s269773] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 09/10/2020] [Indexed: 12/18/2022] Open
Abstract
Recently, immunotherapies that target the interactions of programmed cell death 1 (PD-1) with its major ligands, programmed death ligand 1 (PD-L1) and programmed death ligand 2 (PD-L2), have achieved significant success. To date, several immune checkpoint inhibitors targeting the PD-1/PD-L1 pathway have been developed to treat melanoma, non-small cell lung cancer, head and neck cancer, renal cell carcinoma, and urothelial carcinoma. Despite promising outcomes with immunotherapy, there are many limitations to several current immune biomarkers for predicting immune benefits and to traditional imaging for evaluating the efficacy and prognosis of immunotherapy and monitoring adverse reactions. In this review, we recommend a novel imaging method, molecular imaging. This paper reviews the application and prospects of molecular imaging in the context of current immunotherapies in regard to the following aspects: 1) detecting the expression of PD-1/PD-L1; 2) evaluating the efficacy of immunotherapy; 3) assessing patient prognosis with immunotherapy; 4) monitoring the toxicity of immunotherapy; and 5) other targets imaging.
Collapse
Affiliation(s)
- Weiqing Wang
- School of Clinical Medicine, Weifang Medical University, Weifang, Shandong 261053, People's Republic of China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong, People's Republic of China
| | - Zhenhua Gao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong, People's Republic of China
| | - Lu Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong, People's Republic of China
| | - Jianing Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong, People's Republic of China
| | - Jinming Yu
- School of Clinical Medicine, Weifang Medical University, Weifang, Shandong 261053, People's Republic of China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong, People's Republic of China
| | - Shumei Han
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong, People's Republic of China
| | - Xue Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong, People's Republic of China
| |
Collapse
|
49
|
Nabet BY, Esfahani MS, Moding EJ, Hamilton EG, Chabon JJ, Rizvi H, Steen CB, Chaudhuri AA, Liu CL, Hui AB, Almanza D, Stehr H, Gojenola L, Bonilla RF, Jin MC, Jeon YJ, Tseng D, Liu C, Merghoub T, Neal JW, Wakelee HA, Padda SK, Ramchandran KJ, Das M, Plodkowski AJ, Yoo C, Chen EL, Ko RB, Newman AM, Hellmann MD, Alizadeh AA, Diehn M. Noninvasive Early Identification of Therapeutic Benefit from Immune Checkpoint Inhibition. Cell 2020; 183:363-376.e13. [PMID: 33007267 PMCID: PMC7572899 DOI: 10.1016/j.cell.2020.09.001] [Citation(s) in RCA: 213] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 06/18/2020] [Accepted: 08/28/2020] [Indexed: 12/30/2022]
Abstract
Although treatment of non-small cell lung cancer (NSCLC) with immune checkpoint inhibitors (ICIs) can produce remarkably durable responses, most patients develop early disease progression. Furthermore, initial response assessment by conventional imaging is often unable to identify which patients will achieve durable clinical benefit (DCB). Here, we demonstrate that pre-treatment circulating tumor DNA (ctDNA) and peripheral CD8 T cell levels are independently associated with DCB. We further show that ctDNA dynamics after a single infusion can aid in identification of patients who will achieve DCB. Integrating these determinants, we developed and validated an entirely noninvasive multiparameter assay (DIREct-On, Durable Immunotherapy Response Estimation by immune profiling and ctDNA-On-treatment) that robustly predicts which patients will achieve DCB with higher accuracy than any individual feature. Taken together, these results demonstrate that integrated ctDNA and circulating immune cell profiling can provide accurate, noninvasive, and early forecasting of ultimate outcomes for NSCLC patients receiving ICIs.
Collapse
Affiliation(s)
- Barzin Y Nabet
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA; Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Mohammad S Esfahani
- Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Everett J Moding
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA; Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Emily G Hamilton
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA; Program in Cancer Biology, Stanford University, Stanford, CA, USA
| | - Jacob J Chabon
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Hira Rizvi
- Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chloe B Steen
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Aadel A Chaudhuri
- Department of Radiation Oncology, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Chih Long Liu
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA; Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Angela B Hui
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA; Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Diego Almanza
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA; Program in Cancer Biology, Stanford University, Stanford, CA, USA
| | - Henning Stehr
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Linda Gojenola
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Rene F Bonilla
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Michael C Jin
- Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Young-Jun Jeon
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA; Stanford Cancer Institute, Stanford University, Stanford, CA, USA; Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Diane Tseng
- Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Cailian Liu
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Taha Merghoub
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell School of Medicine, New York, NY, USA; Parker Institute for Cancer Immunotherapy at MSK, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joel W Neal
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA; Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Heather A Wakelee
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA; Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Sukhmani K Padda
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA; Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Kavitha J Ramchandran
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA; Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Millie Das
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA; Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, CA, USA; Department of Medicine, VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Andrew J Plodkowski
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christopher Yoo
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Emily L Chen
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Ryan B Ko
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Aaron M Newman
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA; Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Matthew D Hellmann
- Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell School of Medicine, New York, NY, USA; Parker Institute for Cancer Immunotherapy at MSK, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Ash A Alizadeh
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA; Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA.
| | - Maximilian Diehn
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA; Stanford Cancer Institute, Stanford University, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
50
|
Functional Imaging of Immunotherapy: Response Criteria, Imaging Characteristics, and Novel Immunoimaging of Advanced Malignancies. CURRENT RADIOLOGY REPORTS 2020. [DOI: 10.1007/s40134-020-00369-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|