1
|
Peeters E, van Genugten EAJ, Heskamp S, de Vries IJM, van Herpen C, Koenen HJPM, Kneilling M, van der Post RS, van Dop WA, Westdorp H, Aarntzen E. Exploring molecular imaging to investigate immune checkpoint inhibitor-related toxicity. J Immunother Cancer 2025; 13:e011009. [PMID: 40341021 PMCID: PMC12060888 DOI: 10.1136/jitc-2024-011009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 04/18/2025] [Indexed: 05/10/2025] Open
Abstract
Immune checkpoint inhibitors (ICI) boost the endogenous anticancer immunity, evoking long-lasting anticancer responses in a subset of patients with solid tumors. Simultaneously, ICI are also associated with serious toxicities, impacting treatment duration and the quality of life. The proposed processes underlying ICI-related toxicity include T-cell activation and recruitment to non-tumor tissues, involvement of other immune cells and fibroblasts and the host' microbiome composition. However, the exact mechanisms of these processes remain incompletely understood, hindering clinicians' ability to predict and identify ICI-related toxicity in the early stages of treatment. Molecular imaging may play a role as a non-invasive biomarker, providing a tool to study ICI-related toxicity. This review discusses the applications of molecular imaging to answer questions regarding the mechanisms, detection, and prediction of ICI-related toxicity. Potential targets and the current state of development of suitable imaging techniques are discussed.
Collapse
Affiliation(s)
- Eva Peeters
- Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Sandra Heskamp
- Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carla van Herpen
- Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hans J P M Koenen
- Laboratory of Medical Immunology, Radboud University Medical Center, Nijmegen, Gelderland, The Netherlands
| | - Manfred Kneilling
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University of Tübingen, Tubingen, Baden-Württemberg, Germany
- Department of Dermatology, University of Tübingen, Tubingen, Baden-Württemberg, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls Universität Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Rachel S van der Post
- Department of Pathology, Radboud University Medical Center, Nijmegen, Gelderland, The Netherlands
| | - Willemijn A van Dop
- Department of Gastroenterology, Radboud University Medical Center, Nijmegen, Gelderland, The Netherlands
| | - Harm Westdorp
- Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Erik Aarntzen
- Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Nuclear Medicine, Eberhard Karls Universität Tübingen, Tübingen, Baden-Württemberg, Germany
- Department of Nuclear Medicine and Molecular Imaging, University Medical Centre Groningen, Groningen, Groningen, The Netherlands
| |
Collapse
|
2
|
Nakahara T, Fujimoto S, Jinzaki M. Molecular imaging of cardiovascular disease: Current status and future perspective. J Cardiol 2025; 85:386-398. [PMID: 39922562 DOI: 10.1016/j.jjcc.2025.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/15/2025] [Accepted: 01/28/2025] [Indexed: 02/10/2025]
Abstract
Advancements in knowledge of cardiovascular disease, pharmacology, and chemistry have led to the development of newer radiopharmaceuticals and targets for new and more suitable molecules. Molecular imaging encompasses multiple imaging techniques for identifying the characteristics of key components involved in disease. Despite its limitations in spatial resolution, the affinity for key molecules compensates for disadvantages in diagnosing diseases and elucidating their pathophysiology. This review introduce established molecular tracers involved in clinical practice and emerging tracers already applied in clinical studies, classifying the key component in A: artery, specifically those vulnerable plaque (A-I) inflammatory cells [18F-FDG]; A-II) lipid/fatty acid; A-III) hypoxia; A-IV) angiogenesis; A-V) protease [18F/68Ga-FAPI]; A-VI) thrombus/hemorrhage; A-VII) apoptosis and A-VIII) microcalcification [18F-NaF]) and B: myocardium, including myocardial ischemia, infarction and myocardiopathy (B-I) myocardial ischemia; B-II) myocardial infarction (myocardial damage and fibrosis); B-III) myocarditis and endocarditis; B-IV) sarcoidosis; B-V) amyloidosis; B-VI) metabolism; B-VII) innervation imaging). In addition to cardiovascular-specific tracers tested in animal models, many radiotracers may have been developed in other areas, such as oncology imaging or neuroimaging. While this review does not cover all available tracers, some of them hold potential for future use assessing cardiovascular disease. Advances in molecular biology, pharmaceuticals, and imaging sciences will facilitate the identification of precise disease mechanisms, enabling precise diagnoses, better assessment of disease status, and enhanced therapeutic evaluation in this multi-modality era.
Collapse
Affiliation(s)
- Takehiro Nakahara
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan.
| | - Shinichiro Fujimoto
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masahiro Jinzaki
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
3
|
Luo Y, Yang Q, Qin X, Yu B, Jiang S, Liu Y. The Imaging Value of 18F-FAPI PET/CT in Sorafenib-Induced Cardiac Dysfunction in Patients with Hepatocellular Carcinoma: Compared with 18F-FDG PET/CT. Mol Imaging Biol 2025:10.1007/s11307-025-02005-4. [PMID: 40307657 DOI: 10.1007/s11307-025-02005-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 03/13/2025] [Accepted: 04/01/2025] [Indexed: 05/02/2025]
Abstract
PURPOSE Evaluation of 18F-FAPI PET/CT imaging in sorafenib-induced cardiac dysfunction in hepatocellular carcinoma (HCC) patients, and compared with 18F-FDG PET/CT. PROCEDURES This retrospective study enrolled 75 HCC patients treated with sorafenib at our institution from June 2021 to June 2023. All patients underwent 18F-FDG PET/CT six months after treatment, followed by 18F-FAPI PET/CT within the subsequent week. Patients were divided into cardiac dysfunction group and control group based on the definition of cancer therapy-related cardiac dysfunction (CTRCD). Myocardial uptake parameters on 18F-FDG and 18F-FAPI PET/CT were compared between the two groups. The primary endpoint was the occurrence of major adverse cardiac events (MACEs), and the secondary endpoint was all-cause mortality, with follow-up at 30, 90, and 180 days after the PET/CT examinations. RESULTS This study ultimately enrolled 47 patients, with the cardiac dysfunction group (n = 9) and control group (n = 38) demonstrating significant differences in myocardial 18F-FAPI high uptake, left ventricular (LV) 18F-FDG SUV, LV/liver 18F-FDG SUV, myocardial 18F-FAPI SUV, myocardial/aorta 18F-FAPI SUV, and myocardial/liver 18F-FAPI SUV. One year after treatment, the incidence of MACEs was slightly higher in the group with high 18F-FAPI myocardial uptake compared to the low uptake group (19.5% vs. 14.0%, log-rank p = 0.621), and the overall survival rate was lower in the high uptake group compared to the low uptake group (57.9% vs. 65.8%, log-rank p = 0.503). CONCLUSIONS The myocardial uptake parameters of 18F-FDG and 18F-FAPI PET/CT are helpful in evaluating sorafenib-induced cardiac dysfunction in HCC patients. The level of 18F-FAPI myocardial uptake has potential value in predicting post-treatment cardiotoxicity and overall survival prognosis in HCC patients.
Collapse
Affiliation(s)
- Yingqi Luo
- Department of Nuclear Medicine, The Second Affiliated Hospital, Guangzhou Medical University, No. 250 Changgang East Road, Haizhu District, Guangzhou City, Guangdong Province, China
| | - Qingqi Yang
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaowen Qin
- Department of Nuclear Medicine, The Second Affiliated Hospital, Guangzhou Medical University, No. 250 Changgang East Road, Haizhu District, Guangzhou City, Guangdong Province, China
| | - Boyang Yu
- School of Clinical Medicine, Guangzhou Medical University, Guangzhou, China
| | - Shengnan Jiang
- Department of Nuclear Medicine, The Second Affiliated Hospital, Guangzhou Medical University, No. 250 Changgang East Road, Haizhu District, Guangzhou City, Guangdong Province, China
| | - Ying Liu
- Department of Nuclear Medicine, The Second Affiliated Hospital, Guangzhou Medical University, No. 250 Changgang East Road, Haizhu District, Guangzhou City, Guangdong Province, China.
| |
Collapse
|
4
|
Filippi L, Perrone MA, Schillaci O. FAPI-Targeted Molecular Imaging: Transforming Insights into Post-Ischemic Myocardial Remodeling? Mol Diagn Ther 2025:10.1007/s40291-025-00778-6. [PMID: 40263181 DOI: 10.1007/s40291-025-00778-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2025] [Indexed: 04/24/2025]
Abstract
Post-ischemic myocardial remodeling significantly impacts clinical outcomes after acute myocardial infarction (MI), involving structural and functional changes such as ventricular dilation, infarct wall thinning, and fibrosis development. These processes, driven by inflammatory cascades, neurohormonal activation, and extracellular matrix remodeling, result in impaired cardiac output and an increased risk of heart failure. Imaging with fibroblast activation protein inhibitors (FAPI) has emerged as a promising non-invasive tool for assessing myocardial fibrosis via positron emission tomography (PET) or single-photon emission computed tomography (SPECT), targeting activated fibroblasts; the mediators of reparative and fibrotic processes. This innovative approach enables precise visualization and quantification of fibrosis dynamics, surpassing traditional imaging modalities. Preclinical studies using [68Ga]Ga-FAPI PET/computed tomography (CT) demonstrated the tracer's specificity for fibroblast activation and its peak uptake in the infarct border zone at day 6 post-MI. These findings, corroborated by histology and autoradiography, highlight its potential for tracking reparative fibrosis. Clinical translation of FAPI imaging was recently achieved with [68Ga]Ga-FAPI-46 PET/magnetic resonance imaging (MRI), showing persistent fibroblast activity beyond infarct zones and strong correlations with myocardial injury markers. Complementary research on [99mTc]Tc-HFAPi SPECT imaging in patients post-MI established its predictive value for left ventricular remodeling, emphasizing its cost-effectiveness and accessibility compared with PET. These advancements underscore FAPI-based imaging's potential to transform risk stratification and therapeutic guidance in post-MI care.
Collapse
Affiliation(s)
- Luca Filippi
- Department of Biomedicine and Prevention, University Tor Vergata, Via Montpellier 1, 0133, Rome, Italy.
| | - Marco Alfonso Perrone
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University Tor Vergata, Via Montpellier 1, 0133, Rome, Italy
| |
Collapse
|
5
|
Patel S, Dave K, Garcia MJ, Gongora CA, Travin MI, Zhang L. Multimodal Imaging of Immune Checkpoint Inhibitor Myocarditis. J Clin Med 2025; 14:2850. [PMID: 40283680 PMCID: PMC12028134 DOI: 10.3390/jcm14082850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have dramatically changed the landscape of cancer treatment and are increasingly used either as monotherapy or in combination with other ICIs, chemotherapy, and molecularly targeted agents. ICI myocarditis is a rare but potentially fatal irAE associated with the use of ICI characterized by T-cell mediated cardiomyocyte death. Diagnosing ICI myocarditis can be intricate as its atypical presentations. Multimodal imaging plays a crucial role in the diagnosis and risk stratification of ICI myocarditis. Current management strategies for ICI myocarditis include corticosteroids and immunosuppressants. Multidisciplinary collaboration is vital in these cases-combining oncology expertise with cardiology insights.
Collapse
Affiliation(s)
- Shreyans Patel
- Division of Cardiology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; (M.J.G.); (C.A.G.); (L.Z.)
| | - Kartikeya Dave
- Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA;
| | - Mario J. Garcia
- Division of Cardiology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; (M.J.G.); (C.A.G.); (L.Z.)
| | - Carlos A. Gongora
- Division of Cardiology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; (M.J.G.); (C.A.G.); (L.Z.)
| | - Mark I. Travin
- Division of Nuclear Medicine, Department of Radiology, Montefiore Medical Center and the Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA;
| | - Lili Zhang
- Division of Cardiology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; (M.J.G.); (C.A.G.); (L.Z.)
| |
Collapse
|
6
|
De Perna ML, Rigamonti E, Zannoni R, Espeli V, Moschovitis G. Immune Checkpoint Inhibitors and Cardiovascular Adverse Events. ESC Heart Fail 2025. [PMID: 40205958 DOI: 10.1002/ehf2.15281] [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: 10/27/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 04/11/2025] Open
Abstract
In the last years, we assisted to a tremendous increase in therapeutic options for the management of cancers, with immunotherapy at the forefront of this innovation. Immune checkpoint inhibitors (ICIs) have been developed to enhance the activity of the immune system against cancer cells (1) and the number of approvals for ICIs has rapidly increased. ICIs have also been associated with disinhibited cytotoxic T cells that damage healthy tissue in multiple organs, causing immune-related adverse events (AEs). Cardiovascular AEs (CVAe) are increasingly reported: myocarditis, Takotsubo syndrome, pericarditis and pericardial effusion, worsening of atherosclerosis, acute coronary syndromes, non-inflammatory heart failure, and ischaemic stroke. They are classified into five grades, based on presenting symptoms, level of cardiac biomarkers, and imaging. Even though myocarditis occurs more frequently than previously thought, clinically relevant myocarditis is a rare irAE compared to other irAE (0.5-1.2%). The clinical manifestations range from mild symptoms such as to chest pain, heart failure, and cardiogenic shock. The prognosis is severe, with mortality rates ranging from 25% to 50%. It is frequently associated with the concomitant use of combination of checkpoint inhibitors. The treatment strategies are tripartite: (i) holding ICI to prevent further toxicity, (ii) immunosuppression to alleviate inflammatory changes, and (iii) supportive therapy to address cardiac complications. Glucocorticoids represent the first-line treatment. In hemodynamically unstable patients, treatment with high-dose steroids should be initiated (intravenous methylprednisolone 1000 or 1250 mg oral methylprednisolone during 4 days). ICI-associated pericarditis can be accompanied by no/mild pericardial effusion up to cardiac tamponade. The treatment is made of nonsteroidal anti-inflammatory drugs and colchicine, corticosteroids if needed, and pericardiocentesis for the large effusions. ICIs could be continued for Grade 1 pericarditis, while temporary suspension of ICI is warranted for more severe cases. There is significant potential for accelerated atherosclerosis with ICIs as a long-term effect, but atherosclerosis-related CVAEs are not frequent, especially during treatment; increasing evidence associates ICIs with progression of atherosclerosis and increased atherosclerotic cardiovascular disease. ICIs can lead to arrhythmias: atrial fibrillation, supraventricular and ventricular tachycardias. Non-inflammatory heart failure syndrome have been observed in ICI-treated patients. Immune checkpoint inhibitors seem to be involved in the development of right ventricular dysfunction and pulmonary arterial hypertension. It is of the outmost importance to improve the collaboration among the different medical figures, such as cardiologists, oncologists, endocrinologists, and immunologists, both in clinical practice and in basic science research, to better recognize these adverse events, to understand their pathophysiological mechanisms, and to improve the overall survival and quality of life of the affected patients.
Collapse
Affiliation(s)
- Maria Luisa De Perna
- Istituto Cardiocentro Ticino, Department of Cardiology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Elia Rigamonti
- Istituto Cardiocentro Ticino, Department of Cardiology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Raffaele Zannoni
- Service de Médicine Interne générale, Département de médecine interne, Hopitaux Universitaires de Genève (HUG), rue Gabrielle-Perret-Gentil 4, Genève, Switzerland
| | - Vittoria Espeli
- Department of Oncology, Ente Ospedaliero Cantonale, Istituto Oncologico della Svizzera Italiana, Lugano, Switzerland
| | - Giorgio Moschovitis
- Istituto Cardiocentro Ticino, Department of Cardiology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| |
Collapse
|
7
|
McGale J, Khurana S, Howell H, Nakhla A, Roa T, Doshi P, Shirini D, Huang A, Duong P, Backhaus P, Liao M, Kaur H, Fontani AM, Hung I, Pandit-Taskar N, Haberkorn U, Gulati A, Naim A, Sinigaglia M, Bebawy M, Girard A, Seban RD, Dercle L. FAP-Targeted SPECT/CT and PET/CT Imaging for Breast Cancer Patients. Clin Nucl Med 2025; 50:e138-e145. [PMID: 39780367 DOI: 10.1097/rlu.0000000000005617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
ABSTRACT Breast cancer presents a significant global health challenge, necessitating continued innovation in diagnostic and therapeutic approaches. Recent advances have led to the identification of cancer-associated fibroblasts, which are highly prevalent in breast cancers and express fibroblast activation proteins (FAPs), as critical targets. FAP-specific radiotracers, when used with PET/CT and SPECT/CT, have significant potential for improving early breast cancer detection, staging, treatment response monitoring, and therapeutic intervention. This review provides insight into FAP-targeted molecular imaging, exploring advanced techniques for protein status assessment, development of early-phase targeted therapies, and other emerging applications. The advent of FAP-targeted imaging stands to significantly enhance personalized oncologic care, leading to improved breast cancer management and overall patient outcomes.
Collapse
Affiliation(s)
- Jeremy McGale
- From the Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Sakshi Khurana
- From the Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Harrison Howell
- From the Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Abanoub Nakhla
- Department of Surgery, Maimonides Medical Center, New York, NY
| | - Tina Roa
- From the Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Parth Doshi
- Department of Internal Medicine, Lewis Katz School of Medicine, Philadelphia, PA
| | - Dorsa Shirini
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alice Huang
- From the Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Phuong Duong
- From the Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Philipp Backhaus
- European Institute for Molecular Imaging, University of Münster, Münster, Germany and Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Matthew Liao
- From the Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Harleen Kaur
- From the Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | | | | | - Neeta Pandit-Taskar
- Department of Radiology, Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Uwe Haberkorn
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Amit Gulati
- Department of Internal Medicine, Maimonides Medical Center, New York, NY
| | - Asmâa Naim
- Université Mohammed VI des Sciences et de la Santé, Casablanca, Morocco
| | | | - Maria Bebawy
- Morristown Medical Center, OBGYN Department, Morristown, NJ
| | - Antoine Girard
- Department of Nuclear Medicine, CHU Amiens-Picardie, Amiens, France
| | - Romain-David Seban
- Department of Nuclear Medicine and Endocrine Oncology, Institut Curie, Saint-Cloud, France and Laboratory of Translational Imaging in Oncology, Paris Sciences et Lettres (PSL) Research University, Institut Curie, Orsay, France
| | - Laurent Dercle
- From the Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| |
Collapse
|
8
|
Crosio S, Treglia G, Imbimbo M, Froesch P, Grazioli Gauthier L, Arangalage D, Bergamaschi L, Györik SA, Viani GM, Caretta A, Leo LA, Pedrazzini G, Moschovitis G, Pavon AG. Multimodality Imaging and Immune-Related Adverse Events During Immune Checkpoint Inhibitors Treatment: Where Do We Stand? Echocardiography 2025; 42:e70115. [PMID: 40028736 DOI: 10.1111/echo.70115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/06/2025] [Accepted: 02/23/2025] [Indexed: 03/05/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy, significantly improving survival across various malignancies. However, these therapies are associated with various types of immune-related adverse events (irAEs), including cardiotoxicity, a spectrum of rare but potentially life-threatening complications impacting significantly morbidity and mortality. Cardiovascular imaging has become key in cardio-oncology, providing essential diagnostic tools for early detection and monitoring. This review synthesizes current evidence and underlines the pivotal role of early and tailored imaging strategies in managing ICI-induced cardiotoxicity. By bridging the knowledge gap, it aims to provide targetable insights to optimize the clinical management in patients undergoing immunotherapy.
Collapse
Affiliation(s)
- Stephanie Crosio
- Department of Cardiology, Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Giorgio Treglia
- Division of Nuclear Medicine, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università Della Svizzera Italiana, Lugano, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Martina Imbimbo
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Patrizia Froesch
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Lorenzo Grazioli Gauthier
- Department of Cardiology, Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Dimitri Arangalage
- Department of Cardiology, Bichat-Claude Bernard Hospital and Université Paris Cité, Paris, France
| | - Luca Bergamaschi
- Department of Cardiology, IRCCS Policlinico St. Orsola-Malpighi, Department of Experimental, Diagnostic and Specialty Medicine-DIMES, University of Bologna, Bologna, Italy
| | - Sándor A Györik
- Departement of Pneumology, Hospital of Bellinzona, Bellinzona, Switzerland
| | - Giacomo Maria Viani
- Department of Cardiology, Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Alessandro Caretta
- Department of Cardiology, Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Laura Anna Leo
- Department of Cardiology, Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Giovanni Pedrazzini
- Department of Cardiology, Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università Della Svizzera Italiana, Lugano, Switzerland
| | - Giorgio Moschovitis
- Department of Cardiology, Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Anna Giulia Pavon
- Department of Cardiology, Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università Della Svizzera Italiana, Lugano, Switzerland
| |
Collapse
|
9
|
Huang YV, Sun Y, Chou H, Wagner N, Vitale MR, Bayer AL, Xu B, Lee D, Lin Z, Branche C, Waliany S, Neal JW, Wakelee HA, Witteles RM, Nguyen PK, Graves EE, Berry GJ, Alcaide P, Wu SM, Zhu H. Novel Therapeutic Approach Targeting CXCR3 to Treat Immunotherapy Myocarditis. Circ Res 2025; 136:473-490. [PMID: 39931812 PMCID: PMC11867805 DOI: 10.1161/circresaha.124.325652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 01/12/2025] [Accepted: 01/16/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are successful in treating many cancers but may cause immune-related adverse events. ICI-mediated myocarditis has a high fatality rate with severe cardiovascular consequences. Targeted therapies for ICI myocarditis are currently limited. METHODS We used a genetic mouse model of PD1 deletion (MRL/Pdcd1-/-) along with a novel drug-treated ICI myocarditis mouse model to recapitulate the disease phenotype. We performed single-cell RNA-sequencing, single-cell T-cell receptor sequencing, and cellular indexing of transcriptomes and epitopes on immune cells isolated from MRL and MRL/Pdcd1-/- mice at serial time points. We assessed the impact of macrophage deletion in MRL/Pdcd1-/- mice, then inhibited CXCR3 (C-X-C motif chemokine receptor 3) in ICI-treated mice to assess the therapeutic effect on myocarditis phenotype. Furthermore, we delineated the functional and mechanistic effects of CXCR3 blockade on T-cell and macrophage interactions. We then correlated the results in human single-cell multiomics data from blood and heart biopsy data from patients with ICI myocarditis. RESULTS Single-cell multiomics demonstrated expansion of CXCL (C-X-C motif chemokine ligand) 9/10+CCR2+ macrophages and CXCR3hi (C-X-C motif chemokine receptor 3 high-expressing) CD8+ (cluster of differentiation) effector T lymphocytes in the hearts of MRL/Pdcd1-/- mice correlating with onset of myocarditis development. Both depletion of CXCL9/10+CCR2+ (C-C motif chemokine receptor) macrophages and CXCR3 blockade, respectively, led to decreased CXCR3hi CD8+ T-cell infiltration into the heart and significantly improved survival. Transwell migration assays demonstrated that the selective blockade of CXCR3 and its ligand, CXCL10, reduced CXCR3+CD8+ T-cell migration toward macrophages, implicating this interaction in T-cell cardiotropism toward cardiac macrophages. Furthermore, cardiomyocyte apoptosis was induced by CXCR3hi CD8+ T cells. Cardiac biopsies from patients with confirmed ICI myocarditis demonstrated infiltrating CXCR3+ T cells and CXCL9+/CXCL10+ macrophages. Both mouse cardiac immune cells and patient peripheral blood immune cells revealed expanded TCRs (T-cell receptors) correlating with CXCR3hi CD8+ T cells in ICI myocarditis samples. CONCLUSIONS These findings bring forth the CXCR3-CXCL9/10 axis as an attractive therapeutic target for ICI myocarditis treatment, and more broadly as a druggable pathway in cardiac inflammation.
Collapse
Affiliation(s)
- Yuhsin Vivian Huang
- Stanford Cardiovascular Institute, CA (Y.V.H., Y.S., H.C., N.W., M.R.V., B.X., Z.L., C.B., R.M.W., P.K.N., S.M.W., H.Z.)
| | - Yin Sun
- Stanford Cardiovascular Institute, CA (Y.V.H., Y.S., H.C., N.W., M.R.V., B.X., Z.L., C.B., R.M.W., P.K.N., S.M.W., H.Z.)
| | - Harrison Chou
- Stanford Cardiovascular Institute, CA (Y.V.H., Y.S., H.C., N.W., M.R.V., B.X., Z.L., C.B., R.M.W., P.K.N., S.M.W., H.Z.)
| | - Noah Wagner
- Stanford Cardiovascular Institute, CA (Y.V.H., Y.S., H.C., N.W., M.R.V., B.X., Z.L., C.B., R.M.W., P.K.N., S.M.W., H.Z.)
| | - Maria Rosaria Vitale
- Stanford Cardiovascular Institute, CA (Y.V.H., Y.S., H.C., N.W., M.R.V., B.X., Z.L., C.B., R.M.W., P.K.N., S.M.W., H.Z.)
| | | | - Bruce Xu
- Stanford Cardiovascular Institute, CA (Y.V.H., Y.S., H.C., N.W., M.R.V., B.X., Z.L., C.B., R.M.W., P.K.N., S.M.W., H.Z.)
| | - Daniel Lee
- F. Edward Hebert School of Medicine at Uniformed Services University, Bethesda, MD (D.L.)
| | - Zachary Lin
- Stanford Cardiovascular Institute, CA (Y.V.H., Y.S., H.C., N.W., M.R.V., B.X., Z.L., C.B., R.M.W., P.K.N., S.M.W., H.Z.)
| | - Corynn Branche
- Stanford Cardiovascular Institute, CA (Y.V.H., Y.S., H.C., N.W., M.R.V., B.X., Z.L., C.B., R.M.W., P.K.N., S.M.W., H.Z.)
| | - Sarah Waliany
- Division of Cardiovascular Medicine, Stanford, CA (S.W., R.M.W., P.K.N., S.M.W., H.Z.)
- Massachusetts General Hospital Cancer Center, Boston, MA (S.W.)
| | - Joel W. Neal
- Division of Oncology, Stanford, CA (J.W.N., H.A.W.)
- Stanford Cancer Institute, CA (J.W.N., H.A.W.)
| | - Heather A. Wakelee
- Division of Oncology, Stanford, CA (J.W.N., H.A.W.)
- Stanford Cancer Institute, CA (J.W.N., H.A.W.)
| | - Ronald M. Witteles
- Stanford Cardiovascular Institute, CA (Y.V.H., Y.S., H.C., N.W., M.R.V., B.X., Z.L., C.B., R.M.W., P.K.N., S.M.W., H.Z.)
- Division of Cardiovascular Medicine, Stanford, CA (S.W., R.M.W., P.K.N., S.M.W., H.Z.)
| | - Patricia K. Nguyen
- Stanford Cardiovascular Institute, CA (Y.V.H., Y.S., H.C., N.W., M.R.V., B.X., Z.L., C.B., R.M.W., P.K.N., S.M.W., H.Z.)
- Division of Cardiovascular Medicine, Stanford, CA (S.W., R.M.W., P.K.N., S.M.W., H.Z.)
| | | | | | - Pilar Alcaide
- Tufts University School of Medicine, Boston, MA (A.L.B., P.A.)
| | - Sean M. Wu
- Stanford Cardiovascular Institute, CA (Y.V.H., Y.S., H.C., N.W., M.R.V., B.X., Z.L., C.B., R.M.W., P.K.N., S.M.W., H.Z.)
- Division of Cardiovascular Medicine, Stanford, CA (S.W., R.M.W., P.K.N., S.M.W., H.Z.)
| | - Han Zhu
- Stanford Cardiovascular Institute, CA (Y.V.H., Y.S., H.C., N.W., M.R.V., B.X., Z.L., C.B., R.M.W., P.K.N., S.M.W., H.Z.)
- Division of Cardiovascular Medicine, Stanford, CA (S.W., R.M.W., P.K.N., S.M.W., H.Z.)
| |
Collapse
|
10
|
Panuccio G, Correale P, d'Apolito M, Mutti L, Giannicola R, Pirtoli L, Giordano A, Labate D, Macheda S, Carabetta N, Abdelwahed YS, Landmesser U, Tassone P, Tagliaferri P, De Rosa S, Torella D. Immuno-related cardio-vascular adverse events associated with immuno-oncological treatments: an under-estimated threat for cancer patients. Basic Res Cardiol 2025; 120:153-169. [PMID: 39225869 PMCID: PMC11790807 DOI: 10.1007/s00395-024-01077-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/16/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024]
Abstract
Immunotherapy represents an emergent and heterogeneous group of anticancer treatments harnessing the human immune-surveillance system, including immune-checkpoint inhibitor monoclonal antibodies (mAbs), Chimeric Antigen Receptor T Cells (CAR-T) therapy, cancer vaccines and lymphocyte activation gene-3 (LAG-3) therapy. While remarkably effective against several malignancies, these therapies, often in combination with other cancer treatments, have showed unforeseen toxicity, including cardiovascular complications. The occurrence of immuno-mediated adverse (irAEs) events has been progressively reported in the last 10 years. These irAEs present an extended range of severity, from self-limiting to life-threatening conditions. Although recent guidelines in CardioOncology have provided important evidence in managing cancer treatments, they often encompass general approaches. However, a specific focus is required due to the particular etiology, unique risk factors, and associated side effects of immunotherapy. This review aims to deepen the understanding of the prevalence and nature of cardiovascular issues in patients undergoing immunotherapy, offering insights into strategies for risk stratification and management.
Collapse
Affiliation(s)
- Giuseppe Panuccio
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité Berlin, 12200, Berlin, Germany.
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| | - Pierpaolo Correale
- Medical Oncology Unit, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
| | - Maria d'Apolito
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
- Medical Oncology Unit, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
| | - Luciano Mutti
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
- Department of Applied Sciences and Biotechnology, Università dell'Aquila, L'Aquila, Italy
| | - Rocco Giannicola
- Medical Oncology Unit, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
| | - Luigi Pirtoli
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
- Department of Medical Biotechnology, University of Siena, 53100, Siena, Italy
| | - Demetrio Labate
- Unit of Intensive Care Medicine and Anesthesia, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
| | - Sebastiano Macheda
- Unit of Intensive Care Medicine and Anesthesia, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
| | - Nicole Carabetta
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Youssef S Abdelwahed
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité Berlin, 12200, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), 10785, Berlin, Germany
| | - Ulf Landmesser
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité Berlin, 12200, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), 10785, Berlin, Germany
- Berlin Institute of Health (BIH), 10178, Berlin, Germany
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Salvatore De Rosa
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| |
Collapse
|
11
|
Cook GJR, Alberts IL, Wagner T, Fischer BM, Nazir MS, Lilburn D. The impact of long axial field of view (LAFOV) PET on oncologic imaging. Eur J Radiol 2025; 183:111873. [PMID: 39647272 PMCID: PMC11904125 DOI: 10.1016/j.ejrad.2024.111873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/10/2024]
Abstract
The development of long axial field of view (LAFOV) positron emission tomography coupled with computed tomography (PET/CT) scanners might be considered the biggest step forward in PET imaging since it became a mainstream clinical modality. Despite increased capital and maintenance costs and data storage requirements, the improvement in image quality, significantly faster acquisition times and lower radiopharmaceutical administered activities, allow a high quality and more efficient clinical service. This step change in technology overcomes some of the limitations of standard short axial field of view scanners. It allows simultaneous imaging of all body systems, and with the ability to obtain high temporal resolution data, it increases potential research applications, particularly in multisystem disease or for dosimetry measurements of novel radiopharmaceuticals. The improvements in sensitivity and signal-to-noise facilitates the use of tracers with long half-lives and low administered activity (e.g. [89Zr]-labelled monoclonal antibodies) or very short half-lives (e.g. [82Rb]), opening up applications that hitherto have been challenging. It is early in the evolution of LAFOV PET/CT and the advantages these systems offer have still to be fully realised in providing additional impact in clinical practice. In this article we describe the potential advantages of LAFOV PET technology and some of the clinical and research applications where it has been applied as well as some of the future developments that may enhance the modality further.
Collapse
Affiliation(s)
- Gary J R Cook
- King's College London & Guy's and St Thomas' PET Centre, School of Biomedical Engineering and Imaging Sciences, King's College, London SE1 7EH, UK.
| | - Ian L Alberts
- Molecular Imaging and Therapy, BC Cancer Agency, Vancouver, BC, Canada; Department of Radiology, University of British Columbia, Vancouver, BC, Canada.
| | - Thomas Wagner
- Department of Nuclear Medicine, Royal Free London NHS Trust, London NW3 1TX, UK.
| | - B Malene Fischer
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital-Rigshospitalet, 2100 Copenhagen, Denmark.
| | - Muhummad Sohaib Nazir
- Department of Cardiovascular Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK; Cardio-Oncology Centre of Excellence, Royal Brompton Hospital, Guy's and St Thomas' NHS Foundation Trust, London, UK.
| | - David Lilburn
- King's College London & Guy's and St Thomas' PET Centre, School of Biomedical Engineering and Imaging Sciences, King's College, London SE1 7EH, UK.
| |
Collapse
|
12
|
Lucinian YA, Martineau P, Abikhzer G, Harel F, Pelletier-Galarneau M. Novel tracers to assess myocardial inflammation with radionuclide imaging. J Nucl Cardiol 2024; 42:102012. [PMID: 39069249 DOI: 10.1016/j.nuclcard.2024.102012] [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: 02/16/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 07/30/2024]
Abstract
Myocardial inflammation plays a central role in the pathophysiology of various cardiac diseases. While FDG-PET is currently the primary method for molecular imaging of myocardial inflammation, its effectiveness is hindered by physiological myocardial uptake as well as its propensity for uptake by multiple disease-specific mechanisms. Novel radiotracers targeting diverse inflammatory immune cells and molecular pathways may provide unique insight through the visualization of underlying mechanisms central to the pathogenesis of inflammatory cardiac diseases, offering opportunities for increased understanding of immunocardiology. Moreover, the potentially enhanced specificity may lead to better quantification of disease activity, aiding in the guidance and monitoring of immunomodulatory therapy. This review aims to provide an update on advancements in non-FDG radiotracers for imaging myocardial inflammatory diseases, with a focus on cardiac sarcoidosis, myocarditis, and acute myocardial infarction.
Collapse
Affiliation(s)
| | | | - Gad Abikhzer
- Jewish General Hospital, Montreal, Quebec, Canada
| | | | | |
Collapse
|
13
|
Kiani M, Jokar S, Hassanzadeh L, Behnammanesh H, Bavi O, Beiki D, Assadi M. Recent Clinical Implications of FAPI: Imaging and Therapy. Clin Nucl Med 2024; 49:e538-e556. [PMID: 39025634 DOI: 10.1097/rlu.0000000000005348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
ABSTRACT The fibroblast activation protein (FAP) is a biomarker that is selectively overexpressed on cancer-associated fibroblasts (CAFs) in various types of tumoral tissues and some nonmalignant diseases, including fibrosis, arthritis, cardiovascular, and metabolic diseases. FAP plays a critical role in tumor microenvironment through facilitating proliferation, invasion, angiogenesis, immunosuppression, and drug resistance. Recent studies reveal that FAP might be regarded as a promising target for cancer diagnosis and treatment. FAP-targeted imaging modalities, especially PET, have shown high sensitivity and specificity in detecting FAP-expressing tumors. FAP-targeted imaging can potentially enhance tumor detection, staging, and monitoring of treatment response, and facilitate the development of personalized treatment strategies. This study provides a comprehensive view of FAP and its function in the pathophysiology of cancer and nonmalignant diseases. It also will discuss the characteristics of radiolabeled FAP inhibitors, particularly those based on small molecules, their recent clinical implications in imaging and therapy, and the associated clinical challenges with them. In addition, we present the results of imaging and biodistribution radiotracer 68 Ga-FAPI-46 in patients with nonmalignant diseases, including interstitial lung disease, primary biliary cirrhosis, and myocardial infarction, who were referred to our department. Our results show that cardiac FAP-targeted imaging can provide a novel potential biomarker for managing left ventricle remodeling. Moreover, this study has been organized and presented in a manner that offers a comprehensive overview of the current status and prospects of FAPI inhibitors in the diagnosis and treatment of diseases.
Collapse
Affiliation(s)
- Mahshid Kiani
- From the Department of Nuclear Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Safura Jokar
- From the Department of Nuclear Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Hassanzadeh
- Department of Nuclear Medicine, School of Medicine, Rajaie Cardiovascular, Medical & Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Omid Bavi
- Department of Mechanical Engineering, Shiraz University of Technology, Shiraz, Iran
| | - Davood Beiki
- Research Center for Nuclear Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Assadi
- The Persian Gulf Nuclear Medicine Research Center, Department of Molecular Imaging and Radionuclide Therapy, Bushehr Medical University Hospital, Bushehr University of Medical Sciences, Bushehr, Iran
| |
Collapse
|
14
|
Ali A, Lu Y, Khalaf S, Iliescu C, Koutroumpakis E, Yusuf SW, Siddiqui BA, Subudhi SK, Deswal A, Palaskas NL. Use of positron emission tomography for the diagnosis of immune-checkpoint inhibitor| myocarditis. J Nucl Cardiol 2024; 39:101909. [PMID: 39053574 DOI: 10.1016/j.nuclcard.2024.101909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 06/26/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Affiliation(s)
- Abdelrahman Ali
- Department of Cardiology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. https://twitter.com/@Abdelcards
| | - Yang Lu
- Department of Nuclear Medicine, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shaden Khalaf
- Department of Cardiology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cezar Iliescu
- Department of Cardiology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Efstratios Koutroumpakis
- Department of Cardiology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Syed Wamique Yusuf
- Department of Cardiology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bilal A Siddiqui
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sumit K Subudhi
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anita Deswal
- Department of Cardiology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. https://twitter.com/@anita_deswal
| | - Nicolas L Palaskas
- Department of Cardiology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
15
|
Telli T, Hosseini A, Settelmeier S, Kersting D, Kessler L, Weber WA, Rassaf T, Herrmann K, Varasteh Z. Imaging of Cardiac Fibrosis: How Far Have We Moved From Extracellular to Cellular? Semin Nucl Med 2024; 54:686-700. [PMID: 38493001 DOI: 10.1053/j.semnuclmed.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 02/29/2024] [Indexed: 03/18/2024]
Abstract
Cardiovascular disease is the leading cause of morbidity and mortality worldwide. Myocardial fibrosis plays an important role in adverse outcomes such as heart failure and arrhythmias. As the pathological response and degree of scarring, and therefore clinical presentation varies from patient to patient, early detection of fibrosis is crucial for identifying the appropriate treatment approach and forecasting the progression of a disease along with the likelihood of disease-related mortality. Current imaging modalities provides information about either decreased function or extracellular signs of fibrosis. Targeting activated fibroblasts represents a burgeoning approach that could offer insights prior to observable functional alterations, presenting a promising focus for potential anti-fibrotic therapeutic interventions at cellular level. In this article, we provide an overview of imaging cardiac fibrosis and discuss the role of different advanced imaging modalities with the focus on novel non-invasive imaging of activated fibroblasts.
Collapse
Affiliation(s)
- Tugce Telli
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Atefeh Hosseini
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Stephan Settelmeier
- Westgerman Heart- and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, Essen, Germany
| | - David Kersting
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Lukas Kessler
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany; Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Wolfgang A Weber
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Tienush Rassaf
- Westgerman Heart- and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, Essen, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Zohreh Varasteh
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany; Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University Munich, Munich, Germany.
| |
Collapse
|
16
|
Tong J, Vogiatzakis N, Andres MS, Senechal I, Badr A, Ramalingam S, Rosen SD, Lyon AR, Nazir MS. Complementary use of cardiac magnetic resonance and 18 F-FDG positron emission tomography imaging in suspected immune checkpoint inhibitor myocarditis. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:53. [PMID: 39175028 PMCID: PMC11340117 DOI: 10.1186/s40959-024-00250-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 07/19/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND Immune checkpoint inhibitor (ICI) myocarditis is an uncommon but potentially fatal complication of immunotherapy. Cardiac imaging is essential to make timely diagnoses as there are critical downstream implications for patients. OBJECTIVE To determine the agreement of cardiac magnetic resonance (CMR) and 18 F-fluorodeoxyglucose Positron Emission Tomography (FDG-PET) in patients with suspected ICI myocarditis. METHODS Patients with suspected ICI myocarditis, who underwent CMR and 18 F-FDG-PET imaging at a single cardio-oncology service from 2017 to 2023, were enrolled. CMR was performed according to recommended guidelines for assessment of myocarditis. 18 F-FDG-PET imaging was performed following 18 h carbohydrate-free fast. Imaging was analysed by independent reviewers to determine the presence or absence of ICI myocarditis. RESULTS Twelve patients (mean age 60 ± 15 years old, 7 [58%] male) underwent both CMR and 18 F-FDG-PET imaging. Three (25%) met the 2018 Lake Louise Criteria for CMR diagnosis of myocarditis; 4 (33%) had evidence of myocardial inflammation as determined by 18 F-FDG-PET. Amongst those with positive 18 F-FDG-PET, mean standard uptake value (SUV) was 3.5 ± 1.7. There was agreement between CMR and PET in 7 cases (CMR and PET positive (n = 1), CMR and PET negative (n = 6)) and discordance in 5 cases (CMR positive and PET negative (n = 2), CMR negative and PET positive (n = 3)). CONCLUSION Both CMR and PET provide complementary clinical information in diagnostic of ICI myocarditis. CMR informs on myocardial oedema, whilst 18 F-FDG-PET provides information on glucose metabolism reflecting monocyte and lymphocytic activity. Future studies should investigate the role of hybrid PET-CMR for the timely diagnosis of ICI myocarditis.
Collapse
Affiliation(s)
- Jieli Tong
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
- Department of Cardiology, Tan Tock Seng Hospital, Singapore, Singapore
| | - Nikolaos Vogiatzakis
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Maria Sol Andres
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Isabelle Senechal
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Ahmed Badr
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Sivatharshini Ramalingam
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Stuart D Rosen
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Alexander R Lyon
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Muhummad Sohaib Nazir
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK.
- School of Biomedical Engineering and Imaging Sciences, King's College London, Guy's and St Thomas' Hospital, London, UK.
| |
Collapse
|
17
|
Pozzessere C, Mazini B, Omoumi P, Jreige M, Noirez L, Digklia A, Fasquelle F, Sempoux C, Dromain C. Immune-Related Adverse Events Induced by Immune Checkpoint Inhibitors and CAR-T Cell Therapy: A Comprehensive Imaging-Based Review. Cancers (Basel) 2024; 16:2585. [PMID: 39061225 PMCID: PMC11274393 DOI: 10.3390/cancers16142585] [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: 05/07/2024] [Revised: 07/09/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Immunotherapy has revolutionized oncology care, improving patient outcomes in several cancers. However, these therapies are also associated with typical immune-related adverse events due to the enhanced inflammatory and immune response. These toxicities can arise at any time during treatment but are more frequent within the first few months. Any organ and tissue can be affected, ranging from mild to life-threatening. While some manifestations are common and more often mild, such as dermatitis and colitis, others are rarer and more severe, such as myocarditis. Management depends on the severity, with treatment being held for >grade 2 toxicities. Steroids are used in more severe cases, and immunosuppressive treatment may be considered for non-responsive toxicities, along with specific organ support. A multidisciplinary approach is mandatory for prompt identification and management. The diagnosis is primarily of exclusion. It often relies on imaging features, and, when possible, cytologic and/or pathological analyses are performed for confirmation. In case of clinical suspicion, imaging is required to assess the presence, extent, and features of abnormalities and to evoke and rule out differential diagnoses. This imaging-based review illustrates the diverse system-specific toxicities associated with immune checkpoint inhibitors and chimeric antigen receptor T-cells with a multidisciplinary perspective. Clinical characteristics, imaging features, cytological and histological patterns, as well as the management approach, are presented with insights into radiological tips to distinguish these toxicities from the most important differential diagnoses and mimickers-including tumor progression, pseudoprogression, inflammation, and infection-to guide imaging and clinical specialists in the pathway of diagnosing immune-related adverse events.
Collapse
Affiliation(s)
- Chiara Pozzessere
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital (CHUV), CH-1011 Lausanne, Switzerland
| | - Bianca Mazini
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital (CHUV), CH-1011 Lausanne, Switzerland
| | - Patrick Omoumi
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital (CHUV), CH-1011 Lausanne, Switzerland
| | - Mario Jreige
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital (CHUV), CH-1011 Lausanne, Switzerland
| | - Leslie Noirez
- Department of Pulmonology, Lausanne University Hospital (CHUV), CH-1011 Lausanne, Switzerland
| | - Antonia Digklia
- Department of Oncology, Lausanne University Hospital (CHUV), CH-1011 Lausanne, Switzerland
| | - François Fasquelle
- Department of Pathology, Lausanne University Hospital (CHUV), CH-1011 Lausanne, Switzerland
| | - Christine Sempoux
- Department of Pathology, Lausanne University Hospital (CHUV), CH-1011 Lausanne, Switzerland
| | - Clarisse Dromain
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital (CHUV), CH-1011 Lausanne, Switzerland
| |
Collapse
|
18
|
Albano D, Rizzo A, Slart RHJA, Hess S, Noriega-Álvarez E, Wakfie-Corieh CG, Leccisotti L, Glaudemans AWJM, Gheysens O, Treglia G. The Role of Fibroblast Activation Protein Inhibitor Positron Emission Tomography in Inflammatory and Infectious Diseases: An Updated Systematic Review. Pharmaceuticals (Basel) 2024; 17:716. [PMID: 38931383 PMCID: PMC11206476 DOI: 10.3390/ph17060716] [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: 05/16/2024] [Revised: 05/25/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
The role of fibroblast activation protein inhibitor (FAPI) positron emission tomography/computed tomography (PET/CT) is emerging for the assessment of non-oncological diseases, such as inflammatory and infectious diseases, even if the evidence in the literature is still in its initial phases. We conducted a systematic search of Scopus, PubMed/MEDLINE, Embase, and Cochrane library databases for studies published before 31 December 2023 reporting infectious and inflammatory disease imaging with FAPI PET/CT. We included twenty-one studies for a total of 1046 patients. The most frequent disease studied was lung interstitial disease, investigated in six studies for a total of 200 patients, followed by bone and joint diseases in two studies and 185 patients, IgG4-related disease in 53 patients, and Crohn's disease in 30 patients. Despite the heterogeneity of studies in terms of study design and technical features, FAPI PET/CT showed a high detection rate and diagnostic role. Moreover, when compared with 2-[18F]FDG PET/CT (n = 7 studies), FAPI PET/CT seems to have better diagnostic performances. The presence of chronic inflammation and tissue remodeling, typical of immune-mediated inflammatory conditions, may be the underlying mechanism of FAPI uptake.
Collapse
Affiliation(s)
- Domenico Albano
- Nuclear Medicine, ASST Spedali Civili Brescia, 25128 Brescia, Italy;
- Nuclear Medicine Department, University of Brescia, 25121 Brescia, Italy
| | - Alessio Rizzo
- Nuclear Medicine Division, Candiolo Cancer Institute, FPO-IRCCS, 10060 Turin, Italy;
| | - Riemer H. J. A. Slart
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (R.H.J.A.S.); (A.W.J.M.G.)
- Biomedical Photonic Imaging Group, Faculty of Science and Technology, University of Twente, 7522 NB Enschede, The Netherlands
| | - Søren Hess
- Department of Nuclear Medicine, Odense University Hospital, 5000 Odense, Denmark;
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, 5230 Odense, Denmark
| | - Edel Noriega-Álvarez
- Department of Nuclear Medicine, University Hospital of Guadalajara, 19002 Guadalajara, Spain;
| | - Cristina Gamila Wakfie-Corieh
- Department of Nuclear Medicine, Hospital Clínico San Carlos, 28040 Madrid, Spain;
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Lucia Leccisotti
- Section of Nuclear Medicine, Department of Radiological Sciences and Haematology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
- Unit of Nuclear Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Andor W. J. M. Glaudemans
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (R.H.J.A.S.); (A.W.J.M.G.)
| | - Olivier Gheysens
- Department of Nuclear Medicine, Cliniques Universitaires Saint-Luc and Institute of Clinical and Experimental Research (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium;
| | - Giorgio Treglia
- Division of Nuclear Medicine, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, 6900 Lugano, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland
| |
Collapse
|
19
|
Palaskas NL, Ali HJ, Koutroumpakis E, Ganatra S, Deswal A. Cardiovascular toxicity of immune therapies for cancer. BMJ 2024; 385:e075859. [PMID: 38749554 DOI: 10.1136/bmj-2023-075859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
In addition to conventional chemoradiation and targeted cancer therapy, the use of immune based therapies, specifically immune checkpoint inhibitors (ICIs) and chimeric antigen receptor T cell therapy (CAR-T), has increased exponentially across a wide spectrum of cancers. This has been paralleled by recognition of off-target immune related adverse events that can affect almost any organ system including the cardiovascular system. The use of ICIs has been associated with myocarditis, a less common but highly fatal adverse effect, pericarditis and pericardial effusions, vasculitis, thromboembolism, and potentially accelerated atherosclerosis. CAR-T resulting in a systemic cytokine release syndrome has been associated with myriad cardiovascular consequences including arrhythmias, myocardial infarction, and heart failure. This review summarizes the current state of knowledge regarding adverse cardiovascular effects associated with ICIs and CAR-T.
Collapse
Affiliation(s)
| | - Hyeon-Ju Ali
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Sarju Ganatra
- Lahey Hospital and Medical Center, Burlington, MA 01805
| | - Anita Deswal
- University of Texas MD Anderson Cancer Center, Houston, TX, USA 01805
| |
Collapse
|
20
|
Su Y, Liu X, Xie B, Zhang B, Yang Q, Yang MF. Comparison of Cardiac Activated Fibroblast Imaging and Magnetic Resonance Imaging in Patients with COVID-19-Related Myocarditis. Rev Cardiovasc Med 2024; 25:161. [PMID: 39076498 PMCID: PMC11267212 DOI: 10.31083/j.rcm2505161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/15/2023] [Accepted: 12/28/2023] [Indexed: 07/31/2024] Open
Abstract
Background This study aimed to explore the association between cardiac fibroblast activation and cardiac magnetic resonance (CMR) imaging parameters in patients with myocarditis following infection with coronavirus 2019 (COVID-19). Methods In this prospective study, four patients with COVID-19-related myocarditis underwent 99mTc-labeled-hydrazinonicotinamide-fibroblast activation protein inhibitor-04 (99mTc-HFAPi) single photon emission computed tomography/computed tomography (SPECT/CT) and CMR imaging. Segmental 99mTc-HFAPi activity was quantified as the percentage of average segmental myocardial count × global left ventricular target-to-background ratio. T1/T2 values, extracellular volume (ECV), and late gadolinium enhancement (LGE) were analyzed by CMR. The consistency between myocardial 99mTc-HFAPi activity and CMR parameters was explored. Results In patients with myocarditis, the proportion of segments with abnormal 99mTc-HFAPi activity was significantly higher than in those with abnormal LGE (81.25% vs. 60.93%, p = 0.011), abnormal T2 (81.25% vs. 50.00%, p < 0.001), and abnormal ECV (81.25% vs. 59.38%, p = 0.007); however, they were similar in those with abnormal native T1 (81.25% vs. 73.43%, p = 0.291). Meanwhile, 99mTc-HFAPi imaging exhibited good consistency with native T1 (kappa = 0.69). Conclusions Increased cardiac 99mTc-HFAPi activity is present in COVID-19-related myocarditis, which is correlated with the native T1 values in CMR.
Collapse
Affiliation(s)
- Yao Su
- Department of Nuclear Medicine, Beijing Chaoyang Hospital, Capital Medical University, 100020 Beijing, China
| | - Xin Liu
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, 100020 Beijing, China
| | - Boqia Xie
- Cardiac Center, Beijing Chaoyang Hospital, Capital Medical University, 100020 Beijing, China
| | - Bowen Zhang
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, 100020 Beijing, China
| | - Qi Yang
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, 100020 Beijing, China
| | - Min-Fu Yang
- Department of Nuclear Medicine, Beijing Chaoyang Hospital, Capital Medical University, 100020 Beijing, China
| |
Collapse
|
21
|
Zatarain-Nicolás E, Martín P, Márquez Rodas I, Virizuela J, Martín García A, Mitroi C, Cosín Sales J, Barrios V, Sánchez-Cabo F, Ibañez B, de Castro Carpeño J, López Fernández T. Cardiovascular toxicity of checkpoint inhibitors: review of associated toxicity and design of the Spanish Immunotherapy Registry of Cardiovascular Toxicity. Clin Transl Oncol 2023; 25:3073-3085. [PMID: 37227656 DOI: 10.1007/s12094-023-03217-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 04/29/2023] [Indexed: 05/26/2023]
Abstract
Immune checkpoint inhibitors (ICI) have changed the prognosis of many tumors. However, concerning associated cardiotoxicity has been reported. Little is known about the real-life incidence-specific surveillance protocols or the translational correlation between the underlying mechanisms and the clinical presentation of ICI-induced cardiotoxicity. The lack of data from prospective studies led us to review the current knowledge and to present the creation of the Spanish Immunotherapy Registry of Cardiovascular Toxicity (SIR-CVT), a prospective registry of patients receiving ICI that aims to examine the role of hsa-miR-Chr8:96, (a specific serum biomarker of myocarditis) in the early diagnosis of ICI-induced myocarditis. An exhaustive prospective cardiac imaging study will be performed before and during the first 12 months of treatment. The correlation between clinical, imaging, and immunologic parameters may improve our understanding of ICI-induced cardiotoxicity and enable simpler surveillance protocols. We assess ICI-induced cardiovascular toxicity and describe the rationale of the SIR-CVT.
Collapse
Affiliation(s)
- Eduardo Zatarain-Nicolás
- Cardiology Department, Hospital General Universitario Gregorio Marañón, CIBER-CV (Instituto de Salud Carlos III), Universidad Complutense, Madrid, Spain.
| | - Pilar Martín
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), CIBER-CV (Instituto de Salud Carlos III), Madrid, Spain
| | - Iván Márquez Rodas
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Juan Virizuela
- Medical Oncology Department, Hospital Universitario Virgen de la Macarena, Seville, Spain
| | - Ana Martín García
- Cardiology Department, Complejo Asistencial Universitario de Salamanca, IBSAL, USAL, CIBER-CV (Instituto de Salud Carlos III), Salamanca, Spain
| | - Cristina Mitroi
- Cardiology Department, Hospital Universitario Puerta de Hierro Majadahonda, IDIPHISA, CIBER-CV (Instituto de Salud Carlos III), Madrid, Spain
| | - Juan Cosín Sales
- Agencia de Investigación de la Sociedad Española de Cardiología (AISEC), Madrid, Spain
- Cardiology Department, Hospital Arnau de Vilanova, Universidad CEU-Cardenal Herrera, Valencia, Spain
| | - Vivencio Barrios
- Agencia de Investigación de la Sociedad Española de Cardiología (AISEC), Madrid, Spain
- Cardiology Department, Hospital Universitario Ramon y Cajal, Universidad de Alcalá, Madrid, Spain
| | - Fátima Sánchez-Cabo
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), CIBER-CV (Instituto de Salud Carlos III), Madrid, Spain
| | - Borja Ibañez
- Cardiology Department, IIS-Fundación Jiménez Díaz University Hospital, CIBER-CV (Instituto de Salud Carlos III), CNIC, Madrid, Spain
| | | | - Teresa López Fernández
- Cardiology Service, Cardio-Oncology Unit, La Paz University Hospital and IdiPAz Research Institute, Madrid, Spain.
| |
Collapse
|
22
|
Bengel FM, Diekmann J, Hess A, Jerosch-Herold M. Myocardial Fibrosis: Emerging Target for Cardiac Molecular Imaging and Opportunity for Image-Guided Therapy. J Nucl Med 2023; 64:49S-58S. [PMID: 37918842 DOI: 10.2967/jnumed.122.264867] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/25/2023] [Indexed: 11/04/2023] Open
Abstract
Myocardial fibrosis is a major contributor to the development and progression of heart failure. Significant progress in the understanding of its pathobiology has led to the introduction and preclinical testing of multiple highly specific antifibrotic therapies. Because the mechanisms of fibrosis are highly dynamic, and because the involved cell populations are heterogeneous and plastic, there is increasing emphasis that any therapy directed specifically against myocardial fibrosis will require personalization and guidance by equally specific diagnostic testing for successful clinical translation. Noninvasive imaging techniques have undergone significant progress and provide increasingly specific information about the quantity, quality, and activity of myocardial fibrosis. Cardiac MRI can precisely map the extracellular space of the myocardium, whereas nuclear imaging characterizes activated fibroblasts and immune cells as the cellular components contributing to fibrosis. Existing techniques may be used in complementarity to provide the imaging biomarkers needed for the success of novel targeted therapies. This review provides a road map on how progress in basic fibrosis research, antifibrotic drug development, and high-end noninvasive imaging may come together to facilitate the success of fibrosis-directed cardiovascular medicine.
Collapse
Affiliation(s)
- Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany; and
| | - Johanna Diekmann
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany; and
| | - Annika Hess
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany; and
| | | |
Collapse
|
23
|
Kersting D, Mavroeidi IA, Settelmeier S, Seifert R, Schuler M, Herrmann K, Rassaf T, Rischpler C. Molecular Imaging Biomarkers in Cardiooncology: A View on Established Technologies and Future Perspectives. J Nucl Med 2023; 64:29S-38S. [PMID: 37918843 DOI: 10.2967/jnumed.122.264868] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 09/05/2023] [Indexed: 11/04/2023] Open
Abstract
Novel therapeutic options have significantly improved survival and long-term outcomes in many cancer entities. Unfortunately, this improvement in outcome is often accompanied by new and increasingly relevant therapy-related cardiovascular toxicity. In this context, cardiooncology has emerged as a new field of interdisciplinary individual patient care. Important tasks are pretherapeutic risk stratification and early detection and treatment of cardiotoxicity, which comprises cardiac damage in relation to cardiovascular comorbidities, the tumor disease, and cancer treatment. Clinical manifestations can cover a broad spectrum, ranging from subtle and usually asymptomatic abnormalities to serious acute or chronic complications. Typical manifestations include acute and chronic heart failure, myo- and pericarditis, arrythmias, ischemia, and endothelial damage. They can be related to almost all current cancer treatments, including cytotoxic chemotherapy, targeted therapy, immunotherapy, hormonal therapy, and radiotherapy. Molecular imaging biomarkers can aid in pretherapeutic cardiooncologic assessment for primary prevention and personalized surveillance, detection, and differential diagnosis of cardiotoxic complications. Potential advantages over conventional diagnostics are the higher detection sensitivity for subtle changes in cardiac homeostasis, higher reproducibility, and better observer independence. Hybrid imaging with highly sensitive PET/MRI may be particularly suited for early diagnosis. Important technologies that are encouraged in current multidisciplinary guidelines are equilibrium radionuclide angiography for evaluation of ventricular function and chamber morphology, as well as myocardial perfusion imaging for additional detection of ischemia. Novel modalities that may detect even earlier signs of cardiotoxicity comprise 123I-metaiodobenzylguanidine SPECT to visualize sympathetic innervation, 18F-FDG and somatostatin receptor (68Ga-DOTATOC/DOTATATE) PET to indicate a metabolic shift and inflammation, and 68Ga-fibroblast activation protein inhibitor PET to monitor cardiac remodeling. In addition, PET imaging of mitochondrial function has recently been introduced in preclinical models and will potentially broaden the field of application through higher sensitivity and specificity and by enabling higher individualization of diagnostic concepts.
Collapse
Affiliation(s)
- David Kersting
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany;
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Ilektra-Antonia Mavroeidi
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; and
| | - Stephan Settelmeier
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Robert Seifert
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Martin Schuler
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; and
| | - Ken Herrmann
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Christoph Rischpler
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| |
Collapse
|
24
|
Addison D, Neilan TG, Barac A, Scherrer-Crosbie M, Okwuosa TM, Plana JC, Reding KW, Taqueti VR, Yang EH, Zaha VG. Cardiovascular Imaging in Contemporary Cardio-Oncology: A Scientific Statement From the American Heart Association. Circulation 2023; 148:1271-1286. [PMID: 37732422 DOI: 10.1161/cir.0000000000001174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Advances in cancer therapeutics have led to dramatic improvements in survival, now inclusive of nearly 20 million patients and rising. However, cardiovascular toxicities associated with specific cancer therapeutics adversely affect the outcomes of patients with cancer. Advances in cardiovascular imaging have solidified the critical role for robust methods for detecting, monitoring, and prognosticating cardiac risk among patients with cancer. However, decentralized evaluations have led to a lack of consensus on the optimal uses of imaging in contemporary cancer treatment (eg, immunotherapy, targeted, or biological therapy) settings. Similarly, available isolated preclinical and clinical studies have provided incomplete insights into the effectiveness of multiple modalities for cardiovascular imaging in cancer care. The aims of this scientific statement are to define the current state of evidence for cardiovascular imaging in the cancer treatment and survivorship settings and to propose novel methodological approaches to inform the optimal application of cardiovascular imaging in future clinical trials and registries. We also propose an evidence-based integrated approach to the use of cardiovascular imaging in routine clinical settings. This scientific statement summarizes and clarifies available evidence while providing guidance on the optimal uses of multimodality cardiovascular imaging in the era of emerging anticancer therapies.
Collapse
|
25
|
Samuel Y, Babu A, Karagkouni F, Ismail A, Choi S, Boussios S. Cardiac Toxicities in Oncology: Elucidating the Dark Box in the Era of Precision Medicine. Curr Issues Mol Biol 2023; 45:8337-8358. [PMID: 37886969 PMCID: PMC10605822 DOI: 10.3390/cimb45100526] [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: 10/01/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 10/28/2023] Open
Abstract
Despite current advancements in chemotherapy, immunotherapy and targeted treatments, the potential for major adverse cardiovascular events, regardless of previous cardiac history, persists. Scoring systems, such as the Heart Failure Association-International Cardio-Oncology Society (HFA-ICOS) risk assessment tool, can be utilized to evaluate several factors including prior cardiac history, risk factors and cardiac biomarkers to categorize patients into low, moderate, high, and very high-risk groups. Common cardiotoxicity complications include new or worsening left ventricular ejection fraction (LVEF), QT interval prolongation, myocardial ischaemia, hypertension, thromboembolic disease, cardiac device malfunction and valve disease. Baseline electrocardiogram (ECG) and transthoracic echocardiogram (TTE) are routinely performed for all patients commenced on cardiotoxic treatment, while other imaging modalities and biochemical markers have proven useful for monitoring. Management mainly includes early risk stratification and prompt identification of cardiovascular complications, with patient-specific surveillance throughout treatment. A multidisciplinary approach is crucial in determining the relationship between potential treatment benefits and cardiotoxicity, and whether the continuation of treatment is appropriate on a case-by-case basis. Early risk stratification, optimizing the patient's cardiovascular status prior to treatment, and prompt identification of suspected cardiotoxicity are key in significantly reducing risk. This article provides a comprehensive review of the various types of treatment-related cardiotoxicity, offering guidance on identifying high-risk patients, recognizing early signs of cardiotoxicity, and outlining appropriate treatment approaches and follow-up care for such cases.
Collapse
Affiliation(s)
- Younan Samuel
- Department of Cardiology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, Kent, UK; (Y.S.); (A.B.); (F.K.)
| | - Aswin Babu
- Department of Cardiology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, Kent, UK; (Y.S.); (A.B.); (F.K.)
| | - Foteini Karagkouni
- Department of Cardiology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, Kent, UK; (Y.S.); (A.B.); (F.K.)
| | - Ayden Ismail
- GKT School of Medicine, King’s College London, London SE1 9RT, UK;
| | - Sunyoung Choi
- Department of Cardiology, Hampshire Hospitals NHS Foundation Trust, Aldermaston Road, Basingstoke RG24 9NA, Hampshire, UK;
| | - Stergios Boussios
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, Kent, UK
- Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, King’s College London, London SE1 9RT, UK
- Kent Medway Medical School, University of Kent, Canterbury CT2 7LX, Kent, UK
- AELIA Organization, 9th Km Thessaloniki—Thermi, 57001 Thessaloniki, Greece
| |
Collapse
|
26
|
Su X, Zhao Y, Zhou X. Radiolabeled fibroblast activation protein inhibitors in the evaluation of myocardial fibrosis. Chin Med J (Engl) 2023; 136:2377-2379. [PMID: 37464383 PMCID: PMC10538885 DOI: 10.1097/cm9.0000000000002590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Indexed: 07/20/2023] Open
Affiliation(s)
- Xiangyu Su
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | | | | |
Collapse
|
27
|
Cui Y, Wang Y, Wang S, Du B, Li X, Li Y. Highlighting Fibroblasts Activation in Fibrosis: The State-of-The-Art Fibroblast Activation Protein Inhibitor PET Imaging in Cardiovascular Diseases. J Clin Med 2023; 12:6033. [PMID: 37762974 PMCID: PMC10531835 DOI: 10.3390/jcm12186033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Fibrosis is a common healing process that occurs during stress and injury in cardiovascular diseases. The evolution of fibrosis is associated with cardiovascular disease states and causes adverse effects. Fibroblast activation is responsible for the formation and progression of fibrosis. The incipient detection of activated fibroblasts is important for patient management and prognosis. Fibroblast activation protein (FAP), a membrane-bound serine protease, is almost specifically expressed in activated fibroblasts. The development of targeted FAP-inhibitor (FAPI) positron emission tomography (PET) imaging enabled the visualisation of FAP, that is, incipient fibrosis. Recently, research on FAPI PET imaging in cardiovascular diseases increased and is highly sought. Hence, we comprehensively reviewed the application of FAPI PET imaging in cardiovascular diseases based on the state-of-the-art published research. These studies provided some insights into the value of FAPI PET imaging in the early detection of cardiovascular fibrosis, risk stratification, response evaluation, and prediction of the evolution of left ventricular function. Future studies should be conducted with larger populations and multicentre patterns, especially for response evaluation and outcome prediction.
Collapse
Affiliation(s)
| | | | | | | | - Xuena Li
- Department of Nuclear Medicine, The First Hospital of China Medical University, Shenyang 110001, China; (Y.C.); (Y.W.); (S.W.); (B.D.)
| | - Yaming Li
- Department of Nuclear Medicine, The First Hospital of China Medical University, Shenyang 110001, China; (Y.C.); (Y.W.); (S.W.); (B.D.)
| |
Collapse
|
28
|
Rezaei S, Gharapapagh E, Dabiri S, Heidari P, Aghanejad A. Theranostics in targeting fibroblast activation protein bearing cells: Progress and challenges. Life Sci 2023; 329:121970. [PMID: 37481033 PMCID: PMC10773987 DOI: 10.1016/j.lfs.2023.121970] [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: 05/16/2023] [Revised: 07/03/2023] [Accepted: 07/19/2023] [Indexed: 07/24/2023]
Abstract
Cancer cells are surrounded by a complex and highly dynamic tumor microenvironment (TME). Cancer-associated fibroblasts (CAFs), a critical component of TME, contribute to cancer cell proliferation as well as metastatic spread. CAFs express a variety of biomarkers, which can be targeted for detection and therapy. Most importantly, CAFs express high levels of fibroblast activation protein (FAP) which contributes to progression of cancer, invasion, metastasis, migration, immunosuppression, and drug resistance. As a consequence, FAP is an attractive theranostic target. In this review, we discuss the latest advancement in targeting FAP in oncology using theranostic biomarkers and imaging modalities such as single-photon emission computed tomography (SPECT), positron emission tomography (PET), computed tomography (CT), fluorescence imaging, and magnetic resonance imaging (MRI).
Collapse
Affiliation(s)
- Sahar Rezaei
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Nuclear Medicine, Faculty of Medicine, Imam Reza General Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Esmaeil Gharapapagh
- Department of Nuclear Medicine, Faculty of Medicine, Imam Reza General Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahram Dabiri
- Department of Nuclear Medicine, Faculty of Medicine, Imam Reza General Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pedram Heidari
- Departments of Radiology, Massachusetts General Hospital, Boston, United States
| | - Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Nuclear Medicine, Faculty of Medicine, Imam Reza General Hospital, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
29
|
Bentestuen M, Al-Obaydi N, Zacho HD. FAPI-avid nonmalignant PET/CT findings: An expedited systematic review. Semin Nucl Med 2023; 53:694-705. [PMID: 36813670 DOI: 10.1053/j.semnuclmed.2023.02.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 02/01/2023] [Indexed: 02/22/2023]
Abstract
Fibroblast activation protein inhibitor (FAPI) is a promising tracer in oncologic positron emission tomography/computed tomography (PET/CT). Numerous studies have demonstrated the superior sensitivity of FAPI PET/CT over fluorodeoxyglucose (FDG) PET/CT in several types of cancer. However, the cancer specificity of FAPI uptake remains understudied, and several cases of false-positive FAPI PET/CT findings have been reported. A systematic search of PubMed, Embase, and Web of Science was conducted for studies published prior to April 2022 reporting nonmalignant FAPI PET/CT findings. We included original peer-reviewed articles of studies in humans using FAPI tracers radiolabeled with 68Ga or 18F that were published in English. Papers without original data and studies with insufficient information were excluded. Nonmalignant findings were presented on a per-lesion basis and grouped according to the type of organ or tissue involved. The search identified a total of 1.178 papers, of which 108 studies were eligible. Eighty studies were case reports (74%), and the remaining 28 were cohort studies (26%). A total of 2.372 FAPI-avid nonmalignant findings were reported, with the most frequent being uptake in the arteries, e.g., related to plaques (n = 1178, 49%). FAPI uptake was also frequently related to degenerative and traumatic bone and joint lesions (n = 147, 6%) or arthritis (n = 92, 4%). For organs, diffuse or focal uptake was often seen in cases of inflammation, infection, fibrosis, and IgG4-related disease (n = 157, 7%). FAPI-avid inflammatory/reactive lymph nodes (n = 121, 5%) and tuberculosis lesions (n = 51, 2%) have been reported and could prove to be potential pitfalls in cancer staging. Periodontitis (n = 76, 3%), hemorrhoids (n = 47, 2%), and scarring/wound healing (n = 35, 2%) also presented as focal uptake on FAPI PET/CT. The present review provides an overview of the reported FAPI-avid nonmalignant PET/CT findings to date. A large number of benign clinical entities may show FAPI uptake and should be kept in mind when interpreting FAPI PET/CT findings in patients with cancer.
Collapse
Affiliation(s)
- Morten Bentestuen
- Department of Nuclear Medicine and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, North Jutland Region, Denmark.
| | - Noor Al-Obaydi
- Department of Nuclear Medicine and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, North Jutland Region, Denmark
| | - Helle D Zacho
- Department of Nuclear Medicine and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, North Jutland Region, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, North Jutland Region, Denmark
| |
Collapse
|
30
|
Chitsazan M, Amin A, Ladel L, Baig A, Chitsazan M. Cardiovascular Toxicity Associated With Immune Checkpoint Inhibitor Therapy: A Comprehensive Review. Crit Pathw Cardiol 2023; 22:69-82. [PMID: 37363862 DOI: 10.1097/hpc.0000000000000327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Immune checkpoint inhibitors (ICIs), a significant breakthrough treatment of cancer, exert their function through enhancing the immune system's ability to recognize and attack cancer cells. However, these revolutionary cancer treatments have been associated with a range of immune-related adverse effects, including cardiovascular toxicity. The most commonly reported cardiovascular toxicities associated with ICIs are myocarditis, pericarditis, arrhythmias, and vasculitis. These cardiovascular manifestations are often severe and can lead to life-threatening complications. Therefore, prompt identification and management of these toxicities is critical, and a multidisciplinary teamwork by cardiologists and oncologists are required to ensure optimal patient outcomes. In this review, we summarize the current knowledge on the mechanisms underlying ICI-associated cardiovascular toxicity, clinical presentations of these toxicities, potential risk factors, diagnosis, management, and surveillance strategies during ICI therapy. While ICIs have already transformed cancer treatment, further research is needed to better understand and manage their immune-related cardiovascular effects, and possibly, to identify biomarkers which can predict the occurrence of these cardiovascular complications.
Collapse
Affiliation(s)
| | - Ahmad Amin
- Medstar Union Memorial Hospital, Baltimore, MD
| | - Luisa Ladel
- From the Department of Medicine, Norwalk Hospital, Norwalk, CT
| | - Alyza Baig
- From the Department of Medicine, Norwalk Hospital, Norwalk, CT
| | - Mitra Chitsazan
- Rajaie Cardiovascular, Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Mikail N, Chequer R, Imperiale A, Meisel A, Bengs S, Portmann A, Gimelli A, Buechel RR, Gebhard C, Rossi A. Tales from the future-nuclear cardio-oncology, from prediction to diagnosis and monitoring. Eur Heart J Cardiovasc Imaging 2023; 24:1129-1145. [PMID: 37467476 PMCID: PMC10501471 DOI: 10.1093/ehjci/jead168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/07/2023] [Indexed: 07/21/2023] Open
Abstract
Cancer and cardiovascular diseases (CVD) often share common risk factors, and patients with CVD who develop cancer are at high risk of experiencing major adverse cardiovascular events. Additionally, cancer treatment can induce short- and long-term adverse cardiovascular events. Given the improvement in oncological patients' prognosis, the burden in this vulnerable population is slowly shifting towards increased cardiovascular mortality. Consequently, the field of cardio-oncology is steadily expanding, prompting the need for new markers to stratify and monitor the cardiovascular risk in oncological patients before, during, and after the completion of treatment. Advanced non-invasive cardiac imaging has raised great interest in the early detection of CVD and cardiotoxicity in oncological patients. Nuclear medicine has long been a pivotal exam to robustly assess and monitor the cardiac function of patients undergoing potentially cardiotoxic chemotherapies. In addition, recent radiotracers have shown great interest in the early detection of cancer-treatment-related cardiotoxicity. In this review, we summarize the current and emerging nuclear cardiology tools that can help identify cardiotoxicity and assess the cardiovascular risk in patients undergoing cancer treatments and discuss the specific role of nuclear cardiology alongside other non-invasive imaging techniques.
Collapse
Affiliation(s)
- Nidaa Mikail
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Renata Chequer
- Department of Nuclear Medicine, Bichat University Hospital, AP-HP, University Diderot, 75018 Paris, France
| | - Alessio Imperiale
- Nuclear Medicine, Institut de Cancérologie de Strasbourg Europe (ICANS), University Hospitals of Strasbourg, 67093 Strasbourg, France
- Molecular Imaging-DRHIM, IPHC, UMR 7178, CNRS/Unistra, 67093 Strasbourg, France
| | - Alexander Meisel
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Kantonsspital Glarus, Burgstrasse 99, 8750 Glarus, Switzerland
| | - Susan Bengs
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Angela Portmann
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Alessia Gimelli
- Imaging Department, Fondazione CNR/Regione Toscana Gabriele Monasterio, Via G. Moruzzi 1, 56124 Pisa, Italy
| | - Ronny R Buechel
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Cathérine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- Department of Cardiology, University Hospital Inselspital Bern, Freiburgstrasse 18, 3010 Bern, Switzerland
| | - Alexia Rossi
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| |
Collapse
|
32
|
Leo I, Vidula M, Bisaccia G, Procopio MC, Licordari R, Perotto M, La Vecchia G, Miaris N, Bravo PE, Bucciarelli-Ducci C. The Role of Advanced Cardiovascular Imaging Modalities in Cardio-Oncology: From Early Detection to Unravelling Mechanisms of Cardiotoxicity. J Clin Med 2023; 12:4945. [PMID: 37568347 PMCID: PMC10419705 DOI: 10.3390/jcm12154945] [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: 05/15/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Advances in cancer therapies have led to a global improvement in patient survival rates. Nevertheless, the price to pay is a concomitant increase in cardiovascular (CV) morbidity and mortality in this population. Increased inflammation and disturbances of the immune system are shared by both cancer and CV diseases. Immunological effects of anti-cancer treatments occur with both conventional chemotherapy and, to a greater extent, with novel biological therapies such as immunotherapy. For these reasons, there is growing interest in the immune system and its potential role at the molecular level in determining cardiotoxicity. Early recognition of these detrimental effects could help in identifying patients at risk and improve their oncological management. Non-invasive imaging already plays a key role in evaluating baseline CV risk and in detecting even subclinical cardiac dysfunction during surveillance. The aim of this review is to highlight the role of advanced cardiovascular imaging techniques in the detection and management of cardiovascular complications related to cancer treatment.
Collapse
Affiliation(s)
- Isabella Leo
- Royal Brompton and Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 7EH, UK; (I.L.)
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Mahesh Vidula
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA (P.E.B.)
- Divisions of Nuclear Medicine and Cardiothoracic Imaging, Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Giandomenico Bisaccia
- Royal Brompton and Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 7EH, UK; (I.L.)
- Department of Neuroscience, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Maria Cristina Procopio
- Royal Brompton and Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 7EH, UK; (I.L.)
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Roberto Licordari
- Royal Brompton and Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 7EH, UK; (I.L.)
- Department of Biomedical and Dental Sciences and of Morphological and Functional Images, University of Messina, 98122 Messina, Italy
| | - Maria Perotto
- Royal Brompton and Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 7EH, UK; (I.L.)
| | - Giulia La Vecchia
- Royal Brompton and Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 7EH, UK; (I.L.)
- Department of Cardiovascular and Pulmonary Science, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Nikolaos Miaris
- Royal Brompton and Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 7EH, UK; (I.L.)
| | - Paco E. Bravo
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA (P.E.B.)
- Divisions of Nuclear Medicine and Cardiothoracic Imaging, Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chiara Bucciarelli-Ducci
- Royal Brompton and Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 7EH, UK; (I.L.)
- School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences and Medicine, King’s College London, London WC2R 2LS, UK
| |
Collapse
|
33
|
Mpanya D, Sathekge M, Klug E, Damelin J, More S, Hadebe B, Vorster M, Tsabedze N. Gallium-68 fibroblast activation protein inhibitor positron emission tomography in cardiovascular disease. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2023; 3:1224905. [PMID: 39355018 PMCID: PMC11440833 DOI: 10.3389/fnume.2023.1224905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 06/29/2023] [Indexed: 10/03/2024]
Abstract
Gallium-68 fibroblast activation protein inhibitor [(68Ga)Ga-FAPI] is a new radiopharmaceutical positioning itself as the preferred agent in patients with malignant tumours, competing with 2-Deoxy-2-[18F]fluoro-d-glucose [2-(18F)FDG] using positron emission tomography (PET). While imaging oncology patients with [68Ga]Ga-FAPI PET, incidental uptake of [68Ga]Ga-FAPI has been detected in the myocardium. This review summarises original research studies associating the visualisation of FAPI-based tracers in the myocardium with underlying active cardiovascular disease.
Collapse
Affiliation(s)
- Dineo Mpanya
- Division of Cardiology, Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Mike Sathekge
- Department of Nuclear Medicine, University of Pretoria, Pretoria, Gauteng, South Africa
- Nuclear Medicine Research Infrastructure, Steve Biko Academic Hospital, Pretoria, South Africa
| | - Eric Klug
- Division of Cardiology, Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Netcare Sunninghill, Sunward Park Hospitals, Johannesburg, South Africa
| | - Jenna Damelin
- Division of Cardiology, Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Stuart More
- Division of Nuclear Medicine, Department of Radiation Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Bawinile Hadebe
- Department of Nuclear Medicine, College of Health Sciences, University of KwaZulu Natal, Durban, South Africa
- Department of Nuclear Medicine, Inkosi Albert Luthuli Central Hospital, Durban, South Africa
| | - Mariza Vorster
- Department of Nuclear Medicine, College of Health Sciences, University of KwaZulu Natal, Durban, South Africa
- Department of Nuclear Medicine, Inkosi Albert Luthuli Central Hospital, Durban, South Africa
| | - Nqoba Tsabedze
- Division of Cardiology, Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
34
|
Chandekar KR, Prashanth A, Vinjamuri S, Kumar R. FAPI PET/CT Imaging-An Updated Review. Diagnostics (Basel) 2023; 13:2018. [PMID: 37370912 PMCID: PMC10297281 DOI: 10.3390/diagnostics13122018] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Despite revolutionizing the field of oncological imaging, Positron Emission Tomography (PET) with [18F]Fluorodeoxyglucose (FDG) as its workhorse is limited by a lack of specificity and low sensitivity in certain tumor subtypes. Fibroblast activation protein (FAP), a type II transmembrane glycoprotein, is expressed by cancer-associated fibroblasts (CAFs) that form a major component of the tumor stroma. FAP holds the promise to be a pan-cancer target, owing to its selective over-expression in a vast majority of neoplasms, particularly epithelial cancers. Several radiolabeled FAP inhibitors (FAPI) have been developed for molecular imaging and potential theranostic applications. Preliminary data on FAPI PET/CT remains encouraging, with extensive multi-disciplinary clinical research currently underway. This review summarizes the existing literature on FAPI PET/CT imaging with an emphasis on diagnostic applications, comparison with FDG, pitfalls, and future directions.
Collapse
Affiliation(s)
- Kunal Ramesh Chandekar
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi 110029, India;
| | - Arun Prashanth
- Department of Nuclear Medicine, MIOT International Hospital, Chennai 600089, India;
| | - Sobhan Vinjamuri
- Department of Nuclear Medicine, Royal Liverpool and Broadgreen University Hospital, Liverpool L7-8YE, UK;
| | - Rakesh Kumar
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi 110029, India;
| |
Collapse
|
35
|
Tersalvi G, Beltrani V, Grübler MR, Molteni A, Cristoforetti Y, Pedrazzini G, Treglia G, Biasco L. Positron Emission Tomography in Heart Failure: From Pathophysiology to Clinical Application. J Cardiovasc Dev Dis 2023; 10:220. [PMID: 37233187 PMCID: PMC10218989 DOI: 10.3390/jcdd10050220] [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: 03/31/2023] [Revised: 05/13/2023] [Accepted: 05/15/2023] [Indexed: 05/27/2023] Open
Abstract
Imaging modalities are increasingly being used to evaluate the underlying pathophysiology of heart failure. Positron emission tomography (PET) is a non-invasive imaging technique that uses radioactive tracers to visualize and measure biological processes in vivo. PET imaging of the heart uses different radiopharmaceuticals to provide information on myocardial metabolism, perfusion, inflammation, fibrosis, and sympathetic nervous system activity, which are all important contributors to the development and progression of heart failure. This narrative review provides an overview of the use of PET imaging in heart failure, highlighting the different PET tracers and modalities, and discussing fields of present and future clinical application.
Collapse
Affiliation(s)
- Gregorio Tersalvi
- Department of Cardiology, Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, 6900 Lugano, Switzerland
- Department of Internal Medicine, Ente Ospedaliero Cantonale, 6850 Mendrisio, Switzerland
| | - Vittorio Beltrani
- Department of Cardiology, Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, 6900 Lugano, Switzerland
- Department of Internal Medicine, Ente Ospedaliero Cantonale, 6850 Mendrisio, Switzerland
| | - Martin R. Grübler
- Department of Cardiology, Regional Hospital Neustadt, 2700 Wiener Neustadt, Austria
- Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Alessandra Molteni
- Department of Internal Medicine, Ente Ospedaliero Cantonale, 6850 Mendrisio, Switzerland
| | - Yvonne Cristoforetti
- Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), 6900 Lugano, Switzerland
| | - Giovanni Pedrazzini
- Department of Cardiology, Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, 6900 Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), 6900 Lugano, Switzerland
| | - Giorgio Treglia
- Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), 6900 Lugano, Switzerland
- Clinic of Nuclear Medicine, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
- Faculty of Biology and Medicine, University of Lausanne (UNIL), 1015 Lausanne, Switzerland
| | - Luigi Biasco
- Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), 6900 Lugano, Switzerland
- Division of Cardiology, Azienda Sanitaria Locale Torino 4, 10073 Ospedale di Ciriè, Italy
| |
Collapse
|
36
|
Hicks RJ, Giesel F, Herrmann K. Fibroblast Activation Protein as a Diagnostic and Therapeutic Target: Where Do We Go from Here? PET Clin 2023:S1556-8598(23)00032-9. [PMID: 37121834 DOI: 10.1016/j.cpet.2023.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Affiliation(s)
- Rodney J Hicks
- The Department of Medicine, St Vincent's Medical School, University of Melbourne, Melbourne, Australia; The Department of Medicine, Central Clinical School the Alfred Hospital, Monash University, Melbourne, Australia; The Melbourne Theranostic Innovation Centre, Level 8, 14-20 Blackwood Street, North Melbourne, Victoria 3051, Australia.
| | - Frederik Giesel
- Nuclear Medicine, University Hospital Duesseldorf, Duessldorf, Germany.
| | - Ken Herrmann
- Nuclear Medicine, Universitätsmedizin Essen, Essen, Germany.
| |
Collapse
|
37
|
Diekmann J, Bengel FM. Cardiac Applications of Fibroblast Activation Protein Imaging. PET Clin 2023:S1556-8598(23)00030-5. [PMID: 37117121 DOI: 10.1016/j.cpet.2023.03.004] [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: 04/30/2023]
Abstract
Several promising applications of cardiac molecular fibroblast activation protein (FAP) imaging are emerging. Myocardial fibrosis plays a key role in the complex process of cardiac remodeling and can lead to adverse clinical outcomes such as left ventricular dysfunction, propensity to arrhythmias, and reduction of perfusion. If fibrosis becomes irreversible, patients can develop heart failure. Therefore identification and early fibrosis treatment is highly warranted. FAP-targeted imaging enables new insights into pathogenesis and treatment response in various cardiac diseases such as myocardial infarction, heart failure or systemic diseases being a new selective biomarker.
Collapse
Affiliation(s)
- Johanna Diekmann
- Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Street. 1, Hannover 30625, Germany.
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Street. 1, Hannover 30625, Germany
| |
Collapse
|
38
|
Cannizzaro MT, Inserra MC, Passaniti G, Celona A, D'Angelo T, Romeo P, Basile A. Role of advanced cardiovascular imaging in chemotherapy-induced cardiotoxicity. Heliyon 2023; 9:e15226. [PMID: 37095987 PMCID: PMC10121465 DOI: 10.1016/j.heliyon.2023.e15226] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/08/2023] Open
Abstract
The development of cardiotoxicity induced by cancer treatments has emerged as a significant clinical problem, both in the short run, as it may influence drug administration in chemotherapeutic protocols, and in the long run, because it may determine adverse cardiovascular outcomes in survivors of various malignant diseases. Therefore, early detection of anticancer drug-related cardiotoxicity is an important clinical target to improve prevention of adverse effects and patient care. Today, echocardiography is the first-line cardiac imaging techniques used for identifying cardiotoxicity. Cardiac dysfunction, clinical and subclinical, is generally diagnosed by the reduction of left ventricular ejection fraction (LVEF) and global longitudinal strain (GLS). However, myocardial injury detected by echocardiography is preceded by other alterations, such as myocardial perfusion and mitochondrial and metabolic dysfunction, that can only be recognized by second-level imaging techniques, like cardiac magnetic resonance (CMR) and nuclear imaging, which, using targeted radiotracers, may help to provide information on the specific mechanisms of cardiotoxicity. In this review, we focus on the current and emerging role of CMR, as a critical diagnostic tool of cardiotoxicity in the very early phase, due to its availability and because it allows the contemporary detection of functional alterations, tissue alterations (mainly performed using T1, T2 mapping with the evaluation of extracellular volume-ECV) and perfusional alteration (evaluated with rest-stress perfusion) and, in the next future, even metabolic changes. Moreover, in the subsequent future, the use of Artificial Intelligence and big data on imaging parameters (CT, CMR) and oncoming molecular imaging datasets, including differences for gender and countries, may help predict cardiovascular toxicity at its earliest stages, avoiding its progression, with precise tailoring of patients' diagnostic and therapeutic pathways.
Collapse
Affiliation(s)
| | | | | | | | - Tommaso D'Angelo
- Diagnostic and Interventional Radiology Unit, BIOMORF Department, University Ospital “Policlinico G. Martino”, Messina, Italy
| | - Placido Romeo
- Radiology Department of AO “San Marco”, A.U.O. Policlinico “G.Rodolico-San Marco”, Catania, Italy
| | - Antonio Basile
- University of Catania, Department of Surgical and Medical Sciences and Advanced Technologies ‘G.F. Ingrassia’, Italy
| |
Collapse
|
39
|
Caobelli F, Nappi C. A spotlight on fibroblast-activated protein inhibitor (FAPi) cardiovascular imaging. Clin Transl Imaging 2023. [DOI: 10.1007/s40336-023-00548-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
|
40
|
Barton AK, Tzolos E, Bing R, Singh T, Weber W, Schwaiger M, Varasteh Z, Slart RHJA, Newby DE, Dweck MR. Emerging molecular imaging targets and tools for myocardial fibrosis detection. Eur Heart J Cardiovasc Imaging 2023; 24:261-275. [PMID: 36575058 PMCID: PMC9936837 DOI: 10.1093/ehjci/jeac242] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 11/20/2022] [Indexed: 12/29/2022] Open
Abstract
Myocardial fibrosis is the heart's common healing response to injury. While initially seeking to optimize the strength of diseased tissue, fibrosis can become maladaptive, producing stiff poorly functioning and pro-arrhythmic myocardium. Different patterns of fibrosis are associated with different myocardial disease states, but the presence and quantity of fibrosis largely confer adverse prognosis. Current imaging techniques can assess the extent and pattern of myocardial scarring, but lack specificity and detect the presence of established fibrosis when the window to modify this process may have ended. For the first time, novel molecular imaging methods, including gallium-68 (68Ga)-fibroblast activation protein inhibitor positron emission tomography (68Ga-FAPI PET), may permit highly specific imaging of fibrosis activity. These approaches may facilitate earlier fibrosis detection, differentiation of active vs. end-stage disease, and assessment of both disease progression and treatment-response thereby improving patient care and clinical outcomes.
Collapse
Affiliation(s)
- Anna K Barton
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Chancellor’s Building, Little France Crescent, Edinburgh EH16 4SB, UK
| | - Evangelos Tzolos
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Chancellor’s Building, Little France Crescent, Edinburgh EH16 4SB, UK
| | - Rong Bing
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Chancellor’s Building, Little France Crescent, Edinburgh EH16 4SB, UK
| | - Trisha Singh
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Chancellor’s Building, Little France Crescent, Edinburgh EH16 4SB, UK
| | - Wolfgang Weber
- Department of Nuclear Medicine, Clinikum rechts der Isar, Technical University of Munich, Ismaniger Straße 22, 81675 Munich, Germany
| | - Markus Schwaiger
- Department of Nuclear Medicine, Clinikum rechts der Isar, Technical University of Munich, Ismaniger Straße 22, 81675 Munich, Germany
| | - Zohreh Varasteh
- Department of Nuclear Medicine, Clinikum rechts der Isar, Technical University of Munich, Ismaniger Straße 22, 81675 Munich, Germany
| | - Riemer H J A Slart
- Faculty of Medical Sciences, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - David E Newby
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Chancellor’s Building, Little France Crescent, Edinburgh EH16 4SB, UK
| | - Marc R Dweck
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Chancellor’s Building, Little France Crescent, Edinburgh EH16 4SB, UK
| |
Collapse
|
41
|
Sun LY, Echefu G, Doshi K, Roberts ML, Hamid A, Cheng RK, Olson J, Brown SA. Commentary: "Multimodality advanced cardiovascular and molecular imaging for early detection and monitoring of cancer therapy-associated cardiotoxicity and the role of artificial intelligence and big data". Front Cardiovasc Med 2023; 10:982028. [PMID: 36923958 PMCID: PMC10009261 DOI: 10.3389/fcvm.2023.982028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 01/31/2023] [Indexed: 03/02/2023] Open
Affiliation(s)
- Louise Y Sun
- Division of Cardiothoracic Anesthesiology, Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Gift Echefu
- Department of Internal Medicine, Baton Rouge General Medical Center, Baton Rouge, LA, United States
| | - Krishna Doshi
- Department of Internal Medicine, Advocate Lutheran General Hospital, Park Ridge, IL, United States
| | - Michelle L Roberts
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | | | - Richard K Cheng
- Cardio-Oncology Program, Division of Cardiology, University of Washington, Seattle, WA, United States
| | - Jessica Olson
- Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Sherry-Ann Brown
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
42
|
Wang G, Yang Q, Wu S, Xu X, Li X, Liang S, Pan G, Zuo C, Zhao X, Cheng C, Liu S. Molecular imaging of fibroblast activity in pressure overload heart failure using [ 68 Ga]Ga-FAPI-04 PET/CT. Eur J Nucl Med Mol Imaging 2023; 50:465-474. [PMID: 36171409 DOI: 10.1007/s00259-022-05984-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/20/2022] [Indexed: 01/11/2023]
Abstract
PURPOSE We aimed to evaluate whether [68 Ga]Ga-FAPI-04 PET/CT could characterize the early stages of cardiac fibrosis in pressure overload heart failure. METHODS Sprague-Dawley rats underwent abdominal aortic constriction (AAC) (n = 12) and sham surgery (n = 10). All rats were scanned with [68 Ga]Ga-FAPI-04 PET/CT at 2, 4, and 8 weeks after surgery. The expression of fibroblast activation protein (FAP) in the myocardium was detected by immunohistochemistry. [68 Ga]Ga-FAPI-04 PET signal and FAP expression were compared between two groups. RESULTS Compared with the sham group, the AAC group presented with decreased ejection fraction (EF) and fractional shortening (FS) and increased left ventricular internal dimensions in diastole (LVIDd) and systole (LVIDs) at 4 and 8 weeks (all p < 0.01). The AAC group showed higher [68 Ga]Ga-FAPI-04 accumulation in the heart than the sham group at 2, 4, and 8 weeks, and FAPI increased significantly from 2 to 8 weeks (all p < 0.001). Immunohistochemistry confirmed the higher density of the FAP+ area in the AAC group. The intensity of the [68 Ga]Ga-FAPI-04 correlated with the density of the FAP+ area (p < 0.001). The expression of the [68 Ga]Ga-FAPI-04 at 4 weeks correlated with the deterioration of cardiac function at 8 weeks (EF: R = - 0.87; FS: R = - 0.72; LVIDd: R = 0.77; LVIDs: R = 0.79; all p < 0.001). The AAC group also showed an increased [68 Ga]Ga-FAPI-04 signal in the liver, peaking at 4 weeks and then declining. Cardiac and liver PET signals correlated at 4 weeks in the AAC group (R = 0.69, p = 0.0010), suggesting an early fibrotic link between organs. A combination of the [68 Ga]Ga-FAPI-04 intensity in the heart and liver at 4 weeks better predicted the deterioration of cardiac function at 8 weeks. CONCLUSIONS The activated fibroblasts in the heart and liver after pressure overload can be monitored by [68 Ga]Ga-FAPI-04 PET/CT, which reveals an early fibrotic link in cardio-liver interactions and could better predict nonischemic heart failure prognosis.
Collapse
Affiliation(s)
- Guokun Wang
- Department of Cardiovascular Surgery, the First Affiliated Hospital (Changhai Hospital) of Naval Medical University, Shanghai, China
| | - Qinqin Yang
- Department of Nuclear Medicine, the First Affiliated Hospital (Changhai Hospital) of Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Shengyong Wu
- Department of Military Health Statistics, Naval Medical University, Shanghai, China
| | - Xudong Xu
- Department of Cardiology, the First Affiliated Hospital (Changhai Hospital) of Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Xiao Li
- Department of Nuclear Medicine, the First Affiliated Hospital (Changhai Hospital) of Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Siyu Liang
- Department of Nuclear Medicine, the First Affiliated Hospital (Changhai Hospital) of Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Guixia Pan
- Department of Nuclear Medicine, the First Affiliated Hospital (Changhai Hospital) of Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Changjing Zuo
- Department of Nuclear Medicine, the First Affiliated Hospital (Changhai Hospital) of Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Xianxian Zhao
- Department of Cardiology, the First Affiliated Hospital (Changhai Hospital) of Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Chao Cheng
- Department of Nuclear Medicine, the First Affiliated Hospital (Changhai Hospital) of Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| | - Suxuan Liu
- Department of Cardiology, the First Affiliated Hospital (Changhai Hospital) of Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
43
|
Lee IK, Noguera-Ortega E, Xiao Z, Todd L, Scholler J, Song D, Liousia M, Lohith K, Xu K, Edwards KJ, Farwell MD, June CH, Albelda SM, Puré E, Sellmyer MA. Monitoring Therapeutic Response to Anti-FAP CAR T Cells Using [18F]AlF-FAPI-74. Clin Cancer Res 2022; 28:5330-5342. [PMID: 35972732 PMCID: PMC9771904 DOI: 10.1158/1078-0432.ccr-22-1379] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/28/2022] [Accepted: 08/12/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE Despite the success of chimeric antigen receptor (CAR) T-cell therapy against hematologic malignancies, successful targeting of solid tumors with CAR T cells has been limited by a lack of durable responses and reports of toxicities. Our understanding of the limited therapeutic efficacy in solid tumors could be improved with quantitative tools that allow characterization of CAR T-targeted antigens in tumors and accurate monitoring of response. EXPERIMENTAL DESIGN We used a radiolabeled FAP inhibitor (FAPI) [18F]AlF-FAPI-74 probe to complement ongoing efforts to develop and optimize FAP CAR T cells. The selectivity of the radiotracer for FAP was characterized in vitro, and its ability to monitor changes in FAP expression was evaluated using rodent models of lung cancer. RESULTS [18F]AlF-FAPI-74 showed selective retention in FAP+ cells in vitro, with effective blocking of the uptake in presence of unlabeled FAPI. In vivo, [18F]AlF-FAPI-74 was able to detect FAP expression on tumor cells as well as FAP+ stromal cells in the tumor microenvironment with a high target-to-background ratio. We further demonstrated the utility of the tracer to monitor changes in FAP expression following FAP CAR T-cell therapy, and the PET imaging findings showed a robust correlation with ex vivo analyses. CONCLUSIONS This noninvasive imaging approach to interrogate the tumor microenvironment represents an innovative pairing of a diagnostic PET probe with solid tumor CAR T-cell therapy and has the potential to serve as a predictive and pharmacodynamic response biomarker for FAP as well as other stroma-targeted therapies. A PET imaging approach targeting FAP expressed on activated fibroblasts of the tumor stroma has the potential to predict and monitor therapeutic response to FAP-targeted CAR T-cell therapy. See related commentary by Weber et al., p. 5241.
Collapse
Affiliation(s)
- Iris K. Lee
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Estela Noguera-Ortega
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Zebin Xiao
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pensnsylvania, Philadelphia, PA, USA
| | - Leslie Todd
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pensnsylvania, Philadelphia, PA, USA
| | - John Scholler
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Decheng Song
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maria Liousia
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Katheryn Lohith
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kexiang Xu
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kimberly J. Edwards
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael D. Farwell
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H. June
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Steven M. Albelda
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ellen Puré
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pensnsylvania, Philadelphia, PA, USA
| | - Mark A. Sellmyer
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- The Deparment of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
44
|
Lin K, Chen X, Xue Q, Yao S, Miao W. Diffuse uptake of [ 68Ga]Ga-FAPI in the left heart in a patient with hypertensive heart disease by PET/CT. J Nucl Cardiol 2022; 29:3596-3598. [PMID: 33988808 DOI: 10.1007/s12350-021-02646-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 04/08/2021] [Indexed: 01/18/2023]
Affiliation(s)
- Kaixian Lin
- Department of Nuclear Medicine, Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian Province, China
| | - Xin Chen
- Department of Cardiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian Province, China
| | - Qianqian Xue
- Department of Nuclear Medicine, Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian Province, China
| | - Shaobo Yao
- Department of Nuclear Medicine, Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian Province, China
| | - Weibing Miao
- Department of Nuclear Medicine, Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian Province, China.
| |
Collapse
|
45
|
Thuny F, Naidoo J, Neilan TG. Cardiovascular complications of immune checkpoint inhibitors for cancer. Eur Heart J 2022; 43:4458-4468. [PMID: 36040835 PMCID: PMC10263267 DOI: 10.1093/eurheartj/ehac456] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/07/2022] [Accepted: 08/03/2022] [Indexed: 12/14/2022] Open
Abstract
Over the last decade or so, there has been a paradigm shift in the oncologic care of patients with a range of solid tumour and haematologic malignancies, away from traditional cytotoxic chemotherapy and towards personalized cancer treatments, using both targeted therapy and immunotherapy. This shift has contributed to the remarkable and sustained increase in the number of cancer survivors and the longevity of patients with a cancer diagnosis. This review will focus on the cardiovascular effects of immune checkpoint inhibitors and will present a background on immune checkpoint inhibition for cancer, the epidemiology, potential mechanisms, the potential insights into cardiovascular biology, and a diagnostic and therapeutic approach to potential cases. Our understanding of the cardiovascular effects of immune checkpoint inhibitors needs to improve. However, the evolution necessarily needs to be rapid. Initial observations noted that immune checkpoint inhibitor therapy can lead to a fulminant myocarditis. Recent reports have expanded the effect of immune checkpoint inhibitor therapy on the cardiovascular system to include an increase in cardiac dysfunction without myocarditis, arrhythmias, venous thromboembolic disease, accelerated atherosclerosis, and atherosclerosis-related cardiovascular events. The association between immune checkpoint inhibitor therapy and an increase in these cardiovascular events is not only limited to events occurring within the first few weeks after starting therapy but can also include events that occur months to years after therapy. The latter observation is especially of relevance in those treated with adjuvant or neoadjuvant therapy. There needs to be a shift from recognition of an increase in cardiovascular events to currently approved immune checkpoint inhibitor therapies to understanding the mechanisms that lead to adverse cardiovascular effects, understanding who is at risk, and understanding what we can do about it.
Collapse
Affiliation(s)
- Franck Thuny
- Aix-Marseille University, University Mediterranean Center of Cardio-Oncology, Unit of Heart Failure and Valvular Heart Diseases, Department of Cardiology, North Hospital, Assistance Publique - Hôpitaux de Marseille, Centre for CardioVascular and Nutrition Research (C2VN), Inserm 1263, Inrae 1260, Marseille, France
| | - Jarushka Naidoo
- Sidney Kimmel Comprehensive Cancer Center, John Hopkins University, Baltimore, MD, USA
- Beaumont Hospital and RCSI University of Health Sciences, Dublin, Ireland
| | - Tomas G Neilan
- Division of Cardiology and Department of Radiology, Cardiovascular Imaging Research Center (CIRC), Massachusetts General Hospital, Boston, MA, USA
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
46
|
Lyon AR, López-Fernández T, Couch LS, Asteggiano R, Aznar MC, Bergler-Klein J, Boriani G, Cardinale D, Cordoba R, Cosyns B, Cutter DJ, de Azambuja E, de Boer RA, Dent SF, Farmakis D, Gevaert SA, Gorog DA, Herrmann J, Lenihan D, Moslehi J, Moura B, Salinger SS, Stephens R, Suter TM, Szmit S, Tamargo J, Thavendiranathan P, Tocchetti CG, van der Meer P, van der Pal HJH. 2022 ESC Guidelines on cardio-oncology developed in collaboration with the European Hematology Association (EHA), the European Society for Therapeutic Radiology and Oncology (ESTRO) and the International Cardio-Oncology Society (IC-OS). Eur Heart J 2022; 43:4229-4361. [PMID: 36017568 DOI: 10.1093/eurheartj/ehac244] [Citation(s) in RCA: 1218] [Impact Index Per Article: 406.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
|
47
|
Xing Q, Zhang Z, Zhu B, Lin Q, Shen L, Li F, Xia Z, Zhao Z. Case Report: Treatment for steroid-refractory immune-related myocarditis with tofacitinib. Front Immunol 2022; 13:944013. [PMID: 36189247 PMCID: PMC9521497 DOI: 10.3389/fimmu.2022.944013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 08/30/2022] [Indexed: 12/19/2022] Open
Abstract
Introduction Immune therapy has ushered in a new era of tumor treatment, at the expense of immune-related adverse events, including rare but fatal adverse cardiovascular events, such as myocarditis. Steroids remain the cornerstone of therapy for immune-related myocarditis, with no clear consensus on additional immunosuppressive treatment for steroid-refractory cases yet. Case report Here, we report a patient with stage IV nasopharyngeal carcinoma who developed immune-related myocarditis in the fourth course of therapy with immune checkpoint inhibitors. The patient presented with precordial discomfort with elevation of cardiac enzymes and interleukin-6, atypical electrocardiographic abnormalities, and reduced left ventricular ejection fraction. Coronary computed tomography angiography excluded the possibility of acute coronary syndrome. The therapy with tofacitinib targeting the Janus kinase-signal transducer and activator of transcription signal pathway was successfully conducted, since there was no significant improvement in troponin under high-dose steroid and intravenous immunoglobulin treatment. The patient recovered without major adverse cardiac events during hospitalization. Discussion The safety and efficacy of tofacitinib in a patient with steroid-refractory immune-related myocarditis were investigated, hoping to provide a basis for prospective therapeutic strategies. Tofacitinib led to remarkable remissions in primary autoimmune disease by blocking the inflammatory cascade, indicating its potential therapeutic use in immune-related adverse events.
Collapse
Affiliation(s)
- Qian Xing
- Department of Critical Care, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhongwei Zhang
- Department of Critical Care, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- *Correspondence: Zhongwei Zhang, ; Biao Zhu,
| | - Biao Zhu
- Department of Critical Care, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- *Correspondence: Zhongwei Zhang, ; Biao Zhu,
| | - Qionghua Lin
- Department of Critical Care, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lihua Shen
- Department of Critical Care, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fangfang Li
- Department of Critical Care, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhili Xia
- Department of Critical Care, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhiyong Zhao
- Department of Critical Care, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
48
|
Lyon AR, López-Fernández T, Couch LS, Asteggiano R, Aznar MC, Bergler-Klein J, Boriani G, Cardinale D, Cordoba R, Cosyns B, Cutter DJ, de Azambuja E, de Boer RA, Dent SF, Farmakis D, Gevaert SA, Gorog DA, Herrmann J, Lenihan D, Moslehi J, Moura B, Salinger SS, Stephens R, Suter TM, Szmit S, Tamargo J, Thavendiranathan P, Tocchetti CG, van der Meer P, van der Pal HJH. 2022 ESC Guidelines on cardio-oncology developed in collaboration with the European Hematology Association (EHA), the European Society for Therapeutic Radiology and Oncology (ESTRO) and the International Cardio-Oncology Society (IC-OS). Eur Heart J Cardiovasc Imaging 2022; 23:e333-e465. [PMID: 36017575 DOI: 10.1093/ehjci/jeac106] [Citation(s) in RCA: 156] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
49
|
Li Y, Liu PJ, Zhang ZL, Wang YN. Cardiac imaging techniques for the assessment of immune checkpoint inhibitor-induced cardiotoxicity and their potential clinical applications. Am J Cancer Res 2022; 12:3548-3560. [PMID: 36119829 PMCID: PMC9442027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 07/09/2022] [Indexed: 06/15/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have encouraged a paradigm shift in the clinical management of patients with cancer. Despite the dramatically improved tumor response and patient prognosis, ICIs have been associated with ICI-related myocarditis, which has a high fatality rate. Cardiac imaging plays a critical role in the assessment of cardiac injury. Echocardiography, cardiac magnetic resonance imaging, and targeted tracer-based cardiac molecular imaging techniques alone or in combination reflect pathophysiology and depict different aspects of lesions at different clinical stages, i.e., they have potentially complementary value. Imaging techniques for identifying ICI-induced cardiotoxicity at the early stage may reduce the incidence of adverse cardiovascular events. Particularly in planned ICI therapy among patients with cancer, improved monitoring approaches to identify patients who are at the highest risk of ICI-related myocarditis may help in refining clinical decisions, allowing treatment to be more accurately targeted toward patients who are most likely to benefit. In this study, we systematically reviewed the studies on cardiac imaging techniques for assessing ICI-induced cardiotoxicity. We elaborated about the potential applications of cardiac imaging techniques for the optimized management of patients with ICI-related myocarditis, including risk stratification, diagnosis, and prognosis.
Collapse
Affiliation(s)
- Yi Li
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
- Medical Science Research Center (MRC), Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Pei-Jun Liu
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Zhuo-Li Zhang
- Radiological Sciences, Chao Family Comprehensive Cancer Center, University of California (Irvine)USA
| | - Yi-Ning Wang
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| |
Collapse
|
50
|
Fibroblast Activation Protein Inhibitor Theranostics. PET Clin 2022; 17:453-464. [PMID: 35717101 DOI: 10.1016/j.cpet.2022.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The evolution of the fibroblast activation protein inhibitor molecules over the past decade has brought into the forefront a novel theranostic agent that has the potential of matching the workhorse of PET/computed tomography, [fluorine-18] fluoro-2-deoxy-d-glucose (18F-FDG). It is hoped that in the next decade it can act as a complementary tracer to 18F-FDG, in providing phenotypic and biomarker information and also in directing fibroblast activation protein-targeted therapies.
Collapse
|