1
|
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
|
2
|
Tamboline M, Collins J, Jackson W, Gu W, Worssam M, Cheng P, David J, Taschereau R, Chatziioannou AF, Jackson S, Xu S, Ikotun OF. Preclinical evaluation of high-resolution CT, 18F-FDG, and 18F-NaF PET imaging for longitudinal monitoring of atherosclerosis. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07276-1. [PMID: 40289041 DOI: 10.1007/s00259-025-07276-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/07/2025] [Indexed: 04/29/2025]
Abstract
RATIONALE Detection of atherosclerosis is essential to the management and prevention of life-threatening cardiovascular events. Although non-invasive imaging modalities, such as 18F-sodium fluoride (18F-NaF), 18F-fluorodeoxyglucose (18F-FDG) PET, and CT, visualize distinct hallmarks of atherosclerosis, there has yet to be a singular multi-cohort interrogation of their strengths and limitations. Thus, we focused on identifying the optimal approach for visualizing atherosclerosis at different stages of disease progression. METHODS In this study, 6-week-old, male, ApoE deficient mice (Apoe-/-) were placed on a high cholesterol diet for 12-20 weeks to induce calcific atherosclerotic disease. Age-matched, male, wildtype (WT) C57BL/6 mice fed with regular chow served as the control group. Mice were imaged at 12, 15, 18, and 20 weeks after starting their respective diets. To follow the progression of calcified atherosclerotic lesions, at each time point, in vivo, 18F-NaF microPET/CT images were acquired 1 h and 3 h post tracer i.v. injection. In a separate cohort, in vivo 18F-FDG PET/CT images were acquired at 3 and 5 h post tracer i.v. injection to follow inflammation as a result of progressive atherosclerotic lesion formation. High-resolution microCT images were acquired for all mice to visualize aorta calcification. After each imaging session, a subset (n = 3) was euthanized from each group and histological analysis of the aorta was performed to confirm disease progression. RESULTS In this comparative study, within the same cohort, 18F-NaF PET detected atherosclerotic calcification earlier than microCT. At both 1 and 3 h post-injection (p.i.), calcified lesions were clearly detected by 18F-NaF with a six-fold higher signal in Apoe-/- compared to WT mice. Interestingly, 18F-NaF signal peaked at week 18, whereas aortic CT signal progressively increased with a 13-, 16-, and 29-fold at 15, 18, and 20 weeks, respectively. 18F-FDG arortic accumulation at weeks 12 and 15, were significantly greater in Apoe -/- mice than WT control when images were acquired at 5 h but not at 3 h p.i.. In contrast to histological analysis, at ≥ 16 weeks where inflammation is significantly elevated, 18F-FDG was equivalent in Apoe-/- and WT control mice and significantly reduced with disease progression. CONCLUSIONS Our results show that 18F-NaF PET and 18F-FDG PET are sensitive imaging modalities for the early detection of atherosclerotic lesions. However, both 18F-NaF PET and high-resolution microCT prove to be effective methods for monitoring late-stage and progressive disease.
Collapse
Affiliation(s)
- Mikayla Tamboline
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, 90025, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90025, USA
| | - Jeffrey Collins
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, 90025, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90025, USA
| | - William Jackson
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Wenduo Gu
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Matthew Worssam
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Paul Cheng
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
| | - John David
- Cardiovascular, Amgen Inc., Thousand Oaks, CA, 91320, USA
| | - Richard Taschereau
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, 90025, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90025, USA
| | - Arion F Chatziioannou
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, 90025, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90025, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90025, USA
| | - Simon Jackson
- Cardiovascular, Amgen Inc., Thousand Oaks, CA, 91320, USA
| | - Shili Xu
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, 90025, USA.
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90025, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90025, USA.
- , Los Angeles, USA.
| | - Oluwatayo F Ikotun
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, 90025, USA.
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90025, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90025, USA.
- , Los Angeles, USA.
| |
Collapse
|
3
|
Sanchez S, Mossa-Basha M, Anagnostakou V, Liebeskind DS, Samaniego EA. Comprehensive imaging analysis of intracranial atherosclerosis. J Neurointerv Surg 2025; 17:311-320. [PMID: 38719445 DOI: 10.1136/jnis-2023-020622] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/20/2024] [Indexed: 01/26/2025]
Abstract
Intracranial atherosclerotic disease (ICAD) involves the build-up of atherosclerotic plaques in cerebral arteries, significantly contributing to stroke worldwide. Diagnosing ICAD entails various techniques that measure arterial stenosis severity. Digital subtraction angiography, CT angiography, and magnetic resonance angiography are established methods for assessing stenosis. High-resolution MRI offers additional insights into plaque morphology including plaque burden, hemorrhage, remodeling, and contrast enhancement. These metrics and plaque traits help identify symptomatic plaques. Techniques like transcranial Doppler, CT perfusion, computational fluid dynamics, and quantitative MRA analyze blood flow restrictions due to ICAD. Intravascular ultrasound or optical coherence tomography have a very high spatial resolution and can assess the structure of the arterial wall and the plaque from the lumen of the target vascular territory. Positron emission tomography could further detect inflammation markers. This review aims to provide a comprehensive overview of the spectrum of current modalities for atherosclerotic plaque analysis and risk stratification.
Collapse
Affiliation(s)
| | | | - Vania Anagnostakou
- Radiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - David S Liebeskind
- Department of Neurology, University of California Los Angeles, Los Angeles, California, USA
| | - Edgar A Samaniego
- Neurology, Neurosurgery and Radiology, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| |
Collapse
|
4
|
Bacmeister L, Hempfling N, Maier A, Weber S, Buellesbach A, Heidenreich A, Bojti I, Gissler MC, Hilgendorf I, von Zur Muehlen C, Westermann D, Meyer PT, Goetz C, Wolf D. Longitudinal Assessment of Subclinical Arterial Inflammation in Patients Receiving Immune Checkpoint Inhibitors by Sequential [ 18F]FDG PET Scans. Circ Cardiovasc Imaging 2025; 18:e016851. [PMID: 39902567 DOI: 10.1161/circimaging.124.016851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 11/25/2024] [Indexed: 02/05/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs), though revolutionary in cancer treatment, may accelerate atherosclerosis by inducing arterial inflammation. Due to a lack of controlled studies, the capacity of arterial 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) uptake in patients with cancer to detect this arterial inflammation remains unclear. METHODS Arterial [18F]FDG uptake at 6 anatomic landmarks was assessed on serial positron emission tomography scans in patients with cancer treated at a German University Hospital between January 2010 and May 2023. Patients aged ≥65 years with at least 4 sequential scans within 30 months were included. Linear mixed regression analyses were used to evaluate the change in arterial tracer uptake in patients who received ICI or not. RESULTS Of the 156 patients included, 50 (30.1%) received ICIs after the baseline scan. Baseline arterial [18F]FDG uptake correlated with traditional cardiovascular risk factors, such as body mass index and male sex. Cross-sectional analyses suggested a negative effect of cholesterol-lowering medication on arterial [18F]FDG uptake at follow-up. In a time-dependent interaction analysis, arterial [18F]FDG uptake increased by 0.8% annually in patients without ICIs (95% CI, 0.2%-1.4%), potentially reflecting the background progression of arterial inflammation in patients with cancer. In ICI users, [18F]FDG uptake increased by 2.5% annually (95% CI, 1.7%-3.3%; P=0.001 for interaction with no ICI). Higher annual increase rates in ICI users were consistent across several anatomic landmarks, preexisting cardiovascular disease status, arterial calcification status, and concomitant chemotherapy or steroid use. However, this effect did not reach statistical significance in patients with melanoma and those with prior irradiation therapy. CONCLUSIONS This is the first controlled clinical study supporting the role of ICIs in accelerating atherosclerosis through low-grade arterial inflammation. However, although detectable by repeated [18F]FDG scans, the increase in tracer uptake associated with ICI use was modest compared with individual variability, questioning whether [18F]FDG captures the full pathophysiological process of ICI-induced, lymphocyte-driven inflammation.
Collapse
Affiliation(s)
- Lucas Bacmeister
- Clinic for Cardiology and Angiology, Medical Center - University of Freiburg, University Heart Center Freiburg Bad Krozingen, University Medical Center, Germany (L.B., N.H., A.M., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, D. Wolf)
- Faculty of Medicine, University of Freiburg, Germany (L.B., N.H., A.M., S.W., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, P.T.M., C.G., D. Wolf)
| | - Niklas Hempfling
- Clinic for Cardiology and Angiology, Medical Center - University of Freiburg, University Heart Center Freiburg Bad Krozingen, University Medical Center, Germany (L.B., N.H., A.M., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, D. Wolf)
- Faculty of Medicine, University of Freiburg, Germany (L.B., N.H., A.M., S.W., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, P.T.M., C.G., D. Wolf)
| | - Alexander Maier
- Clinic for Cardiology and Angiology, Medical Center - University of Freiburg, University Heart Center Freiburg Bad Krozingen, University Medical Center, Germany (L.B., N.H., A.M., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, D. Wolf)
- Faculty of Medicine, University of Freiburg, Germany (L.B., N.H., A.M., S.W., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, P.T.M., C.G., D. Wolf)
| | - Susanne Weber
- Faculty of Medicine, University of Freiburg, Germany (L.B., N.H., A.M., S.W., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, P.T.M., C.G., D. Wolf)
- Institute of Medical Biometry and Statistics, Medical Center - University of Freiburg, Germany (S.W.)
| | - Annette Buellesbach
- Clinic for Cardiology and Angiology, Medical Center - University of Freiburg, University Heart Center Freiburg Bad Krozingen, University Medical Center, Germany (L.B., N.H., A.M., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, D. Wolf)
- Faculty of Medicine, University of Freiburg, Germany (L.B., N.H., A.M., S.W., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, P.T.M., C.G., D. Wolf)
| | - Adrian Heidenreich
- Clinic for Cardiology and Angiology, Medical Center - University of Freiburg, University Heart Center Freiburg Bad Krozingen, University Medical Center, Germany (L.B., N.H., A.M., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, D. Wolf)
- Faculty of Medicine, University of Freiburg, Germany (L.B., N.H., A.M., S.W., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, P.T.M., C.G., D. Wolf)
| | - Istvan Bojti
- Clinic for Cardiology and Angiology, Medical Center - University of Freiburg, University Heart Center Freiburg Bad Krozingen, University Medical Center, Germany (L.B., N.H., A.M., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, D. Wolf)
- Faculty of Medicine, University of Freiburg, Germany (L.B., N.H., A.M., S.W., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, P.T.M., C.G., D. Wolf)
| | - Mark Colin Gissler
- Clinic for Cardiology and Angiology, Medical Center - University of Freiburg, University Heart Center Freiburg Bad Krozingen, University Medical Center, Germany (L.B., N.H., A.M., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, D. Wolf)
- Faculty of Medicine, University of Freiburg, Germany (L.B., N.H., A.M., S.W., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, P.T.M., C.G., D. Wolf)
| | - Ingo Hilgendorf
- Clinic for Cardiology and Angiology, Medical Center - University of Freiburg, University Heart Center Freiburg Bad Krozingen, University Medical Center, Germany (L.B., N.H., A.M., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, D. Wolf)
- Faculty of Medicine, University of Freiburg, Germany (L.B., N.H., A.M., S.W., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, P.T.M., C.G., D. Wolf)
| | - Constantin von Zur Muehlen
- Clinic for Cardiology and Angiology, Medical Center - University of Freiburg, University Heart Center Freiburg Bad Krozingen, University Medical Center, Germany (L.B., N.H., A.M., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, D. Wolf)
- Faculty of Medicine, University of Freiburg, Germany (L.B., N.H., A.M., S.W., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, P.T.M., C.G., D. Wolf)
| | - Dirk Westermann
- Clinic for Cardiology and Angiology, Medical Center - University of Freiburg, University Heart Center Freiburg Bad Krozingen, University Medical Center, Germany (L.B., N.H., A.M., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, D. Wolf)
- Faculty of Medicine, University of Freiburg, Germany (L.B., N.H., A.M., S.W., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, P.T.M., C.G., D. Wolf)
| | - Philipp Tobias Meyer
- Faculty of Medicine, University of Freiburg, Germany (L.B., N.H., A.M., S.W., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, P.T.M., C.G., D. Wolf)
- Department of Nuclear Medicine, Medical Center-University of Freiburg, Germany (P.T.M., C.G.)
| | - Christian Goetz
- Faculty of Medicine, University of Freiburg, Germany (L.B., N.H., A.M., S.W., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, P.T.M., C.G., D. Wolf)
- Department of Nuclear Medicine, Medical Center-University of Freiburg, Germany (P.T.M., C.G.)
| | - Dennis Wolf
- Clinic for Cardiology and Angiology, Medical Center - University of Freiburg, University Heart Center Freiburg Bad Krozingen, University Medical Center, Germany (L.B., N.H., A.M., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, D. Wolf)
- Faculty of Medicine, University of Freiburg, Germany (L.B., N.H., A.M., S.W., A.B., A.H., I.B., M.C.G., I.H., C.v.z.M., D. Westermann, P.T.M., C.G., D. Wolf)
| |
Collapse
|
5
|
Mujahid A, Alotaibi B, DeMellier C, Gallegos C, Sherwani M, Alexandrian A, Sorace A, Brady A, George R. Characterization of Cutaneous Radiation Syndrome in a Mouse Model Using [18F]F- Fluorodeoxyglucose Positron Emission Tomography. HEALTH PHYSICS 2025:00004032-990000000-00222. [PMID: 39817868 DOI: 10.1097/hp.0000000000001947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
ABSTRACT Ionizing radiation on the skin has the potential to cause various sequelae affecting quality of life and even leading to death due to multi-system failure. The development of radiation dermatitis is attributed to oxidative damage to the skin's basal layer and alterations in immune response, leading to inflammation. Past studies have shown that [18F]F-2-fluoro-2-deoxyglucose positron emission tomography-computed tomography ([18F]F-FDG PET/CT) can be used effectively for the detection of inflammatory activity, especially in conditions like hidradenitis suppurativa, psoriasis, and early atherosclerosis. Since currently there are no specific tests for radiation dermatitis, our study aimed to validate whether radiation dermatitis induced in mice can be accurately visualized and measured using [18F]F-FDG PET/CT. We induced cutaneous radiation syndrome in BALB/c mice with different radiation absorbed doses and monitored symptom development through photography, PET imaging, and histopathology, marking the first attempt at non-invasively quantifying radiation dermatitis effects at the molecular level using PET imaging. Our results showed that there were progressive changes in the dorsal skin of irradiated mice, with notable differences between those exposed to varying doses of radiation. Erythema, epilation, and desquamation were more pronounced in mice exposed to lower doses (25 Gy and 35 Gy) than at 45 Gy; however, by the third week, severe skin deterioration, including ulceration and dermal atrophy, was evident in mice irradiated with 35 Gy and 45 Gy. PET/CT imaging revealed increased [18F]F-FDG uptake in the irradiated dorsal skin area of all mice compared to controls, with more pronounced avidity for the lesion in the 25 Gy and 35 Gy than the 45 Gy. Comparison of tissue-normalized SUVMax values showed that both the 25 Gy and 35 Gy mice exhibited fourfold [18F]F-FDG uptake in the dorsal skin compared to controls, while a twofold uptake was seen at 45 Gy, thus indicating substantial metabolic changes in the dorsal skin induced by radiation exposure. Histopathological analyses correlated with the above findings, demonstrating generalized hypertrophy and epidermal thickening in all irradiated mice compared to controls, with thicker epidermis observed with higher radiation doses and increased destruction of microvasculature. In conclusion, PET/CT emerges as a successful tool for imaging cutaneous radiation syndrome, with the observed dermal changes in irradiated mice closely aligning with metabolic alterations of the affected area.
Collapse
Affiliation(s)
- Alaa Mujahid
- Nuclear Medicine and Molecular Imaging Sciences Program, Department of Clinical & Diagnostic Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL
| | - Bashayr Alotaibi
- Nuclear Medicine and Molecular Imaging Sciences Program, Department of Clinical & Diagnostic Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL
| | - Chloe DeMellier
- Department of Radiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Carlos Gallegos
- Department of Biomedical Engineering, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mohammad Sherwani
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Ara Alexandrian
- Medical Physics, Mary Bird Perkins Cancer Center, Baton Rouge, LA
- Health Physics Program, Department of Clinical & Diagnostic Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL
| | - Anna Sorace
- Department of Radiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
- Department of Biomedical Engineering, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Graduate Biomedical Sciences, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
- Center of Clinical and Translational Sciences, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
- O’Neal Comprehensive Cancer center, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Amy Brady
- Nuclear Medicine and Molecular Imaging Sciences Program, Department of Clinical & Diagnostic Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL
| | - Remo George
- Nuclear Medicine and Molecular Imaging Sciences Program, Department of Clinical & Diagnostic Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL
- Graduate Biomedical Sciences, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
- Center of Clinical and Translational Sciences, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
- O’Neal Comprehensive Cancer center, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
6
|
Rankin S, Fountain C, Gemmell AJ, Quinn D, Henderson A, McClure J, Small S, Venugopal B, McKay P, Slomka PJ, Colville D, Petrie MC, Meléndez GC, Lang NN. Arterial effects of anthracycline: structural and inflammatory assessments in non-human primates and lymphoma patients. Clin Sci (Lond) 2025; 139:29-41. [PMID: 39680089 DOI: 10.1042/cs20241529] [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: 07/29/2024] [Revised: 12/04/2024] [Accepted: 12/16/2024] [Indexed: 12/17/2024]
Abstract
Anthracyclines, such as doxorubicin, are important anti-cancer therapies but are associated with arterial injury. Histopathological insights have been limited to small animal models, and the role of inflammation in the arterial toxic effects of anthracycline is unclear in humans. Our aims were (1) to evaluate aortic media fibrosis and injury in non-human primates treated with anthracyclines; (2) to assess the effect of anthracycline on aortic inflammation in patients treated for lymphoma. African Green monkeys (AGMs) received doxorubicin (30-60 mg/m2/biweekly intravenously, cumulative dose: 240 mg/m2). Blinded histopathologic analyses of the ascending aorta were performed 15 weeks after the last doxorubicin dose and compared to five age- and gender-matched healthy, untreated AGMs. Analysis of the thoracic aorta of patients with diffuse large B-cell lymphoma (DLBCL), at baseline and after doxorubicin exposure, was performed using 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) in this observational study by maximal tissue-to-background ratio (TBRmax). In AGMs, doxorubicin exposure was associated with greater aortic fibrosis (collagen deposition: doxorubicin 6.23 ± 0.88% vs. controls 4.67 ± 0.54%; P=0.01) and intracellular vacuolization (doxorubicin 66.3 ± 10.1 vs. controls 11.5 ± 4.2 vacuoles/field, P<0.0001) than untreated controls. In 101 patients with DLBCL, there was no change in aortic TBRmax after anthracycline exposure (TBRmax 1.46 ± 0.16 vs. 1.44 ± 0.14, respectively, P=0.14). Univariate analyses yielded similar results. In a large animal model, anthracycline exposure was associated with aortic fibrosis. In patients with lymphoma, anthracycline exposure was not associated with aortic inflammation. Further research is required to elucidate the mechanisms of anthracycline-related vascular harm.
Collapse
Affiliation(s)
- Stephen Rankin
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, U.K
| | - Caitlin Fountain
- Departments of Internal Medicine, Section on Cardiology and Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, U.S.A
| | - Alastair J Gemmell
- Department of Clinical Physics & Bioengineering, NHS Greater Glasgow & Clyde, Glasgow
- School of Medicine, Dentistry and Nursing, University of Glasgow
| | - Daire Quinn
- The Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Alasdair Henderson
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, U.K
| | - John McClure
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, U.K
| | - Sandy Small
- Department of Clinical Physics & Bioengineering, NHS Greater Glasgow & Clyde, Glasgow
- School of Medicine, Dentistry and Nursing, University of Glasgow
| | - Balaji Venugopal
- School of Medicine, Dentistry and Nursing, University of Glasgow
- The Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Pamela McKay
- The Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Piotr J Slomka
- Cedars-Sinai, Division of Artificial Intelligence in Medicine, Department of Medicine, Los Angeles, U.S.A
| | - David Colville
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, U.K
- School of Medicine, Dentistry and Nursing, University of Glasgow
| | - Mark C Petrie
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, U.K
| | - Giselle C Meléndez
- Departments of Internal Medicine, Section on Cardiology and Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, U.S.A
| | - Ninian N Lang
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, U.K
| |
Collapse
|
7
|
Zahergivar A, Golagha M, Stoddard G, Anderson PS, Woods L, Newman A, Carter MR, Wang L, Ibrahim M, Chamberlin J, Auffermann WF, Kabakus I, Burt JR. Prognostic value of coronary artery calcium scoring in patients with non-small cell lung cancer using initial staging computed tomography. BMC Med Imaging 2024; 24:350. [PMID: 39731094 DOI: 10.1186/s12880-024-01544-6] [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: 07/11/2024] [Accepted: 12/19/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND Lung cancer is a leading cause of cancer-related mortality worldwide, with non-small cell lung cancer (NSCLC) comprising 85% of cases. Due to the lack of early clinical signs, metastasis often occurs before diagnosis, impacting treatment and prognosis. Cardiovascular disease (CVD) is a common comorbidity in lung cancer patients, with shared risk factors exacerbating outcomes. METHODS This study investigates the association between coronary artery calcium (CAC) scores, major adverse cardiovascular events (MACE), and survival outcomes in NSCLC patients, utilizing positron emission tomography-computed tomography (PET-CT) for CAC scoring. A retrospective cohort study of 154 NSCLC patients (mean age 66.3 years, 52% women) at the University of Utah (2005-2022) was conducted. Baseline PET-CT or CT imaging was used to quantify CAC scores, categorized into five risk levels. Cox proportional hazards and logistic regression analyses assessed the impact of CAC scores on survival and cardiovascular events, adjusting for confounders such as age, gender, and smoking status. RESULTS Higher CAC scores were significantly associated with increased MACE, acute myocardial infarction (MI), and poorer overall survival. The severe risk CAC score group had significantly lower survival (p = 0.022). Logistic regression revealed a strong association between higher CAC scores and MI incidence (moderate: OR = 13.8, severe: OR = 21.2) and MACE (severe: OR = 10.2). Smoking history was a significant predictor of overall survival (p = 0.006). CONCLUSION CAC scoring via PET-CT provides valuable prognostic insights in NSCLC patients, highlighting the need for integrated cardiovascular risk management in this population. Further research and advanced technologies like machine learning could enhance CAC scoring application in clinical practice. TRIAL REGISTRATION Retrospectively registered.
Collapse
Affiliation(s)
- Aryan Zahergivar
- Internal Medicine Department, District of Columbia, George Washington University, District of Columbia, Washington, USA
| | - Mahshid Golagha
- Urology Oncology Branch, National Cancer Institutes, National Institutes of Health, Bethesda, MD, USA
| | - Greg Stoddard
- Biomedical Informatics, University of Utah, Salt Lake City, Utah, USA
| | - Parker Sage Anderson
- Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Lacey Woods
- Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Anna Newman
- Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Malorie R Carter
- Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Libo Wang
- Department of Internal Medicine, Division of Cardiology, University of Utah, Salt Lake City, Utah, USA
| | - Mark Ibrahim
- Department of Internal Medicine, Division of Cardiology, University of Utah, Salt Lake City, Utah, USA
| | - Jordan Chamberlin
- Department of Radiology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - William F Auffermann
- Department of Radiology, Cardiothoracic Imaging, University of Utah, 30 N 1900 E #1A71, Salt Lake City, Utah, 84132, USA
| | - Ismail Kabakus
- Department of Radiology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jeremy R Burt
- Department of Radiology, Cardiothoracic Imaging, University of Utah, 30 N 1900 E #1A71, Salt Lake City, Utah, 84132, USA.
| |
Collapse
|
8
|
Sartin SL, Shetty DR, Strange CD, Gayer G, Ahuja J, Agrawal R, Truong MT. Pitfalls in Positron Emission Tomography/Computed Tomography in the Thorax and Abdomen. Semin Ultrasound CT MR 2024; 45:488-495. [PMID: 39069275 DOI: 10.1053/j.sult.2024.07.012] [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: 07/30/2024]
Abstract
Positron emission tomography/computed tomography (PET/CT) using [18F]-fluoro-2-deoxy-D-glucose (FDG) has become the mainstay imaging modality for evaluating oncology patients with certain cancers. The most common FDG PET/CT applications include staging/restaging, assessing response to therapy and detecting tumor recurrence. It is important to be aware of potential pitfalls and technical artifacts on PET/CT in the chest and abdomen to ensure accurate interpretation, avoid unnecessary intervention and optimize patient care.
Collapse
Affiliation(s)
- Stephen L Sartin
- Department of Thoracic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Dhanwin R Shetty
- Department of Thoracic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Chad D Strange
- Department of Thoracic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gabriela Gayer
- Department of Radiology, Beth Israel Deaconess Medical Center, Boston, MA
| | - Jitesh Ahuja
- Department of Thoracic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Rishi Agrawal
- Department of Thoracic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Mylene T Truong
- Department of Thoracic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
9
|
Cau R, Anzalone N, Mannelli L, Edjlali M, Balestrieri A, Nardi V, Lanzino G, Lerman A, Suri JS, Saba L. Pericarotid Fat as a Marker of Cerebrovascular Risk. AJNR Am J Neuroradiol 2024; 45:1635-1641. [PMID: 39147585 PMCID: PMC11543090 DOI: 10.3174/ajnr.a8300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/27/2024] [Indexed: 08/17/2024]
Abstract
Vascular inflammation is widely recognized as an important factor in the atherosclerotic process, particularly in terms of plaque development and progression. Conventional tests, such as measuring circulating inflammatory biomarkers, lack the precision to identify specific areas of vascular inflammation. In this context, noninvasive imaging modalities can detect perivascular fat changes, serving as a marker of vascular inflammation. This review aims to provide a comprehensive overview of the key concepts related to perivascular carotid fat and its pathophysiology. Additionally, we examine the existing literature on the association of pericarotid fat with features of plaque vulnerability and cerebrovascular events. Finally, we scrutinize the advantages and limitations of the noninvasive assessment of pericarotid fat.
Collapse
Affiliation(s)
- Riccardo Cau
- From the Department of Radiology (R.C., A.B., L.S.), Azienda Ospedaliero Universitaria (A.O.U.), Cagliari, Italy
| | - Nicoletta Anzalone
- Vita-Salute San Raffaele University (N.A.), Milan, Italy
- Neuroradiology Unit and CERMAC (N.A.), IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Myriam Edjlali
- Department of Neuroradiology (M.E.), Université Paris-Descartes-Sorbonne-Paris-Cité, IMABRAIN-INSERM-UMR1266, DHU-Neurovasc, Centre Hospitalier Sainte-Anne, Paris, France
| | - Antonella Balestrieri
- From the Department of Radiology (R.C., A.B., L.S.), Azienda Ospedaliero Universitaria (A.O.U.), Cagliari, Italy
| | - Valentina Nardi
- Department of Neurosurgery (V.N., G.L.), Mayo Clinic, Rochester, Minnesota
| | - Giuseppe Lanzino
- Department of Neurosurgery (V.N., G.L.), Mayo Clinic, Rochester, Minnesota
| | - Amir Lerman
- Department of Cardiovascular Medicine (A.L.), Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Jasjit S Suri
- Stroke Monitoring and Diagnostic Division (J.S.S.), AtheroPoint, Roseville, California
| | - Luca Saba
- From the Department of Radiology (R.C., A.B., L.S.), Azienda Ospedaliero Universitaria (A.O.U.), Cagliari, Italy
| |
Collapse
|
10
|
Wang Z, Zhang P. Novel imaging modalities for the identification of vulnerable plaques. Front Cardiovasc Med 2024; 11:1450252. [PMID: 39328242 PMCID: PMC11424440 DOI: 10.3389/fcvm.2024.1450252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
Atherosclerosis is a slow, progressive disease that is closely associated with major adverse cardiovascular events. Early diagnosis and risk assessment of atherosclerosis can effectively improve the prognosis and reduce the occurrence of adverse cardiovascular events in the later stage. A variety of invasive and non-invasive imaging modalities are important tools for diagnosing lesions, monitoring the efficacy of treatments, and predicting associated risk events. This review mainly introduces the four commonly used non-invasive imaging modalities in clinical practice and intravascular imaging such as optical coherence tomography, intravascular ultrasound imaging, and near-infrared spectroscopy, compares the advantages and disadvantages in the diagnosis of vulnerable plaques, and briefly summarizes the new progressions of each.
Collapse
Affiliation(s)
- Ziyan Wang
- Department of Cardiovascular Ultrasound, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Pingyang Zhang
- Department of Cardiovascular Ultrasound, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
11
|
Gao M, Wen W, Li H, Zheng Y, Yun M, Meng J, Wang S, Wang B, Hu B, Mou T, Yu Y, Zhang X, Li X. Coronary sodium [ 18F]fluoride activity predicts outcomes post-CABG: a comparative evaluation with conventional metrics. Eur J Nucl Med Mol Imaging 2024; 51:3235-3251. [PMID: 38730084 DOI: 10.1007/s00259-024-06736-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/26/2024] [Indexed: 05/12/2024]
Abstract
PURPOSE The value of preoperative multidisciplinary approach remains inadequately delineated in forecasting postoperative outcomes of patients undergoing coronary artery bypass grafting (CABG). Herein, we aimed to ascertain the efficacy of multi-modality cardiac imaging in predicting post-CABG cardiovascular outcomes. METHODS Patients with triple coronary artery disease underwent cardiac sodium [18F]fluoride ([18F]NaF) positron emission tomography/computed tomography (PET/CT), coronary angiography, and CT-based coronary artery calcium scoring before CABG. The maximum coronary [18F]NaF activity (target-to-blood ratio [TBR]max) and the global coronary [18F]NaF activity (TBRglobal) was determined. The primary endpoint was perioperative myocardial infarction (PMI) within 7-day post-CABG. Secondary endpoint included major adverse cardiac and cerebrovascular events (MACCEs) and recurrent angina. RESULTS This prospective observational study examined 101 patients for a median of 40 months (interquartile range: 19-47 months). Both TBRmax (odds ratio [OR] = 1.445; p = 0.011) and TBRglobal (OR = 1.797; P = 0.018) were significant predictors of PMI. TBRmax>3.0 (area under the curve [AUC], 0.65; sensitivity, 75.0%; specificity, 56.8%; p = 0.036) increased PMI risk by 3.661-fold, independent of external confounders. Kaplan-Meier test revealed a decrease in MACCE survival rate concomitant with an escalating TBRmax. TBRmax>3.6 (AUC, 0.70; sensitivity, 76.9%; specificity, 73.9%; p = 0.017) increased MACCEs risk by 5.520-fold. Both TBRmax (hazard ratio [HR], 1.298; p = 0.004) and TBRglobal (HR = 1.335; p = 0.011) were significantly correlated with recurrent angina. No significant associations were found between CAC and SYNTAX scores and between PMI occurrence and long-term MACCEs. CONCLUSION Quantification of coronary microcalcification activity via [18F]NaF PET displayed a strong ability to predict early and long-term post-CABG cardiovascular outcomes, thereby outperforming conventional metrics of coronary macrocalcification burden and stenosis severity. TRIAL REGISTRATION The trial was registered with the Chinese Clinical Trial Committee (number: ChiCTR1900022527; URL: www.chictr.org.cn/showproj.html?proj=37933 ).
Collapse
Affiliation(s)
- Mingxin Gao
- Department of Nuclear Medicine, Molecular Imaging Lab, Beijing Anzhen Hospital, Capital Medical University, No.2, Anzhen Road, Chaoyang District, Beijing, 10029, China
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Wanwan Wen
- Department of Nuclear Medicine, Molecular Imaging Lab, Beijing Anzhen Hospital, Capital Medical University, No.2, Anzhen Road, Chaoyang District, Beijing, 10029, China
| | - Haiyang Li
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Yaqi Zheng
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Mingkai Yun
- Department of Nuclear Medicine, Molecular Imaging Lab, Beijing Anzhen Hospital, Capital Medical University, No.2, Anzhen Road, Chaoyang District, Beijing, 10029, China
| | - Jingjing Meng
- Department of Nuclear Medicine, Molecular Imaging Lab, Beijing Anzhen Hospital, Capital Medical University, No.2, Anzhen Road, Chaoyang District, Beijing, 10029, China
| | - Shipan Wang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Bolin Wang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Biao Hu
- Department of Nuclear Medicine, Molecular Imaging Lab, Beijing Anzhen Hospital, Capital Medical University, No.2, Anzhen Road, Chaoyang District, Beijing, 10029, China
| | - Tiantian Mou
- Department of Nuclear Medicine, Molecular Imaging Lab, Beijing Anzhen Hospital, Capital Medical University, No.2, Anzhen Road, Chaoyang District, Beijing, 10029, China
| | - Yang Yu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2, Anzhen Road, Chaoyang District, Beijing, 100029, China.
| | - Xiaoli Zhang
- Department of Nuclear Medicine, Molecular Imaging Lab, Beijing Anzhen Hospital, Capital Medical University, No.2, Anzhen Road, Chaoyang District, Beijing, 10029, China.
| | - Xiang Li
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Vienna General Hospital, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
12
|
Wang X, Liu X, Wu W, Liao L, Zhou M, Wang X, Tan Z, Zhang G, Bai Y, Li X, Zhao M. Hypoxia activates macrophage-NLRP3 inflammasome promoting atherosclerosis via PFKFB3-driven glycolysis. FASEB J 2024; 38:e23854. [PMID: 39096131 DOI: 10.1096/fj.202400283r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/01/2024] [Accepted: 07/21/2024] [Indexed: 08/04/2024]
Abstract
The onset and progression of atherosclerosis are closely linked to the involvement of macrophages. While the contribution of NLRP3 inflammasome activation to the creation of a local highly inflammatory microenvironment is well recognized, the precise triggers remain unclear. In this study, we aimed to investigate the regulatory mechanism of NLRP3 inflammasome activation in response to hypoxia-induced glycolysis involving PFKFB3 in the development of atherosclerosis. To develop an atherosclerosis model, we selected ApoE knockout mice treated with a high-fat western diet. We then quantified the expression of HIF-1α, PFKFB3, and NLRP3. In addition, we administered the PFKFB3 inhibitor PFK158 during atherosclerosis modeling. The glycolytic activity was subsequently determined through 18F-FDG micro-PET/CT, ex vivo glucose uptake, and ECAR analysis. Furthermore, we employed lipopolysaccharide (LPS) and TNF-α to induce the differentiation of bone marrow-derived macrophages (BMDMs) into M1-like phenotypes under both hypoxic and normoxic conditions. Our histological analyses revealed the accumulation of PFKFB3 in human atherosclerotic plaques, demonstrating colocalization with NLRP3 expression and macrophages. Treatment with PFK158 reduced glycolytic activity and NLRP3 inflammasome activation, thereby mitigating the occurrence of atherosclerosis. Mechanistically, hypoxia promoted glycolytic reprogramming and NLRP3 inflammasome activation in BMDMs. Subsequent blocking of either HIF-1α or PFKFB3 downregulated the NLRP3/Caspase-1/IL-1β pathway in hypoxic BMDMs. Our study demonstrated that the HIF-1α/PFKFB3/NLRP3 axis serves as a crucial mechanism for macrophage inflammation activation in the emergence of atherosclerosis. The therapeutic potential of PFKFB3 inhibition may represent a promising strategy for atheroprotection.
Collapse
Affiliation(s)
- Xuan Wang
- Department of Nuclear Medicine, The Third Xiangya Hospital, Central South University, Changsha, P.R. China
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Xiangbin Liu
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Wanzhou Wu
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Longshen Liao
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Ming Zhou
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Xiaobo Wang
- Department of Nuclear Medicine, The Third Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Zeming Tan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Guogang Zhang
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Yongping Bai
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Xiang Li
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Vienna General Hospital, Medical University of Vienna, Vienna, Austria
| | - Min Zhao
- Department of Nuclear Medicine, The Third Xiangya Hospital, Central South University, Changsha, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, P.R. China
| |
Collapse
|
13
|
Dai Y, Junho CVC, Schieren L, Wollenhaupt J, Sluimer JC, van der Vorst EPC, Noels H. Cellular metabolism changes in atherosclerosis and the impact of comorbidities. Front Cell Dev Biol 2024; 12:1446964. [PMID: 39188527 PMCID: PMC11345199 DOI: 10.3389/fcell.2024.1446964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 07/17/2024] [Indexed: 08/28/2024] Open
Abstract
Cell activation and nutrient dysregulation are common consequences of atherosclerosis and its preceding risk factors, such as hypertension, dyslipidemia, and diabetes. These diseases may also impact cellular metabolism and consequently cell function, and the other way around, altered cellular metabolism can impact disease development and progression through altered cell function. Understanding the contribution of altered cellular metabolism to atherosclerosis and how cellular metabolism may be altered by co-morbidities and atherosclerosis risk factors could support the development of novel strategies to lower the risk of CVD. Therefore, we briefly review disease pathogenesis and the principles of cell metabolic pathways, before detailing changes in cellular metabolism in the context of atherosclerosis and comorbidities. In the hypoxic, inflammatory and hyperlipidemic milieu of the atherosclerotic plaque riddled with oxidative stress, metabolism shifts to increase anaerobic glycolysis, the pentose-phosphate pathway and amino acid use. We elaborate on metabolic changes for macrophages, neutrophils, vascular endothelial cells, vascular smooth muscle cells and lymphocytes in the context of atherosclerosis and its co-morbidities hypertension, dyslipidemia, and diabetes. Since causal relationships of specific key genes in a metabolic pathway can be cell type-specific and comorbidity-dependent, the impact of cell-specific metabolic changes must be thoroughly explored in vivo, with a focus on also systemic effects. When cell-specific treatments become feasible, this information will be crucial for determining the best metabolic intervention to improve atherosclerosis and its interplay with co-morbidities.
Collapse
Affiliation(s)
- Yusang Dai
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital, RWTH Aachen University, Aachen, Germany
- Physical Examination Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Carolina Victoria Cruz Junho
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital, RWTH Aachen University, Aachen, Germany
| | - Luisa Schieren
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital, RWTH Aachen University, Aachen, Germany
| | - Julia Wollenhaupt
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital, RWTH Aachen University, Aachen, Germany
| | - Judith C. Sluimer
- Department of Nephrology and Clinical Immunology, University Hospital RWTH Aachen, Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands
| | - Emiel P. C. van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital, RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for Cardiorenal Disease (AMICARE), RWTH Aachen Campus, Aachen, Germany
- Interdisciplinary Centre for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital, RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for Cardiorenal Disease (AMICARE), RWTH Aachen Campus, Aachen, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands
| |
Collapse
|
14
|
Nicholls SJ. Serial PET imaging to evaluate medical therapies: Is it ready for prime time? Atherosclerosis 2024; 395:117517. [PMID: 38582638 DOI: 10.1016/j.atherosclerosis.2024.117517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 04/08/2024]
Affiliation(s)
- Stephen J Nicholls
- Victorian Heart Institute, Monash University, Melbourne, 631 Blackburn Road, Clayton, VIC, 3168, Australia.
| |
Collapse
|
15
|
Rankin S, Fountain C, Gemmell AJ, Quinn D, Henderson A, McClure J, Small S, Venugopal B, McKay P, Slomka PJ, Colville D, Petrie MC, Meléndez GC, Lang NN. Arterial effects of anthracycline: structural and inflammatory assessments in non-human primates and lymphoma patients using 18F-FDG positron emission tomography. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.30.596741. [PMID: 38895275 PMCID: PMC11185566 DOI: 10.1101/2024.05.30.596741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Background Anthracyclines, such as doxorubicin, are important anti-cancer therapies but are associated with arterial injury. Histopathological insights have been limited to small animal models and the role of inflammation in the arterial toxic effects of anthracycline is unclear in humans. Our aims were: 1) To evaluate aortic media fibrosis and injury in non-human primates treated with anthracyclines; 2) To assess the effect of anthracycline on aortic inflammation in patients treated for lymphoma. Methods 1) African Green monkeys (AGM) received doxorubicin (30-60 mg/m2/biweekly IV, cumulative dose: 240 mg/m2). Blinded histopathologic analyses of collagen deposition and cell vacuolization in the ascending aorta were performed 15 weeks after the last doxorubicin dose and compared to 5 age- and gender-matched healthy, untreated AGMs. 2) Analysis of the thoracic aorta of patients with diffuse large B-cell lymphoma (DLBCL), at baseline and after doxorubicin exposure, was performed using 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) in this observational study. The primary outcome was change in maximal tissue-to-background ratio (TBRmax) of the thoracic aorta from baseline to their end-of-treatment clinical PET/CT. Results In AGMs, doxorubicin exposure was associated with greater aortic fibrosis (collagen deposition: doxorubicin cohort 6.23±0.88% vs. controls 4.67±0.54%; p=0.01) and increased intracellular vacuolization (doxorubicin 66.3 ± 10.1 vs controls 11.5 ± 4.2 vacuoles/field, p<0.0001) than untreated controls.In 101 patients with DLBCL, there was no change in aortic TBRmax after anthracycline exposure (pre-doxorubicin TBRmax 1.46±0.16 vs post-doxorubicin TBRmax 1.44±0.14, p=0.14). The absence of change in TBRmax was consistent across all univariate analyses. Conclusions In a large animal model, anthracycline exposure was associated with aortic fibrosis. In patients with lymphoma, anthracycline exposure was not associated with aortic inflammation.Further research is required to elucidate the mechanisms of anthracycline-related vascular harm.
Collapse
Affiliation(s)
- Stephen Rankin
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow UK
| | - Caitlin Fountain
- Departments of Internal Medicine, Section on Cardiology and Pathology, Section on Comparative Medicine. Wake Forest University School of Medicine, Winston-Salem, USA
| | - Alastair J Gemmell
- Department of Clinical Physics & Bioengineering, NHS Greater Glasgow & Clyde, Glasgow
- School of Medicine, Dentistry and Nursing, University of Glasgow
| | - Daire Quinn
- The Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Alasdair Henderson
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow UK
| | - John McClure
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow UK
| | - Sandy Small
- Department of Clinical Physics & Bioengineering, NHS Greater Glasgow & Clyde, Glasgow
- School of Medicine, Dentistry and Nursing, University of Glasgow
| | - Balaji Venugopal
- The Beatson West of Scotland Cancer Centre, Glasgow, UK
- School of Medicine, Dentistry and Nursing, University of Glasgow
| | - Pamela McKay
- The Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Piotr J Slomka
- Cedars-Sinai, Division of Artificial Intelligence in Medicine, Department of Medicine, Los Angeles, USA
| | - David Colville
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow UK
- School of Medicine, Dentistry and Nursing, University of Glasgow
| | - Mark C Petrie
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow UK
| | - Giselle C. Meléndez
- Departments of Internal Medicine, Section on Cardiology and Pathology, Section on Comparative Medicine. Wake Forest University School of Medicine, Winston-Salem, USA
| | - Ninian N Lang
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow UK
| |
Collapse
|
16
|
Ganizada BH, Veltrop RJA, Akbulut AC, Koenen RR, Accord R, Lorusso R, Maessen JG, Reesink K, Bidar E, Schurgers LJ. Unveiling cellular and molecular aspects of ascending thoracic aortic aneurysms and dissections. Basic Res Cardiol 2024; 119:371-395. [PMID: 38700707 PMCID: PMC11143007 DOI: 10.1007/s00395-024-01053-1] [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: 02/05/2024] [Revised: 04/03/2024] [Accepted: 04/26/2024] [Indexed: 06/01/2024]
Abstract
Ascending thoracic aortic aneurysm (ATAA) remains a significant medical concern, with its asymptomatic nature posing diagnostic and monitoring challenges, thereby increasing the risk of aortic wall dissection and rupture. Current management of aortic repair relies on an aortic diameter threshold. However, this approach underestimates the complexity of aortic wall disease due to important knowledge gaps in understanding its underlying pathologic mechanisms.Since traditional risk factors cannot explain the initiation and progression of ATAA leading to dissection, local vascular factors such as extracellular matrix (ECM) and vascular smooth muscle cells (VSMCs) might harbor targets for early diagnosis and intervention. Derived from diverse embryonic lineages, VSMCs exhibit varied responses to genetic abnormalities that regulate their contractility. The transition of VSMCs into different phenotypes is an adaptive response to stress stimuli such as hemodynamic changes resulting from cardiovascular disease, aging, lifestyle, and genetic predisposition. Upon longer exposure to stress stimuli, VSMC phenotypic switching can instigate pathologic remodeling that contributes to the pathogenesis of ATAA.This review aims to illuminate the current understanding of cellular and molecular characteristics associated with ATAA and dissection, emphasizing the need for a more nuanced comprehension of the impaired ECM-VSMC network.
Collapse
MESH Headings
- Humans
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/physiopathology
- Aortic Dissection/pathology
- Aortic Dissection/genetics
- Aortic Dissection/metabolism
- Animals
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/metabolism
- Aorta, Thoracic/pathology
- Aorta, Thoracic/physiopathology
- Vascular Remodeling
- Extracellular Matrix/pathology
- Extracellular Matrix/metabolism
- Phenotype
Collapse
Affiliation(s)
- Berta H Ganizada
- Department of Cardiothoracic Surgery, Heart and Vascular Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands
| | - Rogier J A Veltrop
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands
| | - Asim C Akbulut
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands
| | - Rory R Koenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands
| | - Ryan Accord
- Department of Cardiothoracic Surgery, Center for Congenital Heart Disease, University Medical Center Groningen, Groningen, The Netherlands
| | - Roberto Lorusso
- Department of Cardiothoracic Surgery, Heart and Vascular Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands
| | - Jos G Maessen
- Department of Cardiothoracic Surgery, Heart and Vascular Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands
| | - Koen Reesink
- Department of Biomedical Engineering, Heart and Vascular Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands
| | - Elham Bidar
- Department of Cardiothoracic Surgery, Heart and Vascular Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands
| | - Leon J Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands.
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands.
| |
Collapse
|
17
|
Omery BM, Kalirao AS. Myocardial Infarction and Cardiac Arrest During Acquisition of 18 F-FDG PET/CT. Clin Nucl Med 2024; 49:580-581. [PMID: 38389214 DOI: 10.1097/rlu.0000000000005124] [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: 02/24/2024]
Abstract
ABSTRACT An 83-year-old man with multiple myeloma and history of coronary artery disease and ischemic cardiomyopathy experienced cardiac arrest during the 18 F-FDG PET/CT examination. The patient had not yet been treated for multiple myeloma. The PET/CT demonstrated focal FDG uptake about the left anterior descending coronary artery. Diffuse intense FDG uptake in the liver and less than normal uptake in other organs of usual FDG distribution were also present, likely due to altered hemodynamics and heart failure in the setting of an acute coronary syndrome.
Collapse
Affiliation(s)
- Bilal M Omery
- From the Department of Radiology, Aurora St Luke's Medical Center, Milwaukee, WI
| | | |
Collapse
|
18
|
Guimarães J, de Almeida J, Mendes PL, Ferreira MJ, Gonçalves L. Advancements in non-invasive imaging of atherosclerosis: Future perspectives. J Clin Lipidol 2024; 18:e142-e152. [PMID: 38142178 DOI: 10.1016/j.jacl.2023.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 11/11/2023] [Accepted: 11/14/2023] [Indexed: 12/25/2023]
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the buildup of plaques in arterial walls, leading to cardiovascular diseases and high morbidity and mortality rates worldwide. Non-invasive imaging techniques play a crucial role in evaluating patients with suspected or established atherosclerosis. However, there is a growing body of evidence suggesting the need to visualize the underlying processes of plaque progression and rupture to enhance risk stratification. This review explores recent advancements in non-invasive assessment of atherosclerosis, focusing on computed tomography, magnetic resonance imaging, and nuclear imaging. These advancements provide valuable insights into the assessment and management of atherosclerosis, potentially leading to better risk stratification and improved patient outcomes.
Collapse
Affiliation(s)
- Joana Guimarães
- Cardiology Department, Coimbra's Hospital and University Center, Praceta Mota Pinto, 3000-561 Coimbra, Portugal.
| | - José de Almeida
- Cardiology Department, Coimbra's Hospital and University Center, Praceta Mota Pinto, 3000-561 Coimbra, Portugal
| | - Paulo Lázaro Mendes
- Cardiology Department, Coimbra's Hospital and University Center, Praceta Mota Pinto, 3000-561 Coimbra, Portugal
| | - Maria João Ferreira
- Cardiology Department, Coimbra's Hospital and University Center, Praceta Mota Pinto, 3000-561 Coimbra, Portugal; Faculty of Medicine, Coimbra's University, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Lino Gonçalves
- Cardiology Department, Coimbra's Hospital and University Center, Praceta Mota Pinto, 3000-561 Coimbra, Portugal; Faculty of Medicine, Coimbra's University, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| |
Collapse
|
19
|
Chen LH, Spagnolo-Allende A, Yang D, Qiao Y, Gutierrez J. Epidemiology, Pathophysiology, and Imaging of Atherosclerotic Intracranial Disease. Stroke 2024; 55:311-323. [PMID: 38252756 PMCID: PMC10827355 DOI: 10.1161/strokeaha.123.043630] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Intracranial atherosclerotic disease (ICAD) is one of the most common causes of stroke worldwide. Among people with stroke, those of East Asia descent and non-White populations in the United States have a higher burden of ICAD-related stroke compared with Whites of European descent. Disparities in the prevalence of asymptomatic ICAD are less marked than with symptomatic ICAD. In addition to stroke, ICAD increases the risk of dementia and cognitive decline, magnifying ICAD societal burden. The risk of stroke recurrence among patients with ICAD-related stroke is the highest among those with confirmed stroke and stenosis ≥70%. In fact, the 1-year recurrent stroke rate of >20% among those with stenosis >70% is one of the highest rates among common causes of stroke. The mechanisms by which ICAD causes stroke include plaque rupture with in situ thrombosis and occlusion or artery-to-artery embolization, hemodynamic injury, and branch occlusive disease. The risk of stroke recurrence varies by the presumed underlying mechanism of stroke, but whether techniques such as quantitative magnetic resonance angiography, computed tomographic angiography, magnetic resonance perfusion, or transcranial Doppler can help with risk stratification beyond the degree of stenosis is less clear. The diagnosis of ICAD is heavily reliant on lumen-based studies, such as computed tomographic angiography, magnetic resonance angiography, or digital subtraction angiography, but newer technologies, such as high-resolution vessel wall magnetic resonance imaging, can help distinguish ICAD from stenosing arteriopathies.
Collapse
Affiliation(s)
- Li Hui Chen
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Antonio Spagnolo-Allende
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Dixon Yang
- Department of Neurology, Rush University, Chicago, IL, USA
| | - Ye Qiao
- Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
| | - Jose Gutierrez
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| |
Collapse
|
20
|
Ryu J, Han SA, Han S, Choi S, Moon DH, Oh M. Comparison of SUV A/V and SUV A-V for Evaluating Atherosclerotic Inflammation in 18F-FDG PET/CT. Nucl Med Mol Imaging 2024; 58:25-31. [PMID: 38261882 PMCID: PMC10796899 DOI: 10.1007/s13139-023-00822-5] [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: 05/26/2023] [Revised: 08/03/2023] [Accepted: 08/23/2023] [Indexed: 01/25/2024] Open
Abstract
Purpose This study aimed to compare the clinical significance of two parameters, division of standardized uptake value (SUV) of target arterial activity by background venous blood pool activity (SUVA/V) and subtraction of background venous blood pool activity from SUV of target arterial activity (SUVA-V) of carotid arteries with atherosclerotic plaques using 18F-fluorodeoxyglucose (FDG) positron emission tomography and computed tomography (PET/CT). Methods Patients aged 50 years or more who were diagnosed with carotid artery stenosis of 50% or more with carotid Doppler ultrasonography and had torso 18F-FDG PET/CT were enrolled retrospectively and classified patients who developed cerebrovascular events (CVEs) within 5 years after 18F-FDG PET/CT scan as the active group and patients who did not experience the CVE within 5 years as an inactive group. We calculated SUVA/V and SUVA-V using measurements of SUVmax of carotid arteries and mean SUV of superior vena cava (SVC). Results SUVA-V, SUVA-V_high, and SUVA-V_low were significantly higher in the active group than in the inactive group, but neither SUVA/V, SUVA/V_high, nor SUVA/V_low showed significant differences between the active and inactive groups. The difference in rank between groups of SUVA/V_high and SUVA/V_low was greater than the difference in rank between groups of SUVA-V_high and SUVA-V_low. The CVE incidence differed between SUVA/V_high and SUVA/V_low of high carotid FDG uptake, but the CVE incidence did not differ between SUVA-V_high and SUVA-V_low of high carotid FDG uptake. Conclusion SUVA-V may be a more rational solution than SUVA/V for evaluating atherosclerotic plaque inflammation on 18F-FDG PET/CT.
Collapse
Affiliation(s)
- Jeongryul Ryu
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505 Republic of Korea
| | - Shin Ae Han
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505 Republic of Korea
| | - Sangwon Han
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505 Republic of Korea
| | - Sunju Choi
- Department of Nuclear Medicine, Kyung Hee University School of Medicine, Seoul, Republic of Korea
| | - Dae Hyuk Moon
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505 Republic of Korea
| | - Minyoung Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505 Republic of Korea
| |
Collapse
|
21
|
Chlorogiannis DD, Pargaonkar S, Papanagiotou P, Bakogiannis NC, Bakoyiannis C, Kokkinidis DG. Inflammation, anti-inflammatory agents, and the role of colchicine in carotid artery stenosis. VASA 2024; 53:4-12. [PMID: 38079179 DOI: 10.1024/0301-1526/a001104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Cardiovascular disease is a major cause of morbidity and mortality worldwide. In the last few years, the role of inflammation and inflammatory modulatory medications is investigated for the optimal treatment of coronary artery disease. It can be hypothesized that since inflammation is also involved in carotid artery stenosis development and progression, the same class of medication could be useful. Our objective with this review is to present the available evidence, published studies and promising ongoing trials on the role of anti-inflammatory medications - with a special emphasis on the most commonly used drug of this class: colchicine - in patients with carotid artery stenosis.
Collapse
Affiliation(s)
| | - Sumant Pargaonkar
- Department of Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, New York Ciry, NY, USA
| | - Panagiotis Papanagiotou
- First Department of Radiology, School of Medicine, National & Kapodistrian University of Athens, Areteion Hospital, Athens, Greece
- Department of Diagnostic and Interventional Neuroradiology, Hospital Bremen-Mitte/Bremen-Ost, Bremen, Germany
| | - Nikolaos C Bakogiannis
- Division of Vascular Surgery, Laiko General Hospital/University of Athens School of Medicine, Athens, Greece
| | - Christos Bakoyiannis
- Division of Vascular Surgery, Laiko General Hospital/University of Athens School of Medicine, Athens, Greece
| | - Damianos G Kokkinidis
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
22
|
Khare HA, Binderup T, Hag AMF, Kjaer A. Longitudinal imaging of murine atherosclerosis with 2-deoxy-2-[ 18F]fluoro-D-glucose and [ 18F]-sodium fluoride in genetically modified Apolipoprotein E knock-out and wild type mice. Sci Rep 2023; 13:22983. [PMID: 38151517 PMCID: PMC10752895 DOI: 10.1038/s41598-023-49585-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 12/09/2023] [Indexed: 12/29/2023] Open
Abstract
In a longitudinal design, four arterial segments in mice were followed by positron emission tomography/computed tomography (PET/CT) imaging. We aimed to determine how the tracers reflected the development of atherosclerosis via the uptake of 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) for imaging inflammation and [18F]-sodium fluoride (Na[18F]F) for imaging active microcalcification in a murine model of atherosclerosis. Apolipoprotein E knock-out (ApoE) mice and C57 BL/6NtaC (B6) mice were divided into four groups. They received either normal chow (N = 7, ApoE mice and N = 6, B6 mice) for 32 weeks or a high-fat diet (N = 6, ApoEHFD mice and N = 9, B6HFD mice) for 32 weeks. The mice were scanned with [18F]FDG and Na[18F]F using a dedicated small animal PET/CT scanner at three timepoints. The tracer uptakes in four aortic segments (abdominal aorta, aortic arch, ascending aorta, and thoracic aorta) were measured and reported as SUVmax values. The uptake of [18F]FDG (SUVmax: 5.7 ± 0.5 vs 1.9 ± 0.2, 230.3%, p = < 0.0001) and Na[18F]F (SUVmax: 9.6 ± 1.8 vs 4.0 ± 0.3, 175%, p = 0.007) was significantly increased in the abdominal aorta of ApoEHFD mice at Week 32 compared to baseline abdominal aorta values of ApoEHFD mice. [18F]FDG uptake in the aortic arch, ascending aorta and the thoracic aorta of B6HFD mice at Week 32 showed a robust resemblance to the abdominal aorta uptake whereas the Na[18F]F uptake only resembled in the thoracic aorta of B6HFD mice at Week 32 compared to the abdominal aorta. The uptake of both [18F]FDG and Na[18F]F increased as the disease progressed over time, and the abdominal aorta provided a robust measure across mouse strain and diet. Therefore, it seems to be the preferred region for image readout. For [18F]FDG-PET, both B6 and ApoE mice provide valuable information and either mouse strain may be used in preclinical cardiovascular studies, whereas for Na[18F]F -PET, ApoE mice should be preferred.
Collapse
Affiliation(s)
- Harshvardhan A Khare
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Tina Binderup
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne Mette Fisker Hag
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
23
|
Zhang J, Yuan Z, Li X, Wang F, Wei X, Kang Y, Mo C, Jiang J, Liang H, Ye L. Activation of the JNK/COX-2/HIF-1α axis promotes M1 macrophage via glycolytic shift in HIV-1 infection. Life Sci Alliance 2023; 6:e202302148. [PMID: 37798121 PMCID: PMC10556724 DOI: 10.26508/lsa.202302148] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/07/2023] Open
Abstract
Chronic inflammation is recognized as a major risk factor for the severity of HIV infection. Whether metabolism reprogramming of macrophages caused by HIV-1 is related to chronic inflammatory activation, especially M1 polarization of macrophages, is inconclusive. Here, we show that HIV-1 infection induces M1 polarization and enhanced glycolysis in macrophages. Blockade of glycolysis inhibits M1 polarization of macrophages, indicating that HIV-1-induced M1 polarization is supported by enhanced glycolysis. Moreover, we find that this immunometabolic adaptation is dependent on hypoxia-inducible factor 1α (HIF-1α), a strong inducer of glycolysis. HIF-1α-target genes, including HK2, PDK1, and LDHA, are also involved in this process. Further research discovers that COX-2 regulates HIF-1α-dependent glycolysis. However, the elevated expression of COX-2, enhanced glycolysis, and M1 polarization of macrophages could be reversed by inactivation of JNK in the context of HIV-1 infection. Our study mechanistically elucidates that the JNK/COX-2/HIF-1α axis is activated to strengthen glycolysis, thereby promoting M1 polarization in macrophages in HIV-1 infection, providing a new idea for resolving chronic inflammation in clinical AIDS patients.
Collapse
Affiliation(s)
- Junhan Zhang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, China
| | - Zongxiang Yuan
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, China
| | - Xuanrong Li
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, China
| | - Fengyi Wang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, China
| | - Xueqin Wei
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, China
| | - Yiwen Kang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, China
| | - Chuye Mo
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, China
| | - Junjun Jiang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, China
| | - Hao Liang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, China
| | - Li Ye
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, China
| |
Collapse
|
24
|
Blanchard I, Vootukuru N, Bhattaru A, Patil S, Rojulpote C. PET Radiotracers in Atherosclerosis: A Review. Curr Probl Cardiol 2023; 48:101925. [PMID: 37392979 DOI: 10.1016/j.cpcardiol.2023.101925] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 06/27/2023] [Accepted: 06/27/2023] [Indexed: 07/03/2023]
Abstract
Traditional atherosclerosis imaging modalities are limited to late stages of disease, prior to which patients are frequently asymptomatic. Positron emission tomography (PET) imaging allows for the visualization of metabolic processes underscoring disease progression via radioactive tracer, allowing earlier-stage disease to be identified. 2-deoxy-2-[fluorine-18]fluoro-D-glucose (18F-FDG) uptake largely reflects the metabolic activity of macrophages, but is unspecific and limited in its utility. By detecting areas of microcalcification, 18F-Sodium Fluoride (18F-NaF) uptake also provides insight into atherosclerosis pathogenesis. Gallium-68 DOTA-0-Tyr3-Octreotate (68Ga-DOTATATE) PET has also shown potential in identifying vulnerable atherosclerotic plaques with high somatostatin receptor expression. Finally, 11-carbon (11C)-choline and 18F-fluoromethylcholine (FMCH) tracers may identify high-risk atherosclerotic plaques by detecting increased choline metabolism. Together, these radiotracers quantify disease burden, assess treatment efficacy, and stratify risk for adverse cardiac events.
Collapse
Affiliation(s)
| | - Nishita Vootukuru
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ
| | - Abhijit Bhattaru
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ; Department of Radiology, University of Pennsylvania, Philadelphia, PA
| | | | - Chaitanya Rojulpote
- Department of Radiology, University of Pennsylvania, Philadelphia, PA; Department of Medicine, The Wright Center for Graduate Medical Education, Scranton, PA.
| |
Collapse
|
25
|
Hupe J, Worthmann H, Ravenberg KK, Grosse GM, Ernst J, Haverich A, Bengel FM, Weissenborn K, Schmitto JD, Hanke JS, Derlin T, Gabriel MM. Interplay between driveline infection, vessel wall inflammation, cerebrovascular events and mortality in patients with left ventricular assist device. Sci Rep 2023; 13:18552. [PMID: 37899422 PMCID: PMC10613624 DOI: 10.1038/s41598-023-45110-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 10/16/2023] [Indexed: 10/31/2023] Open
Abstract
In patients with left ventricular assist device (LVAD), infections and thrombotic events represent severe complications. We investigated device-specific local and systemic inflammation and its impact on cerebrovascular events (CVE) and mortality. In 118 LVAD patients referred for 18F-FDG-PET/CT, metabolic activity of LVAD components, thoracic aortic wall, lymphoid and hematopoietic organs, was quantified and correlated with clinical characteristics, laboratory findings, and outcome. Driveline infection was detected in 92/118 (78%) patients by 18F-FDG-PET/CT. Activity at the driveline entry site was associated with increased signals in aortic wall (r = 0.32, p < 0.001), spleen (r = 0.20, p = 0.03) and bone marrow (r = 0.20, p = 0.03), indicating systemic interactions. Multivariable analysis revealed independent associations of aortic wall activity with activity of spleen (β = 0.43, 95% CI 0.18-0.68, p < 0.001) and driveline entry site (β = 0.04, 95% CI 0.01-0.06, p = 0.001). Twenty-two (19%) patients suffered CVE after PET/CT. In a binary logistic regression analysis metabolic activity at the driveline entry site missed the level of significance as an influencing factor for CVE after adjusting for anticoagulation (OR = 1.16, 95% CI 1-1.33, p = 0.05). Metabolic activity of the subcutaneous driveline (OR = 1.13, 95% CI 1.02-1.24, p = 0.016) emerged as independent risk factor for mortality. Molecular imaging revealed systemic inflammatory interplay between thoracic aorta, hematopoietic organs, and infected device components in LVAD patients, the latter predicting CVE and mortality.
Collapse
Affiliation(s)
- Juliane Hupe
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Hans Worthmann
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Kim K Ravenberg
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Gerrit M Grosse
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Johanna Ernst
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Axel Haverich
- Department of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Karin Weissenborn
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Jan D Schmitto
- Department of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Jasmin S Hanke
- Department of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Maria M Gabriel
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
26
|
Craig NJ, Dweck MR. Positron emission tomography imaging of inflammation in diabetes mellitus. Cardiovasc Res 2023; 119:1887-1888. [PMID: 37463514 DOI: 10.1093/cvr/cvad109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/09/2023] [Accepted: 06/11/2023] [Indexed: 07/20/2023] Open
Affiliation(s)
- Neil J Craig
- Centre for Cardiovascular Sciences, University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 6SB, UK
| | - Marc R Dweck
- Centre for Cardiovascular Sciences, University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 6SB, UK
| |
Collapse
|
27
|
McCabe JJ, Evans NR, Gorey S, Bhakta S, Rudd JHF, Kelly PJ. Imaging Carotid Plaque Inflammation Using Positron Emission Tomography: Emerging Role in Clinical Stroke Care, Research Applications, and Future Directions. Cells 2023; 12:2073. [PMID: 37626883 PMCID: PMC10453446 DOI: 10.3390/cells12162073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/11/2023] [Accepted: 08/13/2023] [Indexed: 08/27/2023] Open
Abstract
Atherosclerosis is a chronic systemic inflammatory condition of the vasculature and a leading cause of stroke. Luminal stenosis severity is an important factor in determining vascular risk. Conventional imaging modalities, such as angiography or duplex ultrasonography, are used to quantify stenosis severity and inform clinical care but provide limited information on plaque biology. Inflammatory processes are central to atherosclerotic plaque progression and destabilization. 18F-fluorodeoxyglucose (FDG) positron emission tomography (PET) is a validated technique for quantifying plaque inflammation. In this review, we discuss the evolution of FDG-PET as an imaging modality to quantify plaque vulnerability, challenges in standardization of image acquisition and analysis, its potential application to routine clinical care after stroke, and the possible role it will play in future drug discovery.
Collapse
Affiliation(s)
- John J. McCabe
- Health Research Board Stroke Clinical Trials Network Ireland, Catherine McAuley Centre, Nelson Street, D07 KX5K Dublin, Ireland; (S.G.); (P.J.K.)
- Neurovascular Unit for Applied Translational and Therapeutics Research, Catherine McAuley Centre, Nelson Street, D07 KX5K Dublin, Ireland
- School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
- Stroke Service, Department of Medicine for the Elderly, Mater Misericordiae University Hospital, Eccles Street, D07 R2WY Dublin, Ireland
| | - Nicholas R. Evans
- Department of Clinical Neurosciences, Box 83, Addenbrooke’s Hospital, University of Cambridge, Hills Road, Cambridge CB2 0QQ, UK; (N.R.E.); (S.B.)
| | - Sarah Gorey
- Health Research Board Stroke Clinical Trials Network Ireland, Catherine McAuley Centre, Nelson Street, D07 KX5K Dublin, Ireland; (S.G.); (P.J.K.)
- Neurovascular Unit for Applied Translational and Therapeutics Research, Catherine McAuley Centre, Nelson Street, D07 KX5K Dublin, Ireland
- School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
- Stroke Service, Department of Medicine for the Elderly, Mater Misericordiae University Hospital, Eccles Street, D07 R2WY Dublin, Ireland
| | - Shiv Bhakta
- Department of Clinical Neurosciences, Box 83, Addenbrooke’s Hospital, University of Cambridge, Hills Road, Cambridge CB2 0QQ, UK; (N.R.E.); (S.B.)
| | - James H. F. Rudd
- Division of Cardiovascular Medicine, Addenbrooke’s Hospital, University of Cambridge, Hills Road, Cambridge CB2 0QQ, UK;
| | - Peter J. Kelly
- Health Research Board Stroke Clinical Trials Network Ireland, Catherine McAuley Centre, Nelson Street, D07 KX5K Dublin, Ireland; (S.G.); (P.J.K.)
- Neurovascular Unit for Applied Translational and Therapeutics Research, Catherine McAuley Centre, Nelson Street, D07 KX5K Dublin, Ireland
- School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
- Stroke Service, Department of Medicine for the Elderly, Mater Misericordiae University Hospital, Eccles Street, D07 R2WY Dublin, Ireland
| |
Collapse
|
28
|
Healy J, Searle E, Panta RK, Chernoglazov A, Roake J, Butler P, Butler A, Gieseg SP. Ex-vivo atherosclerotic plaque characterization using spectral photon-counting CT: Comparing material quantification to histology. Atherosclerosis 2023; 378:117160. [PMID: 37495488 DOI: 10.1016/j.atherosclerosis.2023.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 07/28/2023]
Abstract
BACKGROUND AND AIMS Atherosclerotic plaques are characterized as being vulnerable to rupture based on a series of histologically defined features, including a lipid-rich necrotic core, spotty calcification and ulceration. Existing imaging modalities have limitations in their ability to distinguish between different materials and structural features. We examined whether X-ray spectral photon-counting computer tomography (SPCCT) images were able to distinguish key plaque features in a surgically excised specimen from the carotid artery with comparison to histological images. METHODS An excised carotid plaque was imaged in the diagnostic X-ray energy range of 30-120 keV using a small-bore SPCCT scanner equipped with a Medipix3RX photon-counting spectral X-ray detector with a cadmium telluride (CdTe) sensor. Material identification and quantification (MIQ) images of the carotid plaque were generated using proprietary MIQ software at 0.09 mm volumetric pixels (voxels). The plaque was sectioned, stained and photographed at high resolution for comparison. RESULTS A lipid-rich core with spotty calcification was identified in the MIQ images and confirmed by histology. MIQ showed a core region containing lipid, with a mean concentration of 260 mg lipid/ml corresponding to a mean value of -22HU. MIQ showed calcified regions with mean concentration of 41 mg Ca/ml corresponded to a mean value of 123HU. An ulceration of the carotid wall at the bifurcation was identified to be lipid-lined, with a small calcification identified near the breach of the artery wall. CONCLUSIONS SPCCT derived material identification and quantification images showed hallmarks of vulnerable plaque including a lipid-rich necrotic core, spotty calcifications and ulcerations.
Collapse
Affiliation(s)
- Joe Healy
- Free Radical Biochemistry Laboratory, School of Biological Sciences, University of Canterbury, Christchurch, New Zealand; MARS Bioimaging Ltd., Christchurch, New Zealand
| | - Emily Searle
- Free Radical Biochemistry Laboratory, School of Biological Sciences, University of Canterbury, Christchurch, New Zealand; MARS Bioimaging Ltd., Christchurch, New Zealand
| | - Raj Kumar Panta
- Department of Radiology, University of Otago, Christchurch, New Zealand; MARS Bioimaging Ltd., Christchurch, New Zealand; European Organization for Nuclear Research (CERN), Geneva, Switzerland
| | | | - Justin Roake
- Department of Surgery, University of Otago, Christchurch, New Zealand
| | - Phil Butler
- Department of Physics and Astronomy, University of Canterbury, New Zealand; MARS Bioimaging Ltd., Christchurch, New Zealand; European Organization for Nuclear Research (CERN), Geneva, Switzerland
| | - Anthony Butler
- Department of Physics and Astronomy, University of Canterbury, New Zealand; Department of Radiology, University of Otago, Christchurch, New Zealand; MARS Bioimaging Ltd., Christchurch, New Zealand; European Organization for Nuclear Research (CERN), Geneva, Switzerland
| | - Steven P Gieseg
- Free Radical Biochemistry Laboratory, School of Biological Sciences, University of Canterbury, Christchurch, New Zealand; Department of Radiology, University of Otago, Christchurch, New Zealand; European Organization for Nuclear Research (CERN), Geneva, Switzerland.
| |
Collapse
|
29
|
Boczar KE, Beanlands RS, Glassman SJ, Wang J, Zeng W, deKemp RA, Ward NC, Fehlmann CA, Wells GA, Karsh J, Dwivedi G. Anti-inflammatory effect of biologic therapy in patients with psoriatic disease: A prospective cohort FDG PET study. J Nucl Cardiol 2023; 30:1642-1652. [PMID: 36754934 PMCID: PMC10372102 DOI: 10.1007/s12350-023-03204-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 12/07/2022] [Indexed: 02/10/2023]
Abstract
AIM The aim of the study was to evaluate the changes in central vascular inflammation measured by FDG PET and myocardial blood flow reserve (MFR) determined by 82Rb PET following therapy with biologic agents for 6 months in patients with psoriatic arthritis (PsA) and/or cutaneous psoriasis (PsO) (group 1) and compare with PsO subjects receiving non-biologic therapy (group 2) and controls (group 3). METHODS AND RESULTS Target-to-background ratio (TBR) by FDG PET in the most diseased segment of the ascending aorta (TBRmax) was measured to assess vascular inflammation. 82Rb PET studies were used to assess changes in left ventricular MFR. A total of 34 participants were enrolled in the study (11 in group 1, 13 in group 2, and 10 controls). A significant drop in the thoracic aorta uptake was observed in the biologic-treated group (ΔTBRmax: - .46 ± .55) compared to the PsO group treated with non-biologic therapy (ΔTBRmax: .23 ± .67). Those showing response to biologic agents maintained MFR compared to who showed no response. CONCLUSION In a cohort of psoriasis patients treated with biologics, FDG uptake in the thoracic aorta decreased over the study period. Patients who demonstrated a significant anti-inflammatory response on FDG PET imaging maintained their MFR compared to non-responders.
Collapse
Affiliation(s)
- Kevin E Boczar
- University of Ottawa Heart Institute, Ottawa, ON, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
- University of Ottawa, Ottawa, ON, Canada
- Division of Cardiology, Department of Medicine, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Jerry Wang
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Wanzhen Zeng
- University of Ottawa Heart Institute, Ottawa, ON, Canada
- University of Ottawa, Ottawa, ON, Canada
| | | | - Natalie C Ward
- School of Medicine, University of Western Australia, Perth, WA, Australia
| | - Christophe A Fehlmann
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
- The Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Division of Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - George A Wells
- University of Ottawa Heart Institute, Ottawa, ON, Canada
- Research Methods Centre, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Jacob Karsh
- Division of Rheumatology, The Ottawa Hospital, Ottawa, ON, Canada
| | - Girish Dwivedi
- University of Ottawa Heart Institute, Ottawa, ON, Canada.
- University of Ottawa, Ottawa, ON, Canada.
- School of Medicine, University of Western Australia, Perth, WA, Australia.
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Murdoch, Australia.
- Department of Cardiology, Fiona Stanley Hospital, Murdoch, WA, Australia.
- School of Biomedical Sciences, Curtin University, Bentley, WA, Australia.
| |
Collapse
|
30
|
Jensen JK, Madsen JS, Jensen MEK, Kjaer A, Ripa RS. [ 64Cu]Cu-DOTATATE PET metrics in the investigation of atherosclerotic inflammation in humans. J Nucl Cardiol 2023; 30:986-1000. [PMID: 36045250 PMCID: PMC10261263 DOI: 10.1007/s12350-022-03084-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/19/2022] [Indexed: 11/25/2022]
Abstract
PURPOSE The aim of this study was to assess and compare the arterial uptake of the inflammatory macrophage targeting PET tracer [64Cu]Cu-DOTATATE in patients with no or known cardiovascular disease (CVD) to investigate potential differences in uptake. METHODS Seventy-nine patients who had undergone [64Cu]Cu-DOTATATE PET/CT imaging for neuroendocrine neoplasm disease were retrospectively allocated to three groups: controls with no known CVD risk factors (n = 22), patients with CVD risk factors (n = 24), or patients with known ischemic CVD (n = 33). Both maximum, mean of max and most-diseased segment (mds) standardized uptake value (SUV) and target-to-background ratio (TBR) uptake metrics were measured and reported for the carotid arteries and the aorta. To assess reproducibility between different reviewers, Bland-Altman plots were made. RESULTS For the carotid arteries, SUVmax (P = .03), SUVmds (0.05), TBRmax (P < .01), TBRmds (P < .01), and mean-of-max TBR (P = .01) were overall shown to provide a group-wise difference in uptake. When measuring uptake values in the aorta, a group-wise difference was only observed with TBRmds (P = .04). Overall, reproducibility of the reported uptake metrics was excellent for SUVs and good to excellent for TBRs for both the carotid arteries and the aorta. CONCLUSION Using [64Cu]Cu-DOTATATE PET imaging as a marker of atherosclerotic inflammation, we were able to demonstrate differences in some of the most frequently reported uptake metrics in patients with different degrees of CVD. Measurements of the carotid artery as either maximum uptake values or most-diseased segment analysis showed the best ability to discriminate between the groups.
Collapse
Affiliation(s)
- Jacob K. Jensen
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital – Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Johanne S. Madsen
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital – Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Malte E. K. Jensen
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital – Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital – Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rasmus S. Ripa
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital – Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
31
|
Whittington B, Dweck MR, van Beek EJR, Newby D, Williams MC. PET-MRI of Coronary Artery Disease. J Magn Reson Imaging 2023; 57:1301-1311. [PMID: 36524452 DOI: 10.1002/jmri.28554] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022] Open
Abstract
Simultaneous positron emission tomography and magnetic resonance imaging (PET-MRI) combines the anatomical detail and tissue characterization of MRI with the functional information from PET. Within the coronary arteries, this hybrid technique can be used to identify biological activity combined with anatomically high-risk plaque features to better understand the processes underlying coronary atherosclerosis. Furthermore, the downstream effects of coronary artery disease on the myocardium can be characterized by providing information on myocardial perfusion, viability, and function. This review will describe the current capabilities of PET-MRI in coronary artery disease and discuss the limitations and future directions of this emerging technique. LEVEL OF EVIDENCE: 5 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Beth Whittington
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging Facility QMRI, University of Edinburgh, Edinburgh, UK
| | - Marc R Dweck
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging Facility QMRI, University of Edinburgh, Edinburgh, UK
| | | | - David Newby
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging Facility QMRI, University of Edinburgh, Edinburgh, UK
| | - Michelle C Williams
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging Facility QMRI, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
32
|
Singh SB, Ng SJ, Lau HC, Khanal K, Bhattarai S, Paudyal P, Shrestha BB, Naseer R, Sandhu S, Gokhale S, Raynor WY. Emerging PET Tracers in Cardiac Molecular Imaging. Cardiol Ther 2023; 12:85-99. [PMID: 36593382 PMCID: PMC9986170 DOI: 10.1007/s40119-022-00295-1] [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: 10/12/2022] [Accepted: 11/26/2022] [Indexed: 01/04/2023] Open
Abstract
18F-fluorodeoxyglucose (FDG) and 18F-sodium fluoride (NaF) represent emerging PET tracers used to assess atherosclerosis-related inflammation and molecular calcification, respectively. By localizing to sites with high glucose utilization, FDG has been used to assess myocardial viability for decades, and its role in evaluating cardiac sarcoidosis has come to represent a major application. In addition to determining late-stage changes such as loss of perfusion or viability, by targeting mechanisms present in atherosclerosis, PET-based techniques have the ability to characterize atherogenesis in the early stages to guide intervention. Although it was once thought that FDG would be a reliable indicator of ongoing plaque formation, micro-calcification as portrayed by NaF-PET/CT appears to be a superior method of monitoring disease progression. PET imaging with NaF has the additional advantage of being able to determine abnormal uptake due to coronary artery disease, which is obscured by physiologic myocardial activity on FDG-PET/CT. In this review, we discuss the evolving roles of FDG, NaF, and other PET tracers in cardiac molecular imaging.
Collapse
Affiliation(s)
- Shashi Bhushan Singh
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Sze Jia Ng
- Department of Medicine, Crozer-Chester Medical Center, 1 Medical Center Boulevard, Upland, PA, 19013, USA
| | - Hui Chong Lau
- Department of Medicine, Crozer-Chester Medical Center, 1 Medical Center Boulevard, Upland, PA, 19013, USA
| | - Kishor Khanal
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
- Division of Cardiology, Memorial Healthcare System, 3501 Johnson Street, Hollywood, FL, 33021, USA
| | - Sanket Bhattarai
- Department of Medicine, KIST Medical College, Mahalaxmi 01, Lalitpur, Bagmati, Nepal
| | - Pranita Paudyal
- West China Hospital, Sichuan University, 37 Guoxue Lane, Wuhou District, Chengdu, 610041, Sichuan, China
| | - Bimash Babu Shrestha
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Rizwan Naseer
- Department of Medicine, Crozer-Chester Medical Center, 1 Medical Center Boulevard, Upland, PA, 19013, USA
| | - Simran Sandhu
- College of Health and Human Development, Pennsylvania State University, 10 East College Avenue, University Park, PA, 16802, USA
| | - Saket Gokhale
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - William Y Raynor
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA.
- Department of Radiology, Rutgers Robert Wood Johnson Medical School, 1 Robert Wood Johnson Place, MEB #404, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
33
|
Derlin T, Spencer BA, Mamach M, Abdelhafez Y, Nardo L, Badawi RD, Cherry SR, Bengel FM. Exploring Vessel Wall Biology In Vivo by Ultrasensitive Total-Body PET. J Nucl Med 2023; 64:416-422. [PMID: 36175139 PMCID: PMC10071799 DOI: 10.2967/jnumed.122.264550] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/28/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
Ultrasensitive, high-resolution, extended-field-of-view total-body (TB) PET using the first-of-its-kind 194-cm axial-field-of-view uEXPLORER may facilitate the interrogation of biologic hallmarks of hitherto difficult-to-evaluate low-signal vessel wall pathology in cardiovascular disease. Methods: Healthy volunteers were imaged serially for up to 12 h after a standard dose of 18F-FDG (n = 15) or for up to 3 h after injection of a very low dose (about 5% of a standard dose; n = 15). A cohort undergoing standard 18F-FDG PET (n = 15) on a conventional scanner with a 22-cm axial field of view served as a comparison group. Arterial wall signal, crosstalk with hematopoietic and lymphoid organs, and image quality were analyzed using standardized techniques. Results: TB PET depicted the large vessel walls with excellent quality. The arterial wall could be imaged with high contrast up to 12 h after tracer injection. Ultralow-dose TB 18F-FDG images yielded a vessel wall signal and target-to-background ratio comparable to those of conventional-dose, short-axial-field-of-view PET. Crosstalk between vessel wall and lymphoid organs was identified with better accuracy in both TB PET cohorts than in conventional PET. Conclusion: TB PET enables detailed assessment of in vivo vessel wall biology and its crosstalk with other organs over an extended time window after tracer injection or at an ultralow tracer dose. These initial observations support the feasibility of serial imaging in low-risk populations and will stimulate future mechanistic studies or therapy monitoring in atherosclerosis and other vessel wall pathologies.
Collapse
Affiliation(s)
- Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany;
| | - Benjamin A Spencer
- Department of Biomedical Engineering, University of California, Davis, Davis, California
| | - Martin Mamach
- Department of Medical Physics and Radiation Protection, Hannover Medical School, Hannover, Germany; and
| | - Yasser Abdelhafez
- Department of Radiology, University of California, Davis, Davis, California
| | - Lorenzo Nardo
- Department of Radiology, University of California, Davis, Davis, California
| | - Ramsey D Badawi
- Department of Biomedical Engineering, University of California, Davis, Davis, California
- Department of Radiology, University of California, Davis, Davis, California
| | - Simon R Cherry
- Department of Biomedical Engineering, University of California, Davis, Davis, California
- Department of Radiology, University of California, Davis, Davis, California
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| |
Collapse
|
34
|
Nakahara T, Strauss HW, Narula J, Jinzaki M. Vulnerable Plaque Imaging. Semin Nucl Med 2023; 53:230-240. [PMID: 36333157 DOI: 10.1053/j.semnuclmed.2022.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022]
Abstract
Atherosclerotic plaques progress as a result of inflammation. Both invasive and noninvasive imaging techniques have been developed to identify and characterize plaque as vulnerable (more likely to rupture and cause a clinical event). Imaging techniques to identify vulnerable include identifying vessels with focal subendothelial collections of I) inflammatory cells; II) lipid/ fatty acid; III) local regions of hypoxia; IV) local expression of angiogenesis factors; V) local expression of protease; VI) intravascular foci of thrombus; hemorrhage (most often seen in the aftermath of a clinical event); VII) apoptosis and VIII) microcalcification. This review provides an overview of atherosclerotic plaque progression and tracers which can visualize specific molecules associated with vulnerability.
Collapse
Affiliation(s)
- Takehiro Nakahara
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan.
| | - H William Strauss
- Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jagat Narula
- Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Mahahiro Jinzaki
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
35
|
Shah N, Reid M, Mani V, Kundel V, Kaplan RC, Kizer JR, Fayad ZA, Shea S, Redline S. Sleep apnea and carotid atherosclerosis in the Multi-Ethnic Study of Atherosclerosis (MESA): leveraging state-of-the-art vascular imaging. THE INTERNATIONAL JOURNAL OF CARDIOVASCULAR IMAGING 2023; 39:621-630. [PMID: 36316593 DOI: 10.1007/s10554-022-02743-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 10/07/2022] [Indexed: 11/05/2022]
Abstract
PURPOSE To further characterize the relationship between obstructive sleep apnea (OSA) and carotid atherosclerosis, we examined the structural and metabolic features of carotid plaque using hybrid 18-F-fluorodeoxyglucose (FDG) Positron Emission Tomography/Magnetic Resonance Imaging (PET/MRI) in the Multi-Ethnic Study of Atherosclerosis (MESA). METHODS We studied 46 individuals from the MESA-PET and MESA-Sleep ancillary studies. OSA was defined as an apnea hypopnea index [AHI] ≥ 15 events per hour (4% desaturation). PET/MRI was used to measure carotid plaque inflammation (using target-to-background-ratios [TBR]) and carotid wall thickness (CWT). Linear regression was used to assess the associations between OSA, CWT and TBR. RESULTS The mean age was 67.9 years (SD 8.53) and the mean BMI was 28.9 kg/m2 (SD 4.47). There was a trend toward a higher mean CWT in the OSA (n = 11) vs. non-OSA group (n = 35), 1.51 vs. 1.41 (p = 0.098). TBR did not differ by OSA groups, and there was no significant association between OSA and carotid plaque inflammation (TBR) in adjusted analyses. Although there was a significant interaction between OSA and obesity, there were no statistically significant associations between OSA and vascular inflammation in stratified analysis by obesity. CONCLUSION Despite a trend toward a higher carotid wall thickness in OSA vs. non-OSA participants, we did not find an independent association between OSA and carotid plaque inflammation using PET/MRI in MESA. Our findings suggest that simultaneous assessments of structural and metabolic features of atherosclerosis may fill current knowledge gaps pertaining to the influence of OSA on atherosclerosis prevalence and progression.
Collapse
Affiliation(s)
- Neomi Shah
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Michelle Reid
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
| | - Venkatesh Mani
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vaishnavi Kundel
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert C Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA.,Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jorge R Kizer
- Departments of Medicine, Epidemiology and Biostatistics, San Francisco Veterans Affairs Health Care System and University of California San Francisco, San Francisco, CA, USA
| | - Zahi A Fayad
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steven Shea
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, and Department of Epidemiology, Mailman School of Public Health, New York, NY, USA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
36
|
Nakamura E, Maekawa K, Saito Y, Matsumoto T, Ogawa M, Komohara Y, Asada Y, Yamashita A. Altered choline level in atherosclerotic lesions: Upregulation of choline transporter-like protein 1 in human coronary unstable plaque. PLoS One 2023; 18:e0281730. [PMID: 36800352 PMCID: PMC9937458 DOI: 10.1371/journal.pone.0281730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Inflammatory activity and hypoxia in atherosclerotic plaques are associated with plaque instability and thrombotic complications. Recent studies show that vascular cell metabolism affects atherogenesis and thrombogenicity. This study aimed to identify the metabolites in macrophage-rich unstable plaques that modulate atherogenesis and serve as potential markers of plaque instability. Atherosclerotic plaques were induced by balloon injury in the iliofemoral arteries of rabbits fed on a conventional or 0.5% cholesterol diet. At 3 months post-balloon injury, the arteries and cardiac tissues were subjected to histological, quantitative real-time polymerase chain reaction, and metabolomic analyses. The identified metabolite-related proteins were immunohistochemically analyzed in stable and unstable plaques from human coronary arteries. The factors modulating the identified metabolites were examined in macrophages derived from human peripheral blood mononuclear cells. Metabolomic analysis revealed that choline and guanine levels in macrophage-rich arteries were upregulated compared with those in non-injured arteries and cardiac tissues. Vascular choline levels, but not guanine levels, were positively correlated with the areas immunopositive for macrophages and tumor necrosis factor (TNF)-α and matrix metalloproteinase (MMP) 9 mRNA levels in injured arteries. In human coronary arteries, choline transporter-like protein (CTL) 1 was mainly localized to macrophages within plaques. The area that was immunopositive for CTL1 in unstable plaques was significantly higher than that in stable plaques. Intracellular choline levels were upregulated upon stimulation with TNF-α but were downregulated under hypoxia in cultured macrophages. Administration of choline upregulated the expression of TNF-α and CTL1 mRNA in cultured macrophages. The transfection of CTL1 small interfering RNA decreased CTL1, TNF-α, and MMP9 mRNA levels in cultured macrophages. These results suggest that choline metabolism is altered in macrophage-rich atherosclerotic lesions and unstable plaques. Thus, CTL1 may be potential markers of plaque instability.
Collapse
Affiliation(s)
- Eriko Nakamura
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Kazunari Maekawa
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Yoichi Saito
- Bioengineering Lab, Faculty of Advanced Science and Technology, Kumamoto University, Kumamoto, Japan
| | - Tomoko Matsumoto
- Center for Collaborative Research and Community Cooperation, University of Miyazaki, Miyazaki, Japan
| | - Mikako Ogawa
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Science, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yujiro Asada
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- Department of Pathology, Miyazaki Medical Association Hospital, Miyazaki, Japan
| | - Atsushi Yamashita
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- * E-mail:
| |
Collapse
|
37
|
Advances in the Assessment of Coronary Artery Disease Activity with PET/CT and CTA. Tomography 2023; 9:328-341. [PMID: 36828378 PMCID: PMC9962109 DOI: 10.3390/tomography9010026] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 01/30/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Non-invasive testing plays a pivotal role in the diagnosis, assessment of progression, response to therapy, and risk stratification of coronary artery disease. Although anatomical plaque imaging by computed tomography angiography (CTA) and ischemia detection with myocardial perfusion imaging studies are current standards of care, there is a growing body of evidence that imaging of the processes which drive atherosclerotic plaque progression and rupture has the potential to further enhance risk stratification. In particular, non-invasive imaging of coronary plaque inflammation and active calcification has shown promise in this regard. Positron emission tomography (PET) with newly-adopted radiotracers provides unique insights into atheroma activity acting as a powerful independent predictor of myocardial infarctions. Similarly, by providing a quantitative measure of coronary inflammation, the pericoronary adipose tissue density (PCAT) derived from standard coronary CTA enhances cardiac risk prediction and allows re-stratification over and above current state-of-the-art assessments. In this review, we shall discuss the recent advances in the non-invasive methods of assessment of disease activity by PET and CTA, highlighting how these methods could improve risk stratification and ultimately benefit patients with coronary artery disease.
Collapse
|
38
|
Kwiecinski J. Novel PET Applications and Radiotracers for Imaging Cardiovascular Pathophysiology. Cardiol Clin 2023; 41:129-139. [PMID: 37003671 DOI: 10.1016/j.ccl.2023.01.002] [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: 02/22/2023]
Abstract
PET allows the assessment of cardiovascular pathophysiology across a wide range of cardiovascular conditions. By imaging processes directly involved in disease progression and adverse events, such as inflammation and developing calcifications (microcalcifications), PET can not only enhance our understanding of cardiovascular disease, but also, as shown for 18F-sodium fluoride, has the potential to predict hard endpoints. In this review, the recent advances in disease activity assessment with cardiovascular PET, which provide hope that this promising technology could be leveraged in the clinical setting, shall be discussed.
Collapse
Affiliation(s)
- Jacek Kwiecinski
- Department of Interventional Cardiology and Angiology, KKiAI, Institute of Cardiology, Alpejska 42, Warsaw 04-628, Poland.
| |
Collapse
|
39
|
Zheng Y, Lim MJR, Tan BYQ, Chan BPL, Paliwal P, Jonathan OJY, Bharatendu C, Chan ACY, Yeo LLL, Vijayan J, Hong CS, Chee YH, Wong LYH, Chen J, Chong VYF, Dong Y, Tan CH, Sunny S, Teoh HL, Sinha AK, Sharma VK. Role of plaque inflammation in symptomatic carotid stenosis. Front Neurol 2023; 14:1086465. [PMID: 36761341 PMCID: PMC9902904 DOI: 10.3389/fneur.2023.1086465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/09/2023] [Indexed: 01/25/2023] Open
Abstract
Objective Prior studies have shown that plaque inflammation on FDG-PET and the symptomatic carotid atheroma inflammation lumen-stenosis (SCAIL) score were associated with recurrent ischemic events, but the findings have thus far not been widely validated. Therefore, we aimed to validate the findings of prior studies. Methods A single-center prospective cohort study that recruited patients with (1) recent TIA or ischemic stroke within the past 30 days, (2) ipsilateral carotid artery stenosis of ≥50%, and (3) were not considered for early carotid revascularization. The (1) maximum standardized uptake value (SUVmax) of the symptomatic carotid plaque, (2) the SCAIL score, and (3) stenosis severity of the symptomatic carotid artery were measured for all patients. The outcomes were (1) a 90-day ipsilateral ischemic stroke and (2) a 90-day ipsilateral symptomatic TIA or major adverse cardiovascular event (MACE). Results Among the 131 patients included in the study, the commonest cardiovascular risk factor was hypertension (95 patients, 72.5%), followed by diabetes mellitus (77 patients, 58.8%) and being a current smoker (64 patients, 48.9%). The median (IQR) duration between the index cerebral ischemic event and recruitment to the study was 1 (0, 2.5) days. The median (IQR) duration between the index cerebral ischemic event and FDG-PET was 5 (4, 7) days. A total of 14 (10.7%) patients had a 90-day stroke, and 41 (31.3%) patients had a 90-day TIA or MACE. On comparison of the predictive performances of the SCAIL score and SUVmax, SUVmax was found to be superior to the SCAIL score for predicting both 90-day ipsilateral ischemic stroke (AUC: SCAIL = 0.79, SUVmax = 0.92; p < 0.001; 95% CI = 0.072, 0.229) and 90-day TIA or MACE (AUC: SCAIL = 0.76, SUVmax = 0.84; p = 0.009; 95% CI = 0.020, 0.143). Conclusion Plaque inflammation as quantified on FDG-PET may serve as a reliable biomarker for risk stratification among patients with ECAD and recent TIA or ischemic stroke. Future studies should evaluate whether patients with significant plaque inflammation as quantified on FDG-PET benefit from carotid revascularization and/or anti-inflammatory therapy.
Collapse
Affiliation(s)
- Yilong Zheng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mervyn Jun Rui Lim
- Division of Neurosurgery, National University Health System, Singapore, Singapore
| | - Benjamin Yong-Qiang Tan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore,Division of Neurology, National University Health System, Singapore, Singapore
| | | | | | | | - Chandra Bharatendu
- Division of Neurology, National University Health System, Singapore, Singapore
| | | | | | - Joy Vijayan
- Division of Neurology, National University Health System, Singapore, Singapore
| | - Chiew S. Hong
- Division of Neurology, National University Health System, Singapore, Singapore
| | - Young Heng Chee
- Division of Neurology, National University Health System, Singapore, Singapore
| | - Lily Y. H. Wong
- Division of Neurology, National University Health System, Singapore, Singapore
| | - Jintao Chen
- Division of Neurology, National University Health System, Singapore, Singapore
| | | | - Yanhong Dong
- Alice Lee Centre for Nursing Studies, Singapore, Singapore
| | - Chi Hsien Tan
- Division of Neurology, National University Health System, Singapore, Singapore
| | - Sibi Sunny
- Division of Neurology, National University Health System, Singapore, Singapore
| | - Hock Luen Teoh
- Division of Neurology, National University Health System, Singapore, Singapore
| | - Arvind Kumar Sinha
- Department of Diagnostic Imaging, National University Health System, Singapore, Singapore
| | - Vijay Kumar Sharma
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore,Division of Neurology, National University Health System, Singapore, Singapore,*Correspondence: Vijay Kumar Sharma ✉
| |
Collapse
|
40
|
Ng SJ, Lau HC, Naseer R, Sandhu S, Raynor WY, Werner TJ, Alavi A. Atherosclerosis Imaging. PET Clin 2023; 18:71-80. [DOI: 10.1016/j.cpet.2022.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
41
|
Zhang Y, Cui B, Yang H, Ma J, Yang Y, Yang B, Ma Y, Jiao L, Li X, Lu J. Morphological feature and mapping inflammation in classified carotid plaques in symptomatic and asymptomatic patients: A hybrid 18F-FDG PET/MR study. Front Neurosci 2023; 17:1144248. [PMID: 37025371 PMCID: PMC10070967 DOI: 10.3389/fnins.2023.1144248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 03/06/2023] [Indexed: 04/08/2023] Open
Abstract
Purpose To investigate morphological and inflamed-metabolism features of carotid atherosclerotic plaques between symptomatic and asymptomatic patients with hybrid 18F-FDG PET/MR imaging. Methods A total of 20 symptomatic and 20 asymptomatic patients with carotid plaques underwent hybrid 18F-FDG PET/MR scans. American heart association (AHA) lesion types were classified, and plaque compositions were further determined on consecutive MRI axial sections in both carotid arteries. 18F-FDG uptake in carotid arteries was quantified using region of interest (ROI) methods based on maximum standardized uptake values (SUVmax) and target-to-background ratio (TBR) on corresponding positron emission tomography (PET) images. Results A total of seventy-one carotid plaques were quantified. AHA type VI was the most common (23, 32.4%), and the region of carotid bifurcation was the most common place presenting lesions (32, 45.1%). Compared with the asymptomatic group, the prevalence of high-risk features including plaque burden, lumen stenosis, maximum necrotic core area, and maximum intra-plaque hemorrhage area increased in the symptomatic group. Carotid TBR values of plaque in symptomatic group (TBR = 2.56 ± 0.34) was significantly higher than that in asymptomatic group (TBR = 1.57 ± 0.14) (P < 0.05). hs-CRP is an independent risk factor for the stability of carotid plaque. The correlation between normalized wall index (NWI) and TBR values was significantly positive in both the symptomatic and the asymptomatic groups (P < 0.01), and both NWI and TBR were significantly correlated with the level of hs-CRP (P < 0.01). Conclusion Integrated 18F-FDG PET/MR scans presented distinct risk features between symptomatic and asymptomatic patients. Hybrid 18F-FDG PET/MR systems combined with clinical serum hs-CRP may help distinguish vulnerable carotid plaques.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Bixiao Cui
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Hongwei Yang
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Jie Ma
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Yu Yang
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Bin Yang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yan Ma
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiang Li
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- *Correspondence: Jie Lu,
| |
Collapse
|
42
|
Totzeck M, Aide N, Bauersachs J, Bucerius J, Georgoulias P, Herrmann K, Hyafil F, Kunikowska J, Lubberink M, Nappi C, Rassaf T, Saraste A, Sciagra R, Slart RHJA, Verberne H, Rischpler C. Nuclear medicine in the assessment and prevention of cancer therapy-related cardiotoxicity: prospects and proposal of use by the European Association of Nuclear Medicine (EANM). Eur J Nucl Med Mol Imaging 2023; 50:792-812. [PMID: 36334105 PMCID: PMC9852191 DOI: 10.1007/s00259-022-05991-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022]
Abstract
Cardiotoxicity may present as (pulmonary) hypertension, acute and chronic coronary syndromes, venous thromboembolism, cardiomyopathies/heart failure, arrhythmia, valvular heart disease, peripheral arterial disease, and myocarditis. Many of these disease entities can be diagnosed by established cardiovascular diagnostic pathways. Nuclear medicine, however, has proven promising in the diagnosis of cardiomyopathies/heart failure, and peri- and myocarditis as well as arterial inflammation. This article first outlines the spectrum of cardiotoxic cancer therapies and the potential side effects. This will be complemented by the definition of cardiotoxicity using non-nuclear cardiovascular imaging (echocardiography, CMR) and biomarkers. Available nuclear imaging techniques are then presented and specific suggestions are made for their application and potential role in the diagnosis of cardiotoxicity.
Collapse
Affiliation(s)
- Matthias Totzeck
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Nicolas Aide
- Nuclear Medicine Department, University Hospital, Caen, France
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Jan Bucerius
- Department of Nuclear Medicine, University Medicine Göttingen, Georg-August-University Göttingen, Göttingen, Germany
| | - Panagiotis Georgoulias
- Department of Nuclear Medicine, Faculty of Medicine, University of Thessaly, University Hospital of Larissa, Larissa, Greece
| | - Ken Herrmann
- Clinic for Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Fabien Hyafil
- Department of Nuclear Medicine, DMU IMAGINA, Georges-Pompidou European Hospital, Assistance-Publique – Hôpitaux de Paris, University of Paris, Paris, France
| | - Jolanta Kunikowska
- Nuclear Medicine Department, Medical University of Warsaw, Warsaw, Poland
| | - Mark Lubberink
- Medical Physics, Uppsala University Hospital, Uppsala, Sweden
| | - Carmela Nappi
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, Naples, Italy
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Antti Saraste
- Heart Center, Turku University Hospital and University of Turku, Turku, Finland
| | - Roberto Sciagra
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Riemer H. J. A. Slart
- Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands ,Department of Biomedical Photonic Imaging, Faculty of Science and Technology, Enschede, The Netherlands
| | - Hein Verberne
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Christoph Rischpler
- Clinic for Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
43
|
Parametric Imaging of Biologic Activity of Atherosclerosis Using Dynamic Whole-Body Positron Emission Tomography. JACC. CARDIOVASCULAR IMAGING 2022; 15:2098-2108. [PMID: 36481078 DOI: 10.1016/j.jcmg.2022.05.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 04/26/2022] [Accepted: 05/11/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND For molecular imaging of atherosclerotic vessel wall activity, tracer kinetic analysis may yield improved contrast versus blood, more robust quantitative parameters, and more reliable characterization of systems biology. OBJECTIVES The authors introduce a novel dynamic whole-body positron emission tomography (PET) protocol that is enabled by rapid continuous camera table motion, followed by reconstruction of parametric data sets using voxel-based Patlak graphical analysis. METHODS Twenty-five subjects were prospectively enrolled and underwent dynamic PET up to 90 minutes after injection of 2-[18F]fluoro-2-deoxy-D-glucose (FDG). Two sets of images were generated: 1) the established standard of static standardized uptake value (SUV) images; and 2) parametric images of the metabolic rate of FDG (MRFDG) using the Patlak plot-derived influx rate. Arterial wall signal was measured and compared using the volume-of-interest technique, and its association with hematopoietic and lymphoid organ signal and atherosclerotic risk factors was explored. RESULTS Parametric MRFDG images provided excellent arterial wall visualization, with elimination of blood-pool activity, and enhanced focus detectability and reader confidence. Target-to-background ratio (TBR) from MRFDG images was significantly higher compared with SUV images (2.6 ± 0.8 vs 1.4 ± 0.2; P < 0.0001), confirming improved arterial wall contrast. On MRFDG images, arterial wall signal showed improved correlation with hematopoietic and lymphoid organ activity (spleen P = 0.0009; lymph nodes P = 0.0055; and bone marrow P = 0.0202) and increased with the number of atherosclerotic risk factors (r = 0.49; P = 0.0138), where signal from SUV images (SUVmaxP = 0.9754; TBRmaxP = 0.8760) did not. CONCLUSIONS Absolute quantification of MRFDG is feasible for arterial wall using dynamic whole-body PET imaging. Parametric images provide superior arterial wall contrast, and they might be better suited to explore the relationship between arterial wall activity, systemic organ networks, and cardiovascular risk. This novel methodology may serve as a platform for future diagnostic and therapeutic clinical studies targeting the biology of arterial wall disease.
Collapse
|
44
|
Fayad ZA, Calcagno C. Exploring Atherosclerosis Imaging With FDG-PET in Motion. JACC. CARDIOVASCULAR IMAGING 2022; 15:2109-2111. [PMID: 36481079 DOI: 10.1016/j.jcmg.2022.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022]
Affiliation(s)
- Zahi A Fayad
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| | - Claudia Calcagno
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
45
|
Figtree GA, Adamson PD, Antoniades C, Blumenthal RS, Blaha M, Budoff M, Celermajer DS, Chan MY, Chow CK, Dey D, Dwivedi G, Giannotti N, Grieve SM, Hamilton-Craig C, Kingwell BA, Kovacic JC, Min JK, Newby DE, Patel S, Peter K, Psaltis PJ, Vernon ST, Wong DT, Nicholls SJ. Noninvasive Plaque Imaging to Accelerate Coronary Artery Disease Drug Development. Circulation 2022; 146:1712-1727. [PMID: 36441819 DOI: 10.1161/circulationaha.122.060308] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 08/29/2022] [Indexed: 11/30/2022]
Abstract
Coronary artery disease (CAD) remains the leading cause of adult mortality globally. Targeting known modifiable risk factors has had substantial benefit, but there remains a need for new approaches. Improvements in invasive and noninvasive imaging techniques have enabled an increasing recognition of distinct quantitative phenotypes of coronary atherosclerosis that are prognostically relevant. There are marked differences in plaque phenotype, from the high-risk, lipid-rich, thin-capped atheroma to the low-risk, quiescent, eccentric, nonobstructive calcified plaque. Such distinct phenotypes reflect different pathophysiologic pathways and are associated with different risks for acute ischemic events. Noninvasive coronary imaging techniques, such as computed tomography, positron emission tomography, and coronary magnetic resonance imaging, have major potential to accelerate cardiovascular drug development, which has been affected by the high costs and protracted timelines of cardiovascular outcome trials. This may be achieved through enrichment of high-risk phenotypes with higher event rates or as primary end points of drug efficacy, at least in phase 2 trials, in a manner historically performed through intravascular coronary imaging studies. Herein, we provide a comprehensive review of the current technology available and its application in clinical trials, including implications for sample size requirements, as well as potential limitations. In its effort to accelerate drug development, the US Food and Drug Administration has approved surrogate end points for 120 conditions, but not for CAD. There are robust data showing the beneficial effects of drugs, including statins, on CAD progression and plaque stabilization in a manner that correlates with established clinical end points of mortality and major adverse cardiovascular events. This, together with a clear mechanistic rationale for using imaging as a surrogate CAD end point, makes it timely for CAD imaging end points to be considered. We discuss the importance of global consensus on these imaging end points and protocols and partnership with regulatory bodies to build a more informed, sustainable staged pathway for novel therapies.
Collapse
Affiliation(s)
- Gemma A Figtree
- Kolling Institute of Medical Research, Sydney, Australia (G.A.F., S.T.V.)
- Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Australia (G.A.F., S.T.V.)
- Charles Perkins Centre (G.A.F., C.K.C.), University of Sydney, Australia
- Faculty of Medicine and Health (G.A.F., D.S.C., N.G., S.P., S.T.V.), University of Sydney, Australia
| | - Philip D Adamson
- Christchurch Heart Institute, University of Otago Christchurch, New Zealand (P.D.A.)
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (P.D.A., D.E.N.)
| | - Charalambos Antoniades
- Acute Vascular Imaging Centre (C.A.), Radcliffe Department of Medicine, University of Oxford, UK
- Division of Cardiovascular Medicine (C.A.), Radcliffe Department of Medicine, University of Oxford, UK
| | - Roger S Blumenthal
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Baltimore, MD (R.S.B., M. Blaha)
| | - Michael Blaha
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Baltimore, MD (R.S.B., M. Blaha)
| | | | - David S Celermajer
- Faculty of Medicine and Health (G.A.F., D.S.C., N.G., S.P., S.T.V.), University of Sydney, Australia
- Departments of Cardiology (D.S.C., S.P.), Royal Prince Alfred Hospital, Sydney, Australia
| | - Mark Y Chan
- Department of Cardiology, National University Heart Centre, Singapore (M.Y.C.)
| | - Clara K Chow
- Westmead Applied Research Centre (C.K.C.), University of Sydney, Australia
- Charles Perkins Centre (G.A.F., C.K.C.), University of Sydney, Australia
| | - Damini Dey
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA (D.D.)
| | - Girish Dwivedi
- Harry Perkins Institute of Medical Research, University of Western Australia (G.D.)
- Department of Cardiology, Fiona Stanley Hospital, Perth, Australia (G.D.)
| | - Nicola Giannotti
- Faculty of Medicine and Health (G.A.F., D.S.C., N.G., S.P., S.T.V.), University of Sydney, Australia
| | - Stuart M Grieve
- Imaging and Phenotyping Laboratory (S.M.G.), University of Sydney, Australia
- Radiology (S.M.G.), Royal Prince Alfred Hospital, Sydney, Australia
| | - Christian Hamilton-Craig
- Faculty of Medicine and Centre for Advanced Imaging, University of Queensland and School of Medicine, Griffith University Sunshine Coast, Australia (C.H.-C.)
| | | | - Jason C Kovacic
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia (J.C.K.)
- St Vincent's Clinical School, University of NSW, Australia (J.C.K.)
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY (J.C.K.)
| | | | - David E Newby
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (P.D.A., D.E.N.)
| | - Sanjay Patel
- Faculty of Medicine and Health (G.A.F., D.S.C., N.G., S.P., S.T.V.), University of Sydney, Australia
- Departments of Cardiology (D.S.C., S.P.), Royal Prince Alfred Hospital, Sydney, Australia
| | - Karlheinz Peter
- Baker Heart and Diabetes Institute, Melbourne, Australia (K.P.)
- Department of Cardiology, The Alfred Hospital, Melbourne, Australia (K.P.)
| | - Peter J Psaltis
- Lifelong Health, South Australian Health and Medical Research Institute, Adelaide (P.J.P.)
- Department of Cardiology, Royal Adelaide Hospital, Australia (P.J.P.)
| | - Stephen T Vernon
- Kolling Institute of Medical Research, Sydney, Australia (G.A.F., S.T.V.)
- Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Australia (G.A.F., S.T.V.)
- Faculty of Medicine and Health (G.A.F., D.S.C., N.G., S.P., S.T.V.), University of Sydney, Australia
| | - Dennis T Wong
- Monash Heart, Clayton, Australia (D.T.W., S.J.N.)
- Victorian Heart Institute, Monash University, Melbourne, Australia (D.T.W., S.J.N.)
| | - Stephen J Nicholls
- Monash Heart, Clayton, Australia (D.T.W., S.J.N.)
- Victorian Heart Institute, Monash University, Melbourne, Australia (D.T.W., S.J.N.)
| |
Collapse
|
46
|
Liu Y, Hua J, Liu L, Zhang W, Xu S, Chen X. The value of the SUV ratio between lymph node and bone marrow in predicting pelvic lymphatic metastasis of patients with locally advanced cervical cancer: an integrated PET/CT study. Nucl Med Commun 2022; 43:1155-1160. [PMID: 36003034 PMCID: PMC9575580 DOI: 10.1097/mnm.0000000000001613] [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: 06/08/2022] [Accepted: 08/16/2022] [Indexed: 02/04/2023]
Abstract
PURPOSE This study aimed to evaluate the value of the standardized uptake value (SUV) ratio between lymph nodes and bone marrow (BM) measured by Fluorine-18-fluorodeoxyglucose PET and computed tomography ( 18 F-FDG PET/CT) for predicting pelvic lymph node (PLN) metastasis in patients with locally advanced cervical cancer (LACC). MATERIALS AND METHODS A total of 62 patients with pathological stage Ib-IVa cervical cancer who underwent 18 F-FDG PET/CT before treatment were reviewed retrospectively. We measured the metabolic and morphological parameters of lymph nodes and primary tumors, bone marrow SUV (SUVBM) and calculated the ratio of lymph nodes maximum SUV (SUVmax) to bone marrow SUV (SUVLN/BM) and the ratio of short-axis diameter to long-axis diameter (Ds/l) of lymph nodes. A receiver operating characteristic (ROC) curve was performed to evaluate the diagnostic efficacy of each parameter. RESULTS There were 180 lymph nodes with pathological evidence included in the study. Our results indicated that Ds/l, SUVmax of lymph nodes (SUVLN) and SUVLN/BM were independent risk factors for PLN metastasis in LACC ( P < 0.05), and SUVLN/BM showed the best diagnostic performance by ROC curve analysis. The SUVBM in the anemia group was significantly higher than that in the nonanemia group (3.05 vs. 2.40, P < 0.05); furthermore, false-positive cases decreased when the SUVLN/BM was used as the diagnostic criterion instead of SUVLN, especially in the anemia group. ROC curve analysis showed that the area under the curve value of the combination of SUVLN/BM and Ds/l was 0.884 ( P < 0.05), which was higher than Ds/l or SUVLN/BM alone. CONCLUSIONS SUVLN/BM could improve the ability to predicting PLN metastasis in patients with LACC, and the diagnostic efficacy of the combination of SUVLN/BM and Ds/l might be better than that of a single parameter.
Collapse
Affiliation(s)
- Ying Liu
- Department of Nuclear Medicine, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, People’s Republic of China
| | - Jun Hua
- Department of Nuclear Medicine, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, People’s Republic of China
| | - Lisheng Liu
- Department of Nuclear Medicine, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, People’s Republic of China
| | - Wei Zhang
- Department of Nuclear Medicine, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, People’s Republic of China
| | - Shufan Xu
- Department of Nuclear Medicine, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, People’s Republic of China
| | - Xiaoliang Chen
- Department of Nuclear Medicine, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, People’s Republic of China
| |
Collapse
|
47
|
Puig N, Camps-Renom P, Camacho M, Aguilera-Simón A, Jiménez-Altayó F, Fernández-León A, Marín R, Martí-Fàbregas J, Sánchez-Quesada JL, Jiménez-Xarrié E, Benitez S. Plasma sICAM-1 as a Biomarker of Carotid Plaque Inflammation in Patients with a Recent Ischemic Stroke. Transl Stroke Res 2022; 13:745-756. [PMID: 35237947 PMCID: PMC9391243 DOI: 10.1007/s12975-022-01002-x] [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: 10/19/2021] [Revised: 01/24/2022] [Accepted: 02/23/2022] [Indexed: 11/12/2022]
Abstract
18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET) identifies carotid plaque inflammation and predicts stroke recurrence in patients with atherothrombotic stroke. The aim of the study was to identify plasma inflammatory biomarkers associated with plaque inflammation according to 18F-FDG uptake. We conducted a prospective study of consecutive adult patients with a recent (< 7 days) anterior circulation ischemic stroke and at least one atherosclerotic plaque in the ipsilateral internal carotid artery. We included 64 patients, 57.8% of whom showed a carotid stenosis ≥ 50%. All patients underwent an early (< 15 days from inclusion) 18F-FDG PET, and a blood sample was obtained at days 7 ± 1 from the stroke. The plasma concentration of 16 inflammation-related molecules was analyzed in a Luminex using xMAP technology. Multivariable linear regression was used to assess the association between plasma biomarkers and the standardized uptake value (SUV) of 18F-FDG uptake. Soluble intercellular adhesion molecule-1 (sICAM-1), soluble vascular adhesion molecule-1 (sVCAM-1), and fractalkine (FKN) were independently associated with plaque inflammation (β = 0.121, 95% CI 0.061-0.181, p < 0.001; β = 0.144, 95% CI 0.012-0.276, p = 0.033; β = 0.136, 95% CI 0.037-0.235, p = 0.008). In a multivariable logistic regression analysis, sICAM-1 was associated with SUVmax ≥ 2.85 (OR = 1.02, 95% CI 1.00-1.03, p = 0.020). Multivariable Cox regression was used to assess the association between biomarkers and stroke recurrence. sICAM-1 was associated with stroke recurrence (HR = 1.03, 95% CI 1.00-1.05, p = 0.002). In summary, elevated concentrations of sICAM-1 were associated with carotid plaque inflammation and an increased risk of stroke recurrence in patients with recent ischemic stroke and carotid atherosclerosis.
Collapse
Affiliation(s)
- Núria Puig
- Cardiovascular Biochemistry, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Building M, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Barcelona, Spain
| | - Pol Camps-Renom
- Stroke Unit, Department of Neurology, Hospital de La Santa Creu I Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Mercedes Camacho
- Genetic of Complexes Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Ana Aguilera-Simón
- Stroke Unit, Department of Neurology, Hospital de La Santa Creu I Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Francesc Jiménez-Altayó
- Department of Pharmacology, Neuroscience Institute, Faculty of Medicine, UAB, Cerdanyola del Vallès, Barcelona, Spain
| | - Alejandro Fernández-León
- Department of Nuclear Medicine, Hospital de La Santa Creu I Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Rebeca Marín
- Stroke Unit, Department of Neurology, Hospital de La Santa Creu I Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Joan Martí-Fàbregas
- Stroke Unit, Department of Neurology, Hospital de La Santa Creu I Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Jose Luis Sánchez-Quesada
- Cardiovascular Biochemistry, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain
- CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Madrid, Spain
| | - Elena Jiménez-Xarrié
- Stroke Unit, Department of Neurology, Hospital de La Santa Creu I Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Sonia Benitez
- Cardiovascular Biochemistry, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain.
| |
Collapse
|
48
|
Cerne JW, Liu S, Umair M, Pathrose A, Moore JE, Allen BD, Markl M, Carr JC, Savas H, Wilsbacher L, Avery R. Combined modality PET/MR for the detection of severe large vessel vasculitis. Eur J Hybrid Imaging 2022; 6:16. [PMID: 35965266 PMCID: PMC9376186 DOI: 10.1186/s41824-022-00136-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/10/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Large vessel vasculitis (LVV) can be characterized based on symptom severity, and this characterization helps clinicians decide upon treatment approach. Our aim was to compare the imaging findings of combined modality positron emission tomography/magnetic resonance (PET/MR) and inflammatory markers between severe and non-severe LVV. A retrospective query was performed to identify all patients with LVV who underwent PET/MR at our institution between January 2015 and January 2021.
Results
Eleven patients (nine females; age 62.2 ± 16.4 years) underwent 15 PET/MR scans. Positivity was defined by findings indicative of active LVV on each modality: PET positive if vessel metabolic activity > liver metabolic activity; MR positive if wall thickening or contrast enhancement. When positive PET or positive MR findings were considered a positive scan, LVV patients with severe disease (n = 9 scans) showed a higher number of positive scans (n = 9) compared to the number of positive scans in non-severe patients (n = 3) (p < 0.05). The sensitivity and specificity for the detection of severe LVV were 1.00 and 0.50, respectively. When only the presence of both positive PET and positive MR findings were considered a positive scan, inflammatory marker levels were not significantly different between severe and non-severe LVV groups (severe: erythrocyte sedimentation rate (ESR) = 9.8 ± 10.6 mm/h; C-reactive protein (CRP) = 0.6 ± 0.4 mg/dL) (non-severe: ESR = 14.3 ± 22.4 mm/h; CRP = 0.5 ± 0.6 mg/dL). Blood- and liver-normalized maximum standardized uptake values were not significantly different between severe and non-severe patients (1.4 ± 0.3 vs 1.5 ± 0.4; 1.1 ± 0.4 vs 1.0 ± 0.3, respectively).
Conclusions
Because of the differences observed, PET/MR appears to be better suited to facilitate the characterization of LVV as severe or non-severe compared to inflammatory marker measurements and quantitative measurements of metabolic activity. Qualitative assessment of PET and MR positivity by 18F-fluorodeoxyglucose PET/MR may be able to supplement clinical symptoms-based LVV classification decisions and may be helpful when clinical symptoms overlap with other disease processes.
Collapse
|
49
|
Robson PM, Kaufman A, Pruzan A, Dweck MR, Trivieri MG, Abgral R, Karakatsanis NA, Brunner PM, Guttman E, Fayad ZA, Mani V. Scan-rescan measurement repeatability of 18F-FDG PET/MR imaging of vascular inflammation. J Nucl Cardiol 2022; 29:1660-1670. [PMID: 34046803 DOI: 10.1007/s12350-021-02627-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 03/07/2021] [Indexed: 12/27/2022]
Abstract
Non-invasive positron emission tomography (PET) of vascular inflammation and atherosclerotic plaque by identifying increased uptake of 18F-fluordeoxyglucose (18F-FDG) is a powerful tool for monitoring disease activity, progression, and its response to therapy. 18F-FDG PET/computed tomography (PET/CT) of the aorta and carotid arteries has become widely used to assess changes in inflammation in clinical trials. However, the recent advent of hybrid PET/magnetic resonance (PET/MR) scanners has advantages for vascular imaging due to the reduction in radiation exposure and improved soft tissue contrast of MR compared to CT. Important for research and clinical use is an understanding of the scan-rescan repeatability of the PET measurement. While this has been studied for PET/CT, no data is currently available for vascular PET/MR imaging. In this study, we determined the scan-rescan measurement repeatability of 18F-FDG PET/MR in the aorta and carotid arteries was less than 5%, comparable to similar findings for 18F-FDG PET/CT.
Collapse
Affiliation(s)
- Philip M Robson
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Audrey Kaufman
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alison Pruzan
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marc R Dweck
- British Heart Foundation/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Maria-Giovanna Trivieri
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ronan Abgral
- Department of Nuclear Medicine, European University of Brittany, EA3878 GETBO, IFR 148, CHRU Brest, Brest, France
| | - Nicolas A Karakatsanis
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Patrick M Brunner
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY, USA
| | - Emma Guttman
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zahi A Fayad
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Venkatesh Mani
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
50
|
Taglieri N, Bonfiglioli R, Bon I, Malosso P, Corovic A, Bruno M, Le E, Granozzi B, Palmerini T, Ghetti G, Tamburello M, Bruno AG, Saia F, Tarkin JM, Rudd JHF, Calza L, Fanti S, Re MC, Galié N. Pattern of arterial inflammation and inflammatory markers in people living with HIV compared with uninfected people. J Nucl Cardiol 2022; 29:1566-1575. [PMID: 33569752 PMCID: PMC9345795 DOI: 10.1007/s12350-020-02522-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/25/2020] [Indexed: 11/03/2022]
Abstract
STUDY DESIGN To compare arterial inflammation (AI) between people living with HIV (PLWH) and uninfected people as assessed by 18F-Fluorodeoxyglucose (18F-FDG)-positron emission tomography (PET). METHODS We prospectively enrolled 20 PLWH and 20 uninfected people with no known cardiovascular disease and at least 3 traditional cardiovascular risk factors. All patients underwent 18F-FDG-PET/computed tomography (CT) of the thorax and neck. Biomarkers linked to inflammation and atherosclerosis were also determined. The primary outcome was AI in ascending aorta (AA) measured as mean maximum target-to-background ratio (TBRmax). The independent relationships between HIV status and both TBRmax and biomarkers were evaluated by multivariable linear regression adjusted for body mass index, creatinine, statin therapy, and atherosclerotic cardiovascular 10-year estimated risk (ASCVD). RESULTS Unadjusted mean TBRmax in AA was slightly higher but not statistically different (P = .18) in PLWH (2.07; IQR 1.97, 2.32]) than uninfected people (2.01; IQR 1.85, 2.16]). On multivariable analysis, PLWH had an independent risk of increased mean log-TBRmax in AA (coef = 0.12; 95%CI 0.01,0.22; P = .032). HIV infection was independently associated with higher values of interleukin-10 (coef = 0.83; 95%CI 0.34, 1.32; P = .001), interferon-γ (coef. = 0.90; 95%CI 0.32, 1.47; P = .003), and vascular cell adhesion molecule-1 (VCAM-1) (coef. = 0.75; 95%CI: 0.42, 1.08, P < .001). CONCLUSIONS In patients with high cardiovascular risk, HIV status was an independent predictor of increased TBRmax in AA. PLWH also had an increased independent risk of IFN-γ, IL-10, and VCAM-1 levels.
Collapse
Affiliation(s)
- Nevio Taglieri
- Division of Cardiology, Department of Experimental Diagnostic and Specialty Medicine, IRCCS Policlinico di St.Orsola, Alma Mater Studiorum-University of Bologna, Via Massarenti 9, 40138, Bologna, Italy.
| | - Rachele Bonfiglioli
- Division of Nuclear Medicine, Department of Experimental Diagnostic and Specialty Medicine, IRCCS Policlinico di St. Orsola, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Isabella Bon
- Division of Microbiology, Department of Experimental Diagnostic and Specialty Medicine, IRCCS Policlinico di St. Orsola, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Pietro Malosso
- Department of Medical and Surgical Sciences, Clinics of Infectious Diseases, IRCCS Policlinico di St. Orsola, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Andrej Corovic
- Division of Cardiovascular Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, UK
| | - Matteo Bruno
- Division of Cardiology, Department of Experimental Diagnostic and Specialty Medicine, IRCCS Policlinico di St.Orsola, Alma Mater Studiorum-University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Elizabeth Le
- Division of Cardiovascular Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, UK
| | - Bianca Granozzi
- Department of Medical and Surgical Sciences, Clinics of Infectious Diseases, IRCCS Policlinico di St. Orsola, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Tullio Palmerini
- Division of Cardiology, Department of Experimental Diagnostic and Specialty Medicine, IRCCS Policlinico di St.Orsola, Alma Mater Studiorum-University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Gabriele Ghetti
- Division of Cardiology, Department of Experimental Diagnostic and Specialty Medicine, IRCCS Policlinico di St.Orsola, Alma Mater Studiorum-University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Martina Tamburello
- Division of Microbiology, Department of Experimental Diagnostic and Specialty Medicine, IRCCS Policlinico di St. Orsola, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Antonio Giulio Bruno
- Division of Cardiology, Department of Experimental Diagnostic and Specialty Medicine, IRCCS Policlinico di St.Orsola, Alma Mater Studiorum-University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Francesco Saia
- Division of Cardiology, Department of Experimental Diagnostic and Specialty Medicine, IRCCS Policlinico di St.Orsola, Alma Mater Studiorum-University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Jason M Tarkin
- Division of Cardiovascular Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, UK
| | - James H F Rudd
- Division of Cardiovascular Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, UK
| | - Leonardo Calza
- Department of Medical and Surgical Sciences, Clinics of Infectious Diseases, IRCCS Policlinico di St. Orsola, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Stefano Fanti
- Division of Nuclear Medicine, Department of Experimental Diagnostic and Specialty Medicine, IRCCS Policlinico di St. Orsola, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Maria Carla Re
- Division of Microbiology, Department of Experimental Diagnostic and Specialty Medicine, IRCCS Policlinico di St. Orsola, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Nazzareno Galié
- Division of Cardiology, Department of Experimental Diagnostic and Specialty Medicine, IRCCS Policlinico di St.Orsola, Alma Mater Studiorum-University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| |
Collapse
|