1
|
Cui J, Wang G, Yip LX, Dong M, Mu M, Tian L, Gao Y, Fan Q, Zhu Q, Zhao X, Xu X, Leong DT, Sun X. Enhanced Ferritin‐Manganese Interaction by Nanoplatinum Growth Enabling Liver Fibrosis 3D Magnetic Resonance Visualization and Synergistic Therapy with Real‐Time Monitoring. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202410748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Indexed: 04/09/2025]
Abstract
AbstractEarly detection and timely intervention are essential to prevent liver fibrosis from progressing to cirrhosis or hepatocellular carcinoma. Herein, utilizing the enhanced ferritin‐manganese interaction by nanoplatinum growth, a novel ferritin‐platinum‐manganese magnetic resonance nanoplatform with RGD grafting and metformin loading (FNMMR) is developed. RGD can enhance the targeting ability of the nanoplatform toward integrin αVβ3 on activated hepatic stellate cells (aHSCs) in liver fibrosis. Systemic delivery of FNMMR shows clear degree‐dependent magnetic resonance contrast enhancement in liver fibrosis. 3D reconstruction techniques and histogram‐based features are achieved to qualitatively and quantitatively analyze the inhomogeneous liver fibrosis areas. FNMMR with catalase‐like activity can catalyze the generation of O2 to alleviate the liver fibrosis hypoxia and inhibit the expression of HIF‐1α, blocking the TGF‐β1/Smad signaling pathway. In addition, metformin shows synergy with HIF‐1α reduction in blocking the TGF‐β1/Smad pathway, effectively inhibiting the activation of HSCs and reducing collagen formation. Furthermore, FNMMR can achieve real‐time anti‐fibrotic therapy monitoring by magnetic resonance imaging. Importantly, no obvious side effects can be observed in both histological and hematology examinations. Therefore, this work presents a novel nanoplatform for accurate liver fibrosis diagnosis and synergistic anti‐fibrotic therapy with real‐time monitoring.
Collapse
Affiliation(s)
- Jin Cui
- Department of Radiology Shandong Provincial Hospital affiliated to Shandong First Medical University China 250021 China
| | - Gongzheng Wang
- Department of Radiology Shandong Provincial Hospital affiliated to Shandong First Medical University China 250021 China
| | - Li Xian Yip
- Department of Chemical and Biomolecular Engineering National University of Singapore Singapore 117585 Singapore
| | - Mengzhen Dong
- Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences Jinan 250117 China
| | - Mengyao Mu
- Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences Jinan 250117 China
| | - Liya Tian
- Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences Jinan 250117 China
| | - Yuan Gao
- Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences Jinan 250117 China
| | - Qing Fan
- Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences Jinan 250117 China
| | - Qiang Zhu
- Department of Radiology Shandong Provincial Hospital affiliated to Shandong First Medical University China 250021 China
| | - Xinya Zhao
- Department of Radiology Shandong Provincial Hospital affiliated to Shandong First Medical University China 250021 China
| | - Xueli Xu
- School of Science Shandong Jianzhu University Jinan 250101 China
| | - David Tai Leong
- Department of Chemical and Biomolecular Engineering National University of Singapore Singapore 117585 Singapore
| | - Xiao Sun
- Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences Jinan 250117 China
| |
Collapse
|
2
|
Song Y, Qin C, Chen Y, Ruan W, Gai Y, Song W, Gao Y, Hu W, Qiao P, Song X, Lv X, Zheng D, Chu H, Jiang D, Yang L, Lan X. Non-invasive visualization of liver fibrosis with [ 68Ga]Ga-DOTA-FAPI-04 PET from preclinical insights to clinical translation. Eur J Nucl Med Mol Imaging 2024; 51:3572-3584. [PMID: 38850311 DOI: 10.1007/s00259-024-06773-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 04/26/2024] [Indexed: 06/10/2024]
Abstract
PURPOSE The reversibility of early liver fibrosis highlights the need for improved early detection and monitoring techniques. Fibroblast activation protein (FAP) is a promising theranostics target significantly upregulated during fibrosis. This preclinical and preliminary clinical study investigated a FAP-targeted probe, gallium-68-labeled FAP inhibitor 04 ([68Ga]Ga-DOTA-FAPI-04), for its capability to visualize liver fibrosis. METHODS The preclinical study employed [68Ga]Ga-DOTA-FAPI-04 micro-positron emission tomography (PET)/computed tomography (CT) on carbon tetrachloride-induced mice model (n = 34) and olive oil-treated control group (n = 26), followed by validation of the probe's biodistribution. Hepatic uptake was correlated with fibrosis and inflammation levels, quantified through histology and serum assays. FAP and α-smooth muscle actin expression were determined by immunohistochemistry, as well as immunofluorescence. The subsequent clinical trial enrolled 26 patients with suspected or confirmed liver fibrosis to undergo [68Ga]Ga-DOTA-FAPI-04 PET/magnetic resonance imaging or PET/CT. Key endpoints included correlating [68Ga]Ga-DOTA-FAPI-04 uptake with histological inflammation grades and fibrosis stages, and evaluating its diagnostic and differential efficacy compared to established serum markers and liver stiffness measurement (LSM). RESULTS [68Ga]Ga-DOTA-FAPI-04 mean uptake in mice livers was notably higher than in control mice, increasing from week 6 [0.70 ± 0.11 percentage injected dose per cubic centimeter (%ID/cc)], peaking at week 10 (0.97 ± 0.15%ID/cc) and slightly reducing at week 12 (0.89 ± 0.28%ID/cc). The hepatic biodistribution and FAP expression showed a consistent trend. In the patient cohort, hepatic [68Ga]Ga-DOTA-FAPI-04 uptake presented moderate correlations with inflammation grades (r = 0.517 to 0.584, all P < 0.05) and fibrosis stages (r = 0.653 to 0.698, all P < 0.01). The average SUVmax to background ratio in the liver showed superior discriminative ability, especially between stage 0 and stage 1, outperforming LSM (area under curve 0.984 vs. 0.865). CONCLUSION [68Ga]Ga-DOTA-FAPI-04 PET shows significant potential for non-invasive visualization and dynamic monitoring of liver fibrosis in both preclinical experiment and preliminary clinical trial, especially outperforming other common clinical indicators in the early stage. TRIAL REGISTRATION NCT04605939. Registered October 25, 2020, https://clinicaltrials.gov/study/NCT04605939.
Collapse
Affiliation(s)
- Yangmeihui Song
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
- Hubei Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
| | - Chunxia Qin
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
- Hubei Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
- Key Laboratory of Biological Targeting Therapy, Ministry of Education, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
| | - Yixiong Chen
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
| | - Weiwei Ruan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
- Hubei Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
- Key Laboratory of Biological Targeting Therapy, Ministry of Education, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
| | - Yongkang Gai
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
- Hubei Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
- Key Laboratory of Biological Targeting Therapy, Ministry of Education, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
| | - Wenyu Song
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
- Hubei Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
| | - Yu Gao
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
- Hubei Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
| | - Wenzhu Hu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
- Hubei Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
| | - Pengxin Qiao
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
- Hubei Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
| | - Xiangming Song
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
- Hubei Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
| | - Xiaoying Lv
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
- Hubei Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
| | - Danzha Zheng
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
- Hubei Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
| | - Huikuan Chu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
- Hubei Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
- Key Laboratory of Biological Targeting Therapy, Ministry of Education, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
| | - Ling Yang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China.
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China.
- Hubei Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China.
- Key Laboratory of Biological Targeting Therapy, Ministry of Education, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China.
| |
Collapse
|
3
|
Che T, Yang X, Zhang Y, Zheng Y, Zhang Y, Zhang X, Wu Z. Mitochondria-Regulated Information Processing Nanosystem Promoting Immune Cell Communication for Liver Fibrosis Regression. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400413. [PMID: 38721946 DOI: 10.1002/smll.202400413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/23/2024] [Indexed: 10/04/2024]
Abstract
Liver fibrosis is a coordinated response to tissue injury that is mediated by immune cell interactions. A mitochondria-regulated information-processing (MIP) nanosystem that promotes immune cell communication and interactions to inhibit liver fibrosis is designed. The MIP nanosystem mimics the alkaline amino acid domain of mitochondrial precursor proteins, providing precise targeting of the mitochondria. The MIP nanosystem is driven by light to modulate the mitochondria of hepatic stellate cells, resulting in the release of mitochondrial DNA into the fibrotic microenvironment, as detected by macrophages. By activating the STING signaling pathway, the developed nanosystem-induced macrophage phenotype switches to a reparative subtype (Ly6Clow) and downstream immunostimulatory transcriptional activity, fully restoring the fibrotic liver to its normal tissue state. The MIP nanosystem serves as an advanced information transfer system, allowing precise regulation of trained immunity, and offers a promising approach for effective liver fibrosis immunotherapy with the potential for clinical translation.
Collapse
Affiliation(s)
- Tingting Che
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
- Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, 300134, China
| | - Xiaopeng Yang
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
- Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, 300134, China
| | - Yuanyuan Zhang
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
- Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, 300134, China
| | - Yin Zheng
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Shandong Institute of Endocrine and Metabolic Diseases, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Jinan, Shandong, 250012, China
| | - Yufei Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, Tianjin Key Laboratory of functional polymer materials, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Xinge Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, Tianjin Key Laboratory of functional polymer materials, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Zhongming Wu
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
- Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, 300134, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Shandong Institute of Endocrine and Metabolic Diseases, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Jinan, Shandong, 250012, China
| |
Collapse
|
4
|
Liu W, Xu H, Zhang H, Xie M, Liu Y, Wang L, Wu X, Feng Y, Chen K. Noninvasive assessment of liver fibrosis in mini pigs using an 18F-AlF-NOTA-RGD2 PET/CT molecular probe. Heliyon 2024; 10:e35502. [PMID: 39170113 PMCID: PMC11336738 DOI: 10.1016/j.heliyon.2024.e35502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024] Open
Abstract
To evaluate the efficacy of the 18F-AlF-NOTA-RGD2 positron emission tomography (PET)/computed tomography (CT) molecular probe for the noninvasive staging of liver fibrosis in mini pigs, a potential alternative to invasive diagnostic methods was revealed. This study used 18F-AlF-NOTA-RGD2 PET/CT imaging of mini pigs to assess liver fibrosis. The methods included synthesis and quality control of the molecular probe, establishment of an animal model of liver fibrosis, blood serum enzymatic tests, histopathological examination, PET/CT imaging, collagen content and expression, and mitochondrial reserve function assessment. The 18F-AlF-NOTA-RGD2 PET/CT molecular probe effectively differentiated various stages of liver fibrosis in mini pigs. Blood serum enzymatic tests revealed distinct stages of liver fibrosis, revealing significant increases in AST, ALT, TBIL, and DBIL levels as fibrosis advanced. Notably, ALT levels increased markedly in severe fibrosis patients. A gradual increase in collagen deposition and increasing α-SMA RNA expression and protein levels effectively differentiated between mild and severe fibrosis stages. Pathological examinations and Sirius Red staining confirmed these findings, highlighting substantial increases in collagen accumulation. PET/CT imaging results aligned with histopathological findings, showing that increased radiotracer uptake correlated with fibrosis severity. Assessments of mitochondrial function revealed a decrease in total liver glutathione content and mitochondrial reserve capacity, especially in patients with severe fibrosis. The 18F-AlF-NOTA-RGD2 PET/CT molecular probe is a promising tool for the noninvasive assessment of liver fibrosis, offering potential benefits over traditional diagnostic methods in hepatology.
Collapse
Affiliation(s)
- Wenrui Liu
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan, 610041, China
| | - Hongwei Xu
- Department of Liver Surgery, Center of Liver Transplantation, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Haili Zhang
- Department of Liver Surgery, Center of Liver Transplantation, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Maodi Xie
- Laboratory of Mitochondria and Metabolis, Wmest China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yundi Liu
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan, 610041, China
| | - Li Wang
- Jiangsu Xinrui Pharmaceutical Co., Ltd, Jiangsu, 226500, China
| | - Xiaoai Wu
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yinrui Feng
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan, 610041, China
| | - Kefei Chen
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan, 610041, China
- Department of Liver Surgery, Center of Liver Transplantation, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
5
|
Huang W, Peng Y, Kang L. Advancements of non‐invasive imaging technologies for the diagnosis and staging of liver fibrosis: Present and future. VIEW 2024; 5. [DOI: 10.1002/viw.20240010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 06/28/2024] [Indexed: 01/04/2025] Open
Abstract
AbstractLiver fibrosis is a reparative response triggered by liver injury. Non‐invasive assessment and staging of liver fibrosis in patients with chronic liver disease are of paramount importance, as treatment strategies and prognoses depend significantly on the degree of fibrosis. Although liver fibrosis has traditionally been staged through invasive liver biopsy, this method is prone to sampling errors, particularly when biopsy sizes are inadequate. Consequently, there is an urgent clinical need for an alternative to biopsy, one that ensures precise, sensitive, and non‐invasive diagnosis and staging of liver fibrosis. Non‐invasive imaging assessments have assumed a pivotal role in clinical practice, enjoying growing popularity and acceptance due to their potential for diagnosing, staging, and monitoring liver fibrosis. In this comprehensive review, we first delved into the current landscape of non‐invasive imaging technologies, assessing their accuracy and the transformative impact they have had on the diagnosis and management of liver fibrosis in both clinical practice and animal models. Additionally, we provided an in‐depth exploration of recent advancements in ultrasound imaging, computed tomography imaging, magnetic resonance imaging, nuclear medicine imaging, radiomics, and artificial intelligence within the field of liver fibrosis research. We summarized the key concepts, advantages, limitations, and diagnostic performance of each technique. Finally, we discussed the challenges associated with clinical implementation and offer our perspective on advancing the field, hoping to provide alternative directions for the future research.
Collapse
Affiliation(s)
- Wenpeng Huang
- Department of Nuclear Medicine Peking University First Hospital Beijing China
| | - Yushuo Peng
- Department of Nuclear Medicine Peking University First Hospital Beijing China
| | - Lei Kang
- Department of Nuclear Medicine Peking University First Hospital Beijing China
| |
Collapse
|
6
|
Chen S, Zhuang D, Jia Q, Guo B, Hu G. Advances in Noninvasive Molecular Imaging Probes for Liver Fibrosis Diagnosis. Biomater Res 2024; 28:0042. [PMID: 38952717 PMCID: PMC11214848 DOI: 10.34133/bmr.0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/08/2024] [Indexed: 07/03/2024] Open
Abstract
Liver fibrosis is a wound-healing response to chronic liver injury, which may lead to cirrhosis and cancer. Early-stage fibrosis is reversible, and it is difficult to precisely diagnose with conventional imaging modalities such as magnetic resonance imaging, positron emission tomography, single-photon emission computed tomography, and ultrasound imaging. In contrast, probe-assisted molecular imaging offers a promising noninvasive approach to visualize early fibrosis changes in vivo, thus facilitating early diagnosis and staging liver fibrosis, and even monitoring of the treatment response. Here, the most recent progress in molecular imaging technologies for liver fibrosis is updated. We start by illustrating pathogenesis for liver fibrosis, which includes capillarization of liver sinusoidal endothelial cells, cellular and molecular processes involved in inflammation and fibrogenesis, as well as processes of collagen synthesis, oxidation, and cross-linking. Furthermore, the biological targets used in molecular imaging of liver fibrosis are summarized, which are composed of receptors on hepatic stellate cells, macrophages, and even liver collagen. Notably, the focus is on insights into the advances in imaging modalities developed for liver fibrosis diagnosis and the update in the corresponding contrast agents. In addition, challenges and opportunities for future research and clinical translation of the molecular imaging modalities and the contrast agents are pointed out. We hope that this review would serve as a guide for scientists and students who are interested in liver fibrosis imaging and treatment, and as well expedite the translation of molecular imaging technologies from bench to bedside.
Collapse
Affiliation(s)
- Shaofang Chen
- Department of Radiology, Shenzhen People’s Hospital (The Second Clinical Medical College,
Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Danping Zhuang
- Department of Radiology, Shenzhen People’s Hospital (The Second Clinical Medical College,
Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Qingyun Jia
- Department of Radiology, Shenzhen People’s Hospital (The Second Clinical Medical College,
Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Bing Guo
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application,
Harbin Institute of Technology, Shenzhen 518055, China
| | - Genwen Hu
- Department of Radiology, Shenzhen People’s Hospital (The Second Clinical Medical College,
Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| |
Collapse
|
7
|
Baniasadi A, Das JP, Prendergast CM, Beizavi Z, Ma HY, Jaber MY, Capaccione KM. Imaging at the nexus: how state of the art imaging techniques can enhance our understanding of cancer and fibrosis. J Transl Med 2024; 22:567. [PMID: 38872212 PMCID: PMC11177383 DOI: 10.1186/s12967-024-05379-1] [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: 02/11/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024] Open
Abstract
Both cancer and fibrosis are diseases involving dysregulation of cell signaling pathways resulting in an altered cellular microenvironment which ultimately leads to progression of the condition. The two disease entities share common molecular pathophysiology and recent research has illuminated the how each promotes the other. Multiple imaging techniques have been developed to aid in the early and accurate diagnosis of each disease, and given the commonalities between the pathophysiology of the conditions, advances in imaging one disease have opened new avenues to study the other. Here, we detail the most up-to-date advances in imaging techniques for each disease and how they have crossed over to improve detection and monitoring of the other. We explore techniques in positron emission tomography (PET), magnetic resonance imaging (MRI), second generation harmonic Imaging (SGHI), ultrasound (US), radiomics, and artificial intelligence (AI). A new diagnostic imaging tool in PET/computed tomography (CT) is the use of radiolabeled fibroblast activation protein inhibitor (FAPI). SGHI uses high-frequency sound waves to penetrate deeper into the tissue, providing a more detailed view of the tumor microenvironment. Artificial intelligence with the aid of advanced deep learning (DL) algorithms has been highly effective in training computer systems to diagnose and classify neoplastic lesions in multiple organs. Ultimately, advancing imaging techniques in cancer and fibrosis can lead to significantly more timely and accurate diagnoses of both diseases resulting in better patient outcomes.
Collapse
Affiliation(s)
- Alireza Baniasadi
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA.
| | - Jeeban P Das
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Conor M Prendergast
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA
| | - Zahra Beizavi
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA
| | - Hong Y Ma
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA
| | | | - Kathleen M Capaccione
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA
| |
Collapse
|
8
|
Li Z, Yang H, Li X, She T, Tao Z, Zhong Y, Su T, Feng Y, Shi Q, Li L, Tian R, Wang S, Cheng J, Cai H, Lu X. Platelet-derived growth factor receptor β-targeted positron emission tomography imaging for the noninvasive monitoring of liver fibrosis. Eur J Nucl Med Mol Imaging 2024; 51:1530-1543. [PMID: 38189910 DOI: 10.1007/s00259-023-06577-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 12/15/2023] [Indexed: 01/09/2024]
Abstract
PURPOSE Noninvasive quantifying activated hepatic stellate cells (aHSCs) by molecular imaging is helpful for assessing disease progression and therapeutic responses of liver fibrosis. Our purpose is to develop platelet-derived growth factor receptor β (PDGFRβ)-targeted radioactive tracer for assessing liver fibrosis by positron emission tomography (PET) imaging of aHSCs. METHODS Comparative transcriptomics, immunofluorescence staining and flow cytometry were used to evaluate PDGFRβ as biomarker for human aHSCs and determine the correlation of PDGFRβ with the severity of liver fibrosis. The high affinity affibody for PDGFRβ (ZPDGFRβ) was labeled with gallium-68 (68Ga) for PET imaging of mice with carbon tetrachloride (CCl4)-induced liver fibrosis. Binding of the [68Ga]Ga-labeled ZPDGFRβ ([68Ga]Ga-DOTA-ZPDGFRβ) for aHSCs in human liver tissues was measured by autoradiography. RESULTS PDGFRβ overexpressed in aHSCs was highly correlated with the severity of liver fibrosis in patients and CCl4-treated mice. The 68Ga-labeled ZPDGFRβ affibody ([68Ga]Ga-DOTA-ZPDGFRβ) showed PDGFRβ-dependent binding to aHSCs. According to the PET imaging, hepatic uptake of [68Ga]Ga-DOTA-ZPDGFRβ increased with the accumulation of aHSCs and collagens in the fibrotic livers of mice. In contrast, hepatic uptake of [68Ga]Ga-DOTA-ZPDGFRβ decreased with spontaneous recovery or treatment of liver fibrosis, indicating that the progression and therapeutic responses of liver fibrosis in mice could be visualized by PDGFRβ-targeted PET imaging. [68Ga]Ga-DOTA-ZPDGFRβ also bound human aHSCs and visualized fibrosis in patient-derived liver tissues. CONCLUSIONS PDGFRβ is a reliable biomarker for both human and mouse aHSCs. PDGFRβ-targeted PET imaging could be used for noninvasive monitoring of liver fibrosis in mice and has great potential for clinical translation.
Collapse
Affiliation(s)
- Zhao Li
- Department of Nuclear Medicine, NHC Key Lab of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Nuclear Medicine , West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hao Yang
- Department of Nuclear Medicine, NHC Key Lab of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University, Chengdu, 610041, China
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xin Li
- Department of Nuclear Medicine , West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tianshan She
- Department of Nuclear Medicine, NHC Key Lab of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ze Tao
- Department of Nuclear Medicine, NHC Key Lab of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University, Chengdu, 610041, China
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yi Zhong
- Laboratory of Clinical Proteomics and Metabolomics, Institutes for Systems Genetics , West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tao Su
- Laboratory of Clinical Proteomics and Metabolomics, Institutes for Systems Genetics , West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yanru Feng
- Department of Nuclear Medicine, NHC Key Lab of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiuxiao Shi
- Department of Nuclear Medicine, NHC Key Lab of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lin Li
- Department of Nuclear Medicine , West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rong Tian
- Department of Nuclear Medicine , West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shisheng Wang
- Laboratory of Clinical Proteomics and Metabolomics, Institutes for Systems Genetics , West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jingqiu Cheng
- Department of Nuclear Medicine, NHC Key Lab of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University, Chengdu, 610041, China
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Huawei Cai
- Department of Nuclear Medicine , West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Xiaofeng Lu
- Department of Nuclear Medicine, NHC Key Lab of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
9
|
Pan Y, Dang H, Zhou H, Fu H, Wu S, Liu H, Zhang J, Wang R, Tian Y, Xu B. A comparison study of dynamic [ 18F]Alfatide II imaging and [ 11C]MET in orthotopic rat models of glioblastoma. J Cancer Res Clin Oncol 2024; 150:208. [PMID: 38647690 PMCID: PMC11035414 DOI: 10.1007/s00432-024-05688-4] [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: 01/10/2024] [Accepted: 03/05/2024] [Indexed: 04/25/2024]
Abstract
PURPOSE To investigate and compare the dynamic positron emission tomography (PET) imaging with [18F]Alfatide II Imaging and [11C]Methionine ([11C]MET) in orthotopic rat models of glioblastoma multiforme (GBM), and to assess the utility of [18F]Alfatide II in detecting and evaluating neoangiogenesis in GBM. METHODS [18F]Alfatide II and [11C]MET were injected into the orthotopic GBM rat models (n = 20, C6 glioma cells), followed by dynamic PET/MR scans 21 days after surgery of tumor implantation. On the PET image with both radiotracers, the MRI-based volume-of-interest (VOI) was manually delineated encompassing glioblastoma. Time-activity curves were expressed as tumor-to-normal brain ratio (TNR) parameters and PET pharmacokinetic modeling (PKM) performed using 2-tissue-compartment models (2TCM). Immunofluorescent staining (IFS), western blotting and blocking experiment of tumor tissue were performed for the validation. RESULTS Compared to 11C-MET, [18F]Alfatide II presented a persistent accumulation in the tumor, albeit with a slightly lower SUVmean of 0.79 ± 0.25, and a reduced uptake in the contralateral normal brain tissue, respectively. This resulted in a markedly higher tumor-to-normal brain ratio (TNR) of 18.22 ± 1.91. The time-activity curve (TACs) showed a significant increase in radioactive uptake in tumor tissue, followed by a plateau phase up to 60 min for [18F]Alfatide II (time to peak:255 s) and 40 min for [11C]MET (time to peak:135 s) post injection. PKM confirmed significantly higher K1 (0.23/0.07) and K3 (0.26/0.09) in the tumor region compared to the normal brain with [18F]Alfatide II. Compared to [11C]MET imaging, PKM confirmed both significantly higher K1/K2 (1.24 ± 0.79/1.05 ± 0.39) and K3/K4 (11.93 ± 4.28/3.89 ± 1.29) in the tumor region with [18F]Alfatide II. IFS confirmed significant expression of integrin and tumor vascularization in tumor region. CONCLUSION [18F]Alfatide II demonstrates potential in imaging tumor-associated neovascularization in the context of glioblastoma multiforme (GBM), suggesting its utility as a tool for further exploration in neovascular characterization.
Collapse
Affiliation(s)
- Yue Pan
- Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China
- Department of Nuclear Medicine, First Medical Center of Chinese PLA General Hospital, Fuxing Road 28, Beijing, 100853, China
| | - Haodan Dang
- Department of Nuclear Medicine, First Medical Center of Chinese PLA General Hospital, Fuxing Road 28, Beijing, 100853, China
| | - Haoxi Zhou
- Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China
| | - Huaping Fu
- Department of Nuclear Medicine, First Medical Center of Chinese PLA General Hospital, Fuxing Road 28, Beijing, 100853, China
| | - Shina Wu
- Department of Nuclear Medicine, First Medical Center of Chinese PLA General Hospital, Fuxing Road 28, Beijing, 100853, China
| | - Huanhuan Liu
- Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China
| | - Jinming Zhang
- Department of Nuclear Medicine, First Medical Center of Chinese PLA General Hospital, Fuxing Road 28, Beijing, 100853, China
| | - Ruimin Wang
- Department of Nuclear Medicine, First Medical Center of Chinese PLA General Hospital, Fuxing Road 28, Beijing, 100853, China
| | - Yuan Tian
- Department of Radiology, The 8th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Baixuan Xu
- Department of Nuclear Medicine, First Medical Center of Chinese PLA General Hospital, Fuxing Road 28, Beijing, 100853, China.
| |
Collapse
|
10
|
Li H, Dai R, Huang Y, Zhong J, Yan Q, Yang J, Hu K, Zhong Y. [18F]AlF-ND-bisFAPI PET imaging of fibroblast activation protein as a biomarker to monitor the progression of liver fibrosis. Hepatol Commun 2024; 8:e0407. [PMID: 38466884 PMCID: PMC10932522 DOI: 10.1097/hc9.0000000000000407] [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] [Received: 11/14/2023] [Accepted: 02/01/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND Hepatic fibrosis is a progressive disease, which is reversible in the early stages. The current monitoring methods have notable limitations that pose a challenge to early detection. In this study, we evaluated the utility of [18F]AlF-ND-bisFAPI positron emission tomography imaging of fibroblast activation protein (FAP) to monitor the progression of liver fibrosis. METHODS Two mouse models of liver fibrosis were established by bile duct ligation and carbon tetrachloride administration, respectively. Positron emission tomography imaging was performed with the FAP-specific radiotracer [18F]AlF-ND-bisFAPI for the evaluation of rat HSCs and mouse models of fibrosis and combined with histopathology, immunohistochemical staining, and immunoblotting to elucidate the relationships among radioactivity uptake, FAP levels, and liver fibrosis progression. Furthermore, [18F]AlF-ND-bisFAPI autoradiography was performed to assess tracer binding in liver sections from patients with varying degrees of liver fibrosis. RESULTS Cell experiments demonstrated that [18F]AlF-ND-bisFAPI uptake was specific in activated HSCs. Compared with control mice, [18F]AlF-ND-bisFAPI uptake in livers increased in the early stages of fibrosis and increased significantly further with disease progression. Immunohistochemistry and western blot analyses demonstrated that FAP expression increased with fibrosis severity. In accordance with the findings in animal models, ex vivo autoradiography on human fibrotic liver sections showed that radioactivity increased as fibrosis progressed from mild to severe. CONCLUSIONS [18F]AlF-ND-bisFAPI positron emission tomography imaging is a promising noninvasive method for monitoring the progression of liver fibrosis.
Collapse
Affiliation(s)
- Hongsheng Li
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ruoxue Dai
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yueqi Huang
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiawei Zhong
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qingsong Yan
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiaqi Yang
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kongzhen Hu
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Yuhua Zhong
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
11
|
Jeppesen TE, Shao T, Chen J, Patel JS, Zhou X, Kjaer A, Liang SH. Poly(ADP-ribose) polymerase (PARP)-targeted PET imaging in non-oncology application: a pilot study in preclinical models of nonalcoholic steatohepatitis. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2024; 14:41-47. [PMID: 38500745 PMCID: PMC10944370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 02/01/2024] [Indexed: 03/20/2024]
Abstract
Poly(ADP-ribose) polymerase (PARP) activation often indicates a disruptive signal to lipid metabolism, the physiological alteration of which may be implicated in the development of non-alcoholic fatty liver disease. The objective of this study was to evaluate the capability of [68Ga]DOTA-PARPi PET to detect hepatic PARP expression in a non-alcoholic steatohepatitis (NASH) mouse model. In this study, male C57BL/6 mice were subjected to a choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD) for a 12-week period to establish preclinical NASH models. [68Ga]DOTA-PARPi PET imaging of the liver was conducted at the 12-week mark after CDAHFD feeding. Comprehensive histopathological analysis, covering hepatic steatosis, inflammation, fibrosis, along with blood biochemistry, was performed in both NASH models and control groups. Despite the induction of severe inflammation, steatosis and fibrosis in the liver of mice with the CDAHFD-NASH model, PET imaging of NASH with [68Ga]-DOTA-PARPi did not reveal a significantly higher uptake in NASH models compared to the control. This underscores the necessity for further development of new chelator-based PARP1 tracers with high binding affinity to enable the visualization of PARP1 changes in NASH pathology.
Collapse
Affiliation(s)
- Troels E Jeppesen
- Division of Nuclear Medicine, Department of Radiology, Harvard Medical School and Massachusetts General HospitalBoston, MA, USA
- Department of Clinical Physiology, Nuclear Medicine and PET and Cluster for Molecular Imaging, Copenhagen University Hospital - RigshospitaletCopenhagen, Denmark
- Department of Biomedical Sciences, University of CopenhagenCopenhagen, Denmark
| | - Tuo Shao
- Division of Nuclear Medicine, Department of Radiology, Harvard Medical School and Massachusetts General HospitalBoston, MA, USA
- Division of Liver Center and Gastrointestinal, Department of Medicine, Massachusetts General HospitalBoston, MA, USA
| | - Jiahui Chen
- Division of Nuclear Medicine, Department of Radiology, Harvard Medical School and Massachusetts General HospitalBoston, MA, USA
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA, USA
| | - Jimmy S Patel
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA, USA
- Department of Radiation Oncology, Winship Cancer Institute of Emory UniversityAtlanta, GA, USA
| | - Xin Zhou
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA, USA
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine and PET and Cluster for Molecular Imaging, Copenhagen University Hospital - RigshospitaletCopenhagen, Denmark
- Department of Biomedical Sciences, University of CopenhagenCopenhagen, Denmark
| | - Steven H Liang
- Division of Nuclear Medicine, Department of Radiology, Harvard Medical School and Massachusetts General HospitalBoston, MA, USA
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA, USA
| |
Collapse
|
12
|
Eriksson O, Velikyan I. Radiotracers for Imaging of Fibrosis: Advances during the Last Two Decades and Future Directions. Pharmaceuticals (Basel) 2023; 16:1540. [PMID: 38004406 PMCID: PMC10674214 DOI: 10.3390/ph16111540] [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: 08/28/2023] [Revised: 10/13/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
Fibrosis accompanies various pathologies, and there is thus an unmet medical need for non-invasive, sensitive, and quantitative methods for the assessment of fibrotic processes. Currently, needle biopsy with subsequent histological analysis is routinely used for the diagnosis along with morphological imaging techniques, such as computed tomography (CT), magnetic resonance imaging (MRI), and ultrasound (US). However, none of these imaging techniques are sufficiently sensitive and accurate to detect minor changes in fibrosis. More importantly, they do not provide information on fibrotic activity on the molecular level, which is critical for fundamental understanding of the underlying biology and disease course. Molecular imaging technology using positron emission tomography (PET) offers the possibility of imaging not only physiological real-time activity, but also high-sensitivity and accurate quantification. This diagnostic tool is well established in oncology and has exhibited exponential development during the last two decades. However, PET diagnostics has only recently been widely applied in the area of fibrosis. This review presents the progress of development of radiopharmaceuticals for non-invasive detection of fibrotic processes, including the fibrotic scar itself, the deposition of new fibrotic components (fibrogenesis), or the degradation of existing fibrosis (fibrolysis).
Collapse
Affiliation(s)
- Olof Eriksson
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, 751 83 Uppsala, Sweden;
- Antaros Tracer AB, Dragarbrunnsgatan 46, 2 tr, 753 20 Uppsala, Sweden
| | - Irina Velikyan
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, 751 83 Uppsala, Sweden;
- Nuclear Medicine and PET, Department of Surgical Sciences, Uppsala University, 752 85 Uppsala, Sweden
| |
Collapse
|
13
|
Miao M, Miao J, Zhang Y, Zhang J, She M, Zhao M, Miao Q, Yang L, Zhou K, Li Q. An activatable near-infrared molecular reporter for fluoro-photoacoustic imaging of liver fibrosis. Biosens Bioelectron 2023; 235:115399. [PMID: 37210842 DOI: 10.1016/j.bios.2023.115399] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/09/2023] [Accepted: 05/13/2023] [Indexed: 05/23/2023]
Abstract
Noninvasive and accurate detection of liver fibrosis is extremely significant for well-timed intervention and treatment to prevent or reverse its progression. Fluorescence imaging probes hold great potential for imaging of liver fibrosis, but they always encounter the inherent limitation of shallow penetration depth, which compromises their ability of in vivo detection. To overcome this issue, an activatable fluoro-photoacoustic bimodal imaging probe (IP) is herein developed for specific visualization of liver fibrosis. The probe IP is constructed on a near-infrared thioxanthene-hemicyanine dye that is caged with gamma-glutamyl transpeptidase (GGT) responsive substrate and linked with integrin-targeted peptide (cRGD). Such molecular design permits IP to effectively accumulate in the liver fibrosis region through specific recognition of cRGD towards integrin and activate its fluoro-photoacoustic signal after interaction with overexpressed GGT to precisely monitor the liver fibrosis. Thus, our study presents a potential strategy to design dual-target fluoro-photoacoustic imaging probes for noninvasive detection of early-stage liver fibrosis.
Collapse
Affiliation(s)
- Minqian Miao
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Jia Miao
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Yuan Zhang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Jinglin Zhang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Meng She
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Min Zhao
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Qingqing Miao
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Li Yang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China.
| | - Kailong Zhou
- Department of Hand and Foot Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Qing Li
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
14
|
Ren K, Yuan Y, Shao Q, Qiao H. A novel LC-MS/MS approach to [ 19 F]-alfatide and alfatide for application in preclinical pharmacokinetics. Biomed Chromatogr 2023; 37:e5584. [PMID: 36645674 DOI: 10.1002/bmc.5584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/04/2023] [Accepted: 01/11/2023] [Indexed: 01/17/2023]
Abstract
For the further study of [19 F]-alfatide, the development of highly sensitive analytical methods for its determination is an urgent issue. In this paper, a method for simultaneously determining [19 F]-alfatide and alfatide using liquid chromatography/tandem mass spectrometry was created and validated. The plasma samples were pretreated using the protein precipitation method. Poroshell 120 EC-C18 (4.6 × 50 mm, 2.7 μm, Agilent) was used for the separation of the analytes to be measured. The mobile phase, consisting of acetonitrile and water (0.1% acetic acid, 50:50, v/v), was delivered at a flow rate of 0.60 ml/min for sample analysis. Positive electrospray ionization was performed using multiple-reaction monitoring with transitions of m/z 715.2 → 636.6 for [19 F]-alfatide, the ion pairs 700.3 → 851.5 for alfatide and the ion pairs 237.1 → 194.2 for carbamazepine (internal standard). According to the results, the method had high specificity, precision and accuracy as well as an extended linear range. The matrix effect and extraction recovery were also acceptable. The method was successfully applied to the pharmacokinetic study of [19 F]-alfatide in rats.
Collapse
Affiliation(s)
- Kaiyue Ren
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, China
| | - Yanjuan Yuan
- Jiangsu Provincial Institute Of Materia Medica, Nanjing, China
| | - Qing Shao
- Jiangsu Provincial Institute Of Materia Medica, Nanjing, China
| | - Hongqun Qiao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, China
| |
Collapse
|
15
|
Pirasteh A, Periyasamy S, Meudt JJ, Liu Y, Lee LM, Schachtschneider KM, Schook LB, Gaba RC, Mao L, Said A, McMillan AB, Laeseke PF, Shanmuganayagam D. Staging Liver Fibrosis by Fibroblast Activation Protein Inhibitor PET in a Human-Sized Swine Model. J Nucl Med 2022; 63:1956-1961. [PMID: 35450958 PMCID: PMC9730920 DOI: 10.2967/jnumed.121.263736] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/07/2022] [Indexed: 01/11/2023] Open
Abstract
Current methods of staging liver fibrosis have notable limitations. We investigated the utility of PET in staging liver fibrosis by correlating liver uptake of 68Ga-labeled fibroblast activation protein inhibitor (FAPI) with histology in a human-sized swine model. Methods: Five pigs underwent baseline 68Ga-FAPI-46 (68Ga-FAPI) PET/MRI and liver biopsy, followed by liver parenchymal embolization, 8 wk of oral alcohol intake, endpoint 68Ga-FAPI PET/MRI, and necropsy. Regions of interest were drawn on baseline and endpoint PET images, and SUVmean was recorded. At the endpoint, liver sections corresponding to regions of interest were identified and cut out. Fibrosis was histologically evaluated using a modified METAVIR score for swine liver and quantitatively using collagen proportionate area (CPA). Box-and-whisker plots and linear regression were used to correlate SUVmean with METAVIR score and CPA, respectively. Results: Liver 68Ga-FAPI uptake strongly correlated with CPA (r = 0.89, P < 0.001). 68Ga-FAPI uptake was significantly and progressively higher across F2 and F3/F4 fibrosis stages, with a respective median SUVmean of 2.9 (interquartile range [IQR], 2.7-3.8) and 7.6 (IQR, 6.7-10.2) (P < 0.001). There was no significant difference between 68Ga-FAPI uptake of baseline liver and endpoint liver sections staged as F0/F1, with a respective median SUVmean of 1.7 (IQR, 1.3-2.0) and 1.7 (IQR, 1.5-1.8) (P = 0.338). Conclusion: The strong correlation between liver 68Ga-FAPI uptake and the histologic stage of liver fibrosis suggests that 68Ga-FAPI PET can play an impactful role in noninvasive staging of liver fibrosis, pending validation in patients.
Collapse
Affiliation(s)
- Ali Pirasteh
- Radiology and Medical Physics, University of Wisconsin–Madison, Madison, Wisconsin
| | - Sarvesh Periyasamy
- Radiology and Biomedical Engineering, University of Wisconsin–Madison, Madison, Wisconsin
| | - Jennifer Jean Meudt
- Animal and Dairy Sciences, University of Wisconsin–Madison, Madison, Wisconsin
| | - Yongjun Liu
- Pathology and Laboratory Medicine, University of Wisconsin–Madison, Madison, Wisconsin
| | - Laura M. Lee
- Research Animal Resources and Compliance, University of Wisconsin–Madison, Madison, Wisconsin
| | - Kyle M. Schachtschneider
- Radiology and Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois;,National Center for Supercomputing Applications, University of Illinois at Urbana–Champaign, Champaign, Illinois
| | - Lawrence B. Schook
- Animal Sciences, University of Illinois at Chicago, Chicago, Illinois;,Radiology/Interventional Radiology, University of Illinois at Chicago, Chicago, Illinois
| | - Ron C. Gaba
- Radiology/Interventional Radiology, University of Illinois at Chicago, Chicago, Illinois
| | - Lu Mao
- Biostatistics and Medical Informatics, University of Wisconsin–Madison, Madison, Wisconsin
| | - Adnan Said
- Medicine, Gastroenterology, and Hepatology, University of Wisconsin–Madison, Madison, Wisconsin;,William S. Middleton VA Medical Center, Madison, Wisconsin
| | - Alan Blair McMillan
- Radiology and Medical Physics, University of Wisconsin–Madison, Madison, Wisconsin
| | - Paul F. Laeseke
- Radiology and Biomedical Engineering, University of Wisconsin–Madison, Madison, Wisconsin
| | - Dhanansayan Shanmuganayagam
- Animal and Dairy Sciences, University of Wisconsin–Madison, Madison, Wisconsin;,Surgery, University of Wisconsin–Madison, Madison, Wisconsin; and,Center for Biomedical Swine Research and Innovation, University of Wisconsin–Madison, Madison, Wisconsin
| |
Collapse
|
16
|
Zang J, Wen X, Lin R, Zeng X, Wang C, Shi M, Zeng X, Zhang J, Wu X, Zhang X, Miao W, Xu P, Guo Z, Zhang J, Chen X. Synthesis, preclinical evaluation and radiation dosimetry of a dual targeting PET tracer [ 68Ga]Ga-FAPI-RGD. Am J Cancer Res 2022; 12:7180-7190. [PMID: 36276644 PMCID: PMC9576616 DOI: 10.7150/thno.79144] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 09/30/2022] [Indexed: 11/23/2022] Open
Abstract
To enhance tumor uptake and retention, we designed and developed bi-specific heterodimeric radiotracers targeting both FAP and αvβ3, [68Ga]Ga-FAPI-RGD. The present study aimed to evaluate the specificity, pharmacokinetics, and dosimetry of [68Ga]Ga-FAPI-RGD by preclinical and preliminary clinical studies. Methods: FAPI-RGD was designed and synthesized with the quinoline-based FAPI-02 and the cyclic RGDfK peptide. Preclinical pharmacokinetics were determined in Panc02 xenograft model using microPET and biodistribution experiments. The safety and effective dosimetry of [68Ga]Ga-FAPI-RGD was evaluated in 6 cancer patients, and compared with 2-[18F]FDG imaging. Results: The [68Ga]Ga-FAPI-RGD had good stability in saline for at least 4 h, and showed favorable binding affinity and specificity in vitro and in vivo. Compared to [68Ga]Ga-FAPI-02 and [68Ga]Ga-RGDfK, the tumor uptake and retention of [68Ga]Ga-FAPI-RGD were very much enhanced than its monomeric counterparts at all the time points examined by microPET imaging. A total of 6 patients with various malignant tumors were prospectively enrolled. The effective dose of [68Ga]Ga-FAPI-RGD was 1.94E-02 mSv/MBq. The biodistribution of [68Ga]Ga-FAPI-RGD from 0 to 2 h after injection demonstrated rapid and high tumor uptake, prolonged tumor retention, and high tumor-to-background ratios (TBRs) which further increased over time. No significant difference in mean SUVmax of [68Ga]Ga-FAPI-RGD and 2-[18F]FDG was present in primary tumors (8.9±3.2 vs. 10.3 ± 6.9; p = 0.459). Conclusion: The dual targeting PET tracer [68Ga]Ga-FAPI-RGD showed significantly improved tumor uptake and retention, as well as cleaner background over 68Ga-labeled FAPI and RGD monospecific tracers. The first-in-human biodistribution study showed high TBRs over time, suggesting high diagnostic performance and favorable tracer kinetics for potential therapeutic applications.
Collapse
Affiliation(s)
- Jie Zang
- Department of Nuclear Medicine, the First Affiliated Hospital, Fujian Medical University, No. 20 Chazhong Road, Taijiang District, Fuzhou 350005, Fujian Province, China
| | - Xuejun Wen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Rong Lin
- Department of Nuclear Medicine, the First Affiliated Hospital, Fujian Medical University, No. 20 Chazhong Road, Taijiang District, Fuzhou 350005, Fujian Province, China
| | - Xinying Zeng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Chao Wang
- Department of Nuclear Medicine, the First Affiliated Hospital, Fujian Medical University, No. 20 Chazhong Road, Taijiang District, Fuzhou 350005, Fujian Province, China
| | - Mengqi Shi
- Departments of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore.,Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Xueyuan Zeng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Jiaying Zhang
- Department of Nuclear Medicine, the First Affiliated Hospital, Fujian Medical University, No. 20 Chazhong Road, Taijiang District, Fuzhou 350005, Fujian Province, China
| | - Xiaoming Wu
- College of Nuclear Science and Technology, Harbin Engineering University, Harbin 150001, China
| | - Xianzhong Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Weibing Miao
- Department of Nuclear Medicine, the First Affiliated Hospital, Fujian Medical University, No. 20 Chazhong Road, Taijiang District, Fuzhou 350005, Fujian Province, China.,Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, Fujian Province, China
| | - Pengfei Xu
- Departments of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore.,Institute of Clinical Pharmacy & Pharmacology, Jining First People's Hospital, Jining Medical University, Jining 272000, China
| | - Zhide Guo
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Jingjing Zhang
- Departments of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore.,Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.,Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore.,Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.,Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.,Departments of Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117597, Singapore.,Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (ASTAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| |
Collapse
|
17
|
Wan Y, Hu Z, Liu Q, Wang L, Sullivan MA, Gilbert RG. Liver fibrosis alters the molecular structures of hepatic glycogen. Carbohydr Polym 2022; 278:118991. [PMID: 34973794 DOI: 10.1016/j.carbpol.2021.118991] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 01/06/2023]
Abstract
Liver fibrosis (LF) leads to liver failure and short survival. Liver glycogen is a hyperbranched glucose polymer, comprising individual β particles, which can bind together to form aggregated α particles. Glycogen functionality depends on its molecular structure. This study compared the molecular structure of liver glycogen from both LF and healthy rats, and explored underlying mechanisms for observed differences. Glycogen from both groups contained α and β particles; the LF group contained a higher proportion of β particles, with the glycogen containing fewer long chains than seen in the control group. Both glycogen branching enzyme and glycogen phosphorylase showed a significant decrease of activity in the LF group. Transcriptomics and proteomics revealed a functional deficiency of mitochondria in the LF group, which may lead to changes in glycogen structure. These results provide for the first time an understanding of how liver fibrosis affects liver glycogen metabolism and glycogen structure. HYPOTHESIS: We hypothesized that the molecular structure of liver glycogen from a rat model of liver fibrosis would be altered compared to the control group.
Collapse
Affiliation(s)
- Yujun Wan
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Zhenxia Hu
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Qinghua Liu
- Jiangsu Provincial Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu Province 221000, China; State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Liang Wang
- Jiangsu Provincial Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu Province 221000, China; Department of Bioinformatics, School of Medical Informatics and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu Province 221000, China
| | - Mitchell A Sullivan
- Glycation and Diabetes Group, Mater Research Institute, Translational Research Institute, The University of Queensland, Brisbane, Queensland 4072, Australia.
| | - Robert G Gilbert
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Brisbane, Queensland 4072, Australia; Joint International Research Laboratory of Agriculture and Agri-Product Safety, College of Agriculture, Yangzhou University, Yangzhou, Jiangsu 225009, China.
| |
Collapse
|
18
|
Shao T, Chen Z, Rong J, Belov V, Chen J, Jeyarajan A, Deng X, Fu H, Yu Q, Rwema SH, Lin W, Papisov M, Josephson L, Chung RT, Liang SH. [ 18F]MAGL-4-11 positron emission tomography molecular imaging of monoacylglycerol lipase changes in preclinical liver fibrosis models. Acta Pharm Sin B 2022; 12:308-315. [PMID: 35127387 PMCID: PMC8799882 DOI: 10.1016/j.apsb.2021.07.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/06/2021] [Accepted: 07/01/2021] [Indexed: 01/11/2023] Open
Abstract
Monoacylglycerol lipase (MAGL) is a pivotal enzyme in the endocannabinoid system, which metabolizes 2-arachidonoylglycerol (2-AG) into the proinflammatory eicosanoid precursor arachidonic acid (AA). MAGL and other endogenous cannabinoid (EC) degrading enzymes are involved in the fibrogenic signaling pathways that induce hepatic stellate cell (HSC) activation and ECM accumulation during chronic liver disease. Our group recently developed an 18F-labeled MAGL inhibitor ([18F]MAGL-4-11) for PET imaging and demonstrated highly specific binding in vitro and in vivo. In this study, we determined [18F]MAGL-4-11 PET enabled imaging MAGL levels in the bile duct ligation (BDL) and carbon tetrachloride (CCl4) models of liver cirrhosis; we also assessed the hepatic gene expression of the enzymes involved with EC system including MAGL, NAPE-PLD, FAAH and DAGL that as a function of disease severity in these models; [18F]MAGL-4-11 autoradiography was performed to assess tracer binding in frozen liver sections both in animal and human. [18F]MAGL-4-11 demonstrated reduced PET signals in early stages of fibrosis and further significantly decreased with disease progression compared with control mice. We confirmed MAGL and FAAH expression decreases with fibrosis severity, while its levels in normal liver tissue are high; in contrast, the EC synthetic enzymes NAPE-PLD and DAGL are enhanced in these different fibrosis models. In vitro autoradiography further supported that [18F]MAGL-4-11 bound specifically to MAGL in both animal and human fibrotic liver tissues. Our PET ligand [18F]MAGL-4-11 shows excellent sensitivity and specificity for MAGL visualization in vivo and accurately reflects the histological stages of liver fibrosis in preclinical models and human liver tissues.
Collapse
Affiliation(s)
- Tuo Shao
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA,Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Zhen Chen
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Jian Rong
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Vasily Belov
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA,Shriners Hospitals for Children-Boston Boston, MA 02114, USA
| | - Jiahui Chen
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Andre Jeyarajan
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Xiaoyun Deng
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Hualong Fu
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Qingzhen Yu
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Steve H. Rwema
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Wenyu Lin
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Mikhail Papisov
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA,Shriners Hospitals for Children-Boston Boston, MA 02114, USA
| | - Lee Josephson
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Raymond T. Chung
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Corresponding authors. Tel.: +1 617 724 7562, fax: +1 617 643 0446 (Raymond T. Chung); Tel.: +1 617 726 6107, fax: +1 617 726 6165 (Steven H. Liang).
| | - Steven H. Liang
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA,Corresponding authors. Tel.: +1 617 724 7562, fax: +1 617 643 0446 (Raymond T. Chung); Tel.: +1 617 726 6107, fax: +1 617 726 6165 (Steven H. Liang).
| |
Collapse
|
19
|
Wegrzyniak O, Rosestedt M, Eriksson O. Recent Progress in the Molecular Imaging of Nonalcoholic Fatty Liver Disease. Int J Mol Sci 2021; 22:7348. [PMID: 34298967 PMCID: PMC8306605 DOI: 10.3390/ijms22147348] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/30/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
Pathological fibrosis of the liver is a landmark feature in chronic liver diseases, including nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH). Diagnosis and assessment of progress or treatment efficacy today requires biopsy of the liver, which is a challenge in, e.g., longitudinal interventional studies. Molecular imaging techniques such as positron emission tomography (PET) have the potential to enable minimally invasive assessment of liver fibrosis. This review will summarize and discuss the current status of the development of innovative imaging markers for processes relevant for fibrogenesis in liver, e.g., certain immune cells, activated fibroblasts, and collagen depositions.
Collapse
Affiliation(s)
- Olivia Wegrzyniak
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, SE-751 83 Uppsala, Sweden; (O.W.); (M.R.)
| | - Maria Rosestedt
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, SE-751 83 Uppsala, Sweden; (O.W.); (M.R.)
| | - Olof Eriksson
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, SE-751 83 Uppsala, Sweden; (O.W.); (M.R.)
- Antaros Medical AB, SE-431 83 Mölndal, Sweden
| |
Collapse
|
20
|
Dai X, Zeng Y, Zhang H, Gu Z, Gong Q, Luo K. Advances on Nanomedicines for Diagnosis and Theranostics of Hepatic Fibrosis. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202000091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- Xinghang Dai
- Huaxi MR Research Center (HMRRC) Department of Radiology Functional and molecular imaging Key Laboratory of Sichuan Province West China Hospital Sichuan University Chengdu 610041 China
- West China School of Medicine Sichuan University Chengdu 610041 China
| | - Yujun Zeng
- Huaxi MR Research Center (HMRRC) Department of Radiology Functional and molecular imaging Key Laboratory of Sichuan Province West China Hospital Sichuan University Chengdu 610041 China
| | - Hu Zhang
- Huaxi MR Research Center (HMRRC) Department of Radiology Functional and molecular imaging Key Laboratory of Sichuan Province West China Hospital Sichuan University Chengdu 610041 China
- Amgen Bioprocessing Centre Keck Graduate Institute CA 91711 USA
| | - Zhongwei Gu
- Research Unit of Psychoradiology Chinese Academy of Medical Sciences Chengdu 610041 China
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC) Department of Radiology Functional and molecular imaging Key Laboratory of Sichuan Province West China Hospital Sichuan University Chengdu 610041 China
- Research Unit of Psychoradiology Chinese Academy of Medical Sciences Chengdu 610041 China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC) Department of Radiology Functional and molecular imaging Key Laboratory of Sichuan Province West China Hospital Sichuan University Chengdu 610041 China
- Research Unit of Psychoradiology Chinese Academy of Medical Sciences Chengdu 610041 China
| |
Collapse
|
21
|
Koumarianou A, Alexandraki KI, Wallin G, Kaltsas G, Daskalakis K. Pathogenesis and Clinical Management of Mesenteric Fibrosis in Small Intestinal Neuroendocine Neoplasms: A Systematic Review. J Clin Med 2020; 9:E1777. [PMID: 32521677 PMCID: PMC7357094 DOI: 10.3390/jcm9061777] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/04/2020] [Accepted: 06/04/2020] [Indexed: 02/07/2023] Open
Abstract
Mesenteric fibrosis (MF) constitutes an underrecognized sequela in patients with small intestinal neuroendocrine neoplasms (SI-NENs), often complicating the disease clinical course. The aim of the present systematic review, carried out by Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) methodology, is to provide an update in evolving aspects of MF pathogenesis and its clinical management in SI-NENs. Complex and dynamic interactions are present in the microenvironment of tumor deposits in the mesentery. Serotonin, as well as the signaling pathways of certain growth factors play a pivotal, yet not fully elucidated role in the pathogenesis of MF. Clinically, MF often results in significant morbidity by causing either acute complications, such as intestinal obstruction and/or acute ischemia or more chronic conditions involving abdominal pain, venous stasis, malabsorption and malnutrition. Surgical resection in patients with locoregional disease only or symptomatic distant stage disease, as well as palliative minimally invasive interventions in advanced inoperable cases seem clinically meaningful, whereas currently available systemic and/or targeted treatments do not unequivocally affect the development of MF in SI-NENs. Increased awareness and improved understanding of the molecular pathogenesis of MF in SI-NENs may provide better diagnostic and predictive tools for its timely recognition and intervention and also facilitates the development of agents targeting MF.
Collapse
Affiliation(s)
- Anna Koumarianou
- Hematology Oncology Unit, Fourth Department of Internal Medicine, Attikon University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Krystallenia I. Alexandraki
- 1st Department of Propaedeutic Internal Medicine, Endocrine Unit, National and Kapodistrian, University of Athens, 11527 Athens, Greece; (K.I.A.); (G.K.); (K.D.)
| | - Göran Wallin
- Department of Surgery, Faculty of Medicine and Health, Örebro University, 701 85 Örebro, Sweden;
| | - Gregory Kaltsas
- 1st Department of Propaedeutic Internal Medicine, Endocrine Unit, National and Kapodistrian, University of Athens, 11527 Athens, Greece; (K.I.A.); (G.K.); (K.D.)
| | - Kosmas Daskalakis
- 1st Department of Propaedeutic Internal Medicine, Endocrine Unit, National and Kapodistrian, University of Athens, 11527 Athens, Greece; (K.I.A.); (G.K.); (K.D.)
- Department of Surgery, Faculty of Medicine and Health, Örebro University, 701 85 Örebro, Sweden;
| |
Collapse
|