1
|
Shen F, Liu Q, Wang Y, Chen C, Ma H. Comparison of [ 18F] FDG PET/CT and [ 18F]FDG PET/MRI in the Detection of Distant Metastases in Breast Cancer: A Meta-Analysis. Clin Breast Cancer 2025; 25:e113-e123.e4. [PMID: 39438190 DOI: 10.1016/j.clbc.2024.09.015] [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: 03/04/2024] [Revised: 09/03/2024] [Accepted: 09/20/2024] [Indexed: 10/25/2024]
Abstract
PURPOSE This meta-analysis aims to assess and compare the diagnostic effectiveness of [18F] FDG PET/CT and [18F] FDG PET/MRI for distant metastases in breast cancer patients. METHODS A comprehensive search of the PubMed and Embase databases was performed to identify relevant articles until September 22, 2023. Studies were eligible to be included if they assessed the diagnostic performance of [18F] FDG PET/CT and/or [18F] FDG PET/MRI in detecting distant metastases of breast cancer patients. The DerSimonian and Laird method was used to assess sensitivity and specificity, and then transformed through the Freeman-Tukey double arcsine transformation. RESULTS 29 articles consisting of 3779 patients were finally included in this study. The overall sensitivity of [18F] FDG PET/CT in diagnosing distant metastases of breast cancer was 0.96 (95% CI: 0.93-0.98), and the overall specificity was 0.95 (95% CI: 0.92-0.97). The overall sensitivity of [18F] FDG PET/MRI was 1.00 (95% CI: 0.97-1.00), and the specificity was 0.97 (95% CI: 0.94-1.00). The results suggested that [18F] FDG PET/CT and [18F] FDG PET/MRI appears to have similar sensitivity (P = .16) and specificity (P = .30) in diagnosing distant metastases of breast cancer. CONCLUSIONS The results of our meta-analysis indicated that [18F] FDG PET/CT and [18F] FDG PET/MRI in diagnosing distant metastases of breast cancer appear to have similar sensitivity and specificity. Patients who have access to only one of these modalities will not have the accuracy of their staging compromised. In clinical practice, both of these imaging techniques have their respective strengths and limitations, and physicians should take these into account when making the most suitable choice for patients.
Collapse
Affiliation(s)
- Fangqian Shen
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Qi Liu
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yishuang Wang
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Can Chen
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Hu Ma
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
2
|
Shin J, Kim N, Jeon GH, Yun T, Kang BT, Jeong DH. Diagnostic Application of Fluorine-18 Fluorodeoxyglucose Positron Emission Tomography/Computed Tomography in the Treatment of Oral Squamous Cell Carcinoma in an African Pygmy Hedgehog. Animals (Basel) 2024; 14:3679. [PMID: 39765582 PMCID: PMC11672570 DOI: 10.3390/ani14243679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Neoplastic diseases are common in African pygmy hedgehogs (Atelerix albiventris), with squamous cell carcinoma (SCC) being the most prevalent oral tumor. Traditional imaging techniques like computed tomography (CT) have limitations in accurately defining tumor extent and detecting metastasis. In this study, a hedgehog with a suspected oral tumor underwent 18F-FDG PET/CT imaging, revealing a hypermetabolic lesion consistent with SCC and indications of metastatic activity. The lesion was too extensive for a lumpectomy, and suspected metastasis led to the decision for euthanasia due to the impracticability of further treatment. A histopathological examination after autopsy confirmed the diagnosis and metastatic spread, as suggested by PET/CT findings. This is the first case report demonstrating the potential use of 18F-FDG PET/CT in hedgehogs, facilitating the accurate staging and management of oral SCC. Further research is needed to establish PET/CT as a standard diagnostic tool for neoplastic diseases in hedgehogs.
Collapse
Affiliation(s)
- Jaegyeong Shin
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (J.S.); (N.K.); (T.Y.); (B.-T.K.)
| | - Nari Kim
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (J.S.); (N.K.); (T.Y.); (B.-T.K.)
| | - Gwi-Ho Jeon
- Su Veterinary Clinic, Cheongju 28665, Republic of Korea;
| | - Taesik Yun
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (J.S.); (N.K.); (T.Y.); (B.-T.K.)
| | - Byeong-Teck Kang
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (J.S.); (N.K.); (T.Y.); (B.-T.K.)
| | - Dong-Hyuk Jeong
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (J.S.); (N.K.); (T.Y.); (B.-T.K.)
- Wildlife Center of Chungbuk, Cheongju 28116, Republic of Korea
| |
Collapse
|
3
|
Prasad V, Parihar AS. Asymmetric 18F-FDG Uptake in Oropharynx-PET Parameters to Minimize Unnecessary Interventions. JAMA Otolaryngol Head Neck Surg 2024; 150:1087-1088. [PMID: 39207819 DOI: 10.1001/jamaoto.2024.2639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Affiliation(s)
- Vikas Prasad
- Division of Nuclear Medicine, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Missouri
| | - Ashwin Singh Parihar
- Division of Nuclear Medicine, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
4
|
Berriel Diaz M, Rohm M, Herzig S. Cancer cachexia: multilevel metabolic dysfunction. Nat Metab 2024; 6:2222-2245. [PMID: 39578650 DOI: 10.1038/s42255-024-01167-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 10/16/2024] [Indexed: 11/24/2024]
Abstract
Cancer cachexia is a complex metabolic disorder marked by unintentional body weight loss or 'wasting' of body mass, driven by multiple aetiological factors operating at various levels. It is associated with many malignancies and significantly contributes to cancer-related morbidity and mortality. With emerging recognition of cancer as a systemic disease, there is increasing awareness that understanding and treatment of cancer cachexia may represent a crucial cornerstone for improved management of cancer. Here, we describe the metabolic changes contributing to body wasting in cachexia and explain how the entangled action of both tumour-derived and host-amplified processes induces these metabolic changes. We discuss energy homeostasis and possible ways that the presence of a tumour interferes with or hijacks physiological energy conservation pathways. In that context, we highlight the role played by metabolic cross-talk mechanisms in cachexia pathogenesis. Lastly, we elaborate on the challenges and opportunities in the treatment of this devastating paraneoplastic phenomenon that arise from the complex and multifaceted metabolic cross-talk mechanisms and provide a status on current and emerging therapeutic approaches.
Collapse
Affiliation(s)
- Mauricio Berriel Diaz
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Inner Medicine, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Maria Rohm
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Inner Medicine, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Stephan Herzig
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Inner Medicine, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Chair Molecular Metabolic Control, Technical University of Munich, Munich, Germany.
| |
Collapse
|
5
|
Olawuni B, Bode BP. Asparagine as a signal for glutamine sufficiency via asparagine synthetase: a fresh evidence-based framework in physiology and oncology. Am J Physiol Cell Physiol 2024; 327:C1335-C1346. [PMID: 39344414 DOI: 10.1152/ajpcell.00316.2024] [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: 05/12/2024] [Revised: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
Among the 20 proteinogenic amino acids, glutamine (GLN) and asparagine (ASN) represent a unique cohort in containing a terminal amide in their side chain, and share a direct metabolic relationship, with glutamine generating asparagine through the ATP-dependent asparagine synthetase (ASNS) reaction. Circulating glutamine levels and metabolic flux through cells and tissues greatly exceed those for asparagine, and "glutamine addiction" in cancer has likewise received considerable attention. However, historic and recent evidence collectively suggest that in spite of its modest presence, asparagine plays an outsized regulatory role in cellular function. Here, we present a unifying evidence-based hypothesis that the amides constitute a regulatory signaling circuit, with glutamine as a driver and asparagine as a second messenger that allosterically regulates key biochemical and physiological functions, particularly cell growth and survival. Specifically, it is proposed that ASNS serves as a sensor of substrate sufficiency for S-phase entry and progression in proliferating cells. ASNS-generated asparagine serves as a subsequent second messenger that modulates the activity of key regulatory proteins and promotes survival in the face of cellular stress, and serves as a feed-forward driver of S-phase progression in cell growth. We propose that this signaling pathway be termed the amide signaling circuit (ASC) in homage to the SLC1A5-encoded ASCT2 that transports both glutamine and asparagine in a bidirectional manner, and has been implicated in the pathogenesis of a broad spectrum of human cancers. Support for the ASC model is provided by the recent discovery that glutamine is sensed in primary cilia via ASNS during metabolic stress.
Collapse
Affiliation(s)
- Babatunde Olawuni
- Department of Biological Sciences, Northern Illinois University, DeKalb, Illinois, United States
| | - Barrie P Bode
- Department of Biological Sciences, Northern Illinois University, DeKalb, Illinois, United States
- Division of Research and Innovation Partnerships, Northern Illinois University, DeKalb, Illinois, United States
| |
Collapse
|
6
|
Fraum TJ, Sari H, Dias AH, Munk OL, Pyka T, Smith AM, Mawlawi OR, Laforest R, Wang G. Whole-Body Multiparametric PET in Clinical Oncology: Current Status, Challenges, and Opportunities. AJR Am J Roentgenol 2024; 223:e2431712. [PMID: 39230403 DOI: 10.2214/ajr.24.31712] [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: 09/05/2024]
Abstract
The interpretation of clinical oncologic PET studies has historically used static reconstructions based on SUVs. SUVs and SUV-based images have important limitations, including dependence on uptake times and reduced conspicuity of tracer-avid lesions in organs with high background uptake. The acquisition of dynamic PET images enables additional PET reconstructions via Patlak modeling, which assumes that a tracer is irreversibly trapped by tissues of interest. The resulting multiparametric PET images capture a tracer's net trapping rate and apparent volume of distribution, separating the contributions of bound and free tracer fractions to the PET signal captured in the SUV. Potential benefits of multiparametric PET include higher quantitative stability, superior lesion conspicuity, and greater accuracy for differentiating malignant and benign lesions. However, the imaging protocols necessary for multiparametric PET are inherently more complex and time intensive, despite the recent introduction of automated or semiautomated scanner-based reconstruction packages. In this Review, we examine the current state of multiparametric PET in whole-body oncologic imaging. We summarize the Patlak method and relevant tracer kinetics, discuss clinical workflows and protocol considerations, and highlight clinical challenges and opportunities. We aim to help oncologic imagers make informed decisions about whether to implement multiparametric PET in their clinical practices.
Collapse
Affiliation(s)
- Tyler J Fraum
- Department of Radiology, Washington University School of Medicine, 510 S Kingshighway Blvd, St. Louis, MO 63110
| | - Hasan Sari
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Siemens Healthineers International AG, Zurich, Switzerland
| | - André H Dias
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Ole L Munk
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Thomas Pyka
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- TUM School of Medicine and Health, Munich, Germany
| | | | - Osama R Mawlawi
- Department of Imaging Physics, UT MD Anderson Cancer Center, Houston, TX
| | - Richard Laforest
- Department of Radiology, Washington University School of Medicine, 510 S Kingshighway Blvd, St. Louis, MO 63110
| | - Guobao Wang
- Department of Radiology, University of California Davis Health, Sacramento, CA
| |
Collapse
|
7
|
Pashikanti G, Chavan LN, Liebeskind LS, Goodman MM. Synthetic Efforts toward the Synthesis of a Fluorinated Analog of 5-Aminolevulinic Acid: Practical Synthesis of Racemic and Enantiomerically Defined 3-Fluoro-5-aminolevulinic Acid. J Org Chem 2024; 89:12176-12186. [PMID: 39189689 PMCID: PMC11382157 DOI: 10.1021/acs.joc.4c01070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/08/2024] [Accepted: 08/15/2024] [Indexed: 08/28/2024]
Abstract
In 2017, the FDA authorized 5-aminolevulinic acid (5-ALA) for intraoperative optical imaging of suspected high-grade gliomas. This was the first authorized optical imaging agent for brain tumor surgery to enhance the visualization of malignant tissue. Herein we report the synthesis of a racemic and enantiopure fluorinated analog of 5-ALA, i.e., 3-fluoro-5-aminolevulinic acid (3F-5-ALA). We anticipate that these studies will provide the foundation for the future construction of a fluorine-18-labeled 5-ALA PET tracer to be used for functional and metabolic imaging of gliomas.
Collapse
Affiliation(s)
- Gouthami Pashikanti
- Department
of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Lahu N. Chavan
- Department
of Radiology and Imaging Sciences, School of Medicine, Emory University, 1364 Clifton Road NE, Atlanta, Georgia 30322, United States
- Center
for Systems Imaging, Emory University, 1841 Clifton Rd NE, Atlanta, Georgia 30322, United States
| | - Lanny S. Liebeskind
- Department
of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Mark M. Goodman
- Department
of Radiology and Imaging Sciences, School of Medicine, Emory University, 1364 Clifton Road NE, Atlanta, Georgia 30322, United States
- Center
for Systems Imaging, Emory University, 1841 Clifton Rd NE, Atlanta, Georgia 30322, United States
| |
Collapse
|
8
|
Courault P, Zimmer L, Lancelot S. Toward Functional PET Imaging of the Spinal Cord. Semin Nucl Med 2024:S0001-2998(24)00066-7. [PMID: 39181820 DOI: 10.1053/j.semnuclmed.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 07/25/2024] [Indexed: 08/27/2024]
Abstract
At present, spinal cord imaging primarily uses magnetic resonance imaging (MRI) or computed tomography (CT), but the greater sensitivity of positron emission tomography (PET) techniques and the development of new radiotracers are paving the way for a new approach. The substantial rise in publications on PET radiotracers for spinal cord exploration indicates a growing interest in the functional and molecular imaging of this organ. The present review aimed to provide an overview of the various radiotracers used in this indication, in preclinical and clinical settings. Firstly, we outline spinal cord anatomy and associated target pathologies. Secondly, we present the state-of-the-art of spinal cord imaging techniques used in clinical practice, with their respective strengths and limitations. Thirdly, we summarize the literature on radiotracers employed in functional PET imaging of the spinal cord. In conclusion, we propose criteria for an ideal radiotracer for molecular spinal cord imaging, emphasizing the relevance of multimodal hybrid cameras, and particularly the benefits of PET-MRI integration.
Collapse
Affiliation(s)
- Pierre Courault
- Lyon Neuroscience Research Center (CRNL), INSERM, CNRSx, Lyon, France; Hospices Civils de Lyon (HCL), Lyon, France; CERMEP-Imaging Platform, Lyon, France
| | - Luc Zimmer
- Lyon Neuroscience Research Center (CRNL), INSERM, CNRSx, Lyon, France; Hospices Civils de Lyon (HCL), Lyon, France; CERMEP-Imaging Platform, Lyon, France; National Institute for Nuclear Science and Technology (INSTN), CEA, Saclay, France.
| | - Sophie Lancelot
- Lyon Neuroscience Research Center (CRNL), INSERM, CNRSx, Lyon, France; Hospices Civils de Lyon (HCL), Lyon, France; CERMEP-Imaging Platform, Lyon, France
| |
Collapse
|
9
|
Weyts K, Lequesne J, Johnson A, Curcio H, Parzy A, Coquan E, Lasnon C. The impact of introducing deep learning based [ 18F]FDG PET denoising on EORTC and PERCIST therapeutic response assessments in digital PET/CT. EJNMMI Res 2024; 14:72. [PMID: 39126532 DOI: 10.1186/s13550-024-01128-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 07/06/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND [18F]FDG PET denoising by SubtlePET™ using deep learning artificial intelligence (AI) was previously found to induce slight modifications in lesion and reference organs' quantification and in lesion detection. As a next step, we aimed to evaluate its clinical impact on [18F]FDG PET solid tumour treatment response assessments, while comparing "standard PET" to "AI denoised half-duration PET" ("AI PET") during follow-up. RESULTS 110 patients referred for baseline and follow-up standard digital [18F]FDG PET/CT were prospectively included. "Standard" EORTC and, if applicable, PERCIST response classifications by 2 readers between baseline standard PET1 and follow-up standard PET2 as a "gold standard" were compared to "mixed" classifications between standard PET1 and AI PET2 (group 1; n = 64), or between AI PET1 and standard PET2 (group 2; n = 46). Separate classifications were established using either standardized uptake values from ultra-high definition PET with or without AI denoising (simplified to "UHD") or EANM research limited v2 (EARL2)-compliant values (by Gaussian filtering in standard PET and using the same filter in AI PET). Overall, pooling both study groups, in 11/110 (10%) patients at least one EORTCUHD or EARL2 or PERCISTUHD or EARL2 mixed vs. standard classification was discordant, with 369/397 (93%) concordant classifications, unweighted Cohen's kappa = 0.86 (95% CI: 0.78-0.94). These modified mixed vs. standard classifications could have impacted management in 2% of patients. CONCLUSIONS Although comparing similar PET images is preferable for therapy response assessment, the comparison between a standard [18F]FDG PET and an AI denoised half-duration PET is feasible and seems clinically satisfactory.
Collapse
Affiliation(s)
- Kathleen Weyts
- Nuclear Medicine Department, François Baclesse Comprehensive Cancer Centre, UNICANCER, Caen, 3 Avenue du General Harris, BP 45026, Caen Cedex 5, 14076, France.
| | - Justine Lequesne
- Biostatistics Department, François Baclesse Comprehensive Cancer Centre, UNICANCER, Caen, France
| | - Alison Johnson
- Medical Oncology Department, François Baclesse Comprehensive Cancer Centre, UNICANCER, Caen, France
| | - Hubert Curcio
- Medical Oncology Department, François Baclesse Comprehensive Cancer Centre, UNICANCER, Caen, France
| | - Aurélie Parzy
- Medical Oncology Department, François Baclesse Comprehensive Cancer Centre, UNICANCER, Caen, France
| | - Elodie Coquan
- Medical Oncology Department, François Baclesse Comprehensive Cancer Centre, UNICANCER, Caen, France
| | - Charline Lasnon
- Nuclear Medicine Department, François Baclesse Comprehensive Cancer Centre, UNICANCER, Caen, 3 Avenue du General Harris, BP 45026, Caen Cedex 5, 14076, France
- UNICAEN, INSERM 1086 ANTICIPE, Normandy University, Caen, France
| |
Collapse
|
10
|
Lue KH, Chen YH, Chu SC, Lin CB, Wang TF, Liu SH. Prognostic value of combining clinical factors, 18F-FDG PET-based intensity, volumetric features, and deep learning predictor in patients with EGFR-mutated lung adenocarcinoma undergoing targeted therapies: a cross-scanner and temporal validation study. Ann Nucl Med 2024; 38:647-658. [PMID: 38704786 DOI: 10.1007/s12149-024-01936-2] [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: 03/07/2024] [Accepted: 04/22/2024] [Indexed: 05/07/2024]
Abstract
OBJECTIVE To investigate the prognostic value of 18F-FDG PET-based intensity, volumetric features, and deep learning (DL) across different generations of PET scanners in patients with epidermal growth factor receptor (EGFR)-mutated lung adenocarcinoma receiving tyrosine kinase inhibitor (TKI) treatment. METHODS We retrospectively analyzed the pre-treatment 18F-FDG PET of 217 patients with advanced-stage lung adenocarcinoma and actionable EGFR mutations who received TKI as first-line treatment. Patients were separated into analog (n = 166) and digital (n = 51) PET cohorts. 18F-FDG PET-derived intensity, volumetric features, ResNet-50 DL of the primary tumor, and clinical variables were used to predict progression-free survival (PFS). Independent prognosticators were used to develop prediction model. Model was developed and validated in the analog and digital PET cohorts, respectively. RESULTS In the analog PET cohort, female sex, stage IVB status, exon 19 deletion, SUVmax, metabolic tumor volume, and positive DL prediction independently predicted PFS. The model devised from these six prognosticators significantly predicted PFS in the analog (HR = 1.319, p < 0.001) and digital PET cohorts (HR = 1.284, p = 0.001). Our model provided incremental prognostic value to staging status (c-indices = 0.738 vs. 0.558 and 0.662 vs. 0.598 in the analog and digital PET cohorts, respectively). Our model also demonstrated a significant prognostic value for overall survival (HR = 1.198, p < 0.001, c-index = 0.708 and HR = 1.256, p = 0.021, c-index = 0.664 in the analog and digital PET cohorts, respectively). CONCLUSIONS Combining 18F-FDG PET-based intensity, volumetric features, and DL with clinical variables may improve the survival stratification in patients with advanced EGFR-mutated lung adenocarcinoma receiving TKI treatment. Implementing the prediction model across different generations of PET scanners may be feasible and facilitate tailored therapeutic strategies for these patients.
Collapse
Affiliation(s)
- Kun-Han Lue
- Department of Medical Imaging and Radiological Sciences, Tzu Chi University of Science and Technology, No.880, Sec.2, Chien-kuo Rd., Hualien, 970302, Taiwan
| | - Yu-Hung Chen
- Department of Medical Imaging and Radiological Sciences, Tzu Chi University of Science and Technology, No.880, Sec.2, Chien-kuo Rd., Hualien, 970302, Taiwan.
- Department of Nuclear Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No.707, Sec.3, Zhongyang Rd, Hualien, 970473, Taiwan.
- School of Medicine, College of Medicine, Tzu Chi University, No.701, Sec.3, Zhongyang Rd, Hualien, 970473, Taiwan.
| | - Sung-Chao Chu
- School of Medicine, College of Medicine, Tzu Chi University, No.701, Sec.3, Zhongyang Rd, Hualien, 970473, Taiwan
- Department of Hematology and Oncology, Buddhist Tzu Chi Medical Foundation, Hualien Tzu Chi Hospital, Hualien, Taiwan
| | - Chih-Bin Lin
- School of Medicine, College of Medicine, Tzu Chi University, No.701, Sec.3, Zhongyang Rd, Hualien, 970473, Taiwan
- Department of Internal Medicine, Buddhist Tzu Chi Medical Foundation, Hualien Tzu Chi Hospital, Hualien, Taiwan
| | - Tso-Fu Wang
- School of Medicine, College of Medicine, Tzu Chi University, No.701, Sec.3, Zhongyang Rd, Hualien, 970473, Taiwan
- Department of Hematology and Oncology, Buddhist Tzu Chi Medical Foundation, Hualien Tzu Chi Hospital, Hualien, Taiwan
| | - Shu-Hsin Liu
- Department of Medical Imaging and Radiological Sciences, Tzu Chi University of Science and Technology, No.880, Sec.2, Chien-kuo Rd., Hualien, 970302, Taiwan
- Department of Nuclear Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No.707, Sec.3, Zhongyang Rd, Hualien, 970473, Taiwan
| |
Collapse
|
11
|
Saha PS, Yan J, Zhu C. Diffuse reflectance spectroscopy for optical characterizations of orthotopic head and neck cancer models in vivo. BIOMEDICAL OPTICS EXPRESS 2024; 15:4176-4189. [PMID: 39022549 PMCID: PMC11249676 DOI: 10.1364/boe.528608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 07/20/2024]
Abstract
We demonstrated an easy-to-build, portable diffuse reflectance spectroscopy device along with a Monte Carlo inverse model to quantify tissue absorption and scattering-based parameters of orthotopic head and neck cancer models in vivo. Both tissue-mimicking phantom studies and animal studies were conducted to verify the optical spectroscopy system and Monte Carlo inverse model for the accurate extraction of tissue optical properties. For the first time, we reported the tissue absorption and scattering coefficients of mouse normal tongue tissues and tongue tumor tissues. Our in vivo animal studies showed reduced total hemoglobin concentration, lower tissue vascular oxygen saturation, and increased tissue scattering in the orthotopic tongue tumors compared to the normal tongue tissues. Our data also showed that mice tongue tumors with different sizes may have significantly different tissue absorption and scattering-based parameters. Small tongue tumors (volume was ∼60 mm3) had increased absorption coefficients, decreased reduced-scattering coefficients, and increased total hemoglobin concentrations compared to tiny tongue tumors (volume was ∼18 mm3). These results demonstrated the potential of diffuse reflectance spectroscopy to noninvasively evaluate tumor biology using orthotopic tongue cancer models for future head and neck cancer research.
Collapse
Affiliation(s)
- Pranto Soumik Saha
- Department of Biomedical Engineering, University of Kentucky
, Lexington, KY 40506, USA
| | - Jing Yan
- Department of Biomedical Engineering, University of Kentucky
, Lexington, KY 40506, USA
| | - Caigang Zhu
- Department of Biomedical Engineering, University of Kentucky
, Lexington, KY 40506, USA
| |
Collapse
|
12
|
Salva de Torres C, Baraibar I, Saoudi González N, Ros J, Salva F, Rodríguez-Castells M, Alcaraz A, García A, Tabernero J, Élez E. Current and Emerging Treatment Paradigms in Colorectal Cancer: Integrating Hallmarks of Cancer. Int J Mol Sci 2024; 25:6967. [PMID: 39000083 PMCID: PMC11241496 DOI: 10.3390/ijms25136967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
The treatment of unresectable metastatic colorectal cancer has evolved over the last two decades, as knowledge of cancer biology has broadened and new targets have emerged. 'The Hallmarks of Cancer' illustrate the crucial capabilities acquired by cells to become malignant and represent the evolution of knowledge of tumor biology. This review integrates these novel targets and therapies into selected hallmarks: sustaining proliferative signaling, inducing vasculature, avoiding immune destruction, genome instability and mutation, reprogramming cellular metabolism, and resisting cell death. The different strategies and combinations under study are based on treatments with anti-EGFR, anti-VEGF, and anti-HER2 agents, KRAS G12C inhibitors, BRAF and MEK inhibitors, and immune checkpoint inhibitors. However, new approaches are emerging, including vaccines, WEE1 inhibitors, and PARP inhibitors, among others. The further deciphering of cancer biology will unravel new targets, develop novel therapies, and improve patients' outcomes.
Collapse
Affiliation(s)
| | - Iosune Baraibar
- Medical Oncology Department, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology (VHIO), E-08035 Barcelona, Spain; (I.B.); (N.S.G.); (J.R.); (F.S.); (M.R.-C.), (J.T.)
| | - Nadia Saoudi González
- Medical Oncology Department, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology (VHIO), E-08035 Barcelona, Spain; (I.B.); (N.S.G.); (J.R.); (F.S.); (M.R.-C.), (J.T.)
| | - Javier Ros
- Medical Oncology Department, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology (VHIO), E-08035 Barcelona, Spain; (I.B.); (N.S.G.); (J.R.); (F.S.); (M.R.-C.), (J.T.)
| | - Francesc Salva
- Medical Oncology Department, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology (VHIO), E-08035 Barcelona, Spain; (I.B.); (N.S.G.); (J.R.); (F.S.); (M.R.-C.), (J.T.)
| | - Marta Rodríguez-Castells
- Medical Oncology Department, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology (VHIO), E-08035 Barcelona, Spain; (I.B.); (N.S.G.); (J.R.); (F.S.); (M.R.-C.), (J.T.)
| | - Adriana Alcaraz
- Vall d’Hebron Institute of Oncology (VHIO), E-08035 Barcelona, Spain; (A.A.); (A.G.)
| | - Ariadna García
- Vall d’Hebron Institute of Oncology (VHIO), E-08035 Barcelona, Spain; (A.A.); (A.G.)
| | - Josep Tabernero
- Medical Oncology Department, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology (VHIO), E-08035 Barcelona, Spain; (I.B.); (N.S.G.); (J.R.); (F.S.); (M.R.-C.), (J.T.)
| | - Elena Élez
- Medical Oncology Department, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology (VHIO), E-08035 Barcelona, Spain; (I.B.); (N.S.G.); (J.R.); (F.S.); (M.R.-C.), (J.T.)
| |
Collapse
|
13
|
Zhou M, Yin X, Chen B, Hu S, Zhou W. A PET probe targeting polyamine transport system for precise tumor diagnosis and therapy. Asian J Pharm Sci 2024; 19:100924. [PMID: 38903130 PMCID: PMC11186966 DOI: 10.1016/j.ajps.2024.100924] [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: 12/25/2023] [Revised: 02/02/2024] [Accepted: 03/04/2024] [Indexed: 06/22/2024] Open
Abstract
Polyamine metabolism dysregulation is a hallmark of many cancers, offering a promising avenue for early tumor theranostics. This study presents the development of a nuclear probe derived from spermidine (SPM) for dual-purpose tumor PET imaging and internal radiation therapy. The probe, radiolabeled with either [68Ga]Ga for diagnostic applications or [177Lu]Lu for therapeutic use, was synthesized with exceptional purity, stability, and specific activity. Extensive testing involving 12 different tumor cell lines revealed remarkable specificity towards B16 melanoma cells, showcasing outstanding tumor localization and target-to-non-target ratio. Mechanistic investigations employing polyamines, non-labeled precursor, and polyamine transport system (PTS) inhibitor, consistently affirmed the probe's targetability through recognition of the PTS. Notably, while previous reports indicated PTS upregulation in various tumor types for targeted therapy, this study observed no positive signals, highlighting a concentration-dependent discrepancy between targeting for therapy and diagnosis. Furthermore, when labeled with [177Lu], the probe demonstrated its therapeutic potential by effectively controlling tumor growth and extending mouse survival. Investigations into biodistribution, excretion, and biosafety in healthy humans laid a robust foundation for clinical translation. This study introduces a versatile SPM-based nuclear probe with applications in precise tumor theranostics, offering promising prospects for clinical implementation.
Collapse
Affiliation(s)
- Ming Zhou
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Xiaoqin Yin
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Bei Chen
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Shuo Hu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha 410008, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha 410008, China
| |
Collapse
|
14
|
Ayat P, Sawass Najjar D, Alkaissi H, Gill H, Otey J, AlFaraj M, McFarlane SI. Differential Effect of Hyperglycemia on the Odds of Cancer Among the Adult Population of the United States. Cureus 2024; 16:e63061. [PMID: 39050345 PMCID: PMC11268948 DOI: 10.7759/cureus.63061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
Objective Accumulating evidence indicates a relationship between diabetes and cancer risk, with obesity, insulin resistance, and hyperglycemia being implicated as the major underlying pathogenetic mechanisms of increased cancer risk among people with diabetes. We aim to assess the differential effect of dysglycemia (prediabetes and diabetes) on the strength of association (odds) of cancer amongst the adult US diabetic population. Material and methods We analyzed data from the 1997-2013 National Health Interview Survey (NHIS) dataset, which applies a multistage area probability sampling design. We used descriptive statistics and logistic regression analyses to test the strengths of the association between diabetes, prediabetes, and cancer before and after adjusting for major risk factors for cancer, including age and body mass index (BMI). Results A total of 722,532 individuals were surveyed, with a mean age of 47.18 ±0.3 years (±SEM) and a BMI of 26.9 ±0.01 kg/m2. Between 1997 and 2013, BMI increased from 26.0 to 27.4 kg/m2, the diabetes rate increased from 4.1% to 7.6%, and associated cancer rates increased from 6.6% to 9.0%. Body mass index was 27.1 vs. 26.8 kg/m2, P < 0.01, for those with and without cancer, respectively. The unadjusted odds ratio for cancer was 1.92 (1.78-2.08) (95% CI) and 2.20 (2.13-2.27) for prediabetes and diabetes, respectively. After adjusting for age, BMI, race, and cigarette smoking, the odds ratio for cancer was 1.12 (1.03-1.22), P < 0.01, and 1.15 (1.11-1.18), P <0.01, for prediabetes and diabetes, respectively. Conclusion Among US adults, the increasing rate of diabetes over the years was associated with an increased rate of cancer. Diabetes and prediabetes have a graduated effect on cancer risk. While obesity is generally implicated as an underlying pathophysiologic link between diabetes and cancer, our study showed a modest difference in BMI between those with and without cancer. In addition, the effect of diabetes and prediabetes on the odds of cancer persisted after adjusting for BMI. These data collectively suggest that hyperglycemia is an attractive pathophysiologic mechanism that may play a role in increasing the odds of cancer among diabetic and prediabetic populations. Our study is consistent with the accumulating evidence implicating hyperglycemia in the pathogenesis of cancer, where glucose is used in PET scanning to detect cancer (the Warburg effect), and the ketogenic diet appears to be useful in cancer management, enhancing the effect of chemotherapeutic agents.
Collapse
Affiliation(s)
- Parinaz Ayat
- Internal Medicine, State University of New York (SUNY) Downstate Health Science University, New York City, USA
| | - Diana Sawass Najjar
- Internal Medicine, State University of New York (SUNY) Downstate Health Science University, New York City, USA
| | - Hussam Alkaissi
- Internal Medicine, National Institutes of Health (NIH) National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Bethesda, USA
| | - Harjinder Gill
- Internal Medicine, State University of New York (SUNY) Downstate Health Science University, New York City, USA
| | - Jennifer Otey
- Medicine, State University of New York (SUNY) Downstate Health Science University, New York City, USA
| | - Marwa AlFaraj
- Integrative Medicine, State University of New York (SUNY) Downstate Health Science University, New York City, USA
| | - Samy I McFarlane
- Internal Medicine, Endocrine Division, State University of New York (SUNY) Downstate Health Science University, New York City, USA
| |
Collapse
|
15
|
Zhang YQ, Zhang W, Kong XT, Hai WX, Guo R, Zhang M, Zhang SL, Li B. The therapeutic effect of a novel GAPDH inhibitor in mouse model of breast cancer and efficacy monitoring by molecular imaging. Cancer Cell Int 2024; 24:188. [PMID: 38811918 PMCID: PMC11138053 DOI: 10.1186/s12935-024-03361-x] [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: 12/07/2023] [Accepted: 05/07/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Breast cancer is a serious threat to women's health with high morbidity and mortality. The development of more effective therapies for the treatment of breast cancer is strongly warranted. Growing evidence suggests that targeting glucose metabolism may be a promising cancer treatment strategy. We previously identified a new glyceraldehyde-3-phosphate dehydrogenase (GAPDH) inhibitor, DC-5163, which shows great potential in inhibiting tumor growth. Here, we evaluated the anticancer potential of DC-5163 in breast cancer cells. METHODS The effects of DC-5163 on breast cancer cells were investigated in vitro and in vivo. Seahorse, glucose uptake, lactate production, and cellular ATP content assays were performed to examine the impact of DC-5163 on cellular glycolysis. Cell viability, colony-forming ability, cell cycle, and apoptosis were assessed by CCK8 assay, colony formation assay, flow cytometry, and immunoblotting respectively. The anticancer activity of DC-5163 in vivo was evaluated in a mouse breast cancer xenograft model. RESULTS DC-5163 suppressed aerobic glycolysis and reduced energy supply of breast cancer cells, thereby inhibiting breast cancer cell growth, inducing cell cycle arrest in the G0/G1 phase, and increasing apoptosis. The therapeutic efficacy was assessed using a breast cancer xenograft mouse model. DC-5163 treatment markedly suppressed tumor growth in vivo without inducing evident systemic toxicity. Micro-PET/CT scans revealed a notable reduction in tumor 18F-FDG and 18F-FLT uptake in the DC-5163 treatment group compared to the DMSO control group. CONCLUSIONS Our results suggest that DC-5163 is a promising GAPDH inhibitor for suppressing breast cancer growth without obvious side effects. 18F-FDG and 18F-FLT PET/CT can noninvasively assess the levels of glycolysis and proliferation in tumors following treatment with DC-5163.
Collapse
Affiliation(s)
- Yun-Qi Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, 030000, China
| | - Wei Zhang
- Drug Discovery and Design Canter, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Xiang-Tai Kong
- Drug Discovery and Design Canter, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Wang-Xi Hai
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, 030000, China
| | - Rui Guo
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, 030000, China
| | - Min Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, 030000, China
| | - Su-Lin Zhang
- Drug Discovery and Design Canter, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China.
| | - Biao Li
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, China.
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, 030000, China.
| |
Collapse
|
16
|
Lee H, Hwang KH. Focal incidental colorectal fluorodeoxyglucose uptake: Should it be spotlighted? World J Clin Cases 2024; 12:2466-2474. [PMID: 38817235 PMCID: PMC11135452 DOI: 10.12998/wjcc.v12.i15.2466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/17/2024] [Accepted: 04/12/2024] [Indexed: 05/14/2024] Open
Abstract
Fluorine-18 fluorodeoxyglucose (F-18 FDG) positron emission tomography/computed tomography (PET/CT) has emerged as a cornerstone in cancer evaluation imaging, with a well-established history spanning several years. This imaging modality, encompassing the examination of the body from the base of the skull to the upper thighs, comprehensively covers the chest and abdominopelvic regions in a singular scan, allowing for a holistic assessment of nearly the entire body, including areas of marginal interest. The inherent advantage of this expansive scan range lies in its potential to unveil unexpected incidental abnormal hypermetabolic areas. The identification of incidental focal FDG uptake within colorectal regions during PET/CT scans is not an uncommon occurrence, albeit fraught with challenges associated with non-specific FDG uptake. The presence of benign colorectal lesions or physiological uptake poses a particular obstacle, as these may manifest with FDG uptake levels that mimic malignancy. Consequently, physicians are confronted with a diagnostic dilemma when encountering abnormal FDG uptake in unexpected colorectal areas. Existing studies have presented divergent results concerning these uptakes. Standardized uptake value and its derivatives have served as pivotal metrics in quantifying FDG uptake in PET images. In this article, we aim to succinctly explore the distinctive characteristics of FDG, delve into imaging findings, and elucidate the clinical significance of incidental focal colorectal uptake. This discussion aims to contribute valuable insights into the nuanced interpretation of such findings, fostering a comprehensive understanding.
Collapse
Affiliation(s)
- Haejun Lee
- Department of Nuclear Medicine, Gachon University College of Medicine, Gil Medical Center, Incheon 21565, South Korea
| | - Kyung-Hoon Hwang
- Department of Nuclear Medicine, Gachon University College of Medicine, Gil Medical Center, Incheon 21565, South Korea
| |
Collapse
|
17
|
Lee H, Hwang KH. Unexpected focal fluorodeoxyglucose uptake in main organs; pass through or pass by? World J Clin Cases 2024; 12:1885-1899. [PMID: 38660550 PMCID: PMC11036514 DOI: 10.12998/wjcc.v12.i11.1885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/31/2024] [Accepted: 03/21/2024] [Indexed: 04/11/2024] Open
Abstract
Since the inception of fluorine-18 fluorodeoxyglucose (F-18 FDG), positron emission tomography/computed tomography (PET/CT) utilizing F-18 FDG has become widely accepted as a valuable imaging modality in the field of oncology, with global prevalence in clinical practice. Given that a single Torso PET/CT scan encompasses the anatomical region from the skull base to the upper thigh, the detection of incidental abnormal focal hypermetabolism in areas of limited clinical interest is both feasible and not uncommon. Numerous investigations have been undertaken to delineate the distinctive features of these findings, yet the outcomes have proven inconclusive. The incongruent results of these studies present a challenge for physicians, leaving them uncertain about the appropriate course of action. This article provides a succinct overview of the characteristics of fluorodeoxyglucose, followed by a comprehensive discussion of the imaging findings and clinical significance associated with incidental focal abnormal F-18 FDG activity in several representative organs. In conclusion, while the prevalence of unrecognized malignancy varies across organs, malignancies account for a substantial proportion, ranging from approximately one-third to over half, of incidental focal uptake. In light of these rates, physicians are urged to exercise vigilance in not disregarding unexpected uptake, facilitating more assured clinical decisions, and advocating for further active evaluation.
Collapse
Affiliation(s)
- Haejun Lee
- Department of Nuclear Medicine, Gachon University College of Medicine, Gil Medical Center, Incheon 21565, South Korea
| | - Kyung-Hoon Hwang
- Department of Nuclear Medicine, Gachon University College of Medicine, Gil Medical Center, Incheon 21565, South Korea
| |
Collapse
|
18
|
Chen T, Mao S, Ma J, Tang X, Zhu R, Mao D, Zhu X, Pan Q. Proximity-Enhanced Functional Imaging Analysis of Engineered Tumors. Angew Chem Int Ed Engl 2024; 63:e202319117. [PMID: 38305848 DOI: 10.1002/anie.202319117] [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: 12/12/2023] [Revised: 02/01/2024] [Accepted: 02/01/2024] [Indexed: 02/03/2024]
Abstract
Functional imaging (FI) techniques have revolutionized tumor imaging by providing information on specific tumor functions, such as glycometabolism. However, tumor cells lack unique molecular characteristics at the molecular level and metabolic pathways, resulting in limited metabolic differences compared to normal cells and increased background signals from FI. To address this limitation, we developed a novel imaging technique termed proximity-enhanced functional imaging (PEFI) for accurate visualization of tumors. By using "two adjacent chemically labeled glycoproteins" as output signals, we significantly enhance the metabolic differences between tumor and normal cells by PEFI, thereby reducing the background signals for analysis and improving the accuracy of tumor functional imaging. Our results demonstrate that PEFI can accurately identify tumors at the cellular, tissue, and animal level, and has potential value in clinical identification and analysis of tumor cells and tissues, as well as in the guidance of clinical tumor resection surgery.
Collapse
Affiliation(s)
- Tianshu Chen
- Clinical Laboratory, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Shanghai Key Laboratory of Clinical Molecular Diagnostics for Pediatrics, Shanghai, 200127, P. R. China
| | - Siwei Mao
- Clinical Laboratory, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Shanghai Key Laboratory of Clinical Molecular Diagnostics for Pediatrics, Shanghai, 200127, P. R. China
| | - Ji Ma
- Clinical Laboratory, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Shanghai Key Laboratory of Clinical Molecular Diagnostics for Pediatrics, Shanghai, 200127, P. R. China
| | - Xiaochen Tang
- Clinical Laboratory, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Shanghai Key Laboratory of Clinical Molecular Diagnostics for Pediatrics, Shanghai, 200127, P. R. China
| | - Rui Zhu
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, 200072, P. R. China
| | - Dongsheng Mao
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, 200072, P. R. China
| | - Xiaoli Zhu
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, 200072, P. R. China
| | - Qiuhui Pan
- Clinical Laboratory, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Shanghai Key Laboratory of Clinical Molecular Diagnostics for Pediatrics, Shanghai, 200127, P. R. China
- Sanya Women and Children's Hospital Managed by Shanghai Children's Medical Center, Sanya, 572000, P. R. China
| |
Collapse
|
19
|
Nitta N, Yoshimatsu R, Iwasa H, Nakaji K, Miyatake K, Nishimori M, Matsumoto T, Yamanishi T, Yamagami T. Difference in arterial FDG accumulation in healthy study participants between digital PET/CT and standard PET/CT. Ann Nucl Med 2024; 38:96-102. [PMID: 37851300 DOI: 10.1007/s12149-023-01875-4] [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: 05/15/2023] [Accepted: 09/27/2023] [Indexed: 10/19/2023]
Abstract
OBJECTIVE To evaluate the differences in FDG accumulation in arteries throughout the body between digital and standard PET/CT. METHODS Forty-six people who had FDG-PET examinations with a digital PET/CT scanner for health screening between April 2020 and March 2021 and had previous examinations with a standard PET/CT scanner were the study participants. FDG accumulation in arteries throughout the body was visually assessed in each segment. Scan was considered positive when arterial FDG accumulation was equal to or greater than that of the liver. The positivity rates for general arteries and each arterial segment were compared between the two kinds of scanners. If any one of the arterial segments was considered positive, the general arteries were classified as positive. Moreover, the rate of change in results from the standard PET/CT to the digital scanner in the same individual (negative to positive, positive to negative) was examined. RESULTS In the evaluation of general arteries, the positivity rates were 21.7% (10 cases) for the standard PET/CT, whereas positive rates were 97.8% (45 cases) for the digital PET/CT (p < 0.001). In all arterial segments, the positivity rate was significantly higher with the digital PET/CT compared to the standard PET/CT; those with the digital PET/CT were, respectively, 95.7%, 87.0%, 73.9%, 37.0%, 34.8%, and 21.7% in the femoral, brachial, aortic, subclavian, iliac, and carotid arteries. On the other hand, those with the standard PET/CT were 13.0%, 13.0%, 19.6%, 2.2%, 0%, and 4.4% in segments in the above order. Changes from negative to positive were shown in many cases; 35 cases (76.0%) of general arteries, 38 cases (82.6%) for the femoral artery, and 34 cases (73.9%) for the brachial artery. The exception was one case in which a change from positive to negative was confirmed in the carotid artery. In all arteries considered to be positive, FDG accumulation was not greater than but was equal to that in the liver with both scanners. CONCLUSIONS Arterial FDG accumulation was significantly higher with digital PET/CT compared to conventional PET/CT. With digital PET/CT, an arterial FDG accumulation equal to the liver may not to be considered as abnormal accumulation.
Collapse
Affiliation(s)
- Noriko Nitta
- Department of Diagnostic and Interventional Radiology, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| | - Rika Yoshimatsu
- Department of Diagnostic and Interventional Radiology, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan.
| | - Hitomi Iwasa
- Department of Diagnostic and Interventional Radiology, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| | - Kousuke Nakaji
- Department of Diagnostic and Interventional Radiology, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| | - Kana Miyatake
- Department of Diagnostic and Interventional Radiology, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| | - Miki Nishimori
- Department of Diagnostic and Interventional Radiology, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| | - Tomohiro Matsumoto
- Department of Diagnostic and Interventional Radiology, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| | - Tomoaki Yamanishi
- Department of Diagnostic and Interventional Radiology, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| | - Takuji Yamagami
- Department of Diagnostic and Interventional Radiology, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| |
Collapse
|
20
|
Ouvrard E, Kaseb A, Poterszman N, Porot C, Somme F, Imperiale A. Nuclear medicine imaging for bone metastases assessment: what else besides bone scintigraphy in the era of personalized medicine? Front Med (Lausanne) 2024; 10:1320574. [PMID: 38288299 PMCID: PMC10823373 DOI: 10.3389/fmed.2023.1320574] [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: 10/12/2023] [Accepted: 12/28/2023] [Indexed: 01/31/2024] Open
Abstract
Accurate detection and reliable assessment of therapeutic responses in bone metastases are imperative for guiding treatment decisions, preserving quality of life, and ultimately enhancing overall survival. Nuclear imaging has historically played a pivotal role in this realm, offering a diverse range of radiotracers and imaging modalities. While the conventional bone scan using 99mTc marked bisphosphonates has remained widely utilized, its diagnostic performance is hindered by certain limitations. Positron emission tomography, particularly when coupled with computed tomography, provides improved spatial resolution and diagnostic performance with various pathology-specific radiotracers. This review aims to evaluate the performance of different nuclear imaging modalities in clinical practice for detecting and monitoring the therapeutic responses in bone metastases of diverse origins, addressing their limitations and implications for image interpretation.
Collapse
Affiliation(s)
- Eric Ouvrard
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
| | - Ashjan Kaseb
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
- Radiology, College of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Nathan Poterszman
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
| | - Clémence Porot
- Radiopharmacy, Institut de Cancérologie Strasbourg Europe (ICANS), Strasbourg, France
| | - Francois Somme
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
| | - Alessio Imperiale
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
- IPHC, UMR 7178, CNRS/Unistra, Strasbourg, France
| |
Collapse
|
21
|
Omokawa M, Kimura H, Arimitsu K, Yagi Y, Hattori Y, Kawashima H, Naito Y, Yasui H. Synthesis and Biological Evaluation of a Novel Sugar-Conjugated Platinum(II) Complex Having a Tumor-Targeting Effect. ACS OMEGA 2024; 9:879-886. [PMID: 38222559 PMCID: PMC10785272 DOI: 10.1021/acsomega.3c06922] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/23/2023] [Accepted: 12/05/2023] [Indexed: 01/16/2024]
Abstract
We designed and synthesized a novel platinum complex conjugated with 2-fluorinated 2-deoxyglucoside, named FGC-Pt, to capitalize on the Warburg effect and metabolic trapping properties of [18F]2-deoxy-2-fluoro-d-glucose ([18F]FDG). Then, we conducted comprehensive in vitro and in vivo studies to evaluate the effects of FGC-Pt. In vitro cytotoxicity assays using HeLa cells revealed that FGC-Pt exhibited concentration-dependent cytotoxicity, even though its cytotoxic effect was less pronounced than that of cisplatin. In the evaluation of in vivo biodistribution in mice, platinum concentration in tumors and major organs (muscle, bone, blood, liver, and kidney) and the ratio of platinum concentration in tumors to major organs following the tail vein injection of FGC-Pt and cisplatin suggest that FGC-Pt is more retained in tumors than in other organs and tends to accumulate in tumors more than cisplatin. Furthermore, an in vivo assessment of the antitumor effect conducted in A549 cell-bearing mice demonstrated that FGC-Pt possesses substantial potential as an antitumor agent. It exhibited a tumor growth-inhibitory effect comparable to that of cisplatin while inducing lower toxicity, as evidenced by lower weight loss after administration. Herein, we successfully produced a novel compound with a tumor-growth-inhibitory effect comparable to that of cisplatin and low toxicity.
Collapse
Affiliation(s)
- Marina Omokawa
- Laboratory
of Analytical and Bioinorganic Chemistry, Division of Analytical and
Physical Sciences, Kyoto Pharmaceutical
University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan
| | - Hiroyuki Kimura
- Laboratory
of Analytical and Bioinorganic Chemistry, Division of Analytical and
Physical Sciences, Kyoto Pharmaceutical
University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan
- Division
of Probe Chemistry for Disease Analysis/Central Institute for Radioisotope
Science, Research Center for Experimental Modeling of Human Disease, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8640, Japan
| | - Kenji Arimitsu
- Laboratory
of Analytical and Bioinorganic Chemistry, Division of Analytical and
Physical Sciences, Kyoto Pharmaceutical
University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan
- Laboratory
of Medicinal Chemistry, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka 584-8540, Japan
| | - Yusuke Yagi
- Laboratory
of Analytical and Bioinorganic Chemistry, Division of Analytical and
Physical Sciences, Kyoto Pharmaceutical
University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan
- Department
of Radiological Technology, Faculty of Medicinal Science, Kyoto College of Medical Science, 1-3 Imakita, Oyama-higashi, Sonobe, Nantan 622-0041, Kyoto, Japan
| | - Yasunao Hattori
- Center
for Instrumental Analysis, Kyoto Pharmaceutical
University, 1 Shichono-cho,
Misasagi, Yamashina-ku, Kyoto 607-8412, Japan
| | - Hidekazu Kawashima
- Radioisotope
Research Center, Kyoto Pharmaceutical University, 1 Shichono-cho, Misasagi, Yamashina-ku, Kyoto 607-8412, Japan
| | - Yuki Naito
- Laboratory
of Analytical and Bioinorganic Chemistry, Division of Analytical and
Physical Sciences, Kyoto Pharmaceutical
University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan
| | - Hiroyuki Yasui
- Laboratory
of Analytical and Bioinorganic Chemistry, Division of Analytical and
Physical Sciences, Kyoto Pharmaceutical
University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan
| |
Collapse
|
22
|
Ali MU, Chaudhary BN, Panja S, Gendelman HE. Theranostic Diagnostics. Results Probl Cell Differ 2024; 73:551-578. [PMID: 39242393 DOI: 10.1007/978-3-031-62036-2_22] [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: 09/09/2024]
Abstract
Diagnosing and then treating disease defines theranostics. The approach holds promise by facilitating targeted disease outcomes. The simultaneous analysis of finding the presence of disease pathophysiology while providing a parallel in treatment is a novel and effective strategy for seeking improved medical care. We discuss how theranostics improves disease outcomes is discussed. The chapter reviews the delivery of targeted therapies. Bioimaging techniques are highlighted as early detection and tracking systems for microbial infections, degenerative diseases, and cancers.
Collapse
Affiliation(s)
- Mohammad Uzair Ali
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bharat N Chaudhary
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sudipta Panja
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
23
|
Wang Y, Harada‐Shoji N, Kitamura N, Yamazaki Y, Ebata A, Amari M, Watanabe M, Miyashita M, Tada H, Abe T, Suzuki T, Gonda K, Ishida T. Mitochondrial dynamics as a novel treatment strategy for triple-negative breast cancer. Cancer Med 2024; 13:e6987. [PMID: 38334464 PMCID: PMC10854452 DOI: 10.1002/cam4.6987] [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: 11/04/2023] [Revised: 12/22/2023] [Accepted: 01/19/2024] [Indexed: 02/10/2024] Open
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC), recognized as the most heterogeneous type of breast cancer (BC), exhibits a worse prognosis than other subtypes. Mitochondria dynamics play a vital role as mediators in tumorigenesis by adjusting to the cell microenvironments. However, the relationship between mitochondrial dynamics and metabophenotype exhibits discrepancies and divergence across various research and BC models. Therefore, this study aims to explore the role of mitochondrial dynamics in TNBC drug resistance and tumorigenesis. METHODS The Wst-8 test was conducted to assess doxorubicin sensitivity in HCC38, MDA-MB-231 (TNBC), and MCF-7 (luminal). Confocal microscopy and FACS were used to quantify the mitochondrial membrane potential (ΔφM), mitophagy, and reactive oxygen species (ROS) production. Agilent Seahorse XF Analyzer was utilized to measure metabolic characteristics. Dynamin-related protein-1 (DRP1), Parkin, and p62 immunohistochemistry staining were performed using samples from 107 primary patients with BC before and after neoadjuvant chemotherapy (NAC). RESULTS MDA-MB-231, a TNBC cell line with reduced sensitivity to doxorubicin, reduced ΔφM, and enhanced mitophagy to maintain ROS production through oxidative phosphorylation (OXPHOS)-based metabolism. HCC38, a doxorubicin-sensitive cell line, exhibited no alterations in ΔφM or mitophagy. However, it demonstrated an increase in ROS production and glycolysis. Clinicopathological studies revealed that pretreatment (before NAC) expression of DRP1 was significant in TNBC, as was pretreatment expression of Parkin in the hormone receptor-negative group. Furthermore, low p62 levels seem to be a risk factor for recurrence-free survival. CONCLUSION Our findings indicated that the interplay between mitophagy, linked to a worse clinical prognosis, and OXPHOS metabolism promoted chemotherapy resistance in TNBC. Mitochondrial fission is prevalent in TNBC. These findings suggest that targeting the unique mitochondrial metabolism and dynamics in TNBC may offer a novel therapeutic strategy for patients with TNBC.
Collapse
Affiliation(s)
- Yuechen Wang
- Department of Breast and Endocrine Surgical OncologyTohoku University Graduate School of MedicineSendaiJapan
| | - Narumi Harada‐Shoji
- Department of Breast and Endocrine Surgical OncologyTohoku University Graduate School of MedicineSendaiJapan
| | - Narufumi Kitamura
- Department of Medical Physics, Graduate School of MedicineTohoku UniversitySendaiJapan
| | - Yuto Yamazaki
- Department of PathologyTohoku University Graduate School of MedicineSendaiJapan
| | - Akiko Ebata
- Department of Breast and Endocrine Surgical OncologyTohoku University Graduate School of MedicineSendaiJapan
| | - Masakazu Amari
- Department of Breast SurgeryTohoku Kosai HospitalSendaiJapan
| | - Mika Watanabe
- Department of PathologyTohoku Kosai HospitalSendaiJapan
| | - Minoru Miyashita
- Department of Breast and Endocrine Surgical OncologyTohoku University Graduate School of MedicineSendaiJapan
| | - Hiroshi Tada
- Department of Breast and Endocrine Surgical OncologyTohoku University Graduate School of MedicineSendaiJapan
| | - Takaaki Abe
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
- Department of Medical ScienceTohoku University Graduate School of Biomedical Engineering, Tohoku UniversitySendaiJapan
- Department of Clinical Biology and Hormonal RegulationTohoku University Graduate School of MedicineSendaiJapan
| | - Takashi Suzuki
- Department of PathologyTohoku University Graduate School of MedicineSendaiJapan
| | - Kohsuke Gonda
- Department of Medical Physics, Graduate School of MedicineTohoku UniversitySendaiJapan
- International Center for Synchrotron Radiation Innovation Smart (SRIS)Tohoku UniversitySendaiJapan
| | - Takanori Ishida
- Department of Breast and Endocrine Surgical OncologyTohoku University Graduate School of MedicineSendaiJapan
| |
Collapse
|
24
|
Bomhals B, Cossement L, Maes A, Sathekge M, Mokoala KMG, Sathekge C, Ghysen K, Van de Wiele C. Principal Component Analysis Applied to Radiomics Data: Added Value for Separating Benign from Malignant Solitary Pulmonary Nodules. J Clin Med 2023; 12:7731. [PMID: 38137800 PMCID: PMC10743692 DOI: 10.3390/jcm12247731] [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: 10/16/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Here, we report on the added value of principal component analysis applied to a dataset of texture features derived from 39 solitary pulmonary lung nodule (SPN) lesions for the purpose of differentiating benign from malignant lesions, as compared to the use of SUVmax alone. Texture features were derived using the LIFEx software. The eight best-performing first-, second-, and higher-order features for separating benign from malignant nodules, in addition to SUVmax (MaximumGreyLevelSUVbwIBSI184IY), were included for PCA. Two principal components (PCs) were retained, of which the contributions to the total variance were, respectively, 87.6% and 10.8%. When included in a logistic binomial regression analysis, including age and gender as covariates, both PCs proved to be significant predictors for the underlying benign or malignant character of the lesions under study (p = 0.009 for the first PC and 0.020 for the second PC). As opposed to SUVmax alone, which allowed for the accurate classification of 69% of the lesions, the regression model including both PCs allowed for the accurate classification of 77% of the lesions. PCs derived from PCA applied on selected texture features may allow for more accurate characterization of SPN when compared to SUVmax alone.
Collapse
Affiliation(s)
- Birte Bomhals
- Department of Diagnostic Sciences, University Ghent, 9000 Ghent, Belgium; (B.B.); (L.C.)
| | - Lara Cossement
- Department of Diagnostic Sciences, University Ghent, 9000 Ghent, Belgium; (B.B.); (L.C.)
| | - Alex Maes
- Department of Morphology and Functional Imaging, University Hospital Leuven, 3000 Leuven, Belgium;
- Department of Nuclear Medicine, Katholieke University Leuven, AZ Groeninge, President Kennedylaan 4, 8500 Kortrijk, Belgium
| | - Mike Sathekge
- Department of Nuclear Medicine, Steve Biko Academic Hospital and Nuclear Medicine Research Infrastructure (NuMeRi), University of Pretoria, Pretoria 0002, South Africa
| | - Kgomotso M. G. Mokoala
- Department of Nuclear Medicine, Steve Biko Academic Hospital and Nuclear Medicine Research Infrastructure (NuMeRi), University of Pretoria, Pretoria 0002, South Africa
| | - Chabi Sathekge
- Department of Nuclear Medicine, Steve Biko Academic Hospital and Nuclear Medicine Research Infrastructure (NuMeRi), University of Pretoria, Pretoria 0002, South Africa
| | - Katrien Ghysen
- Department of Pneumology, AZ Groeninge, 8500 Kortrijk, Belgium
| | - Christophe Van de Wiele
- Department of Diagnostic Sciences, University Ghent, 9000 Ghent, Belgium; (B.B.); (L.C.)
- Department of Nuclear Medicine, Katholieke University Leuven, AZ Groeninge, President Kennedylaan 4, 8500 Kortrijk, Belgium
| |
Collapse
|
25
|
Alam S, Doherty E, Ortega-Prieto P, Arizanova J, Fets L. Membrane transporters in cell physiology, cancer metabolism and drug response. Dis Model Mech 2023; 16:dmm050404. [PMID: 38037877 PMCID: PMC10695176 DOI: 10.1242/dmm.050404] [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] [Indexed: 12/02/2023] Open
Abstract
By controlling the passage of small molecules across lipid bilayers, membrane transporters influence not only the uptake and efflux of nutrients, but also the metabolic state of the cell. With more than 450 members, the Solute Carriers (SLCs) are the largest transporter super-family, clustering into families with different substrate specificities and regulatory properties. Cells of different types are, therefore, able to tailor their transporter expression signatures depending on their metabolic requirements, and the physiological importance of these proteins is illustrated by their mis-regulation in a number of disease states. In cancer, transporter expression is heterogeneous, and the SLC family has been shown to facilitate the accumulation of biomass, influence redox homeostasis, and also mediate metabolic crosstalk with other cell types within the tumour microenvironment. This Review explores the roles of membrane transporters in physiological and malignant settings, and how these roles can affect drug response, through either indirect modulation of sensitivity or the direct transport of small-molecule therapeutic compounds into cells.
Collapse
Affiliation(s)
- Sara Alam
- Drug Transport and Tumour Metabolism Lab, MRC Laboratory of Medical Sciences, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Emily Doherty
- Drug Transport and Tumour Metabolism Lab, MRC Laboratory of Medical Sciences, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Paula Ortega-Prieto
- Drug Transport and Tumour Metabolism Lab, MRC Laboratory of Medical Sciences, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Julia Arizanova
- Drug Transport and Tumour Metabolism Lab, MRC Laboratory of Medical Sciences, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Louise Fets
- Drug Transport and Tumour Metabolism Lab, MRC Laboratory of Medical Sciences, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
26
|
Rowe SP, Auwaerter PG, Sheikhbahaei S, Solnes LB, Wright WF. Molecular Imaging of Infections: Emerging Techniques for Pathogen-Specific Diagnosis and Guided Therapy. J Infect Dis 2023; 228:S241-S248. [PMID: 37788504 DOI: 10.1093/infdis/jiad092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023] Open
Abstract
Evaluation of patients that may be infected is challenging. Imaging to identify or localize a site of infection is often limited because of the nonspecific nature of the findings on conventional imaging modalities. Available imaging methods lack the ability to determine if antibiotics are reaching the site of infection and are not optimized to follow response to therapy. Positron emission tomography (PET) is a method by which radiolabeled molecules can be used to detect metabolic perturbations or levels of expression of specific targets. The most common PET agent is the glucose analog 2-deoxy-2-[18F]fluoro-D-glucose (18F-FDG). 18F-FDG has some applicability to localizing a site of infection, but its lack of specificity limits its usefulness. There is a need for the development of pathogen-specific PET radiotracers to address the imaging shortcomings noted above. Preclinical and clinical progress has been made, but significant challenges remain.
Collapse
Affiliation(s)
- Steven P Rowe
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Paul G Auwaerter
- The Sherrilyn and Ken Fisher Center for Environmental Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Division of Infectious Disease, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sara Sheikhbahaei
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lilja B Solnes
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - William F Wright
- The Sherrilyn and Ken Fisher Center for Environmental Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Division of Infectious Disease, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
27
|
Sunassee ED, Jardim-Perassi BV, Madonna MC, Ordway B, Ramanujam N. Metabolic Imaging as a Tool to Characterize Chemoresistance and Guide Therapy in Triple-Negative Breast Cancer (TNBC). Mol Cancer Res 2023; 21:995-1009. [PMID: 37343066 PMCID: PMC10592445 DOI: 10.1158/1541-7786.mcr-22-1004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 04/07/2023] [Accepted: 06/15/2023] [Indexed: 06/23/2023]
Abstract
After an initial response to chemotherapy, tumor relapse is frequent. This event is reflective of both the spatiotemporal heterogeneities of the tumor microenvironment as well as the evolutionary propensity of cancer cell populations to adapt to variable conditions. Because the cause of this adaptation could be genetic or epigenetic, studying phenotypic properties such as tumor metabolism is useful as it reflects molecular, cellular, and tissue-level dynamics. In triple-negative breast cancer (TNBC), the characteristic metabolic phenotype is a highly fermentative state. However, during treatment, the spatial and temporal dynamics of the metabolic landscape are highly unstable, with surviving populations taking on a variety of metabolic states. Thus, longitudinally imaging tumor metabolism provides a promising approach to inform therapeutic strategies, and to monitor treatment responses to understand and mitigate recurrence. Here we summarize some examples of the metabolic plasticity reported in TNBC following chemotherapy and review the current metabolic imaging techniques available in monitoring chemotherapy responses clinically and preclinically. The ensemble of imaging technologies we describe has distinct attributes that make them uniquely suited for a particular length scale, biological model, and/or features that can be captured. We focus on TNBC to highlight the potential of each of these technological advances in understanding evolution-based therapeutic resistance.
Collapse
Affiliation(s)
- Enakshi D. Sunassee
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | | | - Megan C. Madonna
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Bryce Ordway
- Department of Cancer Physiology, Moffitt Cancer Center, Tampa, FL 33612, USA
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Nirmala Ramanujam
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27708, USA
| |
Collapse
|
28
|
Maxouri O, Bodalal Z, Daal M, Rostami S, Rodriguez I, Akkari L, Srinivas M, Bernards R, Beets-Tan R. How to 19F MRI: applications, technique, and getting started. BJR Open 2023; 5:20230019. [PMID: 37953866 PMCID: PMC10636348 DOI: 10.1259/bjro.20230019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 07/26/2023] [Accepted: 08/01/2023] [Indexed: 11/14/2023] Open
Abstract
Magnetic resonance imaging (MRI) plays a significant role in the routine imaging workflow, providing both anatomical and functional information. 19F MRI is an evolving imaging modality where instead of 1H, 19F nuclei are excited. As the signal from endogenous 19F in the body is negligible, exogenous 19F signals obtained by 19F radiofrequency coils are exceptionally specific. Highly fluorinated agents targeting particular biological processes (i.e., the presence of immune cells) have been visualised using 19F MRI, highlighting its potential for non-invasive and longitudinal molecular imaging. This article aims to provide both a broad overview of the various applications of 19F MRI, with cancer imaging as a focus, as well as a practical guide to 19F imaging. We will discuss the essential elements of a 19F system and address common pitfalls during acquisition. Last but not least, we will highlight future perspectives that will enhance the role of this modality. While not an exhaustive exploration of all 19F literature, we endeavour to encapsulate the broad themes of the field and introduce the world of 19F molecular imaging to newcomers. 19F MRI bridges several domains, imaging, physics, chemistry, and biology, necessitating multidisciplinary teams to be able to harness this technology effectively. As further technical developments allow for greater sensitivity, we envision that 19F MRI can help unlock insight into biological processes non-invasively and longitudinally.
Collapse
Affiliation(s)
| | | | | | | | | | - Leila Akkari
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - René Bernards
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | |
Collapse
|
29
|
Bulut G, Atilgan HI, Çınarer G, Kılıç K, Yıkar D, Parlar T. Prediction of pathological complete response to neoadjuvant chemotherapy in locally advanced breast cancer by using a deep learning model with 18F-FDG PET/CT. PLoS One 2023; 18:e0290543. [PMID: 37708209 PMCID: PMC10501592 DOI: 10.1371/journal.pone.0290543] [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: 12/07/2022] [Accepted: 08/10/2023] [Indexed: 09/16/2023] Open
Abstract
OBJECTIVES The aim of the study is 18F-FDG PET/CT imaging by using deep learning method are predictive for pathological complete response pCR after Neoadjuvant chemotherapy (NAC) in locally advanced breast cancer (LABC). INTRODUCTION NAC is the standard treatment for locally advanced breast cancer (LABC). Pathological complete response (pCR) after NAC is considered a good predictor of disease-free survival (DFS) and overall survival (OS).Therefore, there is a need to develop methods that can predict the pCR at the time of diagnosis. METHODS This article was designed as a retrospective chart study.For the convolutional neural network model, a total of 355 PET/CT images of 31 patients were used. All patients had primary breast surgery after completing NAC. RESULTS Pathological complete response was obtained in a total of 9 patients. The study results show that our proposed deep convolutional neural networks model achieved a remarkable success with an accuracy of 84.79% to predict pathological complete response. CONCLUSION It was concluded that deep learning methods can predict breast cancer treatment.
Collapse
Affiliation(s)
- Gülcan Bulut
- Division of Medical Oncology, International Medicana Hospital, Izmir, Turkey
| | - Hasan Ikbal Atilgan
- Department of Nuclear Medicine, Mustafa Kemal University Medical School, Hatay, Turkey
| | - Gökalp Çınarer
- Department of Computer Engineering, Faculty of Engineering and Architecture, Bozok University, Yozgat, Turkey
| | - Kazım Kılıç
- Department of Computer Programming, Yozgat Vocational High School, Bozok University, Yozgat, Turkey
| | - Deniz Yıkar
- Division of Nuclear Medicine, Hatay Training and Research Hospital, Hatay, Turkey
| | - Tuba Parlar
- Department of Computer Technologies, Mustafa Kemal University, Hatay, Türkiye
| |
Collapse
|
30
|
Metelerkamp Cappenberg T, De Schepper S, Vangestel C, De Lombaerde S, Wyffels L, Van den Wyngaert T, Mattis J, Gray B, Pak K, Stroobants S, Elvas F. First-in-human study of a novel cell death tracer [ 99mTc]Tc-Duramycin: safety, biodistribution and radiation dosimetry in healthy volunteers. EJNMMI Radiopharm Chem 2023; 8:20. [PMID: 37646865 PMCID: PMC10468453 DOI: 10.1186/s41181-023-00207-1] [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: 07/24/2023] [Accepted: 08/23/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Imaging of cell death can provide an early indication of treatment response in cancer. [99mTc]Tc-Duramycin is a small-peptide SPECT tracer that recognizes both apoptotic and necrotic cells by binding to phosphatidylethanolamine present in the cell membrane. Preclinically, this tracer has shown to have favorable pharmacokinetics and selective tumor accumulation early after the onset of anticancer therapy. In this first-in-human study, we report the safety, biodistribution and internal radiation dosimetry of [99mTc]Tc-Duramycin in healthy human volunteers. RESULTS Six healthy volunteers (3 males, 3 females) were injected intravenously with [99mTc]Tc-Duramycin (dose: 6 MBq/kg; 473 ± 36 MBq). [99mTc]Tc-Duramycin was well tolerated in all subjects, with no serious adverse events reported. Following injection, a 30-min dynamic planar imaging of the abdomen was performed, and whole-body (WB) planar scans were acquired at 1, 2, 3, 6 and 23 h post-injection (PI), with SPECT acquisitions after each WB scan and one low-dose CT after the first SPECT. In vivo 99mTc activities were determined from semi-quantitative analysis of the images, and time-activity curves were generated. Residence times were calculated from the dynamic and WB planar scans. The mean effective dose was 7.61 ± 0.75 µSv/MBq, with the kidneys receiving the highest absorbed dose (planar analysis: 43.82 ± 4.07 µGy/MBq, SPECT analysis: 19.72 ± 3.42 μGy/MBq), followed by liver and spleen. The median effective dose was 3.61 mSv (range, 2.85-4.14). The tracer cleared slowly from the blood (effective half-life of 2.0 ± 0.4 h) due to high plasma protein binding with < 5% free tracer 3 h PI. Excretion was almost exclusively renal. CONCLUSION [99mTc]Tc-Duramycin demonstrated acceptable dosimetry (< 5 mSv) and a favorable safety profile. Due to slow blood clearance, optimal target-to-background ratios are expected 5 h PI. These data support the further assessment of [99mTc]Tc-Duramycin for clinical treatment response evaluation. TRIAL REGISTRATION NCT05177640, Registered April 30, 2021, https://clinicaltrials.gov/study/NCT05177640 .
Collapse
Affiliation(s)
| | - Stijn De Schepper
- Department of Nuclear Medicine, Antwerp University Hospital (UZA), Edegem, Belgium
| | - Christel Vangestel
- Department of Nuclear Medicine, Antwerp University Hospital (UZA), Edegem, Belgium
- Molecular Imaging and Radiology (MIRA), University of Antwerp, Wilrijk, Belgium
| | - Stef De Lombaerde
- Department of Nuclear Medicine, Antwerp University Hospital (UZA), Edegem, Belgium
- Molecular Imaging and Radiology (MIRA), University of Antwerp, Wilrijk, Belgium
| | - Leonie Wyffels
- Department of Nuclear Medicine, Antwerp University Hospital (UZA), Edegem, Belgium
- Molecular Imaging and Radiology (MIRA), University of Antwerp, Wilrijk, Belgium
| | - Tim Van den Wyngaert
- Department of Nuclear Medicine, Antwerp University Hospital (UZA), Edegem, Belgium
- Molecular Imaging and Radiology (MIRA), University of Antwerp, Wilrijk, Belgium
| | - Jeffrey Mattis
- Molecular Targeting Technologies, Inc., West Chester, PA, USA
| | - Brian Gray
- Molecular Targeting Technologies, Inc., West Chester, PA, USA
| | - Koon Pak
- Molecular Targeting Technologies, Inc., West Chester, PA, USA
| | - Sigrid Stroobants
- Department of Nuclear Medicine, Antwerp University Hospital (UZA), Edegem, Belgium
- Molecular Imaging and Radiology (MIRA), University of Antwerp, Wilrijk, Belgium
| | - Filipe Elvas
- Department of Nuclear Medicine, Antwerp University Hospital (UZA), Edegem, Belgium.
- Molecular Imaging and Radiology (MIRA), University of Antwerp, Wilrijk, Belgium.
| |
Collapse
|
31
|
Kolokotroni A, Gkikopoulou E, Rinotas V, Ntari L, Zareifi D, Rouchota M, Sarpaki S, Lymperopoulos I, Alexopoulos LG, Loudos G, Denis MC, Karagianni N, Douni E. Α Humanized RANKL Transgenic Mouse Model of Progestin-Induced Mammary Carcinogenesis for Evaluation of Novel Therapeutics. Cancers (Basel) 2023; 15:4006. [PMID: 37568820 PMCID: PMC10417415 DOI: 10.3390/cancers15154006] [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: 07/16/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Receptor activator of nuclear factor-κB ligand (RANKL) is critically involved in mammary gland pathophysiology, while its pharmaceutical inhibition is being currently investigated in breast cancer. Herein, we investigated whether the overexpression of human RANKL in transgenic mice affects hormone-induced mammary carcinogenesis, and evaluated the efficacy of anti-RANKL treatments, such as OPG-Fc targeting both human and mouse RANKL or Denosumab against human RANKL. We established novel MPA/DMBA-driven mammary carcinogenesis models in TgRANKL mice that express both human and mouse RANKL, as well as in humanized humTgRANKL mice expressing only human RANKL, and compared them to MPA/DMBA-treated wild-type (WT) mice. Our results show that TgRANKL and WT mice have similar levels of susceptibility to mammary carcinogenesis, while OPG-Fc treatment restored mammary ductal density, and prevented ductal branching and the formation of neoplastic foci in both genotypes. humTgRANKL mice also developed MPA/DMBA-induced tumors with similar incidence and burden to those of WT and TgRANKL mice. The prophylactic treatment of humTgRANKL mice with Denosumab significantly prevented the rate of appearance of mammary tumors from 86.7% to 15.4% and the early stages of carcinogenesis, whereas therapeutic treatment did not lead to any significant attenuation of tumor incidence or tumor burden compared to control mice, suggesting the importance of RANKL primarily in the initial stages of tumorigenesis. Overall, we provide unique genetic tools for investigating the involvement of RANKL in breast carcinogenesis, and allow the preclinical evaluation of novel therapeutics that target hormone-related breast cancers.
Collapse
Affiliation(s)
- Anthi Kolokotroni
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece
- Institute for Bioinnovation, Biomedical Sciences Research Center “Alexander Fleming”, Fleming 34, 16672 Vari, Greece
| | - Evi Gkikopoulou
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece
- Institute for Bioinnovation, Biomedical Sciences Research Center “Alexander Fleming”, Fleming 34, 16672 Vari, Greece
| | - Vagelis Rinotas
- Institute for Bioinnovation, Biomedical Sciences Research Center “Alexander Fleming”, Fleming 34, 16672 Vari, Greece
| | - Lydia Ntari
- Biomedcode Hellas SA, Fleming 34, 16672 Vari, Greece (M.C.D.)
| | - Danae Zareifi
- Department of Mechanical Engineering, National Technical University of Athens, 10682 Athens, Greece
| | - Maritina Rouchota
- BIOEMTECH, Lefkippos Attica Technology Park, NCSR “Demokritos”, Ag. Paraskevi, 15343 Athens, Greece (G.L.)
| | - Sophia Sarpaki
- BIOEMTECH, Lefkippos Attica Technology Park, NCSR “Demokritos”, Ag. Paraskevi, 15343 Athens, Greece (G.L.)
| | | | - Leonidas G. Alexopoulos
- Department of Mechanical Engineering, National Technical University of Athens, 10682 Athens, Greece
| | - George Loudos
- BIOEMTECH, Lefkippos Attica Technology Park, NCSR “Demokritos”, Ag. Paraskevi, 15343 Athens, Greece (G.L.)
| | - Maria C. Denis
- Biomedcode Hellas SA, Fleming 34, 16672 Vari, Greece (M.C.D.)
| | - Niki Karagianni
- Biomedcode Hellas SA, Fleming 34, 16672 Vari, Greece (M.C.D.)
| | - Eleni Douni
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece
- Institute for Bioinnovation, Biomedical Sciences Research Center “Alexander Fleming”, Fleming 34, 16672 Vari, Greece
| |
Collapse
|
32
|
Campbell E, Jordan C, Gilmour R. Fluorinated carbohydrates for 18F-positron emission tomography (PET). Chem Soc Rev 2023; 52:3599-3626. [PMID: 37171037 PMCID: PMC10243284 DOI: 10.1039/d3cs00037k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Indexed: 05/13/2023]
Abstract
Carbohydrate diversity is foundational in the molecular literacy that regulates cellular function and communication. Consequently, delineating and leveraging this structure-function interplay continues to be a core research objective in the development of candidates for biomedical diagnostics. A totemic example is the ubiquity of 2-deoxy-2-[18F]-fluoro-D-glucose (2-[18F]-FDG) as a radiotracer for positron emission tomography (PET), in which metabolic trapping is harnessed. Building on this clinical success, more complex sugars with unique selectivities are gaining momentum in molecular recognition and personalised medicine: this reflects the opportunities that carbohydrate-specific targeting affords in a broader sense. In this Tutorial Review, key milestones in the development of 2-[18F]-FDG and related glycan-based radiotracers for PET are described, with their diagnostic functions, to assist in navigating this rapidly expanding field of interdisciplinary research.
Collapse
Affiliation(s)
- Emma Campbell
- Organisch-Chemisches Institut, Westfälische Wilhelms-Universität Münster Corrensstraße 36, 48149, Münster, Germany.
- Cells in Motion Interfaculty Centre, Westfälische Wilhelms-Universität Münster, Röntgenstraße 16, 48149, Münster, Germany
| | - Christina Jordan
- Organisch-Chemisches Institut, Westfälische Wilhelms-Universität Münster Corrensstraße 36, 48149, Münster, Germany.
- Cells in Motion Interfaculty Centre, Westfälische Wilhelms-Universität Münster, Röntgenstraße 16, 48149, Münster, Germany
| | - Ryan Gilmour
- Organisch-Chemisches Institut, Westfälische Wilhelms-Universität Münster Corrensstraße 36, 48149, Münster, Germany.
- Cells in Motion Interfaculty Centre, Westfälische Wilhelms-Universität Münster, Röntgenstraße 16, 48149, Münster, Germany
| |
Collapse
|
33
|
Shankar LK, Schöder H, Sharon E, Wolchok J, Knopp MV, Wahl RL, Ellingson BM, Hall NC, Yaffe MJ, Towbin AJ, Farwell MD, Pryma D, Poussaint TY, Wright CL, Schwartz L, Harisinghani M, Mahmood U, Wu AM, Leung D, de Vries EGE, Tang Y, Beach G, Reeves SA. Harnessing imaging tools to guide immunotherapy trials: summary from the National Cancer Institute Cancer Imaging Steering Committee workshop. Lancet Oncol 2023; 24:e133-e143. [PMID: 36858729 PMCID: PMC10119769 DOI: 10.1016/s1470-2045(22)00742-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/18/2022] [Accepted: 11/30/2022] [Indexed: 03/02/2023]
Abstract
As the immuno-oncology field continues the rapid growth witnessed over the past decade, optimising patient outcomes requires an evolution in the current response-assessment guidelines for phase 2 and 3 immunotherapy clinical trials and clinical care. Additionally, investigational tools-including image analysis of standard-of-care scans (such as CT, magnetic resonance, and PET) with analytics, such as radiomics, functional magnetic resonance agents, and novel molecular-imaging PET agents-offer promising advancements for assessment of immunotherapy. To document current challenges and opportunities and identify next steps in immunotherapy diagnostic imaging, the National Cancer Institute Clinical Imaging Steering Committee convened a meeting with diverse representation among imaging experts and oncologists to generate a comprehensive review of the state of the field.
Collapse
Affiliation(s)
- Lalitha K Shankar
- Clinical Trials Branch, National Cancer Institute, Rockville, MD, USA.
| | - Heiko Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elad Sharon
- Investigational Drug Branch, National Cancer Institute, Rockville, MD, USA
| | - Jedd Wolchok
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Michael V Knopp
- Department of Radiology, Ohio State University, Columbus, OH, USA
| | - Richard L Wahl
- Department of Radiology, Washington University, St Louis, MO, USA
| | - Benjamin M Ellingson
- Department of Radiological Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Nathan C Hall
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Martin J Yaffe
- Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Alexander J Towbin
- Department of Radiology and Medical Imaging, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Michael D Farwell
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Pryma
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | | - Umar Mahmood
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Anna M Wu
- Department of Immunology & Theranostics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | | | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | | | | | - Steven A Reeves
- Coordinating Center for Clinical Trials, National Cancer Institute, Rockville, MD, USA
| |
Collapse
|
34
|
Singh G, Thakur N, Kumar U. RAS: Circuitry and therapeutic targeting. Cell Signal 2023; 101:110505. [PMID: 36341985 DOI: 10.1016/j.cellsig.2022.110505] [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: 08/05/2022] [Revised: 10/05/2022] [Accepted: 10/21/2022] [Indexed: 11/26/2022]
Abstract
Cancer has affected the lives of millions worldwide and is truly regarded as a devastating disease process. Despite advanced understanding of the genomic underpinning of cancer development and progression, therapeutic challenges are still persistent. Among all the human cancers, around 33% are attributed to mutations in RAS oncogene, a crucial component of the signaling pathways. With time, our understanding of RAS circuitry has improved and now the fact that it activates several downstream effectors, depending on the type and grades of cancer has been established. The circuitry is controlled via post-transcriptional mechanisms and frequent distortions in these mechanisms lead to important metabolic as well as immunological states that favor cancer cells' growth, survival, plasticity and metastasis. Therefore, understanding RAS circuitry can help researchers/clinicians to develop novel and potent therapeutics that, in turn, can save the lives of patients suffering from RAS-mutant cancers. There are many challenges presented by resistance and the potential strategies with a particular focus on novel combinations for overcoming these, that could move beyond transitory responses in the direction of treatment. Here in this review, we will look at how understanding the circuitry of RAS can be put to use in making strategies for developing therapeutics against RAS- driven malignancies.
Collapse
Affiliation(s)
- Gagandeep Singh
- Department of Biosciences (UIBT), Chandigarh University, NH-05, Ludhiana - Chandigarh State Hwy, Sahibzada Ajit Singh Nagar, Punjab 140413, India
| | - Neelam Thakur
- Department of Biosciences (UIBT), Chandigarh University, NH-05, Ludhiana - Chandigarh State Hwy, Sahibzada Ajit Singh Nagar, Punjab 140413, India; Department of Zoology, Sardar Patel University, Vallabh Government College Campus, Paddal, Kartarpur, Mandi, Himachal Pradesh 175001, India.
| | - Umesh Kumar
- School of Biosciences, Institute of Management Studies Ghaziabad (University Courses Campus), Adhyatmik Nagar, NH09, Ghaziabad, Uttar Pradesh 201015, India.
| |
Collapse
|
35
|
Wang D, Li X, Gong G, Lu Y, Guo Z, Chen R, Huang H, Li Z, Bian J. An updated patent review of glutaminase inhibitors (2019-2022). Expert Opin Ther Pat 2023; 33:17-28. [PMID: 36698323 DOI: 10.1080/13543776.2023.2173573] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Kidney-type glutaminase (GLS1), a key enzyme controlling the hydrolysis of glutamine to glutamate to resolve the 'glutamine addiction' of cancer cells, has been shown to play a central role in supporting cancer growth and proliferation. Therefore, the inhibition of GLS1 as a novel cancer treating strategy is of great interest. AREAS COVERED This review covers recent patents (2019-present) involving GLS1 inhibitors, which are mostly focused on their chemical structures, molecular mechanisms of action, pharmacokinetic properties, and potential clinical applications. EXPERT OPINION Currently, despite significant efforts, the search for potent GLS1 inhibitors has not resulted in the development of compounds for therapeutic applications. Most recent patents and literature focus on GLS1 inhibitors IPN60090 and DRP104, which have entered clinical trials. While other patent disclosures during this period have not generated any drug candidates, the clinical update will inform the potential of these inhibitors as promising therapeutic agents either as single or as combination interventions.
Collapse
Affiliation(s)
- Danni Wang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaohong Li
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Guangyue Gong
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yulong Lu
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ziming Guo
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Rui Chen
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Huidan Huang
- Department of Pharmaceutical Engineering, School of Pharmacy, Wannan Medical College, Wuhu, China
| | - Zhiyu Li
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jinlei Bian
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
36
|
Yang Y, Ren P, Liu X, Sun X, Zhang C, Du X, Xing B. PPP1R26 drives hepatocellular carcinoma progression by controlling glycolysis and epithelial-mesenchymal transition. J Exp Clin Cancer Res 2022; 41:101. [PMID: 35292107 PMCID: PMC8922775 DOI: 10.1186/s13046-022-02302-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/25/2022] [Indexed: 01/17/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is usually diagnosed at an advanced stage due to rapid progression. Glycolysis supports anabolic growth and metastasis to promote HCC progression. However, the molecular mechanisms linking glycolysis and metastasis in HCC are not completely defined. Methods The expression of PPP1R26 in human HCC tissues was evaluated by immunohistochemistry, and the clinical significance of PPP1R26 in the progression and prognosis of the HCC patients were analyzed. The PPP1R26-binding proteins were determined by mass spectrometry analysis. The function of PPP1R26 in glycolysis, EMT and tumorigenesis were evaluated in HCC cells. Glucose uptake and tumor growth were evaluated using PET imaging in mouse xenografts in vivo. Protein binding was confirmed by co-immunoprecipitation and immunofluorescence co-localization. Protein-RNA binding was determined by RNA-immunoprecipitation (RIP) experiment. The binding of protein on the promoter was evaluated by chromatin immunoprecipitation assay (ChIP). Results PPP1R26 is upregulated in human HCC tissues and its upregulation is significantly associated with metastasis and the poor survival of the patients. PPP1R26 activates glycolysis in HCC cells and in mouse xenografts in vivo. PPP1R26 drives glycolysis by binding to PTBP1 to facilitate the mRNA splicing of PKM2. Simultaneously, overexpressed PPP1R26 induces the nuclear accumulation of PKM2 to inhibit the expression of E-cadherin further to drive EMT. Mechanistically, PPP1R26 binds with Ser37-phosphorylated PKM2 and TGIF2 in the nucleus and blocks the binding of TGIF2 with CDH1 promoter to inhibit the transcription of CDH1. Conclusion PPP1R26 promotes glycolysis by enhancing PKM2 splicing and simultaneously activates EMT by forming a PPP1R26-PKM2-TGIF2 complex to drive HCC progression. Therefore, targeting PPP1R26 attenuates HCC progression and provides a potential therapeutic strategy for the HCC patients with upregulation of PPP1R26. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02302-8.
Collapse
|
37
|
Capozzi A. Design and performance of a small bath cryostat with NMR capability for transport of hyperpolarized samples. Sci Rep 2022; 12:19260. [PMID: 36357496 PMCID: PMC9649762 DOI: 10.1038/s41598-022-23890-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 11/07/2022] [Indexed: 11/12/2022] Open
Abstract
As of today, dissolution Dynamic Nuclear Polarization (dDNP) is the only clinically available hyperpolarization technique for 13C-MRI. Despite the clear path towards personalized medicine that dDNP is paving as an alternative and/or complement to Positron Emission Tomography (PET), the technique struggles to enter everyday clinical practice. Because of the minute-long hyperpolarization lifetime after dissolution, one of the reasons lies in the need and consequent complexities of having the machine that generates the hyperpolarization (i.e. the dDNP polarizer) on site. Since some years, research groups are working to make hyperpolarization transportable. Two different methods have been developed that allow "freezing" of the nuclear spin state prior to samples extraction from the polarizer. Nevertheless, so far, all attempts of transport have been limited to a very small scale and to the level of proof-of-principle experiments. The main reason for that is the lack of adequate hardware, strategy, and control on most of the crucial parameters. To bridge the technical gap with PET and provide MRI facilities with hours long relaxing hyperpolarized compounds at controlled conditions, a new generation of low cost/small footprint liquid He cryostats equipped with a magnetically enforced cryogenic probe is needed. In this paper, we detail the theoretical and practical construction of a hyperpolarized samples transportation device small enough to fit in a car and able to hold a sample at 4.2 K for almost 8 h despite the presence of a cryogenically-demanding purpose-built probe that provides enough magnetic field upon insertion of the sample and NMR quality homogeneity at storage position. Should transportable hyperpolarization via DNP become a reality, we herein provide important details to make it possible.
Collapse
Affiliation(s)
- Andrea Capozzi
- SB IPHYS LIFMET, Institute of Physics, EPFL, CH F0 632, Bâtiment CH, Station 6, CH-1015, Lausanne, Switzerland.
- Department of Health Technology, HYPERMAG, Technical University of Denmark, Building 349, 2800, Kgs Lyngby, Denmark.
| |
Collapse
|
38
|
Jiao JW, Zhan XH, Wang JJ, He LX, Guo ZC, Xu XE, Liao LD, Huang X, Wen B, Xu YW, Hu H, Neufeld G, Chang ZJ, Zhang K, Xu LY, Li EM. LOXL2-dependent deacetylation of aldolase A induces metabolic reprogramming and tumor progression. Redox Biol 2022; 57:102496. [PMID: 36209516 PMCID: PMC9547286 DOI: 10.1016/j.redox.2022.102496] [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: 09/07/2022] [Revised: 09/29/2022] [Accepted: 10/02/2022] [Indexed: 02/05/2023] Open
Abstract
Lysyl-oxidase like-2 (LOXL2) regulates extracellular matrix remodeling and promotes tumor invasion and metastasis. Altered metabolism is a core hallmark of cancer, however, it remains unclear whether and how LOXL2 contributes to tumor metabolism. Here, we found that LOXL2 and its catalytically inactive L2Δ13 splice variant boost glucose metabolism of esophageal tumor cells, facilitate tumor cell proliferation and promote tumor development in vivo. Consistently, integrated transcriptomic and metabolomic analysis of a knock-in mouse model expressing L2Δ13 gene revealed that LOXL2/L2Δ13 overexpression perturbs glucose and lipid metabolism. Mechanistically, we identified aldolase A, glyceraldehyde-3-phosphate dehydrogenase and enolase as glycolytic proteins that interact physically with LOXL2 and L2Δ13. In the case of aldolase A, LOXL2/L2Δ13 stimulated its mobilization from the actin cytoskeleton to enhance aldolase activity during malignant transformation. Using stable isotope labeling of amino acids in cell culture (SILAC) followed by proteomic analysis, we identified LOXL2 and L2Δ13 as novel deacetylases that trigger metabolic reprogramming. Both LOXL2 and L2Δ13 directly catalyzed the deacetylation of aldolase A at K13, resulting in enhanced glycolysis which subsequently reprogramed tumor metabolism and promoted tumor progression. High level expression of LOXL2/L2Δ13 combined with decreased acetylation of aldolase-K13 predicted poor clinical outcome in patients with esophageal cancer. In summary, we have characterized a novel molecular mechanism that mediates the pro-tumorigenic activity of LOXL2 independently of its classical amine oxidase activity. These findings may enable the future development of therapeutic agents targeting the metabolic machinery via LOXL2 or L2Δ13. HIGHLIGHT OF THE STUDY: LOXL2 and its catalytically inactive isoform L2Δ13 function as new deacetylases to promote metabolic reprogramming and tumor progression in esophageal cancer by directly activating glycolytic enzymes such as aldolase A.
Collapse
Affiliation(s)
- Ji-Wei Jiao
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, China
| | - Xiu-Hui Zhan
- Department of Orthopedics, Research Center of Translational Medicine, The Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Juan-Juan Wang
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, China
| | - Li-Xia He
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, China
| | - Zhen-Chang Guo
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, China; Department of Biochemistry and Molecular Biology, Tianjin Medical University, Tianjin, 300070, China
| | - Xiu-E Xu
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, China
| | - Lian-Di Liao
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, China
| | - Xin Huang
- Department of Clinical Laboratory, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Bing Wen
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, China
| | - Yi-Wei Xu
- Department of Clinical Laboratory Medicine, Cancer Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Hai Hu
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Gera Neufeld
- Technion Integrated Cancer Center, The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, 31096, Israel
| | - Zhi-Jie Chang
- State Key Laboratory of Membrane Biology, School of Medicine, National Engineering Laboratory for Anti-tumor Therapeutics, Tsinghua University, Beijing, 10084, China
| | - Kai Zhang
- Department of Biochemistry and Molecular Biology, Tianjin Medical University, Tianjin, 300070, China.
| | - Li-Yan Xu
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, China.
| | - En-Min Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, China; Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, China.
| |
Collapse
|
39
|
Liu S, Liu Y, Zhang Z, Wang X, Yang Y, Sun K, Yu J, Chiu DT, Wu C. Near-Infrared Optical Transducer for Dynamic Imaging of Cerebrospinal Fluid Glucose in Brain Tumor. Anal Chem 2022; 94:14265-14272. [PMID: 36206033 DOI: 10.1021/acs.analchem.2c02600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Aberrant cerebral glucose metabolism is related to many brain diseases, especially brain tumor. However, it remains challenging to measure the dynamic changes in cerebral glucose. Here, we developed a near-infrared (NIR) optical transducer to sensitively monitor the glucose variations in cerebrospinal fluid in vivo. The transducer consists of an oxygen-sensitive nanoparticle combined with glucose oxidase (GOx), yielding highly sensitive NIR phosphorescence in response to blood glucose change. We demonstrated long-term continuous glucose monitoring by using the NIR transducer. After subcutaneous implantation, the glucose transducer provides a strong luminescence signal that can continuously monitor blood glucose fluctuations for weeks. By using the NIR emission of the transducer, we further observed abnormal dynamic changes in cerebrospinal fluid glucose and quantitatively assessed cerebral glucose uptake rates in transgenic mice bearing brain tumors. This study provides a promising method for the diagnosis of various metabolic diseases with altered glucose metabolism.
Collapse
Affiliation(s)
- Siyang Liu
- Harbin Institute of Technology, Harbin 150001, China.,Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Ye Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zhe Zhang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xiaodong Wang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yicheng Yang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Kai Sun
- Department of Chemistry and Bioengineering, University of Washington, 4000 15th NE, Seattle, Washington 98195, United States
| | - Jiangbo Yu
- Department of Chemistry and Bioengineering, University of Washington, 4000 15th NE, Seattle, Washington 98195, United States
| | - Daniel T Chiu
- Department of Chemistry and Bioengineering, University of Washington, 4000 15th NE, Seattle, Washington 98195, United States
| | - Changfeng Wu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
40
|
Jiang Y, Zeng Q, Jiang Q, Peng X, Gao J, Wan H, Wang L, Gao Y, Zhou X, Lin D, Feng H, Liang S, Zhou H, Ding J, Ai J, Huang R. 18F-FDG PET as an imaging biomarker for the response to FGFR-targeted therapy of cancer cells via FGFR-initiated mTOR/HK2 axis. Am J Cancer Res 2022; 12:6395-6408. [PMID: 36168616 PMCID: PMC9475468 DOI: 10.7150/thno.74848] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 08/16/2022] [Indexed: 11/05/2022] Open
Abstract
Rationale: The overall clinical response to FGFR inhibitor (FGFRi) is far from satisfactory in cancer patients stratified by FGFR aberration, the current biomarker in clinical practice. A novel biomarker to evaluate the therapeutic response to FGFRi in a non-invasive and dynamic manner is thus greatly desired. Methods: Six FGFR-aberrant cancer cell lines were used, including four FGFRi-sensitive ones (NCI-H1581, NCI-H716, RT112 and Hep3B) and two FGFRi-resistant ones (primary for NCI-H2444 and acquired for NCI-H1581/AR). Cell viability and tumor xenograft growth analyses were performed to evaluate FGFRi sensitivities, accompanied by corresponding 18F-fluorodeoxyglucose (18F-FDG) uptake assay. mTOR/PLCγ/MEK-ERK signaling blockade by specific inhibitors or siRNAs was applied to determine the regulation mechanism. Results: FGFR inhibition decreased the in vitro accumulation of 18F-FDG only in four FGFRi-sensitive cell lines, but in neither of FGFRi-resistant ones. We then demonstrated that FGFRi-induced transcriptional downregulation of hexokinase 2 (HK2), a key factor of glucose metabolism and FDG trapping, via mTOR pathway leading to this decrease. Moreover, 18F-FDG PET imaging successfully differentiated the FGFRi-sensitive tumor xenografts from primary or acquired resistant ones by the tumor 18F-FDG accumulation change upon FGFRi treatment. Of note, both 18F-FDG tumor accumulation and HK2 expression could respond the administration/withdrawal of FGFRi in NCI-H1581 xenografts correspondingly. Conclusion: The novel association between the molecular mechanism (FGFR/mTOR/HK2 axis) and radiological phenotype (18F-FDG PET uptake) of FGFR-targeted therapy was demonstrated in multiple preclinical models. The adoption of 18F-FDG PET biomarker-based imaging strategy to assess response/resistance to FGFR inhibition may benefit treatment selection for cancer patients.
Collapse
Affiliation(s)
- Yuchen Jiang
- School of Pharmacy, Nanchang University, Nanchang 330006, China.,Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Qinghe Zeng
- Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Qinghui Jiang
- Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xia Peng
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jing Gao
- Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Haiyan Wan
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Luting Wang
- Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yinglei Gao
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaoyu Zhou
- Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Dongze Lin
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hanyi Feng
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Sheng Liang
- Department of Nuclear Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Hu Zhou
- University of Chinese Academy of Sciences, Beijing 100049, China.,Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jian Ding
- School of Pharmacy, Nanchang University, Nanchang 330006, China.,Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Ai
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ruimin Huang
- Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
41
|
Positron Emission Tomography Probes for Imaging Cytotoxic Immune Cells. Pharmaceutics 2022; 14:pharmaceutics14102040. [PMID: 36297474 PMCID: PMC9610635 DOI: 10.3390/pharmaceutics14102040] [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: 08/20/2022] [Revised: 09/18/2022] [Accepted: 09/19/2022] [Indexed: 11/17/2022] Open
Abstract
Non-invasive positron emission tomography (PET) imaging of immune cells is a powerful approach for monitoring the dynamics of immune cells in response to immunotherapy. Despite the clinical success of many immunotherapeutic agents, their clinical efficacy is limited to a subgroup of patients. Conventional imaging, as well as analysis of tissue biopsies and blood samples do not reflect the complex interaction between tumour and immune cells. Consequently, PET probes are being developed to capture the dynamics of such interactions, which may improve patient stratification and treatment evaluation. The clinical efficacy of cancer immunotherapy relies on both the infiltration and function of cytotoxic immune cells at the tumour site. Thus, various immune biomarkers have been investigated as potential targets for PET imaging of immune response. Herein, we provide an overview of the most recent developments in PET imaging of immune response, including the radiosynthesis approaches employed in their development.
Collapse
|
42
|
Jemaa S, Paulson JN, Hutchings M, Kostakoglu L, Trotman J, Tracy S, de Crespigny A, Carano RAD, El-Galaly TC, Nielsen TG, Bengtsson T. Full automation of total metabolic tumor volume from FDG-PET/CT in DLBCL for baseline risk assessments. Cancer Imaging 2022; 22:39. [PMID: 35962459 PMCID: PMC9373298 DOI: 10.1186/s40644-022-00476-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 07/17/2022] [Indexed: 11/21/2022] Open
Abstract
Background Current radiological assessments of 18fluorodeoxyglucose-positron emission tomography (FDG-PET) imaging data in diffuse large B-cell lymphoma (DLBCL) can be time consuming, do not yield real-time information regarding disease burden and organ involvement, and hinder the use of FDG-PET to potentially limit the reliance on invasive procedures (e.g. bone marrow biopsy) for risk assessment. Methods Our aim is to enable real-time assessment of imaging-based risk factors at a large scale and we propose a fully automatic artificial intelligence (AI)-based tool to rapidly extract FDG-PET imaging metrics in DLBCL. On availability of a scan, in combination with clinical data, our approach generates clinically informative risk scores with minimal resource requirements. Overall, 1268 patients with previously untreated DLBCL from the phase III GOYA trial (NCT01287741) were included in the analysis (training: n = 846; hold-out: n = 422). Results Our AI-based model comprising imaging and clinical variables yielded a tangible prognostic improvement compared to clinical models without imaging metrics. We observed a risk increase for progression-free survival (PFS) with hazard ratios [HR] of 1.87 (95% CI: 1.31–2.67) vs 1.38 (95% CI: 0.98–1.96) (C-index: 0.59 vs 0.55), and a risk increase for overall survival (OS) (HR: 2.16 (95% CI: 1.37–3.40) vs 1.40 (95% CI: 0.90–2.17); C-index: 0.59 vs 0.55). The combined model defined a high-risk population with 35% and 42% increased odds of a 4-year PFS and OS event, respectively, versus the International Prognostic Index components alone. The method also identified a subpopulation with a 2-year Central Nervous System (CNS)-relapse probability of 17.1%. Conclusion Our tool enables an enhanced risk stratification compared with IPI, and the results indicate that imaging can be used to improve the prediction of central nervous system relapse in DLBCL. These findings support integration of clinically informative AI-generated imaging metrics into clinical workflows to improve identification of high-risk DLBCL patients. Trial Registration Registered clinicaltrials.gov number: NCT01287741. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s40644-022-00476-0.
Collapse
Affiliation(s)
- S Jemaa
- 1PHC Imaging, Genentech, Inc, South San Francisco, CA, USA
| | - J N Paulson
- Biostatistics, Genentech, Inc, South San Francisco, CA, USA
| | - M Hutchings
- Department of HaematologyRigshospitalet, Copenhagen, Denmark
| | - L Kostakoglu
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | - J Trotman
- Department of Haematology, Concord Repatriation General Hospital, University of Sydney, Concord, NSW, Australia
| | - S Tracy
- Biostatistics, Genentech, Inc, South San Francisco, CA, USA
| | - A de Crespigny
- Clinical Imaging Group, Genentech, Inc, South San Francisco, CA, USA
| | - R A D Carano
- 1PHC Imaging, Genentech, Inc, South San Francisco, CA, USA
| | - T C El-Galaly
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark
| | - T G Nielsen
- Pharmaceutical Development Clinical Oncology, F. Hoffmann-La Roche Ltd, Bldg 1, Grenzarcherstrasse 124m, CH-4070, Basel, Switzerland.
| | - T Bengtsson
- 1PHC Imaging, Genentech, Inc, South San Francisco, CA, USA.,Department of Statistics, University of California-Berkeley, Berkeley, CA, USA
| |
Collapse
|
43
|
Hormuth DA, Farhat M, Christenson C, Curl B, Chad Quarles C, Chung C, Yankeelov TE. Opportunities for improving brain cancer treatment outcomes through imaging-based mathematical modeling of the delivery of radiotherapy and immunotherapy. Adv Drug Deliv Rev 2022; 187:114367. [PMID: 35654212 PMCID: PMC11165420 DOI: 10.1016/j.addr.2022.114367] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/25/2022] [Accepted: 05/25/2022] [Indexed: 11/01/2022]
Abstract
Immunotherapy has become a fourth pillar in the treatment of brain tumors and, when combined with radiation therapy, may improve patient outcomes and reduce the neurotoxicity. As with other combination therapies, the identification of a treatment schedule that maximizes the synergistic effect of radiation- and immune-therapy is a fundamental challenge. Mechanism-based mathematical modeling is one promising approach to systematically investigate therapeutic combinations to maximize positive outcomes within a rigorous framework. However, successful clinical translation of model-generated combinations of treatment requires patient-specific data to allow the models to be meaningfully initialized and parameterized. Quantitative imaging techniques have emerged as a promising source of high quality, spatially and temporally resolved data for the development and validation of mathematical models. In this review, we will present approaches to personalize mechanism-based modeling frameworks with patient data, and then discuss how these techniques could be leveraged to improve brain cancer outcomes through patient-specific modeling and optimization of treatment strategies.
Collapse
Affiliation(s)
- David A Hormuth
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX 78712, USA; Departments of Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX 78712, USA.
| | - Maguy Farhat
- Departments of Radiation Oncology, MD Anderson Cancer Center, Houston, TX 77230, USA
| | - Chase Christenson
- Departments of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Brandon Curl
- Departments of Radiation Oncology, MD Anderson Cancer Center, Houston, TX 77230, USA
| | - C Chad Quarles
- Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Caroline Chung
- Departments of Radiation Oncology, MD Anderson Cancer Center, Houston, TX 77230, USA
| | - Thomas E Yankeelov
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX 78712, USA; Departments of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; Departments of Diagnostic Medicine, The University of Texas at Austin, Austin, TX 78712, USA; Departments of Oncology, The University of Texas at Austin, Austin, TX 78712, USA; Departments of Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX 78712, USA; Departments of Imaging Physics, MD Anderson Cancer Center, Houston, TX 77230, USA
| |
Collapse
|
44
|
Edmonds CE, O'Brien SR, Mankoff DA, Pantel AR. Novel applications of molecular imaging to guide breast cancer therapy. Cancer Imaging 2022; 22:31. [PMID: 35729608 PMCID: PMC9210593 DOI: 10.1186/s40644-022-00468-0] [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: 04/03/2022] [Accepted: 05/30/2022] [Indexed: 11/10/2022] Open
Abstract
The goals of precision oncology are to provide targeted drug therapy based on each individual’s specific tumor biology, and to enable the prediction and early assessment of treatment response to allow treatment modification when necessary. Thus, precision oncology aims to maximize treatment success while minimizing the side effects of inadequate or suboptimal therapies. Molecular imaging, through noninvasive assessment of clinically relevant tumor biomarkers across the entire disease burden, has the potential to revolutionize clinical oncology, including breast oncology. In this article, we review breast cancer positron emission tomography (PET) imaging biomarkers for providing early response assessment and predicting treatment outcomes. For 2-18fluoro-2-deoxy-D-glucose (FDG), a marker of cellular glucose metabolism that is well established for staging multiple types of malignancies including breast cancer, we highlight novel applications for early response assessment. We then review current and future applications of novel PET biomarkers for imaging the steroid receptors, including the estrogen and progesterone receptors, the HER2 receptor, cellular proliferation, and amino acid metabolism.
Collapse
Affiliation(s)
- Christine E Edmonds
- Department of Radiology, Hospital of the University if Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA.
| | - Sophia R O'Brien
- Department of Radiology, Hospital of the University if Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - David A Mankoff
- Department of Radiology, Hospital of the University if Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Austin R Pantel
- Department of Radiology, Hospital of the University if Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| |
Collapse
|
45
|
Jeffers CD, Lawhn-Heath C, Butterfield RI, Hoffman JM, Scott PJH. SNMMI Clinical Trials Network Research Series for Technologists: Clinical Research Primer- Use of Imaging Agents in Therapeutic Drug Development and Approval. J Nucl Med Technol 2022; 50:jnmt.122.264372. [PMID: 35701219 DOI: 10.2967/jnmt.122.264372] [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: 05/04/2022] [Accepted: 06/11/2022] [Indexed: 11/16/2022] Open
Abstract
The process of bringing a new drug to market is complex and has recently necessitated a new drug discovery paradigm for the pharmaceutical industry that is both more efficient and more economical. Key to this has been the increasing use of nuclear medicine and molecular imaging to support drug discovery efforts by answering critical questions on the pathway for development and approval of a new therapeutic drug. Some of these questions include: (i) Does the new drug reach its intended target in the body at sufficient levels to effectively treat or diagnose disease without unacceptable toxicity? (ii) How is the drug absorbed, metabolized, and excreted? (iii) What is the effective dose in humans? To conduct the appropriate imaging studies to answer such questions, pharmaceutical companies are increasingly partnering with molecular imaging departments. Nuclear medicine technologists are critical to this process as they perform scans to collect the qualitative and quantitative imaging data used to measure study endpoints. This article describes preclinical and clinical research trials and provides an overview of the different ways that radiopharmaceuticals are used to answer critical questions during therapeutic drug development.
Collapse
|
46
|
m6A methylation mediates LHPP acetylation as a tumour aerobic glycolysis suppressor to improve the prognosis of gastric cancer. Cell Death Dis 2022; 13:463. [PMID: 35568711 PMCID: PMC9107493 DOI: 10.1038/s41419-022-04859-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 04/06/2022] [Accepted: 04/14/2022] [Indexed: 12/14/2022]
Abstract
LHPP, a histidine phosphatase, has been implicated in tumour progression. However, its role, underlying mechanisms, and prognostic significance in human gastric cancer (GC) are elusive. Here, we obtained GC tissues and corresponding normal tissues from 48 patients and identified LHPP as a downregulated gene via RNA-seq. qRT-PCR and western blotting were applied to examine LHPP levels in normal and GC tissues. The prognostic value of LHPP was elucidated using tissue microarray and IHC analyses in two independent GC cohorts. The functional roles and mechanistic insights of LHPP in GC growth and metastasis were evaluated in vitro and in vivo. The results showed that LHPP expression was significantly decreased in GC tissues at both the mRNA and protein levels. Multivariate Cox regression analysis revealed that LHPP was an independent prognostic factor and effective predictor in patients with GC. The low expression of LHPP was significantly related to the poor prognosis and chemotherapy sensitivity of gastric cancer patients. Moreover, elevated LHPP expression effectively suppressed GC growth and metastasis in vitro and in vivo. Mechanistically, the m6A modification of LHPP mRNA by METTL14 represses its expression; LHPP inhibits the phosphorylation of GSK3b through acetylation and mediates HIF1A to inhibit glycolysis, proliferation, invasion and metastasis of gastric cancer cells. Together, our findings suggest that LHPP is regulated by m6A methylation and regulates the metabolism of GC by changing the acetylation level. Thus, LHPP is a potential predictive biomarker and therapeutic target for GC.
Collapse
|
47
|
Li Y. Inactivation of PDH can Reduce Anaplastic Thyroid Cancer Cells' Sensitivity to Artemisinin. Anticancer Agents Med Chem 2022; 22:1753-1760. [PMID: 34515013 DOI: 10.2174/1871520621666210910100803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 08/02/2021] [Accepted: 08/09/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Anaplastic Thyroid Cancer (ATC) is a rare subtype of thyroid tumors with a high mortality rate. Targeted therapies against ATC are ineffective and mostly transient. Artemisinin has shown excellent anti-tumor activity in several cancers, but its effects on ATC are still unknown. OBJECTIVE To evaluate the effects of artemisinin on ATC cells and assess the mechanism underlying drug resistance. METHODS The viability and proliferation rates of the artemisinin-treated CAL-62 and BHT-101 cells were analyzed by MTT and EdU incorporation assays. The protein expression levels were determined by Tandem Mass Tag (TMT) labeling quantitative proteomics and western blotting. RESULTS Artemisinin treatment significantly decreased the expression levels of COX2 and COX7A2 and increased that of COX14, YEM1l1, ALAS1, and OAT after 48h. In addition, FTL was upregulated in the CAL-62 cells and downregulated in BHT-101 cells. The CAL-62 cells showed transient and reversible resistance to artemisinin, which was correlated to time-dependent changes in HIF1α, PDK1, and PDHA levels. CONCLUSION Artemisinin targets the mitochondrial respiratory chain proteins in ATC cells. CAL-62 cells show transient resistance to artemisinin via PDH downregulation, indicating that PDH activation may enhance the cytotoxic effects of artemisinin on ATC cells.
Collapse
Affiliation(s)
- Yitian Li
- Research Department of Jining Medical University, Jining, Shandong, China
| |
Collapse
|
48
|
Cember ATJ, Wilson NE, Rich LJ, Bagga P, Nanga RPR, Swago S, Swain A, Thakuri D, Elliot M, Schnall MD, Detre JA, Reddy R. Integrating 1H MRS and deuterium labeled glucose for mapping the dynamics of neural metabolism in humans. Neuroimage 2022; 251:118977. [PMID: 35143973 PMCID: PMC9166154 DOI: 10.1016/j.neuroimage.2022.118977] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/13/2022] [Accepted: 02/05/2022] [Indexed: 11/21/2022] Open
Abstract
In the technique presented here, dubbed 'qMRS', we quantify the change in 1H MRS signal following administration of 2H-labeled glucose. As in recent human DMRS studies, we administer [6,6'-2H2]-glucose orally to healthy subjects. Since 2H is not detectable by 1H MRS, the transfer of the 2H label from glucose to a downstream metabolite leads to a reduction in the corresponding 1H MRS resonance of the metabolite, even if the total concentration of both isoforms remains constant. Moreover, introduction of the deuterium label alters the splitting pattern of the proton resonances, making indirect detection of the deuterated forms- as well as the direct detection of the decrease in unlabeled form- possible even without a 2H coil. Because qMRS requires only standard 1H MRS acquisition methods, it can be performed using commonly implemented single voxel spectroscopy (SVS) and chemical shift imaging (CSI) sequences. In this work, we implement qMRS in semi-LASER based CSI, generating dynamic maps arising from the fitted spectra, and demonstrating the feasibility of using qMRS and qCSI to monitor dynamic metabolism in the human brain using a 7T scanner with no auxiliary hardware.
Collapse
Affiliation(s)
- Abigail T J Cember
- Department of Radiology, Center for Advanced Metabolic Imaging in Precision Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Graduate Group in Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Neil E Wilson
- Department of Radiology, Center for Advanced Metabolic Imaging in Precision Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Siemens Medical Solutions USA, Malvern, PA, USA
| | - Laurie J Rich
- Department of Radiology, Center for Advanced Metabolic Imaging in Precision Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Puneet Bagga
- Department of Radiology, Center for Advanced Metabolic Imaging in Precision Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ravi Prakash Reddy Nanga
- Department of Radiology, Center for Advanced Metabolic Imaging in Precision Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Sophia Swago
- Department of Radiology, Center for Advanced Metabolic Imaging in Precision Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Anshuman Swain
- Department of Radiology, Center for Advanced Metabolic Imaging in Precision Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Deepa Thakuri
- Department of Radiology, Center for Advanced Metabolic Imaging in Precision Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Mark Elliot
- Department of Radiology, Center for Advanced Metabolic Imaging in Precision Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Mitchell D Schnall
- Department of Radiology, Center for Advanced Metabolic Imaging in Precision Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - John A Detre
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Ravinder Reddy
- Department of Radiology, Center for Advanced Metabolic Imaging in Precision Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
49
|
Xu J, Peng C, Yao B, Xu HJ, Xie Q. Direct Deoxyfluorination of Alcohols with KF as the Fluorine Source. J Org Chem 2022; 87:6471-6478. [PMID: 35442691 DOI: 10.1021/acs.joc.2c00388] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
This report describes a method for the deoxyfluorination of alcohols with KF as the fluorine source via in situ generation of highly active CF3SO2F. Diverse functionalities, including halogen, nitro, ketone, ester, alkene, and alkyne, are well tolerated. Mild conditions, a short reaction time, and a wide substrate scope make this method an excellent choice for the construction of C-F bonds.
Collapse
Affiliation(s)
- Jun Xu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Chao Peng
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Bolin Yao
- Department of Nuclear Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei 230001, P. R. China
| | - Hua-Jian Xu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Qiang Xie
- Department of Nuclear Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei 230001, P. R. China
| |
Collapse
|
50
|
Jiang M, Zhang X, Chen Y, Chen P, Guo X, Ma L, Gao Q, Mei W, Zhang J, Zheng J. A Review of the Correlation Between Epidermal Growth Factor Receptor Mutation Status and 18F-FDG Metabolic Activity in Non-Small Cell Lung Cancer. Front Oncol 2022; 12:780186. [PMID: 35515138 PMCID: PMC9065410 DOI: 10.3389/fonc.2022.780186] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 03/25/2022] [Indexed: 11/15/2022] Open
Abstract
PET/CT with 18F-2-fluoro-2-deoxyglucose (18F-FDG) has been proposed as a promising modality for diagnosing and monitoring treatment response and evaluating prognosis for patients with non-small cell lung cancer (NSCLC). The status of epidermal growth factor receptor (EGFR) mutation is a critical signal for the treatment strategies of patients with NSCLC. Higher response rates and prolonged progression-free survival could be obtained in patients with NSCLC harboring EGFR mutations treated with tyrosine kinase inhibitors (TKIs) when compared with traditional cytotoxic chemotherapy. However, patients with EGFR mutation treated with TKIs inevitably develop drug resistance, so predicting the duration of resistance is of great importance for selecting individual treatment strategies. Several semiquantitative metabolic parameters, e.g., maximum standard uptake value (SUVmax), metabolic tumor volume (MTV), and total lesion glycolysis (TLG), measured by PET/CT to reflect 18F-FDG metabolic activity, have been demonstrated to be powerful in predicting the status of EGFR mutation, monitoring treatment response of TKIs, and assessing the outcome of patients with NSCLC. In this review, we summarize the biological and clinical correlations between EGFR mutation status and 18F-FDG metabolic activity in NSCLC. The metabolic activity of 18F-FDG, as an extrinsic manifestation of NSCLC, could reflect the mutation status of intrinsic factor EGFR. Both of them play a critical role in guiding the implementation of treatment modalities and evaluating therapy efficacy and outcome for patients with NSCLC.
Collapse
Affiliation(s)
- Maoqing Jiang
- Department of PET/CT Center, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
- Department of Nuclear Medicine, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Xiaohui Zhang
- Department of PET/CT Center, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| | - Yan Chen
- Department of Physical Examination Center, Ningbo First Hospital, Ningbo, China
| | - Ping Chen
- Department of Nephrology, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Xiuyu Guo
- Department of PET/CT Center, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| | - Lijuan Ma
- Department of PET/CT Center, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| | - Qiaoling Gao
- Department of PET/CT Center, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| | - Weiqi Mei
- Department of Nuclear Medicine, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Jingfeng Zhang
- Department of Education, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Jianjun Zheng
- Department of PET/CT Center, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| |
Collapse
|