1
|
Zhang W, Liang X, Zhang X, Tong W, Shi G, Guo H, Jin Z, Tian J, Du Y, Xue H. Magnetic-optical dual-modality imaging monitoring chemotherapy efficacy of pancreatic ductal adenocarcinoma with a low-dose fibronectin-targeting Gd-based contrast agent. Eur J Nucl Med Mol Imaging 2024; 51:1841-1855. [PMID: 38372766 DOI: 10.1007/s00259-024-06617-w] [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: 10/19/2023] [Accepted: 01/15/2024] [Indexed: 02/20/2024]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) is a lethal hypovascular tumor surrounded by dense fibrosis. Albumin-bound paclitaxel and gemcitabine (AG) chemotherapy is the mainstay of PDAC treatment through depleting peritumoral fibrosis and killing tumor cells; however, it remains challenging due to the lack of a noninvasive imaging method evaluating fibrotic changes during AG chemotherapy. In this study, we developed a dual-modality imaging platform that enables noninvasive, dynamic, and quantitative assessment of chemotherapy-induced fibrotic changes through near-infrared fluorescence molecular imaging (FMI) and magnetic resonance imaging (MRI) using an extradomain B fibronectin (EDB-FN)-targeted imaging probe (ZD2-Gd-DOTA-Cy7). METHODS The ZD2-Gd-DOTA-Cy7 probe was constructed by conjugating a peptide (Cys-TVRTSAD) to Gd-DOTA and the near-infrared dye Cy7. PDAC murine xenograft models were intravenously injected with ZD2-Gd-DOTA-Cy7 at a Gd concentration of 0.05 mmol/kg or free Cy7 and Gd-DOTA as control. The normalized tumor background ratio (TBR) on FMI and the T1 reduction ratio on MRI were quantitatively analyzed. For models receiving AG chemotherapy or saline, MRI/FMI was performed before and after treatment. Histological analyses were performed for validation. RESULTS The ZD2-Gd-DOTA-Cy7 concentration showed a linear correlation with the fluorescence intensity and T1 relaxation time in vitro. The optimal imaging time was 30 min after injection of the ZD2-Gd-DOTA-Cy7 (0.05 mmol/kg), only half of the clinic dosage of gadolinium. Additionally, ZD2-Gd-DOTA-Cy7 generated a 1.44-fold and 1.90-fold robust contrast enhancement compared with Cy7 (P < 0.05) and Gd-DOTA (P < 0.05), respectively. For AG chemotherapy monitoring, the T1 reduction ratio and normalized TBR in the fibrotic tumor areas were significantly increased by 1.99-fold (P < 0.05) and 1.78-fold (P < 0.05), respectively, in the control group compared with those in the AG group. CONCLUSION MRI/FMI with a low dose of ZD2-Gd-DOTA-Cy7 enables sensitive imaging of PDAC and the quantitative assessment of fibrotic changes during AG chemotherapy, which shows potential clinical applications for precise diagnosis, post-treatment monitoring, and disease management.
Collapse
Affiliation(s)
- Wenjia Zhang
- Department of Radiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Beijing, 100730, China
- Department of Radiology, Peking University People's Hospital, Beijing, 100032, China
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, No. 95 Zhongguancun East Road, Beijing, 100190, China
| | - Xiaolong Liang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Xinyu Zhang
- Department of Radiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Beijing, 100730, China
| | - Wei Tong
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, No. 95 Zhongguancun East Road, Beijing, 100190, China
| | - Guangyuan Shi
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, No. 95 Zhongguancun East Road, Beijing, 100190, China
| | - Haozhuo Guo
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, No. 95 Zhongguancun East Road, Beijing, 100190, China
| | - Zhengyu Jin
- Department of Radiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Beijing, 100730, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, No. 95 Zhongguancun East Road, Beijing, 100190, China.
- Beijing Advanced Innovation Centre for Big Data-Based Precision Medicine, School of Medicine, Beihang University, Beijing, 100191, China.
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, No. 95 Zhongguancun East Road, Beijing, 100190, China.
- The University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Huadan Xue
- Department of Radiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Beijing, 100730, China.
| |
Collapse
|
2
|
Ju J, Xu D, Mo X, Miao J, Xu L, Ge G, Zhu X, Deng H. Multifunctional polysaccharide nanoprobes for biological imaging. Carbohydr Polym 2023; 317:121048. [PMID: 37364948 DOI: 10.1016/j.carbpol.2023.121048] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/19/2023] [Accepted: 05/20/2023] [Indexed: 06/28/2023]
Abstract
Imaging and tracking biological targets or processes play an important role in revealing molecular mechanisms and disease states. Bioimaging via optical, nuclear, or magnetic resonance techniques enables high resolution, high sensitivity, and high depth imaging from the whole animal down to single cells via advanced functional nanoprobes. To overcome the limitations of single-modality imaging, multimodality nanoprobes have been engineered with a variety of imaging modalities and functionalities. Polysaccharides are sugar-containing bioactive polymers with superior biocompatibility, biodegradability, and solubility. The combination of polysaccharides with single or multiple contrast agents facilitates the development of novel nanoprobes with enhanced functions for biological imaging. Nanoprobes constructed with clinically applicable polysaccharides and contrast agents hold great potential for clinical translations. This review briefly introduces the basics of different imaging modalities and polysaccharides, then summarizes the recent progress of polysaccharide-based nanoprobes for biological imaging in various diseases, emphasizing bioimaging with optical, nuclear, and magnetic resonance techniques. The current issues and future directions regarding the development and applications of polysaccharide nanoprobes are further discussed.
Collapse
Affiliation(s)
- Jingxuan Ju
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Danni Xu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xuan Mo
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiaqian Miao
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Li Xu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Guangbo Ge
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Hongping Deng
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
3
|
Łopuszyńska N, Węglarz WP. Contrasting Properties of Polymeric Nanocarriers for MRI-Guided Drug Delivery. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2163. [PMID: 37570481 PMCID: PMC10420849 DOI: 10.3390/nano13152163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/20/2023] [Accepted: 07/20/2023] [Indexed: 08/13/2023]
Abstract
Poor pharmacokinetics and low aqueous solubility combined with rapid clearance from the circulation of drugs result in their limited effectiveness and generally high therapeutic doses. The use of nanocarriers for drug delivery can prevent the rapid degradation of the drug, leading to its increased half-life. It can also improve the solubility and stability of drugs, advance their distribution and targeting, ensure a sustained release, and reduce drug resistance by delivering multiple therapeutic agents simultaneously. Furthermore, nanotechnology enables the combination of therapeutics with biomedical imaging agents and other treatment modalities to overcome the challenges of disease diagnosis and therapy. Such an approach is referred to as "theranostics" and aims to offer a more patient-specific approach through the observation of the distribution of contrast agents that are linked to therapeutics. The purpose of this paper is to present the recent scientific reports on polymeric nanocarriers for MRI-guided drug delivery. Polymeric nanocarriers are a very broad and versatile group of materials for drug delivery, providing high loading capacities, improved pharmacokinetics, and biocompatibility. The main focus was on the contrasting properties of proposed polymeric nanocarriers, which can be categorized into three main groups: polymeric nanocarriers (1) with relaxation-type contrast agents, (2) with chemical exchange saturation transfer (CEST) properties, and (3) with direct detection contrast agents based on fluorinated compounds. The importance of this aspect tends to be downplayed, despite its being essential for the successful design of applicable theranostic nanocarriers for image-guided drug delivery. If available, cytotoxicity and therapeutic effects were also summarized.
Collapse
Affiliation(s)
- Natalia Łopuszyńska
- Department of Magnetic Resonance Imaging, Institute of Nuclear Physics Polish Academy of Sciences, 31-342 Cracow, Poland
| | - Władysław P. Węglarz
- Department of Magnetic Resonance Imaging, Institute of Nuclear Physics Polish Academy of Sciences, 31-342 Cracow, Poland
| |
Collapse
|
4
|
Longo DL, Carella A, Corrado A, Pirotta E, Mohanta Z, Singh A, Stabinska J, Liu G, McMahon MT. A snapshot of the vast array of diamagnetic CEST MRI contrast agents. NMR IN BIOMEDICINE 2023; 36:e4715. [PMID: 35187749 PMCID: PMC9724179 DOI: 10.1002/nbm.4715] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 05/11/2023]
Abstract
Since the inception of CEST MRI in the 1990s, a number of compounds have been identified as suitable for generating contrast, including paramagnetic lanthanide complexes, hyperpolarized atom cages and, most interesting, diamagnetic compounds. In the past two decades, there has been a major emphasis in this field on the identification and application of diamagnetic compounds that have suitable biosafety profiles for usage in medical applications. Even in the past five years there has been a tremendous growth in their numbers, with more and more emphasis being placed on finding those that can be ultimately used for patient studies on clinical 3 T scanners. At this point, a number of endogenous compounds present in tissue have been identified, and also natural and synthetic organic compounds that can be administered to highlight pathology via CEST imaging. Here we will provide a very extensive snapshot of the types of diamagnetic compound that can generate CEST MRI contrast, together with guidance on their utility on typical preclinical and clinical scanners and a review of the applications that might benefit the most from this new technology.
Collapse
Affiliation(s)
- Dario Livio Longo
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Turin, Italy
| | - Antonella Carella
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Turin, Italy
| | - Alessia Corrado
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Turin, Italy
| | - Elisa Pirotta
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Turin, Italy
| | - Zinia Mohanta
- F.M. Kirby Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aruna Singh
- F.M. Kirby Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Julia Stabinska
- F.M. Kirby Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Guanshu Liu
- F.M. Kirby Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael T. McMahon
- F.M. Kirby Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
5
|
Knutsson L, Xu X, van Zijl PCM, Chan KWY. Imaging of sugar-based contrast agents using their hydroxyl proton exchange properties. NMR IN BIOMEDICINE 2023; 36:e4784. [PMID: 35665547 PMCID: PMC9719573 DOI: 10.1002/nbm.4784] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/30/2022] [Accepted: 06/03/2022] [Indexed: 05/13/2023]
Abstract
The ability of CEST MRI to detect the presence of millimolar concentrations of non-metallic contrast agents has made it possible to study, non-invasively, important biological molecules such as proteins and sugars, as well as drugs already approved for clinical use. Here, we review efforts to use sugar and sugar polymers as exogenous contrast agents, which is possible based on the exchange of their hydroxyl protons with water protons. While this capability has raised early enthusiasm, for instance about the possibility of imaging D-glucose metabolism with MRI in a way analogous to PET, experience over the past decade has shown that this is not trivial. On the other hand, many studies have confirmed the possibility of imaging a large variety of sugar analogues, each with potentially interesting applications to assess tissue physiology. Some promising applications are the study of (i) sugar delivery and transport to assess blood-brain barrier integrity and (ii) sugar uptake by cells for their characterization (e.g., cancer versus healthy), as well as (iii) clearance of sugars to assess tissue drainage-for instance, through the glymphatic system. To judge these opportunities and their challenges, especially in the clinic, it is necessary to understand the technical aspects of detecting the presence of rapidly exchanging protons through the water signal in MRI, especially as a function of magnetic field strength. We expect that novel approaches in terms of MRI detection (both saturation transfer and relaxation based), MRI data analysis, and sugar design will push this young field forward in the next decade.
Collapse
Affiliation(s)
- Linda Knutsson
- Department of Medical Radiation Physics, Lund University, Lund, Sweden
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, US
| | - Xiang Xu
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Peter CM van Zijl
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, US
| | - Kannie WY Chan
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong, China
- Tung Biomedical Sciences Centre, City University of Hong Kong
- City University of Hong Kong Shenzhen Institute, Shenzhen, China
| |
Collapse
|
6
|
Liu G, Ling J, He L, Xu Y, Chen T, Shi C, Luo L. Theranostic Cancer Treatment Using Lentinan-Coated Selenium Nanoparticles and Label-Free CEST MRI. Pharmaceutics 2022; 15:pharmaceutics15010120. [PMID: 36678748 PMCID: PMC9864256 DOI: 10.3390/pharmaceutics15010120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/20/2022] [Accepted: 12/24/2022] [Indexed: 01/01/2023] Open
Abstract
Selenium nanoparticle (SeNP)-based nanotherapeutics have become an emerging cancer therapy, while effective drug delivery remains a technical hurdle. A theranostic approach, through which imaging companions are integrated with SeNPs, will allow image-guided drug delivery and, therefore, is highly desirable. Traditional methods require the chemical conjugation of imaging agents to the surface of nanoparticles, which may impede the later clinical translation. In this study, we developed a label-free strategy in which lentinan-functionalized SeNPs (LNT-SeNPs) are detected using MRI by the hydroxyl protons carried on LNT molecules. The in vitro phantom study showed that LNT and LNT-SeNPs have a strong CEST signal at 1.0 ppm apart from the water resonance, suggesting an in vivo detectability in the µM concentration range. Demonstrated on CT26 colon tumor cells, LNT-SeNPs exert a strong anticancer effect (IC50 = 4.8 μM), prominently attributed to the ability to generate intracellular reactive oxygen species. However, when testing in a mouse model of CT26 tumors, administration of LNT-SeNPs alone was found unable to deliver sufficient drugs to the tumor, leading to poor treatment responses. To improve the drug delivery, we co-injected LNT-SeNPs and TNF-α, a previously reported drug that could effectively damage the endothelial cells in the tumor vasculature, thereby increasing drug delivery to the tumor. Our results revealed a 75% increase in the intratumoral CEST MRI signal, indicating a markedly increased delivery efficiency of LNT-SeNPs when combined with TNF-α. The combination therapy also resulted in a significantly enhanced treatment outcome, as revealed by the tumor growth study. Taken together, our study demonstrates the first label-free, SeNP-based theranostic system, in which LNT was used for both functional surface coating and CEST MRI signal generating. Such a theranostic LNT-SeNP system is advantageous because it requires chemical labeling and, therefore, has high biocompatibility and low translatable barriers.
Collapse
Affiliation(s)
- Guanfu Liu
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Jiabao Ling
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Lizhen He
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Yuan Xu
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Tianfeng Chen
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
- Correspondence: (T.C.); (C.S.); Tel.: +86-022-85223393 (T.C.); +86-020-38688848 (C.S.)
| | - Changzheng Shi
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
- Correspondence: (T.C.); (C.S.); Tel.: +86-022-85223393 (T.C.); +86-020-38688848 (C.S.)
| | - Liangping Luo
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| |
Collapse
|
7
|
Liu J, Chu C, Zhang J, Bie C, Chen L, Aafreen S, Xu J, Kamson DO, van Zijl PCM, Walczak P, Janowski M, Liu G. Label-Free Assessment of Mannitol Accumulation Following Osmotic Blood-Brain Barrier Opening Using Chemical Exchange Saturation Transfer Magnetic Resonance Imaging. Pharmaceutics 2022; 14:pharmaceutics14112529. [PMID: 36432721 PMCID: PMC9695341 DOI: 10.3390/pharmaceutics14112529] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/02/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022] Open
Abstract
PURPOSE Mannitol is a hyperosmolar agent for reducing intracranial pressure and inducing osmotic blood-brain barrier opening (OBBBO). There is a great clinical need for a non-invasive method to optimize the safety of mannitol dosing. The aim of this study was to develop a label-free Chemical Exchange Saturation Transfer (CEST)-based MRI approach for detecting intracranial accumulation of mannitol following OBBBO. METHODS In vitro MRI was conducted to measure the CEST properties of D-mannitol of different concentrations and pH. In vivo MRI and MRS measurements were conducted on Sprague-Dawley rats using a Biospec 11.7T horizontal MRI scanner. Rats were catheterized at the internal carotid artery (ICA) and randomly grouped to receive either 1 mL or 3 mL D-mannitol. CEST MR images were acquired before and at 20 min after the infusion. RESULTS In vitro MRI showed that mannitol has a strong, broad CEST contrast at around 0.8 ppm with a mM CEST MRI detectability. In vivo studies showed that CEST MRI could effectively detect mannitol in the brain. The low dose mannitol treatment led to OBBBO but no significant mannitol accumulation, whereas the high dose regimen resulted in both OBBBO and mannitol accumulation. The CEST MRI findings were consistent with 1H-MRS and Gd-enhanced MRI assessments. CONCLUSION We demonstrated that CEST MRI can be used for non-invasive, label-free detection of mannitol accumulation in the brain following BBBO treatment. This method may be useful as a rapid imaging tool to optimize the dosing of mannitol-based OBBBO and improve its safety and efficacy.
Collapse
Affiliation(s)
- Jing Liu
- Department of Radiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510230, China
- Russell H. Morgan Department of Radiology and Radiological Sciences, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Chengyan Chu
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Jia Zhang
- Russell H. Morgan Department of Radiology and Radiological Sciences, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Chongxue Bie
- Russell H. Morgan Department of Radiology and Radiological Sciences, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Lin Chen
- Russell H. Morgan Department of Radiology and Radiological Sciences, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Safiya Aafreen
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jiadi Xu
- Russell H. Morgan Department of Radiology and Radiological Sciences, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - David O. Kamson
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Peter C. M. van Zijl
- Russell H. Morgan Department of Radiology and Radiological Sciences, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Piotr Walczak
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Miroslaw Janowski
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Guanshu Liu
- Russell H. Morgan Department of Radiology and Radiological Sciences, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD 21205, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21218, USA
- Correspondence: ; Tel.: +1-443-923-9500; Fax: +1-410-614-3147
| |
Collapse
|
8
|
Lu ZR, Laney V, Li Y. Targeted Contrast Agents for Magnetic Resonance Molecular Imaging of Cancer. Acc Chem Res 2022; 55:2833-2847. [PMID: 36121350 DOI: 10.1021/acs.accounts.2c00346] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Magnetic resonance imaging (MRI) is a clinical imaging modality that provides high-resolution images of soft tissues, including cancerous lesions. Stable gadolinium(III) chelates have been used as contrast agents (CA) in MRI to enhance the contrast between the tissues of interest and surrounding tissues for accurate diagnostic imaging. Magnetic resonance molecular imaging (MRMI) of cancer requires targeted CA to specifically elucidate cancer-associated molecular processes and can provide high-resolution delineation and characterization of cancer for precision medicine. The main challenge for MRMI is the lack of sufficient sensitivity to detect the low concentration of the cellular oncogenic markers. In addition, targeted CA must satisfy regulatory safety requirements prior to clinical development. Up to now, there is no FDA-approved targeted CA for MRMI of cancer.In this Account, we discuss the latest developments in the design and development of clinically translatable targeted CA for MRMI of cancer, with an emphasis on our own research. The primary limitation of MRMI can be overcome by designing small molecular targeted CA to target abundant cancer-specific targets found in the tumor microenvironment (TME). For example, aggressive tumors have a unique extracellular matrix (ECM) composed of oncoproteins, which can be used as targetable markers for MRMI. We have designed and prepared small peptide conjugates of clinical contrast agents, including Gd-DTPA and Gd-DOTA, to target fibrin-fibronectin clots in tumors. These small molecular CA have been effective in enhancing MRMI detection of solid tumors and have demonstrated the ability to detect submillimeter cancer micrometastases in mouse tumor models, exceeding the detection limit of current clinical imaging modalities. We have also identified extradomain B fibronectin (EDB-FN), an oncofetal subtype of fibronectin, as a promising TME target to leverage in the design and development of small peptide targeted CA for clinical translation. The expression level of EDB-FN is correlated with invasiveness of cancer cells and poor patient survival of multiple cancer types. ZD2 peptide with a sequence of seven amino acids (TVRTSAD) was identified to specifically bind to the EDB protein fragment. Several ZD2 conjugates of macrocyclic GBCA, including Gd-DOTA and Gd(HP-DO3A), have been synthesized and tested in mouse tumor models. ZD2-N3-Gd(HP-DO3A) (MT218) with a high r1 relaxivity was selected as the lead agent for clinical translation. The physicochemical properties and preclinical assessments of MT218 are summarized in this Account. MRMI of EDB-FN with MT218 can effectively detect invasive tumors of multiple cancers with risk-stratification and monitor tumor response to anticancer therapies in mouse models. Currently, MT218 is in clinical trials for precision cancer MRMI. Herein, we will show that using targeted MRI contrast agents specific to abundant TME biomarkers is a pragmatic solution for effective precision cancer imaging in high spatial resolution. And thus, we illustrate a replicable approach for CA development that is vital for cancer MRMI.
Collapse
Affiliation(s)
- Zheng-Rong Lu
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Wickenden Building, Cleveland, Ohio 44106, United States.,Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Ave, Cleveland, Ohio 44106, United States
| | - Victoria Laney
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Wickenden Building, Cleveland, Ohio 44106, United States
| | - Yajuan Li
- Molecular Theranostics, 7100 Euclid Ave, Suite 152, Cleveland, Ohio 44114, United States
| |
Collapse
|
9
|
Mohan T, Kleinschek KS, Kargl R. Polysaccharide peptide conjugates: Chemistry, properties and applications. Carbohydr Polym 2022; 280:118875. [PMID: 35027118 DOI: 10.1016/j.carbpol.2021.118875] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/25/2021] [Accepted: 11/05/2021] [Indexed: 11/02/2022]
Abstract
The intention of this publication is to give an overview on research related to conjugates of polysaccharides and peptides. Dextran, chitosan, and alginate were selected, to cover four of the most often encountered functional groups known to be present in polysaccharides. These groups are the hydroxyl, the amine, the carboxyl, and the acetal functionality. A collection of the commonly used chemical reactions for conjugation is provided. Conjugation results into distinct properties compared to the parent polysaccharide, and a number of these characteristics are highlighted. This review aims at demonstrating the applicability of said conjugates with a strong emphasis on biomedical applications, drug delivery, biosensing, and tissue engineering. Some suggestions are made for more rigorous chemistries and analytics that could be investigated. Finally, an outlook is given into which direction the field could be developed further. We hope that this survey provides the reader with a comprehensive summary and contributes to the progress of works that aim at synthetically combining two of the main building blocks of life into supramolecular structures with unprecedented biological response.
Collapse
Affiliation(s)
- Tamilselvan Mohan
- Institute for Chemistry and Technology of Biobased Systems (IBIOSYS), Graz University of Technology, Stremayrgasse 9, 8010 Graz, Austria
| | - Karin Stana Kleinschek
- Institute for Chemistry and Technology of Biobased Systems (IBIOSYS), Graz University of Technology, Stremayrgasse 9, 8010 Graz, Austria
| | - Rupert Kargl
- Institute for Chemistry and Technology of Biobased Systems (IBIOSYS), Graz University of Technology, Stremayrgasse 9, 8010 Graz, Austria; Institute for Automation, Faculty of Electrical Engineering and Computer Science, University of Maribor, Smetanova ulica 17, 2000 Maribor, Slovenia.
| |
Collapse
|
10
|
Han Z, Chen C, Xu X, Bai R, Staedtke V, Huang J, Chan KW, Xu J, Kamson DO, Wen Z, Knutsson L, van Zijl PC, Liu G. Dynamic contrast-enhanced CEST MRI using a low molecular weight dextran. NMR IN BIOMEDICINE 2022; 35:e4649. [PMID: 34779550 PMCID: PMC8828685 DOI: 10.1002/nbm.4649] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 09/27/2021] [Accepted: 10/19/2021] [Indexed: 05/25/2023]
Abstract
Natural and synthetic sugars have great potential for developing highly biocompatible and translatable chemical exchange saturation transfer (CEST) MRI contrast agents. In this study, we aimed to develop the smallest clinically available form of dextran, Dex1 (molecular weight, MW ~ 1 kDa), as a new CEST agent. We first characterized the CEST properties of Dex1 in vitro at 11.7 T and showed that the Dex1 had a detectable CEST signal at ~1.2 ppm, attributed to hydroxyl protons. In vivo CEST MRI studies were then carried out on C57BL6 mice bearing orthotopic GL261 brain tumors (n = 5) using a Bruker BioSpec 11.7 T MRI scanner. Both steady-state full Z-spectral images and single offset (1.2 ppm) dynamic dextran-enhanced (DDE) images were acquired before and after the intravenous injection of Dex1 (2 g/kg). The steady-state Z-spectral analysis showed a significantly higher CEST contrast enhancement in the tumor than in contralateral brain (∆MTRasym1.2 ppm = 0.010 ± 0.006 versus 0.002 ± 0.008, P = 0.0069) at 20 min after the injection of Dex1. Pharmacokinetic analyses of DDE were performed using the area under the curve (AUC) in the first 10 min after Dex1 injection, revealing a significantly higher uptake of Dex1 in the tumor than in brain tissue for tumor-bearing mice (AUC[0-10 min] = 21.9 ± 4.2 versus 5.3 ± 6.4%·min, P = 0.0294). In contrast, no Dex1 uptake was foundling in the brains of non-tumor-bearing mice (AUC[0-10 min] = -1.59 ± 2.43%·min). Importantly, the CEST MRI findings were consistent with the measurements obtained using DCE MRI and fluorescence microscopy, demonstrating the potential of Dex1 as a highly translatable CEST MRI contrast agent for assessing tumor hemodynamics.
Collapse
Affiliation(s)
- Zheng Han
- Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Chuheng Chen
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, USA
| | - Xiang Xu
- Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Renyuan Bai
- Department of Neurology and Neurosurgery, Johns Hopkins University, Baltimore, MD, USA
| | - Verena Staedtke
- Department of Neurology and Neurosurgery, Johns Hopkins University, Baltimore, MD, USA
| | - Jianpan Huang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Kannie W.Y. Chan
- Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Jiadi Xu
- Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - David O. Kamson
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Zhibo Wen
- Department of Radiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Linda Knutsson
- Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
- Department of Medical Radiation Physics, Lund University, Lund, Sweden
| | - Peter C.M. van Zijl
- Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Guanshu Liu
- Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| |
Collapse
|
11
|
Gao T, Zou C, Li Y, Jiang Z, Tang X, Song X. A Brief History and Future Prospects of CEST MRI in Clinical Non-Brain Tumor Imaging. Int J Mol Sci 2021; 22:11559. [PMID: 34768990 PMCID: PMC8584005 DOI: 10.3390/ijms222111559] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/12/2021] [Accepted: 10/23/2021] [Indexed: 02/08/2023] Open
Abstract
Chemical exchange saturation transfer (CEST) MRI is a promising molecular imaging tool which allows the specific detection of metabolites that contain exchangeable amide, amine, and hydroxyl protons. Decades of development have progressed CEST imaging from an initial concept to a clinical imaging tool that is used to assess tumor metabolism. The first translation efforts involved brain imaging, but this has now progressed to imaging other body tissues. In this review, we summarize studies using CEST MRI to image a range of tumor types, including breast cancer, pelvic tumors, digestive tumors, and lung cancer. Approximately two thirds of the published studies involved breast or pelvic tumors which are sites that are less affected by body motion. Most studies conclude that CEST shows good potential for the differentiation of malignant from benign lesions with a number of reports now extending to compare different histological classifications along with the effects of anti-cancer treatments. Despite CEST being a unique 'label-free' approach with a higher sensitivity than MR spectroscopy, there are still some obstacles for implementing its clinical use. Future research is now focused on overcoming these challenges. Vigorous ongoing development and further clinical trials are expected to see CEST technology become more widely implemented as a mainstream imaging technology.
Collapse
Affiliation(s)
- Tianxin Gao
- School of Life Science, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, China; (T.G.); (C.Z.); (Z.J.)
| | - Chuyue Zou
- School of Life Science, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, China; (T.G.); (C.Z.); (Z.J.)
| | - Yifan Li
- Center for Biomedical Imaging Research, School of Medicine, Tsinghua University, Beijing 100084, China;
| | - Zhenqi Jiang
- School of Life Science, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, China; (T.G.); (C.Z.); (Z.J.)
| | - Xiaoying Tang
- School of Life Science, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, China; (T.G.); (C.Z.); (Z.J.)
| | - Xiaolei Song
- Center for Biomedical Imaging Research, School of Medicine, Tsinghua University, Beijing 100084, China;
| |
Collapse
|
12
|
Zhao X, Yang X, Wang X, Zhao X, Zhang Y, Liu S, Anderson GJ, Kim SJ, Li Y, Nie G. Penetration Cascade of Size Switchable Nanosystem in Desmoplastic Stroma for Improved Pancreatic Cancer Therapy. ACS NANO 2021; 15:14149-14161. [PMID: 34478262 DOI: 10.1021/acsnano.0c08860] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) cells are surrounded by a dense extracellular matrix (ECM), which greatly restricts the access of therapeutic agents, resulting in poor clinical response to chemotherapy. Transforming growth factor-β1 (TGF-β1) signaling plays a crucial role in construction of the desmoplastic stroma and provides potential targets for PDAC therapy. To surmount the pathological obstacle, we developed a size switchable nanosystem based on PEG-PLGA nanospheres encapsulated within liposomes for the combined delivery of vactosertib (VAC), a TGF-β1 receptor kinase inhibitor, and the cytotoxic drug paclitaxel (TAX). By surface modification of the liposomes with a peptide, APTEDB, the nanosystem can be anchored to abundant tumor-associated fibronectin in PDAC stroma and decreases its size by releasing encapsulated TAX-loaded nanospheres, as well as VAC after collapse of the liposomes. The inhibition of ECM hyperplasia by VAC allows TAX more ready access to the cancer cells in addition to its small size, thereby shrinking pancreatic tumor xenografts more effectively than a combination of the free drugs. This size switchable nanosystem enables sequential delivery of drugs at a fixed dose combination with simplified administration and provides an encouraging cascade approach of drug penetration for enhanced chemotherapy in cancers with a dense desmoplastic stroma.
Collapse
Affiliation(s)
- Xiaozheng Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Xiao Yang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
| | - Xudong Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
| | - Yinlong Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
| | - Shaoli Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
| | - Gregory J Anderson
- QIMR Berghofer Medical Research Institute, Royal Brisbane Hospital, Brisbane, Queensland 4029, Australia
| | - Seong-Jin Kim
- GILO Institute, GILO Foundation, Seoul 06668, Republic of Korea
- Medpacto Inc., 92 Myeongdal-ro, Seocho-gu, Seoul 06668, Republic of Korea
| | - Yiye Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- GBA Research Innovation Institute for Nanotechnology, Guangdong 510700, China
| |
Collapse
|
13
|
Ahmad RS, Eubank TD, Lukomski S, Boone BA. Immune Cell Modulation of the Extracellular Matrix Contributes to the Pathogenesis of Pancreatic Cancer. Biomolecules 2021; 11:biom11060901. [PMID: 34204306 PMCID: PMC8234537 DOI: 10.3390/biom11060901] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/07/2021] [Accepted: 06/13/2021] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy with a five-year survival rate of only 9%. PDAC is characterized by a dense, fibrotic stroma composed of extracellular matrix (ECM) proteins. This desmoplastic stroma is a hallmark of PDAC, representing a significant physical barrier that is immunosuppressive and obstructs penetration of cytotoxic chemotherapy agents into the tumor microenvironment (TME). Additionally, dense ECM promotes hypoxia, making tumor cells refractive to radiation therapy and alters their metabolism, thereby supporting proliferation and survival. In this review, we outline the significant contribution of fibrosis to the pathogenesis of pancreatic cancer, with a focus on the cross talk between immune cells and pancreatic stellate cells that contribute to ECM deposition. We emphasize the cellular mechanisms by which neutrophils and macrophages, specifically, modulate the ECM in favor of PDAC-progression. Furthermore, we investigate how activated stellate cells and ECM influence immune cells and promote immunosuppression in PDAC. Finally, we summarize therapeutic strategies that target the stroma and hinder immune cell promotion of fibrogenesis, which have unfortunately led to mixed results. An enhanced understanding of the complex interactions between the pancreatic tumor ECM and immune cells may uncover novel treatment strategies that are desperately needed for this devastating disease.
Collapse
Affiliation(s)
- Ramiz S. Ahmad
- Department of Surgery, West Virginia University, Morgantown, WV 26506, USA;
| | - Timothy D. Eubank
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV 26506, USA; (T.D.E.); (S.L.)
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Slawomir Lukomski
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV 26506, USA; (T.D.E.); (S.L.)
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Brian A. Boone
- Department of Surgery, West Virginia University, Morgantown, WV 26506, USA;
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV 26506, USA; (T.D.E.); (S.L.)
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA
- Correspondence:
| |
Collapse
|
14
|
Abstract
Mounting evidence shows the great promise of nanoparticle drug delivery systems (nano-DDSs) to improve delivery efficiency and reduce off-target adverse effects. By tracking drug delivery and distribution, monitoring nanoparticle degradation and drug release, aiding and optimizing treatment planning, and directing the design of more robust nano-DDSs, image guidance has become a vital component of nanomedicine. Recently, chemical exchange saturation transfer (CEST) magnetic resonance imaging (MRI) has emerged as an attempting imaging method for achieving image-guided drug delivery. One of the unbeatable advantages of CEST MRI is its ability to detect diamagnetic compounds that cannot be detected using conventional MRI methods, making a broad spectrum of bioorganic agents, natural compounds, even nano-carriers directly MRI detectable in a high-spatial-resolution manner. To date, CEST MRI has become a versatile and powerful imaging technology for non-invasive in vivo tracking of nanoparticles and their loaded drugs. In this review, we will provide a concise overview of different forms of recently developed, CEST MRI trackable nano-DDSs, including liposomes, polymeric nanoparticles, self-assembled drug-based nanoparticles, and carbon dots. The potential applications and future perspectives will also be discussed.
Collapse
Affiliation(s)
- Zheng Han
- Russell H. Morgan Department of Radiology & Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | | |
Collapse
|
15
|
Jugniot N, Bam R, Meuillet EJ, Unger EC, Paulmurugan R. Current status of targeted microbubbles in diagnostic molecular imaging of pancreatic cancer. Bioeng Transl Med 2021; 6:e10183. [PMID: 33532585 PMCID: PMC7823123 DOI: 10.1002/btm2.10183] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/19/2020] [Accepted: 08/19/2020] [Indexed: 12/14/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is often associated with a poor prognosis due to silent onset, resistance to therapies, and rapid spreading. Most patients are ineligible for curable surgery as they present with advanced disease at the time of diagnosis. Present diagnostic methods relying on anatomical changes have various limitations including difficulty to discriminate between benign and malignant conditions, invasiveness, the ambiguity of imaging results, or the inability to detect molecular biomarkers of PDAC initiation and progression. Therefore, new imaging technologies with high sensitivity and specificity are critically needed for accurately detecting PDAC and noninvasively characterizing molecular features driving its pathogenesis. Contrast enhanced targeted ultrasound (CETUS) is an upcoming molecular imaging modality that specifically addresses these issues. Unlike anatomical imaging modalities such as CT and MRI, molecular imaging using CETUS is promising for early and accurate detection of PDAC. The use of molecularly targeted microbubbles that bind to neovascular targets can enhance the ultrasound signal specifically from malignant PDAC tissues. This review discusses the current state of diagnostic imaging modalities for pancreatic cancer and places a special focus on ultrasound targeted-microbubble technology together with its clinical translatability for PDAC detection.
Collapse
Affiliation(s)
- Natacha Jugniot
- Department of RadiologyMolecular Imaging Program at Stanford, Stanford UniversityPalo AltoCaliforniaUSA
| | - Rakesh Bam
- Department of RadiologyMolecular Imaging Program at Stanford, Stanford UniversityPalo AltoCaliforniaUSA
| | | | | | - Ramasamy Paulmurugan
- Department of RadiologyMolecular Imaging Program at Stanford, Stanford UniversityPalo AltoCaliforniaUSA
| |
Collapse
|
16
|
Molecular and Functional Imaging and Theranostics of the Tumor Microenvironment. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00069-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
17
|
Liu G, van Zijl PC. CEST (Chemical Exchange Saturation Transfer) MR Molecular Imaging. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00012-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
18
|
Repurposing Clinical Agents for Chemical Exchange Saturation Transfer Magnetic Resonance Imaging: Current Status and Future Perspectives. Pharmaceuticals (Basel) 2020; 14:ph14010011. [PMID: 33374213 PMCID: PMC7824058 DOI: 10.3390/ph14010011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/19/2020] [Accepted: 12/21/2020] [Indexed: 12/24/2022] Open
Abstract
Molecular imaging is becoming an indispensable tool to pursue precision medicine. However, quickly translating newly developed magnetic resonance imaging (MRI) agents into clinical use remains a formidable challenge. Recently, Chemical Exchange Saturation Transfer (CEST) MRI is emerging as an attractive approach with the capability of directly using low concentration, exchangeable protons-containing agents for generating quantitative MRI contrast. The ability to utilize diamagnetic compounds has been extensively exploited to detect many clinical compounds, such as FDA approved drugs, X-ray/CT contrast agents, nutrients, supplements, and biopolymers. The ability to directly off-label use clinical compounds permits CEST MRI to be rapidly translated to clinical settings. In this review, the current status of CEST MRI based on clinically available compounds will be briefly introduced. The advancements and limitations of these studies are reviewed in the context of their pre-clinical or clinical applications. Finally, future directions will be briefly discussed.
Collapse
|
19
|
Qiao P, Ayat NR, Vaidya A, Gao S, Sun W, Chou S, Han Z, Gilmore H, Winter JM, Lu ZR. Magnetic Resonance Molecular Imaging of Extradomain B Fibronectin Improves Imaging of Pancreatic Cancer Tumor Xenografts. Front Oncol 2020; 10:586727. [PMID: 33194740 PMCID: PMC7661967 DOI: 10.3389/fonc.2020.586727] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/16/2020] [Indexed: 12/16/2022] Open
Abstract
The survival of pancreatic cancer patients can be greatly improved if their disease is detected at an early, potentially curable stage. Magnetic resonance molecular imaging (MRMI) of oncoproteins is a promising strategy for accurate, early detection of the disease. Here, we test the hypothesis that MRMI of extradomain-B fibronectin (EDB-FN), an abundant oncoprotein in the tumor extracellular matrix, can overcome the stromal barriers of pancreatic cancer to facilitate effective molecular imaging and detection of small tumors. Specimens of normal, premalignant, and malignant human pancreatic tissues were stained with a peptide-fluorophore conjugate (ZD2-Cy5.5) to assess EDB-FN binding and expression. MRMI with ZD2-N3-Gd(HP-DO3A) (MT218) specific to EDB-FN and MRI with Gd(HP-DO3A) were performed in three murine models bearing human pancreatic cancer xenografts, including a Capan-1 flank model, a BxPC3-GFP-Luc and a PANC-1-GFP-Luc intrapancreatic xenograft model. Tumor enhancement of the contrast agents was analyzed and compared. Staining of human tissue samples with ZD2-Cy5.5 revealed high EDB-FN expression in pancreatic tumors, moderate expression in premalignant tissue, and little expression in normal tissue. MRMI with MT218 generated robust intratumoral contrast, clearly detected and delineated small tumors (smallest average size: 6.1 mm2), and out-performed conventional contrast enhanced MRI with Gd(HP-DO3A). Quantitative analysis of signal enhancement revealed that MT218 produced 2.7, 2.1, and 1.6 times greater contrast-to-noise ratio (CNR) than the clinical agent in the Capan-1 flank, BxPC3-GFP-Luc and PANC-1-GFP-Luc intrapancreatic models, respectively (p < 0.05). MRMI of the ECM oncoprotein EDB-FN with MT218 is able to generate superior contrast enhancement in small pancreatic tumors and provide accurate tumor delineation in animal models. Early, accurate detection and delineation of pancreatic cancer with high-resolution MRMI has the potential to guide timely treatment and significantly improve the long-term survival of pancreatic cancer patients.
Collapse
Affiliation(s)
- Peter Qiao
- Department of Biomedical Engineering, Case School of Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Nadia R Ayat
- Department of Biomedical Engineering, Case School of Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Amita Vaidya
- Department of Biomedical Engineering, Case School of Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Songqi Gao
- Department of Biomedical Engineering, Case School of Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Wenyu Sun
- Department of Biomedical Engineering, Case School of Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Samuel Chou
- Department of Biomedical Engineering, Case School of Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Zheng Han
- Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Hannah Gilmore
- Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States.,Case Comprehensive Cancer Center, Case Western Reserve School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Jordan M Winter
- Case Comprehensive Cancer Center, Case Western Reserve School of Medicine, Case Western Reserve University, Cleveland, OH, United States.,Department of Surgery, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Zheng-Rong Lu
- Department of Biomedical Engineering, Case School of Engineering, Case Western Reserve University, Cleveland, OH, United States.,Case Comprehensive Cancer Center, Case Western Reserve School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
20
|
Vaidya A, Ayat N, Buford M, Wang H, Shankardass A, Zhao Y, Gilmore H, Wang Z, Lu ZR. Noninvasive assessment and therapeutic monitoring of drug-resistant colorectal cancer by MR molecular imaging of extradomain-B fibronectin. Theranostics 2020; 10:11127-11143. [PMID: 33042274 PMCID: PMC7532678 DOI: 10.7150/thno.47448] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 06/23/2020] [Indexed: 12/16/2022] Open
Abstract
Antineoplastic resistance represents a multifaceted challenge for cancer therapy and diagnostics. Extensive molecular heterogeneity, even within neoplasms of the same type, can elicit distinct outcomes of administering therapeutic pressures, frequently leading to the development of drug-resistant populations. Improved success of oncotherapies merits the exploration of precise molecular imaging technologies that can detect not only anatomical but also molecular changes in tumors and their microenvironment, early on in the treatment regimen. To this end, we developed magnetic resonance molecular imaging (MRMI) strategies to target the extracellular matrix oncoprotein, extradomain-B fibronectin (EDB-FN), for non-invasive assessment and therapeutic monitoring of drug-resistant colorectal cancer (CRC). Methods: Two drug-resistant CRC lines generated from parent DLD-1 and RKO cells by long-term treatment with 5'-FU and 5'-FU plus CB-839 respectively, were characterized for functional and gene expression changes using 3D culture, transwell invasion, qRT-PCR, and western blot assays. Contrast-enhanced MRMI of EDB-FN was performed in athymic nu/nu mice bearing subcutaneous tumor xenografts with 40 µmol/kg dose of macrocyclic ZD2-targeted contrast agent MT218 [ZD2-N3-Gd (HP-DO3A)] on a 3T MRS 3000 scanner. Immunohistochemistry was conducted on patient specimens and xenografts using anti-EDB-FN antibody G4. Results: Analyses of TCGA and GTEx databases revealed poor prognosis of colon cancer patients with higher levels of EDB-FN. Similarly, immunohistochemical staining of patient specimens showed increased EDB-FN expression in primary colon adenocarcinoma and hepatic metastases, but none in normal adjacent tissues. Drug-resistant DLD1-DR and RKO-DR cells were also found to demonstrate enhanced invasive potential and significantly elevated EDB-FN expression over their parent counterparts. MRMI of EDB-FN with 40 µmol/kg dose of MT218 (60% lower than the clinical dose) resulted in robust signal enhancement in the drug-resistant CRC xenografts with 84-120% increase in their contrast-to-noise ratios (CNRs) over the non-resistant counterparts. The feasibility of non-invasive therapeutic monitoring using MRMI of EDB-FN was also evaluated in drug-resistant DLD1-DR tumors treated with a pan-AKT inhibitor MK2206-HCl. The treated drug-resistant tumors failed to respond to therapy, which was accurately detected by MRMI with MT218, demonstrating higher signal enhancement and increased CNRs in the 4-week follow-up scans over the pre-treatment scans. Conclusions: EDB-FN is a promising molecular marker for assessing drug resistance. MRMI of EDB-FN with MT218 at a significantly reduced dose can facilitate effective non-invasive assessment and treatment response monitoring of drug-resistant CRC, highlighting its translational potential for active surveillance and management of CRC and other malignancies.
Collapse
Affiliation(s)
- Amita Vaidya
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Nadia Ayat
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Megan Buford
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Helen Wang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Aman Shankardass
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Yiqing Zhao
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Hannah Gilmore
- Department of Pathology, University Hospitals of Cleveland, Cleveland, OH 44106, USA
| | - Zhenghe Wang
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Zheng-Rong Lu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
21
|
Li H, Zeng Y, Zhang H, Gu Z, Gong Q, Luo K. Functional gadolinium-based nanoscale systems for cancer theranostics. J Control Release 2020; 329:482-512. [PMID: 32898594 DOI: 10.1016/j.jconrel.2020.08.064] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/25/2020] [Accepted: 08/30/2020] [Indexed: 02/07/2023]
Abstract
Cancer theranostics is a new strategy for combating cancer that integrates cancer imaging and treatment through theranostic agents to provide an efficient and safe way to improve cancer prognosis. Design and synthesis of these cancer theranostic agents are crucial since these agents are required to be biocompatible, tumor-specific, imaging distinguishable and therapeutically efficacious. In this regard, several types of gadolinium (Gd)-based nanomaterials have been introduced to combine different therapeutic agents with Gd to enhance the efficacy of therapeutic agents. At the same time, the entire treatment procedure could be monitored via imaging tools due to incorporation of Gd ions, Gd chelates and Gd/other imaging probes in the theranostic agents. This review aims to overview recent advances in the Gd-based nanomaterials for cancer theranostics and perspectives for Gd nanomaterial-based cancer theranostics are provided.
Collapse
Affiliation(s)
- Haonan Li
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yujun Zeng
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, Claremont, CA 91711, USA
| | - Zhongwei Gu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
22
|
Vaidya A, Wang H, Qian V, Gilmore H, Lu ZR. Overexpression of Extradomain-B Fibronectin is Associated with Invasion of Breast Cancer Cells. Cells 2020; 9:cells9081826. [PMID: 32756405 PMCID: PMC7463489 DOI: 10.3390/cells9081826] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 12/18/2022] Open
Abstract
Breast tumor heterogeneity is a major impediment to oncotherapy. Cancer cells undergo rapid clonal evolution, thereby acquiring significant growth and invasive advantages. The absence of specific markers of these high-risk populations precludes efficient therapeutic and diagnostic management of the disease. Given the critical function of tumor microenvironment in the oncogenic circuitry, we sought to determine the expression profile of the extracellular matrix oncoprotein, extradomain-B fibronectin (EDB-FN) in invasive breast cancer. Analyses of TCGA/GTEx databases and immunostaining of clinical samples found a significant overexpression of EDB-FN in breast tumors, which correlated with poor overall survival. Significant upregulation of EDB-FN was observed in invasive cell populations generated from relatively less invasive MCF7 and MDA-MB-468 cells by long-term TGF-β treatment and acquired chemoresistance. Treatment of the invasive cell populations with an AKT inhibitor (MK2206-HCl) reduced their invasive potential, with a concomitant decrease in their EDB-FN expression, partly through the phosphoAKT-SRp55 pathway. EDB-FN downregulation, with direct RNAi of EDB-FN or indirectly through RNAi of SRp55, also resulted in reduced motility of the invasive cell populations, validating the correlation between EDB-FN expression and invasion of breast cancer cells. These data establish EDB-FN as a promising molecular marker for non-invasive therapeutic surveillance of aggressive breast cancer.
Collapse
Affiliation(s)
- Amita Vaidya
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; (A.V.); (H.W.); (V.Q.)
| | - Helen Wang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; (A.V.); (H.W.); (V.Q.)
| | - Victoria Qian
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; (A.V.); (H.W.); (V.Q.)
| | - Hannah Gilmore
- Department of Pathology, University Hospitals of Cleveland, Cleveland, OH 44106, USA;
| | - Zheng-Rong Lu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; (A.V.); (H.W.); (V.Q.)
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Correspondence: ; Tel.: +1-216-368-0187
| |
Collapse
|
23
|
Chen Z, Krishnamachary B, Pachecho-Torres J, Penet MF, Bhujwalla ZM. Theranostic small interfering RNA nanoparticles in cancer precision nanomedicine. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 12:e1595. [PMID: 31642207 DOI: 10.1002/wnan.1595] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 09/10/2019] [Accepted: 09/11/2019] [Indexed: 12/24/2022]
Abstract
Due to their ability to effectively downregulate the expression of target genes, small interfering RNA (siRNA) have emerged as promising candidates for precision medicine in cancer. Although some siRNA-based treatments have advanced to clinical trials, challenges such as poor stability during circulation, and less than optimal pharmacokinetics and biodistribution of siRNA in vivo present barriers to the systemic delivery of siRNA. In recent years, theranostic nanomedicine integrating siRNA delivery has attracted significant attention for precision medicine. Theranostic nanomedicine takes advantage of the high capacity of nanoplatforms to ferry cargo with imaging and therapeutic capabilities. These theranostic nanoplatforms have the potential to play a major role in gene specific treatments. Here we have reviewed recent advances in the use of theranostic nanoplatforms to deliver siRNA, and discussed the opportunities as well as challenges associated with this exciting technology. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Implantable Materials and Surgical Technologies > Nanomaterials and Implants.
Collapse
Affiliation(s)
- Zhihang Chen
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Balaji Krishnamachary
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jesus Pachecho-Torres
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Marie-France Penet
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Zaver M Bhujwalla
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|