1
|
Wen X, Zhang C, Tian Y, Miao Y, Liu S, Xu JJ, Ye D, He J. Smart Molecular Imaging and Theranostic Probes by Enzymatic Molecular In Situ Self-Assembly. JACS AU 2024; 4:2426-2450. [PMID: 39055152 PMCID: PMC11267545 DOI: 10.1021/jacsau.4c00392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/15/2024] [Accepted: 06/18/2024] [Indexed: 07/27/2024]
Abstract
Enzymatic molecular in situ self-assembly (E-MISA) that enables the synthesis of high-order nanostructures from synthetic small molecules inside a living subject has emerged as a promising strategy for molecular imaging and theranostics. This strategy leverages the catalytic activity of an enzyme to trigger probe substrate conversion and assembly in situ, permitting prolonging retention and congregating many molecules of probes in the targeted cells or tissues. Enhanced imaging signals or therapeutic functions can be achieved by responding to a specific enzyme. This E-MISA strategy has been successfully applied for the development of enzyme-activated smart molecular imaging or theranostic probes for in vivo applications. In this Perspective, we discuss the general principle of controlling in situ self-assembly of synthetic small molecules by an enzyme and then discuss the applications for the construction of "smart" imaging and theranostic probes against cancers and bacteria. Finally, we discuss the current challenges and perspectives in utilizing the E-MISA strategy for disease diagnoses and therapies, particularly for clinical translation.
Collapse
Affiliation(s)
- Xidan Wen
- Department
of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital
of Medical School, Nanjing University, Nanjing 210008, China
- State
Key Laboratory of Analytical Chemistry for Life Science, Chemistry
and Biomedicine Innovation Center (ChemBIC), School of Chemistry and
Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Chao Zhang
- Department
of Neurosurgery, Zhujiang Hospital, Southern
Medical University, Guangzhou 510282, China
| | - Yuyang Tian
- State
Key Laboratory of Analytical Chemistry for Life Science, Chemistry
and Biomedicine Innovation Center (ChemBIC), School of Chemistry and
Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Yinxing Miao
- State
Key Laboratory of Analytical Chemistry for Life Science, Chemistry
and Biomedicine Innovation Center (ChemBIC), School of Chemistry and
Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Shaohai Liu
- State
Key Laboratory of Analytical Chemistry for Life Science, Chemistry
and Biomedicine Innovation Center (ChemBIC), School of Chemistry and
Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Jing-Juan Xu
- State
Key Laboratory of Analytical Chemistry for Life Science, Chemistry
and Biomedicine Innovation Center (ChemBIC), School of Chemistry and
Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Deju Ye
- State
Key Laboratory of Analytical Chemistry for Life Science, Chemistry
and Biomedicine Innovation Center (ChemBIC), School of Chemistry and
Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Jian He
- Department
of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital
of Medical School, Nanjing University, Nanjing 210008, China
| |
Collapse
|
2
|
Ma X, Mao M, He J, Liang C, Xie HY. Nanoprobe-based molecular imaging for tumor stratification. Chem Soc Rev 2023; 52:6447-6496. [PMID: 37615588 DOI: 10.1039/d3cs00063j] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
The responses of patients to tumor therapies vary due to tumor heterogeneity. Tumor stratification has been attracting increasing attention for accurately distinguishing between responders to treatment and non-responders. Nanoprobes with unique physical and chemical properties have great potential for patient stratification. This review begins by describing the features and design principles of nanoprobes that can visualize specific cell types and biomarkers and release inflammatory factors during or before tumor treatment. Then, we focus on the recent advancements in using nanoprobes to stratify various therapeutic modalities, including chemotherapy, radiotherapy (RT), photothermal therapy (PTT), photodynamic therapy (PDT), chemodynamic therapy (CDT), ferroptosis, and immunotherapy. The main challenges and perspectives of nanoprobes in cancer stratification are also discussed to facilitate probe development and clinical applications.
Collapse
Affiliation(s)
- Xianbin Ma
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Mingchuan Mao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Jiaqi He
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Chao Liang
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Hai-Yan Xie
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Chemical Biology Center, Peking University, Beijing, 100191, P. R. China.
| |
Collapse
|
3
|
Vladimirov N, Perlman O. Molecular MRI-Based Monitoring of Cancer Immunotherapy Treatment Response. Int J Mol Sci 2023; 24:3151. [PMID: 36834563 PMCID: PMC9959624 DOI: 10.3390/ijms24043151] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 01/29/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
Immunotherapy constitutes a paradigm shift in cancer treatment. Its FDA approval for several indications has yielded improved prognosis for cases where traditional therapy has shown limited efficiency. However, many patients still fail to benefit from this treatment modality, and the exact mechanisms responsible for tumor response are unknown. Noninvasive treatment monitoring is crucial for longitudinal tumor characterization and the early detection of non-responders. While various medical imaging techniques can provide a morphological picture of the lesion and its surrounding tissue, a molecular-oriented imaging approach holds the key to unraveling biological effects that occur much earlier in the immunotherapy timeline. Magnetic resonance imaging (MRI) is a highly versatile imaging modality, where the image contrast can be tailored to emphasize a particular biophysical property of interest using advanced engineering of the imaging pipeline. In this review, recent advances in molecular-MRI based cancer immunotherapy monitoring are described. Next, the presentation of the underlying physics, computational, and biological features are complemented by a critical analysis of the results obtained in preclinical and clinical studies. Finally, emerging artificial intelligence (AI)-based strategies to further distill, quantify, and interpret the image-based molecular MRI information are discussed in terms of perspectives for the future.
Collapse
Affiliation(s)
- Nikita Vladimirov
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Or Perlman
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
4
|
Chavda VP, Khadela A, Shah Y, Postwala H, Balar P, Vora L. Current status of Cancer Nanotheranostics: Emerging strategies for cancer management. Nanotheranostics 2023; 7:368-379. [PMID: 37151802 PMCID: PMC10161386 DOI: 10.7150/ntno.82263] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/20/2023] [Indexed: 05/09/2023] Open
Abstract
Cancer diagnosis and management have been a slow-evolving area in medical science. Conventional therapies have by far proved to have various limitations. Also, the concept of immunotherapy which was thought to revolutionize the management of cancer has presented its range of drawbacks. To overcome these limitations nanoparticulate-derived diagnostic and therapeutic strategies are emerging. These nanomaterials are to be explored as they serve as a prospect for cancer theranostics. Nanoparticles have a significant yet unclear role in screening as well as therapy of cancer. However, nanogels and Photodynamic therapy is one such approach to be developed in cancer theranostics. Photoactive cancer theranostics is a vivid area that might prove to help manage cancer. Also, the utilization of the quantum dots as a diagnostic tool and to selectively kill cancer cells, especially in CNS tumors. Additionally, the redox-sensitive micelles targeting the tumor microenvironment of the cancer are also an important theranostic tool. This review focuses on exploring various agents that are currently being studied or can further be studied as cancer theranostics.
Collapse
Affiliation(s)
- Vivek P Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L.M. College of Pharmacy, Ahmedabad, Gujarat 380009, India
- ✉ Corresponding author: Vivek P. Chavda, Department of Pharmaceutics and Pharmaceutical Technology, L.M. College of Pharmacy, Niangua, Ahmedabad (Gujarat)-380009. +91 7030919407; ; ORCID ID: https://orcid.org/0000-0002-7701-8597
| | - Avinash Khadela
- Department of Pharmacology, L. M. College of Pharmacy, Niangua, Ahmedabad, Gujarat 380009, India
| | - Yasha Shah
- PharmD Section, L.M. College of Pharmacy, Ahmedabad, Gujarat 380009, India
| | - Humzah Postwala
- PharmD Section, L.M. College of Pharmacy, Ahmedabad, Gujarat 380009, India
| | - Pankti Balar
- Pharmacy Section, L.M. College of Pharmacy, Ahmedabad, Gujarat 380009, India
| | - Lalit Vora
- School of Pharmacy, Queen's University Belfast, 97 Lilburn Road, BT9 7BL, U.K
| |
Collapse
|
5
|
Xie J, El Rami F, Zhou K, Simonetta F, Chen Z, Zheng X, Chen M, Balakrishnan PB, Dai SY, Murty S, Alam IS, Baker J, Negrin RS, Gambhir SS, Rao J. Multiparameter Longitudinal Imaging of Immune Cell Activity in Chimeric Antigen Receptor T Cell and Checkpoint Blockade Therapies. ACS CENTRAL SCIENCE 2022; 8:590-602. [PMID: 35647285 PMCID: PMC9136971 DOI: 10.1021/acscentsci.2c00142] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Indexed: 05/17/2023]
Abstract
Longitudinal multimodal imaging presents unique opportunities for noninvasive surveillance and prediction of treatment response to cancer immunotherapy. In this work we first designed a novel granzyme B activated self-assembly small molecule, G-SNAT, for the assessment of cytotoxic T lymphocyte mediated cancer cell killing. G-SNAT was found to specifically detect the activity of granzyme B within the cytotoxic granules of activated T cells and engaged cancer cells in vitro. In lymphoma tumor-bearing mice, the retention of cyanine 5 labeled G-SNAT-Cy5 correlated to CAR T cell mediated granzyme B exocytosis and tumor eradication. In colorectal tumor-bearing transgenic mice with hematopoietic cells expressing firefly luciferase, longitudinal bioluminescence and fluorescence imaging revealed that after combination treatment of anti-PD-1 and anti-CTLA-4, the dynamics of immune cell trafficking, tumor infiltration, and cytotoxic activity predicted the therapeutic outcome before tumor shrinkage was evident. These results support further development of G-SNAT for imaging early immune response to checkpoint blockade and CAR T-cell therapy in patients and highlight the utility of multimodality imaging for improved mechanistic insights into cancer immunotherapy.
Collapse
Affiliation(s)
- Jinghang Xie
- Department
of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Fadi El Rami
- Department
of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Kaixiang Zhou
- Department
of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Federico Simonetta
- Division
of Blood and Marrow Transplantation, Department of Medicine, Stanford University Medical Center, Stanford, California 94305, United States
| | - Zixin Chen
- Department of Chemistry, Department of Bioengineering, and Department of Materials Science
& Engineering, Stanford University, Stanford, California 94305, United States
| | - Xianchuang Zheng
- Department
of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Min Chen
- Department
of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Preethi B. Balakrishnan
- Department
of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Sheng-Yao Dai
- Department
of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Surya Murty
- Department
of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
- Department of Chemistry, Department of Bioengineering, and Department of Materials Science
& Engineering, Stanford University, Stanford, California 94305, United States
| | - Israt S. Alam
- Department
of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Jeanette Baker
- Division
of Blood and Marrow Transplantation, Department of Medicine, Stanford University Medical Center, Stanford, California 94305, United States
| | - Robert S. Negrin
- Division
of Blood and Marrow Transplantation, Department of Medicine, Stanford University Medical Center, Stanford, California 94305, United States
| | - Sanjiv S. Gambhir
- Department
of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
- Department of Chemistry, Department of Bioengineering, and Department of Materials Science
& Engineering, Stanford University, Stanford, California 94305, United States
| | - Jianghong Rao
- Department
of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
- Department of Chemistry, Department of Bioengineering, and Department of Materials Science
& Engineering, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
6
|
Fang J, Zhao Y, Wang A, Zhang Y, Cui C, Ye S, Mao Q, Feng Y, Li J, Xu C, Shi H. In Vivo Quantitative Assessment of a Radiation Dose Based on Ratiometric Photoacoustic Imaging of Tumor Apoptosis. Anal Chem 2022; 94:5149-5158. [PMID: 35311264 DOI: 10.1021/acs.analchem.2c00098] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Accurately assessing the radiation level of tumors and surrounding tissues is of great significance for the optimization of clinical therapeutic interventions as well as minimizing the radiation-induced side effects. Therefore, the development of noninvasive and sensitive biological dosimeters is vital to achieve quantitative detection of a radiation dose in a living system. Herein, as a proof of concept, we report a tumor-targeted and caspase-3-activatable NIR fluorogenic probe AcDEVD-Cy-RGD consisting of a hemicyanine fluorophore as a signal reporter, a caspase-3 specific Asp-Glu-Val-Asp (DEVD) peptide, and a cyclic Arg-Gly-Asp peptide (cRGD) for tumor targeting. Upon cleavage with activated caspase-3, this probe not only displays the lighted-up NIR fluorescence, but also ratiometric photoacoustic (PA710/PA680) signals concurrently in a caspase-3 concentration-dependent manner, allowing for sensitive and quantitative detection of caspase-3 activity through both fluorescence and PA imaging, which provides the possibility for real-time monitoring of tumor cell apoptosis in a living system. More notably, we utilized this probe to successfully realize the direct visualization of tumor response to chemo- or radiotherapy and, for the first time, achieve the accurate estimation of radiation doses imparted to the tumors. We thus believe that our current strategy would offer an attractive and valuable means for the precise assessment of locally delivered radiation doses in various clinical settings.
Collapse
Affiliation(s)
- Jing Fang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| | - Yan Zhao
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| | - Anna Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| | - Yuqi Zhang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| | - Chaoxiang Cui
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| | - Shuyue Ye
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| | - Qiulian Mao
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| | - Yali Feng
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| | - Jiachen Li
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| | - Chenjie Xu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
| | - Haibin Shi
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| |
Collapse
|
7
|
Guo W, Chen Z, Tan L, Gu D, Ren X, Fu C, Wu Q, Meng X. Emerging biocompatible nanoplatforms for the potential application in diagnosis and therapy of deep tumors. VIEW 2022. [DOI: 10.1002/viw.20200174] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Wenna Guo
- Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry Chinese Academy of Sciences Beijing P.R. China
- School of Optoelectronic Science and Engineering University of Electronic Science and Technology of China Chengdu Sichuan P.R. China
- CAS Key Laboratory of Cryogenics Technical Institute of Physics and Chemistry Beijing P.R. China
| | - Zengzhen Chen
- Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry Chinese Academy of Sciences Beijing P.R. China
- CAS Key Laboratory of Cryogenics Technical Institute of Physics and Chemistry Beijing P.R. China
- University of Chinese Academy of Sciences Beijing P.R. China
| | - Longfei Tan
- Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry Chinese Academy of Sciences Beijing P.R. China
- CAS Key Laboratory of Cryogenics Technical Institute of Physics and Chemistry Beijing P.R. China
| | - Deen Gu
- School of Optoelectronic Science and Engineering University of Electronic Science and Technology of China Chengdu Sichuan P.R. China
| | - Xiangling Ren
- Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry Chinese Academy of Sciences Beijing P.R. China
- CAS Key Laboratory of Cryogenics Technical Institute of Physics and Chemistry Beijing P.R. China
| | - Changhui Fu
- Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry Chinese Academy of Sciences Beijing P.R. China
- CAS Key Laboratory of Cryogenics Technical Institute of Physics and Chemistry Beijing P.R. China
| | - Qiong Wu
- Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry Chinese Academy of Sciences Beijing P.R. China
- CAS Key Laboratory of Cryogenics Technical Institute of Physics and Chemistry Beijing P.R. China
| | - Xianwei Meng
- Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry Chinese Academy of Sciences Beijing P.R. China
- CAS Key Laboratory of Cryogenics Technical Institute of Physics and Chemistry Beijing P.R. China
| |
Collapse
|
8
|
Dou Y, Zhao F, Li X, Guo Y. Monitoring Nitric Oxide-Induced Hypoxic Tumor Radiosensitization by Radiation-Activated Nanoagents under BOLD/DWI Imaging. ACS Biomater Sci Eng 2021; 7:5242-5254. [PMID: 34612040 DOI: 10.1021/acsbiomaterials.1c00543] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Tumor heterogeneity leads to unpredictable radiotherapeutic outcomes although multiple sensitization strategies have been developed. Real-time monitoring of treatment response through noninvasive imaging methods is critical and a great challenge in optimizing radiotherapy. Herein, we propose a combined functional magnetic resonance imaging approach (blood-oxygen-level-dependent/diffusion-weighted (BOLD/DWI) imaging) for monitoring tumor response to nitric oxide (NO)-induced hypoxic radiosensitization achieved by radiation-activated nanoagents (NSC@SiO2-SNO NPs). This nanoagent carrying NO donors can efficiently concentrate in tumors and specifically produce high concentrations of NO under radiation. In vitro and in vivo studies show that this nanoagent can effectively reduce tumor hypoxia, promote radiation-induced apoptosis and DNA damage under hypoxia, and ultimately inhibit tumor growth. In vivo BOLD/DWI imaging enables noninvasive monitoring of improvements in tumor oxygen levels and radiosensitivity during treatment with this nanostrategy by quantifying functional parameters. This work demonstrates that BOLD/DWI imaging is a useful tool for evaluating tumor response and monitoring the effectiveness of radiotherapeutic strategies aimed at improving hypoxia, with great clinical potential.
Collapse
Affiliation(s)
- Yan Dou
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Fangshi Zhao
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Xue Li
- Department of Radiology and Department of Radiation Oncology, Tianjin Medical University Second Hospital, Tianjin 300211, P. R. China
| | - Yanyan Guo
- Department of Radiology and Department of Radiation Oncology, Tianjin Medical University Second Hospital, Tianjin 300211, P. R. China
| |
Collapse
|
9
|
Xie J, Rice MA, Chen Z, Cheng Y, Hsu EC, Chen M, Song G, Cui L, Zhou K, Castillo JB, Zhang CA, Shen B, Chin FT, Kunder CA, Brooks JD, Stoyanova T, Rao J. In Vivo Imaging of Methionine Aminopeptidase II for Prostate Cancer Risk Stratification. Cancer Res 2021; 81:2510-2521. [PMID: 33637565 DOI: 10.1158/0008-5472.can-20-2969] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/31/2020] [Accepted: 02/24/2021] [Indexed: 11/16/2022]
Abstract
Prostate cancer is one of the most common malignancies worldwide, yet limited tools exist for prognostic risk stratification of the disease. Identification of new biomarkers representing intrinsic features of malignant transformation and development of prognostic imaging technologies are critical for improving treatment decisions and patient survival. In this study, we analyzed radical prostatectomy specimens from 422 patients with localized disease to define the expression pattern of methionine aminopeptidase II (MetAP2), a cytosolic metalloprotease that has been identified as a druggable target in cancer. MetAP2 was highly expressed in 54% of low-grade and 59% of high-grade cancers. Elevated levels of MetAP2 at diagnosis were associated with shorter time to recurrence. Controlled self-assembly of a synthetic small molecule enabled design of the first MetAP2-activated PET imaging tracer for monitoring MetAP2 activity in vivo. The nanoparticles assembled upon MetAP2 activation were imaged in single prostate cancer cells with post-click fluorescence labeling. The fluorine-18-labeled tracers successfully differentiated MetAP2 activity in both MetAP2-knockdown and inhibitor-treated human prostate cancer xenografts by micro-PET/CT scanning. This highly sensitive imaging technology may provide a new tool for noninvasive early-risk stratification of prostate cancer and monitoring the therapeutic effect of MetAP2 inhibitors as anticancer drugs. SIGNIFICANCE: This study defines MetAP2 as an early-risk stratifier for molecular imaging of aggressive prostate cancer and describes a MetAP2-activated self-assembly small-molecule PET tracer for imaging MetAP2 activity in vivo.
Collapse
Affiliation(s)
- Jinghang Xie
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Meghan A Rice
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, California
| | - Zixin Chen
- Department of Chemistry, Stanford University, Stanford, California
| | - Yunfeng Cheng
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - En-Chi Hsu
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, California
| | - Min Chen
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Guosheng Song
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Liyang Cui
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Kaixiang Zhou
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Jessa B Castillo
- Department of Radiology, Cyclotron and Radiochemistry Facility, Stanford University School of Medicine, Stanford, California
| | - Chiyuan A Zhang
- Department of Urology, Stanford University School of Medicine, Stanford, California
| | - Bin Shen
- Department of Radiology, Cyclotron and Radiochemistry Facility, Stanford University School of Medicine, Stanford, California
| | - Frederick T Chin
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California.,Department of Radiology, Cyclotron and Radiochemistry Facility, Stanford University School of Medicine, Stanford, California
| | - Christian A Kunder
- Department of Urology, Stanford University School of Medicine, Stanford, California
| | - James D Brooks
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, California.,Department of Urology, Stanford University School of Medicine, Stanford, California
| | - Tanya Stoyanova
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, California.
| | - Jianghong Rao
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California. .,Department of Chemistry, Stanford University, Stanford, California
| |
Collapse
|
10
|
Magnetic Resonance Imaging Agents. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00037-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
11
|
Untiring Pursuit for Glucarate-Based Molecular Imaging Probes. Mol Imaging Biol 2020; 23:310-322. [PMID: 33206335 DOI: 10.1007/s11307-020-01564-y] [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: 08/11/2020] [Revised: 10/08/2020] [Accepted: 11/05/2020] [Indexed: 10/23/2022]
Abstract
Glucarate, a physiologic end-product of the D-glucuronic acid pathway in mammals, is a six-carbon dicarboxylic acid with a wide range of uses. Glucarate-based molecular imaging probes including [99mTc]glucarate and [18F]glucarate have been developed and demonstrated to have infarct/necrosis-avid and/or tumor-seeking properties, showing potential applications in early detection of myocardial infarction, evaluation of tissue viability, monitoring of therapeutic effectiveness, and noninvasive imaging of certain tumors including drug-resistant ones. The mechanism by which [99mTc]glucarate localizes in acute necrotic tissues has been demonstrated to be largely attributable to its binding to the positively charged histones, which become accessible after the disruption of the cell and nuclear membranes as a result of irreversible damage, while the tumor-seeking mechanism of [99mTc]glucarate has been found to be closely related to glucose transporter 5 expression. Moreover, the recently developed [18F]glucarate provides a new alternative probe for positron emission tomography imaging and may have potential advantages over [99mTc]glucarate. In this review, we present the untiring pursuit for glucarate-based molecular imaging probes as infarct/necrosis-avid agent and/or tumor-seeking agent. Moreover, the limitations and the prospects for future research of glucarate-based molecular probes are also discussed.
Collapse
|
12
|
Bayesian Information-Theoretic Calibration of Radiotherapy Sensitivity Parameters for Informing Effective Scanning Protocols in Cancer. J Clin Med 2020; 9:jcm9103208. [PMID: 33027933 PMCID: PMC7601810 DOI: 10.3390/jcm9103208] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/01/2020] [Accepted: 10/03/2020] [Indexed: 12/03/2022] Open
Abstract
With new advancements in technology, it is now possible to collect data for a variety of different metrics describing tumor growth, including tumor volume, composition, and vascularity, among others. For any proposed model of tumor growth and treatment, we observe large variability among individual patients’ parameter values, particularly those relating to treatment response; thus, exploiting the use of these various metrics for model calibration can be helpful to infer such patient-specific parameters both accurately and early, so that treatment protocols can be adjusted mid-course for maximum efficacy. However, taking measurements can be costly and invasive, limiting clinicians to a sparse collection schedule. As such, the determination of optimal times and metrics for which to collect data in order to best inform proper treatment protocols could be of great assistance to clinicians. In this investigation, we employ a Bayesian information-theoretic calibration protocol for experimental design in order to identify the optimal times at which to collect data for informing treatment parameters. Within this procedure, data collection times are chosen sequentially to maximize the reduction in parameter uncertainty with each added measurement, ensuring that a budget of n high-fidelity experimental measurements results in maximum information gain about the low-fidelity model parameter values. In addition to investigating the optimal temporal pattern for data collection, we also develop a framework for deciding which metrics should be utilized at each data collection point. We illustrate this framework with a variety of toy examples, each utilizing a radiotherapy treatment regimen. For each scenario, we analyze the dependence of the predictive power of the low-fidelity model upon the measurement budget.
Collapse
|
13
|
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: 15] [Impact Index Per Article: 3.0] [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
|
14
|
Wang Y, Du W, Zhang T, Zhu Y, Ni Y, Wang C, Sierra Raya FM, Zou L, Wang L, Liang G. A Self-Evaluating Photothermal Therapeutic Nanoparticle. ACS NANO 2020; 14:9585-9593. [PMID: 32806081 DOI: 10.1021/acsnano.9b10144] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Today, tumor therapy and its therapeutic efficiency evaluation are conducted separately, and current imaging techniques cannot evaluate tumor-therapeutic effects in real time. Therefore, it is of great importance to develop highly efficient theranostic strategies which are able to evaluate their tumor-therapeutic effects in real time. In this work, by rational design of a small molecular near-infrared probe Cys(StBu)-Asp-Glu-Val-Asp-Lys(Cypate)-CBT (Cy-CBT) and using a CBT-Cys click condensation reaction, we facilely prepare an intelligent nanoparticle Cy-CBT-NP which is able to evaluate its photothermal therapy (PTT) efficiency on tumors by fluorescence "Turn-On". Fluorescence of Cy-CBT-NP is quenched and photothermal responsive. Upon caspase 3 (Casp3) cleavage of its DEVD substrates, Cy-CBT-NP disassembles to turn the fluorescence "On", which in turn evaluates the PTT efficiency of the nanoparticle on cells and tumors in real time. We envision that our smart strategy could be applied for PTT and real-time evaluation of the therapeutic efficiency of solid tumors in the near future.
Collapse
Affiliation(s)
- Yanfang Wang
- Hefei National Laboratory of Physical Sciences at Microscale, Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China
| | - Wei Du
- Hefei National Laboratory of Physical Sciences at Microscale, Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China
| | - Tong Zhang
- Hefei National Laboratory of Physical Sciences at Microscale, Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China
| | - Yu Zhu
- Hefei National Laboratory of Physical Sciences at Microscale, Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China
| | - Yanhan Ni
- Hefei National Laboratory of Physical Sciences at Microscale, Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China
| | - Chenchen Wang
- Hefei National Laboratory of Physical Sciences at Microscale, Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China
| | - Fatima Maria Sierra Raya
- Hefei National Laboratory of Physical Sciences at Microscale, Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China
| | - Liwei Zou
- Department of Radiology, the Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui 230601, China
| | - Longsheng Wang
- Department of Radiology, the Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui 230601, China
| | - Gaolin Liang
- Hefei National Laboratory of Physical Sciences at Microscale, Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing, Jiangsu 210096, China
| |
Collapse
|
15
|
Wang Y, Weng J, Wen X, Hu Y, Ye D. Recent advances in stimuli-responsive in situ self-assembly of small molecule probes for in vivo imaging of enzymatic activity. Biomater Sci 2020; 9:406-421. [PMID: 32627767 DOI: 10.1039/d0bm00895h] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Stimuli-responsive in situ self-assembly of small molecule probes into nanostructures has been promising for the construction of molecular probes for in vivo imaging. In the past few years, a number of intelligent molecular imaging probes with fluorescence, magnetic resonance imaging (MRI), positron electron tomography (PET) or photoacoustic imaging (PA) modality have been developed based on the in situ self-assembly strategy. In this minireview, we summarize the recent advances in the development of different modality imaging probes through controlling in situ self-assembly for in vivo imaging of enzymatic activity. This review starts from the brief introduction of two different chemical approaches amenable for in situ self-assembly, including (1) stimuli-mediated proteolysis and (2) stimuli-triggered biocompatible reaction. We then discuss their applications in the design of fluorescence, MRI, PET, PA, and bimodality imaging probes for in vivo imaging of different enzymes, such as caspase-3, furin, gelatinase and phosphatase. Finally, we discuss the current and prospective challenges in the stimuli-responsive in situ self-assembly strategy for in vivo imaging.
Collapse
Affiliation(s)
- Yuqi Wang
- State Key Laboratory of Analytical Chemistry for Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | | | | | | | | |
Collapse
|
16
|
Zhou Z, Deng H, Yang W, Wang Z, Lin L, Munasinghe J, Jacobson O, Liu Y, Tang L, Ni Q, Kang F, Liu Y, Niu G, Bai R, Qian C, Song J, Chen X. Early stratification of radiotherapy response by activatable inflammation magnetic resonance imaging. Nat Commun 2020; 11:3032. [PMID: 32541769 PMCID: PMC7295999 DOI: 10.1038/s41467-020-16771-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 05/14/2020] [Indexed: 12/14/2022] Open
Abstract
Tumor heterogeneity is one major reason for unpredictable therapeutic outcomes, while stratifying therapeutic responses at an early time may greatly benefit the better control of cancer. Here, we developed a hybrid nanovesicle to stratify radiotherapy response by activatable inflammation magnetic resonance imaging (aiMRI) approach. The high Pearson's correlation coefficient R values are obtained from the correlations between the T1 relaxation time changes at 24-48 h and the ensuing adaptive immunity (R = 0.9831) at day 5 and the tumor inhibition ratios (R = 0.9308) at day 18 after different treatments, respectively. These results underscore the role of acute inflammatory oxidative response in bridging the innate and adaptive immunity in tumor radiotherapy. Furthermore, the aiMRI approach provides a non-invasive imaging strategy for early prediction of the therapeutic outcomes in cancer radiotherapy, which may contribute to the future of precision medicine in terms of prognostic stratification and therapeutic planning.
Collapse
Affiliation(s)
- Zijian Zhou
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Hongzhang Deng
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350116, Fujian, China
| | - Weijing Yang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Zhantong Wang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lisen Lin
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350116, Fujian, China
| | - Jeeva Munasinghe
- Laboratory of Functional and Molecular Imaging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Orit Jacobson
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yijing Liu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Longguang Tang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Qianqian Ni
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Fei Kang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yuan Liu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Gang Niu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ruiliang Bai
- Interdisciplinary Institute of Neuroscience and Technology, School of Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Chunqi Qian
- Department of Radiology, Michigan State University, East Lansing, MI, 48824, USA
| | - Jibin Song
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350116, Fujian, China.
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
17
|
Soleymani J, Hasanzadeh M, shadjou N, Somi MH, Jouyban A. The role of nanomaterials on the cancer cells sensing based on folate receptor: Analytical approach. Trends Analyt Chem 2020. [DOI: 10.1016/j.trac.2020.115834] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
18
|
Zhang D, Jin Q, Jiang C, Gao M, Ni Y, Zhang J. Imaging Cell Death: Focus on Early Evaluation of Tumor Response to Therapy. Bioconjug Chem 2020; 31:1025-1051. [PMID: 32150392 DOI: 10.1021/acs.bioconjchem.0c00119] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cell death plays a prominent role in the treatment of cancer, because most anticancer therapies act by the induction of cell death including apoptosis, necrosis, and other pathways of cell death. Imaging cell death helps to identify treatment responders from nonresponders and thus enables patient-tailored therapy, which will increase the likelihood of treatment response and ultimately lead to improved patient survival. By taking advantage of molecular probes that specifically target the biomarkers/biochemical processes of cell death, cell death imaging can be successfully achieved. In recent years, with the increased understanding of the molecular mechanism of cell death, a variety of well-defined biomarkers/biochemical processes of cell death have been identified. By targeting these established cell death biomarkers/biochemical processes, a set of molecular imaging probes have been developed and evaluated for early monitoring treatment response in tumors. In this review, we mainly present the recent advances in identifying useful biomarkers/biochemical processes for both apoptosis and necrosis imaging and in developing molecular imaging probes targeting these biomarkers/biochemical processes, with a focus on their application in early evaluation of tumor response to therapy.
Collapse
Affiliation(s)
- Dongjian Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, P.R. China.,Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, P.R. China
| | - Qiaomei Jin
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, P.R. China.,Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, P.R. China
| | - Cuihua Jiang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, P.R. China.,Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, P.R. China
| | - Meng Gao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, P.R. China.,Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, P.R. China
| | - Yicheng Ni
- Theragnostic Laboratory, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| | - Jian Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, P.R. China.,Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, P.R. China
| |
Collapse
|
19
|
Chen Z, Chen M, Zhou K, Rao J. Pre‐targeted Imaging of Protease Activity through In Situ Assembly of Nanoparticles. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201916352] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Zixin Chen
- Departments of Radiology and Chemistry Molecular Imaging Program at Stanford Stanford University School of Medicine Stanford CA 94305 USA
| | - Min Chen
- Departments of Radiology and Chemistry Molecular Imaging Program at Stanford Stanford University School of Medicine Stanford CA 94305 USA
| | - Kaixiang Zhou
- Departments of Radiology and Chemistry Molecular Imaging Program at Stanford Stanford University School of Medicine Stanford CA 94305 USA
| | - Jianghong Rao
- Departments of Radiology and Chemistry Molecular Imaging Program at Stanford Stanford University School of Medicine Stanford CA 94305 USA
| |
Collapse
|
20
|
Chen Z, Chen M, Zhou K, Rao J. Pre-targeted Imaging of Protease Activity through In Situ Assembly of Nanoparticles. Angew Chem Int Ed Engl 2020; 59:7864-7870. [PMID: 32056345 DOI: 10.1002/anie.201916352] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/11/2020] [Indexed: 02/06/2023]
Abstract
The pre-targeted imaging of enzyme activity has not been reported, likely owing to the lack of a mechanism to retain the injected substrate in the first step for subsequent labeling. Herein, we report the use of two bioorthogonal reactions-the condensation reaction of aromatic nitriles and aminothiols and the inverse-electron demand Diels-Alder reaction between tetrazine and trans-cyclooctene (TCO)-to develop a novel strategy for pre-targeted imaging of the activity of proteases. The substrate probe (TCO-C-SNAT4) can be selectively activated by an enzyme target (e.g. caspase-3/7), which triggers macrocyclization and subsequent in situ self-assembly into nanoaggregates retained at the target site. The tetrazine-imaging tag conjugate labels TCO in the nanoaggregates to generate selective signal retention for imaging in vitro, in cells, and in mice. Owing to the decoupling of enzyme activation and imaging tag immobilization, TCO-C-SNAT4 can be repeatedly injected to generate and accumulate more TCO-nanoaggregates for click labeling.
Collapse
Affiliation(s)
- Zixin Chen
- Departments of Radiology and Chemistry, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Min Chen
- Departments of Radiology and Chemistry, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Kaixiang Zhou
- Departments of Radiology and Chemistry, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jianghong Rao
- Departments of Radiology and Chemistry, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
21
|
Chen Z, Chen M, Cheng Y, Kowada T, Xie J, Zheng X, Rao J. Exploring the Condensation Reaction between Aromatic Nitriles and Amino Thiols To Optimize In Situ Nanoparticle Formation for the Imaging of Proteases and Glycosidases in Cells. Angew Chem Int Ed Engl 2020; 59:3272-3279. [PMID: 31828913 DOI: 10.1002/anie.201913314] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Indexed: 12/31/2022]
Abstract
The condensation reaction between 6-hydroxy-2-cyanobenzothiazole (CBT) and cysteine has been shown for various applications such as site-specific protein labelling and in vivo cancer imaging. This report further expands the substrate scope of this reaction by varying the substituents on aromatic nitriles and amino thiols and testing their reactivity and ability to form nanoparticles for cell imaging. The structure-activity relationship study leads to the identification of the minimum structural requirement for the macrocyclization and assembly process in forming nanoparticles. One of the scaffolds made of 2-pyrimidinecarbonitrile and cysteine joined by a benzyl linker was applied to design fluorescent probes for imaging caspase-3/7 and β-galactosidase activity in live cells. These results demonstrate the generality of this system for imaging hydrolytic enzymes.
Collapse
Affiliation(s)
- Zixin Chen
- Departments of Radiology and Chemistry, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Min Chen
- Departments of Radiology and Chemistry, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Yunfeng Cheng
- Departments of Radiology and Chemistry, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Toshiyuki Kowada
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi, 980-8577, Japan
| | - Jinghang Xie
- Departments of Radiology and Chemistry, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Xianchuang Zheng
- Departments of Radiology and Chemistry, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jianghong Rao
- Departments of Radiology and Chemistry, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
22
|
Chen Z, Chen M, Cheng Y, Kowada T, Xie J, Zheng X, Rao J. Exploring the Condensation Reaction between Aromatic Nitriles and Amino Thiols To Optimize In Situ Nanoparticle Formation for the Imaging of Proteases and Glycosidases in Cells. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201913314] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Zixin Chen
- Departments of Radiology and Chemistry Molecular Imaging Program at Stanford Stanford University School of Medicine Stanford CA 94305 USA
| | - Min Chen
- Departments of Radiology and Chemistry Molecular Imaging Program at Stanford Stanford University School of Medicine Stanford CA 94305 USA
| | - Yunfeng Cheng
- Departments of Radiology and Chemistry Molecular Imaging Program at Stanford Stanford University School of Medicine Stanford CA 94305 USA
| | - Toshiyuki Kowada
- Institute of Multidisciplinary Research for Advanced Materials Tohoku University 2-1-1 Katahira, Aoba-ku Sendai Miyagi 980-8577 Japan
| | - Jinghang Xie
- Departments of Radiology and Chemistry Molecular Imaging Program at Stanford Stanford University School of Medicine Stanford CA 94305 USA
| | - Xianchuang Zheng
- Departments of Radiology and Chemistry Molecular Imaging Program at Stanford Stanford University School of Medicine Stanford CA 94305 USA
| | - Jianghong Rao
- Departments of Radiology and Chemistry Molecular Imaging Program at Stanford Stanford University School of Medicine Stanford CA 94305 USA
| |
Collapse
|
23
|
Li H, Meade TJ. Molecular Magnetic Resonance Imaging with Gd(III)-Based Contrast Agents: Challenges and Key Advances. J Am Chem Soc 2019; 141:17025-17041. [PMID: 31593630 DOI: 10.1021/jacs.9b09149] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In an era of personalized medicine, the clinical community has become increasingly focused on understanding diseases at the cellular and molecular levels. Magnetic resonance imaging (MRI) is a powerful imaging modality for acquiring anatomical and functional information. However, it has limited applications in the field of molecular imaging due to its low sensitivity. To expand the capability of MRI to encompass molecular imaging applications, we introduced bioresponsive Gd(III)-based magnetic resonance contrast agents (GBCAs) in 1997. Since that time, many research groups across the globe have developed new examples of bioresponsive GBCAs. These contrast agents have shown great promise for visualizing several biochemical processes, such as gene expression, neuronal signaling, and hormone secretion. They are designed to be conditionally retained, or activated, in vivo in response to specific biochemical events of interest. As a result, an observed MR signal change can serve as a read-out for molecular events. A significant challenge for these probes is how to utilize them for noninvasive diagnostic and theranostic applications. This Perspective focuses on the design strategies that underlie bioresponsive probes, and describes the key advances made in recent years that are facilitating their application in vivo and ultimately in clinical translation. While the field of bioresponsive agents is embryonic, it is clear that many solutions to the experimental and clinical radiologic problems of today will be overcome by the probes of tomorrow.
Collapse
Affiliation(s)
- Hao Li
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology Northwestern University , Evanston , Illinois 60208 , United States
| | - Thomas J Meade
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology Northwestern University , Evanston , Illinois 60208 , United States
| |
Collapse
|
24
|
Mirniaharikandehei S, VanOsdol J, Heidari M, Danala G, Sethuraman SN, Ranjan A, Zheng B. Developing a Quantitative Ultrasound Image Feature Analysis Scheme to Assess Tumor Treatment Efficacy Using a Mouse Model. Sci Rep 2019; 9:7293. [PMID: 31086267 PMCID: PMC6513863 DOI: 10.1038/s41598-019-43847-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 05/02/2019] [Indexed: 12/16/2022] Open
Abstract
The aim of this study is to investigate the feasibility of identifying and applying quantitative imaging features computed from ultrasound images of athymic nude mice to predict tumor response to treatment at an early stage. A computer-aided detection (CAD) scheme with a graphic user interface was developed to conduct tumor segmentation and image feature analysis. A dataset involving ultrasound images of 23 athymic nude mice bearing C26 mouse adenocarcinomas was assembled. These mice were divided into 7 treatment groups utilizing a combination of thermal and nanoparticle-controlled drug delivery. Longitudinal ultrasound images of mice were taken prior and post-treatment in day 3 and day 6. After tumor segmentation, CAD scheme computed image features and created four feature pools including features computed from (1) prior treatment images only and (2) difference between prior and post-treatment images of day 3 and day 6, respectively. To predict tumor treatment efficacy, data analysis was performed to identify top image features and an optimal feature fusion method, which have a higher correlation to tumor size increase ratio (TSIR) determined at Day 10. Using image features computed from day 3, the highest Pearson Correlation coefficients between the top two features selected from two feature pools versus TSIR were 0.373 and 0.552, respectively. Using an equally weighted fusion method of two features computed from prior and post-treatment images, the correlation coefficient increased to 0.679. Meanwhile, using image features computed from day 6, the highest correlation coefficient was 0.680. Study demonstrated the feasibility of extracting quantitative image features from the ultrasound images taken at an early treatment stage to predict tumor response to therapies.
Collapse
Affiliation(s)
| | - Joshua VanOsdol
- Center for Veterinary Health Science, Oklahoma State University, Stillwater, 74078, OK, USA
| | - Morteza Heidari
- School of Electrical and Computer Engineering, University of Oklahoma, 73019, Norman, OK, USA
| | - Gopichandh Danala
- School of Electrical and Computer Engineering, University of Oklahoma, 73019, Norman, OK, USA
| | | | - Ashish Ranjan
- Center for Veterinary Health Science, Oklahoma State University, Stillwater, 74078, OK, USA
| | - Bin Zheng
- School of Electrical and Computer Engineering, University of Oklahoma, 73019, Norman, OK, USA.
| |
Collapse
|
25
|
Teleanu DM, Chircov C, Grumezescu AM, Volceanov A, Teleanu RI. Contrast Agents Delivery: An Up-to-Date Review of Nanodiagnostics in Neuroimaging. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E542. [PMID: 30987211 PMCID: PMC6523665 DOI: 10.3390/nano9040542] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 03/28/2019] [Accepted: 03/29/2019] [Indexed: 12/14/2022]
Abstract
Neuroimaging is a highly important field of neuroscience, with direct implications for the early diagnosis and progression monitoring of brain-associated diseases. Neuroimaging techniques are categorized into structural, functional and molecular neuroimaging, each possessing advantages and disadvantages in terms of resolution, invasiveness, toxicity of contrast agents and costs. Nanotechnology-based approaches for neuroimaging mostly involve the development of nanocarriers for incorporating contrast agents or the use of nanomaterials as imaging agents. Inorganic and organic nanoparticles, liposomes, micelles, nanobodies and quantum dots are some of the most studied candidates for the delivery of contrast agents for neuroimaging. This paper focuses on describing the conventional modalities used for imaging and the applications of nanotechnology for developing novel strategies for neuroimaging. The aim is to highlight the roles of nanocarriers for enhancing and/or overcome the limitations associated with the most commonly utilized neuroimaging modalities. For future directions, several techniques that could benefit from the increased contrast induced by using imaging probes are presented.
Collapse
Affiliation(s)
- Daniel Mihai Teleanu
- Emergency University Hospital, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania.
| | - Cristina Chircov
- Faculty of Engineering in Foreign Languages, Politehnica University of Bucharest, 060042 Bucharest, Romania.
- Faculty of Applied Chemistry and Materials Science, Politehnica University of Bucharest, 011061 Bucharest, Romania.
| | - Alexandru Mihai Grumezescu
- Faculty of Applied Chemistry and Materials Science, Politehnica University of Bucharest, 011061 Bucharest, Romania.
- ICUB - Research Institute of University of Bucharest, University of Bucharest, 36-46 M. Kogalniceanu Blvd., Bucharest 050107, Romania.
| | - Adrian Volceanov
- Faculty of Applied Chemistry and Materials Science, Politehnica University of Bucharest, 011061 Bucharest, Romania.
| | - Raluca Ioana Teleanu
- "Victor Gomoiu" Clinical Children's Hospital, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania.
| |
Collapse
|
26
|
Schultz C. The Life Science Toolbox Provided by Chemical Biology. Isr J Chem 2019. [DOI: 10.1002/ijch.201900020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
27
|
Vaidya T, Agrawal A, Mahajan S, Thakur MH, Mahajan A. The Continuing Evolution of Molecular Functional Imaging in Clinical Oncology: The Road to Precision Medicine and Radiogenomics (Part II). Mol Diagn Ther 2019; 23:27-51. [PMID: 30387041 DOI: 10.1007/s40291-018-0367-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The present era of precision medicine sees "cancer" as a consequence of molecular derangements occurring at the commencement of the disease process, with morphological changes happening much later in the process of tumourigenesis. Conventional imaging techniques, such as computed tomography (CT), ultrasound (US) and magnetic resonance imaging (MRI) play an integral role in the detection of disease at the macroscopic level. However, molecular functional imaging (MFI) techniques entail the visualisation and quantification of biochemical and physiological processes occurring during tumourigenesis. MFI has the potential to play a key role in heralding the transition from the concept of "one-size-fits-all" treatment to "precision medicine". Integration of MFI with other fields of tumour biology such as genomics has spawned a novel concept called "radiogenomics", which could serve as an indispensable tool in translational cancer research. With recent advances in medical image processing, such as texture analysis, deep learning and artificial intelligence, the future seems promising; however, their clinical utility remains unproven at present. Despite the emergence of novel imaging biomarkers, the majority of these require validation before clinical translation is possible. In this two part review, we discuss the systematic collaboration across structural, anatomical and molecular imaging techniques that constitute MFI. Part I reviews positron emission tomography, radiogenomics, AI, and optical imaging, while part II reviews MRI, CT and ultrasound, their current status, and recent advances in the field of precision oncology.
Collapse
Affiliation(s)
- Tanvi Vaidya
- Department of Radiodiagnosis and Imaging, Tata Memorial Hospital, Tata Memorial Centre, Room No 125, Dr E Borges Road, Parel, Mumbai, Maharashtra, 400012, India
| | - Archi Agrawal
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Parel, Mumbai, Maharashtra, 400 012, India
| | - Shivani Mahajan
- Department of Radiodiagnosis and Imaging, Tata Memorial Hospital, Tata Memorial Centre, Room No 125, Dr E Borges Road, Parel, Mumbai, Maharashtra, 400012, India
| | - M H Thakur
- Department of Radiodiagnosis and Imaging, Tata Memorial Hospital, Tata Memorial Centre, Room No 125, Dr E Borges Road, Parel, Mumbai, Maharashtra, 400012, India
| | - Abhishek Mahajan
- Department of Radiodiagnosis and Imaging, Tata Memorial Hospital, Tata Memorial Centre, Room No 125, Dr E Borges Road, Parel, Mumbai, Maharashtra, 400012, India.
| |
Collapse
|
28
|
Wahsner J, Gale EM, Rodríguez-Rodríguez A, Caravan P. Chemistry of MRI Contrast Agents: Current Challenges and New Frontiers. Chem Rev 2019; 119:957-1057. [PMID: 30350585 PMCID: PMC6516866 DOI: 10.1021/acs.chemrev.8b00363] [Citation(s) in RCA: 953] [Impact Index Per Article: 158.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tens of millions of contrast-enhanced magnetic resonance imaging (MRI) exams are performed annually around the world. The contrast agents, which improve diagnostic accuracy, are almost exclusively small, hydrophilic gadolinium(III) based chelates. In recent years concerns have arisen surrounding the long-term safety of these compounds, and this has spurred research into alternatives. There has also been a push to develop new molecularly targeted contrast agents or agents that can sense pathological changes in the local environment. This comprehensive review describes the state of the art of clinically approved contrast agents, their mechanism of action, and factors influencing their safety. From there we describe different mechanisms of generating MR image contrast such as relaxation, chemical exchange saturation transfer, and direct detection and the types of molecules that are effective for these purposes. Next we describe efforts to make safer contrast agents either by increasing relaxivity, increasing resistance to metal ion release, or by moving to gadolinium(III)-free alternatives. Finally we survey approaches to make contrast agents more specific for pathology either by direct biochemical targeting or by the design of responsive or activatable contrast agents.
Collapse
Affiliation(s)
- Jessica Wahsner
- Athinoula A. Martinos Center for Biomedical Imaging and the Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Eric M. Gale
- Athinoula A. Martinos Center for Biomedical Imaging and the Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Aurora Rodríguez-Rodríguez
- Athinoula A. Martinos Center for Biomedical Imaging and the Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging and the Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| |
Collapse
|
29
|
Anzai Y, Minoshima S, Lee VS. Enhancing Value of MRI: A Call for Action. J Magn Reson Imaging 2018; 49:e40-e48. [PMID: 30431676 DOI: 10.1002/jmri.26239] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 06/06/2018] [Indexed: 12/19/2022] Open
Abstract
As national healthcare spending has spiraled out of control, payment reform that moves from volume to value-based payment has been introduced as a practical solution. Under alternative value-based payment models, physicians and clinical teams must deliver the best care possible at a lower cost. Medical imaging has changed the way we diagnose disease, evaluate severity, assess treatment effects, and provide biological insights for the pathophysiology of many diseases. Over the past 50 years, imaging techniques have become increasingly advanced-from X-ray to computed tomography (CT), magnetic resonance imaging (MRI), positron emission tomography (PET), and multi-modal imaging. Advanced imaging such as MRI has given clinicians remarkable insights into medical conditions and saved innumerable lives. Under the value proposition, however, we must ask if each imaging study changes treatment decisions, improves patient outcomes, and is cost-effective. Imaging research has been focused on developing new technologies and clinical applications to assess diagnostic accuracy. What is needed is the higher-level technology assessment. In this article we review why we need to demonstrate the value of MRI, how we define value, what strategies can enhance MR value through partnership with various stakeholders, and how imaging scientists can contribute to healthcare delivery in the future. Level of Evidence: 5 Technical Efficacy: Stage 3 J. Magn. Reson. Imaging 2019;49:e40-e48.
Collapse
Affiliation(s)
- Yoshimi Anzai
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, Utah, USA
| | - Satoshi Minoshima
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, Utah, USA
| | - Vivian S Lee
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
30
|
Hu X, Li F, Wang S, Xia F, Ling D. Biological Stimulus-Driven Assembly/Disassembly of Functional Nanoparticles for Targeted Delivery, Controlled Activation, and Bioelimination. Adv Healthc Mater 2018; 7:e1800359. [PMID: 29782706 DOI: 10.1002/adhm.201800359] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 04/20/2018] [Indexed: 12/23/2022]
Abstract
Nanoassembly technology has emerged as a powerful tool for targeted drug delivery and provides a basis for fabricating medical theranostic nanosystems. However, it is extremely difficult to concentrate nanoparticles at tumor sites, and the poor target-to-background ratio undoubtedly obstructs the accurate diagnosis and effective therapy of cancerous tissues. Importantly, the addition of biological stimulus-responsive groups to nanoassembly systems can enable a biological stimulus-driven assembly-disassembly mutual switch or structural composition/conformation change, thereby amplifying the imaging signal and/or enhancing the therapeutic effect. This progress report provides an overview of well-designed biological stimulus-responsive nanosystems that can realize precise assembly-disassembly switches by disrupting or rebuilding the intricate balance between the entropy and enthalpy of the nanosystems in response to stimuli (pH, redox, enzymes, etc.) in tumor tissues. The discussion encompasses different biological stimulus-responsive groups, fabrication approaches, and outstanding selective "turn-on" performance for efficient tumor imaging, therapy, and bioelimination. This progress report is expected to inspire more extensive research for the development of smart "turn-on" nanomaterials with increased signal-to-noise (S/N) ratios for diagnosis and drug delivery, which may pave the way for precise nanomedicine with site-specific theranostic features and biocompatibility.
Collapse
Affiliation(s)
- Xi Hu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou 310058 China
| | - Fangyuan Li
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou 310058 China
- Hangzhou Institute of Innovative Medicine; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou 310058 China
| | - Shuying Wang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou 310058 China
| | - Fan Xia
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou 310058 China
- Hangzhou Institute of Innovative Medicine; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou 310058 China
| | - Daishun Ling
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou 310058 China
- Hangzhou Institute of Innovative Medicine; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou 310058 China
- Key Laboratory of Biomedical Engineering of the Ministry of Education; College of Biomedical Engineering and Instrument Science; Zhejiang University; Hangzhou 310027 China
| |
Collapse
|
31
|
Cong Y, Qiao ZY, Wang H. Molecular Self-Assembly Constructed in Physiological Conditions for Cancer Diagnosis and Therapy. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800067] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Yong Cong
- CAS Center for Excellence in Nanoscience; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety; National Center for Nanoscience and Technology; No. 11 Beiyitiao, Zhongguancun Beijing 100190 China
| | - Zeng-Ying Qiao
- CAS Center for Excellence in Nanoscience; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety; National Center for Nanoscience and Technology; No. 11 Beiyitiao, Zhongguancun Beijing 100190 China
| | - Hao Wang
- CAS Center for Excellence in Nanoscience; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety; National Center for Nanoscience and Technology; No. 11 Beiyitiao, Zhongguancun Beijing 100190 China
| |
Collapse
|
32
|
Dang T, Suchy M, Truong YJ, Oakden W, Lam WW, Lazurko C, Facey G, Stanisz GJ, Shuhendler AJ. Hydrazo-CEST: Hydrazone-Dependent Chemical Exchange Saturation Transfer Magnetic Resonance Imaging Contrast Agents. Chemistry 2018; 24:9148-9156. [PMID: 29645309 DOI: 10.1002/chem.201801671] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Indexed: 11/06/2022]
Abstract
The rapid formation of hydrazones under physiological conditions was exploited for the detection of aldehydes through chemical exchange saturation transfer magnetic resonance imaging (CEST-MRI). A metal-free, diamagnetic contrast agent derived from N-amino anthranilic acid was introduced, which selectively "turned-on" upon hydrazone formation through an effect termed Hydrazo-CEST. While the hydrazine form of the probe produced no CEST-MRI signal enhancement, the formation of the aryl hydrazone resulted in >20 % intensity decrease in the bulk water signal through the CEST effect, as measured by 300 MHz 1 H NMR, 3 T and 7 T MRI. Both the electronic contributions of the N-amino anthranilate and the aldehyde binding partner were shown to directly impact the exchange rate of the proton on the ring-proximal nitrogen, and thus the imaging signal. Additionally, the presence of the carboxylic acid moiety ortho to the hydrazine was necessary not only for contrast production, but also for rapid hydrazone formation and prolonged hydrazone product stability under physiological conditions. This work provided the first example of an MRI-based contrast agent capable of a "turn on" response upon reaction with bioactive aldehydes, and outlined both the structural and electronic requirements to expand on Hydrazo-CEST, a novel, hydrazone-dependent subtype of diamagnetic CEST-MRI.
Collapse
Affiliation(s)
- Trina Dang
- Department of Chemistry & Biomolecular Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Mojmír Suchy
- Department of Chemistry & Biomolecular Sciences, University of Ottawa, Ottawa, ON, Canada.,University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Yen J Truong
- Department of Chemistry & Biomolecular Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Wendy Oakden
- Department of Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Wilfred W Lam
- Department of Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Caitlin Lazurko
- Department of Chemistry & Biomolecular Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Glenn Facey
- Department of Chemistry & Biomolecular Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Greg J Stanisz
- Department of Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Biomedical Physics, University of Toronto, Toronto, ON, Canada.,Department of Neurosurgery and Pediatric Neurosurgery, Medical University, Lublin, Poland
| | - Adam J Shuhendler
- Department of Chemistry & Biomolecular Sciences, University of Ottawa, Ottawa, ON, Canada.,University of Ottawa Heart Institute, Ottawa, ON, Canada
| |
Collapse
|
33
|
Prayongrat A, Umegaki K, van der Schaaf A, Koong AC, Lin SH, Whitaker T, McNutt T, Matsufuji N, Graves E, Mizuta M, Ogawa K, Date H, Moriwaki K, Ito YM, Kobashi K, Dekura Y, Shimizu S, Shirato H. Present developments in reaching an international consensus for a model-based approach to particle beam therapy. JOURNAL OF RADIATION RESEARCH 2018; 59:i72-i76. [PMID: 29529229 PMCID: PMC5868183 DOI: 10.1093/jrr/rry008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/27/2017] [Indexed: 06/13/2023]
Abstract
Particle beam therapy (PBT), including proton and carbon ion therapy, is an emerging innovative treatment for cancer patients. Due to the high cost of and limited access to treatment, meticulous selection of patients who would benefit most from PBT, when compared with standard X-ray therapy (XRT), is necessary. Due to the cost and labor involved in randomized controlled trials, the model-based approach (MBA) is used as an alternative means of establishing scientific evidence in medicine, and it can be improved continuously. Good databases and reasonable models are crucial for the reliability of this approach. The tumor control probability and normal tissue complication probability models are good illustrations of the advantages of PBT, but pre-existing NTCP models have been derived from historical patient treatments from the XRT era. This highlights the necessity of prospectively analyzing specific treatment-related toxicities in order to develop PBT-compatible models. An international consensus has been reached at the Global Institution for Collaborative Research and Education (GI-CoRE) joint symposium, concluding that a systematically developed model is required for model accuracy and performance. Six important steps that need to be observed in these considerations include patient selection, treatment planning, beam delivery, dose verification, response assessment, and data analysis. Advanced technologies in radiotherapy and computer science can be integrated to improve the efficacy of a treatment. Model validation and appropriately defined thresholds in a cost-effectiveness centered manner, together with quality assurance in the treatment planning, have to be achieved prior to clinical implementation.
Collapse
Affiliation(s)
- Anussara Prayongrat
- Department of Radiation Oncology, Graduate School of Medicine, Hokkaido University, North-15 West-7, Kita-ku, Sapporo, 0608638, Japan
| | - Kikuo Umegaki
- Faculty of Engineering, Hokkaido University, North-13, West-8, Kita-ku, Sapporo, 0608628, Japan
- Global Station for Quantum Biomedical Science and Engineering, Global Institute for Cooperative Research and Education, Hokkaido University, North-15 West-7, Kita-ku, Sapporo, 0608638, Japan
| | - Arjen van der Schaaf
- Department of Radiation Oncology, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Albert C Koong
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Steven H Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Thomas Whitaker
- Department of Radiation Oncology, Mayo Clinic, 200 1st St SW, Rochester, MN 55902, USA
| | - Todd McNutt
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205, USA
| | - Naruhiro Matsufuji
- National Institute of Radiological Sciences, 4 Chome-9-1 Anagawa, Inage Ward, Chiba, 2630024, Japan
| | - Edward Graves
- Department of Radiation Oncology, Stanford University, 291 Campus Drive, Stanford, CA 94305, USA
| | - Masahiko Mizuta
- Graduate School of Information Science and Technology, Hokkaido University, North-14 West-9, Kita-ku, Sapporo, 0600814, Japan
| | - Kazuhiko Ogawa
- Department of Radiation Oncology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hiroyuki Date
- Faculty of Health Sciences, Hokkaido University, North-12 West-5, Kita-ku, Sapporo, 060-0812, Japan
| | - Kensuke Moriwaki
- Department of Medical Statistics, Kobe Pharmaceutical University, 4-19-1, Motoyamakitamachi, Higashinada-ku, Kobe-shi, Hyogo, 658-8558, Japan
| | - Yoichi M Ito
- Department of Biostatistics, Faculty of Medicine, Hokkaido University, North-15 West-7, Kita-ku, Sapporo, 0608638, Japan
| | - Keiji Kobashi
- Department of Medical Physics, Faculty of Medicine, Hokkaido University, North-15 West-7, Kita-ku, Sapporo, 0608638, Japan
| | - Yasuhiro Dekura
- Department of Radiation Oncology, Graduate School of Medicine, Hokkaido University, North-15 West-7, Kita-ku, Sapporo, 0608638, Japan
| | - Shinichi Shimizu
- Global Station for Quantum Biomedical Science and Engineering, Global Institute for Cooperative Research and Education, Hokkaido University, North-15 West-7, Kita-ku, Sapporo, 0608638, Japan
- Department of Radiation Oncology, Faculty of Medicine, Hokkaido University, North-15 West-7, Kita-ku, Sapporo, 0608638, Japan
| | - Hiroki Shirato
- Global Station for Quantum Biomedical Science and Engineering, Global Institute for Cooperative Research and Education, Hokkaido University, North-15 West-7, Kita-ku, Sapporo, 0608638, Japan
- Department of Radiation Medicine, Faculty of Medicine, Hokkaido University, North-15 West-7, Kita-ku, Sapporo, 0608638, Japan
| |
Collapse
|
34
|
Hartshorn CM, Bradbury MS, Lanza GM, Nel AE, Rao J, Wang AZ, Wiesner UB, Yang L, Grodzinski P. Nanotechnology Strategies To Advance Outcomes in Clinical Cancer Care. ACS NANO 2018; 12:24-43. [PMID: 29257865 PMCID: PMC6589353 DOI: 10.1021/acsnano.7b05108] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Ongoing research into the application of nanotechnology for cancer treatment and diagnosis has demonstrated its advantages within contemporary oncology as well as its intrinsic limitations. The National Cancer Institute publishes the Cancer Nanotechnology Plan every 5 years since 2005. The most recent iteration helped codify the ongoing basic and translational efforts of the field and displayed its breadth with several evolving areas. From merely a technological perspective, this field has seen tremendous growth and success. However, an incomplete understanding of human cancer biology persists relative to the application of nanoscale materials within contemporary oncology. As such, this review presents several evolving areas in cancer nanotechnology in order to identify key clinical and biological challenges that need to be addressed to improve patient outcomes. From this clinical perspective, a sampling of the nano-enabled solutions attempting to overcome barriers faced by traditional therapeutics and diagnostics in the clinical setting are discussed. Finally, a strategic outlook of the future is discussed to highlight the need for next-generation cancer nanotechnology tools designed to address critical gaps in clinical cancer care.
Collapse
Affiliation(s)
- Christopher M Hartshorn
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
- Corresponding Author,
| | - Michelle S Bradbury
- Department of Radiology and Molecular Pharmacology Program, Sloan Kettering Institute for Cancer Research, New York, New York, 10065, United States
| | - Gregory M Lanza
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri 63108, United States
| | - Andre E Nel
- Division of NanoMedicine, Department of Medicine, and California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Jianghong Rao
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, California 94305, United States
| | - Andrew Z. Wang
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Ulrich B Wiesner
- Department of Materials Science and Engineering, Cornell University, Ithaca, New York 14843, United States
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Piotr Grodzinski
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
- Corresponding Author,
| |
Collapse
|
35
|
Wang Y, An R, Luo Z, Ye D. Firefly Luciferin-Inspired Biocompatible Chemistry for Protein Labeling and In Vivo Imaging. Chemistry 2017; 24:5707-5722. [PMID: 29068109 DOI: 10.1002/chem.201704349] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Indexed: 12/27/2022]
Abstract
Biocompatible reactions have emerged as versatile tools to build various molecular imaging probes that hold great promise for the detection of biological processes in vitro and/or in vivo. In this Minireview, we describe the recent advances in the development of a firefly luciferin-inspired biocompatible reaction between cyanobenzothiazole (CBT) and cysteine (Cys), and highlight its versatility to label proteins and build multimodality molecular imaging probes. The review starts from the general introduction of biocompatible reactions, which is followed by briefly describing the development of the firefly luciferin-inspired biocompatible chemistry. We then discuss its applications for the specific protein labeling and for the development of multimodality imaging probes (fluorescence, bioluminescence, MRI, PET, photoacoustic, etc.) that enable high sensitivity and spatial resolution imaging of redox environment, furin and caspase-3/7 activity in living cells and mice. Finally, we offer the conclusions and our perspective on the various and potential applications of this reaction. We hope that this review will contribute to the research of biocompatible reactions for their versatile applications in protein labeling and molecular imaging.
Collapse
Affiliation(s)
- Yuqi Wang
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Ruibing An
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Zhiliang Luo
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Deju Ye
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| |
Collapse
|
36
|
Tremblay ML, Davis C, Bowen CV, Stanley O, Parsons C, Weir G, Karkada M, Stanford MM, Brewer KD. Using MRI cell tracking to monitor immune cell recruitment in response to a peptide-based cancer vaccine. Magn Reson Med 2017; 80:304-316. [PMID: 29193231 DOI: 10.1002/mrm.27018] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/26/2017] [Accepted: 10/28/2017] [Indexed: 12/18/2022]
Abstract
PURPOSE MRI cell tracking can be used to monitor immune cells involved in the immunotherapy response, providing insight into the mechanism of action, temporal progression of tumor growth, and individual potency of therapies. To evaluate whether MRI could be used to track immune cell populations in response to immunotherapy, CD8+ cytotoxic T cells, CD4+ CD25+ FoxP3+ regulatory T cells, and myeloid-derived suppressor cells were labeled with superparamagnetic iron oxide particles. METHODS Superparamagnetic iron oxide-labeled cells were injected into mice (one cell type/mouse) implanted with a human papillomavirus-based cervical cancer model. Half of these mice were also vaccinated with DepoVaxTM (ImmunoVaccine, Inc., Halifax, Nova Scotia, Canada), a lipid-based vaccine platform that was developed to enhance the potency of peptide-based vaccines. RESULTS MRI visualization of CD8+ cytotoxic T cells, regulatory T cells, and myeloid-derived suppressor cells was apparent 24 h post-injection, with hypointensities due to iron-labeled cells clearing approximately 72 h post-injection. Vaccination resulted in increased recruitment of CD8+ cytotoxic T cells, and decreased recruitment of myeloid-derived suppressor cells and regulatory T cells to the tumor. We also found that myeloid-derived suppressor cell and regulatory T cell recruitment were positively correlated with final tumor volume. CONCLUSION This type of analysis can be used to noninvasively study changes in immune cell recruitment in individual mice over time, potentially allowing improved application and combination of immunotherapies. Magn Reson Med 80:304-316, 2018. © 2017 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
| | - Christa Davis
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, Nova Scotia, Canada
| | - Chris V Bowen
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, Nova Scotia, Canada.,Department of Diagnostic Radiology, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Physics and Atmospheric Science, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Olivia Stanley
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, Nova Scotia, Canada
| | - Cathryn Parsons
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, Nova Scotia, Canada
| | | | - Mohan Karkada
- Wyss Institute at Harvard Medical School, Boston, Massachusetts, USA
| | - Marianne M Stanford
- Immunovaccine Inc., Halifax, Nova Scotia, Canada.,Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kimberly D Brewer
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, Nova Scotia, Canada.,Department of Diagnostic Radiology, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Physics and Atmospheric Science, Dalhousie University, Halifax, Nova Scotia, Canada.,School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
37
|
Galiè M, Boschi F, Scambi I, Merigo F, Marzola P, Altabella L, Lavagnolo U, Sbarbati A, Spinelli AE. Theranostic Role of 32P-ATP as Radiopharmaceutical for the Induction of Massive Cell Death within Avascular Tumor Core. Theranostics 2017; 7:4399-4409. [PMID: 29158835 PMCID: PMC5695139 DOI: 10.7150/thno.21403] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/01/2017] [Indexed: 01/04/2023] Open
Abstract
Drug inaccessibility to vast areas of the tumor parenchyma is amongst the major hurdles for conventional therapies. Treatment efficacy rapidly decreases with distance from vessels and most of the tumor cells survive therapy. Also, between subsequent cycles of treatment, spared cancer cells replace those killed near the vessels, improving their access to nutrients, boosting their proliferation rate, and thus enabling tumor repopulation. Because of their property of "acting at a distance," radioisotopes are believed to overcome the physical barrier of vascular inaccessibility. Methods A novel molecular imaging tool called Cerenkov Luminescence Imaging (CLI) was employed for the detection of Cerenkov radiation emitted by beta particles, allowing in vivo tracking of beta-emitters. More precisely we investigated using a xenograft model of colon carcinoma the potential use of 32P-ATP as a novel theranostic radiopharmaceutical for tracing tumor lesions while simultaneously hampering their growth. Results Our analyses demonstrated that 32P-ATP injected into tumor-bearing mice reaches tumor lesions and persists for days and weeks within the tumor parenchyma. Also, the high-penetrating beta particles of 32P-ATP exert a "cross-fire" effect that induces massive cell death throughout the entire tumor parenchyma including core regions. Conclusion Our findings suggest 32P-ATP treatment as a potential approach to complement conventional therapies that fail to reach the tumor core and to prevent tumor repopulation.
Collapse
|
38
|
Chen H, Zhang W, Zhu G, Xie J, Chen X. Rethinking cancer nanotheranostics. NATURE REVIEWS. MATERIALS 2017; 2:17024. [PMID: 29075517 PMCID: PMC5654564 DOI: 10.1038/natrevmats.2017.24] [Citation(s) in RCA: 745] [Impact Index Per Article: 93.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Advances in nanoparticle synthesis and engineering have produced nanoscale agents affording both therapeutic and diagnostic functions that are often referred to by the portmanteau 'nanotheranostics'. The field is associated with many applications in the clinic, especially in cancer management. These include patient stratification, drug-release monitoring, imaging-guided focal therapy and post-treatment response monitoring. Recent advances in nanotheranostics have expanded this notion and enabled the characterization of individual tumours, the prediction of nanoparticle-tumour interactions, and the creation of tailor-designed nanomedicines for individualized treatment. Some of these applications require breaking the dogma that a nanotheranostic must combine both therapeutic and diagnostic agents within a single, physical entity; instead, it can be a general approach in which diagnosis and therapy are interwoven to solve clinical issues and improve treatment outcomes. In this Review, we describe the evolution and state of the art of cancer nanotheranostics, with an emphasis on clinical impact and translation.
Collapse
Affiliation(s)
- Hongmin Chen
- Center for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China
- Department of Chemistry, University of Georgia, Athens, Georgia 30602, USA
- Bio-imaging Research Center, University of Georgia, Athens, Georgia 30602, USA
| | - Weizhong Zhang
- Department of Chemistry, University of Georgia, Athens, Georgia 30602, USA
| | - Guizhi Zhu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Jin Xie
- Department of Chemistry, University of Georgia, Athens, Georgia 30602, USA
- Bio-imaging Research Center, University of Georgia, Athens, Georgia 30602, USA
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
39
|
Shan XH, Wang P, Xiong F, Lu HY, Hu H. Detection of human breast cancer cells using a 2-deoxy-D-glucose-functionalized superparamagnetic iron oxide nanoparticles. Cancer Biomark 2017; 18:367-374. [PMID: 28106540 DOI: 10.3233/cbm-160258] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Xiu-Hong Shan
- Department of Radiology, The Affiliated Renmin Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, China
| | - Peng Wang
- Department of Radiology, The Affiliated Renmin Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, China
| | - Fei Xiong
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Hao-Yue Lu
- Medical college of Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Hui Hu
- Department of Radiology, The Affiliated Renmin Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, China
| |
Collapse
|
40
|
|
41
|
Zhang P, He Y, Liu J, Feng J, Sun Z, Lei P, Yuan Q, Zhang H. Core–shell BaYbF5:Tm@BaGdF5:Yb,Tm nanocrystals for in vivo trimodal UCL/CT/MR imaging. RSC Adv 2016. [DOI: 10.1039/c5ra22991j] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
PEGylated core–shell BaYbF5:Tm@BaGdF5:Yb,Tm nanocrystals have been constructed and successfully applied in UCL imaging, CT imaging and MR imaging.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Radiology
- The Second Hospital of Jilin University
- Changchun 130041
- P. R. China
| | - Yangyang He
- Department of Pathology
- The Second Hospital of Jilin University
- Changchun 130041
- P. R. China
| | - Jianhua Liu
- Department of Radiology
- The Second Hospital of Jilin University
- Changchun 130041
- P. R. China
| | - Jing Feng
- State Key Laboratory of Rare Earth Resource Utilization
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
- P. R. China
| | - Zhiqiang Sun
- Department of Interventional Therapy
- Cancer Hospital of Jilin Province
- Changchun 132000
- P. R. China
| | - Pengpeng Lei
- State Key Laboratory of Rare Earth Resource Utilization
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
- P. R. China
| | - Qinghai Yuan
- Department of Radiology
- The Second Hospital of Jilin University
- Changchun 130041
- P. R. China
| | - Hongjie Zhang
- State Key Laboratory of Rare Earth Resource Utilization
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
- P. R. China
| |
Collapse
|
42
|
Recent advances in cell imaging and cytotoxicity of intracellular stimuli-responsive nanomaterials. Sci Bull (Beijing) 2015. [DOI: 10.1007/s11434-015-0952-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|