1
|
Chavda VP, Balar PC, Nalla LV, Bezbaruah R, Gogoi NR, Gajula SNR, Peng B, Meena AS, Conde J, Prasad R. Conjugated Nanoparticles for Solid Tumor Theranostics: Unraveling the Interplay of Known and Unknown Factors. ACS OMEGA 2023; 8:37654-37684. [PMID: 37867666 PMCID: PMC10586263 DOI: 10.1021/acsomega.3c05069] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/19/2023] [Indexed: 10/24/2023]
Abstract
Cancer diagnoses have been increasing worldwide, and solid tumors are among the leading contributors to patient mortality, creating an enormous burden on the global healthcare system. Cancer is responsible for around 10.3 million deaths worldwide. Solid tumors are one of the most prevalent cancers observed in recent times. On the other hand, early diagnosis is a significant challenge that could save a person's life. Treatment with existing methods has pitfalls that limit the successful elimination of the disorder. Though nanoparticle-based imaging and therapeutics have shown a significant impact in healthcare, current methodologies for solid tumor treatment are insufficient. There are multiple complications associated with the diagnosis and management of solid tumors as well. Recently, surface-conjugated nanoparticles such as lipid nanoparticles, metallic nanoparticles, and quantum dots have shown positive results in solid tumor diagnostics and therapeutics in preclinical models. Other nanotheranostic material platforms such as plasmonic theranostics, magnetotheranostics, hybrid nanotheranostics, and graphene theranostics have also been explored. These nanoparticle theranostics ensure the appropriate targeting of tumors along with selective delivery of cargos (both imaging and therapeutic probes) without affecting the surrounding healthy tissues. Though they have multiple applications, nanoparticles still possess numerous limitations that need to be addressed in order to be fully utilized in the clinic. In this review, we outline the importance of materials and design strategies used to engineer nanoparticles in the treatment and diagnosis of solid tumors and how effectively each method overcomes the drawbacks of the current techniques. We also highlight the gaps in each material platform and how design considerations can address their limitations in future research directions.
Collapse
Affiliation(s)
- Vivek P. Chavda
- Department
of Pharmaceutics and Pharmaceutical Technology, L.M. College of Pharmacy, Ahmedabad 380001, India
| | - Pankti C. Balar
- Pharmacy
Section, L.M. College of Pharmacy, Ahmedabad 380001, India
| | - Lakshmi Vineela Nalla
- Department
of Pharmacy, Koneru Lakshmaiah Education
Foundation, Vaddeswaram, Andhra Pradesh 522302, India
| | - Rajashri Bezbaruah
- Department
of Pharmaceutical Sciences, Faculty of Science
and Engineering, Dibrugarh, 786004 Assam, India
| | - Niva Rani Gogoi
- Department
of Pharmaceutical Sciences, Faculty of Science
and Engineering, Dibrugarh, 786004 Assam, India
| | - Siva Nageswara Rao Gajula
- Department
of Pharmaceutical Analysis, GITAM School of Pharmacy, GITAM (Deemed to be University), Rushikonda, Visakhapatnam, Andhra Pradesh 530045, India
| | - Berney Peng
- Department
of Pathology and Laboratory Medicine, University
of California at Los Angeles, Los
Angeles, California 90095, United States
| | - Avtar S. Meena
- Department
of Biotechnology, All India Institute of
Medical Sciences (AIIMS), Ansari
Nagar, New Delhi 110029, India
| | - João Conde
- ToxOmics,
NOVA Medical School, Faculdade de Ciências Médicas,
NMS|FCM, Universidade Nova de Lisboa, Lisboa 1169-056, Portugal
| | - Rajendra Prasad
- School
of Biochemical Engineering, Indian Institute
of Technology (BHU), Varanasi 221005, India
| |
Collapse
|
2
|
Maxouri O, Bodalal Z, Daal M, Rostami S, Rodriguez I, Akkari L, Srinivas M, Bernards R, Beets-Tan R. How to 19F MRI: applications, technique, and getting started. BJR Open 2023; 5:20230019. [PMID: 37953866 PMCID: PMC10636348 DOI: 10.1259/bjro.20230019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 07/26/2023] [Accepted: 08/01/2023] [Indexed: 11/14/2023] Open
Abstract
Magnetic resonance imaging (MRI) plays a significant role in the routine imaging workflow, providing both anatomical and functional information. 19F MRI is an evolving imaging modality where instead of 1H, 19F nuclei are excited. As the signal from endogenous 19F in the body is negligible, exogenous 19F signals obtained by 19F radiofrequency coils are exceptionally specific. Highly fluorinated agents targeting particular biological processes (i.e., the presence of immune cells) have been visualised using 19F MRI, highlighting its potential for non-invasive and longitudinal molecular imaging. This article aims to provide both a broad overview of the various applications of 19F MRI, with cancer imaging as a focus, as well as a practical guide to 19F imaging. We will discuss the essential elements of a 19F system and address common pitfalls during acquisition. Last but not least, we will highlight future perspectives that will enhance the role of this modality. While not an exhaustive exploration of all 19F literature, we endeavour to encapsulate the broad themes of the field and introduce the world of 19F molecular imaging to newcomers. 19F MRI bridges several domains, imaging, physics, chemistry, and biology, necessitating multidisciplinary teams to be able to harness this technology effectively. As further technical developments allow for greater sensitivity, we envision that 19F MRI can help unlock insight into biological processes non-invasively and longitudinally.
Collapse
Affiliation(s)
| | | | | | | | | | - Leila Akkari
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - René Bernards
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | |
Collapse
|
3
|
Yu H, Guo H, Wang Y, Wang Y, Zhang L. Bismuth nanomaterials as contrast agents for radiography and computed tomography imaging and their quality/safety considerations. WIRES NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1801. [DOI: 10.1002/wnan.1801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 11/24/2022]
Affiliation(s)
- Huan Yu
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD‐X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions Soochow University Suzhou China
| | - Haoxiang Guo
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD‐X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions Soochow University Suzhou China
| | - Yong Wang
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD‐X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions Soochow University Suzhou China
| | - Yangyun Wang
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD‐X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions Soochow University Suzhou China
| | - Leshuai Zhang
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD‐X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions Soochow University Suzhou China
| |
Collapse
|
4
|
Huang Y, He Z, Manyande A, Feng M, Xiang H. Nerve regeneration in transplanted organs and tracer imaging studies: A review. Front Bioeng Biotechnol 2022; 10:966138. [PMID: 36051591 PMCID: PMC9424764 DOI: 10.3389/fbioe.2022.966138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
The technique of organ transplantation is well established and after transplantation the patient might be faced with the problem of nerve regeneration of the transplanted organ. Transplanted organs are innervated by the sympathetic, parasympathetic, and visceral sensory plexuses, but there is a lack of clarity regarding the neural influences on the heart, liver and kidneys and the mechanisms of their innervation. Although there has been considerable recent work exploring the potential mechanisms of nerve regeneration in organ transplantation, there remains much that is unknown about the heterogeneity and individual variability in the reinnervation of organ transplantation. The widespread availability of radioactive nerve tracers has also made a significant contribution to organ transplantation and has helped to investigate nerve recovery after transplantation, as well as providing a direction for future organ transplantation research. In this review we focused on neural tracer imaging techniques in humans and provide some conceptual insights into theories that can effectively support our choice of radionuclide tracers. This also facilitates the development of nuclear medicine techniques and promotes the development of modern medical technologies and computer tools. We described the knowledge of neural regeneration after heart transplantation, liver transplantation and kidney transplantation and apply them to various imaging techniques to quantify the uptake of radionuclide tracers to assess the prognosis of organ transplantation. We noted that the aim of this review is both to provide clinicians and nuclear medicine researchers with theories and insights into nerve regeneration in organ transplantation and to advance imaging techniques and radiotracers as a major step forward in clinical research. Moreover, we aimed to further promote the clinical and research applications of imaging techniques and provide clinicians and research technology developers with the theory and knowledge of the nerve.
Collapse
Affiliation(s)
- Yan Huang
- Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Interventional Therapy, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Zhigang He
- Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Anne Manyande
- School of Human and Social Sciences, University of West London, London, United Kingdom
| | - Maohui Feng
- Department of Gastrointestinal Surgery, Wuhan Peritoneal Cancer Clinical Medical Research Center, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuhan, Hubei, China
- *Correspondence: Maohui Feng, ; Hongbing Xiang,
| | - Hongbing Xiang
- Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- *Correspondence: Maohui Feng, ; Hongbing Xiang,
| |
Collapse
|
5
|
Moonshi SS, Wu Y, Ta HT. Visualizing stem cells in vivo using magnetic resonance imaging. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 14:e1760. [PMID: 34651465 DOI: 10.1002/wnan.1760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/18/2021] [Accepted: 08/31/2021] [Indexed: 12/16/2022]
Abstract
Stem cell (SC) therapies displayed encouraging efficacy and clinical outcome in various disorders. Despite this huge hype, clinical translation of SC therapy has been disheartening due to contradictory results from clinical trials. The ability to monitor migration and engraftment of cells in vivo represents an ideal strategy in cell therapy. Therefore, suitable imaging approach to track MSCs would allow understanding of migratory and homing efficiency, optimal route of delivery and engraftment of cells at targeted location. Hence, longitudinal tracking of SCs is crucial for the optimization of treatment parameters, leading to improved clinical outcome and translation. Magnetic resonance imaging (MRI) represents a suitable imaging modality to observe cells non-invasively and repeatedly. Tracking is achieved when cells are incubated prior to implantation with appropriate contrast agents (CA) or tracers which can then be detected in an MRI scan. This review explores and emphasizes the importance of monitoring the distribution and fate of SCs post-implantation using current contrast agents, such as positive CAs including paramagnetic metals (gadolinium), negative contrast agents such as superparamagnetic iron oxides and 19 F containing tracers, specifically for the in vivo tracking of MSCs using MRI. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Shehzahdi Shebbrin Moonshi
- Queensland Microtechnology and Nanotechnology Centre, Griffith University, Nathan, Queensland, Australia
| | - Yuao Wu
- Queensland Microtechnology and Nanotechnology Centre, Griffith University, Nathan, Queensland, Australia
| | - Hang Thu Ta
- Queensland Microtechnology and Nanotechnology Centre, Griffith University, Nathan, Queensland, Australia.,Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland, Australia.,School of Environment and Science, Griffith University, Nathan, Queensland, Australia
| |
Collapse
|
6
|
Helfer BM, Ponomarev V, Patrick PS, Blower PJ, Feitel A, Fruhwirth GO, Jackman S, Pereira Mouriès L, Park MVDZ, Srinivas M, Stuckey DJ, Thu MS, van den Hoorn T, Herberts CA, Shingleton WD. Options for imaging cellular therapeutics in vivo: a multi-stakeholder perspective. Cytotherapy 2021; 23:757-773. [PMID: 33832818 PMCID: PMC9344904 DOI: 10.1016/j.jcyt.2021.02.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/01/2021] [Accepted: 02/13/2021] [Indexed: 12/13/2022]
Abstract
Cell-based therapies have been making great advances toward clinical reality. Despite the increase in trial activity, few therapies have successfully navigated late-phase clinical trials and received market authorization. One possible explanation for this is that additional tools and technologies to enable their development have only recently become available. To support the safety evaluation of cell therapies, the Health and Environmental Sciences Institute Cell Therapy-Tracking, Circulation and Safety Committee, a multisector collaborative committee, polled the attendees of the 2017 International Society for Cell & Gene Therapy conference in London, UK, to understand the gaps and needs that cell therapy developers have encountered regarding safety evaluations in vivo. The goal of the survey was to collect information to inform stakeholders of areas of interest that can help ensure the safe use of cellular therapeutics in the clinic. This review is a response to the cellular imaging interests of those respondents. The authors offer a brief overview of available technologies and then highlight the areas of interest from the survey by describing how imaging technologies can meet those needs. The areas of interest include imaging of cells over time, sensitivity of imaging modalities, ability to quantify cells, imaging cellular survival and differentiation and safety concerns around adding imaging agents to cellular therapy protocols. The Health and Environmental Sciences Institute Cell Therapy-Tracking, Circulation and Safety Committee believes that the ability to understand therapeutic cell fate is vital for determining and understanding cell therapy efficacy and safety and offers this review to aid in those needs. An aim of this article is to share the available imaging technologies with the cell therapy community to demonstrate how these technologies can accomplish unmet needs throughout the translational process and strengthen the understanding of cellular therapeutics.
Collapse
Affiliation(s)
| | - Vladimir Ponomarev
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - P Stephen Patrick
- Department of Medicine, Centre for Advanced Biomedical Imaging, University College London, London, UK
| | - Philip J Blower
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Alexandra Feitel
- Formerly, Health and Environmental Sciences Institute, US Environmental Protection Agency, Washington, DC, USA
| | - Gilbert O Fruhwirth
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Shawna Jackman
- Charles River Laboratories, Shrewsbury, Massachusetts, USA
| | | | - Margriet V D Z Park
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Mangala Srinivas
- Department of Tumor Immunology, Radboud University Medical Center, Nijmegen, the Netherlands; Cenya Imaging BV, Amsterdam, the Netherlands
| | - Daniel J Stuckey
- Department of Medicine, Centre for Advanced Biomedical Imaging, University College London, London, UK
| | - Mya S Thu
- Visicell Medical Inc, La Jolla, California, USA
| | | | | | | |
Collapse
|
7
|
Lu C, Han L, Wang J, Wan J, Song G, Rao J. Engineering of magnetic nanoparticles as magnetic particle imaging tracers. Chem Soc Rev 2021; 50:8102-8146. [PMID: 34047311 DOI: 10.1039/d0cs00260g] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Magnetic particle imaging (MPI) has recently emerged as a promising non-invasive imaging technique because of its signal linearly propotional to the tracer mass, ability to generate positive contrast, low tissue background, unlimited tissue penetration depth, and lack of ionizing radiation. The sensitivity and resolution of MPI are highly dependent on the properties of magnetic nanoparticles (MNPs), and extensive research efforts have been focused on the design and synthesis of tracers. This review examines parameters that dictate the performance of MNPs, including size, shape, composition, surface property, crystallinity, the surrounding environment, and aggregation state to provide guidance for engineering MPI tracers with better performance. Finally, we discuss applications of MPI imaging and its challenges and perspectives in clinical translation.
Collapse
Affiliation(s)
- Chang Lu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China.
| | - Linbo Han
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen 518118, P. R. China
| | - Joanna Wang
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, 1201 Welch Road, Stanford, California 94305-5484, USA.
| | - Jiacheng Wan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China.
| | - Guosheng Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China.
| | - Jianghong Rao
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, 1201 Welch Road, Stanford, California 94305-5484, USA.
| |
Collapse
|
8
|
Peng X, Wang J, Zhou F, Liu Q, Zhang Z. Nanoparticle-based approaches to target the lymphatic system for antitumor treatment. Cell Mol Life Sci 2021; 78:5139-5161. [PMID: 33963442 PMCID: PMC11072902 DOI: 10.1007/s00018-021-03842-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 03/14/2021] [Accepted: 04/16/2021] [Indexed: 02/07/2023]
Abstract
Immunotherapies have been established as safe and efficient modalities for numerous tumor treatments. The lymphatic system, which is an important system, can modulate the immune system via a complex network, which includes lymph nodes, vessels, and lymphocytes. With the deepening understanding of tumor immunology, a plethora of immunotherapies, which include vaccines, photothermal therapy, and photodynamic therapy, have been established for antitumor treatments. However, the deleterious off-target effects and nonspecific targeting of therapeutic agents result in low efficacy of immunotherapy. Fortunately, nanoparticle-based approaches for targeting the lymphatic system afford a unique opportunity to manufacture drugs that can simultaneously tackle both aspects, thereby improving tumor treatments. Over the past decades, great strides have been made in the development of DC vaccines and nanomedicine as antitumor treatments in the field of lymphatic therapeutics and diagnosis. In this review, we summarize the current strategies through which nanoparticle technology has been designed to target the lymphatic system and describe applications of lymphatic imaging for the diagnosis and image-guided surgery of tumor metastasis. Moreover, improvements in the tumor specificity of nanovaccines and medicines, which have been realized through targeting or stimulating the lymphatic system, can provide amplified antitumor immune responses and reduce side effects, thereby promoting the paradigm of antitumor treatment into the clinic to benefit patients.
Collapse
Affiliation(s)
- Xingzhou Peng
- School of Biomedical Engineering, Hainan University, Haikou, 570228, Hainan, China
| | - Junjie Wang
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Feifan Zhou
- School of Biomedical Engineering, Hainan University, Haikou, 570228, Hainan, China
| | - Qian Liu
- School of Biomedical Engineering, Hainan University, Haikou, 570228, Hainan, China.
| | - Zhihong Zhang
- School of Biomedical Engineering, Hainan University, Haikou, 570228, Hainan, China.
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China.
| |
Collapse
|
9
|
Xiao Z, Puré E. Imaging of T-cell Responses in the Context of Cancer Immunotherapy. Cancer Immunol Res 2021; 9:490-502. [PMID: 33941536 DOI: 10.1158/2326-6066.cir-20-0678] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/18/2020] [Accepted: 02/18/2021] [Indexed: 12/16/2022]
Abstract
Immunotherapy, which promotes the induction of cytotoxic T lymphocytes and enhances their infiltration into and function within tumors, is a rapidly expanding and evolving approach to treating cancer. However, many of the critical denominators for inducing effective anticancer immune responses remain unknown. Efforts are underway to develop comprehensive ex vivo assessments of the immune landscape of patients prior to and during response to immunotherapy. An important complementary approach to these efforts involves the development of noninvasive imaging approaches to detect immune targets, assess delivery of immune-based therapeutics, and evaluate responses to immunotherapy. Herein, we review the merits and limitations of various noninvasive imaging modalities (MRI, PET, and single-photon emission tomography) and discuss candidate targets for cellular and molecular imaging for visualization of T-cell responses at various stages along the cancer-immunity cycle in the context of immunotherapy. We also discuss the potential use of these imaging strategies in monitoring treatment responses and predicting prognosis for patients treated with immunotherapy.
Collapse
Affiliation(s)
- Zebin Xiao
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ellen Puré
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
10
|
Wang XY, Wang Y, Wu Q, Liu JJ, Liu Y, Pan DH, Qi W, Wang LZ, Yan JJ, Xu YP, Wang GJ, Miao LY, Yu L, Yang M. Feasibility study of 68Ga-labeled CAR T cells for in vivo tracking using micro-positron emission tomography imaging. Acta Pharmacol Sin 2021; 42:824-831. [PMID: 32901086 DOI: 10.1038/s41401-020-00511-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 08/17/2020] [Indexed: 12/17/2022] Open
Abstract
Clinical tracking of chimeric antigen receptor (CAR) T cells in vivo by positron emission tomography (PET) imaging is an area of intense interest. But the long-lived positron emitter-labeled CAR T cells stay in the liver and spleen for days or even weeks. Thus, the excessive absorbed effective dose becomes a major biosafety issue leading it difficult for clinical translation. In this study we used 68Ga, a commercially available short-lived positron emitter, to label CAR T cells for noninvasive cell tracking in vivo. CAR T cells could be tracked in vivo by 68Ga-PET imaging for at least 6 h. We showed a significant correlation between the distribution of 89Zr and 68Ga-labeled CAR T cells in the same tissues (lungs, liver, and spleen). The distribution and homing behavior of CAR T cells at the early period is highly correlated with the long-term fate of CAR T cells in vivo. And the effective absorbed dose of 68Ga-labeled CAR T cells is only one twenty-fourth of 89Zr-labeled CAR T cells, which was safe for clinical translation. We conclude the feasibility of 68Ga instead of 89Zr directly labeling CAR T cells for noninvasive tracking of the cells in vivo at an early stage based on PET imaging. This method provides a potential solution to the emerging need for safe and practical PET tracer for cell tracking clinically.
Collapse
|
11
|
Cohen O, Betzer O, Elmaliach-Pnini N, Motiei M, Sadan T, Cohen-Berkman M, Dagan O, Popovtzer A, Yosepovich A, Barhom H, Michaeli S, Popovtzer R. 'Golden' exosomes as delivery vehicles to target tumors and overcome intratumoral barriers: in vivo tracking in a model for head and neck cancer. Biomater Sci 2021; 9:2103-2114. [PMID: 33475633 DOI: 10.1039/d0bm01735c] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Exosomes are promising vectors for anti-tumor therapy, due to their biocompatibility, low immunogenicity, and innate ability to interact with target cells. However, promoting clinical application of exosome-based therapeutics requires elucidation of key issues, including exosome biodistribution, tumor targeting and accumulation, and the ability to overcome tumor barriers that limit the penetration of various nano-carriers and drugs. Here, we examined these parameters in exosomes derived from mesenchymal stem cells (MSC-exo) and from the A431 squamous cell carcinoma line (A431-exo), which both have potential use in cancer therapy. Using our novel technique combining gold nanoparticle (GNP) labeling of exosomes and non-invasive computed tomography imaging (CT), we longitudinally and quantitatively tracked the two intravenously-injected exosome types in A431 tumor-bearing mice. CT imaging up to 48 h and subsequent ex vivo analysis revealed tumor homing abilities of both exosome types, yet there was significantly higher tumor accumulation of MSC-exo as compared to A431-exo. Moreover, MSC-exo demonstrated the ability to penetrate the tumor and distribute throughout its bulk, while non-encapsulated GNPs remained concentrated at the tumor periphery. Histological analysis showed penetration of MSC-exo not only into the tumor tissue, but also into tumor cell cytoplasm. While the proportion of biodistribution between organs at 48 h was similar for both exosome types, more rapid clearance was indicated for A431-exo. Thus, our findings demonstrate an effect of exosome type on tumor targeting abilities and biodistribution, and suggest that MSC-exo may have superior abilities for tumor-targeted therapy.
Collapse
Affiliation(s)
- Oded Cohen
- Department of Otolaryngology, Head and Neck Surgery, Kaplan Medical Center, Rehovot, Israel, affiliated to the Hebrew University, Jerusalem, Israel
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Wu YL. Cardiac MRI Assessment of Mouse Myocardial Infarction and Regeneration. Methods Mol Biol 2021; 2158:81-106. [PMID: 32857368 DOI: 10.1007/978-1-0716-0668-1_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Small animal models are indispensable for cardiac regeneration research. Studies in mouse and rat models have provided important insights into the etiology and mechanisms of cardiovascular diseases and accelerated the development of therapeutic strategies. It is vitally important to be able to evaluate the therapeutic efficacy and have reliable surrogate markers for therapeutic development for cardiac regeneration research. Magnetic resonance imaging (MRI), a versatile and noninvasive imaging modality with excellent penetration depth, tissue coverage, and soft-tissue contrast, is becoming a more important tool in both clinical settings and research arenas. Cardiac MRI (CMR) is versatile, noninvasive, and capable of measuring many different aspects of cardiac functions, and, thus, is ideally suited to evaluate therapeutic efficacy for cardiac regeneration. CMR applications include assessment of cardiac anatomy, regional wall motion, myocardial perfusion, myocardial viability, cardiac function assessment, assessment of myocardial infarction, and myocardial injury. Myocardial infarction models in mice are commonly used model systems for cardiac regeneration research. In this chapter, we discuss various CMR applications to evaluate cardiac functions and inflammation after myocardial infarction.
Collapse
Affiliation(s)
- Yijen L Wu
- Department of Developmental Biology, Rangos Research Center Animal Imaging Core, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
13
|
Tremblay ML, O’Brien-Moran Z, Rioux JA, Nuschke A, Davis C, Kast WM, Weir G, Stanford M, Brewer KD. Quantitative MRI cell tracking of immune cell recruitment to tumors and draining lymph nodes in response to anti-PD-1 and a DPX-based immunotherapy. Oncoimmunology 2020; 9:1851539. [PMID: 33299663 PMCID: PMC7714509 DOI: 10.1080/2162402x.2020.1851539] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/11/2020] [Accepted: 11/11/2020] [Indexed: 11/01/2022] Open
Abstract
DPX is a unique T cell activating formulation that generates robust immune responses (both clinically and preclinically) which can be tailored to various cancers via the use of tumor-specific antigens and adjuvants. While DPX-based immunotherapies may act complementary with checkpoint inhibitors, combination therapy is not always easily predictable based on individual therapeutic responses. Optimizing these combinations can be improved by understanding the mechanism of action underlying the individual therapies. Magnetic Resonance Imaging (MRI) allows tracking of cells labeled with superparamagnetic iron oxide (SPIO), which can yield valuable information about the localization of crucial immune cell subsets. In this work, we evaluated the use of a multi-echo, single point MRI pulse sequence, TurboSPI, for tracking and quantifying cytotoxic T lymphocytes (CTLs) and myeloid lineage cells (MLCs). In a subcutaneous cervical cancer model (C3) we compared untreated mice to mice treated with either a single therapy (anti-PD-1 or DPX-R9F) or a combination of both therapies. We were able to detect, using TurboSPI, significant increases in CTL recruitment dynamics in response to combination therapy. We also observed differences in MLC recruitment to therapy-draining (DPX-R9F) lymph nodes in response to treatment with DPX-R9F (alone or in combination with anti-PD-1). We demonstrated that the therapies presented herein induced time-varying changes in cell recruitment. This work establishes that these quantitative molecular MRI techniques can be expanded to study a number of cancer and immunotherapy combinations to improve our understanding of longitudinal immunological changes and mechanisms of action.
Collapse
Affiliation(s)
- Marie-Laurence Tremblay
- Biomedical Translational Imaging Centre (BIOTIC, IWK Health Centre, Halifax, NS, Canada
- Dalhousie University, Halifax, NS, Canada
- IMV Inc, Halifax, NS, Canada
| | - Zoe O’Brien-Moran
- Biomedical Translational Imaging Centre (BIOTIC, IWK Health Centre, Halifax, NS, Canada
- Department of Physics, Dalhousie University, Halifax, NS, Canada
| | - James A. Rioux
- Biomedical Translational Imaging Centre (BIOTIC, IWK Health Centre, Halifax, NS, Canada
- Department of Physics, Dalhousie University, Halifax, NS, Canada
- Department of Diagnostic Radiology, Dalhousie University, Halifax, NS, Canada
| | - Andrea Nuschke
- Biomedical Translational Imaging Centre (BIOTIC, IWK Health Centre, Halifax, NS, Canada
| | - Christa Davis
- Biomedical Translational Imaging Centre (BIOTIC, IWK Health Centre, Halifax, NS, Canada
| | - W. Martin Kast
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Marianne Stanford
- Dalhousie University, Halifax, NS, Canada
- IMV Inc, Halifax, NS, Canada
| | - Kimberly D. Brewer
- Biomedical Translational Imaging Centre (BIOTIC, IWK Health Centre, Halifax, NS, Canada
- Dalhousie University, Halifax, NS, Canada
- Department of Physics, Dalhousie University, Halifax, NS, Canada
- Department of Diagnostic Radiology, Dalhousie University, Halifax, NS, Canada
- School of Biomedical Engineering, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
14
|
Hsu JC, Nieves LM, Betzer O, Sadan T, Noël PB, Popovtzer R, Cormode DP. Nanoparticle contrast agents for X-ray imaging applications. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1642. [PMID: 32441050 DOI: 10.1002/wnan.1642] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022]
Abstract
X-ray imaging is the most widely used diagnostic imaging method in modern medicine and several advanced forms of this technology have recently emerged. Iodinated molecules and barium sulfate suspensions are clinically approved X-ray contrast agents and are widely used. However, these existing contrast agents provide limited information, are suboptimal for new X-ray imaging techniques and are developing safety concerns. Thus, over the past 15 years, there has been a rapid growth in the development of nanoparticles as X-ray contrast agents. Nanoparticles have several desirable features such as high contrast payloads, the potential for long circulation times, and tunable physicochemical properties. Nanoparticles have also been used in a range of biomedical applications such as disease treatment, targeted imaging, and cell tracking. In this review, we discuss the principles behind X-ray contrast generation and introduce new types of X-ray imaging modalities, as well as potential elements and chemical compositions that are suitable for novel contrast agent development. We focus on the progress in nanoparticle X-ray contrast agents developed to be renally clearable, long circulating, theranostic, targeted, or for cell tracking. We feature agents that are used in conjunction with the newly developed multi-energy computed tomography and mammographic imaging technologies. Finally, we offer perspectives on current limitations and emerging research topics as well as expectations for the future development of the field. This article is categorized under: Diagnostic Tools > in vivo Nanodiagnostics and Imaging Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Jessica C Hsu
- Department of Radiology, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Bioengineering, School of Engineering and Applied Science of the University of Pennsylvania, Pennsylvania, USA
| | - Lenitza M Nieves
- Department of Radiology, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Biochemistry and Molecular Biophysics, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Oshra Betzer
- Faculty of Engineering and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
| | - Tamar Sadan
- Faculty of Engineering and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
| | - Peter B Noël
- Department of Radiology, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rachela Popovtzer
- Faculty of Engineering and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
| | - David P Cormode
- Department of Radiology, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Bioengineering, School of Engineering and Applied Science of the University of Pennsylvania, Pennsylvania, USA.,Department of Biochemistry and Molecular Biophysics, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
15
|
Kritchenkov IS, Zhukovsky DD, Mohamed A, Korzhikov-Vlakh VA, Tennikova TB, Lavrentieva A, Scheper T, Pavlovskiy VV, Porsev VV, Evarestov RA, Tunik SP. Functionalized Pt(II) and Ir(III) NIR Emitters and Their Covalent Conjugates with Polymer-Based Nanocarriers. Bioconjug Chem 2020; 31:1327-1343. [PMID: 32223218 DOI: 10.1021/acs.bioconjchem.0c00020] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Two NIR-emitting platinum [Pt(N^N^C)(phosphine)] and iridium [Ir(N^C)2(N^N)]+ complexes containing reactive succinimide groups were synthesized and characterized with spectroscopic methods (N^N^C, 1-phenyl-3-(pyridin-2-yl)benzo[4,5]imidazo[1,2-a]pyrazine, N^C, 6-(2-benzothienyl)phenanthridine, phosphine-3-(diphenylphosphaneyl)propanoic acid N-hydroxysuccinimide ether, and N^N, 4-oxo-4-((1-(pyridin-2-yl)-1H-1,2,3-triazol-4-yl)methoxy)butanoic acid N-hydroxysuccinimide ether). Their photophysics were carefully studied and analyzed using time-dependent density functional theory calculations. These complexes were used to prepare luminescent micro- and nanoparticles with the "core-shell" morphology, where the core consisted of biodegradable polymers of different hydrophobicity, namely, poly(d,l-lactic acid), poly(ε-caprolactone), and poly(ω-pentadecalactone), whereas the shell was formed by covalent conjugation with poly(l-lysine) covalently labeled with the platinum and iridium emitters. The surface of the species was further modified with heparin to reverse their charge from positive to negative values. The microparticles' size determined with dynamic laser scanning varies considerably from 720 to 1480 nm, but the nanoparticles' diameter falls in a rather narrow range, 210-230 nm. The species with a poly(l-lysine) shell display a high positive (>30 mV) zeta-potential that makes them essentially stable in aqueous media. Inversion of the surface charge to a negative value with the heparin cover did not deteriorate the species' stability. The iridium- and platinum-containing particles displayed emissions the spectral patterns of which were essentially similar to those of unconjugated complexes, which indicate retention of the chromophore nature upon binding to the polymer and further immobilization onto polyester micro- and nanoparticles for drug delivery. The obtained particles were tested to determine their ability to penetrate into different cells types: cancer cells, stem cells, and fibroblasts. It was found that all types of particles could effectively penetrate into all cells types under investigation. Nanoparticles were shown to penetrate into the cells more effectively than microparticles. However, positively charged nanoparticles covered with poly(l-lysine) seem to interact with negatively charged proteins in the medium and enter the inner part of the cells less effectively than nanoparticles covered with poly(l-lysine)/heparin. In the case of microparticles, the species with positive zeta-potentials were more readily up-taken by the cells than those with negative values.
Collapse
Affiliation(s)
- Ilya S Kritchenkov
- Institute of Chemistry, Saint-Petersburg State University, Saint-Petersburg 198504, Russia
| | - Daniil D Zhukovsky
- Institute of Chemistry, Saint-Petersburg State University, Saint-Petersburg 198504, Russia
| | - Abdelrahman Mohamed
- Institute of Chemistry, Saint-Petersburg State University, Saint-Petersburg 198504, Russia.,Faculty of Science, Chemistry Department, Beni-Suef University, 62511 Beni-Suef, Egypt
| | | | - Tatiana B Tennikova
- Institute of Chemistry, Saint-Petersburg State University, Saint-Petersburg 198504, Russia
| | | | - Thomas Scheper
- Institute of Technical Chemistry, Leibniz University, 30167 Hannover, Germany
| | - Vladimir V Pavlovskiy
- Institute of Chemistry, Saint-Petersburg State University, Saint-Petersburg 198504, Russia
| | - Vitaly V Porsev
- Institute of Chemistry, Saint-Petersburg State University, Saint-Petersburg 198504, Russia
| | - Robert A Evarestov
- Institute of Chemistry, Saint-Petersburg State University, Saint-Petersburg 198504, Russia
| | - Sergey P Tunik
- Institute of Chemistry, Saint-Petersburg State University, Saint-Petersburg 198504, Russia
| |
Collapse
|
16
|
Perrin J, Capitao M, Mougin-Degraef M, Guérard F, Faivre-Chauvet A, Rbah-Vidal L, Gaschet J, Guilloux Y, Kraeber-Bodéré F, Chérel M, Barbet J. Cell Tracking in Cancer Immunotherapy. Front Med (Lausanne) 2020; 7:34. [PMID: 32118018 PMCID: PMC7033605 DOI: 10.3389/fmed.2020.00034] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 01/23/2020] [Indexed: 12/19/2022] Open
Abstract
The impressive development of cancer immunotherapy in the last few years originates from a more precise understanding of control mechanisms in the immune system leading to the discovery of new targets and new therapeutic tools. Since different stages of disease progression elicit different local and systemic inflammatory responses, the ability to longitudinally interrogate the migration and expansion of immune cells throughout the whole body will greatly facilitate disease characterization and guide selection of appropriate treatment regiments. While using radiolabeled white blood cells to detect inflammatory lesions has been a classical nuclear medicine technique for years, new non-invasive methods for monitoring the distribution and migration of biologically active cells in living organisms have emerged. They are designed to improve detection sensitivity and allow for a better preservation of cell activity and integrity. These methods include the monitoring of therapeutic cells but also of all cells related to a specific disease or therapeutic approach. Labeling of therapeutic cells for imaging may be performed in vitro, with some limitations on sensitivity and duration of observation. Alternatively, in vivo cell tracking may be performed by genetically engineering cells or mice so that may be revealed through imaging. In addition, SPECT or PET imaging based on monoclonal antibodies has been used to detect tumors in the human body for years. They may be used to detect and quantify the presence of specific cells within cancer lesions. These methods have been the object of several recent reviews that have concentrated on technical aspects, stressing the differences between direct and indirect labeling. They are briefly described here by distinguishing ex vivo (labeling cells with paramagnetic, radioactive, or fluorescent tracers) and in vivo (in vivo capture of injected radioactive, fluorescent or luminescent tracers, or by using labeled antibodies, ligands, or pre-targeted clickable substrates) imaging methods. This review focuses on cell tracking in specific therapeutic applications, namely cell therapy, and particularly CAR (Chimeric Antigen Receptor) T-cell therapy, which is a fast-growing research field with various therapeutic indications. The potential impact of imaging on the progress of these new therapeutic modalities is discussed.
Collapse
Affiliation(s)
- Justine Perrin
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Marisa Capitao
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Marie Mougin-Degraef
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, University Hospital, Nantes, France
| | - François Guérard
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Alain Faivre-Chauvet
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, University Hospital, Nantes, France
| | - Latifa Rbah-Vidal
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Joëlle Gaschet
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Yannick Guilloux
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Françoise Kraeber-Bodéré
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, University Hospital, Nantes, France.,Nuclear Medicine, ICO Cancer Center, Saint-Herblain, France
| | - Michel Chérel
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Nuclear Medicine, ICO Cancer Center, Saint-Herblain, France
| | | |
Collapse
|
17
|
Grippin AJ, Wummer B, Wildes T, Dyson K, Trivedi V, Yang C, Sebastian M, Mendez-Gomez H, Padala S, Grubb M, Fillingim M, Monsalve A, Sayour EJ, Dobson J, Mitchell DA. Dendritic Cell-Activating Magnetic Nanoparticles Enable Early Prediction of Antitumor Response with Magnetic Resonance Imaging. ACS NANO 2019; 13:13884-13898. [PMID: 31730332 PMCID: PMC7182054 DOI: 10.1021/acsnano.9b05037] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Cancer vaccines initiate antitumor responses in a subset of patients, but the lack of clinically meaningful biomarkers to predict treatment response limits their development. Here, we design multifunctional RNA-loaded magnetic liposomes to initiate potent antitumor immunity and function as an early biomarker of treatment response. These particles activate dendritic cells (DCs) more effectively than electroporation, leading to superior inhibition of tumor growth in treatment models. Inclusion of iron oxide enhances DC transfection and enables tracking of DC migration with magnetic resonance imaging (MRI). We show that T2*-weighted MRI intensity in lymph nodes is a strong correlation of DC trafficking and is an early predictor of antitumor response. In preclinical tumor models, MRI-predicted "responders" identified 2 days after vaccination had significantly smaller tumors 2-5 weeks after treatment and lived 73% longer than MRI-predicted "nonresponders". These studies therefore provide a simple, scalable nanoparticle formulation to generate robust antitumor immune responses and predict individual treatment outcome with MRI.
Collapse
Affiliation(s)
- Adam J. Grippin
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, 1149 Newell Drive PO Box 10026, University of Florida, Gainesville, FL, USA 32610
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, 1275 Center Dr, University of Florida, Gainesville, FL, USA 32611-7011
| | - Brandon Wummer
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, 1149 Newell Drive PO Box 10026, University of Florida, Gainesville, FL, USA 32610
| | - Tyler Wildes
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, 1149 Newell Drive PO Box 10026, University of Florida, Gainesville, FL, USA 32610
| | - Kyle Dyson
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, 1149 Newell Drive PO Box 10026, University of Florida, Gainesville, FL, USA 32610
| | - Vrunda Trivedi
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, 1149 Newell Drive PO Box 10026, University of Florida, Gainesville, FL, USA 32610
| | - Changlin Yang
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, 1149 Newell Drive PO Box 10026, University of Florida, Gainesville, FL, USA 32610
| | - Mathew Sebastian
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, 1149 Newell Drive PO Box 10026, University of Florida, Gainesville, FL, USA 32610
| | - Hector Mendez-Gomez
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, 1149 Newell Drive PO Box 10026, University of Florida, Gainesville, FL, USA 32610
| | - Suraj Padala
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, 1149 Newell Drive PO Box 10026, University of Florida, Gainesville, FL, USA 32610
| | - Mackenzie Grubb
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, 1275 Center Dr, University of Florida, Gainesville, FL, USA 32611-7011
| | - Matthew Fillingim
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, 1149 Newell Drive PO Box 10026, University of Florida, Gainesville, FL, USA 32610
| | - Adam Monsalve
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, 1275 Center Dr, University of Florida, Gainesville, FL, USA 32611-7011
| | - Elias J. Sayour
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, 1149 Newell Drive PO Box 10026, University of Florida, Gainesville, FL, USA 32610
| | - Jon Dobson
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, 1275 Center Dr, University of Florida, Gainesville, FL, USA 32611-7011
- Department of Materials Science & Engineering, 100 Rhines Hall, University of Florida, Gainesville, FL, USA 32610
| | - Duane A. Mitchell
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, 1149 Newell Drive PO Box 10026, University of Florida, Gainesville, FL, USA 32610
| |
Collapse
|
18
|
Anna IM, Sathy BN, Ashokan A, Gowd GS, Ramachandran R, Kochugovindan Unni AK, Manohar M, Chulliyath D, Nair S, Bhakoo K, Koyakutty M. nCP:Fe—A Biomineral Magnetic Nanocontrast Agent for Tracking Implanted Stem Cells in Brain Using MRI. ACS APPLIED BIO MATERIALS 2019; 2:5390-5403. [DOI: 10.1021/acsabm.9b00709] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Ida M. Anna
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kochi, Kerala 682041, India
| | - Binulal N. Sathy
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kochi, Kerala 682041, India
| | - Anusha Ashokan
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kochi, Kerala 682041, India
| | - Genekehal Siddaramana Gowd
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kochi, Kerala 682041, India
| | - Ranjith Ramachandran
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kochi, Kerala 682041, India
| | | | - Maneesh Manohar
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kochi, Kerala 682041, India
| | - DeepthiMol Chulliyath
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kochi, Kerala 682041, India
| | - Shantikumar Nair
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kochi, Kerala 682041, India
| | - Kishore Bhakoo
- Singapore Bioimaging Consortium (SBIC), Agency for Science, Technology and Research (A*STAR), Helios, Singapore 138667, Singapore
| | - Manzoor Koyakutty
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kochi, Kerala 682041, India
| |
Collapse
|
19
|
Krekorian M, Fruhwirth GO, Srinivas M, Figdor CG, Heskamp S, Witney TH, Aarntzen EH. Imaging of T-cells and their responses during anti-cancer immunotherapy. Theranostics 2019; 9:7924-7947. [PMID: 31656546 PMCID: PMC6814447 DOI: 10.7150/thno.37924] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 09/30/2019] [Indexed: 12/23/2022] Open
Abstract
Immunotherapy has proven to be an effective approach in a growing number of cancers. Despite durable clinical responses achieved with antibodies targeting immune checkpoint molecules, many patients do not respond. The common denominator for immunotherapies that have successfully been introduced in the clinic is their potential to induce or enhance infiltration of cytotoxic T-cells into the tumour. However, in clinical research the molecules, cells and processes involved in effective responses during immunotherapy remain largely obscure. Therefore, in vivo imaging technologies that interrogate T-cell responses in patients represent a powerful tool to boost further development of immunotherapy. This review comprises a comprehensive analysis of the in vivo imaging technologies that allow the characterisation of T-cell responses induced by anti-cancer immunotherapy, with emphasis on technologies that are clinically available or have high translational potential. Throughout we discuss their respective strengths and weaknesses, providing arguments for selecting the optimal imaging options for future research and patient management.
Collapse
Affiliation(s)
- Massis Krekorian
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
- Department of Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Gilbert O. Fruhwirth
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, Kings' College London, London, United Kingdom
| | - Mangala Srinivas
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Carl G. Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Sandra Heskamp
- Department of Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Timothy H. Witney
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, Kings' College London, London, United Kingdom
| | - Erik H.J.G. Aarntzen
- Department of Radiology and Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| |
Collapse
|
20
|
Scheetz L, Park KS, Li Q, Lowenstein PR, Castro MG, Schwendeman A, Moon JJ. Engineering patient-specific cancer immunotherapies. Nat Biomed Eng 2019; 3:768-782. [PMID: 31406259 PMCID: PMC6783331 DOI: 10.1038/s41551-019-0436-x] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 07/03/2019] [Indexed: 02/06/2023]
Abstract
Research into the immunological processes implicated in cancer has yielded a basis for the range of immunotherapies that are now considered the fourth pillar of cancer treatment (alongside surgery, radiotherapy and chemotherapy). For some aggressive cancers, such as advanced non-small-cell lung carcinoma, combination immunotherapies have resulted in unprecedented treatment efficacy for responding patients, and have become frontline therapies. Individualized immunotherapy, enabled by the identification of patient-specific mutations, neoantigens and biomarkers, and facilitated by advances in genomics and proteomics, promises to broaden the responder patient population. In this Perspective, we give an overview of immunotherapies leveraging engineering approaches, including the design of biomaterials, delivery strategies and nanotechnology solutions, for the realization of individualized cancer treatments such as nanoparticle vaccines customized with neoantigens, cell therapies based on patient-derived dendritic cells and T cells, and combinations of theranostic strategies. Developments in precision cancer immunotherapy will increasingly rely on the adoption of engineering principles.
Collapse
Affiliation(s)
- Lindsay Scheetz
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Kyung Soo Park
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Qiao Li
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Pedro R Lowenstein
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Maria G Castro
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA.
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
21
|
Abstract
Viruses are causative agents for many diseases and infect all living organisms on the planet. Development of effective therapies has relied on our ability to isolate and culture viruses in vitro, allowing mechanistic studies and strategic interventions. While this reductionist approach is necessary, testing the relevance of in vitro findings often takes a very long time. New developments in imaging technologies are transforming our experimental approach where viral pathogenesis can be studied in vivo at multiple spatial and temporal resolutions. Here, we outline a vision of a top-down approach using noninvasive whole-body imaging as a guide for in-depth characterization of key tissues, physiologically relevant cell types, and pathways of spread to elucidate mechanisms of virus spread and pathogenesis. Tool development toward imaging of infectious diseases is expected to transform clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Pradeep D Uchil
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut 06510, USA; , , ,
| | - Kelsey A Haugh
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut 06510, USA; , , ,
| | - Ruoxi Pi
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut 06510, USA; , , ,
| | - Walther Mothes
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut 06510, USA; , , ,
| |
Collapse
|
22
|
Fath-Bayati L, Vasei M, Sharif-Paghaleh E. Optical fluorescence imaging with shortwave infrared light emitter nanomaterials for in vivo cell tracking in regenerative medicine. J Cell Mol Med 2019; 23:7905-7918. [PMID: 31559692 PMCID: PMC6850965 DOI: 10.1111/jcmm.14670] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 07/13/2019] [Accepted: 07/30/2019] [Indexed: 12/13/2022] Open
Abstract
In vivo tracking and monitoring of adoptive cell transfer has a distinct importance in cell‐based therapy. There are many imaging modalities for in vivo monitoring of biodistribution, viability and effectiveness of transferred cells. Some of these procedures are not applicable in the human body because of low sensitivity and high possibility of tissue damages. Shortwave infrared region (SWIR) imaging is a relatively new technique by which deep biological tissues can be potentially visualized with high resolution at cellular level. Indeed, scanning of the electromagnetic spectrum (beyond 1000 nm) of SWIR has a great potential to increase sensitivity and resolution of in vivo imaging for various human tissues. In this review, molecular imaging modalities used for monitoring of biodistribution and fate of administered cells with focusing on the application of non‐invasive optical imaging at shortwave infrared region are discussed in detail.
Collapse
Affiliation(s)
- Leyla Fath-Bayati
- Department of Tissue Engineering & Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran.,Department of Tissue Engineering, School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Mohammad Vasei
- Department of Tissue Engineering & Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran.,Cell-based Therapies Research Institute, Digestive Disease Research Institute (DDRI), Shariati Hospital, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Ehsan Sharif-Paghaleh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Imaging Chemistry and Biology, Faculty of Life Sciences and Medicine, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| |
Collapse
|
23
|
Sharifi S, Zununi Vahed S, Ahmadian E, Maleki Dizaj S, Abedi A, Hosseiniyan Khatibi SM, Samiei M. Stem Cell Therapy: Curcumin Does the Trick. Phytother Res 2019; 33:2927-2937. [DOI: 10.1002/ptr.6482] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 07/21/2019] [Accepted: 08/04/2019] [Indexed: 12/31/2022]
Affiliation(s)
- Simin Sharifi
- Dental and Periodontal Research CenterTabriz University of Medical Sciences Tabriz Iran
| | | | - Elham Ahmadian
- Kidney Research CenterTabriz University of Medical Sciences Tabriz Iran
| | - Solmaz Maleki Dizaj
- Dental and Periodontal Research CenterTabriz University of Medical Sciences Tabriz Iran
| | - Atefeh Abedi
- Faculty of DentistryTabriz University of Medical Sciences Tabriz Iran
| | | | - Mohammad Samiei
- Faculty of DentistryTabriz University of Medical Sciences Tabriz Iran
- Stem Cell Research CenterTabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
24
|
Wang C, Adams SR, Xu H, Zhu W, Ahrens ET. β‑Diketonate-Iron(III) Complex: A Versatile Fluorine-19 MRI Signal Enhancement Agent. ACS APPLIED BIO MATERIALS 2019; 2:3836-3842. [PMID: 33981964 DOI: 10.1021/acsabm.9b00455] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Fluorine-19 magnetic resonance imaging (MRI) has gained considerable momentum as a promising imaging modality for in vivo tracking of cellular therapies and as a diagnostic for inflammatory disease. To further the utility of this technique, we increase imaging probe sensitivity by merging paramagnetic metal chelates with aqueous perfluorocarbon (PFC) nanoemulsions. We prepared a highly fluorinated ferric tris(β-diketonate) chelate (MW = 1265.2 g/mol) at gram scale. This iron chelate is soluble in multiple PFC oils used for MRI and readily reduces the 19F longitudinal relaxation time (T 1) to <100 ms with modest line broadening and displays superior properties for 19F MRI applications. The sensitivity enhancement by Fe(III) laden PFC nanoemulsion was confirmed in MRI phantom studies, where reduced T 1 speeds data acquisition thereby increasing the 19F image sensitivity per time via signal averaging. Additionally, 19F relaxivity of nanoemulsions incorporating other metal ions, including Gd, Er, Ho, Dy, Mn, Cr, and Ni, were evaluated. High-moment lanthanide ions, such as Gd(III), display severe line broadening, but other ions [e.g., Ho(III)] induce pseudocontact chemical shifts (up to 0.5 ppm) of 19F in nanoemulsion, which makes them potentially useful for multichromatic 19F imaging. Formulated nanoemulsions have a shelf life >200 days. Free β-diketonate or its iron complex in formed PFC nanoemulsion did not induce cytotoxicity in intracellularly labeled macrophages. Overall, ferric tris(β-diketonate) chelate provides a scalable approach for boosting sensitivity of PFC-based 19F MRI probes. More generally, it can functionalize PFC oil, whose chemical modification remains challenging.
Collapse
Affiliation(s)
- Chao Wang
- Department of Radiology, University of California, San Diego, La Jolla, California 92093, United States
| | - Stephen R Adams
- Department of Pharmacology, University of California, San Diego, La Jolla, California 92093, United States
| | - Hongyan Xu
- Department of Radiology, University of California, San Diego, La Jolla, California 92093, United States
| | - Wenlian Zhu
- Department of Radiology, University of California, San Diego, La Jolla, California 92093, United States
| | - Eric T Ahrens
- Department of Radiology, University of California, San Diego, La Jolla, California 92093, United States
| |
Collapse
|
25
|
Koshkina O, Lajoinie G, Bombelli FB, Swider E, Cruz LJ, White PB, Schweins R, Dolen Y, van Dinther EAW, van Riessen NK, Rogers SE, Fokkink R, Voets IK, van Eck ERH, Heerschap A, Versluis M, de Korte CL, Figdor CG, de Vries IJM, Srinivas M. Multicore Liquid Perfluorocarbon-Loaded Multimodal Nanoparticles for Stable Ultrasound and 19F MRI Applied to In Vivo Cell Tracking. ADVANCED FUNCTIONAL MATERIALS 2019; 29:1806485. [PMID: 32132881 PMCID: PMC7056356 DOI: 10.1002/adfm.201806485] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Indexed: 05/22/2023]
Abstract
Ultrasound is the most commonly used clinical imaging modality. However, in applications requiring cell-labeling, the large size and short active lifetime of ultrasound contrast agents limit their longitudinal use. Here, 100 nm radius, clinically applicable, polymeric nanoparticles containing a liquid perfluorocarbon, which enhance ultrasound contrast during repeated ultrasound imaging over the course of at least 48 h, are described. The perfluorocarbon enables monitoring the nanoparticles with quantitative 19F magnetic resonance imaging, making these particles effective multimodal imaging agents. Unlike typical core-shell perfluorocarbon-based ultrasound contrast agents, these nanoparticles have an atypical fractal internal structure. The nonvaporizing highly hydrophobic perfluorocarbon forms multiple cores within the polymeric matrix and is, surprisingly, hydrated with water, as determined from small-angle neutron scattering and nuclear magnetic resonance spectroscopy. Finally, the nanoparticles are used to image therapeutic dendritic cells with ultrasound in vivo, as well as with 19F MRI and fluorescence imaging, demonstrating their potential for long-term in vivo multimodal imaging.
Collapse
Affiliation(s)
- Olga Koshkina
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands; Physical Chemistry of Polymers, Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Guillaume Lajoinie
- Physics of Fluids Group, Technical Medical (TechMed) Centre and MESA+ Institute for, Nanotechnology, University of Twente, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands
| | - Francesca Baldelli Bombelli
- Laboratory of Supramolecular and BioNano Materials, (SupraBioNanoLab), Department of Chemistry, Materials, and Chemical Engineering, "Giulio Natta,", Politecnico di Milano, Via Luigi Mancinelli 7, 20131 Milan, Italy
| | - Edyta Swider
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Luis J Cruz
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Centre, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Paul B White
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Ralf Schweins
- Institut Laue - Langevin, DS/LSS, 71 Avenue des Martyrs, CS 20 156, 38042 Grenoble CEDEX 9, France
| | - Yusuf Dolen
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Eric A W van Dinther
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - N Koen van Riessen
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Sarah E Rogers
- ISIS Pulsed Neutron and Muon Source, Science and Technology Facilities Council, Rutherford Appleton Laboratory, Harwell, Oxford OX11 0QX, UK
| | - Remco Fokkink
- Department of Agrotechnology and Food Sciences, Physical Chemistry and Soft Matter, Wageningen University, 6708 WE, Wageningen, Netherlands
| | - Ilja K Voets
- Laboratory of Self-Organizing Soft Matter, Laboratory of Macromolecular and Organic Chemistry, Department of Chemical Engineering and Chemistry and Institute for Complex Molecular Systems, Eindhoven University of Technology, De Rondom 70, 5612 AP, Eindhoven, The Netherlands
| | - Ernst R H van Eck
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Arend Heerschap
- Department of Radiology and Nuclear Medicine, Radboudumc, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Michel Versluis
- Physics of Fluids Group, Technical Medical (TechMed) Centre and MESA+ Institute for, Nanotechnology, University of Twente, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands
| | - Chris L de Korte
- Physics of Fluids Group, Technical Medical (TechMed) Centre and MESA+ Institute for, Nanotechnology, University of Twente, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands; Department of Radiology and Nuclear Medicine, Radboudumc, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Mangala Srinivas
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| |
Collapse
|
26
|
Li W, Song W, Chen B, Matcher SJ. Superparamagnetic graphene quantum dot as a dual-modality contrast agent for confocal fluorescence microscopy and magnetomotive optical coherence tomography. JOURNAL OF BIOPHOTONICS 2019; 12:e201800219. [PMID: 30191684 DOI: 10.1002/jbio.201800219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 09/04/2018] [Indexed: 05/03/2023]
Abstract
A magnetic graphene quantum dot (MGQD) nanoparticle, synthesized by hydrothermally reducing and cutting graphene oxide-iron oxide sheet, was demonstrated to possess the capabilities of simultaneous confocal fluorescence and magnetomotive optical coherence tomography (MMOCT) imaging. This MGQD shows low toxicity, significant tunable blue fluorescence and superparamagnetism, which can thus be used as a dual-modality contrast agent for confocal fluorescence microscopy (CFM) and MMOCT. The feasibility of applying MGQD as a tracer of cells is shown by imaging and visualizing MGQD labeled cells using CFM and our in-house MMOCT. Since MMOCT and CFM can offer anatomical structure and intracellular details, respectively, the MGQD for cell tracking could provide a more comprehensive diagnosis.
Collapse
Affiliation(s)
- Wei Li
- Department of Electronic and Electrical Engineering, The University of Sheffield, Sheffield, UK
| | - Wenxing Song
- Department of Chemical and Biological Engineering, The University of Sheffield, Sheffield, UK
| | - Biqiong Chen
- School of Mechanical and Aerospace Engineering, Queen's University Belfast, Belfast, UK
| | - Stephen J Matcher
- Department of Electronic and Electrical Engineering, The University of Sheffield, Sheffield, UK
| |
Collapse
|
27
|
Madanayake NH, Rienzie R, Adassooriya NM. Nanoparticles in Nanotheranostics Applications. Nanotheranostics 2019. [DOI: 10.1007/978-3-030-29768-8_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
28
|
Affiliation(s)
- Zijuan Hai
- 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
| | - 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
| |
Collapse
|
29
|
Swider E, Koshkina O, Tel J, Cruz LJ, de Vries IJM, Srinivas M. Customizing poly(lactic-co-glycolic acid) particles for biomedical applications. Acta Biomater 2018; 73:38-51. [PMID: 29653217 DOI: 10.1016/j.actbio.2018.04.006] [Citation(s) in RCA: 185] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 03/28/2018] [Accepted: 04/03/2018] [Indexed: 12/22/2022]
Abstract
Nano- and microparticles have increasingly widespread applications in nanomedicine, ranging from drug delivery to imaging. Poly(lactic-co-glycolic acid) (PLGA) particles are the most widely-applied type of particles due to their biocompatibility and biodegradability. Here, we discuss the preparation of PLGA particles, and various modifications to tailor particles for applications in biological systems. We highlight new preparation approaches, including microfluidics and PRINT method, and modifications of PLGA particles resulting in novel or responsive properties, such as Janus or upconversion particles. Finally, we describe how the preparation methods can- and should-be adapted to tailor the properties of particles for the desired biomedical application. Our aim is to enable researchers who work with PLGA particles to better appreciate the effects of the selected preparation procedure on the final properties of the particles and its biological implications. STATEMENT OF SIGNIFICANCE Nanoparticles are increasingly important in the field of biomedicine. Particles made of polymers are in the spotlight, due to their biodegradability, biocompatibility, versatility. In this review, we aim to discuss the range of formulation techniques, manipulations, and applications of poly(lactic-co-glycolic acid) (PLGA) particles, to enable a researcher to effectively select or design the optimal particles for their application. We describe the various techniques of PLGA particle synthesis and their impact on possible applications. We focus on recent developments in the field of PLGA particles, and new synthesis techniques that have emerged over the past years. Overall, we show how the chemistry of PLGA particles can be adapted to solve pressing biological needs.
Collapse
|
30
|
Abstract
"Although there is ample evidence that beneficial results can be obtained from the use of mesenchymal stem cells, several questions regarding their use remain to be answered. For many of these questions, preclinical models will be helpful, but the task of evaluating and implementing these findings for orthopaedic patients falls onto the shoulders of clinical researchers. Evaluation of these questions is a daunting, but such a challenge fits the concept of personalized medicine in today's medicine."
Collapse
|
31
|
Moonshi SS, Zhang C, Peng H, Puttick S, Rose S, Fisk NM, Bhakoo K, Stringer BW, Qiao GG, Gurr PA, Whittaker AK. A unique 19F MRI agent for the tracking of non phagocytic cells in vivo. NANOSCALE 2018; 10:8226-8239. [PMID: 29682654 DOI: 10.1039/c8nr00703a] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
There is currently intense interest in new methods for understanding the fate of therapeutically-relevant cells, such as mesenchymal stem cells (MSCs). The absence of a confounding background signal and consequent unequivocal assignment makes 19F MRI one of the most attractive modalities for the tracking of injected cells in vivo. We describe here the synthesis of novel partly-fluorinated polymeric nanoparticles with small size and high fluorine content as MRI agents. The polymers, constructed from perfluoropolyether methacrylate (PFPEMA) and oligo(ethylene glycol) methacrylate (OEGMA) have favourable cell uptake profiles and excellent MRI performance. To facilitate cell studies the polymer was further conjugated with a fluorescent dye creating a dual-modal imaging agent. The efficacy of labelling of MSCs was assessed using 19F NMR, flow cytometry and confocal microscopy. The labelling efficiency of 2.6 ± 0.1 × 1012 19F atoms per cell, and viability of >90% demonstrates high uptake and good tolerance by the cells. This loading translates to a minimum 19F MRI detection sensitivity of ∼7.4 × 103 cells per voxel. Importantly, preliminary in vivo data demonstrate that labelled cells can be readily detected within a short acquisition scan period (12 minutes). Hence, these copolymers show outstanding potential for 19F MRI cellular tracking and for quantification of non-phagocytic and therapeutically-relevant cells in vivo.
Collapse
Affiliation(s)
- Shehzahdi S Moonshi
- Australian Institute for Bioengineering and Nanotechnology and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, QLD 4072, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Helfen A, Roth J, Ng T, Eisenblaetter M. In Vivo Imaging of Pro- and Antitumoral Cellular Components of the Tumor Microenvironment. J Nucl Med 2018; 59:183-188. [PMID: 29217734 DOI: 10.2967/jnumed.117.198952] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 11/14/2017] [Indexed: 12/17/2022] Open
Abstract
Tumor development and growth, as well as metastatic spread, are strongly influenced by various, mostly innate, immune cells, which are recruited to the tumor site and driven to establish a specific tumor-supportive microenvironment. The contents of this microenvironment, such as myeloid cells, are a major factor in the overall prognosis of malignant disease, addressed by a constantly growing armament of therapeutic interventions targeting tumor-supportive immune cells. Current clinical imaging has long ignored the growing need for diagnostic approaches addressing these microenvironmental contents-approaches enabling a sensitive and specific classification of tumor immune crosstalk and the resulting tumor-associated immune cell activity. In this focus article we review the present status of, and promising developments in, the in vivo molecular imaging of tumor immune components designed to allow for inferences to be made on the cross-talk between tumor cells and the immune system. Current imaging modalities based on the infiltrating cell types are briefly discussed.
Collapse
Affiliation(s)
- Anne Helfen
- Department of Clinical Radiology, University Hospital Muenster, Muenster, Germany
| | - Johannes Roth
- Institute of Immunology, University Hospital Muenster, Muenster, Germany
| | - Tony Ng
- Richard Dimbleby Department of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College London, London, United Kingdom
- Breast Cancer Now Research Unit, Department of Research Oncology, Guy's Hospital, King's College London, London, United Kingdom
- UCL Cancer Institute, University College London, London, United Kingdom; and
| | - Michel Eisenblaetter
- Department of Clinical Radiology, University Hospital Muenster, Muenster, Germany
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, United Kingdom
| |
Collapse
|
33
|
Song G, Chen M, Zhang Y, Cui L, Qu H, Zheng X, Wintermark M, Liu Z, Rao J. Janus Iron Oxides @ Semiconducting Polymer Nanoparticle Tracer for Cell Tracking by Magnetic Particle Imaging. NANO LETTERS 2018; 18:182-189. [PMID: 29232142 PMCID: PMC5995329 DOI: 10.1021/acs.nanolett.7b03829] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Iron oxides nanoparticles tailored for magnetic particle imaging (MPI) have been synthesized, and their MPI signal intensity is three-times that of commercial MPI contrast (Ferucarbotran, also called Vivotrax) and seven-times that of MRI contrast (Feraheme) at the same Fe concentration. MPI tailored iron oxide nanoparticles were encapsulated with semiconducting polymers to produce Janus nanoparticles that possessed optical and magnetic properties for MPI and fluorescence imaging. Janus particles were applied to cancer cell labeling and in vivo tracking, and as few as 250 cells were imaged by MPI after implantation, corresponding to an amount of 7.8 ng of Fe. Comparison with MRI and fluorescence imaging further demonstrated the advantages of our Janus particles for MPI-super sensitivity, unlimited tissue penetration, and linear quantitativity.
Collapse
Affiliation(s)
- Guosheng Song
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, 1201 Welch Road, Stanford, California, 94305-5484, USA
| | - Min Chen
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, 1201 Welch Road, Stanford, California, 94305-5484, USA
| | - Yanrong Zhang
- Department of Radiology, Neuroimaging and Neurointervention Division Stanford University Hospital 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Liyang Cui
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, 1201 Welch Road, Stanford, California, 94305-5484, USA
| | - Haibo Qu
- Department of Radiology, Neuroimaging and Neurointervention Division Stanford University Hospital 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Xianchuang Zheng
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, 1201 Welch Road, Stanford, California, 94305-5484, USA
| | - Max Wintermark
- Department of Radiology, Neuroimaging and Neurointervention Division Stanford University Hospital 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Zhuang Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jianghong Rao
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, 1201 Welch Road, Stanford, California, 94305-5484, USA
| |
Collapse
|
34
|
Oyarce C, Cruz-Gomez S, Galvez-Cancino F, Vargas P, Moreau HD, Diaz-Valdivia N, Diaz J, Salazar-Onfray FA, Pacheco R, Lennon-Dumenil AM, Quest AFG, Lladser A. Caveolin-1 Expression Increases upon Maturation in Dendritic Cells and Promotes Their Migration to Lymph Nodes Thereby Favoring the Induction of CD8 + T Cell Responses. Front Immunol 2017; 8:1794. [PMID: 29326695 PMCID: PMC5733362 DOI: 10.3389/fimmu.2017.01794] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/30/2017] [Indexed: 12/14/2022] Open
Abstract
Dendritic cell (DC) trafficking from peripheral tissues to lymph nodes (LNs) is a key step required to initiate T cell responses against pathogens as well as tumors. In this context, cellular membrane protrusions and the actin cytoskeleton are essential to guide DC migration towards chemotactic signals. Caveolin-1 (CAV1) is a scaffolding protein that modulates signaling pathways leading to remodeling of the actin cytoskeleton and enhanced migration of cancer cells. However, whether CAV1 is relevant for DC function and specifically for DC migration to LNs is unknown. Here, we show that CAV1 expression is upregulated in DCs upon LPS- and TNF-α-induced maturation. CAV1 deficiency did not affect differentiation, maturation, or the ability of DCs to activate CD8+ T cells in vitro. However, CAV1-deficient (CAV1-/-) DCs displayed reduced in vivo trafficking to draining LNs in control and inflammatory conditions. In vitro, CAV1-/- DCs showed reduced directional migration in CCL21 gradients in transwell assays without affecting migration velocity in confined microchannels or three-dimensional collagen matrices. In addition, CAV1-/- DCs displayed reduced activation of the small GTPase Rac1, a regulator of actin cytoskeletal remodeling, and lower numbers of F-actin-forming protrusions. Furthermore, mice adoptively transferred with peptide-pulsed CAV1-/- DCs showed reduced CD8+ T cell responses and antitumor protection. Our results suggest that CAV1 promotes the activation of Rac1 and the formation of membrane protrusions that favor DC chemotactic trafficking toward LNs where they can initiate cytotoxic T cell responses.
Collapse
Affiliation(s)
- Cesar Oyarce
- Laboratory of Gene Immunotherapy, Fundación Ciencia & Vida, Santiago, Chile.,Laboratory of Cellular Communication, Advanced Center for Chronic Diseases (ACCDiS) and Center for Molecular Studies of the Cell (CEMC), Program in Cell and Molecular Biology, Faculty of Medicine, Biomedical Sciences Institute (ICBM), University of Chile, Santiago, Chile
| | - Sebastián Cruz-Gomez
- Laboratory of Gene Immunotherapy, Fundación Ciencia & Vida, Santiago, Chile.,Laboratory of Cellular Communication, Advanced Center for Chronic Diseases (ACCDiS) and Center for Molecular Studies of the Cell (CEMC), Program in Cell and Molecular Biology, Faculty of Medicine, Biomedical Sciences Institute (ICBM), University of Chile, Santiago, Chile
| | | | - Pablo Vargas
- Institut National de la Santé et de la Recherche Médicale Unité 144, Institut Curie/CNRS, Paris, France
| | - Hélène D Moreau
- Institut National de la Santé et de la Recherche Médicale Unité 932, Institut Curie/CNRS, Paris, France
| | - Natalia Diaz-Valdivia
- Laboratory of Cellular Communication, Advanced Center for Chronic Diseases (ACCDiS) and Center for Molecular Studies of the Cell (CEMC), Program in Cell and Molecular Biology, Faculty of Medicine, Biomedical Sciences Institute (ICBM), University of Chile, Santiago, Chile
| | - Jorge Diaz
- Laboratory of Cellular Communication, Advanced Center for Chronic Diseases (ACCDiS) and Center for Molecular Studies of the Cell (CEMC), Program in Cell and Molecular Biology, Faculty of Medicine, Biomedical Sciences Institute (ICBM), University of Chile, Santiago, Chile
| | - Flavio Andres Salazar-Onfray
- Program in Immunology, Faculty of Medicine, Biomedical Sciences Institute (ICBM), University of Chile, Santiago, Chile
| | - Rodrigo Pacheco
- Laboratory of Neuroimmunology, Fundación Ciencia & Vida, Santiago, Chile.,Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andres Bello, Santiago, Chile
| | - Ana Maria Lennon-Dumenil
- Institut National de la Santé et de la Recherche Médicale Unité 932, Institut Curie/CNRS, Paris, France
| | - Andrew F G Quest
- Laboratory of Cellular Communication, Advanced Center for Chronic Diseases (ACCDiS) and Center for Molecular Studies of the Cell (CEMC), Program in Cell and Molecular Biology, Faculty of Medicine, Biomedical Sciences Institute (ICBM), University of Chile, Santiago, Chile
| | - Alvaro Lladser
- Laboratory of Gene Immunotherapy, Fundación Ciencia & Vida, Santiago, Chile
| |
Collapse
|
35
|
Belyanina I, Kolovskaya O, Zamay S, Gargaun A, Zamay T, Kichkailo A. Targeted Magnetic Nanotheranostics of Cancer. Molecules 2017; 22:E975. [PMID: 28604617 PMCID: PMC6152710 DOI: 10.3390/molecules22060975] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/02/2017] [Accepted: 06/06/2017] [Indexed: 12/31/2022] Open
Abstract
Current advances in targeted magnetic nanotheranostics are summarized in this review. Unique structural, optical, electronic and thermal properties of magnetic materials in nanometer scale are attractive in the field of biomedicine. Magnetic nanoparticles functionalized with therapeutic molecules, ligands for targeted delivery, fluorescent and other chemical agents can be used for cancer diagnostic and therapeutic purposes. High selectivity, small size, and low immunogenicity of synthetic nucleic acid aptamers make them attractive delivery agents for therapeutic purposes. Properties, production and functionalization of magnetic nanoparticles and aptamers as ligands for targeted delivery are discussed herein. In recent years, magnetic nanoparticles have been widely used in diagnostic methods, such as scintigraphy, single photon emission computed tomography (SPECT), positron emission tomography (PET), magnetic resonance imaging (MRI), and Raman spectroscopy. Therapeutic purposes of magnetic nanoconstructions are also promising. They are used for effective drug delivery, magnetic mediated hypertermia, and megnetodynamic triggering of apoptosis. Thus, magnetic nanotheranostics opens a new venue for complex differential diagnostics, and therapy of metastatic cancer.
Collapse
Affiliation(s)
- Irina Belyanina
- Krasnoyarsk State Medical University named after prof. V.F. Voino-Yaseneckii, 660022 Krasnoyarsk, Russia.
| | - Olga Kolovskaya
- Krasnoyarsk State Medical University named after prof. V.F. Voino-Yaseneckii, 660022 Krasnoyarsk, Russia.
- Federal Research Center, KSC Siberian Branch of Russian Academy of Science, 660022 Krasnoyarsk, Russia.
| | - Sergey Zamay
- Federal Research Center, KSC Siberian Branch of Russian Academy of Science, 660022 Krasnoyarsk, Russia.
| | - Ana Gargaun
- Independent Researcher Vancouver, Vancouver, BC V6K 1C4, Canada.
| | - Tatiana Zamay
- Krasnoyarsk State Medical University named after prof. V.F. Voino-Yaseneckii, 660022 Krasnoyarsk, Russia.
- Federal Research Center, KSC Siberian Branch of Russian Academy of Science, 660022 Krasnoyarsk, Russia.
| | - Anna Kichkailo
- Krasnoyarsk State Medical University named after prof. V.F. Voino-Yaseneckii, 660022 Krasnoyarsk, Russia.
- Federal Research Center, KSC Siberian Branch of Russian Academy of Science, 660022 Krasnoyarsk, Russia.
| |
Collapse
|
36
|
Asiedu KO, Koyasu S, Szajek LP, Choyke PL, Sato N. Bone Marrow Cell Trafficking Analyzed by 89Zr-oxine Positron Emission Tomography in a Murine Transplantation Model. Clin Cancer Res 2017; 23:2759-2768. [PMID: 27965305 PMCID: PMC5457332 DOI: 10.1158/1078-0432.ccr-16-1561] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 11/29/2016] [Accepted: 12/08/2016] [Indexed: 01/01/2023]
Abstract
Purpose: The success of hematopoietic stem cell transplantation (HSCT) depends on donor cell homing to the bone marrow. However, there is no reliable method of noninvasively monitoring the kinetics and distribution of transferred cells. Using zirconium-89 (89Zr)-oxine cell labeling combined with PET imaging, we sought to visualize and quantify donor cell homing in a mouse bone marrow transplantation model.Experimental Design: The effect of 89Zr-oxine labeling on bone marrow cell viability and differentiation was evaluated in vitro89Zr-labeled bone marrow cells (2 × 107 cells, 16.6 kBq/106 cells) were transferred intravenously, and serial microPET images were obtained (n = 5). The effect of a CXCR4 inhibitor, plerixafor (5 mg/kg) and G-CSF (2.5 μg) on bone marrow homing and mobilization were examined (n = 4). Engraftment of the transferred 89Zr-labeled cells was evaluated (n = 3).Results:89Zr-oxine-labeled bone marrow cells showed delayed proliferation, but differentiated normally. Transferred bone marrow cells rapidly migrated to the bone marrow, spleen, and liver (n = 5). Approximately 36% of donor cells homed to the bone marrow within 4 hours, irrespective of prior bone marrow ablation. Inhibition of CXCR4 by plerixafor alone or with G-CSF significantly blocked the bone marrow homing (P < 0.0001, vs. nontreated, at 2 hours), confirming a crucial role of the CXCR4-CXCL12 system. Mobilization of approximately 0.64% of pretransplanted bone marrow cells induced a 3.8-fold increase of circulating bone marrow cells. 89Zr-labeled donor cells engrafted as well as nonlabeled cells.Conclusions:89Zr-oxine PET imaging reveals rapid bone marrow homing of transferred bone marrow cells without impairment of their stem cell functions, and thus, could provide useful information for optimizing HSCT. Clin Cancer Res; 23(11); 2759-68. ©2016 AACR.
Collapse
Affiliation(s)
| | - Sho Koyasu
- Molecular Imaging Program, NCI, NIH, Bethesda, Maryland
| | - Lawrence P Szajek
- Positron Emission Tomography Department, Warren Grant Magnuson Clinical Center, NIH, Bethesda, Maryland
| | | | - Noriko Sato
- Molecular Imaging Program, NCI, NIH, Bethesda, Maryland.
| |
Collapse
|
37
|
Meir R, Popovtzer R. Cell tracking using gold nanoparticles and computed tomography imaging. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 10. [PMID: 28544497 DOI: 10.1002/wnan.1480] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 04/04/2017] [Accepted: 04/18/2017] [Indexed: 12/28/2022]
Abstract
Cell-based therapies utilize transplantation of living cells with therapeutic traits to alleviate numerous diseases and disorders. The use of such biological agents is an attractive alternative for diseases that existing medicine cannot effectively treat. Although very promising, translating cell therapy to the clinic has proven to be challenging, due to inconsistent results in preclinical and clinical studies. To examine the underlying cause for these inconsistencies, it is crucial to noninvasively monitor the accuracy of cell injection, and cell survival and migration patterns. The combination of classical imaging techniques with cellular contrast agents-mainly nanotechnological-based-has enabled significant developments in cell-tracking methodologies. One novel methodology, based on computed tomography (CT) as an imaging modality and gold nanoparticles (AuNPs) as contrast agents, has recently gained interest for its clinical applicability and cost-effectiveness. Studies have shown that AuNPs can be used to efficiently label a variety of cell types, including stem cells and immune cells, without damaging their therapeutic efficacy. Successful in vivo experiments have demonstrated noninvasive, quantitative and longitudinal cell tracking with high sensitivity. This concept has the potential to be used not only as a research tool, but in clinical settings as well. WIREs Nanomed Nanobiotechnol 2018, 10:e1480. doi: 10.1002/wnan.1480 This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging.
Collapse
Affiliation(s)
- Rinat Meir
- Faculty of Engineering and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
| | - Rachela Popovtzer
- Faculty of Engineering and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
38
|
Li A, Wu Y, Tang F, Li W, Feng X, Yao Z. In Vivo Magnetic Resonance Imaging of CD8+ T Lymphocytes Recruiting to Glioblastoma in Mice. Cancer Biother Radiopharm 2017; 31:317-323. [PMID: 27831762 DOI: 10.1089/cbr.2016.2061] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Noninvasive in vivo tracking of adopted immune cells would help improve immunotherapy on glioblastoma. In this study, the authors tried to track adoptive CD8+ T lymphocytes in an in situ GL261 glioblastoma mouse model with magnetic resonance imaging (MRI). CD8+ T lymphocytes from spleen of preimmunized GL261 glioblastoma mice were labeled with superparamagnetic iron oxide, with polylysine as transfection agent. From Prussian blue staining, the labeling efficiency was 0.77% ± 0.06%, without altering cell viability and function. From anti-CD8, and anti-dextran staining, superparamagnetic iron oxide could be seen in the cytoplasm. In vitro imaging of agar gel mixtures with different concentrations of labeled CD8+ T lymphocytes was done with a 3.0T MR T2*WI sequence. Higher cell concentrations showed lower signal values. Twenty-four hours after tail vein injection of labeled and unlabeled CD8+ T lymphocytes, imaging of GL261 mice brain showed black spots at the periphery of the tumor in the labeled group only. Brain tumor pathology further verified infiltration of labeled CD8+ T lymphocytes in the tumor. Thus, preimmunized CD8+ T lymphocytes could be efficiently labeled with superparamagnetic iron oxide and tracked both in vitro and in vivo with 3.0T MRI.
Collapse
Affiliation(s)
- Anning Li
- 1 Department of Radiology, Qilu Hospital of Shandong University , Jinan, People's Republic of China
| | - Yue Wu
- 2 Department of Radiology, Fudan University , Shanghai, People's Republic of China
| | - Feng Tang
- 3 Department of Radiology, Pathology, Huashan Hospital, Fudan University , Shanghai, People's Republic of China
| | - Wei Li
- 3 Department of Radiology, Pathology, Huashan Hospital, Fudan University , Shanghai, People's Republic of China
| | - Xiaoyuan Feng
- 2 Department of Radiology, Fudan University , Shanghai, People's Republic of China
| | - Zhenwei Yao
- 2 Department of Radiology, Fudan University , Shanghai, People's Republic of China
| |
Collapse
|
39
|
Ramamonjisoa N, Ackerstaff E. Characterization of the Tumor Microenvironment and Tumor-Stroma Interaction by Non-invasive Preclinical Imaging. Front Oncol 2017; 7:3. [PMID: 28197395 PMCID: PMC5281579 DOI: 10.3389/fonc.2017.00003] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/05/2017] [Indexed: 12/13/2022] Open
Abstract
Tumors are often characterized by hypoxia, vascular abnormalities, low extracellular pH, increased interstitial fluid pressure, altered choline-phospholipid metabolism, and aerobic glycolysis (Warburg effect). The impact of these tumor characteristics has been investigated extensively in the context of tumor development, progression, and treatment response, resulting in a number of non-invasive imaging biomarkers. More recent evidence suggests that cancer cells undergo metabolic reprograming, beyond aerobic glycolysis, in the course of tumor development and progression. The resulting altered metabolic content in tumors has the ability to affect cell signaling and block cellular differentiation. Additional emerging evidence reveals that the interaction between tumor and stroma cells can alter tumor metabolism (leading to metabolic reprograming) as well as tumor growth and vascular features. This review will summarize previous and current preclinical, non-invasive, multimodal imaging efforts to characterize the tumor microenvironment, including its stromal components and understand tumor-stroma interaction in cancer development, progression, and treatment response.
Collapse
Affiliation(s)
- Nirilanto Ramamonjisoa
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ellen Ackerstaff
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
40
|
Harrison-Brown M, Liu GJ, Banati R. Checkpoints to the Brain: Directing Myeloid Cell Migration to the Central Nervous System. Int J Mol Sci 2016; 17:E2030. [PMID: 27918464 PMCID: PMC5187830 DOI: 10.3390/ijms17122030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/23/2016] [Accepted: 11/25/2016] [Indexed: 12/13/2022] Open
Abstract
Myeloid cells are a unique subset of leukocytes with a diverse array of functions within the central nervous system during health and disease. Advances in understanding of the unique properties of these cells have inspired interest in their use as delivery vehicles for therapeutic genes, proteins, and drugs, or as "assistants" in the clean-up of aggregated proteins and other molecules when existing drainage systems are no longer adequate. The trafficking of myeloid cells from the periphery to the central nervous system is subject to complex cellular and molecular controls with several 'checkpoints' from the blood to their destination in the brain parenchyma. As important components of the neurovascular unit, the functional state changes associated with lineage heterogeneity of myeloid cells are increasingly recognized as important for disease progression. In this review, we discuss some of the cellular elements associated with formation and function of the neurovascular unit, and present an update on the impact of myeloid cells on central nervous system (CNS) diseases in the laboratory and the clinic. We then discuss emerging strategies for harnessing the potential of site-directed myeloid cell homing to the CNS, and identify promising avenues for future research, with particular emphasis on the importance of untangling the functional heterogeneity within existing myeloid subsets.
Collapse
Affiliation(s)
- Meredith Harrison-Brown
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Health Sciences, The University of Sydney, Sydney, NSW 2141, Australia.
- Australian Nuclear Science and Technology Organisation, Sydney, NSW 2234, Australia.
| | - Guo-Jun Liu
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Health Sciences, The University of Sydney, Sydney, NSW 2141, Australia.
- Australian Nuclear Science and Technology Organisation, Sydney, NSW 2234, Australia.
| | - Richard Banati
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Health Sciences, The University of Sydney, Sydney, NSW 2141, Australia.
- Australian Nuclear Science and Technology Organisation, Sydney, NSW 2234, Australia.
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
41
|
Dunn-Pirio AM, Vlahovic G. Immunotherapy approaches in the treatment of malignant brain tumors. Cancer 2016; 123:734-750. [PMID: 27875627 DOI: 10.1002/cncr.30371] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 07/16/2016] [Accepted: 09/01/2016] [Indexed: 12/28/2022]
Abstract
Glioblastoma is the most common malignant primary brain tumor. Despite standard-of-care treatment, consisting of maximal surgical resection followed by chemoradiation, both morbidity and mortality associated with this disease remain very poor. Therefore, there is an urgent need for more efficacious and well tolerated therapies. Advancing knowledge of the intricate interplay between malignant gliomas and the immune system, coupled with the recent launch of immunotherapy research for other cancers, has led to a veritable increase in immunotherapy investigation for glioblastoma and other malignant gliomas. This clinical review highlights the recent breakthroughs in cancer immunotherapy and the complex correlation of the immune system with primary brain tumors, with special attention to multiple immunotherapy modalities currently being investigated for malignant glioma, including peptide vaccines, dendritic cell vaccines, oncolytic viruses, chimeric T-cell receptors, and checkpoint inhibitors. Cancer 2017;123:734-50. © 2016 American Cancer Society.
Collapse
Affiliation(s)
- Anastasie M Dunn-Pirio
- The Preston Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
| | - Gordana Vlahovic
- The Preston Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
42
|
Ma Y, Mou Q, Wang D, Zhu X, Yan D. Dendritic Polymers for Theranostics. Theranostics 2016; 6:930-47. [PMID: 27217829 PMCID: PMC4876620 DOI: 10.7150/thno.14855] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 03/09/2016] [Indexed: 12/14/2022] Open
Abstract
Dendritic polymers are highly branched polymers with controllable structures, which possess a large population of terminal functional groups, low solution or melt viscosity, and good solubility. Their size, degree of branching and functionality can be adjusted and controlled through the synthetic procedures. These tunable structures correspond to application-related properties, such as biodegradability, biocompatibility, stimuli-responsiveness and self-assembly ability, which are the key points for theranostic applications, including chemotherapeutic theranostics, biotherapeutic theranostics, phototherapeutic theranostics, radiotherapeutic theranostics and combined therapeutic theranostics. Up to now, significant progress has been made for the dendritic polymers in solving some of the fundamental and technical questions toward their theranostic applications. In this review, we briefly summarize how to control the structures of dendritic polymers, the theranostics-related properties derived from their structures and their theranostics-related applications.
Collapse
Affiliation(s)
- Yuan Ma
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Quanbing Mou
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Dali Wang
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Deyue Yan
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| |
Collapse
|
43
|
Pongrac IM, Pavičić I, Milić M, Brkić Ahmed L, Babič M, Horák D, Vinković Vrček I, Gajović S. Oxidative stress response in neural stem cells exposed to different superparamagnetic iron oxide nanoparticles. Int J Nanomedicine 2016; 11:1701-15. [PMID: 27217748 PMCID: PMC4853020 DOI: 10.2147/ijn.s102730] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Biocompatibility, safety, and risk assessments of superparamagnetic iron oxide nanoparticles (SPIONs) are of the highest priority in researching their application in biomedicine. One improvement in the biological properties of SPIONs may be achieved by different functionalization and surface modifications. This study aims to investigate how a different surface functionalization of SPIONs – uncoated, coated with d-mannose, or coated with poly-l-lysine – affects biocompatibility. We sought to investigate murine neural stem cells (NSCs) as important model system for regenerative medicine. To reveal the possible mechanism of toxicity of SPIONs on NSCs, levels of reactive oxygen species, intracellular glutathione, mitochondrial membrane potential, cell-membrane potential, DNA damage, and activities of SOD and GPx were examined. Even in cases where reactive oxygen species levels were significantly lowered in NSCs exposed to SPIONs, we found depleted intracellular glutathione levels, altered activities of SOD and GPx, hyperpolarization of the mitochondrial membrane, dissipated cell-membrane potential, and increased DNA damage, irrespective of the surface coating applied for SPION stabilization. Although surface coating should prevent the toxic effects of SPIONs, our results showed that all of the tested SPION types affected the NSCs similarly, indicating that mitochondrial homeostasis is their major cellular target. Despite the claimed biomedical benefits of SPIONs, the refined determination of their effects on various cellular functions presented in this work highlights the need for further safety evaluations. This investigation helps to fill the knowledge gaps on the criteria that should be considered in evaluating the biocompatibility and safety of novel nanoparticles.
Collapse
Affiliation(s)
- Igor M Pongrac
- School of Medicine, Croatian Institute for Brain Research, University of Zagreb, Zagreb, Croatia
| | - Ivan Pavičić
- Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Mirta Milić
- Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Lada Brkić Ahmed
- School of Medicine, Croatian Institute for Brain Research, University of Zagreb, Zagreb, Croatia
| | - Michal Babič
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Daniel Horák
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | | - Srećko Gajović
- School of Medicine, Croatian Institute for Brain Research, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
44
|
Vu-Quang H, Vinding MS, Xia D, Nielsen T, Ullisch MG, Dong M, Nielsen NC, Kjems J. Chitosan-coated poly(lactic-co-glycolic acid) perfluorooctyl bromide nanoparticles for cell labeling in 19F magnetic resonance imaging. Carbohydr Polym 2016; 136:936-44. [DOI: 10.1016/j.carbpol.2015.09.076] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 09/04/2015] [Accepted: 09/23/2015] [Indexed: 12/11/2022]
|
45
|
Stem Cell Tracking with Nanoparticles for Regenerative Medicine Purposes: An Overview. Stem Cells Int 2015; 2016:7920358. [PMID: 26839568 PMCID: PMC4709786 DOI: 10.1155/2016/7920358] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 10/07/2015] [Accepted: 10/11/2015] [Indexed: 02/07/2023] Open
Abstract
Accurate and noninvasive stem cell tracking is one of the most important needs in regenerative medicine to determine both stem cell destinations and final differentiation fates, thus allowing a more detailed picture of the mechanisms involved in these therapies.
Given the great importance and advances in the field of nanotechnology for stem cell imaging, currently, several nanoparticles have become standardized products and have been undergoing fast commercialization. This review has been intended to summarize the current use of different engineered nanoparticles in stem cell tracking for regenerative medicine purposes, in particular by detailing their main features and exploring their biosafety aspects, the first step for clinical application. Moreover, this review has summarized the advantages and applications of stem cell tracking with nanoparticles in experimental and preclinical studies and investigated present limitations for their employment in the clinical setting.
Collapse
|
46
|
Siloxane Nanoprobes for Labeling and Dual Modality Functional Imaging of Neural Stem Cells. Ann Biomed Eng 2015; 44:816-27. [PMID: 26597417 DOI: 10.1007/s10439-015-1514-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 11/13/2015] [Indexed: 12/15/2022]
Abstract
Cell therapy represents a promising therapeutic for a myriad of medical conditions, including cancer, traumatic brain injury, and cardiovascular disease among others. A thorough understanding of the efficacy and cellular dynamics of these therapies necessitates the ability to non-invasively track cells in vivo. Magnetic resonance imaging (MRI) provides a platform to track cells as a non-invasive modality with superior resolution and soft tissue contrast. We recently reported a new nanoprobe platform for cell labeling and imaging using fluorophore doped siloxane core nanoemulsions as dual modality ((1)H MRI/Fluorescence), dual-functional (oximetry/detection) nanoprobes. Here, we successfully demonstrate the labeling, dual-modality imaging, and oximetry of neural progenitor/stem cells (NPSCs) in vitro using this platform. Labeling at a concentration of 10 μL/10(4) cells with a 40%v/v polydimethylsiloxane core nanoemulsion, doped with rhodamine, had minimal effect on viability, no effect on migration, proliferation and differentiation of NPSCs and allowed for unambiguous visualization of labeled NPSCs by (1)H MR and fluorescence and local pO2 reporting by labeled NPSCs. This new approach for cell labeling with a positive contrast (1)H MR probe has the potential to improve mechanistic knowledge of current therapies, and guide the design of future cell therapies due to its clinical translatability.
Collapse
|
47
|
Gallina C, Capelôa T, Saviozzi S, Accomasso L, Catalano F, Tullio F, Martra G, Penna C, Pagliaro P, Turinetto V, Giachino C. Human mesenchymal stem cells labelled with dye-loaded amorphous silica nanoparticles: long-term biosafety, stemness preservation and traceability in the beating heart. J Nanobiotechnology 2015; 13:77. [PMID: 26510588 PMCID: PMC4625930 DOI: 10.1186/s12951-015-0141-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 10/22/2015] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Treatment of myocardial infarction with mesenchymal stem cells (MSCs) has proven beneficial effects in both animal and clinical studies. Engineered silica nanoparticles (SiO2-NPs) have been extensively used as contrast agents in regenerative medicine, due to their resistance to degradation and ease of functionalization. However, there are still controversies on their effective biosafety on cellular systems. In this perspective, the aims of the present study are: 1) to deeply investigate the impact of amorphous 50 nm SiO2-NPs on viability and function of human bone marrow-derived MSCs (hMSCs); 2) to optimize a protocol of harmless hMSCs labelling and test its feasibility in a beating heart model. RESULTS Optimal cell labelling is obtained after 16 h exposure of hMSCs to fluorescent 50 nm SiO2-NPs (50 µg mL(-1)); interestingly, lysosomal activation consequent to NPs storage is not associated to oxidative stress. During prolonged culture hMSCs do not undergo cyto- or genotoxicity, preserve their proliferative potential and their stemness/differentiation properties. Finally, the bright fluorescence emitted by internalized SiO2-NPs allows both clear visualization of hMSCs in normal and infarcted rat hearts and ultrastructural analysis of cell engraftment inside myocardial tissue. CONCLUSIONS Overall, 50 nm SiO2-NPs display elevated compatibility with hMSCs in terms of lack of cyto- and genotoxicity and maintenance of important features of these cells. The demonstrated biosafety, combined with proper cell labelling and visualization in histological sections, make these SiO2-NPs optimal candidates for the purpose of stem cell tracking inside heart tissue.
Collapse
Affiliation(s)
- Clara Gallina
- Department of Clinical and Biological Sciences, University of Turin, 10, Regione Gonzole, CAP 10043, Orbassano, TO, Italy.
| | - Tânia Capelôa
- Department of Clinical and Biological Sciences, University of Turin, 10, Regione Gonzole, CAP 10043, Orbassano, TO, Italy.
| | - Silvia Saviozzi
- Department of Clinical and Biological Sciences, University of Turin, 10, Regione Gonzole, CAP 10043, Orbassano, TO, Italy.
| | - Lisa Accomasso
- Department of Clinical and Biological Sciences, University of Turin, 10, Regione Gonzole, CAP 10043, Orbassano, TO, Italy.
| | - Federico Catalano
- Department of Clinical and Biological Sciences, University of Turin, 10, Regione Gonzole, CAP 10043, Orbassano, TO, Italy. .,Department of Chemistry, Interdepartmental Centre "Nanostructured Interfaces and Surfaces", University of Turin, 7, Via P. Giuria, CAP 10125, Turin, Italy.
| | - Francesca Tullio
- Department of Clinical and Biological Sciences, University of Turin, 10, Regione Gonzole, CAP 10043, Orbassano, TO, Italy.
| | - Gianmario Martra
- Department of Chemistry, Interdepartmental Centre "Nanostructured Interfaces and Surfaces", University of Turin, 7, Via P. Giuria, CAP 10125, Turin, Italy.
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, 10, Regione Gonzole, CAP 10043, Orbassano, TO, Italy.
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, 10, Regione Gonzole, CAP 10043, Orbassano, TO, Italy.
| | - Valentina Turinetto
- Department of Clinical and Biological Sciences, University of Turin, 10, Regione Gonzole, CAP 10043, Orbassano, TO, Italy.
| | - Claudia Giachino
- Department of Clinical and Biological Sciences, University of Turin, 10, Regione Gonzole, CAP 10043, Orbassano, TO, Italy.
| |
Collapse
|
48
|
Srinivas M, Tel J, Schreibelt G, Bonetto F, Cruz LJ, Amiri H, Heerschap A, Figdor CG, de Vries IJM. PLGA-encapsulated perfluorocarbon nanoparticles for simultaneous visualization of distinct cell populations by 19F MRI. Nanomedicine (Lond) 2015; 10:2339-48. [DOI: 10.2217/nnm.15.76] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim: In vivo imaging using 19F MRI is advantageous, due to its ability to quantify cell numbers, but is limited for a lack of suitable labels. Here, we formulate two stable and clinically applicable labels for tracking two populations of primary human dendritic cells (DCs) simultaneously. Materials & methods: Plasmacytoid and myeloid DCs are able to take up sufficient nanoparticles (200 nm) for imaging (1012 19F's per cell), despite being relatively nonphagocytic. Results: Clinically relevant numbers of labeled DCs could be imaged in about 10 min, even on a clinical scanner. Conclusion: We demonstrate the use of perfluorocarbon nanoparticles for simultaneous 19F MRI of distinct cell populations in a clinical setting, without spectroscopic imaging.
Collapse
Affiliation(s)
- Mangala Srinivas
- Department of Tumor Immunology, & Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Jurjen Tel
- Department of Tumor Immunology, & Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Gerty Schreibelt
- Department of Tumor Immunology, & Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Fernando Bonetto
- Instituto de Física del Litoral (CONICET – UNL), Santa Fe, Argentina
| | - Luis-Javier Cruz
- Molecular Imaging, Leiden University Medical Center, Leiden, The Netherlands
| | - Houshang Amiri
- Department of Radiology, Radboud University Medical Center, Nijmegen, The Netherlands
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Arend Heerschap
- Department of Radiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, & Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Department of Tumor Immunology, & Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| |
Collapse
|
49
|
Sharma VK, Alipour A, Soran-Erdem Z, Aykut ZG, Demir HV. Highly monodisperse low-magnetization magnetite nanocubes as simultaneous T(1)-T(2) MRI contrast agents. NANOSCALE 2015; 7:10519-10526. [PMID: 26010145 DOI: 10.1039/c5nr00752f] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
We report the first study of highly monodisperse and crystalline iron oxide nanocubes with sub-nm controlled size distribution (9.7 ± 0.5 nm in size) that achieve simultaneous contrast enhancement in both T1- and T2-weighted magnetic resonance imaging (MRI). Here, we confirmed the magnetite structure of iron oxide nanocubes by X-ray diffraction (XRD), selected area electron diffraction (SAED) pattern, optical absorption and Fourier transformed infrared (FT-IR) spectra. These magnetite nanocubes exhibit superparamagnetic and paramagnetic behavior simultaneously by virtue of their finely controlled shape and size. The magnetic measurements reveal that the magnetic moment values are favorably much lower because of the small size and cubic shape of the nanoparticles, which results in an enhanced spin canting effect. As a proof-of-concept demonstration, we showed their potential as dual contrast agents for both T1- and T2-weighted MRI via phantom studies, in vivo imaging and relaxivity measurements. Therefore, these low-magnetization magnetite nanocubes, while being non-toxic and bio-compatible, hold great promise as excellent dual-mode T1 and T2 contrast agents for MRI.
Collapse
Affiliation(s)
- V K Sharma
- UNAM-Institute of Materials Science and Nanotechnology, National Magnetic Resonance Research Center (UMRAM), Department of Electrical and Electronics Engineering, Department of Physics, Department of Molecular Biology and Genetics, Bilkent University, Ankara, 06800, Turkey
| | | | | | | | | |
Collapse
|
50
|
Su CW, Chiang CS, Li WM, Hu SH, Chen SY. Multifunctional nanocarriers for simultaneous encapsulation of hydrophobic and hydrophilic drugs in cancer treatment. Nanomedicine (Lond) 2015; 9:1499-515. [PMID: 25253498 DOI: 10.2217/nnm.14.97] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Combination therapy for cancer patients is an important standard of care protocol because it can elicit synergistic therapeutic effects and reduce systemic toxicity by simultaneously modulating multiple cell-signaling pathways and overcoming multidrug resistance. Nanocarriers are expected to play a major role in delivering multiple drugs to tumor tissues by overcoming biological barriers. However, especially considering the different physical chemistry of chemotherapeutic drugs, it is highly desirable to develop a codelivery nanocarrier for controlled and targeted delivery of both hydrophobic and hydrophilic drugs. This review reports the recent developments in various combinational drug delivery systems and the simultaneous use of combinational drug delivery systems with functional agents.
Collapse
Affiliation(s)
- Chia-Wei Su
- Department of Materials Science & Engineering, National Chiao Tung University, Hsinchu, Taiwan
| | | | | | | | | |
Collapse
|