1
|
Astrelina TA, Brunchukov VA, Kodina GE, Bubenshchikov VB, Larenkov AA, Lunev AS, Petrosova KA, Rastorgueva AA, Kobzeva IV, Usupzhanova DY, Nikitina VA, Malsagova KA, Kulikova LI, Samoilov AS, Pustovoyt VI. Biodistribution of Mesenchymal Stromal Cells Labeled with [ 89Zr]Zr-Oxine in Local Radiation Injuries in Laboratory Animals. Molecules 2023; 28:7169. [PMID: 37894647 PMCID: PMC10609482 DOI: 10.3390/molecules28207169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/04/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Tracking the migration pathways of living cells after their introduction into a patient's body is a topical issue in the field of cell therapy. Questions related to studying the possibility of long-term intravital biodistribution of mesenchymal stromal cells in the body currently remain open. METHODS Forty-nine laboratory animals were used in the study. Modeling of local radiation injuries was carried out, and the dynamics of the distribution of mesenchymal stromal cells labeled with [89Zr]Zr-oxine in the rat body were studied. RESULTS the obtained results of the labelled cell distribution allow us to assume that this procedure could be useful for visualization of local radiation injury using positron emission tomography. However, further research is needed to confirm this assumption. CONCLUSIONS intravenous injection leads to the initial accumulation of cells in the lungs and their subsequent redistribution to the liver, spleen, and kidneys. When locally injected into tissues, mesenchymal stromal cells are not distributed systemically in significant quantities.
Collapse
Affiliation(s)
- Tatiana A. Astrelina
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| | - Vitaliy A. Brunchukov
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| | - Galina E. Kodina
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| | - Viktor B. Bubenshchikov
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| | - Anton A. Larenkov
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| | - Aleksandr S. Lunev
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| | - Kristina A. Petrosova
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| | - Anna A. Rastorgueva
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| | - Irina V. Kobzeva
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| | - Daria Y. Usupzhanova
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| | - Victoria A. Nikitina
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| | | | - Ludmila I. Kulikova
- Institute of Biomedical Chemistry, Biobanking Group, 119121 Moscow, Russia;
- Institute of Mathematical Problems of Biology RAS—The Branch of Keldysh Institute of Applied Mathematics of Russian Academy of Sciences, 142290 Pushchino, Russia
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 119991 Pushchino, Russia
| | - Alexander S. Samoilov
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| | - Vasiliy I. Pustovoyt
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123182 Moscow, Russia; (T.A.A.); (V.A.B.); (G.E.K.); (V.B.B.); (A.A.L.); (A.S.L.); (K.A.P.); (A.A.R.); (I.V.K.); (D.Y.U.); (V.A.N.); (A.S.S.); (V.I.P.)
| |
Collapse
|
2
|
Nuschke A, Sobey-Skelton C, Dawod B, Kelly B, Tremblay ML, Davis C, Rioux JA, Brewer K. Use of Magnetotactic Bacteria as an MRI Contrast Agent for In Vivo Tracking of Adoptively Transferred Immune Cells. Mol Imaging Biol 2023; 25:844-856. [PMID: 37715090 DOI: 10.1007/s11307-023-01849-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 09/17/2023]
Abstract
PURPOSE In vivo immune cell tracking using MRI can be a valuable tool for studying the mechanisms underlying successful cancer therapies. Current cell labeling methods using superparamagnetic iron oxide (SPIO) lack the persistence to track the fate and location of transplanted cells long-term. Magnetospirillum magneticum is a commercially available, iron-producing bacterium that can be taken up by and live harmoniously within mammalian cells as magneto-endosymbionts (MEs). MEs have shown promise as labeling agents for in vivo stem and cancer cell tracking but have yet to be evaluated in immune cells. This pilot study examined ME labeling in myeloid-derived suppressor cells (MDSCs), cytotoxic T lymphocytes (CTLs), and dendritic cells (DCs) and its effects on cell purity, function, and MRI contrast. PROCEDURES MDSCs, CTLs, and DCs were incubated with MEs at various ME labeling ratios (MLR), and various biological metrics and iron uptake were assessed. For in vivo imaging, MDSCs were labeled overnight with either MEs or SPIO (Molday ION Rhodamine B) and injected into C3 tumor-bearing mice via tail vein injection 24 days post-implant and scanned daily with MRI for 1 week to assess cellular quantification. RESULTS Following incubations, MDSCs contained > 0.6 pg Fe/cell. CTLs achieved Fe loading of < 0.5 pg/cell, and DCs achieved Fe loading of ~ 1.4 pg/cell. The suppressive functionality of MDSCs at 1000 MLR was not affected by ME labeling but was affected at 2000 MLR. Markers of CTL dysfunction were not markedly affected by ME labeling nor were DC markers. In vivo data demonstrated that the MDSCs labeled with MEs generated sufficient contrast to be detectable using TurboSPI, similar to SPIO-labeled cells. CONCLUSIONS Cells can be labeled with sufficient numbers of MEs to be detectable with MRI without compromising cell viability. Care must be taken at higher concentrations of MEs, which may affect some cell types' functional activity and/or morphology. Immune cells with minimal phagocytic behavior have much lower iron content per cell after incubation with MEs vs SPIO; however, MEs can successfully be used as a contrast agent for phagocytic immune cells.
Collapse
Affiliation(s)
- Andrea Nuschke
- Biomedical MRI Research Laboratory, IWK Health Centre, Halifax, NS, Canada
| | - Caitrin Sobey-Skelton
- Biomedical MRI Research Laboratory, IWK Health Centre, Halifax, NS, Canada
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | - Bassel Dawod
- Biomedical MRI Research Laboratory, IWK Health Centre, Halifax, NS, Canada
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | - Brianna Kelly
- Biomedical MRI Research Laboratory, IWK Health Centre, Halifax, NS, Canada
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | - Marie-Laurence Tremblay
- Biomedical MRI Research Laboratory, IWK Health Centre, Halifax, NS, Canada
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | - Christa Davis
- Biomedical MRI Research Laboratory, IWK Health Centre, Halifax, NS, Canada
| | - James A Rioux
- Biomedical MRI Research Laboratory, IWK Health Centre, Halifax, NS, Canada
- Department of Physics, Dalhousie University, Halifax, NS, Canada
- Department of Diagnostic Radiology, Dalhousie University, Halifax, NS, Canada
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, NS, Canada
| | - Kimberly Brewer
- Biomedical MRI Research Laboratory, IWK Health Centre, Halifax, NS, Canada.
- Department of Microbiology & Immunology, 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
|
3
|
Friberger I, Gontu V, Harris RA, Tran TA, Lundberg J, Holmin S. Phenotyping of Macrophages After Radiolabeling and Safety of Intra-arterial Transplantation Assessed by SPECT/CT and MRI. Cell Transplant 2023; 32:9636897231212780. [PMID: 38009543 PMCID: PMC10683405 DOI: 10.1177/09636897231212780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 10/04/2023] [Accepted: 10/23/2023] [Indexed: 11/29/2023] Open
Abstract
Cell therapy is an integral modality of regenerative medicine. Macrophages are known for their sensitivity to activation stimuli and capability to recruit other immune cells to the sites of injury and healing. In addition, the route of administration can impact engraftment and efficacy of cell therapy, and modern neuro-interventional techniques provide the possibility for selective intra-arterial (IA) delivery to the central nervous system (CNS) with very low risk. The effects of radiolabelling and catheter transport on differentially activated macrophages were evaluated. Furthermore, the safety of selective IA administration of these macrophages to the rabbit brain was assessed by single-photon emission computed tomography/computed tomography (SPECT/CT) and ultra-high-field (9.4 T) magnetic resonance imaging (MRI). Cells were successfully labeled with (111In)In-(oxinate)3 and passed through a microcatheter with preserved phenotype. No cells were retained in the healthy rabbit brain after IA administration, and no adverse events could be observed either 1 h (n = 6) or 24 h (n = 2) after cell administration. The procedure affected both lipopolysaccharide/gamma interferon (LPS/IFNγ) activated cells and interleukin 4 (IL4), interleukin 10 (IL10)/transforming growth factor beta 1 (TGFβ1) activated cells to some degree. The LPS/IFNγ activated cells had a significant increase in their phagocytotic function. Overall, the major impact on the cell phenotypes was due to the radiolabeling and not passage through the catheter. Unstimulated cells were substantially affected by both radiolabeling and catheter administration and are hence not suited for this procedure, while both activated macrophages retained their initial phenotypes. In conclusion, activated macrophages are suitable candidates for targeted IA administration without adverse effects on normal, healthy brain parenchyma.
Collapse
Affiliation(s)
- Ida Friberger
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Vamsi Gontu
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Robert A Harris
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Centre for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Thuy A Tran
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Radiopharmacy, Karolinska University Hospital, Stockholm, Sweden
| | - Johan Lundberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Staffan Holmin
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
4
|
Bulte JWM, Shakeri-Zadeh A. In Vivo MRI Tracking of Tumor Vaccination and Antigen Presentation by Dendritic Cells. Mol Imaging Biol 2022; 24:198-207. [PMID: 34581954 PMCID: PMC8477715 DOI: 10.1007/s11307-021-01647-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/21/2021] [Accepted: 08/26/2021] [Indexed: 01/24/2023]
Abstract
Cancer vaccination using tumor antigen-primed dendritic cells (DCs) was introduced in the clinic some 25 years ago, but the overall outcome has not lived up to initial expectations. In addition to the complexity of the immune response, there are many factors that determine the efficacy of DC therapy. These include accurate administration of DCs in the target tissue site without unwanted cell dispersion/backflow, sufficient numbers of tumor antigen-primed DCs homing to lymph nodes (LNs), and proper timing of immunoadjuvant administration. To address these uncertainties, proton (1H) and fluorine (19F) magnetic resonance imaging (MRI) tracking of ex vivo pre-labeled DCs can now be used to non-invasively determine the accuracy of therapeutic DC injection, initial DC dispersion, systemic DC distribution, and DC migration to and within LNs. Magnetovaccination is an alternative approach that tracks in vivo labeled DCs that simultaneously capture tumor antigen and MR contrast agent in situ, enabling an accurate quantification of antigen presentation to T cells in LNs. The ultimate clinical premise of MRI DC tracking would be to use changes in LN MRI signal as an early imaging biomarker to predict the efficacy of tumor vaccination and anti-tumor response long before treatment outcome becomes apparent, which may aid clinicians with interim treatment management.
Collapse
Affiliation(s)
- Jeff W M Bulte
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, MRB 659, 733 N. Broadway, MD, 21205, Baltimore, USA.
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, MRB 659, 733 N. Broadway, MD, 21205, Baltimore, USA.
- Department of Chemical & Biomolecular Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Ali Shakeri-Zadeh
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, MRB 659, 733 N. Broadway, MD, 21205, Baltimore, USA
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, MRB 659, 733 N. Broadway, MD, 21205, Baltimore, USA
| |
Collapse
|
5
|
Friberger I, Jussing E, Han J, Goos JACM, Siikanen J, Kaipe H, Lambert M, Harris RA, Samén E, Carlsten M, Holmin S, Tran TA. Optimisation of the Synthesis and Cell Labelling Conditions for [ 89Zr]Zr-oxine and [ 89Zr]Zr-DFO-NCS: a Direct In Vitro Comparison in Cell Types with Distinct Therapeutic Applications. Mol Imaging Biol 2021; 23:952-962. [PMID: 34231103 PMCID: PMC8578071 DOI: 10.1007/s11307-021-01622-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/29/2021] [Accepted: 06/03/2021] [Indexed: 11/13/2022]
Abstract
BACKGROUND There is a need to better characterise cell-based therapies in preclinical models to help facilitate their translation to humans. Long-term high-resolution tracking of the cells in vivo is often impossible due to unreliable methods. Radiolabelling of cells has the advantage of being able to reveal cellular kinetics in vivo over time. This study aimed to optimise the synthesis of the radiotracers [89Zr]Zr-oxine (8-hydroxyquinoline) and [89Zr]Zr-DFO-NCS (p-SCN-Bn-Deferoxamine) and to perform a direct comparison of the cell labelling efficiency using these radiotracers. PROCEDURES Several parameters, such as buffers, pH, labelling time and temperature, were investigated to optimise the synthesis of [89Zr]Zr-oxine and [89Zr]Zr-DFO-NCS in order to reach a radiochemical conversion (RCC) of >95 % without purification. Radio-instant thin-layer chromatography (iTLC) and radio high-performance liquid chromatography (radio-HPLC) were used to determine the RCC. Cells were labelled with [89Zr]Zr-oxine or [89Zr]Zr-DFO-NCS. The cellular retention of 89Zr and the labelling impact was determined by analysing the cellular functions, such as viability, proliferation, phagocytotic ability and phenotypic immunostaining. RESULTS The optimised synthesis of [89Zr]Zr-oxine and [89Zr]Zr-DFO-NCS resulted in straightforward protocols not requiring additional purification. [89Zr]Zr-oxine and [89Zr]Zr-DFO-NCS were synthesised with an average RCC of 98.4 % (n = 16) and 98.0 % (n = 13), respectively. Cell labelling efficiencies were 63.9 % (n = 35) and 70.2 % (n = 30), respectively. 89Zr labelling neither significantly affected the cell viability (cell viability loss was in the range of 1-8 % compared to its corresponding non-labelled cells, P value > 0.05) nor the cells' proliferation rate. The phenotype of human decidual stromal cells (hDSC) and phagocytic function of rat bone-marrow-derived macrophages (rMac) was somewhat affected by radiolabelling. CONCLUSIONS Our study demonstrates that [89Zr]Zr-oxine and [89Zr]Zr-DFO-NCS are equally effective in cell labelling. However, [89Zr]Zr-oxine was superior to [89Zr]Zr-DFO-NCS with regard to long-term stability, cellular retention, minimal variation between cell types and cell labelling efficiency.
Collapse
Affiliation(s)
- Ida Friberger
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Emma Jussing
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Radiopharmacy, Karolinska University Hospital, Stockholm, Sweden
| | - Jinming Han
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Centre for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Jeroen A C M Goos
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Radiopharmacy, Karolinska University Hospital, Stockholm, Sweden
| | - Jonathan Siikanen
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Helen Kaipe
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Mélanie Lambert
- Department of Medicine in Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Robert A Harris
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Centre for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Erik Samén
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Radiopharmacy, Karolinska University Hospital, Stockholm, Sweden
| | - Mattias Carlsten
- Department of Medicine in Huddinge, Karolinska Institutet, Stockholm, Sweden
- Center for Cell Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Stockholm, Sweden
| | - Staffan Holmin
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Thuy A Tran
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.
- Department of Radiopharmacy, Karolinska University Hospital, Stockholm, Sweden.
| |
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
|
Preclinical models and technologies to advance nanovaccine development. Adv Drug Deliv Rev 2021; 172:148-182. [PMID: 33711401 DOI: 10.1016/j.addr.2021.03.001] [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: 01/26/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022]
Abstract
The remarkable success of targeted immunotherapies is revolutionizing cancer treatment. However, tumor heterogeneity and low immunogenicity, in addition to several tumor-associated immunosuppression mechanisms are among the major factors that have precluded the success of cancer vaccines as targeted cancer immunotherapies. The exciting outcomes obtained in patients upon the injection of tumor-specific antigens and adjuvants intratumorally, reinvigorated interest in the use of nanotechnology to foster the delivery of vaccines to address cancer unmet needs. Thus, bridging nano-based vaccine platform development and predicted clinical outcomes the selection of the proper preclinical model will be fundamental. Preclinical models have revealed promising outcomes for cancer vaccines. However, only few cases were associated with clinical responses. This review addresses the major challenges related to the translation of cancer nano-based vaccines to the clinic, discussing the requirements for ex vivo and in vivo models of cancer to ensure the translation of preclinical success to patients.
Collapse
|
8
|
Chuntova P, Chow F, Watchmaker PB, Galvez M, Heimberger AB, Newell EW, Diaz A, DePinho RA, Li MO, Wherry EJ, Mitchell D, Terabe M, Wainwright DA, Berzofsky JA, Herold-Mende C, Heath JR, Lim M, Margolin KA, Chiocca EA, Kasahara N, Ellingson BM, Brown CE, Chen Y, Fecci PE, Reardon DA, Dunn GP, Liau LM, Costello JF, Wick W, Cloughesy T, Timmer WC, Wen PY, Prins RM, Platten M, Okada H. Unique challenges for glioblastoma immunotherapy-discussions across neuro-oncology and non-neuro-oncology experts in cancer immunology. Meeting Report from the 2019 SNO Immuno-Oncology Think Tank. Neuro Oncol 2021; 23:356-375. [PMID: 33367885 PMCID: PMC7992879 DOI: 10.1093/neuonc/noaa277] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cancer immunotherapy has made remarkable advances with over 50 separate Food and Drug Administration (FDA) approvals as first- or second-line indications since 2015. These include immune checkpoint blocking antibodies, chimeric antigen receptor-transduced T cells, and bispecific T-cell-engaging antibodies. While multiple cancer types now benefit from these immunotherapies, notable exceptions thus far include brain tumors, such as glioblastoma. As such, it seems critical to gain a better understanding of unique mechanistic challenges underlying the resistance of malignant gliomas to immunotherapy, as well as to acquire insights into the development of future strategies. An Immuno-Oncology Think Tank Meeting was held during the 2019 Annual Society for Neuro-Oncology Scientific Conference. Discussants in the fields of neuro-oncology, neurosurgery, neuro-imaging, medical oncology, and cancer immunology participated in the meeting. Sessions focused on topics such as the tumor microenvironment, myeloid cells, T-cell dysfunction, cellular engineering, and translational aspects that are critical and unique challenges inherent with primary brain tumors. In this review, we summarize the discussions and the key messages from the meeting, which may potentially serve as a basis for advancing the field of immune neuro-oncology in a collaborative manner.
Collapse
Affiliation(s)
- Pavlina Chuntova
- Department of Neurological Surgery, UCSF, San Francisco, California
| | - Frances Chow
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | | | - Mildred Galvez
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, Los Angeles, California
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Evan W Newell
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Aaron Diaz
- Department of Neurological Surgery, UCSF, San Francisco, California
| | - Ronald A DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ming O Li
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - E John Wherry
- Department of Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Duane Mitchell
- Department of Neurosurgery, University of Florida College of Medicine, Gainesville, Florida
| | - Masaki Terabe
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Derek A Wainwright
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Jay A Berzofsky
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | | | | | - Michael Lim
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kim A Margolin
- Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, California
| | - E Antonio Chiocca
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts
| | | | - Benjamin M Ellingson
- Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Christine E Brown
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Yvonne Chen
- Department of Microbiology, Immunology & Molecular Genetics, UCLA, Los Angeles, California
| | - Peter E Fecci
- Department of Neurosurgery, Duke University School of Medicine, Durham, North Carolina
| | - David A Reardon
- Department of Medicine/Medical Oncology, Harvard Medical School, Boston, Massachusetts
| | - Gavin P Dunn
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Linda M Liau
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, California
| | | | - Wolfgang Wick
- Department of Neurology, University Hospital Heidelberg, Heidelberg, Germany
| | - Timothy Cloughesy
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - William C Timmer
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Robert M Prins
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, Los Angeles, California.,Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Michael Platten
- Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany.,DKTK CCU Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hideho Okada
- Department of Neurological Surgery, UCSF, San Francisco, California.,Parker Institute for Cancer Immunotherapy, San Francisco, California
| |
Collapse
|
9
|
Kiraga Ł, Kucharzewska P, Strzemecki D, Rygiel TP, Król M. Non-radioactive imaging strategies for in vivo immune cell tracking. PHYSICAL SCIENCES REVIEWS 2021; 8:385-403. [PMID: 36975764 PMCID: PMC10037928 DOI: 10.1515/psr-2020-0205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
In vivo tracking of administered cells chosen for specific disease treatment may be conducted by diagnostic imaging techniques preceded by cell labeling with special contrast agents. The most commonly used agents are those with radioactive properties, however their use in research is often impossible. This review paper focuses on the essential aspect of cell tracking with the exclusion of radioisotope tracers, therefore we compare application of different types of non-radioactive contrast agents (cell tracers), methods of cell labeling and application of various techniques for cell tracking, which are commonly used in preclinical or clinical studies. We discuss diagnostic imaging methods belonging to three groups: (1) Contrast-enhanced X-ray imaging, (2) Magnetic resonance imaging, and (3) Optical imaging. In addition, we present some interesting data from our own research on tracking immune cell with the use of discussed methods. Finally, we introduce an algorithm which may be useful for researchers planning leukocyte targeting studies, which may help to choose the appropriate cell type, contrast agent and diagnostic technique for particular disease study.
Collapse
Affiliation(s)
- Łukasz Kiraga
- Department of Cancer Biology, Institute of Biology, Warsaw University of Life Sciences, 02-787 Warsaw, Poland
- Cellis AG, 80002 Zurich, Switzerland
| | - Paulina Kucharzewska
- Department of Cancer Biology, Institute of Biology, Warsaw University of Life Sciences, 02-787 Warsaw, Poland
- Cellis AG, 80002 Zurich, Switzerland
| | | | - Tomasz P. Rygiel
- Cellis AG, 80002 Zurich, Switzerland
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Magdalena Król
- Department of Cancer Biology, Institute of Biology, Warsaw University of Life Sciences, 02-787 Warsaw, Poland
- Cellis AG, 80002 Zurich, Switzerland
| |
Collapse
|
10
|
Carreira B, Acúrcio RC, Matos AI, Peres C, Pozzi S, Vaskovich‐Koubi D, Kleiner R, Bento M, Satchi‐Fainaro R, Florindo HF. Nanomedicines as Multifunctional Modulators of Melanoma Immune Microenvironment. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Barbara Carreira
- Research Institute for Medicines (iMed.ULisboa) Faculty of Pharmacy, University of Lisbon Av. Prof. Gama Pinto Lisboa 1649‐003 Portugal
| | - Rita C. Acúrcio
- Research Institute for Medicines (iMed.ULisboa) Faculty of Pharmacy, University of Lisbon Av. Prof. Gama Pinto Lisboa 1649‐003 Portugal
| | - Ana I. Matos
- Research Institute for Medicines (iMed.ULisboa) Faculty of Pharmacy, University of Lisbon Av. Prof. Gama Pinto Lisboa 1649‐003 Portugal
| | - Carina Peres
- Research Institute for Medicines (iMed.ULisboa) Faculty of Pharmacy, University of Lisbon Av. Prof. Gama Pinto Lisboa 1649‐003 Portugal
| | - Sabina Pozzi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine Tel Aviv University Tel Aviv 6997801 Israel
| | - Daniella Vaskovich‐Koubi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine Tel Aviv University Tel Aviv 6997801 Israel
| | - Ron Kleiner
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine Tel Aviv University Tel Aviv 6997801 Israel
| | - Mariana Bento
- Research Institute for Medicines (iMed.ULisboa) Faculty of Pharmacy, University of Lisbon Av. Prof. Gama Pinto Lisboa 1649‐003 Portugal
| | - Ronit Satchi‐Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine Tel Aviv University Tel Aviv 6997801 Israel
| | - Helena F. Florindo
- Research Institute for Medicines (iMed.ULisboa) Faculty of Pharmacy, University of Lisbon Av. Prof. Gama Pinto Lisboa 1649‐003 Portugal
| |
Collapse
|
11
|
Xie T, Chen X, Fang J, Xue W, Zhang J, Tong H, Liu H, Guo Y, Yang Y, Zhang W. Non-invasive monitoring of the kinetic infiltration and therapeutic efficacy of nanoparticle-labeled chimeric antigen receptor T cells in glioblastoma via 7.0-Tesla magnetic resonance imaging. Cytotherapy 2020; 23:211-222. [PMID: 33334686 DOI: 10.1016/j.jcyt.2020.10.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 10/28/2020] [Accepted: 10/30/2020] [Indexed: 01/15/2023]
Abstract
BACKGROUND AIMS Chimeric antigen receptor (CAR) T-cell therapy is a promising treatment strategy in solid tumors. In vivo cell tracking techniques can help us better understand the infiltration, persistence and therapeutic efficacy of CAR T cells. In this field, magnetic resonance imaging (MRI) can achieve high-resolution images of cells by using cellular imaging probes. MRI can also provide various biological information on solid tumors. METHODS The authors adopted the amino alcohol derivatives of glucose-coated nanoparticles, ultra-small superparamagnetic particles of iron oxide (USPIOs), to label CAR T cells for non-invasive monitoring of kinetic infiltration and persistence in glioblastoma (GBM). The specific targeting CARs included anti-human epidermal growth factor receptor variant III and IL13 receptor subunit alpha 2 CARs. RESULTS When using an appropriate concentration, USPIO labeling exerted no negative effects on the biological characteristics and killing efficiency of CAR T cells. Increasing hypointensity signals could be detected in GBM models by susceptibility-weighted imaging MRI ranging from 3 days to 14 days following the injection of USPIO-labeled CAR T cells. In addition, nanoparticles and CAR T cells were found on consecutive histopathological sections. Moreover, diffusion and perfusion MRI revealed significantly increased water diffusion and decreased vascular permeability on day 3 after treatment, which was simultaneously accompanied by a significant decrease in tumor cell proliferation and increase in intercellular tight junction on immunostaining sections. CONCLUSION These results establish an effective imaging technique that can track CAR T cells in GBM models and validate their early therapeutic effects, which may guide the evaluation of CAR T-cell therapies in solid tumors.
Collapse
Affiliation(s)
- Tian Xie
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China; Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, China
| | - Xiao Chen
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China; Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, China
| | - Jingqin Fang
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China; Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, China
| | - Wei Xue
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China; Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, China
| | - Junfeng Zhang
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China; Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, China
| | - Haipeng Tong
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China; Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, China
| | - Heng Liu
- Department of Radiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Yu Guo
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China; Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, China
| | - Yizeng Yang
- Department of Medicine, Division of Gastroenterology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
| | - Weiguo Zhang
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China; Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, China.
| |
Collapse
|
12
|
Man F, Khan AA, Carrascal-Miniño A, Blower PJ, T M de Rosales R. A kit formulation for the preparation of [ 89Zr]Zr(oxinate) 4 for PET cell tracking: White blood cell labelling and comparison with [ 111In]In(oxinate) 3. Nucl Med Biol 2020; 90-91:31-40. [PMID: 32979725 PMCID: PMC7116765 DOI: 10.1016/j.nucmedbio.2020.09.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/08/2020] [Accepted: 09/14/2020] [Indexed: 12/29/2022]
Abstract
BACKGROUND Advances in immunology and cell-based therapies are creating a need to track individual cell types, such as immune cells (neutrophils, eosinophils, chimeric antigen receptor (CAR) T cells, etc.) and stem cells. As the fate of administered cells remains largely unknown, nuclear imaging could determine the migration and survival of cells in patients. [89Zr]Zr(oxinate)4, or [89Zr]Zr-oxine, is a radiotracer for positron emission tomography (PET) that has been evaluated in preclinical models of cell tracking and could improve on [111In]In-oxine, the current gold standard radiotracer for cell tracking by scintigraphy and single-photon emission computed tomography (SPECT), because of the better sensitivity, spatial resolution and quantification of PET. However, a clinically usable formulation of [89Zr]Zr-oxine is lacking. This study demonstrates a 1-step procedure for preparing [89Zr]Zr-oxine and evaluates it against [111In]In-oxine in white blood cell (WBC) labelling. METHODS Commercial [89Zr]Zr-oxalate was added to a formulation containing oxine, a buffering agent, a base and a surfactant or organic solvent. WBC isolated from 10 human volunteers were radiolabelled with [89Zr]Zr-oxine following a clinical radiolabelling protocol. Labelling efficiency, cell viability, chemotaxis and DNA damage were evaluated in vitro, in an intra-individual comparison against [111In]In-oxine. RESULTS An optimised formulation of [89Zr]Zr-oxine containing oxine, polysorbate 80 and 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) was developed. This enabled 1-step radiolabelling of oxine with commercial [89Zr]Zr-oxalate (0.1-25 MBq) in 5 min and radiotracer stability for 1 week. WBC labelling efficiency was 48.7 ± 6.3%, compared to 89.1 ± 9.5% (P < 0.0001, n = 10) for [111In]In-oxine. Intracellular retention of 89Zr and cell viability after radiolabelling were comparable to 111In. There were no significant differences in leukocyte chemotaxis or DNA damage between [89Zr]Zr-oxine or [111In]In-oxine. CONCLUSIONS, ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE: Our results demonstrate that [89Zr]Zr-oxine is a suitable PET alternative to [111In]In-oxine for WBC imaging. Our formulation allows rapid, stable, high-yield, single-step preparation of [89Zr]Zr-oxine from commercially available 89Zr. This will facilitate the clinical translation of cell tracking using [89Zr]Zr-oxine.
Collapse
Affiliation(s)
- Francis Man
- School of Biomedical Engineering & Imaging Sciences, King's College London, London SE1 7EH, UK.
| | - Azalea A Khan
- School of Biomedical Engineering & Imaging Sciences, King's College London, London SE1 7EH, UK
| | - Amaia Carrascal-Miniño
- School of Biomedical Engineering & Imaging Sciences, King's College London, London SE1 7EH, UK
| | - Philip J Blower
- School of Biomedical Engineering & Imaging Sciences, King's College London, London SE1 7EH, UK
| | - Rafael T M de Rosales
- School of Biomedical Engineering & Imaging Sciences, King's College London, London SE1 7EH, UK.
| |
Collapse
|
13
|
Bhargava A, Srivastava RK, Mishra DK, Tiwari RR, Sharma RS, Mishra PK. Dendritic cell engineering for selective targeting of female reproductive tract cancers. Indian J Med Res 2019; 148:S50-S63. [PMID: 30964081 PMCID: PMC6469378 DOI: 10.4103/ijmr.ijmr_224_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Female reproductive tract cancers (FRCs) are considered as one of the most frequently occurring malignancies and a foremost cause of death among women. The late-stage diagnosis and limited clinical effectiveness of currently available mainstay therapies, primarily due to the developed drug resistance properties of tumour cells, further increase disease severity. In the past decade, dendritic cell (DC)-based immunotherapy has shown remarkable success and appeared as a feasible therapeutic alternative to treat several malignancies, including FRCs. Importantly, the clinical efficacy of this therapy is shown to be restricted by the established immunosuppressive tumour microenvironment. However, combining nanoengineered approaches can significantly assist DCs to overcome this tumour-induced immune tolerance. The prolonged release of nanoencapsulated tumour antigens helps improve the ability of DC-based therapeutics to selectively target and remove residual tumour cells. Incorporation of surface ligands and co-adjuvants may further aid DC targeting (in vivo) to overcome the issues associated with the short DC lifespan, immunosuppression and imprecise uptake. We herein briefly discuss the necessity and progress of DC-based therapeutics in FRCs. The review also sheds lights on the future challenges to design and develop clinically effective nanoparticles-DC combinations that can induce efficient anti-tumour immune responses and prolong patients’ survival.
Collapse
Affiliation(s)
- Arpit Bhargava
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | | | - Dinesh Kumar Mishra
- School of Pharmacy & Technology Management, Narsee Monjee Institute of Management & Studies, Shirpur, India
| | - Rajnarayan R Tiwari
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Radhey Shyam Sharma
- Division of Reproductive Biology, Maternal & Child Health, Indian Council of Medical Research, New Delhi, India
| | - Pradyumna Kumar Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| |
Collapse
|
14
|
Affiliation(s)
- Jenny Lou
- Department of Medical BiophysicsUniversity of Toronto Toronto M5G 1L7 Canada
- Princess Margaret Cancer CenterUniversity Health Network Toronto M5G 2C1 Canada
- Centre for Pharmaceutical OncologyUniversity of Toronto Toronto M5S 3M2 Canada
| | - Li Zhang
- Toronto General Hospital Research InstituteUniversity Health Network Toronto M5G 2C4 Canada
- Department of ImmunologyUniversity of Toronto Toronto M5S 1A8 Canada
- Department of Laboratory Medicine and PathobiologyUniversity of Toronto Toronto M5S 1A8 Canada
| | - Gang Zheng
- Department of Medical BiophysicsUniversity of Toronto Toronto M5G 1L7 Canada
- Princess Margaret Cancer CenterUniversity Health Network Toronto M5G 2C1 Canada
- Centre for Pharmaceutical OncologyUniversity of Toronto Toronto M5S 3M2 Canada
| |
Collapse
|
15
|
Zhang H, Chen J. Current status and future directions of cancer immunotherapy. J Cancer 2018; 9:1773-1781. [PMID: 29805703 PMCID: PMC5968765 DOI: 10.7150/jca.24577] [Citation(s) in RCA: 207] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 02/05/2018] [Indexed: 12/16/2022] Open
Abstract
In the past decades, our knowledge about the relationship between cancer and the immune system has increased considerably. Recent years' success of cancer immunotherapy including monoclonal antibodies (mAbs), cancer vaccines, adoptive cancer therapy and the immune checkpoint therapy has revolutionized traditional cancer treatment. However, challenges still exist in this field. Personalized combination therapies via new techniques will be the next promising strategies for the future cancer treatment direction.
Collapse
Affiliation(s)
- Hongming Zhang
- Department of Respiratory Medicine, Yancheng Third People's Hospital, the Affiliated Yancheng Hospital of Southeast University Medical College, Yancheng, Jiangsu, China
| | - Jibei Chen
- Department of Respiratory Medicine, Yancheng Third People's Hospital, the Affiliated Yancheng Hospital of Southeast University Medical College, Yancheng, Jiangsu, China
| |
Collapse
|
16
|
Tremblay ML, Davis C, Bowen CV, Stanley O, Parsons C, Weir G, Karkada M, Stanford MM, Brewer KD. Using MRI cell tracking to monitor immune cell recruitment in response to a peptide-based cancer vaccine. Magn Reson Med 2017; 80:304-316. [PMID: 29193231 DOI: 10.1002/mrm.27018] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/26/2017] [Accepted: 10/28/2017] [Indexed: 12/18/2022]
Abstract
PURPOSE MRI cell tracking can be used to monitor immune cells involved in the immunotherapy response, providing insight into the mechanism of action, temporal progression of tumor growth, and individual potency of therapies. To evaluate whether MRI could be used to track immune cell populations in response to immunotherapy, CD8+ cytotoxic T cells, CD4+ CD25+ FoxP3+ regulatory T cells, and myeloid-derived suppressor cells were labeled with superparamagnetic iron oxide particles. METHODS Superparamagnetic iron oxide-labeled cells were injected into mice (one cell type/mouse) implanted with a human papillomavirus-based cervical cancer model. Half of these mice were also vaccinated with DepoVaxTM (ImmunoVaccine, Inc., Halifax, Nova Scotia, Canada), a lipid-based vaccine platform that was developed to enhance the potency of peptide-based vaccines. RESULTS MRI visualization of CD8+ cytotoxic T cells, regulatory T cells, and myeloid-derived suppressor cells was apparent 24 h post-injection, with hypointensities due to iron-labeled cells clearing approximately 72 h post-injection. Vaccination resulted in increased recruitment of CD8+ cytotoxic T cells, and decreased recruitment of myeloid-derived suppressor cells and regulatory T cells to the tumor. We also found that myeloid-derived suppressor cell and regulatory T cell recruitment were positively correlated with final tumor volume. CONCLUSION This type of analysis can be used to noninvasively study changes in immune cell recruitment in individual mice over time, potentially allowing improved application and combination of immunotherapies. Magn Reson Med 80:304-316, 2018. © 2017 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
| | - Christa Davis
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, Nova Scotia, Canada
| | - Chris V Bowen
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, Nova Scotia, Canada.,Department of Diagnostic Radiology, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Physics and Atmospheric Science, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Olivia Stanley
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, Nova Scotia, Canada
| | - Cathryn Parsons
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, Nova Scotia, Canada
| | | | - Mohan Karkada
- Wyss Institute at Harvard Medical School, Boston, Massachusetts, USA
| | - Marianne M Stanford
- Immunovaccine Inc., Halifax, Nova Scotia, Canada.,Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kimberly D Brewer
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, Nova Scotia, Canada.,Department of Diagnostic Radiology, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Physics and Atmospheric Science, Dalhousie University, Halifax, Nova Scotia, Canada.,School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
17
|
Lee SB, Lee HW, Lee H, Jeon YH, Lee SW, Ahn BC, Lee J, Jeong SY. Tracking dendritic cell migration into lymph nodes by using a novel PET probe 18F-tetrafluoroborate for sodium/iodide symporter. EJNMMI Res 2017; 7:32. [PMID: 28378292 PMCID: PMC5380646 DOI: 10.1186/s13550-017-0280-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 03/22/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Recently, 18F-tetrafluoroborate (TFB) was used as a substrate for the human sodium/iodide symporter (hNIS) reporter gene. This study evaluated the feasibility of performing molecular-genetic imaging by using the new radiotracer (18F-TFB) for the hNIS gene, to track dendritic cell (DC) migration in live mice. A murine dendritic cell line (DC2.4) co-expressing the hNIS and effluc genes (DC/NF) was established. To confirm the functional cellular expression of both effluc and NIS in the inoculated DC/NF cells by bio-medical imaging, combined bioluminescence imaging (BLI) and 18F-TFB positron emission tomography/computed tomography (PET/CT) imaging was performed after intramuscular injection with parental DCs and DC/NF cells. For DC-tracking, parental DCs or DC/NF cells were injected in the left or right mouse footpad, respectively, and 18F-TFB PET/CT and BLI were performed to monitor these cells in live mice. RESULTS In vivo PET/CT and BLI showed a clear signal in DC/NF injection sites but not in parental DC injection sites. The signal intensity in DC/NF cells was correlated with time. In vivo 18F-TFB PET/CT imaging showed higher radiotracer activity in the draining popliteal lymph nodes (DPLNs) in DC/NF injection sites than those in DC injection sites on day 2. BLI also showed DC/NF cell migration to the DPLNs on day 2 after the injection. CONCLUSIONS Migration of DCs to the lymph nodes was successfully monitored using 18F-TFB PET/CT imaging of the NIS gene and optical imaging of the effluc gene in live mice. These data support the feasibility of using 18F-TFB as a substrate for hNIS reporter gene imaging to track the migration of DCs to the lymph nodes in live animals. The use of 18F-TFB may facilitate enhanced PET imaging of the hNIS reporter gene in small animals and humans in future studies.
Collapse
Affiliation(s)
- Sang Bong Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 130 Dongdeok-ro, Jung-gu, Daegu, 41944, Republic of Korea.,Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Medical Center, 807 Hogukro, Buk-gu, Daegu, 41404, Republic of Korea
| | - Ho Won Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 130 Dongdeok-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Hongje Lee
- Department of Nuclear Medicine, Dongnam Institution of Radiological and Medical Sciences, 40, Jwadong-gil, Jangan-eup, Gijang-gun, Busan, 46033, Republic of Korea
| | - Yong Hyun Jeon
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 130 Dongdeok-ro, Jung-gu, Daegu, 41944, Republic of Korea.,Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80 Cheombok-ro, Dong-gu, Daegu, 41061, Republic of Korea
| | - Sang-Woo Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 130 Dongdeok-ro, Jung-gu, Daegu, 41944, Republic of Korea.,Department of Nuclear Medicine, Dongnam Institution of Radiological and Medical Sciences, 40, Jwadong-gil, Jangan-eup, Gijang-gun, Busan, 46033, Republic of Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 130 Dongdeok-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Jaetae Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 130 Dongdeok-ro, Jung-gu, Daegu, 41944, Republic of Korea.,Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80 Cheombok-ro, Dong-gu, Daegu, 41061, Republic of Korea
| | - Shin Young Jeong
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 130 Dongdeok-ro, Jung-gu, Daegu, 41944, Republic of Korea.
| |
Collapse
|
18
|
Makela AV, Murrell DH, Parkins KM, Kara J, Gaudet JM, Foster PJ. Cellular Imaging With MRI. Top Magn Reson Imaging 2016; 25:177-186. [PMID: 27748707 DOI: 10.1097/rmr.0000000000000101] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Cellular magnetic resonance imaging (MRI) is an evolving field of imaging with strong translational and research potential. The ability to detect, track, and quantify cells in vivo and over time allows for studying cellular events related to disease processes and may be used as a biomarker for decisions about treatments and for monitoring responses to treatments. In this review, we discuss methods for labeling cells, various applications for cellular MRI, the existing limitations, strategies to address these shortcomings, and clinical cellular MRI.
Collapse
Affiliation(s)
- Ashley V Makela
- *Imaging Research Laboratories, Robarts Research Institute †Department of Medical Biophysics, Western University, London, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
19
|
Revia RA, Zhang M. Magnetite nanoparticles for cancer diagnosis, treatment, and treatment monitoring: recent advances. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2016; 19:157-168. [PMID: 27524934 PMCID: PMC4981486 DOI: 10.1016/j.mattod.2015.08.022] [Citation(s) in RCA: 340] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
The development of nanoparticles (NPs) for use in all facets of oncological disease detection and therapy has shown great progress over the past two decades. NPs have been tailored for use as contrast enhancement agents for imaging, drug delivery vehicles, and most recently as a therapeutic component in initiating tumor cell death in magnetic and photonic ablation therapies. Of the many possible core constituents of NPs, such as gold, silver, carbon nanotubes, fullerenes, manganese oxide, lipids, micelles, etc., iron oxide (or magnetite) based NPs have been extensively investigated due to their excellent superparamagnetic, biocompatible, and biodegradable properties. This review addresses recent applications of magnetite NPs in diagnosis, treatment, and treatment monitoring of cancer. Finally, some views will be discussed concerning the toxicity and clinical translation of iron oxide NPs and the future outlook of NP development to facilitate multiple therapies in a single formulation for cancer theranostics.
Collapse
Affiliation(s)
- Richard A. Revia
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
20
|
He Q, Guo S, Qian Z, Chen X. Development of individualized anti-metastasis strategies by engineering nanomedicines. Chem Soc Rev 2015; 44:6258-6286. [PMID: 26056688 PMCID: PMC4540626 DOI: 10.1039/c4cs00511b] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Metastasis is deadly and also tough to treat as it is much more complicated than the primary tumour. Anti-metastasis approaches available so far are far from being optimal. A variety of nanomedicine formulae provide a plethora of opportunities for developing new strategies and means for tackling metastasis. It should be noted that individualized anti-metastatic nanomedicines are different from common anti-cancer nanomedicines as they specifically target different populations of malignant cells. This review briefly introduces the features of the metastatic cascade, and proposes a series of nanomedicine-based anti-metastasis strategies aiming to block each metastatic step. Moreover, we also concisely introduce the advantages of several promising nanoparticle platforms and their potential for constructing state-of-the-art individualized anti-metastatic nanomedicines.
Collapse
Affiliation(s)
- Qianjun He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, P. R. China.
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| | - Shengrong Guo
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Zhiyong Qian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, P. R. China.
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
21
|
Bhargava A, Bunkar N, Khare NK, Mishra D, Mishra PK. Nanoengineered strategies to optimize dendritic cells for gastrointestinal tumor immunotherapy: from biology to translational medicine. Nanomedicine (Lond) 2015; 9:2187-202. [PMID: 25405796 DOI: 10.2217/nnm.14.115] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Nanomedicine may play an important role in improving the clinical efficacy of dendritic cell-based immunotherapy against GI tract malignancies. Dendritic cell-based vaccines have proven their effectiveness against different established GI tract tumors, yet their success is mainly hindered by the strong tumor-induced suppressive microenvironment. The sustained and targeted release of tumor antigens to dendritic cells using different nanoengineered approaches would be an efficient strategy to overcome established immune tolerance. Encapsulation would result in low diffusivity, restricted movement, effective crosspresentation and enhanced T-cell responses. These nanotherapy-based approaches will certainly help with the designing of clinically translatable dendritic cell-based therapeutic vaccines and facilitate the selective removal of residual disease in gastrointestinal cancer patients following standard treatments.
Collapse
Affiliation(s)
- Arpit Bhargava
- Translational Research Laboratory, School of Biological Sciences, Dr H. S. Gour Central University, Sagar, India
| | | | | | | | | |
Collapse
|
22
|
Charoenphun P, Meszaros LK, Chuamsaamarkkee K, Sharif-Paghaleh E, Ballinger JR, Ferris TJ, Went MJ, Mullen GED, Blower PJ. [(89)Zr]oxinate4 for long-term in vivo cell tracking by positron emission tomography. Eur J Nucl Med Mol Imaging 2015; 42:278-87. [PMID: 25359636 PMCID: PMC4315484 DOI: 10.1007/s00259-014-2945-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 10/16/2014] [Indexed: 01/08/2023]
Abstract
PURPOSE (111)In (typically as [(111)In]oxinate3) is a gold standard radiolabel for cell tracking in humans by scintigraphy. A long half-life positron-emitting radiolabel to serve the same purpose using positron emission tomography (PET) has long been sought. We aimed to develop an (89)Zr PET tracer for cell labelling and compare it with [(111)In]oxinate3 single photon emission computed tomography (SPECT). METHODS [(89)Zr]Oxinate4 was synthesised and its uptake and efflux were measured in vitro in three cell lines and in human leukocytes. The in vivo biodistribution of eGFP-5T33 murine myeloma cells labelled using [(89)Zr]oxinate4 or [(111)In]oxinate3 was monitored for up to 14 days. (89)Zr retention by living radiolabelled eGFP-positive cells in vivo was monitored by FACS sorting of liver, spleen and bone marrow cells followed by gamma counting. RESULTS Zr labelling was effective in all cell types with yields comparable with (111)In labelling. Retention of (89)Zr in cells in vitro after 24 h was significantly better (range 71 to >90%) than (111)In (43-52%). eGFP-5T33 cells in vivo showed the same early biodistribution whether labelled with (111)In or (89)Zr (initial pulmonary accumulation followed by migration to liver, spleen and bone marrow), but later translocation of radioactivity to kidneys was much greater for (111)In. In liver, spleen and bone marrow at least 92% of (89)Zr remained associated with eGFP-positive cells after 7 days in vivo. CONCLUSION [(89)Zr]Oxinate4 offers a potential solution to the emerging need for a long half-life PET tracer for cell tracking in vivo and deserves further evaluation of its effects on survival and behaviour of different cell types.
Collapse
Affiliation(s)
- Putthiporn Charoenphun
- King’s College London, Division of Imaging Sciences and Biomedical Engineering, 4th Floor Lambeth Wing, St Thomas’ Hospital, London, SE1 7EH UK
| | - Levente K. Meszaros
- King’s College London, Division of Imaging Sciences and Biomedical Engineering, 4th Floor Lambeth Wing, St Thomas’ Hospital, London, SE1 7EH UK
| | - Krisanat Chuamsaamarkkee
- King’s College London, Division of Imaging Sciences and Biomedical Engineering, 4th Floor Lambeth Wing, St Thomas’ Hospital, London, SE1 7EH UK
| | - Ehsan Sharif-Paghaleh
- King’s College London, Division of Imaging Sciences and Biomedical Engineering, 4th Floor Lambeth Wing, St Thomas’ Hospital, London, SE1 7EH UK
| | - James R. Ballinger
- King’s College London, Division of Imaging Sciences and Biomedical Engineering, 4th Floor Lambeth Wing, St Thomas’ Hospital, London, SE1 7EH UK
| | - Trevor J. Ferris
- School of Physical Sciences, University of Kent, Canterbury, CT2 7NH UK
| | - Michael J. Went
- School of Physical Sciences, University of Kent, Canterbury, CT2 7NH UK
| | - Gregory E. D. Mullen
- King’s College London, Division of Imaging Sciences and Biomedical Engineering, 4th Floor Lambeth Wing, St Thomas’ Hospital, London, SE1 7EH UK
| | - Philip J. Blower
- King’s College London, Division of Imaging Sciences and Biomedical Engineering, 4th Floor Lambeth Wing, St Thomas’ Hospital, London, SE1 7EH UK
- Division of Chemistry, King’s College London, Britannia House, 7 Trinity St, London, SE11DB UK
| |
Collapse
|
23
|
Superparamagnetic iron oxide labeling limits the efficacy of rabbit immature dendritic cell vaccination by decreasing their antigen uptake ability in a lysosome-dependent manner. Biotechnol Lett 2014; 37:289-98. [PMID: 25257596 DOI: 10.1007/s10529-014-1681-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 09/08/2014] [Indexed: 10/24/2022]
Abstract
Immature dendritic cells (iDCs) are for cell transplantation; however, no method has yet been developed for in vivo monitoring the transplanted iDCs. We have explored the feasibility of using superparamagnetic iron oxide (SPIO) labeling and magnetic resonance imaging for in vivo tracking of transplanted iDCs and determined the effects of SPIO labeling on iDC vaccination. With up to 50 μg Fe/ml, SPIO effectively labeled the iDCs without affecting their growth. At or above 100 μg Fe/ml, SPIO caused considerable damage to iDCs. SPIO labeling resulted in autophagosome formation and decreased the uptake of oxidized low density lipoprotein (ox-LDL), an exogenous antigen, by iDCs. SPIO and ox-LDL both localized to the lysosomes, and this competition for lysosomes could be partially responsible for the decreased ox-LDL phagocytic capacity of iDCs due to SPIO labeling.
Collapse
|
24
|
Liu J, Wang L, Cao J, Huang Y, Lin Y, Wu X, Wang Z, Zhang F, Xu X, Liu G. Functional investigations on embryonic stem cells labeled with clinically translatable iron oxide nanoparticles. NANOSCALE 2014; 6:9025-33. [PMID: 24969040 DOI: 10.1039/c4nr01004c] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Stem cell based therapies offer significant potential in the field of regenerative medicine. The development of superparamagnetic iron oxide (SPIO) nanoparticle labeling and magnetic resonance imaging (MRI) have been increasingly used to track the transplanted cells, enabling in vivo determination of cell fate. However, the impact of SPIO-labeling on the cell phenotype and differentiation capacity of embryonic stem cells (ESCs) remains unclear. In this study, we wrapped SPIO nanoparticles with stearic acid grafted PEI600, termed as Stearic-LWPEI-SPIO, to generate efficient and non-toxic ESC labeling tools. Our results showed that efficient labeling of ESCs at an optimized low dosage of Stearic-LWPEI-SPIO nanoparticles did not alter the differentiation and self-renewal properties of ESCs. The localization of the transplanted ESCs observed by MRI correlated well with histological studies. These findings demonstrate that Stearic-LWPEI-SPIO nanoparticles have potential to be clinically translatable MRI probes and may enable non-invasive in vivo tracking of ESCs in experimental and clinical settings during cell-based therapies.
Collapse
Affiliation(s)
- Jing Liu
- Institute of Stem Cell and Regenerative Medicine, Medical College, Xiamen University, Xiamen, 361102, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Sharif-Paghaleh E, Leech J, Sunassee K, Ali N, Sagoo P, Lechler RI, Smyth LA, Lombardi G, Mullen GE. Monitoring the efficacy of dendritic cell vaccination by early detection of (99m) Tc-HMPAO-labelled CD4(+) T cells. Eur J Immunol 2014; 44:2188-91. [PMID: 24643793 PMCID: PMC4211358 DOI: 10.1002/eji.201344337] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 02/12/2014] [Accepted: 03/12/2014] [Indexed: 12/02/2022]
Affiliation(s)
- Ehsan Sharif-Paghaleh
- Medical Research Council (MRC) Centre for Transplantation, King's College London, King's Health Partners, Guy's Hospital, London, United Kingdom; Division of Imaging Sciences and Biomedical Engineering, King's College London, St. Thomas Hospital, London, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Superparamagnetic iron oxide based nanoprobes for imaging and theranostics. Adv Colloid Interface Sci 2013; 199-200:95-113. [PMID: 23891347 DOI: 10.1016/j.cis.2013.06.007] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 06/21/2013] [Accepted: 06/27/2013] [Indexed: 12/11/2022]
Abstract
The need to target, deliver and subsequently evaluate the efficacy of therapeutics in the treatment of a disease has provided added impetus in developing novel and highly efficient contrast agents. Superparamagnetic iron oxide nanoparticles (SPIONs) have offered tremendous potential in designing advanced magnetic resonance imaging (MRI) diagnostic agents, due to their unique physicochemical properties. There has been tremendous effort devoted in the recent past in developing synthetic methodologies through which their size, hydrodynamic radii, chemical composition and morphologies could be tailored at the nanoscale. This enables one to fine tune their magnetic behavior, and thus their MRI response. While novel synthetic strategies are being assembled for directing SPIONs to the diseased site as well as imparting them stealth and biocompatibility, it is also essential to evaluate their biological toxicological profiles. This review highlights recent advances that have been made in the synthesis of SPIONs, subsequent functionalization with desired entities, and a discussion on their use as MRI contrast agents in cardiovascular research.
Collapse
|
27
|
Leech JM, Sharif-Paghaleh E, Maher J, Livieratos L, Lechler RI, Mullen GE, Lombardi G, Smyth LA. Whole-body imaging of adoptively transferred T cells using magnetic resonance imaging, single photon emission computed tomography and positron emission tomography techniques, with a focus on regulatory T cells. Clin Exp Immunol 2013; 172:169-77. [PMID: 23574314 DOI: 10.1111/cei.12087] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2013] [Indexed: 01/03/2023] Open
Abstract
Cell-based therapies using natural or genetically modified regulatory T cells (T(regs)) have shown significant promise as immune-based therapies. One of the main difficulties facing the further advancement of these therapies is that the fate and localization of adoptively transferred T(regs) is largely unknown. The ability to dissect the migratory pathway of these cells in a non-invasive manner is of vital importance for the further development of in-vivo cell-based immunotherapies, as this technology allows the fate of the therapeutically administered cell to be imaged in real time. In this review we will provide an overview of the current clinical imaging techniques used to track T cells and T(regs) in vivo, including magnetic resonance imaging (MRI) and positron emission tomography (PET)/single photon emission computed tomography (SPECT). In addition, we will discuss how the finding of these studies can be used, in the context of transplantation, to define the most appropriate T(reg) subset required for cellular therapy.
Collapse
Affiliation(s)
- J M Leech
- Medical Research Council, Centre for Transplantation, King's College London, King's Health Partners, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Ferguson PM, Slocombe A, Tilley RD, Hermans IF. Using magnetic resonance imaging to evaluate dendritic cell-based vaccination. PLoS One 2013; 8:e65318. [PMID: 23734246 PMCID: PMC3667033 DOI: 10.1371/journal.pone.0065318] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Accepted: 04/28/2013] [Indexed: 01/19/2023] Open
Abstract
Cancer immunotherapy with antigen-loaded dendritic cell-based vaccines can induce clinical responses in some patients, but further optimization is required to unlock the full potential of this strategy in the clinic. Optimization is dependent on being able to monitor the cellular events that take place once the dendritic cells have been injected in vivo, and to establish whether antigen-specific immune responses to the tumour have been induced. Here we describe the use of magnetic resonance imaging (MRI) as a simple, non-invasive approach to evaluate vaccine success. By loading the dendritic cells with highly magnetic iron nanoparticles it is possible to assess whether the injected cells drain to the lymph nodes. It is also possible to establish whether an antigen-specific response is initiated by assessing migration of successive rounds of antigen-loaded dendritic cells; in the face of a successfully primed cytotoxic response, the bulk of antigen-loaded cells are eradicated on-route to the node, whereas cells without antigen can reach the node unchecked. It is also possible to verify the induction of a vaccine-induced response by simply monitoring increases in draining lymph node size as a consequence of vaccine-induced lymphocyte trapping, which is an antigen-specific response that becomes more pronounced with repeated vaccination. Overall, these MRI techniques can provide useful early feedback on vaccination strategies, and could also be used in decision making to select responders from non-responders early in therapy.
Collapse
Affiliation(s)
| | - Angela Slocombe
- Department of Radiology, Wellington Hospital, Wellington, New Zealand
| | - Richard D. Tilley
- School of Chemical and Physical Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Ian F. Hermans
- Malaghan Institute of Medical Research, Wellington, New Zealand
- * E-mail:
| |
Collapse
|
29
|
Interaction of tumor cells with the immune system: implications for dendritic cell therapy and cancer progression. Drug Discov Today 2013; 18:35-42. [DOI: 10.1016/j.drudis.2012.07.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 06/30/2012] [Accepted: 07/18/2012] [Indexed: 01/21/2023]
|
30
|
Sagoo P, Ratnasothy K, Tsang Y, Barber LD, Noble A, Lechler RI, Lombardi G. Alloantigen-specific regulatory T cells prevent experimental chronic graft-versus-host disease by simultaneous control of allo- and autoreactivity. Eur J Immunol 2012; 42:3322-33. [DOI: 10.1002/eji.201242770] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 08/02/2012] [Accepted: 09/12/2012] [Indexed: 01/01/2023]
Affiliation(s)
- Pervinder Sagoo
- NIHR Biomedical Research Centre; Guy's & St Thomas’ NHS Foundation Trust & King's College London, London; UK
- MRC Centre for Transplantation; Department of Transplantation; Immunoregulation & Mucosal Biology; King's College London, London; UK
| | - Kulachelvy Ratnasothy
- MRC Centre for Transplantation; Department of Transplantation; Immunoregulation & Mucosal Biology; King's College London, London; UK
| | - Yuen Tsang
- MRC Centre for Transplantation; Department of Transplantation; Immunoregulation & Mucosal Biology; King's College London, London; UK
| | - Linda D. Barber
- Department of Haematological Medicine; King's College London; London UK
| | - Alistair Noble
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma; King's College London; London UK
| | - Robert I. Lechler
- MRC Centre for Transplantation; Department of Transplantation; Immunoregulation & Mucosal Biology; King's College London, London; UK
| | - Giovanna Lombardi
- MRC Centre for Transplantation; Department of Transplantation; Immunoregulation & Mucosal Biology; King's College London, London; UK
| |
Collapse
|
31
|
Bhargava A, Mishra D, Banerjee S, Mishra PK. Engineered dendritic cells for gastrointestinal tumor immunotherapy: opportunities in translational research. J Drug Target 2012; 21:126-36. [PMID: 23061479 DOI: 10.3109/1061186x.2012.731069] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
32
|
Colombo M, Carregal-Romero S, Casula MF, Gutiérrez L, Morales MP, Böhm IB, Heverhagen JT, Prosperi D, Parak WJ. Biological applications of magnetic nanoparticles. Chem Soc Rev 2012; 41:4306-34. [PMID: 22481569 DOI: 10.1039/c2cs15337h] [Citation(s) in RCA: 706] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In this review an overview about biological applications of magnetic colloidal nanoparticles will be given, which comprises their synthesis, characterization, and in vitro and in vivo applications. The potential future role of magnetic nanoparticles compared to other functional nanoparticles will be discussed by highlighting the possibility of integration with other nanostructures and with existing biotechnology as well as by pointing out the specific properties of magnetic colloids. Current limitations in the fabrication process and issues related with the outcome of the particles in the body will be also pointed out in order to address the remaining challenges for an extended application of magnetic nanoparticles in medicine.
Collapse
Affiliation(s)
- Miriam Colombo
- Dipartimento di Biotecnologie e Bioscienze, Università di Milano-Bicocca, Milan, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Xu C, Mu L, Roes I, Miranda-Nieves D, Nahrendorf M, Ankrum JA, Zhao W, Karp JM. Nanoparticle-based monitoring of cell therapy. NANOTECHNOLOGY 2011; 22:494001. [PMID: 22101191 PMCID: PMC3334527 DOI: 10.1088/0957-4484/22/49/494001] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Exogenous cell therapy aims to replace/repair diseased or dysfunctional cells and promises to revolutionize medicine by restoring tissue and organ function. To develop effective cell therapy, the location, distribution and long-term persistence of transplanted cells must be evaluated. Nanoparticle (NP) based imaging technologies have the potential to track transplanted cells non-invasively. Here we summarize the most recent advances in NP-based cell tracking with emphasis on (1) the design criteria for cell tracking NPs, (2) protocols for cell labeling, (3) a comparison of available imaging modalities and their corresponding contrast agents, (4) a summary of preclinical studies on NP-based cell tracking and finally (5) perspectives and future directions.
Collapse
Affiliation(s)
- Chenjie Xu
- Center for Regenerative Therapeutics and Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | - Luye Mu
- Center for Regenerative Therapeutics and Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | - Isaac Roes
- Center for Regenerative Therapeutics and Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | - David Miranda-Nieves
- Center for Regenerative Therapeutics and Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, 185 Cambridge Street, Boston, MA 02114, USA
| | - James A Ankrum
- Center for Regenerative Therapeutics and Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | - Weian Zhao
- Center for Regenerative Therapeutics and Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | - Jeffrey M Karp
- Center for Regenerative Therapeutics and Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, 65 Landsdowne Street, Cambridge, MA 02139, USA
| |
Collapse
|
34
|
Mou Y, Hou Y, Chen B, Hua Z, Zhang Y, Xie H, Xia G, Wang Z, Huang X, Han W, Ni Y, Hu Q. In vivo migration of dendritic cells labeled with synthetic superparamagnetic iron oxide. Int J Nanomedicine 2011; 6:2633-40. [PMID: 22114494 PMCID: PMC3218577 DOI: 10.2147/ijn.s24307] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Successful treatment of cancer with dendritic cell tumor vaccine is highly dependent on how effectively the vaccine migrates into lymph nodes and activates T cells. In this study, a simple method was developed to trace migration of dendritic cells to lymph nodes. METHODS Superparamagnetic iron oxide (SPIO) of γ-Fe(2)O(3) nanoparticles were prepared to label dendritic cells generated from bone marrow of enhanced green fluorescent protein (EGFP) transgenic mice, to explore the fluorescence intensity of EGFP influenced by the SPIO, and to make images of labeled dendritic cells with the help of magnetic resonance imaging in vitro. The SPIO-EGFP-labeled dendritic cells were injected into the footpads of five mice. After 48 hours, magnetic resonance imaging, optical imaging, confocal imaging, and Prussian blue staining were used to confirm migration of the SPIO-EGFP-labeled dendritic cells into draining lymph nodes. RESULTS The synthetic SPIO nanoparticles had a spherical shape and desirable superparamagnetism, and confocal imaging and Prussian blue staining showed perfect labeling efficiency as well. Furthermore, the dendritic cells dual-labeled by SPIO and EGFP could migrate into lymph nodes after footpad injection, and could be detected by both magnetic resonance imaging and optical imaging simultaneously, which was further confirmed by immunohistochemistry and Prussian blue staining. The percentage of dendritic cells migrated to the draining lymph nodes was about 4%. CONCLUSION Synthetic SPIO nanoparticles are strong contrast agents with good biocompatibility, and EGFP transgenic dendritic cells can be labeled efficiently by SPIO, which are suitable for further study of the migratory behavior and biodistribution of dendritic cells in vivo.
Collapse
Affiliation(s)
- Yongbin Mou
- Central Laboratory of Stomatology, Stomatological Hospital Affiliated Medical School, Nanjing University, Nanjing, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Sharif-Paghaleh E, Sunassee K, Tavaré R, Ratnasothy K, Koers A, Ali N, Alhabbab R, Blower PJ, Lechler RI, Smyth LA, Mullen GE, Lombardi G. In vivo SPECT reporter gene imaging of regulatory T cells. PLoS One 2011; 6:e25857. [PMID: 22043296 PMCID: PMC3197183 DOI: 10.1371/journal.pone.0025857] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Accepted: 09/12/2011] [Indexed: 12/24/2022] Open
Abstract
Regulatory T cells (Tregs) were identified several years ago and are key in controlling autoimmune diseases and limiting immune responses to foreign antigens, including alloantigens. In vivo imaging techniques including intravital microscopy as well as whole body imaging using bioluminescence probes have contributed to the understanding of in vivo Treg function, their mechanisms of action and target cells. Imaging of the human sodium/iodide symporter via Single Photon Emission Computed Tomography (SPECT) has been used to image various cell types in vivo. It has several advantages over the aforementioned imaging techniques including high sensitivity, it allows non-invasive whole body studies of viable cell migration and localisation of cells over time and lastly it may offer the possibility to be translated to the clinic. This study addresses whether SPECT/CT imaging can be used to visualise the migratory pattern of Tregs in vivo. Treg lines derived from CD4+CD25+FoxP3+ cells were retrovirally transduced with a construct encoding for the human Sodium Iodide Symporter (NIS) and the fluorescent protein mCherry and stimulated with autologous DCs. NIS expressing self-specific Tregs were specifically radiolabelled in vitro with Technetium-99m pertechnetate (99mTcO4−) and exposure of these cells to radioactivity did not affect cell viability, phenotype or function. In addition adoptively transferred Treg-NIS cells were imaged in vivo in C57BL/6 (BL/6) mice by SPECT/CT using 99mTcO4−. After 24 hours NIS expressing Tregs were observed in the spleen and their localisation was further confirmed by organ biodistribution studies and flow cytometry analysis. The data presented here suggests that SPECT/CT imaging can be utilised in preclinical imaging studies of adoptively transferred Tregs without affecting Treg function and viability thereby allowing longitudinal studies within disease models.
Collapse
Affiliation(s)
- Ehsan Sharif-Paghaleh
- Medical Research Council (MRC) Centre for Transplantation, King's College London, King's Health Partners, Guy's Hospital, London, United Kingdom
- Division of Imaging Sciences, King's College London, St Thomas Hospital, London, United Kingdom
| | - Kavitha Sunassee
- Division of Imaging Sciences, King's College London, St Thomas Hospital, London, United Kingdom
| | - Richard Tavaré
- Division of Imaging Sciences, King's College London, St Thomas Hospital, London, United Kingdom
| | - Kulachelvy Ratnasothy
- Medical Research Council (MRC) Centre for Transplantation, King's College London, King's Health Partners, Guy's Hospital, London, United Kingdom
| | - Alexander Koers
- Division of Imaging Sciences, King's College London, St Thomas Hospital, London, United Kingdom
| | - Niwa Ali
- Medical Research Council (MRC) Centre for Transplantation, King's College London, King's Health Partners, Guy's Hospital, London, United Kingdom
| | - Rowa Alhabbab
- Medical Research Council (MRC) Centre for Transplantation, King's College London, King's Health Partners, Guy's Hospital, London, United Kingdom
| | - Philip J. Blower
- Division of Imaging Sciences, King's College London, St Thomas Hospital, London, United Kingdom
| | - Robert I. Lechler
- Medical Research Council (MRC) Centre for Transplantation, King's College London, King's Health Partners, Guy's Hospital, London, United Kingdom
| | - Lesley A. Smyth
- Medical Research Council (MRC) Centre for Transplantation, King's College London, King's Health Partners, Guy's Hospital, London, United Kingdom
| | - Gregory E. Mullen
- Medical Research Council (MRC) Centre for Transplantation, King's College London, King's Health Partners, Guy's Hospital, London, United Kingdom
- Division of Imaging Sciences, King's College London, St Thomas Hospital, London, United Kingdom
- * E-mail: (GL); (GEM)
| | - Giovanna Lombardi
- Medical Research Council (MRC) Centre for Transplantation, King's College London, King's Health Partners, Guy's Hospital, London, United Kingdom
- * E-mail: (GL); (GEM)
| |
Collapse
|