1
|
Sarkar S, Pham JM, Edwards KJ, Sharma N, Xu K, King AP, Del Castillo AF, Farwell MD, Pryma DA, Schuster SJ, Sellmyer MA. A biorthogonal chemistry approach for high-contrast antibody imaging of lymphoma at early time points. EJNMMI Res 2025; 15:26. [PMID: 40122966 PMCID: PMC11930911 DOI: 10.1186/s13550-025-01213-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 02/27/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Monoclonal antibodies are highly specific for their targets making them effective for cancer therapy. However, their large molecular weight causes slow blood clearance, often requiring weeks to be removed from circulation. This limitation affects companion nuclear imaging and antibody-based diagnostics, necessitating delayed imaging. We report the expansion of a methodology improving positron emission tomography (PET) contrast of the lymphoma biomarker CD20 at early time points after radiolabeled antibody administration. Intact radioimmunoconjugates are allowed to stay in circulation long enough to accumulate in tumors, and then, using a chemical trigger, we induced rapid clearance of the radioactivity from non-target tissues by cleaving the linker between the antibody and the radioactivity. For brevity, we refer to the this as the Tetrazine KnockOut (TKO) method which uses the transcyclooctene-tetrazine (TCO-Tz) reaction, wherein an antibody is conjugated with linker containing TCO and a radioisotope. RESULTS We optimized the TCO linker with several different radioisotopes and evaluated the ability of tetrazines to knockout radioactivity from circulating antibodies. We explored several cell types and antibodies with varying internalization rates, to characterize the parameters of TKO and tested [89Zr]Zr-DFO-TCO-rituximab in a lymphoma model with PET imaging after Tz or vehicle administration. Treatment with Tz induced > 70% cleavage of the TCO linker in vitro within 30 min. Internalizing radioimmunoconjugates exhibited similar cellular uptake with Tz compared to vehicle, whereas decreased uptake was seen with slowly internalizing antibodies. In rodents, Tz rapidly liberated the radioactivity from the antibody, cleared from the blood, and accumulated in the bladder. TKO resulted in > 50% decreased radioactivity in non-target organs following Tz injection. No decrease in tumor uptake was observed when rate of antibody internalization is higher in a lymphoma model, and the target-to-background ratio increased by > twofold in comparison with Tz nontreated groups at 24 h. CONCLUSION The TKO approach potentiates early imaging of rituximab radioimmunoconjugates and has translational potential for lymphoma imaging.
Collapse
Affiliation(s)
- Swarbhanu Sarkar
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 813A Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA, 19104-6059, USA
| | - Jonathan M Pham
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 813A Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA, 19104-6059, USA
| | - Kimberly J Edwards
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 813A Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA, 19104-6059, USA
| | - Nitika Sharma
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 813A Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA, 19104-6059, USA
| | - Kexiang Xu
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 813A Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA, 19104-6059, USA
| | - A Paden King
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 813A Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA, 19104-6059, USA
| | - Andres Fernandez Del Castillo
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 813A Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA, 19104-6059, USA
| | - Michael D Farwell
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 813A Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA, 19104-6059, USA
| | - Daniel A Pryma
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 813A Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA, 19104-6059, USA
| | - Stephen J Schuster
- Lymphoma Program, Abramson Cancer Center at the University of Pennsylvania, Philadelphia, PA, USA
| | - Mark A Sellmyer
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 813A Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA, 19104-6059, USA.
- The Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
2
|
89Zr-labelled Obinutuzumab: a potential immuno-PET radiopharmaceutical. J Radioanal Nucl Chem 2022. [DOI: 10.1007/s10967-022-08614-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
3
|
Can pre-transplant 18F-choline positron emission tomography predict relapse following autologous stem cell transplantation in primary central nervous system lymphoma? Bone Marrow Transplant 2022; 57:113-115. [PMID: 34611292 DOI: 10.1038/s41409-021-01484-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/15/2021] [Accepted: 09/24/2021] [Indexed: 02/08/2023]
|
4
|
In situ lymphoma imaging in a spontaneous mouse model using the Cerenkov Luminescence of F-18 and Ga-67 isotopes. Sci Rep 2021; 11:24002. [PMID: 34907289 PMCID: PMC8671545 DOI: 10.1038/s41598-021-03505-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 10/12/2021] [Indexed: 11/08/2022] Open
Abstract
Cerenkov luminescence imaging (CLI) is a promising approach to image-guided surgery and pathological sampling. It could offer additional advantages when combined to whole-body isotope tomographies. We aimed to obtain evidence of its applicability in lymphoma patho-diagnostics, thus we decided to investigate the radiodiagnostic potential of combined PET or SPECT/CLI in an experimental, novel spontaneous high-grade B-cell lymphoma mouse model (Bc.DLFL1). We monitored the lymphoma dissemination at early stage, and at clinically relevant stages such as advanced stage and terminal stage with in vivo 2-deoxy-2-[18F]fluoro-d-glucose (FDG) positron emission tomography (PET)/magnetic resonance imaging (MRI) and 67Ga-citrate single photon emission computed tomography (SPECT)/MRI. In vivo imaging was combined with ex vivo high resolution CLI. The use of CLI with 18F-Fluorine (F-18) and 67Ga-Gallium isotopes in the selection of infiltrated lymph nodes for tumor staging and pathology was thus tested. At advanced stage, FDG PET/MRI plus ex vivo CLI allowed accurate detection of FDG accumulation in lymphoma-infiltrated tissues. At terminal stage we detected tumorous lymph nodes with SPECT/MRI and we could report in vivo detection of the Cerenkov light emission of 67Ga. CLI with 67Ga-citrate revealed lymphoma accumulation in distant lymph node locations, unnoticeable with only MRI. Flow cytometry and immunohistochemistry confirmed these imaging results. Our study promotes the combined use of PET and CLI in preclinical studies and clinical practice. Heterogeneous FDG distribution in lymph nodes, detected at sampling surgery, has implications for tissue pathology processing and it could direct therapy. The results with 67Ga also point to the opportunities to further apply suitable SPECT radiopharmaceuticals for CLI.
Collapse
|
5
|
Dun Y, Huang G, Liu J, Wei W. ImmunoPET imaging of hematological malignancies: From preclinical promise to clinical reality. Drug Discov Today 2021; 27:1196-1203. [PMID: 34838729 DOI: 10.1016/j.drudis.2021.11.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 10/22/2021] [Accepted: 11/18/2021] [Indexed: 12/23/2022]
Abstract
Immuno-positron emission tomography (immunoPET) imaging is a paradigm-shifting imaging technique for whole-body and all-lesion tumor detection, based on the combined specificity of tumor-targeting vectors [e.g., monoclonal antibodies (mAbs), nanobodies, and bispecific antibodies] and the sensitivity of PET imaging. By noninvasively, comprehensively, and serially revealing heterogeneous tumor antigen expression, immunoPET imaging is gradually improving the theranostic prospects for hematological malignancies. In this review, we summarize the available literature regarding immunoPET in imaging hematological malignancies. We also highlight the pros and cons of current conjugation strategies, and modular chemistry that can be leveraged to develop novel immunoPET probes for hematological malignancies. Lastly, we discuss the use of immunoPET imaging in guiding antibody drug development.
Collapse
Affiliation(s)
- Yiting Dun
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China; Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China.
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China.
| |
Collapse
|
6
|
Kang L, Li C, Yang Q, Sutherlin L, Wang L, Chen Z, Becker KV, Huo N, Qiu Y, Engle JW, Wang R, He C, Jiang D, Xu X, Cai W. 64Cu-labeled daratumumab F(ab') 2 fragment enables early visualization of CD38-positive lymphoma. Eur J Nucl Med Mol Imaging 2021; 49:1470-1481. [PMID: 34677626 DOI: 10.1007/s00259-021-05593-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 10/11/2021] [Indexed: 12/19/2022]
Abstract
PURPOSE Abnormal CD38 expression in some hematologic malignancies, including lymphoma, has made it a biomarker for targeted therapies. Daratumumab (Dara) is the first FDA-approved CD38-specific monoclonal antibody, enabling successfully immunoPET imaging over the past years. Radiolabeled Dara however has a long blood circulation and delayed tumor uptake which can limit its applications. The focus of this study is to develop 64Cu-labeled Dara-F(ab')2 for the visualization of CD38 in lymphoma models. METHODS F(ab')2 fragment was prepared from Dara using an IdeS enzyme and purified with Protein A beads. Western blotting, flow cytometry, and surface plasmon resonance (SPR) were performed for in vitro assay. Probes were labeled with 64Cu after the chelation of 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA). Small animal PET imaging and quantitative analysis were performed after injection of 64Cu-labeled Dara-F(ab')2, IgG-F(ab')2, and Dara for evaluation in lymphoma models. RESULTS Flow cytometry and SPR assay proved the specific binding ability of Dara-F(ab')2 and NOTA-Dara-F(ab')2 in vitro. Radiolabeling yield of [64Cu]Cu-NOTA-Dara-F(ab')2 was over 90% and with a specific activity of 4.0 ± 0.6 × 103 MBq/μmol (n = 5). PET imaging showed [64Cu]Cu-NOTA-Dara-F(ab')2 had a rapid and high tumor uptake as early as 2 h (6.9 ± 1.2%ID/g) and peaked (9.5 ± 0.7%ID/g) at 12 h, whereas [64Cu]Cu-NOTA-Dara reached its tumor uptake peaked at 48 h (8.3 ± 1.4%ID/g, n = 4). In comparison, IgG-F(ab')2 and HBL-1 control groups found no noticeable tumor uptake. [64Cu]Cu-NOTA-Dara-F(ab')2 had significantly lower uptake in blood pool, bone, and muscle than [64Cu]Cu-NOTA-Dara and its tumor-to-blood and tumor-to-muscle ratios were significantly higher than controls. CONCLUSIONS [64Cu]Cu-NOTA-Dara-F(ab')2 showed a rapid and high tumor uptake in CD38-positive lymphoma models with favorable imaging contrast, showing its promise as a potential PET imaging agent for future clinical applications.
Collapse
Affiliation(s)
- Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, Xicheng Dist, No. 8 Xishiku Str, Beijing, 100034, China. .,Departments of Radiology and Medical Physics, University of Wisconsin - Madison, K6/562 Clinical Science Center, 600 Highland Ave, Madison, WI, 53705-2275, USA.
| | - Cuicui Li
- Department of Nuclear Medicine, Peking University First Hospital, Xicheng Dist, No. 8 Xishiku Str, Beijing, 100034, China.,Department of Nuclear Medicine, Beijing Friendship Hospital, Beijing, 100050, China
| | - Qi Yang
- Department of Nuclear Medicine, Peking University First Hospital, Xicheng Dist, No. 8 Xishiku Str, Beijing, 100034, China
| | - Logan Sutherlin
- Departments of Radiology and Medical Physics, University of Wisconsin - Madison, K6/562 Clinical Science Center, 600 Highland Ave, Madison, WI, 53705-2275, USA
| | - Lin Wang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Zhao Chen
- Department of Nuclear Medicine, Peking University First Hospital, Xicheng Dist, No. 8 Xishiku Str, Beijing, 100034, China
| | - Kaelyn V Becker
- Departments of Radiology and Medical Physics, University of Wisconsin - Madison, K6/562 Clinical Science Center, 600 Highland Ave, Madison, WI, 53705-2275, USA
| | - Nan Huo
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, 27 Tai-Ping Rd, Beijing, 100850, China
| | - Yongkang Qiu
- Department of Nuclear Medicine, Peking University First Hospital, Xicheng Dist, No. 8 Xishiku Str, Beijing, 100034, China
| | - Jonathan W Engle
- Departments of Radiology and Medical Physics, University of Wisconsin - Madison, K6/562 Clinical Science Center, 600 Highland Ave, Madison, WI, 53705-2275, USA
| | - Rongfu Wang
- Department of Nuclear Medicine, Peking University First Hospital, Xicheng Dist, No. 8 Xishiku Str, Beijing, 100034, China
| | - Chengzhi He
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Dawei Jiang
- Departments of Radiology and Medical Physics, University of Wisconsin - Madison, K6/562 Clinical Science Center, 600 Highland Ave, Madison, WI, 53705-2275, USA. .,Department of Medical Molecular Biology, Beijing Institute of Biotechnology, 27 Tai-Ping Rd, Beijing, 100850, China.
| | - Xiaojie Xu
- Department of Nuclear Medicine, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin - Madison, K6/562 Clinical Science Center, 600 Highland Ave, Madison, WI, 53705-2275, USA.
| |
Collapse
|
7
|
Kumar K, Ghosh A. Radiochemistry, Production Processes, Labeling Methods, and ImmunoPET Imaging Pharmaceuticals of Iodine-124. Molecules 2021; 26:E414. [PMID: 33466827 PMCID: PMC7830191 DOI: 10.3390/molecules26020414] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/07/2021] [Accepted: 01/07/2021] [Indexed: 01/01/2023] Open
Abstract
Target-specific biomolecules, monoclonal antibodies (mAb), proteins, and protein fragments are known to have high specificity and affinity for receptors associated with tumors and other pathological conditions. However, the large biomolecules have relatively intermediate to long circulation half-lives (>day) and tumor localization times. Combining superior target specificity of mAbs and high sensitivity and resolution of the PET (Positron Emission Tomography) imaging technique has created a paradigm-shifting imaging modality, ImmunoPET. In addition to metallic PET radionuclides, 124I is an attractive radionuclide for radiolabeling of mAbs as potential immunoPET imaging pharmaceuticals due to its physical properties (decay characteristics and half-life), easy and routine production by cyclotrons, and well-established methodologies for radioiodination. The objective of this report is to provide a comprehensive review of the physical properties of iodine and iodine radionuclides, production processes of 124I, various 124I-labeling methodologies for large biomolecules, mAbs, and the development of 124I-labeled immunoPET imaging pharmaceuticals for various cancer targets in preclinical and clinical environments. A summary of several production processes, including 123Te(d,n)124I, 124Te(d,2n)124I, 121Sb(α,n)124I, 123Sb(α,3n)124I, 123Sb(3He,2n)124I, natSb(α, xn)124I, natSb(3He,n)124I reactions, a detailed overview of the 124Te(p,n)124I reaction (including target selection, preparation, processing, and recovery of 124I), and a fully automated process that can be scaled up for GMP (Good Manufacturing Practices) production of large quantities of 124I is provided. Direct, using inorganic and organic oxidizing agents and enzyme catalysis, and indirect, using prosthetic groups, 124I-labeling techniques have been discussed. Significant research has been conducted, in more than the last two decades, in the development of 124I-labeled immunoPET imaging pharmaceuticals for target-specific cancer detection. Details of preclinical and clinical evaluations of the potential 124I-labeled immunoPET imaging pharmaceuticals are described here.
Collapse
Affiliation(s)
- Krishan Kumar
- Laboratory for Translational Research in Imaging Pharmaceuticals, The Wright Center of Innovation in Biomedical Imaging, Department of Radiology, The Ohio State University, Columbus, OH 43212, USA;
| | | |
Collapse
|
8
|
Kang L, Li C, Rosenkrans ZT, Engle JW, Wang R, Jiang D, Xu X, Cai W. Noninvasive Evaluation of CD20 Expression Using 64Cu-Labeled F(ab') 2 Fragments of Obinutuzumab in Lymphoma. J Nucl Med 2020; 62:372-378. [PMID: 32826320 DOI: 10.2967/jnumed.120.246595] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/09/2020] [Indexed: 12/11/2022] Open
Abstract
CD20-overexpressed non-Hodgkin lymphoma typically indicates progressive malignancy. Obinutuzumab is a next-generation Food and Drug Administration-approved humanized monoclonal antibody that targets CD20. Previous studies with 89Zr-labeled obinutuzumab have successfully imaged CD20 in vivo. However, delayed tumor uptake and increased radioactive exposure caused by long blood circulation limit its clinical translation. This study aimed to develop 64Cu-labeled F(ab')2 fragments of obinutuzumab for imaging CD20 in lymphoma xenograft tumor models. Methods: F(ab')2 fragments were produced from obinutuzumab using an IgG-degrading enzyme of Streptococcus pyogenes (IdeS) enzyme and purified with protein A beads. Sodium dodecyl sulfate polyacrylamide gel electrophoresis and high-performance liquid chromatography were performed to evaluate the products and their stability. F(ab')2 products were conjugated with p-SCN-Bn-NOTA (NOTA) for 64Cu radiolabeling. Western blotting was performed to screen the CD20 expression levels of lymphoma cells. Enzyme-linked immunosorbent assay, flow cytometry, and confocal imaging were used to test the binding affinity in vitro. Serial PET imaging and biodistribution studies in subcutaneous lymphoma-bearing mice were performed using 64Cu-NOTA-F(ab')2-obinutuzumab or 64Cu-NOTA-F(ab')2-IgG. Results: F(ab')2-obinutuzumab and F(ab')2-IgG produced by the IdeS digestion system were confirmed with sodium dodecyl sulfate polyacrylamide gel electrophoresis and high-performance liquid chromatography. The radiochemical purity of 64Cu-labeled F(ab')2 fragments was no less than 98%, and the specific activity was 56.3 ± 7.9 MBq/mg (n = 6). Among the 5 lymphoma cell lines, Ramos showed the strongest expression of CD20, and CLL-155 showed the lowest, as confirmed by enzyme-linked immunosorbent assay, flow cytometry, and confocal imaging. PET imaging revealed rapid and sustained tumor uptake of 64Cu-NOTA-F(ab')2-obinutuzumab in Ramos tumor-bearing mice. The peak tumor uptake (9.08 ± 1.67 percentage injected dose per gram of tissue [%ID/g]) in the Ramos model was significantly higher than that in the CCL-155 model (2.78 ± 0.62 %ID/g) or the 64Cu-NOTA-F(ab')2-IgG control (1.93 ± 0.26 %ID/g, n = 4, P < 0.001). The tumor-to-blood and tumor-to-muscle ratios were 7.3 ± 1.6 and 21.9 ± 9.0, respectively, at 48 h after injection in the 64Cu-NOTA-F(ab')2-obinutuzumab group. Of the measured off-target organs, the kidneys showed the highest uptake. Ex vivo immunofluorescent staining verified the differential CD20 expression in the Ramos and CCL-155 tumor models. Conclusion: This study demonstrated that 64Cu-NOTA-F(ab')2-obinutuzumab had a rapid and sustained tumor uptake in CD20-positive lymphoma with high contrast, which could enable noninvasive evaluation of CD20 levels in the clinic.
Collapse
Affiliation(s)
- Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China.,Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Cuicui Li
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - Zachary T Rosenkrans
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - Jonathan W Engle
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Rongfu Wang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - Dawei Jiang
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| | - Xiaojie Xu
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing, China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin .,Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
9
|
Kahle XU, Montes de Jesus FM, Glaudemans AWJM, Lub-de Hooge MN, Jorritsma-Smit A, Plattel WJ, van Meerten T, Diepstra A, van den Berg A, Kwee TC, Noordzij W, de Vries EGE, Nijland M. Molecular imaging in lymphoma beyond 18F-FDG-PET: understanding the biology and its implications for diagnostics and therapy. LANCET HAEMATOLOGY 2020; 7:e479-e489. [PMID: 32470439 DOI: 10.1016/s2352-3026(20)30065-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/31/2020] [Accepted: 02/13/2020] [Indexed: 02/08/2023]
Abstract
Mature lymphoproliferative diseases are a heterogeneous group of neoplasms arising from different stages of B-cell and T-cell development. With improved understanding of the molecular processes in lymphoma and novel treatment options, arises a growing need for the molecular characterisation of tumours. Molecular imaging with single-photon-emission CT and PET using specific radionuclide tracers can provide whole-body information to investigate cancer biology, to evaluate phenotypic heterogeneity, to identify resistance to targeted therapy, and to assess the biodistribution of drugs in patients. In this Review, we evaluate the existing literature on molecular imaging in lymphoma, other than 18F-fluordeoxyglucose molecular imaging. The aim is to examine the contribution of molecular imaging to the understanding of the biology of lymphoma and to discuss potential implications for the diagnostics and therapy of this disease. Finally, we discuss possible applications for molecular imaging of patients with lymphoma in the clinical context.
Collapse
Affiliation(s)
- Xaver U Kahle
- Department of Haematology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Filipe M Montes de Jesus
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Andor W J M Glaudemans
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Marjolijn N Lub-de Hooge
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Annelies Jorritsma-Smit
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Wouter J Plattel
- Department of Haematology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Tom van Meerten
- Department of Haematology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Arjan Diepstra
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Anke van den Berg
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Thomas C Kwee
- Department of Radiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Walter Noordzij
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Marcel Nijland
- Department of Haematology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands.
| |
Collapse
|
10
|
Abstract
Immuno-positron emission tomography (immunoPET) is a paradigm-shifting molecular imaging modality combining the superior targeting specificity of monoclonal antibody (mAb) and the inherent sensitivity of PET technique. A variety of radionuclides and mAbs have been exploited to develop immunoPET probes, which has been driven by the development and optimization of radiochemistry and conjugation strategies. In addition, tumor-targeting vectors with a short circulation time (e.g., Nanobody) or with an enhanced binding affinity (e.g., bispecific antibody) are being used to design novel immunoPET probes. Accordingly, several immunoPET probes, such as 89Zr-Df-pertuzumab and 89Zr-atezolizumab, have been successfully translated for clinical use. By noninvasively and dynamically revealing the expression of heterogeneous tumor antigens, immunoPET imaging is gradually changing the theranostic landscape of several types of malignancies. ImmunoPET is the method of choice for imaging specific tumor markers, immune cells, immune checkpoints, and inflammatory processes. Furthermore, the integration of immunoPET imaging in antibody drug development is of substantial significance because it provides pivotal information regarding antibody targeting abilities and distribution profiles. Herein, we present the latest immunoPET imaging strategies and their preclinical and clinical applications. We also emphasize current conjugation strategies that can be leveraged to develop next-generation immunoPET probes. Lastly, we discuss practical considerations to tune the development and translation of immunoPET imaging strategies.
Collapse
Affiliation(s)
- Weijun Wei
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
| | - Zachary T Rosenkrans
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Quan-Yong Luo
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin 53705, United States
| |
Collapse
|
11
|
Wei W, Jiang D, Rosenkrans ZT, Barnhart TE, Engle JW, Luo Q, Cai W. HER2-targeted multimodal imaging of anaplastic thyroid cancer. Am J Cancer Res 2019; 9:2413-2427. [PMID: 31815043 PMCID: PMC6895447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 10/12/2019] [Indexed: 06/10/2023] Open
Abstract
Clinical management of anaplastic thyroid cancer (ATC) is very challenging due to its dedifferentiation and aggressiveness. We aim to develop HER2-targeted multimodal imaging approaches and assess the diagnostic efficacies of these molecular imaging probes in preclinical ATC models. Flow cytometry was used to detect HER2 expression status in thyroid cancer cell lines. We then developed a HER2-specific immunoPET imaging probe 89Zr-Df-pertuzumab by radiolabeling a HER-2 specific monoclonal antibody (mAb) pertuzumab with 89Zr (t1/2=78.4 h) and a fluorescent imaging probe IRDye 800CW-pertuzumab. The diagnostic efficacies of the probes were assessed in subcutaneous and orthotopic ATC models, followed by ex vivo biodistribution profile and immunofluorescence staining studies. HER2 was highly expressed on the surface of all the four primary thyroid cancer cell lines examined, which included two ATC cell lines (i.e., 8505C and THJ-16T). PET imaging with 89Zr-Df-pertuzumab clearly visualized all the subcutaneous ATCs with a peak tumor uptake of 20.23±6.44 %ID/g (n=3), whereas the highest tumor uptake of the nonspecific probe 89Zr-Df-IgG in subcutaneous ATC models was 6.30±0.95 %ID/g (n=3). More importantly, 89Zr-Df-pertuzumab PET imaging strategy readily delineated all the orthotopic ATCs with a peak tumor uptake of 24.93±8.53 %ID/g (n=3). We also suggested that Cerenkov luminescence imaging (CLI) using 89Zr-Df-pertuzumab and fluorescence imaging using IRDye 800CW-pertuzumab are useful tools for image-guided removal of ATCs. We demonstrate that HER2 is a promising biomarker for ATC, and multimodal imaging using 89Zr-Df-pertuzumab and IRDye 800CW-pertuzumab is useful for identifying HER2-postive ATCs.
Collapse
Affiliation(s)
- Weijun Wei
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital600 Yishan Road, Shanghai 200233, China
- Department of Radiology, University of Wisconsin-MadisonMadison, Wisconsin 53705, United States
| | - Dawei Jiang
- Department of Radiology, University of Wisconsin-MadisonMadison, Wisconsin 53705, United States
| | - Zachary T Rosenkrans
- School of Pharmacy, University of Wisconsin-MadisonMadison, Wisconsin 53705, United States
| | - Todd E Barnhart
- Department of Medical Physics, University of Wisconsin-MadisonMadison, Wisconsin 53705, United States
| | - Jonathan W Engle
- Department of Medical Physics, University of Wisconsin-MadisonMadison, Wisconsin 53705, United States
| | - Quanyong Luo
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital600 Yishan Road, Shanghai 200233, China
| | - Weibo Cai
- School of Pharmacy, University of Wisconsin-MadisonMadison, Wisconsin 53705, United States
- Department of Radiology, University of Wisconsin-MadisonMadison, Wisconsin 53705, United States
- Department of Medical Physics, University of Wisconsin-MadisonMadison, Wisconsin 53705, United States
- University of Wisconsin Carbone Cancer CenterMadison, Wisconsin 53705, United States
| |
Collapse
|
12
|
Comparisons of 131I-rituximab treatment responses in patients with aggressive lymphoma and indolent lymphoma. Ann Nucl Med 2019; 33:881-890. [PMID: 31571043 DOI: 10.1007/s12149-019-01401-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 08/31/2019] [Indexed: 12/16/2022]
Abstract
OBJECTIVE We evaluated the changes in treatment response over time after single 131I-rituximab radioimmunotherapy (RIT) according to non-Hodgkin lymphoma (NHL) types. METHODS Fifteen aggressive and 21 indolent lymphoma cases undergoing RIT were evaluated. All patients underwent 18F-FDG-PET-CT before and 5 days, 1, and 3 months after RIT. The maximum standardized uptake value (SUV) and the sum of the products of the longest perpendicular diameters of tumours (SPD) were evaluated. Treatment responses were evaluated 1 and 3 months after RIT RESULTS: In aggressive lymphoma, SUV decreased at 5 days after RIT but increased after that. SPD decreased at 1 month but significantly increased at 3 months. Complete response (CR), partial response (PR), stable disease (SD), and progressive disease (PD) at 1 month after RIT were changed to PD at 3 months after RIT. In indolent lymphoma, the SUV decreased continuously until 1 month after RIT. The SPD significantly decreased at 1 month and tended to further decrease to 3 months. CR, PR, SD, and PD at 1 month after RIT were achieved in 0, 8, 13, and 0 cases, respectively. Among the 13 SD cases, one changed to CR, three changed to PR, and nine had not changed at 3 months after RIT. CONCLUSIONS The treatment response to single RIT differed depending on NHL type. These findings suggest a need to establish an optimal treatment regimen based on NHL aggressiveness.
Collapse
|
13
|
Wei W, Ni D, Ehlerding EB, Luo QY, Cai W. PET Imaging of Receptor Tyrosine Kinases in Cancer. Mol Cancer Ther 2019; 17:1625-1636. [PMID: 30068751 DOI: 10.1158/1535-7163.mct-18-0087] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 04/19/2018] [Accepted: 06/04/2018] [Indexed: 12/22/2022]
Abstract
Overexpression and/or mutations of the receptor tyrosine kinase (RTK) subfamilies, such as epidermal growth factor receptors (EGFR) and vascular endothelial growth factor receptors (VEGFR), are closely associated with tumor cell growth, differentiation, proliferation, apoptosis, and cellular invasiveness. Monoclonal antibodies (mAb) and tyrosine kinase inhibitors (TKI) specifically inhibiting these RTKs have shown remarkable success in improving patient survival in many cancer types. However, poor response and even drug resistance inevitably occur. In this setting, the ability to detect and visualize RTKs with noninvasive diagnostic tools will greatly refine clinical treatment strategies for cancer patients, facilitate precise response prediction, and improve drug development. Positron emission tomography (PET) agents using targeted radioactively labeled antibodies have been developed to visualize tumor RTKs and are changing clinical decisions for certain cancer types. In the present review, we primarily focus on PET imaging of RTKs using radiolabeled antibodies with an emphasis on the clinical applications of these immunoPET probes. Mol Cancer Ther; 17(8); 1625-36. ©2018 AACR.
Collapse
Affiliation(s)
- Weijun Wei
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Department of Radiology, University of Wisconsin-Madison, Wisconsin
| | - Dalong Ni
- Department of Radiology, University of Wisconsin-Madison, Wisconsin
| | - Emily B Ehlerding
- Department of Medical Physics, University of Wisconsin-Madison, Wisconsin
| | - Quan-Yong Luo
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Weibo Cai
- Department of Radiology, University of Wisconsin-Madison, Wisconsin. .,Department of Medical Physics, University of Wisconsin-Madison, Wisconsin.,University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| |
Collapse
|
14
|
18F-labeled anti-human CD20 cys-diabody for same-day immunoPET in a model of aggressive B cell lymphoma in human CD20 transgenic mice. Eur J Nucl Med Mol Imaging 2018; 46:489-500. [PMID: 30456475 DOI: 10.1007/s00259-018-4214-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 11/04/2018] [Indexed: 01/01/2023]
Abstract
PURPOSE Metabolic imaging using [18F]FDG is the current standard for clinical PET; however, some malignancies (e.g., indolent lymphomas) show low avidity for FDG. The majority of B cell lymphomas express CD20, making it a valuable target both for antibody-based therapy and imaging. We previously developed PET tracers based on the humanised anti-CD20 antibody obinutuzumab (GA101). Preclinical studies showed that the smallest bivalent fragment, the cys-diabody (GAcDb, 54.5 kDa) with a peak uptake at 1-2 h post-injection and a biological half-life of 2-5 h, is compatible with short-lived positron emitters such as fluorine-18 (18F, t1/2 110 min), enabling same-day imaging. METHODS GAcDb was radiolabeled using amine-reactive N-succinimidyl 4-[18F]-fluorobenzoate ([18F]SFB), or thiol-reactive N-[2-(4-[18F]-fluorobenzamido)ethyl]maleimide ([18F]FBEM) for site-specific conjugation to C-terminal cysteine residues. Both tracers were used for immunoPET imaging of the B cell compartment in human CD20 transgenic mice (hCD20TM). [18F]FB-GAcDb immunoPET was further evaluated in a disseminated lymphoma (A20-hCD20) syngeneic for hCD20TM and compared to [18F]FDG PET. Tracer uptake was confirmed by ex vivo biodistribution. RESULTS The GAcDb was successfully 18F-radiolabeled using two different conjugation methods resulting in similar specific activities and without impairing immunoreactivity. Both tracers ([18F]FB-GAcDb and [18F]FBEM-GAcDb) specifically target human CD20-expressing B cells in transgenic mice. Fast blood clearance results in high contrast PET images as early as 1 h post injection enabling same-day imaging. [18F]FB-GAcDb immunoPET detects disseminated lymphoma disease in the context of normal tissue expression of hCD20, with comparable sensitivity as [18F]FDG PET but with added specificity for the therapeutic target. CONCLUSIONS [18F]FB-GAcDb and [18F]FBEM-GAcDb could monitor normal B cells and B cell malignancies non-invasively and quantitatively in vivo. In contrast to [18F]FDG PET, immunoPET provides not only information about the extent of disease but also about presence and localisation of the therapeutic target.
Collapse
|
15
|
|
16
|
Kirienko M, Sollini M, Chiti A. Hodgkin lymphoma and imaging in the era of anti-PD-1/PD-L1 therapy. Clin Transl Imaging 2018. [DOI: 10.1007/s40336-018-0294-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
17
|
Yoon JT, Longtine MS, Marquez-Nostra BV, Wahl RL. Evaluation of Next-Generation Anti-CD20 Antibodies Labeled with 89Zr in Human Lymphoma Xenografts. J Nucl Med 2018; 59:1219-1224. [PMID: 29348316 DOI: 10.2967/jnumed.117.203299] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 12/24/2017] [Indexed: 12/31/2022] Open
Abstract
Radioimmunotherapies with monoclonal antibodies to the B-lymphocyte antigen 20 (CD20) are effective treatments for B-cell lymphomas, but U.S. Food and Drug Administration-approved radioimmunotherapies exclusively use radiolabeled murine antibodies, potentially limiting redosing. The Food and Drug Administration recently approved 2 unlabeled anti-CD20 monoclonal antibodies, obinutuzumab and ofatumumab, termed next generation as they are humanized (obinutuzumab) or fully human (ofatumumab), thus potentially allowing a greater potential for redosing than with previous-generation anti-CD20 antibodies, including rituximab (chimeric) and tositumomab (murine), which contain more murine peptide sequences. We prepared 89Zr-ofatumumab and 89Zr-obinituzumab and assessed their tumor targeting by PET/CT imaging and their biodistribution in a preclinical mouse model with CD20 xenografts to determine whether these antibodies have potential as theranostics or for radioimmunotherapy. Methods: Obinutuzumab, ofatumumab, rituximab, tositumomab, and human IgG (as control) were radiolabeled with 89Zr. Raji Burkitt lymphoma xenografts were established in severe combined immunodeficient mice. Mice with palpable tumors (n = 4-9) were injected with 89Zr-obinutuzumab, 89Zr-ofatumumab, 89Zr-rituximab, 89Zr-tositumomab, or 89Zr-IgG, with small-animal PET/CT images acquired at 1, 3, and 7 d after injection, and then sacrificed for biodistribution analyses. Results: At 1, 3, and 7 d after injection, all anti-CD20 antibodies showed clear tumor uptake on PET/CT, with minimal tumor uptake of IgG. Biodistribution data showed significantly (P < 0.005) higher tumor uptake for obinutuzumab (41.4 ± 7.6 percentage injected dose [%ID]/g), ofatumumab (32.6 ± 17.5 %ID/g), rituximab (28.6 ± 7.6 %ID/g), and tositumomab (28.0 ± 6.5 %ID/g) than IgG (7.2 ± 1.2 %ID/g). Tositumomab had much higher splenic uptake (186.4 ± 49.7 %ID/g, P < 0.001) than the other antibodies. Conclusion:89Zr-labeled obinutuzumab and ofatumumab localized to tumor as well as or better than labeled rituximab and tositumomab, 2 monoclonal antibodies that have been used previously in B-cell lymphoma radioimmunotherapy, and both obinutuzumab and ofatumumab have the potential for repeated dosing.
Collapse
Affiliation(s)
- Jason T Yoon
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri
| | - Mark S Longtine
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri
| | | | - Richard L Wahl
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri
| |
Collapse
|
18
|
Ehlerding EB, Lan X, Cai W. "Albumin Hitchhiking" with an Evans Blue Analog for Cancer Theranostics. Theranostics 2018; 8:812-814. [PMID: 29344308 PMCID: PMC5771095 DOI: 10.7150/thno.24183] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 12/05/2017] [Indexed: 12/16/2022] Open
Abstract
Although 177Lu-DOTA-TATE was recently approved in Europe for the treatment of certain neuroendocrine tumors, continued development and optimization has been ongoing to further improve the therapeutic efficacy of somatostatin receptor 2 targeted peptide receptor radionuclide therapy, as well as reducing the renal toxicity. In this work, the use of an Evans blue analog for “albumin hitchhiking” resulted in significant improvement in both the imaging performance and therapeutic efficacy of radiolabeled octreotate, as well as reducing the toxicity since much less radioactivity was used for therapy. Upon clinical translation, such “albumin hitchhiking” could make significant impact in the near future for cancer patient management.
Collapse
|
19
|
Wei W, Ehlerding EB, Lan X, Luo Q, Cai W. PET and SPECT imaging of melanoma: the state of the art. Eur J Nucl Med Mol Imaging 2018; 45:132-150. [PMID: 29085965 PMCID: PMC5700861 DOI: 10.1007/s00259-017-3839-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 09/18/2017] [Indexed: 12/12/2022]
Abstract
Melanoma represents the most aggressive form of skin cancer, and its incidence continues to rise worldwide. 18F-FDG PET imaging has transformed diagnostic nuclear medicine and has become an essential component in the management of melanoma, but still has its drawbacks. With the rapid growth in the field of nuclear medicine and molecular imaging, a variety of promising probes that enable early diagnosis and detection of melanoma have been developed. The substantial preclinical success of melanin- and peptide-based probes has recently resulted in the translation of several radiotracers to clinical settings for noninvasive imaging and treatment of melanoma in humans. In this review, we focus on the latest developments in radiolabeled molecular imaging probes for melanoma in preclinical and clinical settings, and discuss the challenges and opportunities for future development.
Collapse
Affiliation(s)
- Weijun Wei
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600# Yishan Road, Shanghai, 200233, China
- Department of Radiology, University of Wisconsin-Madison, Room 7137, 1111 Highland Avenue, Madison, WI, 53705-2275, USA
| | - Emily B Ehlerding
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Molecular Imaging, No. 1277 Jiefang Ave, Wuhan, 430022, China.
| | - Quanyong Luo
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600# Yishan Road, Shanghai, 200233, China.
| | - Weibo Cai
- Department of Radiology, University of Wisconsin-Madison, Room 7137, 1111 Highland Avenue, Madison, WI, 53705-2275, USA.
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- University of Wisconsin Carbone Cancer Center, Madison, WI, 53705, USA.
| |
Collapse
|
20
|
Zhao Q, Xie D, Liu H, Wang F, Yan GY, Chen X. SSCMDA: spy and super cluster strategy for MiRNA-disease association prediction. Oncotarget 2017; 9:1826-1842. [PMID: 29416734 PMCID: PMC5788602 DOI: 10.18632/oncotarget.22812] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 10/30/2017] [Indexed: 12/23/2022] Open
Abstract
In the biological field, the identification of the associations between microRNAs (miRNAs) and diseases has been paid increasing attention as an extremely meaningful study for the clinical medicine. However, it is expensive and time-consuming to confirm miRNA-disease associations by experimental methods. Therefore, in recent years, several effective computational models for predicting the potential miRNA-disease associations have been developed. In this paper, we proposed the Spy and Super Cluster strategy for MiRNA-Disease Association prediction (SSCMDA) based on known miRNA-disease associations, integrated disease similarity and integrated miRNA similarity. For problems of mixed unknown miRNA-disease pairs containing both potential associations and real negative associations, which will lead to inaccurate prediction, spy strategy is adopted by SSCMDA to identify reliable negative samples from the unknown miRNA-disease pairs. Moreover, the super-cluster strategy could gather as many positive samples as possible to improve the accuracy of the prediction by overcoming the shortage of lacking sufficient positive training samples. As a result, the AUCs of global leave-one-out cross validation (LOOCV), local LOOCV and 5-fold cross validation were 0.9007, 0.8747 and 0.8806+/-0.0025, respectively. According to the AUC results, SSCMDA has shown a significant improvement compared with some previous models. We further carried out case studies based on various version of HMDD database to test the prediction performance robustness of SSCMDA. We also implemented case study to examine whether SSCMDA was effective for new diseases without any known associated miRNAs. As a result, a large proportion of the predicted miRNAs have been verified by experimental reports.
Collapse
Affiliation(s)
- Qi Zhao
- School of Mathematics, Liaoning University, Shenyang, China.,Research Center for Computer Simulating and Information Processing of Bio-Macromolecules of Liaoning Province, Shenyang, China
| | - Di Xie
- School of Mathematics, Liaoning University, Shenyang, China
| | - Hongsheng Liu
- Research Center for Computer Simulating and Information Processing of Bio-Macromolecules of Liaoning Province, Shenyang, China.,School of Life Science, Liaoning University, Shenyang, China
| | - Fan Wang
- School of Mechatronic Engineering, China University of Mining and Technology, Xuzhou, China.,Jiangsu Key Laboratory of Mine Mechanical and Electrical Equipment, China University of Mining and Technology, Xuzhou, China
| | - Gui-Ying Yan
- Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, China
| | - Xing Chen
- School of Information and Control Engineering, China University of Mining and Technology, Xuzhou, China
| |
Collapse
|