58 results on '"Christopher G. England"'
Search Results
2. Supplementary Methods from Dual Targeting of Tissue Factor and CD105 for Preclinical PET Imaging of Pancreatic Cancer
- Author
-
Weibo Cai, Robert J. Nickles, Hing C. Wong, Charles P. Theuer, Bai Liu, Reinier Hernandez, Stephen A. Graves, Sixiang Shi, Christopher G. England, and Haiming Luo
- Abstract
Determination of NOTA to Fab Fragment Ratio
- Published
- 2023
3. Supplementary Figures from Dual Targeting of Tissue Factor and CD105 for Preclinical PET Imaging of Pancreatic Cancer
- Author
-
Weibo Cai, Robert J. Nickles, Hing C. Wong, Charles P. Theuer, Bai Liu, Reinier Hernandez, Stephen A. Graves, Sixiang Shi, Christopher G. England, and Haiming Luo
- Abstract
Supplementary Figure 1. Purification of heterodimer after click chemistry conjugation of ALT-836-Fab and TRC105-Fab Supplementary Figure 2. MALDI-TOF mass spectra of ALT-836-Fab: [M+H]+ = 47266.0, TRC105-Fab: [M+H]+ = 47454.7, and heterodimer: exact [M+H]+ = 103504.0. Supplementary Figure 3. Flow cytometry analysis in BXPC-3 after 1 h incubation of FITC-Fab conjugates for the comparison of their in vitro targeting Supplementary Figure 4. Radiolabeling efficiency at different copper: immunoconjugate ratios were used to determine the number of NOTA per antibody fragment molecule Supplementary Figure 5. PET imaging and biodistribution of 64CuCl2 in BXPC-3 tumor-bearing mice. Supplementary Figure 6. PET imaging and quantitative ROI analysis of 64Cu-NOTA-heterodimer with TF and CD105 dual-blocking in BXPC-3 tumor-bearing mice. Supplementary Figure 7. Coronal images and transverse PET/CT images of normal nude mice at 3, 15, 24 and 30 h following injection of 64Cu-NOTA-heterodimer Supplementary Figure 8. PET ROI-derived quantification of 64Cu-NOTA-heterodimer uptake in the tumor, normal pancreas, blood pool, liver, kidney and muscle at 3 h, 15 h, 24 h and 30 h p.i. of the tracer
- Published
- 2023
4. Data from Dual Targeting of Tissue Factor and CD105 for Preclinical PET Imaging of Pancreatic Cancer
- Author
-
Weibo Cai, Robert J. Nickles, Hing C. Wong, Charles P. Theuer, Bai Liu, Reinier Hernandez, Stephen A. Graves, Sixiang Shi, Christopher G. England, and Haiming Luo
- Abstract
Purpose: Pancreatic adenocarcinoma is a highly aggressive cancer, currently treated with limited success and dismal outcomes. New diagnostic and treatment strategies offer the potential to reduce cancer mortality. Developing highly specific noninvasive imaging probes for pancreatic cancer is essential to improving diagnostic accuracy and monitoring therapeutic intervention.Experimental Design: A bispecific heterodimer was synthesized by conjugating an anti-tissue factor (TF) Fab with an anti-CD105 Fab, via the bio-orthogonal “click” reaction between tetrazine (Tz) and trans-cyclooctene (TCO). The heterodimer was labeled with 64Cu for PET imaging of nude mice bearing BXPC-3 xenograft and orthotopic pancreatic tumors.Results: PET imaging of BXPC-3 (TF/CD105+/+) xenograft tumors with 64Cu-labeled heterodimer displayed significantly enhanced tumor uptake (28.8 ± 3.2 %ID/g; n = 4; SD) at 30 hours postinjection, as compared with each of their monospecific Fab tracers (12.5 ± 1.4 and 7.1 ± 2.6 %ID/g; n = 3; SD). In addition, the activity–concentration ratio allowed for effective tumor visualization (tumor/muscle ratio 75.2 ± 9.4 at 30 hours postinjection.; n = 4; SD). Furthermore, 64Cu-NOTA-heterodimer enabled sensitive detection of orthotopic pancreatic tumor lesions with an uptake of 17.1 ± 4.9 %ID/g at 30 hours postinjection and tumor/muscle ratio of 72.3 ± 46.7.Conclusions: This study demonstrates that dual targeting of TF and CD105 provided synergistic improvements in binding affinity and tumor localization of the heterodimer. Dual-targeted imaging agents of pancreatic and other cancers may assist in diagnosing pancreatic malignancies as well as reliable monitoring of therapeutic response. Clin Cancer Res; 22(15); 3821–30. ©2016 AACR.
- Published
- 2023
5. Detection of Phosphatidylcholine-Coated Gold Nanoparticles in Orthotopic Pancreatic Adenocarcinoma using Hyperspectral Imaging.
- Author
-
Christopher G England, Justin S Huang, Kurtis T James, Guandong Zhang, André M Gobin, and Hermann B Frieboes
- Subjects
Medicine ,Science - Abstract
Nanoparticle uptake and distribution to solid tumors are limited by reticuloendothelial system systemic filtering and transport limitations induced by irregular intra-tumoral vascularization. Although vascular enhanced permeability and retention can aid targeting, high interstitial fluid pressure and dense extracellular matrix may hinder local penetration. Extravascular diffusivity depends upon nanoparticle size, surface modifications, and tissue vascularization. Gold nanoparticles functionalized with biologically-compatible layers may achieve improved uptake and distribution while enabling cytotoxicity through synergistic combination of chemotherapy and thermal ablation. Evaluation of nanoparticle uptake in vivo remains difficult, as detection methods are limited. We employ hyperspectral imaging of histology sections to analyze uptake and distribution of phosphatidylcholine-coated citrate gold nanoparticles (CGN) and silica-gold nanoshells (SGN) after tail-vein injection in mice bearing orthotopic pancreatic adenocarcinoma. For CGN, the liver and tumor showed 26.5 ± 8.2 and 23.3 ± 4.1 particles/100 μm2 within 10 μm from the nearest source and few nanoparticles beyond 50 μm, respectively. The spleen had 35.5 ± 9.3 particles/100 μm2 within 10 μm with penetration also limited to 50 μm. For SGN, the liver showed 31.1 ± 4.1 particles/100 μm2 within 10 μm of the nearest source with penetration hindered beyond 30 μm. The spleen and tumor showed uptake of 22.1 ± 6.2 and 15.8 ± 6.1 particles/100 μm2 within 10 μm, respectively, with penetration similarly hindered. CGH average concentration (nanoparticles/μm2) was 1.09 ± 0.14 in the liver, 0.74 ± 0.12 in the spleen, and 0.43 ± 0.07 in the tumor. SGN average concentration (nanoparticles/μm2) was 0.43 ± 0.07 in the liver, 0.30 ± 0.06 in the spleen, and 0.20 ± 0.04 in the tumor. Hyperspectral imaging of histology sections enables analysis of phosphatidylcholine-coated gold-based nanoparticles in pancreatic tumors with the goal to improve nanotherapeutic efficacy.
- Published
- 2015
- Full Text
- View/download PDF
6. Efficient renal clearance of DNA tetrahedron nanoparticles enables quantitative evaluation of kidney function
- Author
-
Christopher G. England, Dawei Jiang, Hyung Jun Im, Xiaoli Lan, Dalong Ni, Peng Huang, Lei Kang, Weibo Cai, Jonathan W. Engle, and Madeline E Boleyn
- Subjects
Kidney ,Computer science ,Kidney dysfunction ,Renal function ,02 engineering and technology ,Computational biology ,urologic and male genital diseases ,010402 general chemistry ,021001 nanoscience & nanotechnology ,Condensed Matter Physics ,01 natural sciences ,Article ,Atomic and Molecular Physics, and Optics ,0104 chemical sciences ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,Dna nanostructures ,DNA nanotechnology ,medicine ,General Materials Science ,Electrical and Electronic Engineering ,0210 nano-technology ,DNA ,Clearance - Abstract
DNA tetrahedron nanostructure (DTN) is one of the simplest DNA nanostructures and has been successfully applied for biosensing, imaging, and treatment of cancer. To facilitate its biomedical applications and potential clinical translation, fundamental understanding of DTN's transportation among major organs in living organisms becomes increasingly important. Here, we describe the efficient renal clearance of DTN in healthy mice by using positron emission tomography (PET) imaging. The kidney elimination of DTN was later applied for renal function evaluation in murine models of unilateral ureteral obstruction (UUO). We further established a mathematical program of DTN to validate its changes of transportation pattern in healthy and UUO mice. We believe the establishment of pharmacokinetic profiles and mathematical model of DTN may provide insight for future optimization of DNA nanostructures for biomedical applications.
- Published
- 2018
7. Physiology in Perspective
- Author
-
Christopher G England
- Subjects
Physiology ,Perspective (graphical) ,MEDLINE ,Biology ,Epistemology - Published
- 2022
8. Predictive modeling of in vivo response to gemcitabine in pancreatic cancer.
- Author
-
James J Lee, Justin Huang, Christopher G England, Lacey R McNally, and Hermann B Frieboes
- Subjects
Biology (General) ,QH301-705.5 - Abstract
A clear contradiction exists between cytotoxic in-vitro studies demonstrating effectiveness of Gemcitabine to curtail pancreatic cancer and in-vivo studies failing to show Gemcitabine as an effective treatment. The outcome of chemotherapy in metastatic stages, where surgery is no longer viable, shows a 5-year survival
- Published
- 2013
- Full Text
- View/download PDF
9. DNA origami nanostructures can exhibit preferential renal uptake and alleviate acute kidney injury
- Author
-
Christopher G. England, Hao Yan, Zhilei Ge, Dawei Jiang, Christopher J. Kutyreff, Yan Liu, Steve Y. Cho, Peng Huang, Chunhai Fan, Junjun Hou, Hyung Jun Im, Jiye Shi, Dalong Ni, Weibo Cai, Jonathan W. Engle, Luhao Zhang, and Yongjun Yan
- Subjects
Biodistribution ,Pathology ,medicine.medical_specialty ,medicine.medical_treatment ,Biomedical Engineering ,Medicine (miscellaneous) ,Renal function ,Bioengineering ,02 engineering and technology ,urologic and male genital diseases ,010402 general chemistry ,01 natural sciences ,Nephrotoxicity ,medicine ,DNA origami ,Kidney transplantation ,Dialysis ,Kidney ,urogenital system ,business.industry ,Acute kidney injury ,021001 nanoscience & nanotechnology ,medicine.disease ,0104 chemical sciences ,Computer Science Applications ,medicine.anatomical_structure ,0210 nano-technology ,business ,Biotechnology - Abstract
Patients with acute kidney injury (AKI) frequently require kidney transplantation and supportive therapies, such as rehydration and dialysis. Here, we show that radiolabelled DNA origami nanostructures (DONs) with rectangular, triangular and tubular shapes accumulate preferentially in the kidneys of healthy mice and mice with rhabdomyolysis-induced AKI, and that rectangular DONs have renal-protective properties, with efficacy similar to the antioxidant N-acetylcysteine—a clinically used drug that ameliorates contrast-induced AKI and protects kidney function from nephrotoxic agents. We evaluated the biodistribution of DONs non-invasively via positron emission tomography, and the efficacy of rectangular DONs in the treatment of AKI via dynamic positron emission tomography imaging with 68Ga-EDTA, blood tests and kidney tissue staining. DNA-based nanostructures could become a source of therapeutic agents for the treatment of AKI and other renal diseases. DNA origami nanostructures with different shapes can accumulate preferentially in the kidney, with some being renal-protective, as shown in healthy mice and in a mouse model of acute kidney injury.
- Published
- 2018
10. Evaluation of the biological activities of the IL-15 superagonist complex, ALT-803, following intravenous versus subcutaneous administration in murine models
- Author
-
Emily K. Jeng, Terra Noel, Sarah Alter, Bai Liu, Lin Kong, Jeffrey S. Miller, Weibo Cai, Sixiang Shi, Christopher G. England, Peter R. Rhode, Hing C. Wong, and Monica Brown Jones
- Subjects
0301 basic medicine ,Injections, Subcutaneous ,Recombinant Fusion Proteins ,medicine.medical_treatment ,T cell ,Immunology ,Antineoplastic Agents ,CD8-Positive T-Lymphocytes ,Pharmacology ,Lymphocyte Activation ,Biochemistry ,Article ,Mice ,03 medical and health sciences ,Pharmacokinetics ,In vivo ,Cell Line, Tumor ,medicine ,Animals ,Immunology and Allergy ,Tissue Distribution ,Molecular Biology ,Cell Proliferation ,Interleukin-15 ,Chemistry ,Proteins ,Interleukin ,Hematology ,Immunotherapy ,Xenograft Model Antitumor Assays ,Killer Cells, Natural ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Interleukin 15 ,Toxicity ,Administration, Intravenous ,Female ,CD8 - Abstract
ALT-803 is a fusion protein complex consisting of an interleukin (IL)-15 superagonist and a dimeric IL-15 receptor alpha sushi domain IgG1 Fc fusion protein. When administered to mice, ALT-803 is capable of inducing natural killer (NK) and CD8(+) T cell proliferation and activation, and effectively promoting potent anti-tumor responses. Currently, ALT-803 is in clinical trials for treatment of various solid tumors and hematological malignancies. In the initial phase of these clinical studies, intravenous (iv) injection was used according to the route used in pre-clinical efficacy studies. In order to evaluate the possible advantage of subcutaneous (sc) injection versus iv injection, this study compared the biological activity of the two treatment regimens of ALT-803 in pre-clinical in vivo models. The pharmacokinetics, immune stimulation, and anti-tumor efficacy of iv and sc injection routes of ALT-803 in C57BL/6 mice were compared. The half-life of ALT-803 was 7.5 h for iv versus 7.7 h for sc with the maximal detected serum concentration of ALT-803 to be 3926 ng/ml at 0.5 h time-point following iv injection versus 495 ng/ml at 16 h post sc injection. Biodistribution studies indicated that sc ALT-803, similarly to iv ALT-803 as previously reported, has a greater tissue distribution and longer residence time in lymphoid tissues compared to recombinant IL-15. Notably, ALT-803 when administered either iv or sc induced comparable proliferation and activation of CD8(+) T and NK cells and resulted in similar reductions of tumor burden. A toxicity study of mice receiving multiple injections of ALT-803 for 4 weeks by iv or sc routes revealed equivalent immune-related changes. The gradual absorbance into the blood stream and lower maximal blood levels of ALT-803 in sc-injected mice, along with similar anti-tumor efficacy support the administration of ALT-803 by sc injection in patients with various malignancies and infectious diseases.
- Published
- 2018
11. ImmunoPET imaging of tissue factor expression in pancreatic cancer with 89Zr-Df-ALT-836
- Author
-
Christopher G. England, Reinier Hernandez, Weibo Cai, Todd E. Barnhart, Hector F. Valdovinos, Hing C. Wong, Bai Liu, and Yunan Yang
- Subjects
0301 basic medicine ,Biodistribution ,Pathology ,medicine.medical_specialty ,medicine.drug_class ,Mice, Nude ,Siderophores ,Pharmaceutical Science ,Deferoxamine ,Biology ,Kidney ,Monoclonal antibody ,Article ,Thromboplastin ,Flow cytometry ,03 medical and health sciences ,Tissue factor ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,Pancreatic cancer ,medicine ,Animals ,Humans ,Tissue Distribution ,Radioisotopes ,medicine.diagnostic_test ,medicine.disease ,Recombinant Proteins ,Pancreatic Neoplasms ,030104 developmental biology ,Liver ,Positron emission tomography ,Immunoglobulin G ,Positron-Emission Tomography ,030220 oncology & carcinogenesis ,Female ,Zirconium ,Radiopharmaceuticals ,Spleen ,Ex vivo - Abstract
Overexpression of tissue factor (TF) has been associated with increased tumor growth, tumor angiogenesis, and metastatic potential in many malignancies, including pancreatic cancer. Additionally, high TF expression was shown to strongly correlate with poor prognoses and decreased survival in pancreatic cancer patients. Herein, we exploited the potential targeting of TF for positron emission tomography (PET) imaging of pancreatic cancer. The TF-targeted tracer was developed through radiolabeling of the anti-human TF monoclonal antibody (ALT-836) with 89Zr. The tracer was characterized by fluorescence microscopy and flow cytometry assays in BXPC-3 and PANC-1 cells, two pancreatic cancer cell lines with high and low TF expression levels, respectively. Non-invasive PET scans were acquired in tumor-bearing mice injected with 89Zr-Df-ALT-836. Additionally, ex vivo biodistribution, blocking, and histological studies were performed to establish the affinity and specificity of 89Zr-Df-ALT-836 for TF in vivo. 89Zr-labeling of Df-ALT-836 was achieved in high yield and good specific activity. FACS and microscopy studies revealed no detectable difference in TF-binding affinity between ALT-836 and Df-ALT-836 in vitro. Longitudinal PET scans unveiled a lasting and prominent 89Zr-Df-ALT-836 uptake in BXPC-3 tumors (peak at 31.5 ± 6.0 %ID/g at 48 h post-injection; n=3), which was significantly abrogated (2.3 ± 0.5 %ID/g at 48 h post-injection; n=3) when mice were pre-injected with a blocking dose (50 mg/kg) of unlabeled ALT-836. Ex vivo biodistribution data confirmed the accuracy of the PET results, and histological analysis correlated high tumor uptake with in situ TF expression. Taken together, these results attest to the excellent affinity and TF-specificity of 89Zr-Df-ALT-836 in vivo. With elevated, persistent, and specific accumulation in TF-positive BXPC-3 tumors, PET imaging using 89Zr-Df-ALT-836 promises to open new avenues for improving future diagnosis, stratification, and treatment response assessment in pancreatic cancer patients.
- Published
- 2017
12. 89Zr-labeled nivolumab for imaging of T-cell infiltration in a humanized murine model of lung cancer
- Author
-
Todd E. Barnhart, Emily B. Ehlerding, Paul A. Ellison, Christopher G. England, Reinier Hernandez, Dawei Jiang, Brian T. Rekoske, Peng Huang, Weibo Cai, and Douglas G. McNeel
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Biodistribution ,medicine.drug_class ,business.industry ,medicine.medical_treatment ,General Medicine ,Immunotherapy ,Monoclonal antibody ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,In vivo ,030220 oncology & carcinogenesis ,medicine ,Immunohistochemistry ,Radiology, Nuclear Medicine and imaging ,Nivolumab ,business ,Lung cancer ,Ex vivo - Abstract
Nivolumab is a human monoclonal antibody specific for programmed cell death-1 (PD-1), a negative regulator of T-cell activation and response. Acting as an immune checkpoint inhibitor, nivolumab binds to PD-1 expressed on the surface of many immune cells and prevents ligation by its natural ligands. Nivolumab is only effective in a subset of patients, and there is limited evidence supporting its use for diagnostic, monitoring, or stratification purposes. 89Zr-Df-nivolumab was synthesized to map the biodistribution of PD-1-expressing tumor infiltrating T-cells in vivo using a humanized murine model of lung cancer. The tracer was developed by radiolabeling the antibody with the positron emitter zirconium-89 (89Zr). Imaging results were validated by ex vivo biodistribution studies, and PD-1 expression was validated by immunohistochemistry. Data obtained from PET imaging were used to determine human dosimetry estimations. The tracer showed elevated binding to stimulated PD-1 expressing T-cells in vitro and in vivo. PET imaging of 89Zr-Df-nivolumab allowed for clear delineation of subcutaneous tumors through targeting of localized activated T-cells expressing PD-1 in the tumors and salivary glands of humanized A549 tumor-bearing mice. In addition to tumor uptake, salivary and lacrimal gland infiltration of T-cells was noticeably visible and confirmed via histological analysis. These data support our claim that PD-1-targeted agents allow for tumor imaging in vivo, which may assist in the design and development of new immunotherapies. In the future, noninvasive imaging of immunotherapy biomarkers may assist in disease diagnostics, disease monitoring, and patient stratification.
- Published
- 2017
13. CD38 as a PET Imaging Target in Lung Cancer
- Author
-
Todd E. Barnhart, Emily B. Ehlerding, Saige Lacognata, Dawei Jiang, Lei Kang, Weibo Cai, Stephen A. Graves, and Christopher G. England
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Lung Neoplasms ,Blotting, Western ,Mice, Nude ,Pharmaceutical Science ,Deferoxamine ,CD38 ,Article ,Flow cytometry ,Mice ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,Drug Discovery ,medicine ,Animals ,Humans ,Lung cancer ,Multiple myeloma ,medicine.diagnostic_test ,business.industry ,Antibodies, Monoclonal ,Daratumumab ,Flow Cytometry ,medicine.disease ,ADP-ribosyl Cyclase 1 ,030104 developmental biology ,A549 Cells ,Cell culture ,Positron-Emission Tomography ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Molecular Medicine ,Female ,Zirconium ,business - Abstract
Daratumumab (Darzalex, Janssen Biotech) is a clinically approved antibody targeting CD38 for the treatment of multiple myeloma. However, CD38 is also expressed by other cancer cell types, including lung cancer, where its expression or absence may offer prognostic value. We therefore developed a PET tracer based upon daratumumab for tracking CD38 expression, utilizing murine models of non-small cell lung cancer to verify its specificity. Daratumumab was prepared for radiolabeling with 89Zr (t1/2 = 78.4 h) through conjugation with desferrioxamine (Df). Western blot, flow cytometry, and saturation binding assays were utilized to characterize CD38 expression and binding of daratumumab to three non-small cell lung cancer cell lines: A549, H460, and H358. Murine xenograft models of the cell lines were also generated for further in vivo studies. Longitudinal PET imaging was performed following injection of 89Zr-Df–daratumumab out to 120 h postinjection, and nonspecific uptake was also evaluated through the injection of a radiolabeled control IgG antibody in A549 mice, 89Zr-Df–IgG. Ex vivo biodistribution and histological analyses were also performed after the terminal imaging time point at 120 h postinjection. Through cellular studies, A549 cells were found to express higher levels of CD38 than the H460 or H358 cell lines. PET imaging and ex vivo biodistribution studies verified in vitro trends, with A549 tumor uptake peaking at 8.1 ± 1.2%ID/g at 120 h postinjection according to PET analysis, and H460 and H358 at lower levels at the same time point (6.7 ± 0.7%ID/g and 5.1 ± 0.4%ID/g, respectively; n = 3 or 4). Injection of a nonspecific radiolabeled IgG into A549 tumor-bearing mice also demonstrated lower tracer uptake of 4.4 ± 1.3%ID/g at 120 h. Immunofluorescent staining of tumor tissues showed higher staining levels present in A549 tissues over H460 and H358. Thus, 89Zr-Df–daratumumab is able to image CD38-expressing tissues in vivo using PET, as verified through the exploration of non-small cell lung cancer models in this study. This agent therefore holds potential to image CD38 in other malignancies and aid in patient stratification and elucidation of the biodistribution of CD38.
- Published
- 2017
14. ImmunoPET Imaging of CTLA-4 Expression in Mouse Models of Non-small Cell Lung Cancer
- Author
-
Christopher G. England, Weibo Cai, Dawei Jiang, Robert J. Nickles, Rebecca L. Majewski, Hector F. Valdovinos, Glenn Liu, Douglas G. McNeel, and Emily B. Ehlerding
- Subjects
0301 basic medicine ,Biodistribution ,Pathology ,medicine.medical_specialty ,Lung Neoplasms ,medicine.drug_class ,medicine.medical_treatment ,Pharmaceutical Science ,Ipilimumab ,Biology ,Monoclonal antibody ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Drug Discovery ,medicine ,Animals ,Humans ,Cytotoxic T cell ,CTLA-4 Antigen ,Lung cancer ,neoplasms ,Immunotherapy ,medicine.disease ,030104 developmental biology ,CTLA-4 ,Positron-Emission Tomography ,030220 oncology & carcinogenesis ,Cancer research ,Molecular Medicine ,Ex vivo ,medicine.drug - Abstract
Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) is expressed on the surface of activated T cells and some tumor cells, and is the target of the clinically-approved monoclonal antibody ipilimumab. In this study, we investigate specific binding of radiolabeled ipilimumab to CTLA-4 expressed by human non-small cell lung cancer cells in vivo using positron emission tomography (PET). Ipilimumab was radiolabeled with 64Cu (t1/2 = 12.7 h) through the use of the chelator 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) to formulate 64Cu-DOTA-ipilimumab. CTLA-4 expression in three non-small cell lung cancer (NSCLC) cell lines (A549, H460, and H358) was verified and quantified by Western blot and enzyme-linked immunosorbent assays (ELISA). A receptor binding assay was utilized to monitor the binding and internalization of 64Cu-DOTA-ipilimumab in the NSCLC cell lines. Next, the biodistribution of 64Cu-DOTA-ipilimumab was mapped by longitudinal PET imaging up to 48 h after injection. Ex vivo biodistribution and histological studies were employed to verify PET results. By in vitro analysis, CTLA-4 was found to be expressed on all three NSCLC cell lines with A549 and H358 showing the highest and lowest level of expression, respectively. PET imaging and quantification verified these findings as the tracer accumulated highest in the A549 tumor model (9.80 ± 0.22 %ID/g at 48 h after injection; n=4), followed by H460 and H358 tumors with uptakes of 9.37 ± 0.26 %ID/g and 7.43 ± 0.05 %ID/g, respectively (n=4). The specificity of the tracer was verified by injecting excess ipilimumab in A549 tumor-bearing mice, which decreased tracer uptake to 6.90 ± 0.51 %ID/g at 48 after injection (n=4). Ex vivo analysis following the last imaging session also corroborated these findings. 64Cu-DOTA-ipilimumab showed enhanced and persistent accumulation in CTLA-4-expressing tissues, which will enable researchers further insight into CTLA-4 targeted therapies in the future.
- Published
- 2017
15. Radiolabeled pertuzumab for imaging of human epidermal growth factor receptor 2 expression in ovarian cancer
- Author
-
Hector F. Valdovinos, Robert J. Nickles, Steve Y. Cho, Emily B. Ehlerding, Hyung Jun Im, Christopher G. England, Dong Soo Lee, Haiyan Sun, Weibo Cai, Dawei Jiang, and Peng Huang
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,endocrine system diseases ,Receptor, ErbB-2 ,Antibodies, Monoclonal, Humanized ,medicine.disease_cause ,Article ,Metastasis ,Heterocyclic Compounds, 1-Ring ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Internal medicine ,Animals ,Humans ,Medicine ,Tissue Distribution ,Radiology, Nuclear Medicine and imaging ,Radiometry ,skin and connective tissue diseases ,Human Epidermal Growth Factor Receptor 2 ,Ovarian Neoplasms ,medicine.diagnostic_test ,business.industry ,General Medicine ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,030104 developmental biology ,Copper Radioisotopes ,Positron emission tomography ,Isotope Labeling ,Positron-Emission Tomography ,030220 oncology & carcinogenesis ,Biomarker (medicine) ,Female ,Pertuzumab ,Molecular imaging ,business ,Ovarian cancer ,Carcinogenesis ,medicine.drug - Abstract
Human epidermal growth factor receptor 2 (HER2) is over-expressed in over 30% of ovarian cancer cases, playing an essential role in tumorigenesis and metastasis. Non-invasive imaging of HER2 is of great interest for physicians as a mean to better detect and monitor the progression of ovarian cancer. In this study, HER2 was assessed as a biomarker for ovarian cancer imaging usingHER2 expression and binding were examined in three ovarian cancer cell lines (SKOV3, OVCAR3, Caov3) using in vitro techniques, including western blot and saturation binding assays. PET imaging and biodistribution studies in subcutaneous models of ovarian cancer were performed for non-invasive in vivo evaluation of HER2 expression. Additionally, orthotopic models were employed to further validate the imaging capability ofHER2 expression was highest in SKOV3 cells, while OVCAR3 and Caov3 displayed lower HER2 expression.
- Published
- 2017
16. ImmunoPET imaging of CD38 expression in hepatocellular carcinoma using
- Author
-
Shiyong, Li, Christopher G, England, Emily B, Ehlerding, Christopher J, Kutyreff, Jonathan W, Engle, Dawei, Jiang, and Weibo, Cai
- Subjects
Original Article ,digestive system diseases - Abstract
CD38 is expressed on the surface of many immune cells, which are closely associated with antitumor immunity and immune tolerance of tumor cells. Therefore, monitoring CD38 expression has gained great attention for tracking the progression of tumors and cancer treatment. Herein, we aim to develop a PET tracer using an anti-CD38 monoclonal antibody (daratumumab) to monitor CD38 expression in hepatocellular carcinoma (HCC). In this study, daratumumab was radiolabeled with (64)Cu (t(1/2)=12.7 h) to obtain (64)Cu-NOTA-daratumumab. Relative CD38 expression in HepG2 and Huh7 HCC cell lines was assessed using western blot. The specificity of (64)Cu-NOTA-daratumumab to both cell lines was examined using an in vitro cell-binding assay. PET imaging in subcutaneous models of HCC was performed to evaluate the capability and specificity of (64)Cu-NOTA-daratumumab to target CD38 in vivo. Region-of-interest analysis and ex vivo biodistribution were performed to verify the tracer targeting capability of CD38. Through cellular studies of two HCC cell lines, CD38 expression was found to be higher in HepG2 and minimal in Huh7 cells. (64)Cu-NOTA-daratumumab showed relatively high affinity to CD38 (K(a)=18.21 ± 1.74 nM), while the affinity of Huh7 was in the micromolar range for daratumumab binding to the cells (K(a)=3.98 ± 0.87 μM). At 48 h post-injection, PET imaging of subcutaneous models with (64)Cu-NOTA-daratumumab revealed tumor uptakes of 12.23 ± 2.4 and 2.7 ± 1.2 %ID/g for HepG2 and Huh7, respectively (n=4), which correlated well with relative CD38 expression of the cells. Moreover, the (64)Cu-NOTA-IgG nonspecific analogue showed a significantly lower uptake in HepG2 subcutaneous model in mice, suggesting a specific binding of daratumumab with CD38 in vivo. Our cellular studies and PET imaging confirmed the capability and specificity of (64)Cu-NOTA-daratumumab for the imaging of CD38 in murine models of HCC. This study supports our claim that (64)Cu-NOTA-daratumumab is an effective PET tracer for the non-invasive evaluation of CD38 expression and sensitive detection of CD38-positive tumor lesions in HCC.
- Published
- 2019
17. DNA origami nanostructures can exhibit preferential renal uptake and alleviate acute kidney injury
- Author
-
Dawei, Jiang, Zhilei, Ge, Hyung-Jun, Im, Christopher G, England, Dalong, Ni, Junjun, Hou, Luhao, Zhang, Christopher J, Kutyreff, Yongjun, Yan, Yan, Liu, Steve Y, Cho, Jonathan W, Engle, Jiye, Shi, Peng, Huang, Chunhai, Fan, Hao, Yan, and Weibo, Cai
- Abstract
Patients with acute kidney injury (AKI) frequently require kidney transplantation and supportive therapies, such as rehydration and dialysis. Here, we show that radiolabelled DNA origami nanostructures (DONs) with rectangular, triangular and tubular shapes accumulate preferentially in the kidneys of healthy mice and mice with rhabdomyolysis-induced AKI, and that rectangular DONs have renal-protective properties, with efficacy similar to the antioxidant
- Published
- 2018
18. Lymphoma: current status of clinical and preclinical imaging with radiolabeled antibodies
- Author
-
Weibo Cai, Christopher G. England, and Lixin Rui
- Subjects
Pathology ,medicine.medical_specialty ,Lymphoma ,CD30 ,medicine.medical_treatment ,Article ,030218 nuclear medicine & medical imaging ,03 medical and health sciences ,0302 clinical medicine ,immune system diseases ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,CD20 ,Clinical Trials as Topic ,medicine.diagnostic_test ,biology ,business.industry ,Antibodies, Monoclonal ,General Medicine ,Radioimmunotherapy ,medicine.disease ,Positron emission tomography ,Positron-Emission Tomography ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Radiopharmaceuticals ,Molecular imaging ,Antibody ,business ,Preclinical imaging - Abstract
Lymphoma is a complex disease that arises from cells of the immune system with an intricate pathology. While lymphoma may be classified as Hodgkin or non-Hodgkin, each type of tumor is genetically and phenotypically different and highly invasive tissue biopsies are the only method to investigate these differences. Noninvasive imaging strategies, such as immunoPET, can provide a vital insight into disease staging, monitoring treatment response in patients, and dose planning in radioimmunotherapy. ImmunoPET imaging with radiolabeled antibody-based tracers may also assist physicians in optimizing treatment strategies and enhancing patient stratification. Currently, there are two common biomarkers for molecular imaging of lymphoma, CD20 and CD30, both of which have been considered for investigation in preclinical imaging studies. In this review, we examine the current status of both preclinical and clinical imaging of lymphoma using radiolabeled antibodies. Additionally, we briefly investigate the role of radiolabeled antibodies in lymphoma therapy. As radiolabeled antibodies play critical roles in both imaging and therapy of lymphoma, the development of novel antibodies and the discovery of new biomarkers may greatly affect lymphoma imaging and therapy in the future.
- Published
- 2016
19. DNA nanomaterials for preclinical imaging and drug delivery
- Author
-
Dawei Jiang, Weibo Cai, and Christopher G. England
- Subjects
Base pair ,Computer science ,Pharmaceutical Science ,Nanotechnology ,02 engineering and technology ,010402 general chemistry ,01 natural sciences ,Article ,chemistry.chemical_compound ,Drug Delivery Systems ,DNA nanotechnology ,Animals ,Humans ,Biomedicine ,Bioconjugation ,business.industry ,DNA ,021001 nanoscience & nanotechnology ,Molecular Imaging ,Nanostructures ,0104 chemical sciences ,chemistry ,Drug delivery ,Molecular imaging ,0210 nano-technology ,business ,Preclinical imaging - Abstract
Besides being the carrier of genetic information, DNA is also an excellent biological organizer to establish well-designed nanostructures in the fields of material engineering, nanotechnology, and biomedicine. DNA-based materials represent a diverse nanoscale system primarily due to their predictable base pairing and highly regulated conformations, which greatly facilitate the construction of DNA nanostructures with distinct shapes and sizes. Integrating the emerging advancements in bioconjugation techniques, DNA nanostructures can be readily functionalized with high precision for many purposes ranging from biosensors to imaging to drug delivery. Recent progress in the field of DNA nanotechnology has exhibited collective efforts to employ DNA nanostructures as smart imaging agents or delivery platforms within living organisms. Despite significant improvements in the development of DNA nanostructures, there is limited knowledge regarding the in vivo biological fate of these intriguing nanomaterials. In this review, we summarize the current strategies for designing and purifying highly-versatile DNA nanostructures for biological applications, including molecular imaging and drug delivery. Since DNA nanostructures may elicit an immune response in vivo, we also present a short discussion of their potential toxicities in biomedical applications. Lastly, we discuss future perspectives and potential challenges that may limit the effective preclinical and clinical employment of DNA nanostructures. Due to their unique properties, we predict that DNA nanomaterials will make excellent agents for effective diagnostic imaging and drug delivery, improving patient outcome in cancer and other related diseases in the near future.
- Published
- 2016
20. Biocompatibility and in vivo operation of implantable mesoporous PVDF-based nanogenerators
- Author
-
Feng Chen, Haiyan Sun, Xudong Wang, Weibo Cai, Christopher G. England, Yanhao Yu, and Hakan Orbay
- Subjects
Bioelectronics ,Materials science ,Biocompatibility ,Polydimethylsiloxane ,Renewable Energy, Sustainability and the Environment ,Nanogenerator ,Nanotechnology ,02 engineering and technology ,010402 general chemistry ,021001 nanoscience & nanotechnology ,01 natural sciences ,Polyvinylidene fluoride ,Piezoelectricity ,Article ,0104 chemical sciences ,chemistry.chemical_compound ,chemistry ,General Materials Science ,Electrical and Electronic Engineering ,0210 nano-technology ,Mesoporous material ,Energy harvesting - Abstract
The rapid developments of implantable biomedical electronics give rise to the motivation of exploring efficient and durable self-powered charging system. In this paper, we report a mesoporous polyvinylidene fluoride (PVDF)-based implantable piezoelectric nanogenerator (NG) for in vivo biomechanical energy harvesting. The NG was built with a sponge-like mesoporous PVDF film and encapsulated by polydimethylsiloxane (PDMS). After embedding this NG into rodents, a Voc of ~200 mV was produced from the gentle movement of rodent muscle. Meanwhile, no toxicity or incompatibility sign was found in the host after carrying the packaged NG for 6 weeks. Moreover, the electric output of this NG was extremely stable and exhibited no deterioration after 5 days of in vivo operation or 1.512 × 108 times mechanical deformation. This NG device could practically output a constant voltage of 52 mV via a 1 µF capacitor under living circumstance. The outstanding efficiency, magnificent durability and exceptional biocompatibility promise this mesoporous PVDF-based NG in accomplishing self-powered bioelectronics with potentially lifespan operation period.
- Published
- 2016
21. Re-assessing the enhanced permeability and retention effect in peripheral arterial disease using radiolabeled long circulating nanoparticles
- Author
-
Steve Y. Cho, Zhuang Liu, Feng Chen, Liangzhu Feng, Hyung Jun Im, Weibo Cai, Stephen A. Graves, Dong Soo Lee, Christopher G. England, Cheng Xu, Reinier Hernandez, Hakan Orbay, and Robert J. Nickles
- Subjects
Materials science ,Laser Doppler Imaging ,Biophysics ,Ischemia ,Bioengineering ,02 engineering and technology ,Hindlimb ,Enhanced permeability and retention effect ,030204 cardiovascular system & hematology ,Ferric Compounds ,Permeability ,Article ,Capillary Permeability ,Photoacoustic Techniques ,Biomaterials ,Neovascularization ,Peripheral Arterial Disease ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Animals ,Mice, Inbred BALB C ,medicine.diagnostic_test ,021001 nanoscience & nanotechnology ,medicine.disease ,Copper Radioisotopes ,chemistry ,Mechanics of Materials ,Positron emission tomography ,Positron-Emission Tomography ,Ceramics and Composites ,Nanoparticles ,Female ,Graphite ,medicine.symptom ,0210 nano-technology ,Perfusion ,Iron oxide nanoparticles ,Biomedical engineering - Abstract
As peripheral arterial disease (PAD) results in muscle ischemia and neovascularization, it has been claimed that nanoparticles can passively accumulate in ischemic tissues through the enhanced permeability and retention (EPR) effect. At this time, a quantitative evaluation of the passive targeting capabilities of nanoparticles has not been reported in PAD. Using a murine model of hindlimb ischemia, we quantitatively assessed the passive targeting capabilities of 64Cu-labeled PEGylated reduced graphene oxide – iron oxide nanoparticles (64Cu-RGO-IONP-PEG) through the EPR effect using positron emission tomography (PET) imaging. Serial laser Doppler imaging was performed to monitor changes in blood perfusion upon surgical induction of ischemia. Nanoparticle accumulation was assessed at 3, 10, and 17 days post-surgery and found to be highest at 3 days post-surgery, with the ischemic hindlimb displaying an accumulation of 14.7 ± 0.5% injected dose per gram (%ID/g). Accumulation of 64Cu-RGO-IONP-PEG was lowest at 17 days post-surgery, with the ischemic hindlimb displaying only 5.1 ± 0.5%ID/g. Furthermore, nanoparticle accumulation was confirmed by photoacoustic imaging (PA). The combination of PET and serial Doppler imaging showed that nanoparticle accumulation in the ischemic hindlimb negatively correlated with blood perfusion. Thus, we quantitatively confirmed that 64Cu-RGO-IONP-PEG passively accumulated in ischemic tissue via the EPR effect, which is reduced as the perfusion normalizes. As 64Cu-RGO-IONP-PEG displayed substantial accumulation in the ischemic tissue, this nanoparticle platform may function as a future theranostic agent, providing both imaging and therapeutic applications.
- Published
- 2016
22. Dual Targeting of Tissue Factor and CD105 for Preclinical PET Imaging of Pancreatic Cancer
- Author
-
Bai Liu, Christopher G. England, Charles P. Theuer, Sixiang Shi, Haiming Luo, Reinier Hernandez, Robert J. Nickles, Hing C. Wong, Weibo Cai, and Stephen A. Graves
- Subjects
0301 basic medicine ,Cancer Research ,Pathology ,medicine.medical_specialty ,Article ,Thromboplastin ,Flow cytometry ,Immunoglobulin Fab Fragments ,Mice ,03 medical and health sciences ,Tissue factor ,0302 clinical medicine ,Pancreatic tumor ,Cell Line, Tumor ,Pancreatic cancer ,medicine ,Animals ,Humans ,Tissue Distribution ,medicine.diagnostic_test ,business.industry ,Endoglin ,Flow Cytometry ,medicine.disease ,Pancreatic Neoplasms ,Disease Models, Animal ,030104 developmental biology ,Oncology ,Positron emission tomography ,Positron-Emission Tomography ,030220 oncology & carcinogenesis ,Cancer research ,Adenocarcinoma ,Female ,Neprilysin ,Protein Multimerization ,Radiopharmaceuticals ,Bioorthogonal chemistry ,business ,Biomarkers - Abstract
Purpose: Pancreatic adenocarcinoma is a highly aggressive cancer, currently treated with limited success and dismal outcomes. New diagnostic and treatment strategies offer the potential to reduce cancer mortality. Developing highly specific noninvasive imaging probes for pancreatic cancer is essential to improving diagnostic accuracy and monitoring therapeutic intervention. Experimental Design: A bispecific heterodimer was synthesized by conjugating an anti-tissue factor (TF) Fab with an anti-CD105 Fab, via the bio-orthogonal “click” reaction between tetrazine (Tz) and trans-cyclooctene (TCO). The heterodimer was labeled with 64Cu for PET imaging of nude mice bearing BXPC-3 xenograft and orthotopic pancreatic tumors. Results: PET imaging of BXPC-3 (TF/CD105+/+) xenograft tumors with 64Cu-labeled heterodimer displayed significantly enhanced tumor uptake (28.8 ± 3.2 %ID/g; n = 4; SD) at 30 hours postinjection, as compared with each of their monospecific Fab tracers (12.5 ± 1.4 and 7.1 ± 2.6 %ID/g; n = 3; SD). In addition, the activity–concentration ratio allowed for effective tumor visualization (tumor/muscle ratio 75.2 ± 9.4 at 30 hours postinjection.; n = 4; SD). Furthermore, 64Cu-NOTA-heterodimer enabled sensitive detection of orthotopic pancreatic tumor lesions with an uptake of 17.1 ± 4.9 %ID/g at 30 hours postinjection and tumor/muscle ratio of 72.3 ± 46.7. Conclusions: This study demonstrates that dual targeting of TF and CD105 provided synergistic improvements in binding affinity and tumor localization of the heterodimer. Dual-targeted imaging agents of pancreatic and other cancers may assist in diagnosing pancreatic malignancies as well as reliable monitoring of therapeutic response. Clin Cancer Res; 22(15); 3821–30. ©2016 AACR.
- Published
- 2016
23. Accelerated Blood Clearance Phenomenon Reduces the Passive Targeting of PEGylated Nanoparticles in Peripheral Arterial Disease
- Author
-
Christopher G. England, Dong Soo Lee, Stephen A. Graves, Liangzhu Feng, Weibo Cai, Reinier Hernandez, Robert J. Nickles, Hyung Jun Im, Zhuang Liu, and Steve Y. Cho
- Subjects
0301 basic medicine ,Materials science ,Arterial disease ,Nanoparticle ,Nanotechnology ,02 engineering and technology ,Polyethylene glycol ,Ferric Compounds ,Article ,Polyethylene Glycols ,Mice ,Peripheral Arterial Disease ,03 medical and health sciences ,chemistry.chemical_compound ,In vivo ,PEG ratio ,Animals ,General Materials Science ,Mice, Inbred BALB C ,Oxides ,021001 nanoscience & nanotechnology ,Hindlimb ,Peripheral ,030104 developmental biology ,Copper Radioisotopes ,chemistry ,Positron-Emission Tomography ,Drug delivery ,Biophysics ,Nanoparticles ,Female ,Graphite ,Blood clearance ,0210 nano-technology - Abstract
Peripheral arterial disease (PAD) is a leading global health concern. Due to limited imaging and therapeutic options, PAD and other ischemia-related diseases may benefit from the use of long circulating nanoparticles as imaging probes and/or drug delivery vehicles. Polyethylene glycol (PEG)-conjugated nanoparticles have shown shortened circulation half-lives in vivo when injected multiple times into a single subject. This phenomenon has become known as the accelerated blood clearance (ABC) effect. The phenomenon is of concern for clinical translation of nanomaterials as it limits the passive accumulation of nanoparticles in many diseases, yet it has not been evaluated using inorganic or organic-inorganic hybrid nanoparticles. Herein, we found that the ABC phenomenon was induced by re-injection of PEGylated long circulating organic-inorganic hybrid nanoparticles, which significantly reduced the passive targeting of 64Cu-labeled PEGylated reduced graphene oxide – iron oxide nanoparticles (64Cu-RGO-IONP-PEG) in a murine model of PAD. Positron emission tomography (PET) imaging was performed at 3, 10, and 17 days post-surgical induction of hindlimb ischemia. At Day 3 post-surgery, the nanoparticles displayed a long circulation half-life with enhanced accumulation in the ischemic hindlimb. At Day 10 and 17 post-surgery, re-injected mice displayed a short circulation half-life and lower accumulation of the nanoparticles in the ischemic hindlimb, in comparison to the naïve group. Also, re-injected mice showed significantly higher liver uptake than the naïve group, indicating that the nanoparticles experienced higher sequestration by the liver in the re-injected group. Furthermore, photoacoustic (PA) imaging and Prussian blue staining confirmed the enhanced accumulation of the nanoparticles in the liver tissue of re-injected mice. These findings validate the ABC phenomenon using long circulating organic-inorganic hybrid nanoparticles upon multiple administrations to the same animal, which may provide valuable insight into the future clinical applications of nanoparticles for imaging and treatment of PAD.
- Published
- 2016
24. ImmunoPET Imaging of CD146 Expression in Malignant Brain Tumors
- Author
-
Weibo Cai, Yunan Yang, Haiyan Sun, Christopher G. England, Todd E. Barnhart, Reinier Hernandez, and Hector F. Valdovinos
- Subjects
0301 basic medicine ,Biodistribution ,medicine.drug_class ,Blotting, Western ,Brain tumor ,Pharmaceutical Science ,CD146 Antigen ,Biology ,Monoclonal antibody ,Article ,Flow cytometry ,03 medical and health sciences ,0302 clinical medicine ,Western blot ,In vivo ,Cell Line, Tumor ,Drug Discovery ,medicine ,Humans ,Microscopy, Confocal ,Radiochemistry ,medicine.diagnostic_test ,Brain Neoplasms ,Antibodies, Monoclonal ,Flow Cytometry ,medicine.disease ,Molecular biology ,In vitro ,030104 developmental biology ,Microscopy, Fluorescence ,Positron-Emission Tomography ,030220 oncology & carcinogenesis ,Molecular Medicine ,Ex vivo - Abstract
Recently, the overexpression of CD146 and its potential as a therapeutic target in high-grade gliomas, the most lethal type of brain cancer, was uncovered. In this study, we describe the generation of (89)Zr-Df-YY146, a novel (89)Zr-labeled monoclonal antibody (mAb) for the targeting and quantification of CD146 expression in a mouse model of glioblastoma, using noninvasive immunoPET imaging. YY146, a high affinity anti-CD146 mAb, was conjugated to deferoxamine (Df) for labeling with the long-lived positron emitter (89)Zr (t1/2: 78.4 h). In vitro assays, including flow cytometry, immunofluorescence microscopy, and Western blot, were performed with two glioblastoma cell lines, U87MG and U251, to determine their CD146 expression levels. Also, YY146 and Df-YY146's CD146-binding affinities were compared using flow cytometry. In vivo CD146-targeting of (89)Zr-Df-YY146 was evaluated by sequential PET imaging, in athymic nude mice bearing subcutaneously implanted U87MG or U251 tumors. CD146 blocking, ex vivo biodistribution, and histological studies were carried out to confirm (89)Zr-Df-YY146 specificity, as well as the accuracy of PET data. In vitro studies exposed elevated CD146 expression levels in U87MG cells, but negligible levels in U251 cells. Flow cytometry revealed no differences in affinity between YY146 and Df-YY146. (89)Zr labeling of Df-YY146 proceeded with excellent yield (∼80%), radiochemical purity (>95%), and specific activity (∼44 GBq/μmol). Longitudinal PET revealed prominent and persistent (89)Zr-Df-YY146 uptake in mice bearing U87MG tumors that peaked at 14.00 ± 3.28%ID/g (n = 4), 48 h post injection of the tracer. Conversely, uptake was significantly lower in CD146-negative U251 tumors (5.15 ± 0.99%ID/g, at 48 h p.i.; n = 4; P < 0.05). Uptake in U87MG tumors was effectively blocked in a competitive inhibition experiment, corroborating the CD146 specificity of (89)Zr-Df-YY146. Finally, ex vivo biodistribution validated the accuracy of PET data and histological examination successfully correlated tracer uptake with in situ CD146 expression. Prominent, persistent, and specific uptake of (89)Zr-Df-YY146 was observed in brain tumors, demonstrating the potential of this radiotracer for noninvasive PET imaging of CD146 expression. In a future clinical scenario, (89)Zr-Df-YY146 may serve as a tool to guide intervention and assess response to CD146-targeted therapies.
- Published
- 2016
25. Quantum dot–NanoLuc bioluminescence resonance energy transfer enables tumor imaging and lymph node mapping in vivo
- Author
-
Christopher G. England, Anyanee Kamkaew, Weibo Cai, Zhuang Liu, Liang Cheng, and Haiyan Sun
- Subjects
Bioluminescence Resonance Energy Transfer Techniques ,0301 basic medicine ,Luminescence ,Energy transfer ,Contrast Media ,Conjugated system ,Peptides, Cyclic ,Article ,Catalysis ,Mice ,03 medical and health sciences ,Nuclear magnetic resonance ,In vivo ,Cell Line, Tumor ,Decapoda ,Quantum Dots ,Materials Chemistry ,Animals ,Humans ,Bioluminescence ,Luciferase ,Luciferases ,Fluorescent Dyes ,Microscopy, Confocal ,Brain Neoplasms ,Chemistry ,Metals and Alloys ,Resonance ,General Chemistry ,Surfaces, Coatings and Films ,Electronic, Optical and Magnetic Materials ,030104 developmental biology ,Quantum dot ,Ceramics and Composites ,Lymph Nodes ,Glioblastoma ,Conjugate - Abstract
A small luciferase protein (Nluc) was conjugated to QDs as a bioluminescence resonance energy transfer (BRET) pair. The conjugate showed 76% BRET efficiency and lymph node mapping was successfully performed. The cRGD peptide was conjugated to QD-Nluc for tumor targeting. The self-illuminating QD-Nluc showed excellent energy transfer in a living system and offered an optimal tumor-to-background ratio (>85).
- Published
- 2016
26. ImmunoPET imaging of CD38 in murine lymphoma models using (89)Zr-labeled daratumumab
- Author
-
Christopher G. England, Rongfu Wang, Lei Kang, Bo Yu, Weibo Cai, Todd E. Barnhart, Jonathan W. Engle, Dawei Jiang, Zachary T. Rosenkrans, Peng Huang, and Xiaojie Xu
- Subjects
Biodistribution ,Lymphoma ,Mice, Nude ,Article ,Flow cytometry ,03 medical and health sciences ,Mice ,0302 clinical medicine ,In vivo ,hemic and lymphatic diseases ,Cell Line, Tumor ,medicine ,Animals ,Radiology, Nuclear Medicine and imaging ,Tissue Distribution ,B-cell lymphoma ,Radioisotopes ,medicine.diagnostic_test ,Chemistry ,Daratumumab ,Antibodies, Monoclonal ,General Medicine ,medicine.disease ,ADP-ribosyl Cyclase 1 ,Imaging agent ,030220 oncology & carcinogenesis ,Positron-Emission Tomography ,Cancer research ,Zirconium ,Ex vivo ,030215 immunology - Abstract
PURPOSE: CD38 is considered a potential biomarker for multiple myeloma (MM) and has shown a strong link with chronic lymphocytic leukemia due to high and uniform expression on plasma cells. In vivo evaluation of CD38 expression may provide useful information about lesion detection and prognosis of treatment in MM. In this study, immunoPET imaging with (89)Zr-labeled daratumumab was used for differentiation of CD38 expression in murine lymphoma models to provide a potential noninvasive method for monitoring CD38 in the clinic. METHODS: Daratumumab was radiolabeled with (89)Zr (t(1/2) = 78.4 h) via conjugation with desferrioxamine (Df). After Western blot (WB) was used to screen CD38 expression in five lymphoma cell lines, flow cytometry and cellular binding assays were performed to test the binding ability of labeled or conjugated daratumumab with CD38 in vitro. PET imaging and biodistribution studies were performed to evaluate CD38 expression after injection of (89)Zr-Df-daratumumab. (89)Zr-Df-IgG was also evaluated as a non-specific control group in the Ramos model. Finally, CD38 expression in tumor tissues was verified by histological analysis. RESULTS: Using WB screening, the Ramos cell line was found to express the highest level of CD38 while the HBL-1 cell line had the lowest expression. Df-conjugated and (89)Zr-labeled daratumumab displayed similar high binding affinities with Ramos cells. PET imaging of (89)Zr-Df-daratumumab showed a high tumor uptake of up to 26.6 ± 8.0 %ID/g for Ramos at 120 h post-injection, and only up to 6.6 ± 2.9 %ID/g for HBL-1 (n = 4). Additionally, (89)Zr-Df-IgG demonstrated a low tumor uptake in the Ramos model (only 4.3 ± 0.8 %ID/g at 120 h post-injection). Ex vivo biodistribution studies showed similar trends with imaging results. Immunofluorescent staining of tumor tissues verified higher CD38 expression of Ramos than that of HBL-1. CONCLUSIONS: The role of (89)Zr-Df-daratumumab was investigated for evaluating CD38 expression in lymphoma models noninvasively and was found to be to a promising imaging agent of CD38 positive hematological diseases such as MM in future clinical applications.
- Published
- 2018
27. Molecular Imaging of Pancreatic Cancer with Antibodies
- Author
-
Savo Bou Zein Eddine, Christopher G. England, Reinier Hernandez, and Weibo Cai
- Subjects
0301 basic medicine ,medicine.medical_specialty ,magnetic resonance imaging (MRI) ,pancreatic cancer ,Pharmaceutical Science ,Review ,photoacoustic tomography (PAT) ,Antibodies ,03 medical and health sciences ,optical imaging ,0302 clinical medicine ,Pancreatic cancer ,Drug Discovery ,medicine ,Brain positron emission tomography ,Humans ,positron emission tomography (PET) ,Tomography, Emission-Computed, Single-Photon ,medicine.diagnostic_test ,business.industry ,single-photon emission computed tomography (SPECT) ,Cancer ,Magnetic resonance spectroscopic imaging ,Magnetic resonance imaging ,medicine.disease ,molecular imaging ,Magnetic Resonance Imaging ,3. Good health ,Pancreatic Neoplasms ,030104 developmental biology ,Positron emission tomography ,030220 oncology & carcinogenesis ,Positron-Emission Tomography ,Cancer research ,Molecular Medicine ,Radiology ,Molecular imaging ,business ,Preclinical imaging - Abstract
Development of novel imaging probes for cancer diagnostics remains critical for early detection of disease, yet most imaging agents are hindered by suboptimal tumor accumulation. To overcome these limitations, researchers have adapted antibodies for imaging purposes. As cancerous malignancies express atypical patterns of cell surface proteins in comparison to noncancerous tissues, novel antibody-based imaging agents can be constructed to target individual cancer cells or surrounding vasculature. Using molecular imaging techniques, these agents may be utilized for detection of malignancies and monitoring of therapeutic response. Currently, there are several imaging modalities commonly employed for molecular imaging. These imaging modalities include positron emission tomography (PET), single-photon emission computed tomography (SPECT), magnetic resonance (MR) imaging, optical imaging (fluorescence and bioluminescence), and photoacoustic (PA) imaging. While antibody-based imaging agents may be employed for a broad range of diseases, this review focuses on the molecular imaging of pancreatic cancer, as there are limited resources for imaging and treatment of pancreatic malignancies. Additionally, pancreatic cancer remains the most lethal cancer with an overall 5-year survival rate of approximately 7%, despite significant advances in the imaging and treatment of many other cancers. In this review, we discuss recent advances in molecular imaging of pancreatic cancer using antibody-based imaging agents. This task is accomplished by summarizing the current progress in each type of molecular imaging modality described above. Also, several considerations for designing and synthesizing novel antibody-based imaging agents are discussed. Lastly, the future directions of antibody-based imaging agents are discussed, emphasizing the potential applications for personalized medicine.
- Published
- 2015
28. Evaluation of two novel 64Cu-labeled RGD peptide radiotracers for enhanced PET imaging of tumor integrin αvβ3
- Author
-
Rubel Chakravarty, Christopher G. England, Francisco Valenzuela, Weibo Cai, Reinier Hernandez, Yunan Yang, Andrzej Czerwinski, Stephen A. Graves, and Robert J. Nickles
- Subjects
Biodistribution ,biology ,Chemistry ,business.industry ,Angiogenesis ,Integrin ,General Medicine ,Pharmacokinetics ,PEG ratio ,biology.protein ,Biophysics ,Radiology, Nuclear Medicine and imaging ,Molecular imaging ,Nuclear medicine ,business ,Receptor ,Ex vivo - Abstract
Our goal was to demonstrate that suitably derivatized monomeric RGD peptide-based PET tracers, targeting integrin αvβ3, may offer advantages in image contrast, time for imaging, and low uptake in nontarget tissues. Two cyclic RGDfK derivatives, (PEG)2-c(RGDfK) and PEG4-SAA4-c(RGDfK), were constructed and conjugated to NOTA for 64Cu labeling. Their integrin αvβ3-binding properties were determined via a competitive cell binding assay. Mice bearing U87MG tumors were intravenously injected with each of the 64Cu-labeled peptides, and PET scans were acquired during the first 30 min, and 2 and 4 h after injection. Blocking and ex vivo biodistribution studies were carried out to validate the PET data and confirm the specificity of the tracers. The IC50 values of NOTA-(PEG)2-c(RGDfK) and NOTA-PEG4-SAA4-c(RGDfK) were 444 ± 41 nM and 288 ± 66 nM, respectively. Dynamic PET data of 64Cu-NOTA-(PEG)2-c(RGDfK) and 64Cu-NOTA-PEG4-SAA4-c(RGDfK) showed similar circulation t 1/2 and peak tumor uptake of about 4 %ID/g for both tracers. Due to its marked hydrophilicity, 64Cu-NOTA-PEG4-SAA4-c(RGDfK) provided faster clearance from tumor and normal tissues yet maintained excellent tumor-to-background ratios. Static PET scans at later time-points corroborated the enhanced excretion of the tracer, especially from abdominal organs. Ex vivo biodistribution and receptor blocking studies confirmed the accuracy of the PET data and the integrin αvβ3-specificity of the peptides. Our two novel RGD-based radiotracers with optimized pharmacokinetic properties allowed fast, high-contrast PET imaging of tumor-associated integrin αvβ3. These tracers may facilitate the imaging of abdominal malignancies, normally precluded by high background uptake.
- Published
- 2015
29. HaloTag Technology: A Versatile Platform for Biomedical Applications
- Author
-
Christopher G. England, Weibo Cai, and Haiming Luo
- Subjects
Proteomics ,Recombinant Fusion Proteins ,Biomedical Engineering ,Pharmaceutical Science ,Bioengineering ,Genomics ,Nanotechnology ,Review ,Protein Interaction Mapping ,Protein purification ,Animals ,Humans ,Fluorescent Dyes ,Pharmacology ,Protein function ,Staining and Labeling ,Chemistry ,Cellular imaging ,Optical Imaging ,Organic Chemistry ,Proteins ,DNA ,DNA metabolism ,Proteins metabolism ,Molecular imaging ,Biotechnology - Abstract
Exploration of protein function and interaction is critical for discovering links among genomics, proteomics, and disease state; yet, the immense complexity of proteomics found in biological systems currently limits our investigational capacity. Although affinity and autofluorescent tags are widely employed for protein analysis, these methods have been met with limited success because they lack specificity and require multiple fusion tags and genetic constructs. As an alternative approach, the innovative HaloTag protein fusion platform allows protein function and interaction to be comprehensively analyzed using a single genetic construct with multiple capabilities. This is accomplished using a simplified process, in which a variable HaloTag ligand binds rapidly to the HaloTag protein (usually linked to the protein of interest) with high affinity and specificity. In this review, we examine all current applications of the HaloTag technology platform for biomedical applications, such as the study of protein isolation and purification, protein function, protein-protein and protein-DNA interactions, biological assays, in vitro cellular imaging, and in vivo molecular imaging. In addition, novel uses of the HaloTag platform are briefly discussed along with potential future applications.
- Published
- 2015
30. ImmunoPET Imaging of CD146 in Murine Models of Intrapulmonary Metastasis of Non-Small Cell Lung Cancer
- Author
-
Christopher G. England, Dawei Jiang, Reinier Hernandez, Hector F. Valdovinos, Emily B. Ehlerding, Yunan Yang, Haiyan Sun, Peng Huang, Weibo Cai, and Jonathan W. Engle
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Lung Neoplasms ,medicine.drug_class ,Pharmaceutical Science ,Fluorescent Antibody Technique ,Mice, Nude ,CD146 Antigen ,Monoclonal antibody ,Article ,Flow cytometry ,Metastasis ,03 medical and health sciences ,Heterocyclic Compounds, 1-Ring ,Mice ,0302 clinical medicine ,Heterocyclic Compounds ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Positron Emission Tomography Computed Tomography ,Drug Discovery ,medicine ,Carcinoma ,Biomarkers, Tumor ,Animals ,Humans ,Tissue Distribution ,Radioactive Tracers ,Lung cancer ,Lung ,medicine.diagnostic_test ,business.industry ,Antibodies, Monoclonal ,medicine.disease ,Flow Cytometry ,Xenograft Model Antitumor Assays ,Molecular Imaging ,Platelet Endothelial Cell Adhesion Molecule-1 ,030104 developmental biology ,medicine.anatomical_structure ,Copper Radioisotopes ,030220 oncology & carcinogenesis ,Molecular Medicine ,Biomarker (medicine) ,CD146 ,Female ,business - Abstract
CD146 has been identified as an excellent biomarker for lung cancer as its overexpression in solid tumors has been linked to disease progression, invasion, and metastasis. Previously, our group described a positive correlation between 64Cu-labeled YY146 uptake and increased expression of CD146 in six human lung cancer cell lines using subcutaneous tumor models. In this study, we investigate a monoclonal antibody called YY146 for immunoPET imaging of CD146 in two intrapulmonary metastasis models of non-small cell lung cancer (NSCLC). The binding and immunoreactivity of the tracer were assessed by in vitro assays. Radiolabeling of YY146 with the positron emitting Cu-64 (64Cu-NOTA-YY146) enabled PET imaging of intrapulmonary metastasis. Mice were intravenously injected with two-million tumor cells, and CT imaging was used to verify the presence of lung metastases. 64Cu-NOTA-YY146 was injected into tumor-bearing mice and animals were subjected to PET/CT imaging at 4, 24, and 48 h post-injection. Both the average and maximum lung PET signal intensities were quantified and compared between high and low CD146-expressing metastases. Further validation was accomplished through immunofluorescence imaging of resected tissues with CD31 and CD146. In flow cytometry, YY146 revealed strong binding to CD146 in H460 cells due to its high expression, with minimal binding to CD146-low expressing H358 cells. Both YY146 and NOTA-YY146 showed similar binding, suggesting that NOTA conjugation did not elicit any negative effects on its binding affinity. Imaging of 64Cu-NOTA-YY146 in H460 tumor-bearing mice revealed rapid, persistent, and highly-specific tracer accumulation. Uptake of 64Cu-NOTA-YY146 in the whole lung was calculated for H460 and H358 as 7.43 ± 0.38 and 3.95 ± 0.47 %ID/g at 48 h post-injection (n=4, p
- Published
- 2017
31. Theranostic Nanoparticles for Photothermal Therapy of Cancer
- Author
-
Weibo Cai and Christopher G. England
- Subjects
Materials science ,Theranostic nanoparticles ,medicine ,Cancer ,Nanotechnology ,Photothermal therapy ,medicine.disease - Published
- 2017
32. ImmunoPET and Near-Infrared Fluorescence Imaging of Pancreatic Cancer with a Dual-Labeled Bispecific Antibody Fragment
- Author
-
Christopher G. England, Charles P. Theuer, Hing C. Wong, Robert J. Nickles, Stephen A. Graves, Weibo Cai, Haiming Luo, Fanrong Ai, Shreya Goel, and Bai Liu
- Subjects
0301 basic medicine ,Near-Infrared Fluorescence Imaging ,Biodistribution ,Pathology ,medicine.medical_specialty ,Pharmaceutical Science ,Mice, Nude ,Article ,03 medical and health sciences ,Immunoglobulin Fab Fragments ,Mice ,0302 clinical medicine ,In vivo ,Pancreatic cancer ,Cell Line, Tumor ,Drug Discovery ,Antibodies, Bispecific ,medicine ,Animals ,Humans ,medicine.diagnostic_test ,Chemistry ,medicine.disease ,Flow Cytometry ,Imaging agent ,Immunoconjugate ,Pancreatic Neoplasms ,030104 developmental biology ,Copper Radioisotopes ,Positron emission tomography ,030220 oncology & carcinogenesis ,Positron-Emission Tomography ,Cancer research ,Molecular Medicine ,Female ,Ex vivo - Abstract
Dual-targeted imaging agents have shown improved targeting efficiencies in comparison to single-targeted entities. The purpose of this study was to quantitatively assess the tumor accumulation of a dual-labeled heterobifunctional imaging agent, targeting two overexpressed biomarkers in pancreatic cancer, using positron emission tomography (PET) and near-infrared fluorescence (NIRF) imaging modalities. A bispecific immunoconjugate (heterodimer) of CD105 and tissue factor (TF) Fab′ antibody fragments was developed using click chemistry. The heterodimer was dual-labeled with a radionuclide (64Cu) and fluorescent dye. PET/NIRF imaging and biodistribution studies were performed in four-to-five week old nude athymic mice bearing BxPC-3 (CD105/TF+/+) or PANC-1 (CD105/TF−/−) tumors xenografts. A blocking study was conducted to investigate the specificity of the tracer. Ex vivo tissue staining was performed to compare TF/CD105 expression in tissues with PET tracer uptake to validate in vivo results. PET imaging of 64Cu-NOTA-heterodimer-ZW800 in BxPC-3 tumor xenografts revealed enhanced tumor uptake (21.0 ± 3.4 %ID/g; n = 4) compared to the homodimer of TRC-105 (9.6 ± 2.0 %ID/g; n=4; p < 0.01) and ALT-836 (7.6 ± 3.7 %ID/g; n=4; p < 0.01) at 24 h post-injection. Blocking studies revealed that tracer uptake in BxPC-3 tumors could be decreased by four-fold with TF blocking and two-fold with CD105 blocking. In the negative model (PANC-1), heterodimer uptake was significantly lower than that found in the BxPC-3 model (3.5 ± 1.1 %ID/g; n=4; p < 0.01). The specificity was confirmed by the successful blocking of CD105 or TF, which demonstrated the dual targeting with 64Cu-NOTA-heterodimer-ZW800 provided an improvement in overall tumor accumulation. Also, fluorescence imaging validated the PET imaging, allowing for clear delineation of the xenograft tumors. Dual-labeled heterodimeric imaging agents, like 64Cu-NOTA-heterodimer-ZW800, may increase the overall tumor accumulation in comparison to single-targeted homodimers, leading to improved imaging of cancer and other related diseases.
- Published
- 2017
33. Chelator-Free Radiolabeling of Nanographene: Breaking the Stereotype of Chelation
- Author
-
Haiming Luo, Christopher G. England, Robert J. Nickles, Kai Yang, Cheng Xu, Zhuang Liu, Weibo Cai, Feng Chen, Liang Cheng, Sixiang Shi, Emily B. Ehlerding, Shreya Goel, and Hector F. Valdovinos
- Subjects
Oxide ,Nanoparticle ,Nanotechnology ,Breast Neoplasms ,02 engineering and technology ,010402 general chemistry ,01 natural sciences ,Catalysis ,Article ,law.invention ,chemistry.chemical_compound ,Mice ,law ,Animals ,Chelation ,Particle Size ,Chelating Agents ,Chemistry ,Graphene ,Mammary Neoplasms, Experimental ,General Medicine ,General Chemistry ,Pet imaging ,021001 nanoscience & nanotechnology ,Combinatorial chemistry ,0104 chemical sciences ,Copper Radioisotopes ,Positron-Emission Tomography ,Nanoparticles ,Female ,Graphite ,0210 nano-technology ,Copper - Abstract
Macrocyclic chelators have been widely employed in the realm of nanoparticle-based positron emission tomography (PET) imaging, whereas its accuracy remains questionable. Here, we found that 64Cu can be intrinsically labeled onto nanographene based on interactions between Cu and the π electrons of graphene without the need of chelator conjugation, providing a promising alternative radiolabeling approach that maintains the native in vivo pharmacokinetics of the nanoparticles. Due to abundant π bonds, reduced graphene oxide (RGO) exhibited significantly higher labeling efficiency in comparison with graphene oxide (GO) and exhibited excellent radiostability in vivo. More importantly, nonspecific attachment of 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA) on nanographene was observed, which revealed that chelator-mediated nanoparticle-based PET imaging has its inherent drawbacks and can possibly lead to erroneous imaging results in vivo.
- Published
- 2017
34. Radiomanganese PET Detects Changes in Functional β-cell Mass in Mouse Models of Diabetes
- Author
-
Todd E. Barnhart, Halena R. VanDeusen, Christopher G. England, Gregory Severin, Hector F. Valdovinos, Reinier Hernandez, Matthew J. Merrins, Justin J. Jeffery, Rachel J. Fenske, Trillian Gregg, Haley Wienkes, Michelle E. Kimple, Weibo Cai, Stephen A. Graves, and Robert J. Nickles
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Streptozocin ,Diabetes Mellitus, Experimental ,030218 nuclear medicine & medical imaging ,Glibenclamide ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Tolbutamide ,Nifedipine ,SDG 3 - Good Health and Well-being ,In vivo ,Insulin-Secreting Cells ,Internal medicine ,Internal Medicine ,medicine ,Diazoxide ,Animals ,Humans ,Channel blocker ,Pancreas ,Cell Size ,medicine.diagnostic_test ,Chemistry ,Diabetes Mellitus, Type 1 ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Manganese Compounds ,Islet Studies ,Positron emission tomography ,Case-Control Studies ,Positron-Emission Tomography ,Disease Progression ,Calcium Channels ,Radiopharmaceuticals ,medicine.drug - Abstract
The noninvasive measurement of functional β-cell mass would be clinically valuable for monitoring the progression of type 1 and type 2 diabetes, as well as the viability of transplanted insulin-producing cells. Although previous work employing magnetic resonance imaging has shown promise for functional β-cell mass determination through voltage-dependent Ca2+ channel (VDCC)-mediated internalization of Mn2+, the clinical utility of this technique is limited by the cytotoxic levels of Mn2+ contrast agent. Here, we show that positron emission tomography (PET) is advantageous for determining functional β-cell mass using 52Mn2+ (t1/2: 5.6 d). We investigated the whole-body distribution of 52Mn2+ in healthy adult mice by dynamic and static PET imaging. Pancreatic VDCC uptake of 52Mn2+ was successfully manipulated pharmacologically in vitro and in vivo using glucose, nifedipine (VDCC blocker), the sulfonylureas tolbutamide and glibenclamide (KATP channel blockers), and diazoxide (KATP channel opener). In a mouse model of streptozotocin (STZ)-induced type 1 diabetes, 52Mn2+ uptake in the pancreas was distinguished from healthy controls in parallel with classic histological quantification of β-cell mass from pancreatic sections. 52Mn2+-PET also reported the expected increase in functional β-cell mass in the ob/ob model of pre-type 2 diabetes, a result corroborated by histological β-cell mass measurements and live-cell imaging of β-cell Ca2+ oscillations. These results indicate that 52Mn2+-PET is a sensitive new tool for the non-invasive assessment of functional β-cell mass.
- Published
- 2017
35. Targeting of BRAF resistant melanoma via extracellular matrix metalloproteinase inducer receptor
- Author
-
Christopher G. England, Matthew R. Zeiderman, Kelly M. McMasters, Lacey R. McNally, Tess V. Dupre, Charles W. Kimbrough, and Michael E. Egger
- Subjects
Proto-Oncogene Proteins B-raf ,Indoles ,Article ,Flow cytometry ,Mice ,Drug Delivery Systems ,Cell surface receptor ,Cell Line, Tumor ,medicine ,Animals ,Calgranulin B ,Humans ,Vemurafenib ,Receptor ,neoplasms ,Melanoma ,Sulfonamides ,Liposome ,medicine.diagnostic_test ,Chemistry ,medicine.disease ,Molecular biology ,Targeted drug delivery ,Drug Resistance, Neoplasm ,Liposomes ,Drug delivery ,Basigin ,Cancer research ,Female ,Surgery ,medicine.drug - Abstract
Background The BRAF inhibitor vemurafenib (PLX) has shown promise in treating metastatic melanoma, but most patients develop resistance to treatment after 6 mo. We identified a transmembrane protein, extracellular matrix metalloproteinase inducer (EMMPRIN) as a cell surface receptor highly expressed by PLX-resistant melanoma. Using an S100A9 ligand, we created an EMMPRIN targeted probe and liposome that binds to melanoma cells in vivo, thus designing a novel drug delivery vehicle. Methods PLX-resistant cells were established through continuous treatment with PLX-4032 over the course of 1 y. Both PLX-resistant and sensitive melanoma cell lines were evaluated for the expression of unique cell surface proteins, which identified EMMPRIN as an overexpressed protein in PLX0-resistant cells and S100A9 is a ligand for EMMPRIN. To design a probe for EMMPRIN, S100A9 ligand was conjugated to a CF-750 near-infrared (NIR) dye. EMMPRIN targeted liposomes were created to encapsulate CF-750 NIR dye. Liposomes were characterized by scanning electron microscopy, flow cytometry, and in vivo analysis. A2058PLX and A2058 cells were subcutaneously injected into athymic mice. S100A9 liposomes were intravenously injected and tumor accumulation was evaluated using NIR fluorescent imaging. Results Western blot and flow cytometry demonstrated that PLX sensitive and resistant A2058 and A375 melanoma cells highly express EMMPRIN. S100A9 liposomes were 200 nm diameter and uniformly sized. Flow cytometry demonstrated 100X more intracellular dye uptake by A2058 cells treated with S100A9 liposomes compared with untargeted liposomes. In vivo accumulation of S100A9 liposomes within subcutaneous A2058 and A2058PLX tumors was observed from 6–48 h, with A2058PLX accumulating significantly higher levels (P = 0.001626). Conclusions EMMPRIN-targeted liposomes via an S100A9 ligand are a novel, targeted delivery system which could provide improved EMMPRIN specific drug delivery to a tumor.
- Published
- 2014
36. Molecular Imaging of Immunotherapy Targets in Cancer
- Author
-
Christopher G. England, Weibo Cai, Douglas G. McNeel, and Emily B. Ehlerding
- Subjects
0301 basic medicine ,Noninvasive imaging ,medicine.medical_treatment ,Focus on Molecular Imaging ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,PD-L1 ,Neoplasms ,medicine ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,Molecular Targeted Therapy ,biology ,business.industry ,Cancer ,Immunotherapy ,medicine.disease ,Molecular Imaging ,030104 developmental biology ,CTLA-4 ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,biology.protein ,Molecular imaging ,business ,Preclinical imaging - Abstract
Immunotherapy has emerged as a promising alternative in the arsenal against cancer by harnessing the power of the immune system to specifically target malignant tissues. As the field of immunotherapy continues to expand, researchers will require newer methods for studying the interactions between the immune system, tumor cells, and immunotherapy agents. Recently, several noninvasive imaging strategies have been used to map the biodistribution of immune checkpoint molecules, monitor the efficacy and potential toxicities of the treatments, and identify patients who are likely to benefit from immunotherapies. In this review, we outline the current applications of noninvasive techniques for the preclinical imaging of immunotherapy targets and suggest future pathways for molecular imaging to contribute to this developing field.
- Published
- 2016
37. Dual-Modality Positron Emission Tomography/Optical Image-Guided Photodynamic Cancer Therapy with Chlorin e6-Containing Nanomicelles
- Author
-
Shreya Goel, Christopher G. England, Haiyan Sun, Todd E. Barnhart, Hua Gong, Dawei Jiang, Anyanee Kamkaew, Weibo Cai, Hector F. Valdovinos, and Liang Cheng
- Subjects
Fluorescence-lifetime imaging microscopy ,Materials science ,Porphyrins ,medicine.medical_treatment ,General Physics and Astronomy ,Nanotechnology ,Photodynamic therapy ,02 engineering and technology ,Polyethylene glycol ,Conjugated system ,010402 general chemistry ,01 natural sciences ,Article ,chemistry.chemical_compound ,Mice ,In vivo ,Cell Line, Tumor ,PEG ratio ,medicine ,Animals ,General Materials Science ,Micelles ,medicine.diagnostic_test ,Chlorophyllides ,General Engineering ,021001 nanoscience & nanotechnology ,0104 chemical sciences ,chemistry ,Photochemotherapy ,Positron emission tomography ,Positron-Emission Tomography ,Nanomedicine ,Nanoparticles ,0210 nano-technology ,Biomedical engineering - Abstract
Multifunctional nanoparticles with combined diagnostic and therapeutic functions show great promise in nanomedicine. Herein, we develop an organic photodynamic therapy (PDT) system based on polyethylene glycol (PEG)-coated nanomicelles conjugated with ∼20% chlorin e6 (PEG-Ce 6 nanomicelles), which functions as an optical imaging agent, as well as a PDT agent. The formed PEG-Ce 6 nanomicelles with the size of ∼20 nm were highly stable in various physiological solutions for a long time. Moreover, Ce 6 can also be a (64)Cu chelating agent for in vivo positron emission tomography (PET). By simply mixing, more than 90% of (64)Cu was chelator-free labeled on PEG-Ce 6 nanomicelles, and they also showed high stability in serum conditions. Both fluorescence imaging and PET imaging revealed that PEG-Ce 6 nanomicelles displayed high tumor uptake (13.7 ± 2.2%ID/g) after intravenous injection into tumor-bearing mice at the 48 h time point. In addition, PEG-Ce 6 nanomicelles exhibited excellent PDT properties upon laser irradiation, confirming the theranostic properties of PEG-Ce 6 nanomicelles for imaging and treatment of cancer. In addition, PDT was not shown to render any appreciable toxicity. This work presents a theranostic platform based on polymer nanomicelles with great potential in multimodality imaging-guided photodynamic cancer therapy.
- Published
- 2016
38. ImmunoPET for assessing the differential uptake of a CD146-specific monoclonal antibody in lung cancer
- Author
-
Todd E. Barnhart, Yunan Yang, Anyanee Kamkaew, Weibo Cai, Dawei Jiang, Reinier Hernandez, Christopher G. England, Haiyan Sun, Stephen A. Graves, and Rebecca L. Majewski
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Lung Neoplasms ,medicine.drug_class ,Mice, Nude ,CD146 Antigen ,Monoclonal antibody ,Sensitivity and Specificity ,Article ,Metastasis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Coordination Complexes ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,Lung cancer ,biology ,medicine.diagnostic_test ,business.industry ,Disease progression ,Antibodies, Monoclonal ,Reproducibility of Results ,General Medicine ,medicine.disease ,Molecular Imaging ,030104 developmental biology ,Positron emission tomography ,030220 oncology & carcinogenesis ,Positron-Emission Tomography ,biology.protein ,Cancer research ,CD146 ,Female ,Antibody ,Molecular imaging ,Radiopharmaceuticals ,business ,Peptides - Abstract
Overexpression of CD146 in solid tumors has been linked to disease progression, invasion, and metastasis. We describe the generation of aThe anti-CD146 antibody (YY146) was conjugated to 1,4,7-triazacyclononane-triacetic acid (NOTA) and radiolabeled withFlow cytometry and western blot studies showed similar findings with H460 and H23 cells showing high levels of expression of CD146. Small differences in CD146 expression levels were found among A549, H4006, H522, and H358 cells. Tumor uptake ofThe strong correlation between tumor uptake of
- Published
- 2016
39. Positron emission tomography and nanotechnology: A dynamic duo for cancer theranostics
- Author
-
Feng Chen, Christopher G. England, Weibo Cai, and Shreya Goel
- Subjects
Computer science ,Pharmaceutical Science ,Nanotechnology ,02 engineering and technology ,010402 general chemistry ,01 natural sciences ,Theranostic Nanomedicine ,Article ,Neoplasms ,medicine ,Animals ,Humans ,medicine.diagnostic_test ,Early disease ,Cancer ,021001 nanoscience & nanotechnology ,medicine.disease ,Bench to bedside ,0104 chemical sciences ,Positron emission tomography ,Positron-Emission Tomography ,Nanomedicine ,Nanoparticles ,Molecular imaging ,0210 nano-technology ,In vivo pharmacokinetics - Abstract
Development of novel imaging probes for cancer diagnosis is critical for early disease detection and management. The past two decades have witnessed a surge in the development and evolution of radiolabeled nanoparticles as a new frontier in personalized cancer nanomedicine. The dynamic synergism of positron emission tomography (PET) and nanotechnology combines the sensitivity and quantitative nature of PET with the multifunctionality and tunability of nanomaterials, which can help overcome certain key challenges in the field. In this review, we discuss the recent advances in radionanomedicine, exemplifying the ability to tailor the physicochemical properties of nanomaterials to achieve optimal in vivo pharmacokinetics and targeted molecular imaging in living subjects. Innovations in development of facile and robust radiolabeling strategies and biomedical applications of such radionanoprobes in cancer theranostics are highlighted. Imminent issues in clinical translation of radiolabeled nanomaterials are also discussed, with emphasis on multidisciplinary efforts needed to quickly move these promising agents from bench to bedside.
- Published
- 2016
40. Preclinical Pharmacokinetics and Biodistribution Studies of 89Zr-Labeled Pembrolizumab
- Author
-
Haiyan Sun, Emily B. Ehlerding, Glenn Liu, Douglas G. McNeel, Christopher G. England, Brian T. Rekoske, Stephen A. Graves, Reinier Hernandez, Todd E. Barnhart, and Weibo Cai
- Subjects
Male ,Biodistribution ,medicine.drug_class ,Metabolic Clearance Rate ,Receptor expression ,T-Lymphocytes ,Antineoplastic Agents ,Pembrolizumab ,Pharmacology ,Monoclonal antibody ,Antibodies, Monoclonal, Humanized ,Sensitivity and Specificity ,Whole-Body Counting ,030218 nuclear medicine & medical imaging ,Rats, Sprague-Dawley ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Pharmacokinetics ,In vivo ,medicine ,Animals ,Radiology, Nuclear Medicine and imaging ,Tissue Distribution ,Novel Imaging Probes ,Radioisotopes ,Mice, Inbred ICR ,business.industry ,Reproducibility of Results ,Rats ,Cell Tracking ,Organ Specificity ,030220 oncology & carcinogenesis ,Isotope Labeling ,Positron-Emission Tomography ,Humanized mouse ,Zirconium ,Radiopharmaceuticals ,business - Abstract
Pembrolizumab is a humanized monoclonal antibody targeting programmed cell death protein 1 (PD-1) found on T and pro-B cells. Pembrolizumab prevents PD-1 ligation by both PD-L1 and PD-L2, preventing the immune dysregulation that otherwise occurs when T-cells encounter cells expressing these ligands. Clinically, PD-1 blockade elicits potent antitumor immune responses, and antibodies blocking PD-1 ligation, including pembrolizumab, have recently received Food and Drug Administration approval for the treatment of advanced melanoma, renal cell cancer, and non-small cell lung cancer. Methods In this study, we evaluated the pharmacokinetics, biodistribution, and dosimetry of pembrolizumab in vivo, accomplished through radiolabeling with the positron emitter 89Zr. PET imaging was used to evaluate the whole-body distribution of 89Zr-deferoxamine (Df)-pembrolizumab in two rodent models (mice and rats). Data obtained from PET scans and biodistribution studies were extrapolated to humans to estimate the dosimetry of the tracer. As a proof of concept, the biodistribution of 89Zr-Df-pembrolizumab was further investigated in a humanized murine model. Results The tracer remained stable in blood circulation throughout the study and accumulated the greatest in liver and spleen tissues. Both mice and rats showed similar biodistribution and pharmacokinetics of 89Zr-Df-pembrolizumab. In the humanized mouse model, T-cell infiltration into the salivary and lacrimal glands could be successfully visualized. Conclusion These data will augment our understanding of the pharmacokinetics and biodistribution of radiolabeled pembrolizumab in vivo, while providing detailed dosimetry data that may lead to better dosing strategies in the future. These findings further demonstrate the utility of noninvasive in vivo PET imaging to dynamically track T-cell checkpoint receptor expression and localization in a humanized mouse model.
- Published
- 2016
41. Long circulating reduced graphene oxide-iron oxide nanoparticles for efficient tumor targeting and multimodality imaging
- Author
-
Liangzhu Feng, Emily B. Ehlerding, Christopher G. England, Shreya Goel, Taihong Wang, Haiyan Sun, Zhuang Liu, Weibo Cai, Feng Chen, Robert J. Nickles, Sixiang Shi, Cheng Xu, and Stephen A. Graves
- Subjects
Materials science ,Oxide ,Nanoparticle ,Nanotechnology ,Biocompatible Materials ,02 engineering and technology ,Polyethylene glycol ,010402 general chemistry ,01 natural sciences ,Ferric Compounds ,Multimodal Imaging ,Article ,law.invention ,Polyethylene Glycols ,Photoacoustic Techniques ,chemistry.chemical_compound ,Mice ,law ,Cell Line, Tumor ,Positron Emission Tomography Computed Tomography ,Animals ,General Materials Science ,Mice, Inbred BALB C ,Graphene ,Oxides ,Neoplasms, Experimental ,021001 nanoscience & nanotechnology ,Magnetic Resonance Imaging ,0104 chemical sciences ,chemistry ,Surface modification ,Nanoparticles ,Female ,Graphite ,0210 nano-technology ,Nanoconjugates ,Iron oxide nanoparticles ,Superparamagnetism ,Biomedical engineering - Abstract
Polyethylene glycol (PEG) surface modification is one of the most widely used approaches to improve the solubility of inorganic nanoparticles, prevent their aggregation and prolong their in vivo blood circulation half-life. Herein, we developed double-PEGylated biocompatible reduced graphene oxide nanosheets anchored with iron oxide nanoparticles (RGO-IONP-(1st)PEG-(2nd)PEG). The nanoconjugates exhibited a prolonged blood circulation half-life (∼27.7 h) and remarkable tumor accumulation (11 %ID g(-1)) via an enhanced permeability and retention (EPR) effect. Due to the strong near-infrared absorbance and superparamagnetism of RGO-IONP-(1st)PEG-(2nd)PEG, multimodality imaging combining positron emission tomography (PET) imaging with magnetic resonance imaging (MRI) and photoacoustic (PA) imaging was successfully achieved. The promising results suggest the great potential of these nanoconjugates for multi-dimensional and more accurate tumor diagnosis and therapy in the future.
- Published
- 2016
42. NanoLuc: A Small Luciferase Is Brightening Up the Field of Bioluminescence
- Author
-
Weibo Cai, Emily B. Ehlerding, and Christopher G. England
- Subjects
0301 basic medicine ,Biomedical Research ,Luminescence ,Disease detection ,Biomedical Engineering ,Pharmaceutical Science ,Bioengineering ,Nanotechnology ,02 engineering and technology ,Article ,03 medical and health sciences ,Renilla luciferase ,Bioluminescence imaging ,Bioluminescence ,Animals ,Humans ,Luciferase ,Luciferases ,Pharmacology ,Firefly protocol ,Chemistry ,Organic Chemistry ,021001 nanoscience & nanotechnology ,Therapeutic monitoring ,Nanostructures ,030104 developmental biology ,Molecular imaging ,0210 nano-technology ,Biotechnology - Abstract
The biomedical field has greatly benefited from the discovery of bioluminescent proteins. Currently, scientists employ bioluminescent systems for numerous biomedical applications, ranging from highly sensitive cellular assays to bioluminescence-based molecular imaging. Traditionally, these systems are based on Firefly and Renilla luciferases; however, the applicability of these enzymes is limited by their size, stability, and luminescence efficiency. NanoLuc (NLuc), a novel bioluminescence platform, offers several advantages over established systems, including enhanced stability, smaller size, and >150-fold increase in luminescence. In addition, the substrate for NLuc displays enhanced stability and lower background activity, opening up new possibilities in the field of bioluminescence imaging. The NLuc system is incredibly versatile and may be utilized for a wide array of applications. The increased sensitivity, high stability, and small size of the NLuc system have the potential to drastically change the field of reporter assays in the future. However, as with all such technology, NLuc has limitations (including a non-ideal emission for in vivo applications and its unique substrate) which may cause it to find restricted use in certain areas of molecular biology. As this unique technology continues to broaden, NLuc may have a significant impact in both preclinical and clinical fields, with potential roles in disease detection, molecular imaging, and therapeutic monitoring. This review will present the NLuc technology to the scientific community in a non-biased manner, allowing the audience to adopt their own views of this novel system.
- Published
- 2016
43. CD146-targeted immunoPET and NIRF Imaging of Hepatocellular Carcinoma with a Dual-Labeled Monoclonal Antibody
- Author
-
Weibo Cai, Emily B. Ehlerding, Haiyan Sun, Reinier Hernandez, Yunan Yang, Hector F. Valdovinos, Christopher G. England, and Todd E. Barnhart
- Subjects
0301 basic medicine ,Biodistribution ,Pathology ,medicine.medical_specialty ,Carcinoma, Hepatocellular ,medicine.drug_class ,Medicine (miscellaneous) ,Mice, Nude ,Siderophores ,CD146 Antigen ,Deferoxamine ,Monoclonal antibody ,Cell Line ,03 medical and health sciences ,hepatocellular carcinoma (HCC) ,0302 clinical medicine ,89Zr ,In vivo ,medicine ,Animals ,Humans ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,Fluorescent Dyes ,medicine.diagnostic_test ,Staining and Labeling ,Chemistry ,Liver Neoplasms ,Optical Imaging ,near-infrared fluorescence (NIRF) imaging ,Antibodies, Monoclonal ,medicine.disease ,3. Good health ,Quaternary Ammonium Compounds ,Disease Models, Animal ,030104 developmental biology ,Positron emission tomography ,CD146 ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,Positron-Emission Tomography ,Positron emission tomography (PET) ,dual-modality imaging ,Hepatocytes ,Heterografts ,Molecular imaging ,Sulfonic Acids ,Ex vivo ,Research Paper - Abstract
Overexpression of CD146 has been correlated with aggressiveness, recurrence rate, and poor overall survival in hepatocellular carcinoma (HCC) patients. In this study, we set out to develop a CD146-targeting probe for high-contrast noninvasive in vivo positron emission tomography (PET) and near-infrared fluorescence (NIRF) imaging of HCCs. YY146, an anti-CD146 monoclonal antibody, was employed as a targeting molecule to which we conjugated the zwitterionic near-infrared fluorescence (NIRF) dye ZW800-1 and the chelator deferoxamine (Df). This enabled labeling of Df-YY146-ZW800 with (89)Zr and its subsequent detection using PET and NIRF imaging, all without compromising antibody binding properties. Two HCC cell lines expressing high (HepG2) and low (Huh7) levels of CD146 were employed to generate subcutaneous (s.c.) and orthotopic xenografts in athymic nude mice. Sequential PET and NIRF imaging performed after intravenous injection of (89)Zr-Df-YY146-ZW800 into tumor-bearing mice unveiled prominent and persistent uptake of the tracer in HepG2 tumors that peaked at 31.65 ± 7.15 percentage of injected dose per gram (%ID/g; n=4) 72 h post-injection. Owing to such marked accumulation, tumor delineation was successful by both PET and NIRF, which facilitated the fluorescence image-guided resection of orthotopic HepG2 tumors, despite the relatively high liver background. CD146-negative Huh7 and CD146-blocked HepG2 tumors exhibited significantly lower (89)Zr-Df-YY146-ZW800 accretion (6.1 ± 0.5 and 8.1 ± 1.0 %ID/g at 72 h p.i., respectively; n=4), demonstrating the CD146-specificity of the tracer in vivo. Ex vivo biodistribution and immunofluorescent staining corroborated the accuracy of the imaging data and correlated tracer uptake with in situ CD146 expression. Overall, (89)Zr-Df-YY146-ZW800 showed excellent properties as a PET/NIRF imaging agent, including high in vivo affinity and specificity for CD146-expressing HCC. CD146-targeted molecular imaging using dual-labeled YY146 has great potential for early detection, prognostication, and image-guided surgical resection of liver malignancies.
- Published
- 2016
44. Targeting CD146 with a 64 Cu-labeled antibody enables in vivo immunoPET imaging of high-grade gliomas
- Author
-
Christopher G. England, Yunan Yang, Jinrong Wu, Mary E. Meyerand, Jun Rao, Weibo Cai, Reinier Hernandez, Christina M. Lewis, Yazhuo Qu, Xiu-wu Bian, Stephen A. Graves, Robert J. Nickles, Li Yin, and Pu Wang
- Subjects
Pathology ,medicine.medical_specialty ,Epithelial-Mesenchymal Transition ,Time Factors ,Transcription, Genetic ,medicine.drug_class ,medicine.medical_treatment ,Fluorescent Antibody Technique ,Mice, Nude ,CD146 Antigen ,Biology ,Monoclonal antibody ,Malignancy ,Targeted therapy ,Subcutaneous Tissue ,In vivo ,Cancer stem cell ,Cell Line, Tumor ,Glioma ,medicine ,Animals ,Humans ,Tissue Distribution ,Epithelial–mesenchymal transition ,Multidisciplinary ,Brain Neoplasms ,Antibodies, Monoclonal ,Flow Cytometry ,medicine.disease ,Magnetic Resonance Imaging ,Xenograft Model Antitumor Assays ,Clone Cells ,Phenotype ,PNAS Plus ,Copper Radioisotopes ,Positron-Emission Tomography ,Protein Biosynthesis ,Antibody Formation ,Neoplastic Stem Cells ,CD146 ,Neoplasm Grading ,Tomography, X-Ray Computed - Abstract
Given the highly heterogeneous character of brain malignancies and the associated implication for its proper diagnosis and treatment, finding biomarkers that better characterize this disease from a molecular standpoint is imperative. In this study, we evaluated CD146 as a potential molecular target for diagnosis and targeted therapy of glioblastoma multiforme (GBM), the most common and lethal brain malignancy. YY146, an anti-CD146 monoclonal antibody, was generated and radiolabeled for noninvasive positron-emission tomography (PET) imaging of orthotopic GBM models. (64)Cu-labeled YY146 preferentially accumulated in the tumors of mice bearing U87MG xenografts, which allowed the acquisition of high-contrast PET images of small tumor nodules (∼ 2 mm). Additionally, we found that tumor uptake correlated with the levels of CD146 expression in a highly specific manner. We also explored the potential therapeutic effects of YY146 on the cancer stem cell (CSC) and epithelial-to-mesenchymal (EMT) properties of U87MG cells, demonstrating that YY146 can mitigate those aggressive phenotypes. Using YY146 as the primary antibody, we performed histological studies of World Health Organization (WHO) grades I through IV primary gliomas. The positive correlation found between CD146-positive staining and high tumor grade (χ(2) = 9.028; P = 0.029) concurred with the GBM data available in The Cancer Genome Atlas (TCGA) and validated the clinical value of YY146. In addition, we demonstrate that YY146 can be used to detect CD146 in various cancer cell lines and human resected tumor tissues of multiple other tumor types (gastric, ovarian, liver, and lung), indicating a broad applicability of YY146 in solid tumors.
- Published
- 2015
45. Noninvasive brain cancer imaging with a bispecific antibody fragment, generated via click chemistry
- Author
-
Robert J. Nickles, Haiming Luo, Weibo Cai, Reinier Hernandez, Stephen A. Graves, Christopher G. England, Charles P. Theuer, Yunan Yang, and Hao Hong
- Subjects
Pathology ,medicine.medical_specialty ,Antibodies, Neoplasm ,Contrast Media ,Immunoglobulin Fab Fragments ,Mice ,Growth factor receptor ,Antibodies, Bispecific ,medicine ,Animals ,Multidisciplinary ,biology ,Chemistry ,Brain Neoplasms ,Cancer ,Neoplasms, Experimental ,Endoglin ,Biological Sciences ,medicine.disease ,Immunoconjugate ,Radiography ,Positron-Emission Tomography ,Cancer research ,biology.protein ,Click Chemistry ,Bioorthogonal chemistry ,Antibody ,Ligation - Abstract
Early diagnosis remains a task of upmost importance for reducing cancer morbidity and mortality. Successful development of highly specific companion diagnostics targeting aberrant molecular pathways of cancer is needed for sensitive detection, accurate diagnosis, and opportune therapeutic intervention. Herein, we generated a bispecific immunoconjugate [denoted as Bs-F(ab)2] by linking two antibody Fab fragments, an anti-epidermal growth factor receptor (EGFR) Fab and an anti-CD105 Fab, via bioorthogonal "click" ligation of trans-cyclooctene and tetrazine. PET imaging of mice bearing U87MG (EGFR/CD105(+/+)) tumors with (64)Cu-labeled Bs-F(ab)2 revealed a significantly enhanced tumor uptake [42.9 ± 9.5 percentage injected dose per gram (%ID/g); n = 4] and tumor-to-background ratio (tumor/muscle ratio of 120.2 ± 44.4 at 36 h postinjection; n = 4) compared with each monospecific Fab tracer. Thus, we demonstrated that dual targeting of EGFR and CD105 provides a synergistic improvement on both affinity and specificity of (64)Cu-NOTA-Bs-F(ab)2. (64)Cu-NOTA-Bs-F(ab)2 was able to visualize small U87MG tumor nodules (
- Published
- 2015
46. PET Imaging of VEGFR-2 Expression in Lung Cancer with 64Cu-Labeled Ramucirumab
- Author
-
Christopher G. England, Haiyan Sun, Haiming Luo, Robert J. Nickles, Stephen A. Graves, Weibo Cai, and Glenn Liu
- Subjects
0301 basic medicine ,Biodistribution ,Lung Neoplasms ,Mice, Nude ,Antibodies, Monoclonal, Humanized ,Article ,Flow cytometry ,Ramucirumab ,03 medical and health sciences ,chemistry.chemical_compound ,Heterocyclic Compounds, 1-Ring ,Mice ,0302 clinical medicine ,In vivo ,Heterocyclic Compounds ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,Tissue Distribution ,Lung cancer ,medicine.diagnostic_test ,business.industry ,Antibodies, Monoclonal ,medicine.disease ,Vascular Endothelial Growth Factor Receptor-2 ,Xenograft Model Antitumor Assays ,Imaging agent ,Molecular Imaging ,Vascular endothelial growth factor ,030104 developmental biology ,chemistry ,Copper Radioisotopes ,030220 oncology & carcinogenesis ,Isotope Labeling ,Positron-Emission Tomography ,embryonic structures ,Cancer research ,Radiopharmaceuticals ,business ,Ex vivo - Abstract
Lung cancer accounts for 17% of cancer-related deaths worldwide, and most patients present with locally advanced or metastatic disease. Novel PET imaging agents for assessing vascular endothelial growth factor receptor-2 (VEGFR-2) expression can be used for detecting VEGFR-2–positive malignancies and subsequent monitoring of therapeutic response to VEGFR-2–targeted therapies. Here, we report the synthesis and characterization of an antibody-based imaging agent for PET imaging of VEGFR-2 expression in vivo. Methods: Ramucirumab (named RamAb), a fully humanized IgG1 monoclonal antibody, was conjugated to 2-S-(4-isothiocyanatobenzyl)-1,4,7-triazacyclononane-1,4,7-triacetic acid (p-SCN-Bn-NOTA) and labeled with 64Cu. Flow cytometry analysis and microscopy studies were performed to compare the VEGFR-2 binding affinity of RamAb and NOTA-RamAb. PET imaging and biodistribution studies were performed in nude mice bearing HCC4006 and A549 xenograft tumors. Ex vivo histopathology was performed to elucidate the expression patterns of VEGFR-2 in different tissues and organs to validate in vivo results. Results: Flow cytometry examination revealed the specific binding capacity of fluorescein isothiocyanate-RamAb to VEGFR-2, and no difference in VEGFR-2 binding affinity was seen between RamAb and NOTA-RamAb. After being labeled with 64Cu, PET imaging revealed specific and prominent uptake of 64Cu-NOTA-RamAb in VEGFR-2–positive HCC4006 tumors (9.4 ± 0.5 percentage injected dose per gram at 48 h after injection; n = 4) and significantly lower uptake in VEGFR-2–negative A549 tumors (4.3 ± 0.2 percentage injected dose per gram at 48 h after injection; n = 3). Blocking experiments revealed significantly lower uptake in HCC4006 tumors, along with histology analysis, further confirming the VEGFR-2 specificity of 64Cu-NOTA-RamAb. Conclusion: This study provides initial evidence that 64Cu-NOTA-RamAb can function as a PET imaging agent for visualizing VEGFR-2 expression in vivo, which may also find potential applications in monitoring the treatment response of VEGFR-2–targeted cancer therapy.
- Published
- 2015
47. Release Kinetics of Paclitaxel and Cisplatin from Two and Three Layered Gold Nanoparticles
- Author
-
Christopher G. England, John O. Trent, Hermann B. Frieboes, Ashani Kuttan, and M. Clarke Miller
- Subjects
Drug Liberation ,Paclitaxel ,Chemistry, Pharmaceutical ,Kinetics ,Pharmaceutical Science ,Nanoparticle ,Metal Nanoparticles ,Antineoplastic Agents ,Pharmacology ,Article ,chemistry.chemical_compound ,Phosphatidylcholine ,Cell Line, Tumor ,PEG ratio ,medicine ,Humans ,Sulfhydryl Compounds ,Cisplatin ,General Medicine ,Antineoplastic Agents, Phytogenic ,chemistry ,Models, Chemical ,Colloidal gold ,Delayed-Action Preparations ,Biophysics ,Phosphatidylcholines ,Gold ,Lipoproteins, HDL ,Hydrophobic and Hydrophilic Interactions ,Biotechnology ,medicine.drug - Abstract
Gold nanoparticles functionalized with biologically-compatible layers may achieve stable drug release while avoiding adverse effects in cancer treatment. We study cisplatin and paclitaxel release from gold cores functionalized with hexadecanethiol (TL) and phosphatidylcholine (PC) to form two-layer nanoparticles, or TL, PC, and high density lipoprotein (HDL) to form three-layer nanoparticles. Drug release was monitored for 14 days to assess long term effects of the core surface modifications on release kinetics. Release profiles were fitted to previously developed kinetic models to differentiate possible release mechanisms. The hydrophilic drug (cisplatin) showed an initial (5-hr.) burst, followed by a steady release over 14 days. The hydrophobic drug (paclitaxel) showed a steady release over the same time period. Two layer nanoparticles released 64.0 ± 2.5% of cisplatin and 22.3 ± 1.5% of paclitaxel, while three layer nanoparticles released the entire encapsulated drug. The Korsmeyer-Peppas model best described each release scenario, while the simplified Higuchi model also adequately described paclitaxel release from the two layer formulation. We conclude that functionalization of gold nanoparticles with a combination of TL and PC may help to modulate both hydrophilic and hydrophobic drug release kinetics, while the addition of HDL may enhance long term release of hydrophobic drug.
- Published
- 2015
48. Novel Preparation Methods of 52Mn for ImmunoPET Imaging
- Author
-
Hector F. Valdovinos, Weibo Cai, Christopher G. England, Gregory Severin, Charles P. Theuer, Dennis Ringkjøbing Elema, Todd E. Barnhart, Jesper Fonslet, Paul A. Ellison, Robert J. Nickles, Reinier Hernandez, and Stephen A. Graves
- Subjects
Chromium ,Biodistribution ,Organic anion exchange ,Biomedical Engineering ,Pharmaceutical Science ,chemistry.chemical_element ,Molecular imaging ,Bioengineering ,TRC105 ,Article ,ImmunoPET ,Tumor angiogenesis ,chemistry.chemical_compound ,Heterocyclic Compounds, 1-Ring ,Nuclear magnetic resonance ,In vivo ,DOTA ,Animals ,Chelation ,Tissue Distribution ,DOTA chelation ,Manganese-52 (Mn-52, 52Mn) ,Pharmacology ,Radioisotopes ,Manganese ,Mice, Inbred BALB C ,Aqueous solution ,Radiochemistry ,CD105/Endoglin ,Chemistry ,Organic Chemistry ,Antibodies, Monoclonal ,Magnetic Resonance Imaging ,Xenograft Model Antitumor Assays ,Isotope Labeling ,Positron-Emission Tomography ,Positron emission tomography (PET) ,Trace metal analysis (MP-AES, ICP-OES) ,Female ,Radiopharmaceuticals ,Ex vivo ,Biotechnology ,Conjugate - Abstract
52Mn (t1/2 =5.59 d, ß+ = 29.6%, Eßave = 0.24 MeV) shows promise in positron emission tomography (PET) and in dual-modality manganese-enhanced magnetic resonance imaging (MEMRI) applications including neural tractography, stem cell tracking, and biological toxicity studies. The extension to bioconjugate application requires high specific activity 52Mn in a state suitable for macromolecule labeling. To that end a 52Mn production, purification, and labeling system is presented, and its applicability in preclinical, macromolecule PET is shown using the conjugate 52Mn-DOTA-TRC105. 52Mn is produced by 60 µA, 16 MeV proton irradiation of natural chromium metal pressed into a silver disc support. Radiochemical separation proceeds by strong anion exchange chromatography of the dissolved Cr target, employing a semi-organic mobile phase, 97:3 (v:v) ethanol: HCl (11M, aqueous). The method is 62 ± 14% efficient (n=7) in 52Mn recovery, leading to a separation factor from Cr of (1.6 ± 1.0) x106 (n = 4), and an average effective specific activity of 0.8 GBq/µmol (n = 4) in titration against DOTA. 52Mn-DOTA-TRC105 conjugation and labeling demonstrate the potential for chelation applications. In vivo images acquired using PET/CT in mice bearing 4T1 xenograft tumors are presented. Peak tumor uptake is 18.7 ± 2.7 %ID/g at 24 hours post injection and ex vivo 52Mn biodistribution validates the in vivo PET data. Free 52Mn2+(as chloride or acetate) is used as a control in additional mice to evaluate the non-targeted biodistribution in the tumor model.
- Published
- 2015
49. A Review of Pharmacological Treatment Options for Lung Cancer: Emphasis on Novel Nanotherapeutics and Associated Toxicity
- Author
-
Christopher G. England, Chin F. Ng, Hermann B. Frieboes, and Victor van Berkel
- Subjects
medicine.medical_specialty ,Lung Neoplasms ,Clinical Biochemistry ,Antineoplastic Agents ,Pharmacology ,Tumor response ,Pharmacological treatment ,Drug Discovery ,Tumor Microenvironment ,Medicine ,Humans ,Lung cancer ,Intensive care medicine ,Drug Carriers ,business.industry ,medicine.disease ,Clinical trial ,Nanomedicine ,Solubility ,Drug delivery ,Toxicity ,Molecular Medicine ,Nanoparticles ,business ,Potential toxicity - Abstract
Lung cancer remains a leading cause of death. Current treatment options are generally ineffective, highlighting the dire need for novel approaches. While numerous biologically-active chemotherapeutics have been discovered in the last two decades, biological barriers including minimal water solubility, stability, and cellular resistance hinder in vivo effectiveness. To overcome these limitations, nanoparticles have been designed to deliver chemotherapeutics selectively to cancerous tissue while minimizing pharmacokinetics hindrance. Numerous studies are underway analyzing the efficacy of nanoparticles in drug delivery, theranostic applications, and photothermal therapy. However, while nanoparticles have shown efficacy in treating some cancers, their potential toxicity and lack of targeting may hinder clinical potential. With the aim to help sort through these issues, we conduct a review to describe recent applications of nanotherapeutics for the treatment and diagnosis of lung cancer. We first provide a detailed background of statistics, etiology, histological classification, staging, diagnosis, and current treatment options. This is followed by a description of current applications of nanotherapeutics, focusing primarily on results published during the past five years. The potential toxicity associated with nanoparticles is evaluated, revealing inconclusive information which highlights the need for further studies. Lastly, recent advances in mathematical modeling and computational simulation have shown potential in predicting tumor response to nanotherapeutics. Thus, although nanoparticles have shown promise in treating lung cancer, further multi-disciplinary studies to quantify optimal dosages and assess possible toxicity are still needed. To this end, nanotherapeutic options currently in clinical trials offer hope to help address some of these critical issues.
- Published
- 2014
50. Enhanced penetration into 3D cell culture using two and three layered gold nanoparticles
- Author
-
Xinghua Sun, Christopher G. England, Andre M. Gobin, Dhruvinkumar Patel, Thomas Priest, Victor van Berkel, Hermann B. Frieboes, Lacey R. McNally, and Guandong Zhang
- Subjects
Pathology ,medicine.medical_specialty ,Materials science ,nanovector transport ,Cell Culture Techniques ,Biophysics ,Metal Nanoparticles ,Pharmaceutical Science ,Nanoparticle ,Bioengineering ,Enhanced permeability and retention effect ,Diffusion ,Biomaterials ,3D cell culture ,cancer nanotherapy ,In vivo ,International Journal of Nanomedicine ,Cell Line, Tumor ,Drug Discovery ,medicine ,Humans ,Tissue Distribution ,Original Research ,tumor hypoxia ,Neovascularization, Pathologic ,Tumor hypoxia ,Organic Chemistry ,Neoplasms, Experimental ,General Medicine ,Penetration (firestop) ,Nanoshell ,Colloidal gold ,Gold - Abstract
Christopher G England,1 Thomas Priest,2 Guandong Zhang,2 Xinghua Sun,2 Dhruvinkumar N Patel,2 Lacey R McNally,3,4 Victor van Berkel,4,5 André M Gobin,2 Hermann B Frieboes1,2,41Department of Pharmacology and Toxicology, 2Department of Bioengineering, 3Department of Medicine, 4James Graham Brown Cancer Center, 5Department of Surgery, University of Louisville, KY, USAAbstract: Nano-scale particles sized 10–400 nm administered systemically preferentially extravasate from tumor vasculature due to the enhanced permeability and retention effect. Therapeutic success remains elusive, however, because of inhomogeneous particle distribution within tumor tissue. Insufficient tumor vascularization limits particle transport and also results in avascular hypoxic regions with non-proliferating cells, which can regenerate tissue after nanoparticle-delivered cytotoxicity or thermal ablation. Nanoparticle surface modifications provide for increasing tumor targeting and uptake while decreasing immunogenicity and toxicity. Herein, we created novel two layer gold-nanoshell particles coated with alkanethiol and phosphatidylcholine, and three layer nanoshells additionally coated with high-density-lipoprotein. We hypothesize that these particles have enhanced penetration into 3-dimensional cell cultures modeling avascular tissue when compared to standard poly(ethylene glycol) (PEG)-coated nanoshells. Particle uptake and distribution in liver, lung, and pancreatic tumor cell cultures were evaluated using silver-enhancement staining and hyperspectral imaging with dark field microscopy. Two layer nanoshells exhibited significantly higher uptake compared to PEGylated nanoshells. This multilayer formulation may help overcome transport barriers presented by tumor vasculature, and could be further investigated in vivo as a platform for targeted cancer therapies.Keywords: cancer nanotherapy, tumor hypoxia, nanovector transport
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.