Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jonathan W. Engle is active.

Publication


Featured researches published by Jonathan W. Engle.


ACS Nano | 2012

In Vivo Targeting and Imaging of Tumor Vasculature with Radiolabeled, Antibody-Conjugated Nanographene

Hao Hong; Kai Yang; Yin Zhang; Jonathan W. Engle; Liangzhu Feng; Yunan Yang; Tapas R. Nayak; Shreya Goel; Jero Bean; Charles P. Theuer; Todd E. Barnhart; Zhuang Liu; Weibo Cai

Herein we demonstrate that nanographene can be specifically directed to the tumor neovasculature in vivo through targeting of CD105 (i.e., endoglin), a vascular marker for tumor angiogenesis. The covalently functionalized nanographene oxide (GO) exhibited excellent stability and target specificity. Pharmacokinetics and tumor targeting efficacy of the GO conjugates were investigated with serial noninvasive positron emission tomography imaging and biodistribution studies, which were validated by in vitro, in vivo, and ex vivo experiments. The incorporation of an active targeting ligand (TRC105, a monoclonal antibody that binds to CD105) led to significantly improved tumor uptake of functionalized GO, which was specific for the neovasculature with little extravasation, warranting future investigation of these GO conjugates for cancer-targeted drug delivery and/or photothermal therapy to enhance therapeutic efficacy. Since poor extravasation is a major hurdle for nanomaterial-based tumor targeting in vivo, this study also establishes CD105 as a promising vascular target for future cancer nanomedicine.


Biomaterials | 2012

In Vivo Targeting and Positron Emission Tomography Imaging of Tumor Vasculature with 66Ga-Labeled Nano-Graphene

Hao Hong; Yin Zhang; Jonathan W. Engle; Tapas R. Nayak; Charles P. Theuer; Robert J. Nickles; Todd E. Barnhart; Weibo Cai

The goal of this study was to employ nano-graphene for tumor targeting in an animal tumor model, and quantitatively evaluate the pharmacokinetics and tumor targeting efficacy through positron emission tomography (PET) imaging using (66)Ga as the radiolabel. Nano-graphene oxide (GO) sheets with covalently linked, amino group-terminated six-arm branched polyethylene glycol (PEG; 10 kDa) chains were conjugated to NOTA (1,4,7-triazacyclononane-1,4,7-triacetic acid, for (66)Ga-labeling) and TRC105 (an antibody that binds to CD105). Flow cytometry analyses, size measurements, and serum stability studies were performed to characterize the GO conjugates before in vivo investigations in 4T1 murine breast tumor-bearing mice, which were further validated by histology. TRC105-conjugated GO was specific for CD105 in cell culture. (66)Ga-NOTA-GO-TRC105 and (66)Ga-NOTA-GO exhibited excellent stability in complete mouse serum. In 4T1 tumor-bearing mice, these GO conjugates were primarily cleared through the hepatobiliary pathway. (66)Ga-NOTA-GO-TRC105 accumulated quickly in the 4T1 tumors and tumor uptake remained stable over time (3.8 ± 0.4, 4.5 ± 0.4, 5.8 ± 0.3, and 4.5 ± 0.4 %ID/g at 0.5, 3, 7, and 24 h post-injection respectively; n = 4). Blocking studies with unconjugated TRC105 confirmed CD105 specificity of (66)Ga-NOTA-GO-TRC105, which was corroborated by biodistribution and histology studies. Furthermore, histological examination revealed that targeting of NOTA-GO-TRC105 is tumor vasculature CD105 specific with little extravasation. Successful demonstration of in vivo tumor targeting with GO, along with the versatile chemistry of graphene-based nanomaterials, makes them suitable nanoplatforms for future biomedical research such as cancer theranostics.


Nano Letters | 2011

Cancer-targeted optical imaging with fluorescent zinc oxide nanowires.

Hao Hong; Jian Shi; Yunan Yang; Yin Zhang; Jonathan W. Engle; Robert J. Nickles; Xudong Wang; Weibo Cai

Herein we demonstrate that intrinsically fluorescent zinc oxide (ZnO) nanowires (NWs) can be adopted for molecularly targeted imaging of cancer cells, after they are functionalized to render water solubility, biocompatibility, and low cellular toxicity. Optical imaging of integrin α(v)β(3) on U87MG human glioblastoma cells was achieved with RGD peptide-conjugated green fluorescent ZnO NWs, which opened up new avenues of research for investigating ZnO NW-based agents in tumor vasculature-targeted molecular imaging and drug delivery.


PLOS ONE | 2011

Positron Emission Tomography Imaging of CD105 Expression with a 64Cu-Labeled Monoclonal Antibody: NOTA Is Superior to DOTA

Yin Zhang; Hao Hong; Jonathan W. Engle; Jero Bean; Yunan Yang; Bryan R. Leigh; Todd E. Barnhart; Weibo Cai

Optimizing the in vivo stability of positron emission tomography (PET) tracers is of critical importance to cancer diagnosis. In the case of 64Cu-labeled monoclonal antibodies (mAb), in vivo behavior and biodistribution is critically dependent on the performance of the bifunctional chelator used to conjugate the mAb to the radiolabel. This study compared the in vivo characteristics of 64Cu-labeled TRC105 (a chimeric mAb that binds to both human and murine CD105), through two commonly used chelators: 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA) and 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA). Flow cytometry analysis confirmed that chelator conjugation of TRC105 did not affect its CD105 binding affinity or specificity. PET imaging and biodistribution studies in 4T1 murine breast tumor-bearing mice revealed that 64Cu-NOTA-TRC105 exhibited better stability than 64Cu-DOTA-TRC105 in vivo, which resulted in significantly lower liver uptake without compromising the tumor targeting efficiency. In conclusion, this study confirmed that NOTA is a superior chelator to DOTA for PET imaging with 64Cu-labeled TRC105.


Molecular Pharmaceutics | 2012

Multimodality imaging of breast cancer experimental lung metastasis with bioluminescence and a monoclonal antibody dual-labeled with 89Zr and IRDye 800CW

Hao Hong; Yin Zhang; Gregory W. Severin; Yunan Yang; Jonathan W. Engle; Gang Niu; Robert J. Nickles; Xiaoyuan Chen; Bryan R. Leigh; Todd E. Barnhart; Weibo Cai

Metastatic breast cancer is incurable. The goal of this study was to develop a positron emission tomography (PET)/near-infrared fluorescent (NIRF) probe for imaging CD105 expression in breast cancer experimental lung metastasis. TRC105, a chimeric anti-CD105 antibody, was dual-labeled with a NIRF dye (IRDye 800CW) and (89)Zr to yield (89)Zr-Df-TRC105-800CW. Luciferase-transfected 4T1 murine breast cancer cells were injected intravenously into female mice to establish the tumor model. Bioluminescence imaging (BLI) was carried out to noninvasively monitor the lung tumor burden. PET imaging revealed that 4T1 lung tumor uptake of (89)Zr-Df-TRC105-800CW was 8.7 ± 1.4, 10.9 ± 0.5, and 9.7 ± 1.1% ID/g at 4, 24, and 48 h postinjection (n = 4), with excellent tumor contrast. Biodistribution studies, blocking, control studies with (89)Zr-Df-cetuximab-800CW, ex vivo BLI/PET/NIRF imaging, and histology all confirmed CD105 specificity of the tracer. Broad clinical potential of TRC105-based agents was shown in many tumor types, which also enabled early detection of small metastasis and intraoperative guidance for tumor removal.


Applied Radiation and Isotopes | 2012

Cyclotron Produced 44gSc from Natural Calcium

Gregory W. Severin; Jonathan W. Engle; Hector F. Valdovinos; Todd E. Barnhart; Robert J. Nickles

(44g)Sc was produced by 16MeV proton irradiation of unenriched calcium metal with radionuclidic purity greater than 95%. The thick target yield at saturation for (44g)Sc was 213 MBq/μA, dwarfing the yields of contaminants (43)Sc,(44 m)Sc, (47)Sc and (48)Sc for practical bombardment times of 1-2h. Scandium was isolated from the dissolved calcium target by filtration, and reconstituted in small volumes of dilute HCl. Reactions with the chelate 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) indicated a reactivity of 54 ± 14 Gbq/μmol at end-of-bombardment.


Synapse | 2011

In Vivo Kinetics of (F-18)MEFWAY: A Comparison With (C-11)WAY100635 and (F-18)MPPF in the Nonhuman Primate

Dustin Wooten; J.D. Moraino; Ansel T. Hillmer; Jonathan W. Engle; O.J. DeJesus; Dhanabalan Murali; Todd E. Barnhart; Robert J. Nickles; Richard J. Davidson; Mary L. Schneider; Jogeshwar Mukherjee; Brad T. Christian

[F‐18]Mefway was developed to provide an F‐18 labeled positron emission tomography (PET) neuroligand with high affinity for the serotonin 5‐HT1A receptor to improve the in vivo assessment of the 5‐HT1A system. The goal of this work was to compare the in vivo kinetics of [F‐18]mefway, [F‐18]MPPF, and [C‐11]WAY100635 in the rhesus monkey. Methods: Each of four monkeys were given bolus injections of [F‐18]mefway, [C‐11]WAY100635, and [F‐18]MPPF and scans were acquired with a microPET P4 scanner. Arterial blood was sampled to assay parent compound throughout the time course of the PET experiment. Time activity curves were extracted in the high 5‐HT1A binding areas of the anterior cingulate cortex (ACG), mesial temporal cortex, raphe nuclei, and insula cortex. Time activity curves were also extracted in the cerebellum, which was used as a reference region. The in vivo kinetics of the radiotracers were compared based on the nondisplaceable distribution volume (VND) and binding potential (BPND). Results: At 30 min, the fraction of radioactivity in the plasma due to parent compound was 19%, 28%, and 29% and cleared from the arterial plasma at rates of 0.0031, 0.0078, and 0.0069 (min−1) ([F‐18]mefway, [F‐18]MPPF, [C‐11]WAY100635). The BPND in the brain regions were mesial temporal cortex: 7.4 ± 0.6, 3.1 ± 0.4, 7.0 ± 1.2, ACG: 7.2 ± 1.2, 2.1 ± 0.2, 7.9 ± 1.2; raphe nuclei: 3.7 ± 0.6, 1.3 ± 0.3, 3.3 ± 0.7; and insula cortex: 4.2 ± 0.6, 1.2 ± 0.1, 4.7 ± 1.0 for [F‐18]mefway, [F‐18]MPPF, and [C‐11]WAY100635 respectively. Conclusions: In the rhesus monkey, [F‐18]mefway has similar in vivo kinetics to [C‐11]WAY100635 and yields greater than 2‐fold higher BPND than [F‐18]MPPF. These properties make [F‐18]mefway a promising radiotracer for 5‐HT1A assay, providing higher counting statistics and a greater dynamic range in BPND. Synapse, 2011.


Medicinal Chemistry | 2011

89Zr Radiochemistry for Positron Emission Tomography

Gregory W. Severin; Jonathan W. Engle; Todd E. Barnhart; R. Jerry Nickles

The positron emitting isotope (89)Zr is an ideal radionuclide for use in positron emission tomography (PET) imaging with monoclonal antibodies (mAbs). This article reviews the cyclotron physics of (89)Zr production, and the chemical separation methods for isolating it from yttrium target material. (89)Zr coordination with the bifunctional chelate desferrioxamine B is discussed, along with the common procedures for attaching the chelate to mAbs. The review is intended to detail the procedure for creating (89)Zr labeled mAbs, going from cyclotron to PET.


Molecular Pharmaceutics | 2012

Positron emission tomography and optical imaging of tumor CD105 expression with a dual-labeled monoclonal antibody.

Yin Zhang; Hao Hong; Jonathan W. Engle; Yunan Yang; Charles P. Theuer; Todd E. Barnhart; Weibo Cai

CD105 (endoglin) is an independent prognostic marker for poor prognosis in >10 solid tumor types, including breast cancer. The goal of this study was to develop a CD105-specific agent for both positron emission tomography (PET) and near-infrared fluorescence (NIRF) imaging, which can have potential clinical applications in diagnosis and imaged-guided surgery of breast cancer. TRC105, a chimeric anti-CD105 monoclonal antibody, was labeled with both a NIRF dye (i.e., 800CW) and (64)Cu to yield (64)Cu-NOTA-TRC105-800CW. Flow cytometry analysis revealed no difference in CD105 binding affinity/specificity between TRC105 and NOTA-TRC105-800CW. Serial PET imaging revealed that the 4T1 murine breast tumor uptake of (64)Cu-NOTA-TRC105-800CW was 5.2 ± 2.7, 11.0 ± 1.4, and 13.0 ± 0.4% ID/g at 4, 24, and 48 h postinjection respectively. Tumor uptake as measured by ex vivo NIRF imaging exhibited a good linear correlation with the % ID/g values obtained from PET (R = 0.74). Biodistribution data were consistent with the PET/NIRF findings. Blocking experiments, control studies with dual-labeled cetuximab (an isotype-matched control antibody), and histology confirmed the CD105 specificity of (64)Cu-NOTA-TRC105-800CW. Successful PET/NIRF imaging of CD105 expression warrants further investigation and clinical translation of dual-labeled TRC105-based imaging agents.


Journal of Chromatography A | 2015

Application of ion exchange and extraction chromatography to the separation of actinium from proton-irradiated thorium metal for analytical purposes

Valery Radchenko; Jonathan W. Engle; Justin J. Wilson; Joel R. Maassen; F.M. Nortier; Wayne A. Taylor; Eva R. Birnbaum; L.A. Hudston; Kevin D. John; Michael E. Fassbender

Actinium-225 (t1/2=9.92d) is an α-emitting radionuclide with nuclear properties well-suited for use in targeted alpha therapy (TAT), a powerful treatment method for malignant tumors. Actinium-225 can also be utilized as a generator for (213)Bi (t1/2 45.6 min), which is another valuable candidate for TAT. Actinium-225 can be produced via proton irradiation of thorium metal; however, long-lived (227)Ac (t1/2=21.8a, 99% β(-), 1% α) is co-produced during this process and will impact the quality of the final product. Thus, accurate assays are needed to determine the (225)Ac/(227)Ac ratio, which is dependent on beam energy, irradiation time and target design. Accurate actinium assays, in turn, require efficient separation of actinium isotopes from both the Th matrix and highly radioactive activation by-products, especially radiolanthanides formed from proton-induced fission. In this study, we introduce a novel, selective chromatographic technique for the recovery and purification of actinium isotopes from irradiated Th matrices. A two-step sequence of cation exchange and extraction chromatography was implemented. Radiolanthanides were quantitatively removed from Ac, and no non-Ac radionuclidic impurities were detected in the final Ac fraction. An (225)Ac spike added prior to separation was recovered at ≥ 98%, and Ac decontamination from Th was found to be ≥ 10(6). The purified actinium fraction allowed for highly accurate (227)Ac determination at analytical scales, i.e., at (227)Ac activities of 1-100 kBq (27 nCi to 2.7 μCi).

Collaboration


Dive into the Jonathan W. Engle's collaboration.

Top Co-Authors

Avatar

Todd E. Barnhart

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Weibo Cai

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Eva R. Birnbaum

Los Alamos National Laboratory

View shared research outputs
Top Co-Authors

Avatar

Robert J. Nickles

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

F.M. Nortier

Los Alamos National Laboratory

View shared research outputs
Top Co-Authors

Avatar

Kevin D. John

Los Alamos National Laboratory

View shared research outputs
Top Co-Authors

Avatar

Hao Hong

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Yunan Yang

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Michael E. Fassbender

Los Alamos National Laboratory

View shared research outputs
Top Co-Authors

Avatar

Yin Zhang

University of Wisconsin-Madison

View shared research outputs
Researchain Logo
Decentralizing Knowledge