Network


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

Hotspot


Dive into the research topics where Monica Wang is active.

Publication


Featured researches published by Monica Wang.


Bioorganic & Medicinal Chemistry Letters | 2012

Microfluidic technology: An economical and versatile approach for the synthesis of O-(2-[18F]fluoroethyl)-l-tyrosine ([18F]FET)

Vincent Bouvet; Melinda Wuest; Pui-Hang Tam; Monica Wang; Frank Wuest

A new synthesis of O-(2-[(18)F]fluoroethyl)-L-tyrosine [(18)F]FET was developed using a NanoTek® microfluidic synthesis system (Advion BioSciences, Inc.). Optimal reaction conditions were studied through screening different reaction parameters like temperature, flow rate, reaction time, concentration of the labeling precursor, and the applied volume ratio between the labeling precursor and [(18)F]fluoride. [(18)F]FET was obtained after HPLC purification with 50% decay-corrected radiochemical yield starting from as little as 40 μg of labeling precursor. Small animal PET studies in EMT-6 tumor bearing mice showed radioactivity accumulation in the tumor (SUV(60min) 1.21±0.2) resulting in an slightly increasing tumor-to-muscle ratio over time.


Breast Cancer Research | 2015

Targeting lysyl oxidase for molecular imaging in breast cancer

Melinda Wuest; Manuela Kuchar; Sai Kiran Sharma; Susan Richter; Ingrit Hamann; Monica Wang; Larissa Vos; John R. Mackey; Frank Wuest; Reik Löser

IntroductionLysyl oxidase (LOX; ExPASy ENZYME entry: EC 1.4.3.13) and members of the LOX-like family, LOXL1–LOXL4, are copper-dependent enzymes that can modify proteins of the extracellular matrix. Expression of LOX is elevated in many human cancers, including breast cancer. LOX expression correlates with the level of tissue hypoxia, and it is known to play a critical role in breast cancer metastasis. The goal of the present study was to target LOX with (1) molecular probe fluorescent labeling to visualize LOX in vitro and (2) a radiolabeled peptide to target LOX in vivo in three different preclinical models of breast cancer.MethodsGene expression of all five members of the LOX family was analyzed at the transcript level via microarray analysis using tissue biopsy samples from 176 patients with breast cancer. An oligopeptide sequence (GGGDPKGGGGG) was selected as a substrate-based, LOX-targeting structure. The peptide was labeled with fluorescein isothiocyanate (FITC) for confocal microscopy experiments with the murine breast cancer cell line EMT-6. In vivo molecular imaging experiments were performed using a C-terminal amidated peptide, GGGDPKGGGGG, labeled with a short-lived positron emitter, fluorine-18 (18F), for positron emission tomography (PET) in three different breast cancer models: EMT6, MCF-7 and MDA-MB-231. The PET experiments were carried out in the presence or absence of β-aminopropionitrile (BAPN), an irreversible inhibitor of LOX.ResultsImmunostaining experiments using a LOX-specific antibody on EMT-6 cells cultured under hypoxic conditions confirmed the elevation of LOX expression in these cells. An FITC-labeled oligopeptide, FITC-Ava-GGGDPKGGGGG-NH2, was found to be localized in different cellular compartments under these conditions. After injection of [18F]fluorobenzoate-GGGDPKGGGGG-NH2, radioactivity uptake was visible in all three breast cancer models in vivo. Tumor uptake was reduced by predosing the animals with 2 mg of BAPN 4 h or 24 h before injection of the radiotracer.ConclusionsThe present data support further investigation into the development of LOX-binding radiolabeled peptides as molecular probes for molecular imaging of LOX expression in cancer.


Nuclear Medicine and Biology | 2014

Synthesis and evaluation of 2-amino-5-(4-[18F]fluorophenyl)pent-4-ynoic acid ([18F]FPhPA): A novel 18F-labeled amino acid for oncologic PET imaging

Jenilee D. Way; Monica Wang; Ingrit Hamann; Melinda Wuest; Frank Wuest

INTRODUCTION (18)F-labeled amino acids are important PET radiotracers for molecular imaging of cancer. This study describes synthesis and radiopharmacological evaluation of 2-amino-5-(4-[(18)F]fluorophenyl)pent-4-ynoic acid ([(18)F]FPhPA) as a novel amino acid radiotracer for oncologic imaging. METHODS (18)F]FPhPA was prepared using Pd-mediated Sonogashira cross-coupling reaction between 4-[(18)F]fluoroiodobenzene ([(18)F]FIB) and propargylglycine. The radiopharmacological profile of [(18)F]FPhPA was evaluated in comparison with O-(2-[(18)F]fluoroethyl)-L-tyrosine ([(18)F]FET) using the murine breast cancer cell line EMT6 involving cellular uptake studies, radiotracer uptake competitive inhibition experiments and small animal PET imaging. RESULTS (18)F]FPhPA was prepared in 42±10% decay-corrected radiochemical yield with high radiochemical purity >95% after semi-preparative HPLC purification. Cellular uptake of L-[(18)F]FPhPA reached a maximum of 58±14 % radioactivity/mg protein at 90 min. Lower uptake was observed for racemic and D-[(18)F]FPhPA. Radiotracer uptake inhibition studies by synthetic and naturally occurring amino acids suggested that Na(+)-dependent system ASC, especially ASCT2, and Na(+)-independent system L are important amino acid transporters for [(18)F]FPhPA uptake into EMT6 cells. Small animal PET studies demonstrated similar high tumor uptake of [(18)F]FPhPA in EMT6 tumor-bearing mice compared to [(18)F]FET reaching a maximum standardized uptake value (SUV) of 1.35 after 60 min p.i.. Muscle uptake of [(18)F]FPhPA was higher (SUV30min=0.65) compared to [(18)F]FET (SUV30min=0.40), whereas [(18)F]FPhPA showed a more rapid uptake and clearance from the brain compared to [(18)F]FET. CONCLUSION L-[(18)F]FPhPA is the first (18)F-labeled amino acid prepared through Pd-mediated cross-coupling reaction. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE L-[(18)F]FPhPA displayed promising properties as a novel amino acid radiotracer for molecular imaging of system ASC and system L amino acid transporters in cancer.


EJNMMI research | 2014

Immuno-PET of epithelial ovarian cancer: harnessing the potential of CA125 for non-invasive imaging.

Sai Kiran Sharma; Melinda Wuest; Monica Wang; Darryl D. Glubrecht; Bonnie Andrais; Suzanne E. Lapi; Frank Wuest

BackgroundEpithelial ovarian cancer (EOC) is characterized by the overexpression of cancer antigen 125 (CA125), a mucinous glycoprotein that serves as a tumor biomarker. Early diagnosis of EOC is plagued by its asymptomatic nature of progression and the limitations of currently used immunoassay techniques that detect CA125 as a shed antigen in serum samples. Presently, there is no technique available for the in vivo evaluation of CA125 expression in malignant tissues. Moreover, there could be an unexplored pathophysiological time window for the detection of CA125 in EOC, during which it is expressed on tumor cells prior to being shed into the bloodstream. A method for the in vivo evaluation of CA125 expression on ovarian neoplasms earlier along disease progression and/or recurrence can potentially contribute to better disease management. To this end, the present work utilizes an anti-CA125 monoclonal antibody (MAb) and a single-chain variable fragment (scFv) labeled with the positron-emitting radionuclide 64Cu for preclinical molecular imaging of CA125 expression in vivo.MethodsAnti-CA125 MAb and scFv were prepared and functionally characterized for target binding prior to being tested as radiotracers in a preclinical setting.ResultsImmunoblotting, immunofluorescence, and flow cytometry revealed specific binding of CA125-targeting vectors to NIH:OVCAR-3 cells and no binding to antigen-negative SKOV3 cells. 64Cu-labeled anti-CA125 MAb and scFv were obtained in specific activities of 296 and 122 MBq/mg, respectively. Both radioimmunoconjugate vectors demonstrated highly selective binding to NIH:OVCAR-3 cells and virtually no binding to SKOV3 cells. In vivo radiopharmacological evaluation using xenograft mouse models injected with 64Cu-labeled anti-CA125 MAb provided a standardized uptake value (SUV) of 5.76 (29.70 %ID/g) in OVCAR3 tumors 24 h post-injection (p.i.) versus 1.80 (5.91 %ID/g) in SKOV3 tumors. 64Cu-labeled anti-CA125 scFv provided an SUV of 0.64 (3.21 %ID/g) in OVCAR3 tumors 24 h p.i. versus 0.25 (1.49 %ID/g) in SKOV3 tumors. Results from small-animal PET imaging were confirmed by ex vivo autoradiography and immunohistochemistry.ConclusionsRadiolabeling of anti-CA125 MAb and scFv with 64Cu did not compromise their immunoreactivity. Both radioimmunoconjugates presented specific tumor uptake and expected biological clearance profiles. This renders them as potential immuno-PET probes for targeted in vivo molecular imaging of CA125 in EOC.


Dalton Transactions | 2013

Synthesis, characterisation and evaluation of a novel copper-64 complex with selective uptake in EMT-6 cells under hypoxic conditions

James C. Knight; Melinda Wuest; Fawaz A. Saad; Monica Wang; David W. Chapman; Hans-Soenke Jans; Suzanne E. Lapi; Benson M. Kariuki; Angelo J. Amoroso; Frank Wuest

The radiometal (64)Cu is now widely used in the development of diagnostic imaging agents for positron emission tomography (PET). The present study has led to the development and evaluation of a novel chelating agent for (64)Cu: the new monothiourea tripodal ligand 1-benzoyl-3-{6-[(bis-pyridin-2-ylmethyl-amino)-methyl]-pyridin-2-yl}-thiourea (MTUBo). X-ray crystallographic analysis has shown this ligand forms a mononuclear complex with copper(II) and co-ordinates via a trigonal bipyramidal N4S array of donor atoms. Promisingly, cell uptake studies revealed that (64)Cu-MTUBo selectively accumulates in EMT-6 cells incubated under hypoxic conditions which may result from its relatively high Cu(II/I) redox potential. Small-animal PET imaging and ex vivo biodistribution studies in EMT-6 tumor bearing BALB/c mice revealed significant tumor uptake after 1 h p.i., yielding tumor-to-muscle (T/M) and tumor-to-blood (T/B) ratios of 8.1 and 1.1, respectively. However, injection of (64)Cu-acetate resulted in similar uptake indicating that the observed uptake was most likely non-specific. Despite showing high in vitro stability, it is likely that in vivo the complex undergoes transchelation to proteins within the blood in a relatively short timeframe. For comparison, the hypoxia imaging agent (64)Cu-ATSM was also evaluated in the same murine tumor model and showed about 60% higher tumor uptake than (64)Cu-MTUBo.


Cancer Biotherapy and Radiopharmaceuticals | 2012

In Vitro and In Vivo Evaluation of [18F]F-GAZ, a Novel Oxygen-Mimetic Azomycin-Glucose Conjugate, for Imaging Hypoxic Tumor

Melinda Wuest; Piyush Kumar; Monica Wang; Jennifer Yang; Hans-Soenke Jans; Leonard I. Wiebe

Several F-18-labeled 2-nitroimidazole (azomycin) derivatives have been proposed for imaging hypoxia using positron emission tomography (PET). Their cell penetration is based on passive diffusion, which limits their intracellular concentration maxima. The purpose of this study was to investigate the uptake of N-(2-[(18)F]fluoro-3-(6-O-glucosyl)propyl-azomycin ([(18)F]F-GAZ), a new azomycin-glucose conjugate, in vitro and in vivo. [(18)F]F-GAZ was synthesized from its tetraacetyl nosylate precursor by nucleophilic radiofluorination. [(18)F]F-GAZ was evaluated in vivo in EMT-6 tumor-bearing Balb/C mice utilizing the PET and biodistribution analysis. In vitro uptake of [(18)F]FDG by EMT-6 cells was measured in the presence of unlabeled F-GAZ, 2-FDG, and D-glucose. [(18)F]F-GAZ was rapidly cleared from all tissues, including the blood pool and kidneys, with ultimate accumulation in the urinary bladder. Uptake of tracer doses of [(18)F]F-GAZ into EMT-6 tumors was fast, reaching a standardized uptake value of 0.66±0.05 within 5-6 minutes postinjection (p.i.), and decreased to 0.24±0.04 by 60 minutes p.i. (n=6). A tumor-muscle ratio of 1.87±0.18 was observed after 60 minutes. Total uptake of [(18)F]F-GAZ in tumors (60 minutes) amounted to 1.25%±0.15% ID/g versus 0.61%±0.14% ID/g (n=4) in muscle. Similar biodistribution and excretion were observed using carrier-added (100 mg/kg) doses of F-GAZ. In vitro, D-glucose and unlabeled 2-FDG were two orders of magnitude more potent than F-GAZ as competitive inhibitors of [(18)F]FDG uptake into EMT-6 cells. Besides its interaction with glucose transporters, F-GAZ seems to be not transported in the presence of glucose. Furthermore, [(18)F]F-GAZ is unlikely to be effective as a hypoxia imaging agent. The low in vivo toxicity and substantial retention in tumor observed at high doses of F-GAZ do provide rationale for further testing as a radiosensitizer for external beam radiation therapy of radioresistant, hypoxic tumors.


EJNMMI research | 2016

PET imaging of cyclooxygenase-2 (COX-2) in a pre-clinical colorectal cancer model

Ole Tietz; Melinda Wuest; Alison Marshall; Darryl D. Glubrecht; Ingrit Hamann; Monica Wang; Cody Bergman; Jenilee D. Way; Frank Wuest


American journal of nuclear medicine and molecular imaging | 2016

Synthesis and pre-clinical evaluation of an 18F-labeled single-chain antibody fragment for PET imaging of epithelial ovarian cancer

Sai Kiran Sharma; Melinda Wuest; Jenilee D. Way; Vincent Bouvet; Monica Wang; Frank Wuest


Amino Acids | 2016

18F-Labeled wild-type annexin V: comparison of random and site-selective radiolabeling methods

Amanda Perreault; James C. Knight; Monica Wang; Jenilee Way; Frank Wuest


American journal of nuclear medicine and molecular imaging | 2012

Comparative functional evaluation of immunocompetent mouse breast cancer models established from PyMT-tumors using small animal PET with [(18)F]FDG and [(18)F]FLT.

Alan DeSilva; Melinda Wuest; Monica Wang; Jeff Hummel; Karen L. Mossman; Frank Wuest; Mary Hitt

Collaboration


Dive into the Monica Wang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Suzanne E. Lapi

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge