Network


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

Hotspot


Dive into the research topics where Nathaniel Wilganowski is active.

Publication


Featured researches published by Nathaniel Wilganowski.


Molecular Imaging and Biology | 2012

Dual-Labeling Strategies for Nuclear and Fluorescence Molecular Imaging: A Review and Analysis

Ali Azhdarinia; Pradip Ghosh; Sukhen C. Ghosh; Nathaniel Wilganowski; Eva M. Sevick-Muraca

Molecular imaging is used for the detection of biochemical processes through the development of target-specific contrast agents. Separately, modalities such as nuclear and near-infrared fluorescence (NIRF) imaging have been shown to non-invasively monitor disease. More recently, merging of these modalities has shown promise owing to their comparable detection sensitivity and benefited from the development of dual-labeled imaging agents. Dual-labeled agents hold promise for whole-body and intraoperative imaging and could bridge the gap between surgical planning and image-guided resection with a single, molecularly targeted agent. In this review, we summarized the literature for dual-labeled antibodies and peptides that have been developed and have highlighted key considerations for incorporating NIRF dyes into nuclear labeling strategies. We also summarized our findings on several commercially available NIRF dyes and offer perspectives for developing a toolkit to select the optimal NIRF dye and radiometal combination for multimodality imaging.


The Journal of Nuclear Medicine | 2012

Comparison of mAbs Targeting Epithelial Cell Adhesion Molecule for the Detection of Prostate Cancer Lymph Node Metastases with Multimodal Contrast Agents: Quantitative Small-Animal PET/CT and NIRF

Mary A. Hall; Kenneth L. Pinkston; Nathaniel Wilganowski; Holly Robinson; Pradip Ghosh; Ali Azhdarinia; Karina Vazquez-Arreguin; Arseniy M. Kolonin; Barrett R. Harvey; Eva M. Sevick-Muraca

The proliferation of most carcinomas is associated with an overexpression of epithelial cell adhesion molecule (EpCAM), a 40-kDa type I transmembrane protein found on epithelial cells yet absent from other cell types. The absence of EpCAM in normal lymphatics makes it an attractive marker for studying lymph node (LN) metastases of carcinomas to improve LN staging accuracy. Herein, we developed and quantitatively compared dual-labeled monoclonal antibodies (mAbs) of varying affinities against EpCAM for both noninvasive and intraoperative detection of metastatic LNs in prostate cancer. Methods: A panel of hybridoma-derived anti-EpCAM mAbs was generated and screened. Two high-affinity candidate mAbs with specificity for nonoverlapping epitopes on the EpCAM extracellular domain were chosen for further evaluation. After conjugation with DOTA for 64Cu radiolabeling and IRDye 800CW as a fluorophore, dual-labeled specific or isotype control mAb was administered intravenously to male nu/nu mice at 10–12 wk after orthotopic implantation of DsRed-expressing PC3 cells. Within 18–24 h, noninvasive small-animal PET/CT and in vivo, in situ, and ex vivo DsRed reporter gene and near-infrared fluorescence (NIRF) imaging were performed to detect primary tumors and metastatic LNs. Using DsRed fluorescence as the true indicator of cancer-positive tissue, we performed receiver operating characteristic curve analyses of percentage injected dose per gram measured from quantitative small-animal PET/CT and fluorescence intensity measured from semiquantitative NIRF imaging for each LN examined to compare mAb sensitivity and specificity. Results: mAbs 7 and 153 generated in-house were found to have higher affinity than commercial mAb 9601. Accuracy, as a function of sensitivity and specificity, for the detection of cancer-positive LNs during in vivo small-animal PET/CT was highest for mAbs 7 (87.0%) and 153 (78.0%) and significantly greater (P < 0.001) than random chance (50.0%). Rates for mAb 9601 (60.7%) and control mAb 69 (27.0%) were not significantly different from chance. Similarly, mAb 7 had significant detection accuracy by NIRF imaging (96.0%, P < 0.001). Conclusion: mAbs 7 and 153 are attractive, high-affinity candidates for further multimodal imaging agent optimization aimed at enhancing sensitivity and specificity for detection of metastatic LNs in prostate cancer. Fully quantitative NIRF imaging is needed for comprehensive analyses of NIRF-labeled agent accuracy for intraoperative guidance.


Bioorganic & Medicinal Chemistry | 2011

Characterization of chemical, radiochemical and optical properties of a dual-labeled MMP-9 targeting peptide

Ali Azhdarinia; Nathaniel Wilganowski; Holly Robinson; Pradip Ghosh; Sunkuk Kwon; ZaWaunyka Lazard; Alan R. Davis; Elizabeth A. Olmsted-Davis; Eva M. Sevick-Muraca

Optical imaging possesses similar sensitivity to nuclear imaging and has led to the emergence of multimodal approaches with dual-labeled nuclear/near-infrared (NIR) agents. The growing impact of (68)Ga (t(1/2)=68 min) labeled peptides on preclinical and clinical research offers a promising opportunity to merge the high spatial resolution of NIR imaging with the clinically-accepted positron emission tomography (PET). Previously, dual-labeled agents have been prepared with longer-lived radiometals and showed no detrimental effects on optical properties as a result of radiolabeling. In this study, we selected a peptide (M(2)) that targets MMP-2/9 and is dual-labeled with IRDye 800 CW and (68)Ga. Since (68)Ga chelation typically requires low pH (3.5-4) and elevated heating temperatures (95 °C), we sought to evaluate the impact of (68)Ga labeling on the optical properties of M(2). An efficient method for preparation of (68)Ga-M(2) was developed and reaction conditions were optimized. Stability studies in PBS, DTPA, and serum were performed and high levels of intact agent were evident under each condition. The addition of multiple reporters to a targeting agent adds further complexity to the characterization and validation and thus requires not only testing to ensure the agent is stable chemically and radiochemically, but also optically. Therefore, fluorescence properties were evaluated using a spectrofluorometer as well as by fluorescence detection via HPLC. It was determined that (68)Ga-labeling conditions did not impair the fluorescent properties of the agent. The agent was then used for in vivo imaging in a mouse model of heterotopic ossification (HO) with activated MMP-9 expression as an early biomarker which precedes mineralization. Although (68)Ga-complexation greatly reduced binding affinity of the peptide and negated tracer uptake on PET, NIR imaging showed consistent fluorescent signal that correlated to MMP-9 expression. This attests to the feasibility of using (68)Ga/NIR for dual-labeling of other peptides or small molecules for multimodality molecular imaging.


Journal of Medicinal Chemistry | 2013

Multimodal Chelation Platform for Near-Infrared Fluorescence/Nuclear Imaging

Sukhen C. Ghosh; Pradip Ghosh; Nathaniel Wilganowski; Holly Robinson; Mary A. Hall; Gabriel S. Dickinson; Ken Pinkston; Barrett R. Harvey; Eva M. Sevick-Muraca; Ali Azhdarinia

Dual-labeled compounds containing nuclear and near-infrared fluorescence contrast have the potential to molecularly guide surgical resection of cancer by extending whole-body diagnostic imaging findings into the surgical suite. To simplify the dual labeling process for antibody-based agents, we designed a multimodality chelation (MMC) scaffold which combined a radiometal chelating agent and fluorescent dye into a single moiety. Three dye-derivatized MMC compounds were synthesized and radiolabeled. The IRDye 800CW conjugate, 4, had favorable optical properties and showed rapid clearance in vivo. Using 4, an epithelial cell adhesion molecule (EpCAM) targeting MMC-immunoconjugate was prepared and dual-labeled with (64)Cu. In vitro binding activity was confirmed after MMC conjugation. Multimodal imaging studies showed higher tumor accumulation of (64)Cu-7 compared to nontargeted (64)Cu-4 in a prostate cancer model. Further evaluation in different EpCAM-expressing cell lines is warranted as well as application of the MMC dual labeling approach with other monoclonal antibodies.


Hepatology | 2015

The CaMKK2/CaMKIV Relay Is an Essential Regulator of Hepatic Cancer

Fumin Lin; Kathrina L. Marcelo; Kimal Rajapakshe; Cristian Coarfa; Adam Dean; Nathaniel Wilganowski; Holly Robinson; Eva M. Sevick; Karl-Dimiter Bissig; Lauren C. Goldie; Anthony R. Means; Brian York

Hepatic cancer is one of the most lethal cancers worldwide. Here, we report that the expression of Ca2+/calmodulin‐dependent protein kinase kinase 2 (CaMKK2) is significantly up‐regulated in hepatocellular carcinoma (HCC) and negatively correlated with HCC patient survival. The CaMKK2 protein is highly expressed in all eight hepatic cancer cell lines evaluated and is markedly up‐regulated relative to normal primary hepatocytes. Loss of CaMKK2 function is sufficient to inhibit liver cancer cell growth, and the growth defect resulting from loss of CaMKK2 can be rescued by ectopic expression of wild‐type CaMKK2 but not by kinase‐inactive mutants. Cellular ablation of CaMKK2 using RNA interference yields a gene signature that correlates with improvement in HCC patient survival, and ablation or pharmacological inhibition of CaMKK2 with STO‐609 impairs tumorigenicity of liver cancer cells in vivo. Moreover, CaMKK2 expression is up‐regulated in a time‐dependent manner in a carcinogen‐induced HCC mouse model, and STO‐609 treatment regresses hepatic tumor burden in this model. Mechanistically, CaMKK2 signals through Ca2+/calmodulin‐dependent protein kinase 4 (CaMKIV) to control liver cancer cell growth. Further analysis revealed that CaMKK2 serves as a scaffold to assemble CaMKIV with key components of the mammalian target of rapamycin/ribosomal protein S6 kinase, 70 kDa, pathway and thereby stimulate protein synthesis through protein phosphorylation. Conclusion: The CaMKK2/CaMKIV relay is an upstream regulator of the oncogenic mammalian target of rapamycin/ribosomal protein S6 kinase, 70 kDa, pathway, and the importance of this CaMKK2/CaMKIV axis in HCC growth is confirmed by the potent growth inhibitory effects of genetically or pharmacologically decreasing CaMKK2 activity; collectively, these findings suggest that CaMKK2 and CaMKIV may represent potential targets for hepatic cancer. (Hepatology 2015;62:505–520


Infection and Immunity | 2014

Targeting Pili in Enterococcal Pathogenesis

Kenneth L. Pinkston; Kavindra V. Singh; Peng Gao; Nathaniel Wilganowski; Holly Robinson; Sukhen C. Ghosh; Ali Azhdarinia; Eva M. Sevick-Muraca; Barbara E. Murray; Barrett R. Harvey

ABSTRACT Passive protection, the administration of antibodies to prevent infection, has garnered significant interest in recent years as a potential prophylactic countermeasure to decrease the prevalence of hospital-acquired infections. Pili, polymerized protein structures covalently anchored to the peptidoglycan wall of many Gram-positive pathogens, are ideal targets for antibody intervention, given their importance in establishing infection and their accessibility to antibody interactions. In this work, we demonstrated that a monoclonal antibody to the major component of Enterococcus faecalis pili, EbpC, labels polymerized pilus structures, diminishes biofilm formation, and significantly prevents the establishment of a rat endocarditis infection. The effectiveness of this anti-EbpC monoclonal provides strong evidence in support of its potential as a preventative. In addition, after radiolabeling, this monoclonal identified the site of enterococcal infection, providing a rare example of molecularly specific imaging of an established bacterial infection and demonstrating the versatility of this agent for use in future diagnostic and therapeutic applications.


Molecular Imaging and Biology | 2012

Albumin-Binding Domain Conjugate for Near-Infrared Fluorescence Lymphatic Imaging

Cynthia Davies-Venn; Bonnie Angermiller; Nathaniel Wilganowski; Pradip Ghosh; Barrett R. Harvey; Grace Wu; Sunkuk Kwon; Melissa B. Aldrich; Eva M. Sevick-Muraca

PurposeThe aim of this study was to develop and characterize a novel peptide imaging agent for noninvasive near-infrared fluorescence imaging of protein transport by the lymphatics. An imaging agent consisting of a cyclic albumin-binding domain (cABD) peptide, with sequence, Arg-Leu-Ile-Glu-Asp-Ile-Cys-Leu-Pro-Arg-Trp-Gly-Cys-Leu-Trp-Glu-Asp-Asp-Lys, was conjugated to a near-infrared fluorophore, IRDye800CW, allowing for enhanced vascular uptake, retention, and fluorescence imaging.ProcedureCharacterization of the cABD-IRDye800 peptide conjugate was performed using fluorescence spectroscopy to assess optical properties and SDS-PAGE and Biacore binding assays to determine binding affinity and specificity. Fluorescence imaging of normal C57BL/6 mice was conducted to monitor lymphatic uptake and retention.ResultscABD-IRDye800 exhibited approximately six times greater fluorescent yield and greater stability than indocyanine green, an agent previously used in humans to image lymphatic vasculature. The agent exhibited affinity for albumin with IC50 and Kd in the nanomolar range and demonstrated superior retention characteristics within mouse lymphatics when compared with IRDye800CW.ConclusionscABD-IRDye800 has utility for assessing lymphatic function in mouse models of human lymphatic disease and the potential for use in clinical diagnostic imaging of the lymphatic vasculature.


Nuclear Medicine and Biology | 2015

Comparison of DOTA and NODAGA as chelators for 64Cu-labeled immunoconjugates

Sukhen C. Ghosh; Kenneth L. Pinkston; Holly Robinson; Barrett R. Harvey; Nathaniel Wilganowski; Karen Gore; Eva M. Sevick-Muraca; Ali Azhdarinia

INTRODUCTION Bifunctional chelators have been shown to impact the biodistribution of monoclonal antibody (mAb)-based imaging agents. Recently, radiolabeled 1,4,7-triazacyclononane,1-glutaric acid-4,7-acetic acid (NODAGA)-peptide complexes have demonstrated improved in vivo stability and performance compared to their 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) counterparts. Here, we investigated if similar utility could be achieved with mAbs and compared (64)Cu-labeled DOTA and NODAGA-immunoconjugates for the detection of epithelial cell adhesion molecule (EpCAM) in a prostate cancer model. METHODS DOTA and NODAGA-immunoconjugates of an EpCAM targeting mAb (mAb7) were synthesized and radiolabeled with (64)Cu (DOTA: 40°C for 1hr; NODAGA: 25°C for 1hr). The average number of chelators per mAb was quantified by isotopic dilution, and the biological activity of the immunoconjugates was evaluated by flow cytometry and ELISA. Radioligand assays were performed to compare cellular uptake and determine the dissociation constant (Kd) and maximum number of binding sites (Bmax) for the immunoconjugates using DsRed-transfected PC3-cells. A PC3-DsRed xenograft tumor model was established in nude mice and used to perform biodistribution studies to compare organ uptake and pharmacokinetics. RESULTS (64)Cu-DOTA-mAb7 and (64)Cu-NODAGA-mAb7 were prepared with chelator/protein ratios of 2-3 and obtained in comparable radiochemical yields ranging from 59 to 71%. Similar immunoreactivity was observed with both agents, and mock labeling studies indicated that incubation at room temperature or 40°C did not affect potency. (64)Cu-NODAGA-mAb7 demonstrated higher in vitro cellular uptake while (64)Cu-DOTA-mAb7 had higher Kd and Bmax values. From the biodistribution data, we found similar tumor uptake (13.44±1.21%ID/g and 13.24±4.86%ID/g for (64)Cu-DOTA-mAb7 and (64)Cu-NODAGA-mAb7, respectively) for both agents at 24hr, although normal prostate tissue was significantly lower for (64)Cu-NODAGA-mAb7. (64)Cu-NODAGA-mAb7 also had less accumulation in the liver, suggesting excellent retention of the chelation complex in vivo. This was further confirmed by the higher blood activity of (64)Cu-NODAGA-mAb7, which corresponds to increased bioavailability afforded by the enhanced in vivo stability of the agent. Although tumor/muscle ratios were comparable, tumor/prostate ratios were >2-fold and 1.5-fold higher for (64)Cu-NODAGA-mAb7 at 24 and 48hr, respectively, and suggest better ability to discriminate tumor tissue with (64)Cu-NODAGA-mAb7 in our prostate cancer model. CONCLUSIONS To the best of our knowledge, this study represents the first comparison of (64)Cu-labeled DOTA and NODAGA immunoconjugates in vivo. Our results show favorable in vivo performance for (64)Cu-NODAGA-mAb7 which builds upon previous data on our hybrid mAb7 imaging agent by increasing the detection sensitivity for metastatic prostate tumors, as well as for other types of cancer that express EpCAM.


Journal of Biomedical Optics | 2013

In vivo imaging of orthotopic prostate cancer with far-red gene reporter fluorescence tomography and in vivo and ex vivo validation

Yujie Lu; Chinmay Darne; I-Chih Tan; Grace Wu; Nathaniel Wilganowski; Holly Robinson; Ali Azhdarinia; Banghe Zhu; John C. Rasmussen; Eva M. Sevick-Muraca

Abstract. Fluorescence gene reporters have recently become available for excitation at far-red wavelengths, enabling opportunities for small animal in vivo gene reporter fluorescence tomography (GRFT). We employed multiple projections of the far-red fluorescence gene reporters IFP1.4 and iRFP, excited by a point source in transillumination geometry in order to reconstruct the location of orthotopically implanted human prostate cancer (PC3), which stably expresses the reporter. Reconstruction was performed using a linear radiative-transfer-based regularization-free tomographic method. Positron emission tomography (PET) imaging of a radiolabeled antibody-based agent that targeted epithelial cell adhesion molecule overexpressed on PC3 cells was used to confirm in vivo GRFT results. Validation of GRFT results was also conducted from ex vivo fluorescence imaging of resected prostate tumor. In addition, in mice with large primary prostate tumors, a combination of GRFT and PET showed that the radiolabeled antibody did not penetrate the tumor, consistent with known tumor transport limitations of large (∼150  kDa) molecules. These results represent the first tomography of a living animal using far-red gene reporters.


Molecular Imaging and Biology | 2015

Deglycosylation of mAb by EndoS for Improved Molecular Imaging

Peng Gao; Kenneth L. Pinkston; Nathaniel Wilganowski; Holly Robinson; Ali Azhdarinia; Banghe Zhu; Eva M. Sevick-Muraca; Barrett R. Harvey

PurposeMonoclonal antibodies (mAbs) have been shown preclinically as reliable targeting moieties for antigen imaging using near-infrared fluorescence (NIRF) molecular imaging. However, crystallizable fragment-gamma receptor (FcγRs) expressed on immune cells also bind mAbs through defined epitopes on the constant fragment (Fc) of IgG. Herein, we evaluate the potential impact Fc interactions have on mAb agent imaging specificity.ProcedureThrough the removal of conserved glycans within the Fc domain, shown to have Fc/FcγR interactions, we evaluate their impact on non-specific binding/accumulation of a NIRF-labeled mAb-based imaging agent in lymph nodes (LNs) in inflamed animals and in an orthotopic prostate cancer animal model of LN metastasis.ResultsDeglycosylation of a murine mAb against the human epithelial cell adhesion marker using endoglycosidase EndoS significantly reduced non-specific binding in the LNs of inflamed animals and in cancer-negative LNs of tumor-bearing animals. Sensitivity remained unchanged while improvement in imaging specificity increased imaging accuracy.ConclusionThe reduction of non-specific binding through deglycosylation of a mAb-based imaging agent shows that reducing Fc/FcγR interactions can improve imaging accuracy.

Collaboration


Dive into the Nathaniel Wilganowski's collaboration.

Top Co-Authors

Avatar

Ali Azhdarinia

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Eva M. Sevick-Muraca

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Holly Robinson

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Barrett R. Harvey

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Banghe Zhu

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Pradip Ghosh

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Kenneth L. Pinkston

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Mary A. Hall

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Sukhen C. Ghosh

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Eva Sevick

University of Texas Health Science Center at Houston

View shared research outputs
Researchain Logo
Decentralizing Knowledge