Network


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

Hotspot


Dive into the research topics where Sunkuk Kwon is active.

Publication


Featured researches published by Sunkuk Kwon.


Journal of Biomedical Optics | 2007

Comparison of visible and near-infrared wavelength-excitable fluorescent dyes for molecular imaging of cancer

Kristen E. Adams; Shi Ke; Sunkuk Kwon; Feng Liang; Zhen Fan; Yang Lu; Karen K. Hirschi; Michel E. Mawad; Michael A. Barry; Eva M. Sevick-Muraca

Targeted fluorescent molecular imaging probes may provide an optimal means of detecting disease. Stable, organic fluorophores can be repeatedly excited in vivo by propagated light and consequentially can provide large signal-to-noise ratios (SNRs) for image detection of target tissues. In the literature, many small animal imaging studies are performed with a red excitable dye, Cy5.5, conjugated to the targeting component. We report the comparison of the in vivo fluorescent imaging performance of a near-IR (NIR) and a red-excitable dye. Epidermal growth factor (EGF) was conjugated with Cy5.5 [excitation/emission (ex/em), 660710 nm] or IRDye 800CW (ex/em: 785830 nm) for imaging EGF receptor (EGFr) positive (MDA-MB-468) and/or negative (MDA-MB-435) human breast cancer cell lines in subcutaneous xenograft models. The conjugates were injected intravenously at 1-nmol-dye equivalent with and without anti-EGFr monoclonal antibody C225, preadministered 24 h prior as a competitive ligand to EGFr. Our images show that while both agents target EGFr, the EGF-IRDye 800CW evidenced a significantly reduced background and enhanced the tumor-to-background ratio (TBR) compared to the EGF-Cy5.5. Immunohistochemistry shows that EGF causes activation of the EGFr signaling pathway, suggesting that prior to use as a targeting, diagnostic agent, potential deleterious effects should be considered.


The Journal of Nuclear Medicine | 2007

Dual-Labeled Trastuzumab-Based Imaging Agent for the Detection of Human Epidermal Growth Factor Receptor 2 Overexpression in Breast Cancer

Lakshmi Sampath; Sunkuk Kwon; Shi Ke; Wei Wang; Rachel Schiff; Michel E. Mawad; Eva M. Sevick-Muraca

Overexpression of the human epidermal growth factor receptor (HER) family has been implicated in cancer because of its participation in signaling pathways regulating cellular proliferation, differentiation, motility, and survival. In this work, we exploited the extracellular binding property of trastuzumab, a clinically therapeutic monoclonal antibody to the second member of the HER family (HER2), to design a diagnostic imaging agent, (111In-DTPA)n-trastuzumab-(IRDye 800CW)m, that is dual labeled with 111In, a γ-emitter, and a near-infrared (NIR) fluorescent dye, IRDye 800CW, to detect HER2 overexpression in breast cancer cells. The stoichiometric ratios “n” and “m” refer to the number of diethylenetriaminepentaacetic acid dianhydride (DTPA) and IRDye 800CW molecules bound per trastuzumab molecule, respectively. Methods: Fluorescence microscopy and confocal microscopy were used to determine the molecular specificity of (DTPA)n-trastuzumab-(IRDye800)m in vitro in SKBr3 (HER2-positive) and MDA-MB-231 (HER2-negative) breast cancer cells. SKBr3 cells were incubated with (DTPA)n-trastuzumab-(IRDye800)m or IRDye800CW or pretreated with trastuzumab or human IgG followed by (DTPA)n-trastuzumab-(IRDye800)m and examined under a fluorescence microscope. For in vivo characterization, athymic nude mice bearing HER2-overexpressing SKBr3-luc subcutaneous xenografts were injected intravenously with (111In-DTPA)n-trastuzumab-(IRDye800)m and imaged with SPECT and NIR fluorescence imaging at 48 h. Tumor-bearing mice were also injected intravenously with trastuzumab 24 h before administration of (111In-DTPA)n-trastuzumab-(IRDye800)m. Nonspecific uptake in the SKBr3-luc tumors was analyzed by injecting the mice with IRDye 800CW and (111In-DTPA)p-IgG-(IRDye800)q, where “p” and “q” are the stoichiometric ratios of DTPA and IRDye 800CW bound per IgG antibody, respectively. Results: (DTPA)n-trastuzumab-(IRDye800)m showed significantly greater binding to SKBr3 cells than to MDA-MB-231 cells. Confocal imaging revealed that this binding occurred predominantly around the cell membrane. Competitive binding studies with excess trastuzumab before incubation with (DTPA)n-trastuzumab-(IRDye800)m abolished this binding affinity, but pretreatment with nonspecific IgG did not alter binding. In vivo nuclear and optical imaging of SKBr3-luc xenografts injected with (111In-DTPA)n-trastuzumab-(IRDye800)m revealed significantly more uptake in the tumor region than in the contralateral muscle region. The tumor-to-muscle ratio decreased in mice pretreated with trastuzumab and in mice injected with IRDye 800CW and (111In-DTPA)p-IgG-(IRDye800)q. Ex vivo imaging of dissected organs confirmed these results. Finally, coregistration of histologic hematoxylin–eosin stains with autoradiography signals from tumor and muscle tissue slices indicated that (111In-DTPA)n-trastuzumab-(IRDye800)m bound only in tumor tissue and not to muscle. Conclusion: Dual-labeled (111In-DTPA)n-trastuzumab-(IRDye800)m may be an effective diagnostic biomarker capable of tracking HER2 overexpression in breast cancer patients.


Journal of Immunotherapy | 2009

Virus-like particle (VLP) lymphatic trafficking and immune response generation after immunization by different routes

Rafael Cubas; Sheng Zhang; Sunkuk Kwon; Eva M. Sevick-Muraca; Min Li; Changyi Chen; Qizhi Yao

Virus-like particles (VLPs) have gained increasing interest for their use as vaccines due to their repetitive antigenic structure that is capable of efficiently activating the immune system. The efficacy of VLP immunization may lie in its ability to traffic into draining lymph nodes while activating antigen-presenting cells to initiate the orchestration of signals required for the development of a robust immune response. Currently, there is no comprehensive study showing the correlation of different VLP vaccination routes to immune outcome. In this study, we took an optical imaging approach to directly visualize the trafficking of simian-human immunodeficiency (SHIV) VLPs after immunization by commonly used routes and analyzed the corresponding humoral and cellular immune responses generated. We found that VLPs can easily enter the subcapsular sinus of draining lymph nodes with quantitative differences in the number of lymph node involvement depending on the immunization route used. Intradermal immunization led to the largest level of lymph node involvement for the longest period of time, which correlated with the strongest humoral and cellular immune responses. Flow cytometry analysis from extracted splenocytes showed that intradermal immunization led to the largest population of germinal center and activated B cells, which translated into higher antibody levels and antigen-specific cytotoxic T lymphocyte responses. Our results indicate that VLPs traffic into lymph nodes upon immunization and can be directly visualized by optical imaging techniques. Intradermal immunization showed improved responses and might be a preferable delivery route to use for viral and cancer immunotherapeutic studies involving VLPs.


Journal of Clinical Investigation | 2014

Emerging lymphatic imaging technologies for mouse and man

Eva M. Sevick-Muraca; Sunkuk Kwon; John C. Rasmussen

The lymphatic circulatory system has diverse functions in lipid absorption, fluid homeostasis, and immune surveillance and responds dynamically when presented with infection, inflammation, altered hemodynamics, and cancer. Visualization of these dynamic processes in human disease and animal models of disease is key to understanding the contributory role of the lymphatic circulatory system in disease and to devising effective therapeutic strategies. Longitudinal, non-destructive, and repeated imaging is necessary to expand our understanding of disease progression and regression in basic science and clinical investigations. Herein we summarize recent advances in in vivo lymphatic imaging employing magnetic resonance, computed tomography, lymphoscintigraphy, and emerging optical techniques with respect to their contributory roles in both basic science and clinical research investigations.


Journal of Clinical Investigation | 2012

RASA1 maintains the lymphatic vasculature in a quiescent functional state in mice

Philip E. Lapinski; Sunkuk Kwon; Beth A. Lubeck; John E. Wilkinson; R. Sathish Srinivasan; Eva M. Sevick-Muraca; Philip D. King

RASA1 (also known as p120 RasGAP) is a Ras GTPase-activating protein that functions as a regulator of blood vessel growth in adult mice and humans. In humans, RASA1 mutations cause capillary malformation-arteriovenous malformation (CM-AVM); whether it also functions as a regulator of the lymphatic vasculature is unknown. We investigated this issue using mice in which Rasa1 could be inducibly deleted by administration of tamoxifen. Systemic loss of RASA1 resulted in a lymphatic vessel disorder characterized by extensive lymphatic vessel hyperplasia and leakage and early lethality caused by chylothorax (lymphatic fluid accumulation in the pleural cavity). Lymphatic vessel hyperplasia was a consequence of increased proliferation of lymphatic endothelial cells (LECs) and was also observed in mice in which induced deletion of Rasa1 was restricted to LECs. RASA1-deficient LECs showed evidence of constitutive activation of Ras in situ. Furthermore, in isolated RASA1-deficient LECs, activation of the Ras signaling pathway was prolonged and cellular proliferation was enhanced after ligand binding to different growth factor receptors, including VEGFR-3. Blockade of VEGFR-3 was sufficient to inhibit the development of lymphatic vessel hyperplasia after loss of RASA1 in vivo. These findings reveal a role for RASA1 as a physiological negative regulator of LEC growth that maintains the lymphatic vasculature in a quiescent functional state through its ability to inhibit Ras signal transduction initiated through LEC-expressed growth factor receptors such as VEGFR-3.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Lymphatic abnormalities are associated with RASA1 gene mutations in mouse and man

Patricia E. Burrows; Manuel L. Gonzalez-Garay; John C. Rasmussen; Melissa B. Aldrich; Renie Guilliod; Erik A. Maus; Caroline E. Fife; Sunkuk Kwon; Philip E. Lapinski; Philip D. King; Eva M. Sevick-Muraca

Mutations in gene RASA1 have been historically associated with capillary malformation–arteriovenous malformation, but sporadic reports of lymphatic involvement have yet to be investigated in detail. To investigate the impact of RASA1 mutations in the lymphatic system, we performed investigational near-infrared fluorescence lymphatic imaging and confirmatory radiographic lymphangiography in a Parkes–Weber syndrome (PKWS) patient with suspected RASA1 mutations and correlated the lymphatic abnormalities against that imaged in an inducible Rasa1 knockout mouse. Whole-exome sequencing (WES) analysis and validation by Sanger sequencing of DNA from the patient and unaffected biological parents enabled us to identify an early-frameshift deletion in RASA1 that was shared with the father, who possessed a capillary stain but otherwise no overt disease phenotype. Abnormal lymphatic vasculature was imaged in both affected and unaffected legs of the PKWS subject that transported injected indocyanine green dye to the inguinal lymph node and drained atypically into the abdomen and into dermal lymphocele-like vesicles on the groin. Dermal lymphatic hyperplasia and dilated vessels were observed in Rasa1-deficient mice, with subsequent development of chylous ascites. WES analyses did not identify potential gene modifiers that could explain the variability of penetrance between father and son. Nonetheless, we conclude that the RASA1 mutation is responsible for the aberrant lymphatic architecture and functional abnormalities, as visualized in the PKWS subject and in the animal model. Our unique method to combine investigatory near-infrared fluorescence lymphatic imaging and WES for accurate phenoptyping and unbiased genotyping allows the study of molecular mechanisms of lymphatic involvement of hemovascular disorders.


Journal of Immunological Methods | 2010

Functional lymphatic imaging in tumor-bearing mice

Sunkuk Kwon; Eva M. Sevick-Muraca

The lymphatic system provides a route for dissemination of metastatic cancer cells. Yet to date transient changes in lymphatic drainage pathways and function as a result of tumor growth and metastasis have not been completely elucidated. Herein, we non-invasively imaged functional and architectural lymphatic changes in mice with regional, palpable lymph node (LN) involvement using dynamic near-infrared (NIR) fluorescence imaging with intradermal injection of indocyanine green (ICG) to both tumor-free mice and mice bearing C6/LacZ rat glioma tumors in the tail or hindlimb. We found that lymphatic drainage pathways were transiently altered and the contractile function of regional conducting lymphatic vessels was reduced or lost with progressive disease. Therefore, transient changes in the regional lymphatic architecture and function that occur with progressive disease, can be imaged using NIR fluorescence, and may provide a new method to stage disease.


The Prostate | 2012

Imaging prostate cancer lymph node metastases with a multimodality contrast agent

Mary A. Hall; Sunkuk Kwon; Holly Robinson; Pier Anne Lachance; Ali Azhdarinia; Ranjani Ranganathan; Roger E. Price; Wenyaw Chan; Eva M. Sevick-Muraca

Methods to detect lymph node (LN) metastases in prostate cancer (PCa) are limited. Pelvic LN dissection is commonly performed during prostatectomy, but often followed by morbid complications. More refined methods for detecting LN invasion are needed.


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.


Biomedical Optics Express | 2011

Mouse phenotyping with near-infrared fluorescence lymphatic imaging

Sunkuk Kwon; Eva M. Sevick-Muraca

We demonstrate the ability to non-invasively and quantitatively image lymphatic architecture and contractile function using dynamic near-infrared (NIR) fluorescence imaging with injection of indocyanine green in normal and transgenic mice. Unlike normal mice, which showed well defined lymphatic drainage patterns and orthograde propagation of contraction waves, we observed tortuous and mispatterned lymphatic vessels and persistent retrograde lymph flow in mice with deficiency in Prox1, a transcription factor essential for lymphatic vascular development. NIR fluorescence imaging provides a method for quantifying lymphatic function for future studies designed to discern differences in lymphatic function in murine models of human lymphatic disease.

Collaboration


Dive into the Sunkuk Kwon's collaboration.

Top Co-Authors

Avatar

Eva M. Sevick-Muraca

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

John C. Rasmussen

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Kristen E. Adams

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Shi Ke

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Germaine D. Agollah

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Grace Wu

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

Mary A. Hall

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Melissa B. Aldrich

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Alan R. Davis

Baylor College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge