Network


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

Hotspot


Dive into the research topics where Chi Soo Kang is active.

Publication


Featured researches published by Chi Soo Kang.


Bioconjugate Chemistry | 2011

Efficient Bifunctional Decadentate Ligand 3p-C-DEPA for Targeted α-Radioimmunotherapy Applications

Hyun A. Song; Chi Soo Kang; Kwamena E. Baidoo; Diane E. Milenic; Yunwei Chen; Anzhi Dai; Martin W. Brechbiel; Hyun-Soon Chong

A new bifunctional ligand 3p-C-DEPA was synthesized and evaluated for use in targeted α-radioimmunotherapy. 3p-C-DEPA was efficiently prepared via regiospecific ring opening of an aziridinium ion and conjugated with trastuzumab. The 3p-C-DEPA-trastuzumab conjugate was extremely rapid in binding (205/6)Bi, and the corresponding (205/6)Bi-3p-C-DEPA-trastuzumab complex was stable in human serum. Biodistribution studies were performed to evaluate in vivo stability and tumor targeting of (205/6)Bi-3p-C-DEPA-trastuzumab conjugate in tumor bearing athymic mice. (205/6)Bi-3p-C-DEPA-trastuzumab conjugate displayed excellent in vivo stability and targeting as evidenced by low organ uptake and high tumor uptake. The results of the in vitro and in vivo studies indicate that 3p-C-DEPA is a promising chelator for radioimmunotherapy of (212)Bi and (213)Bi.


Nuclear Medicine and Biology | 2012

Copper-64 radiolabeling and biological evaluation of bifunctional chelators for radiopharmaceutical development.

Ravindra A. De Silva; Sandeep Jain; Kimberly A. Lears; Hyun Soon Chong; Chi Soo Kang; Xiang Sun; Buck E. Rogers

INTRODUCTION The development of novel bifunctional chelates for attaching copper-64 to biomolecules has been an active area of research for several years. However, many of these (64)Cu-chelates have poor in vivo stability or harsh radiolabeling conditions. METHODS In this study, two triazacyclononane analogs; C-NE3TA (4-carboxymethyl-7-[2-(carboxymethyl-amino)-3-(4-nitro-phenyl)-propyl]-[1,4,7]triazo-nan-1-yl-acetic acid) and N-NE3TA (4-carboxymethyl-7-[2-[carboxymethyl-(4-nitro-benzyl)-amino]-ethyl]-[1,4,7]triazonan-1-yl-acetic acid) were evaluated for their labeling efficiency with (64)Cu at room temperature and evaluated in vitro and in vivo. In vitro studies included complexation kinetics with Cu(II) using a spectrophotometric method and rat serum stability, while the in vivo biodistribution was evaluated using SCID mice. RESULTS C-NE3TA and N-NE3TA were labeled at >95% efficiency up to ~3.4Ci/μmol. Both C-NE3TA and N-NE3TA formed complexes with Cu(II) almost immediately, with the Cu(II) complexation by C-NE3TA being faster than the formation of Cu(II)-N-NE3TA. Both (64)Cu-N-NE3TA and (64)Cu-C-NE3TA were 96.1% and 90.5% intact after 48h incubation in rat serum, respectively. This is compared to (64)Cu complexes of the control chelators, p-NH(2)-Bn-DOTA and p-NH(2)-Bn-NOTA, with 93.9% and 97.9% retention of (64)Cu in the complex, respectively. In vivo evaluation of (64)Cu-N-NE3TA and (64)Cu-C-NE3TA demonstrates good clearance from normal tissues except for the liver, where 59% and 51% of the radioactivity is retained at 24h compared to 1h for (64)Cu-N-NE3TA and (64)Cu-C-NE3TA, respectively. This compares to 78% and 3% retention for (64)Cu-p-NH(2)-Bn-DOTA and (64)Cu-p-NH(2)-Bn-NOTA. CONCLUSIONS These studies demonstrate that while N-NE3TA and C-NE3TA appear to be superior chelators for (64)Cu than p-NH(2)-Bn-DOTA, they are not better than p-NH(2)-Bn-NOTA. Nevertheless, it may still be interesting to evaluate these chelators after conjugation to biomolecules.


Nuclear Medicine and Biology | 2013

Preclinical evaluation of NETA-based bifunctional ligand for radioimmunotherapy applications using 212Bi and 213Bi: Radiolabeling, serum stability, and biodistribution and tumor uptake studies

Chi Soo Kang; Hyun A. Song; Diane E. Milenic; Kwamena E. Baidoo; Martin W. Brechbiel; Hyun-Soon Chong

INTRODUCTION Despite the great potential of targeted α-radioimmunotherapy (RIT) as demonstrated by pre-clinical and clinical trials, limited progress has been made on the improvement of chelation chemistry for (212)Bi and (213)Bi. A new bifunctional ligand 3p-C-NETA was evaluated for targeted α RIT using (212)Bi and (213)Bi. METHODS Radiolabeling of 3p-C-NETA with (205/6)Bi, a surrogate of (212)Bi and (213)Bi, was evaluated at pH5.5 and room temperature. In vitro stability of the (205/6)Bi-3p-C-NETA-trastuzumab conjugate was evaluated using human serum (pH7, 37 °C). Immunoreactivity and specific activity of the (205/6)Bi-3p-C-NETA-trastuzumab conjugate were measured. An in vivo biodistribution study was performed to evaluate the in vivo stability and tumor targeting properties of the (205/6)Bi-3p-C-NETA-trastuzumab conjugate in athymic mice bearing subcutaneous LS174T tumor xenografts. RESULT The 3p-C-NETA-trastuzumab conjugate was extremely rapid in complexing with (205/6)Bi, and the corresponding (205/6)Bi-3p-C-NETA-trastuzumab was stable in human serum. (205/6)Bi-3p-C-NETA-trastuzumab was prepared with a high specific activity and retained immunoreactivity. (205/6)Bi-3p-C-NETA-trastuzumab conjugate displayed excellent in vivo stability and targeting as evidenced by low normal organ and high tumor uptake. CONCLUSION The results of the in vitro and in vivo studies indicate that 3p-C-NETA is a promising chelator for RIT applications using (212)Bi and (213)Bi. Further detailed in vivo evaluations of 3p-C-NETA for targeted α RIT are warranted.


Nuclear Medicine and Biology | 2015

Synthesis and evaluation of a new bifunctional NETA chelate for molecular targeted radiotherapy using90Y or177Lu

Chi Soo Kang; Yunwei Chen; Hyunbeom Lee; Dijie Liu; Xiang Sun; Junghun Kweon; Michael R. Lewis; Hyun-Soon Chong

INTRODUCTION Therapeutic potential of β-emitting cytotoxic radionuclides (90)Y and (177)Lu has been demonstrated in numerous preclinical and clinical trials. A bifunctional chelate that can effectively complex with the radioisotopes is a critical component for molecular targeted radiotherapy (90)Y and (177)Lu. A new bifunctional chelate 5p-C-NETA with a relatively long alkyl spacer between the chelating backbone and the functional unit for conjugation to a tumor targeting moiety was synthesized. 5p-C-NETA was conjugated to a model targeting moiety, a cyclic Arg-Gly-Asp-D-Tyr-Lys (RGDyK) peptide binding integrin αvβ3 protein overexpressed on various cancers. 5p-C-NETA was conjugated to c(RGDyK) peptide and evaluated for potential use in molecular targeted radiotherapy of (90)Y and (177)Lu. METHODS 5p-C-NETA conjugated with c(RGDyK) was evaluated in vitro for radiolabeling, serum stability, binding affinity, and the result of the in vitro studies of 5p-C-NETA-c(RGDyK) was compared to that of 3p-C-NETA-c(RGDyK). (177)Lu-5p-C-NETA-c(RGDyK) was further evaluated for in vivo biodistribution using gliobastoma bearing mice. RESULT The new chelate rapidly and tightly bound to a cytotoxic radioisotope for cancer therapy, (90)Y or (177)Lu with excellent radiolabeling efficiency and maximum specific activity under mild condition (>99%, RT, <1 min). (90)Y- and (177)Lu-radiolabeled complexes of the new chelator remained stable in human serum without any loss of the radiolanthanide for 14 days. Introduction of the tumor targeting RGD moiety to the new chelator made little impact on complexation kinetics and stability with (90)Y or (177)Lu. (177)Lu-radiolabeled 5p-C-NETA-c(RGDyK) conjugate was shown to target tumors in mice and produced a favorable in vivo stability profile. CONCLUSION The results of in vitro and in vivo evaluation suggest that 5p-C-NETA is an effective bifunctional chelate of (90)Y and (177)Lu that can be applied for generation of versatile molecular targeted radiopharmaceuticals.


Bioorganic & Medicinal Chemistry Letters | 2011

Synthesis and evaluation of a bifunctional chelate for development of Bi(III)-labeled radioimmunoconjugates

Mamta Dadwal; Chi Soo Kang; Hyun A. Song; Xiang Sun; Anzhi Dai; Kwamena E. Baidoo; Martin W. Brechbiel; Hyun-Soon Chong

A new bifunctional ligand C-DEPA was designed and synthesized as a component for antibody-targeted radiation therapy (radioimmunotherapy, RIT) of cancer. C-DEPA was conjugated to a tumor targeting antibody, trastuzumab, and the corresponding C-DEPA-trastuzumab conjugate was evaluated for radiolabeling kinetics with (205/6)Bi. C-DEPA-trastuzumab conjugate rapidly bound (205/6)Bi, and (205/6)Bi-C-DEPA-trastuzumab conjugate was stable in human serum for 72 h. The in vitro radiolabeling kinetics and serum stability data suggest that C-DEPA is a potential chelate for preclinical RIT applications using (212)Bi and (213)Bi.


Bioorganic & Medicinal Chemistry | 2015

Synthesis and comparative biological evaluation of bifunctional ligands for radiotherapy applications of 90Y and 177Lu

Hyun-Soon Chong; Xiang Sun; Yunwei Chen; Inseok Sin; Chi Soo Kang; Michael R. Lewis; Dijie Liu; Varyanna C. Ruthengael; Yongliang Zhong; Ningjie Wu; Hyun A. Song

Zevalin® is an antibody-drug conjugate radiolabeled with a cytotoxic radioisotope ((90)Y) that was approved for radioimmunotherapy (RIT) of B-cell non-Hodgkins lymphoma. A bifunctional ligand that displays favorable complexation kinetics and in vivo stability is required for effective RIT. New bifunctional ligands 3p-C-DE4TA and 3p-C-NE3TA for potential use in RIT were efficiently prepared by the synthetic route based on regiospecific ring opening of aziridinium ions with prealkylated triaza- or tetraaza-backboned macrocycles. The new bifunctional ligands 3p-C-DE4TA and 3p-C-NE3TA along with the known bimodal ligands 3p-C-NETA and 3p-C-DEPA were comparatively evaluated for potential use in targeted radiotherapy using β-emitting radionuclides (90)Y and (177)Lu. The bifunctional ligands were evaluated for radiolabeling kinetics with (90)Y and (177)Lu, and the corresponding (90)Y or (177)Lu-radiolabeled complexes were studied for in vitro stability in human serum and in vivo biodistribution in mice. The results of the comparative complexation kinetic and stability studies indicate that size of macrocyclic cavity, ligand denticity, and bimodality of donor groups have a substantial impact on complexation of the bifunctional ligands with the radiolanthanides. The new promising bifunctional chelates in the DE4TA and NE3TA series were rapid in binding (90)Y and (177)Lu, and the corresponding (90)Y- and (177)Lu-radiolabeled complexes remained inert in human serum or in mice. The in vitro and in vivo data show that 3p-C-DE4TA and 3p-C-NE3TA are promising bifunctional ligands for targeted radiotherapy applications of (90)Y and (177)Lu.


Bioorganic & Medicinal Chemistry | 2014

Novel hexadentate and pentadentate chelators for 64Cu-based targeted PET imaging

Inseok Sin; Chi Soo Kang; Nilantha Bandara; Xiang Sun; Yongliang Zhong; Buck E. Rogers; Hyun-Soon Chong

A series of new hexadentate and pentadentate chelators were designed and synthesized as chelators of (64)Cu. The new pentadentate and hexadentate chelators contain different types of donor groups and are expected to form neutral complexes with Cu(II). The new chelators were evaluated for complex kinetics and stability with (64)Cu. The new chelators instantly bound to (64)Cu with high labeling efficiency and maximum specific activity. All (64)Cu-radiolabeled complexes in human serum remained intact for 2 days. The (64)Cu-radiolabeled complexes were further challenged by EDTA in a 100-fold molar excess. Among the (64)Cu-radiolabeled complexes evaluated, (64)Cu-complex of the new chelator E was well tolerated with a minimal transfer of (64)Cu to EDTA. (64)Cu-radiolabeled complex of the new chelator E was further evaluated for biodistribution studies using mice and displayed rapid blood clearance and low organ uptake. (64)Cu-chelator E produced a favorable in vitro and in vivo complex stability profiles comparable to (64)Cu complex of the known hexadentate NOTA chelator. The in vitro and in vivo data highlight strong potential of the new chelator E for targeted PET imaging application.


ChemMedChem | 2016

Theranostic Polyaminocarboxylate–Cyanine–Transferrin Conjugate for Anticancer Therapy and Near‐Infrared Optical Imaging

Chi Soo Kang; Siyuan Ren; Xiang Sun; Hyun-Soon Chong

Iron chelation therapy has been recognized as a promising antitumor therapeutic strategy. Herein we report a novel theranostic agent for targeted iron chelation therapy and near‐infrared (NIR) optical imaging of cancers. The theranostic agent was prepared by incorporation of a polyaminocarboxylate‐based cytotoxic chelating agent (N‐NE3TA; 7‐[2‐[(carboxymethyl)amino]ethyl]‐1,4,7‐triazacyclononane‐1,4‐diacetic acid) and a NIR fluorescent cyanine dye (Cy5.5) onto a tumor‐targeting transferrin (Tf). The N‐NE3TA–Tf conjugate (without Cy5.5) was characterized and evaluated for antiproliferative activity in HeLa, HT29, and PC3 cancer cells, which have elevated expression levels of the transferrin receptor (TfR). The N‐NE3TA–Tf conjugate displayed significant inhibitory activity against all three cancer cell lines. The NIR dye Cy5.5 was then incorporated into N‐NE3TA–Tf, and the resulting cytotoxic and fluorescent transferrin conjugate N‐NE3TA–Tf–Cy5.5 was shown by microscopy to enter TfR‐overexpressing cancer cells. This theranostic conjugate has potential application for dual use in targeted iron chelation cancer therapy and NIR fluorescence imaging.


Bioorganic & Medicinal Chemistry Letters | 2015

Novel 64Cu-radiolabeled bile acid conjugates for targeted PET imaging

Hyun-Soon Chong; Yunwei Chen; Chi Soo Kang; Xiang Sun; Ningjie Wu

A promising bifunctional chelate (N-NE3TA) was conjugated to bile acids, cholic acid (CA), deoxycholic acid (DCA), and chenodeoxycholic acid (CDCA) as tumor targeting vectors. Bile acid conjugates of N-NE3TA (CA-N-NE3TA, DCA-N-NE3TA, and CDCA-N-NE3TA) were comparatively evaluated for complexation with (64)Cu, an imaging probe for positron emission tomography (PET). N-NE3TA-bile acid conjugates were evaluated for radiolabeling kinetics with (64)Cu, and the corresponding (64)Cu-radiolabeled conjugates were screened for complex stability in human serum and EDTA solution. The NE3TA-bile acid conjugates instantly bound to (64)Cu with excellent radiolabeling efficiency at room temperature. All NE3TA-bile acid conjugates radiolabeled with (64)Cu remained inert in human serum for 2days without releasing a considerable amount of the radioactivity. The (64)Cu-radiolabeled complexes were further challenged by EDTA in a 100-fold molar excess. Bile acid-N-NE3TA conjugates radiolabeled with (64)Cu were quite stable with a minimal transfer of (64)Cu to EDTA at 4h time point. The in vitro data indicate that the bile acid-N-NE3TA conjugates deserve further biological evaluation for (64)Cu-based targeted PET imaging applications.


Journal of Inorganic Biochemistry | 2016

Transferrin conjugates of triazacyclononane-based bifunctional NE3TA chelates for PET imaging: Synthesis, Cu-64 radiolabeling, and in vitro and in vivo evaluation.

Chi Soo Kang; Ningjie Wu; Yunwei Chen; Xiang Sun; Nilantha Bandara; Dijie Liu; Michael R. Lewis; Buck E. Rogers; Hyun-Soon Chong

Three different polyaminocarboxylate-based bifunctional NE3TA (7-[2-[carboxymethyl)amino]ethyl]-1,4,7-triazacyclononane-1,4-diacetic acid) chelating agents were synthesized for potential use in copper 64-PET imaging applications. The bifunctional chelates were comparatively evaluated using transferrin (Tf) as a model targeting vector that binds to the transferrin receptor overexpressed in many different cancer cells. The transferrin conjugates of the NE3TA-based bifunctional chelates were evaluated for radiolabeling with (64)Cu. In vitro stability and cellular uptake of (64)Cu-radiolabeled conjugates were evaluated in human serum and prostate (PC-3) cancer cells, respectively. Among the three NE3TA-Tf conjugates tested, N-NE3TA-Tf was identified as the best conjugate for radiolabeling with (64)Cu. N-NE3TA-Tf rapidly bound to (64)Cu (>98% radiolabeling efficiency, 1min, RT), and (64)Cu-N-NE3TA-Tf remained stable in human serum for 2days and demonstrated high uptake in PC-3 cancer cells. (64)Cu-N-NE3TA-Tf was shown to have rapid blood clearance and increasing tumor uptake in PC-3 tumor bearing mice over a 24h period. This bifunctional chelate presents highly efficient chelation chemistry with (64)Cu under mild condition that can be applied for radiolabeling of various tumor-specific biomolecules with (64)Cu for potential use in PET imaging applications.

Collaboration


Dive into the Chi Soo Kang's collaboration.

Top Co-Authors

Avatar

Hyun-Soon Chong

Illinois Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Xiang Sun

Illinois Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Yunwei Chen

Illinois Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Hyun A. Song

Illinois Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dijie Liu

University of Missouri

View shared research outputs
Top Co-Authors

Avatar

Ningjie Wu

Illinois Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Inseok Sin

Illinois Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Anzhi Dai

Illinois Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Buck E. Rogers

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge