Network


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

Hotspot


Dive into the research topics where Chan Hyuk Kim is active.

Publication


Featured researches published by Chan Hyuk Kim.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Synthesis of site-specific antibody-drug conjugates using unnatural amino acids

Jun Y. Axup; Krishna M. Bajjuri; Melissa Ritland; Benjamin M. Hutchins; Chan Hyuk Kim; Stephanie A. Kazane; Rajkumar Halder; Jane S. Forsyth; Antonio F. Santidrian; Karin Stafin; Yingchun Lu; Hon Tran; Aaron J. Seller; Sandra L. Biroc; Jason Pinkstaff; Feng Tian; Subhash C. Sinha; Vaughn V. Smider; Peter G. Schultz

Antibody-drug conjugates (ADCs) allow selective targeting of cytotoxic drugs to cancer cells presenting tumor-associated surface markers, thereby minimizing systemic toxicity. Traditionally, the drug is conjugated nonselectively to cysteine or lysine residues in the antibody. However, these strategies often lead to heterogeneous products, which make optimization of the biological, physical, and pharmacological properties of an ADC challenging. Here we demonstrate the use of genetically encoded unnatural amino acids with orthogonal chemical reactivity to synthesize homogeneous ADCs with precise control of conjugation site and stoichiometry. p-Acetylphenylalanine was site-specifically incorporated into an anti-Her2 antibody Fab fragment and full-length IgG in Escherichia coli and mammalian cells, respectively. The mutant protein was selectively and efficiently conjugated to an auristatin derivative through a stable oxime linkage. The resulting conjugates demonstrated excellent pharmacokinetics, potent in vitro cytotoxic activity against Her2+ cancer cells, and complete tumor regression in rodent xenograft treatment models. The synthesis and characterization of homogeneous ADCs with medicinal chemistry-like control over macromolecular structure should facilitate the optimization of ADCs for a host of therapeutic uses.


Current Opinion in Chemical Biology | 2013

Protein conjugation with genetically encoded unnatural amino acids

Chan Hyuk Kim; Jun Y. Axup; Peter G. Schultz

The site-specific incorporation of unnatural amino acids with orthogonal chemical reactivity into proteins enables the synthesis of structurally defined protein conjugates. Amino acids containing ketone, azide, alkyne, alkene, and tetrazine side chains can be genetically encoded in response to nonsense and frameshift codons. These bio-orthogonal chemical handles allow precise control over the site and stoichiometry of conjugation, and have enabled medicinal chemistry-like optimization of the physical and biological properties of protein conjugates, especially the next-generation protein therapeutics.


Journal of the American Chemical Society | 2012

Synthesis of bispecific antibodies using genetically encoded unnatural amino acids.

Chan Hyuk Kim; Jun Y. Axup; Anna Dubrovska; Stephanie A. Kazane; Benjamin A. Hutchins; Erik D. Wold; Vaughn V. Smider; Peter G. Schultz

Bispecific antibodies were constructed using genetically encoded unnatural amino acids with orthogonal chemical reactivity. A two-step process afforded homogeneous products in excellent yield. Using this approach, we synthesized an anti-HER2/anti-CD3 bispecific antibody, which efficiently cross-linked HER2+ cells and CD3+ cells. In vitro effector-cell mediated cytotoxicity was observed at picomolar concentrations.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Switch-mediated activation and retargeting of CAR-T cells for B-cell malignancies.

David T. Rodgers; Magdalena Mazagova; Eric Hampton; Yu Cao; Nitya S. Ramadoss; Ian R. Hardy; Andrew Schulman; Juanjuan Du; Feng Wang; Oded Singer; Vanessa Núñez; Jiayin Shen; Ashley K. Woods; Timothy M. Wright; Peter G. Schultz; Chan Hyuk Kim; Travis S. Young

Significance Chimeric antigen receptor T (CAR-T) cell therapy has produced promising results in clinical trials but has been challenged by the inability to control engineered cells once infused into the patient. Here we present a generalizable method of controlling CAR-T cells using peptide-engrafted antibody-based molecular switches that act as a bridge between the target cell and CAR-T cell. We show that switches specific for CD19 govern the activity, tissue-homing, cytokine release, and phenotype of switchable CAR-T cells in a dose-titratable manner using xenograft mouse models of B-cell leukemia. We expect that this method of tuning CAR-T cell responses will provide improved safety and versatility of CAR–T-cell therapy in the clinic. Chimeric antigen receptor T (CAR-T) cell therapy has produced impressive results in clinical trials for B-cell malignancies. However, safety concerns related to the inability to control CAR-T cells once infused into the patient remain a significant challenge. Here we report the engineering of recombinant antibody-based bifunctional switches that consist of a tumor antigen-specific Fab molecule engrafted with a peptide neo-epitope, which is bound exclusively by a peptide-specific switchable CAR-T cell (sCAR-T). The switch redirects the activity of the bio-orthogonal sCAR-T cells through the selective formation of immunological synapses, in which the sCAR-T cell, switch, and target cell interact in a structurally defined and temporally controlled manner. Optimized switches specific for CD19 controlled the activity, tissue-homing, cytokine release, and phenotype of sCAR-T cells in a dose-titratable manner in a Nalm-6 xenograft rodent model of B-cell leukemia. The sCAR–T-cell dosing regimen could be tuned to provide efficacy comparable to the corresponding conventional CART-19, but with lower cytokine levels, thereby offering a method of mitigating cytokine release syndrome in clinical translation. Furthermore, we demonstrate that this methodology is readily adaptable to targeting CD20 on cancer cells using the same sCAR-T cell, suggesting that this approach may be broadly applicable to heterogeneous and resistant tumor populations, as well as other liquid and solid tumor antigens.


Angewandte Chemie | 2012

Site‐Specific Incorporation of ε‐N‐Crotonyllysine into Histones

Chan Hyuk Kim; Mingchao Kang; Hak Joong Kim; Abhishek Chatterjee; Peter G. Schultz

A novel post-translationally modified amino acid, crotonyllysine (Kcr), was genetically incorporated into proteins in bacterial and mammalian cells using an evolved pyrrolysyl-tRNA/synthetase-tRNA pair. The ability to produce histones with homogenous, site-specific Kcr modifications will be valuable in elucidating the biological role of this recently identified post-translational modification.


Journal of the American Chemical Society | 2013

Self-Assembled Antibody Multimers through Peptide Nucleic Acid Conjugation

Stephanie A. Kazane; Jun Y. Axup; Chan Hyuk Kim; Mihai Ciobanu; Erik D. Wold; Sofia Barluenga; Benjamin A. Hutchins; Peter G. Schultz; Nicolas Winssinger; Vaughn V. Smider

With the recent clinical success of bispecific antibodies, a strategy to rapidly synthesize and evaluate bispecific or higher order multispecific molecules could facilitate the discovery of new therapeutic agents. Here, we show that unnatural amino acids (UAAs) with orthogonal chemical reactivity can be used to generate site-specific antibody-oligonucleotide conjugates. These constructs can then be self-assembled into multimeric complexes with defined composition, valency, and geometry. With this approach, we generated potent bispecific antibodies that recruit cytotoxic T lymphocytes to Her2 and CD20 positive cancer cells, as well as multimeric antibody fragments with enhanced activity. This strategy should accelerate the synthesis and in vitro characterization of antibody constructs with unique specificities and molecular architectures.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Versatile strategy for controlling the specificity and activity of engineered T cells

Ji Young Kim; Stephanie A. Kazane; Seihyun Choi; Hwa Young Yun; Minsoo Kim; David T. Rodgers; Holly Pugh; Oded Singer; Sophie B. Sun; Bryan R. Fonslow; James N. Kochenderfer; Timothy M. Wright; Peter G. Schultz; Travis S. Young; Chan Hyuk Kim; Yu Cao

Significance Despite the unprecedented antileukemic response demonstrated in recent clinical trials, the inability to control the potent chimeric antigen receptor (CAR)—T-cell activity has resulted in several serious adverse incidents. Herein, we demonstrate that a switch-mediated CAR-T approach enables the titration of engineered T-cell antitumor activity, which was observed to be highly advantageous in reducing treatment-related toxicities in vivo. Moreover, we show that the use of optimized antibody-based switches readily enables a single CAR construct to target different antigens, indicating its potential application to treat tumor escape variants and heterogeneous tumors expressing distinct tumor antigens. Our data support the safe application of this potent immune cell-based therapy to target other types of cancer, including solid tumors, as well as nononcology indications. The adoptive transfer of autologous T cells engineered to express a chimeric antigen receptor (CAR) has emerged as a promising cancer therapy. Despite impressive clinical efficacy, the general application of current CAR–T-cell therapy is limited by serious treatment-related toxicities. One approach to improve the safety of CAR-T cells involves making their activation and proliferation dependent upon adaptor molecules that mediate formation of the immunological synapse between the target cancer cell and T-cell. Here, we describe the design and synthesis of structurally defined semisynthetic adaptors we refer to as “switch” molecules, in which anti-CD19 and anti-CD22 antibody fragments are site-specifically modified with FITC using genetically encoded noncanonical amino acids. This approach allows the precise control over the geometry and stoichiometry of complex formation between CD19- or CD22-expressing cancer cells and a “universal” anti-FITC–directed CAR-T cell. Optimization of this CAR–switch combination results in potent, dose-dependent in vivo antitumor activity in xenograft models. The advantage of being able to titrate CAR–T-cell in vivo activity was further evidenced by reduced in vivo toxicity and the elimination of persistent B-cell aplasia in immune-competent mice. The ability to control CAR-T cell and cancer cell interactions using intermediate switch molecules may expand the scope of engineered T-cell therapy to solid tumors, as well as indications beyond cancer therapy.


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

Bispecific small molecule–antibody conjugate targeting prostate cancer

Chan Hyuk Kim; Jun Y. Axup; Brian R. Lawson; Hwayoung Yun; Virginie Tardif; Sei hyun Choi; Quan Zhou; Anna Dubrovska; Sandra L. Biroc; Robin Marsden; Jason Pinstaff; Vaughn V. Smider; Peter G. Schultz

Significance We have developed a semisynthetic method for the production of bispecific antibody-like therapeutics consisting of a small molecule targeting moiety conjugated to an antibody. A highly selective prostate-specific membrane antigen-binding ligand was site specifically conjugated to a mutant α cluster of differentiation 3 (αCD3) Fab containing an unnatural amino acid with orthogonal chemical reactivity. The optimized conjugate showed potent in vitro activity, good serum half-life, and potent in vivo activity in prostate cancer xenograft mouse models. This semisynthetic approach is likely to be applicable to the generation of additional bispecific agents using drug-like ligands selective for other cell-surface receptors. Bispecific antibodies, which simultaneously target CD3 on T cells and tumor-associated antigens to recruit cytotoxic T cells to cancer cells, are a promising new approach to the treatment of hormone-refractory prostate cancer. Here we report a site-specific, semisynthetic method for the production of bispecific antibody-like therapeutics in which a derivative of the prostate-specific membrane antigen-binding small molecule DUPA was selectively conjugated to a mutant αCD3 Fab containing the unnatural amino acid, p-acetylphenylalanine, at a defined site. Homogeneous conjugates were generated in excellent yields and had good solubility. The efficacy of the conjugate was optimized by modifying the linker structure, relative binding orientation, and stoichiometry of the ligand. The optimized conjugate showed potent and selective in vitro activity (EC50 ∼100 pM), good serum half-life, and potent in vivo activity in prophylactic and treatment xenograft mouse models. This semisynthetic approach is likely to be applicable to the generation of additional bispecific agents using drug-like ligands selective for other cell-surface receptors.


Journal of the American Chemical Society | 2015

An immunosuppressive antibody-drug conjugate.

Rongsheng E. Wang; Tao Liu; Ying Wang; Yu Cao; Jintang Du; Xiaozhou Luo; Vishal Deshmukh; Chan Hyuk Kim; Brian R. Lawson; Matthew S. Tremblay; Travis S. Young; Stephanie A. Kazane; Feng Wang; Peter G. Schultz

We have developed a novel antibody-drug conjugate (ADC) that can selectively deliver the Lck inhibitor dasatinib to human T lymphocytes. This ADC is based on a humanized antibody that selectively binds with high affinity to CXCR4, an antigen that is selectively expressed on hematopoietic cells. The resulting dasatinib-antibody conjugate suppresses T-cell-receptor (TCR)-mediated T-cell activation and cytokine expression with low nM EC50 and has minimal effects on cell viability. This ADC may lead to a new class of selective immunosuppressive drugs with improved safety and extend the ADC strategy to the targeted delivery of kinase inhibitors for indications beyond oncology.


Angewandte Chemie | 2014

Targeting Human C‐Type Lectin‐like Molecule‐1 (CLL1) with a Bispecific Antibody for Immunotherapy of Acute Myeloid Leukemia

Hua Lu; Quan Zhou; Vishal Deshmukh; Hardeep Phull; Virginie Tardif; Rahul R. Naik; Claire Bouvard; Yong Zhang; Seihyun Choi; Brian R. Lawson; Shoutian Zhu; Chan Hyuk Kim; Peter G. Schultz

Acute myeloid leukemia (AML), which is the most common acute adult leukemia and the second most common pediatric leukemia, still has a poor prognosis. Human C-type lectin-like molecule-1 (CLL1) is a recently identified myeloid lineage restricted cell surface marker, which is overexpressed in over 90% of AML patient myeloid blasts and in leukemic stem cells. Here, we describe the synthesis of a novel bispecific antibody, αCLL1-αCD3, using the genetically encoded unnatural amino acid, p-acetylphenylalanine. The resulting αCLL1-αCD3 recruits cytotoxic T cells to CLL1 positive cells, and demonstrates potent and selective cytotoxicity against several human AML cell lines and primary AML patient derived cells in vitro. Moreover, αCLL1-αCD3 treatment completely eliminates established tumors in an U937 AML cell line xenograft model. These results validate the clinical potential of CLL1 as an AML-specific antigen for the generation of a novel immunotherapeutic for AML.

Collaboration


Dive into the Chan Hyuk Kim's collaboration.

Top Co-Authors

Avatar

Peter G. Schultz

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yu Cao

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jun Y. Axup

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Vaughn V. Smider

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Seihyun Choi

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brian R. Lawson

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Erik D. Wold

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Feng Wang

Chinese Academy of Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge