Network


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

Hotspot


Dive into the research topics where Hok Seon Kim is active.

Publication


Featured researches published by Hok Seon Kim.


Science Translational Medicine | 2011

Amelioration of Type 2 Diabetes by Antibody-Mediated Activation of Fibroblast Growth Factor Receptor 1

Ai-Luen Wu; Ganesh Kolumam; Scott Stawicki; Yongmei Chen; Jun Li; Jose Zavala-Solorio; Khanhky Phamluong; Bo Feng; Li Li; Scot A. Marsters; Lance Kates; Nicholas van Bruggen; Maya Leabman; Anne Wong; David West; Howard M. Stern; Elizabeth Luis; Hok Seon Kim; Daniel G. Yansura; Andrew S. Peterson; Ellen Filvaroff; Yan Wu; Junichiro Sonoda

Antibody-mediated activation of fibroblast growth factor receptor 1 reverses the diabetic phenotype in mice, likely by affecting brown adipose tissues. Getting at Brown Fat It’s fun to indulge in holiday cheer, if only a holiday miracle allowed one to avoid the often-linked weight gain. At the molecular level, obesity and type 2 diabetes can be linked by the fibroblast growth factor (FGF) family of proteins and their receptors (FGFRs), with some factors showing disease-reversing capabilities. For instance, overweight, diabetic mice treated with FGF21 regain normal metabolism and lose weight, even without spending hours on a treadmill. However, attempts to use this fat-burning factor in humans have not been successful, owing to poor pharmacokinetics as well as concerns over negative effects of modified FGF21 proteins. In this issue, Wu and colleagues describe an antibody-based FGF21 mimic that circumvents these limitations to overcome metabolic disease in mice. The authors reasoned that robust drugs that closely mimic FGF21 function would similarly exert antidiabetic effects. Using phage display technology, Wu et al. identified monoclonal antibodies (R1MAbs) that were specifically targeted tissues that play key roles in diabetes and obesity, including adipose (fat) tissue. In contrast to FGF21, which binds several forms of the FGFR throughout the body, the phage-derived R1MAbs bound only to FGFR1—a receptor present in the pancreas and in brown and white adipose tissues. Diabetic mice with high blood sugar (hyperglycemia) were injected once with either R1MAbs or a control antibody. Within 1 week, blood glucose concentrations in the R1MAb-treated mice were normalized and remained at lower levels compared to placebo-treated mice for more than 1 month without reaching dangerously low blood glucose concentrations (hypoglycemia). The R1MAbs also helped the diabetic mice to lose weight, indicating that this antibody agonist of FGFR1 is a dual-action drug for both diabetes and obesity. Wu et al. also shed light on the mechanism of action of their R1MAbs, showing that they work via FGFR homodimerization in brown adipose tissue. With improved pharmacokinetics over FGF21, in addition to a specific receptor-targeting mechanism, these R1MAbs could enter human clinical trials for diabetes and other obesity-related diseases in the near future. Unfortunately, a miracle drug won’t be available in time for the holidays, so perhaps, this year, opt for the sugar-free egg nog. Clinical use of recombinant fibroblast growth factor 21 (FGF21) for the treatment of type 2 diabetes and other disorders linked to obesity has been proposed; however, its clinical development has been challenging owing to its poor pharmacokinetics. Here, we describe an alternative antidiabetic strategy using agonistic anti-FGFR1 (FGF receptor 1) antibodies (R1MAbs) that mimic the metabolic effects of FGF21. A single injection of R1MAb into obese diabetic mice induced acute and sustained amelioration of hyperglycemia, along with marked improvement in hyperinsulinemia, hyperlipidemia, and hepatosteatosis. R1MAb activated the mitogen-activated protein kinase pathway in adipose tissues, but not in liver, and neither FGF21 nor R1MAb improved glucose clearance in lipoatrophic mice, which suggests that adipose tissues played a central role in the observed metabolic effects. In brown adipose tissues, both FGF21 and R1MAb induced phosphorylation of CREB (cyclic adenosine 5′-monophosphate response element–binding protein), and mRNA expression of PGC-1α (peroxisome proliferator–activated receptor-γ coactivator 1α) and the downstream genes associated with oxidative metabolism. Collectively, we propose FGFR1 in adipose tissues as a major functional receptor for FGF21, as an upstream regulator of PGC-1α, and as a compelling target for antibody-based therapy for type 2 diabetes and other obesity-associated disorders.


Journal of Immunology | 2006

Cutting edge : Novel human dendritic cell- and monocyte-attracting chemokine-like protein identified by fold recognition methods

M. Teresa Pisabarro; Beatrice Leung; Mandy Kwong; Racquel Corpuz; Gretchen Frantz; Nan Chiang; Richard Vandlen; Lauri Diehl; Nicholas J. Skelton; Hok Seon Kim; Dan L. Eaton; Kerstin N. Schmidt

Chemokines play an important role in the immune system by regulating cell trafficking in homeostasis and inflammation. In this study, we report the identification and characterization of a novel cytokine-like protein, DMC (dendritic cell and monocyte chemokine-like protein), which attracts dendritic cells and monocytes. The key to the identification of this putative new chemokine was the application of threading techniques to its uncharacterized sequence. Based on our studies, DMC is predicted to have an IL-8-like chemokine fold and to be structurally and functionally related to CXCL8 and CXCL14. Consistent with our predictions, DMC induces migration of monocytes and immature dendritic cells. Expression studies show that DMC is constitutively expressed in lung, suggesting a potential role for DMC in recruiting monocytes and dendritic cells from blood into lung parenchyma.


EBioMedicine | 2015

Sustained Brown Fat Stimulation and Insulin Sensitization by a Humanized Bispecific Antibody Agonist for Fibroblast Growth Factor Receptor 1/βKlotho Complex

Ganesh Kolumam; Mark Z. Chen; Raymond K. Tong; Jose Zavala-Solorio; Lance Kates; Nicholas van Bruggen; Jed Ross; Shelby K. Wyatt; Vineela D. Gandham; Richard A. D. Carano; Diana Ronai Dunshee; Ai-Luen Wu; Benjamin Haley; Keith R. Anderson; Søren Warming; Xin Y. Rairdan; Nicholas Lewin-Koh; Yingnan Zhang; Johnny Gutierrez; Amos Baruch; Thomas Gelzleichter; Dale Stevens; Sharmila Rajan; Travis W. Bainbridge; Jean-Michel Vernes; Y. Gloria Meng; James Ziai; Robert Soriano; Matthew J. Brauer; Yongmei Chen

Dissipating excess calories as heat through therapeutic stimulation of brown adipose tissues (BAT) has been proposed as a potential treatment for obesity-linked disorders. Here, we describe the generation of a humanized effector-less bispecific antibody that activates fibroblast growth factor receptor (FGFR) 1/βKlotho complex, a common receptor for FGF21 and FGF19. Using this molecule, we show that antibody-mediated activation of FGFR1/βKlotho complex in mice induces sustained energy expenditure in BAT, browning of white adipose tissue, weight loss, and improvements in obesity-associated metabolic derangements including insulin resistance, hyperglycemia, dyslipidemia and hepatosteatosis. In mice and cynomolgus monkeys, FGFR1/βKlotho activation increased serum high-molecular-weight adiponectin, which appears to contribute over time by enhancing the amplitude of the metabolic benefits. At the same time, insulin sensitization by FGFR1/βKlotho activation occurs even before the onset of weight loss in a manner that is independent of adiponectin. Together, selective activation of FGFR1/βKlotho complex with a long acting therapeutic antibody represents an attractive approach for the treatment of type 2 diabetes and other obesity-linked disorders through enhanced energy expenditure, insulin sensitization and induction of high-molecular-weight adiponectin.


Journal of Biological Chemistry | 2017

Effector-attenuating Substitutions That Maintain Antibody Stability and Reduce Toxicity in Mice

Megan Lo; Hok Seon Kim; Raymond K. Tong; Travis W. Bainbridge; Jean-Michel Vernes; Yin Zhang; Yuwen Linda Lin; Shan Chung; Mark S. Dennis; Y. Joy Yu Zuchero; Ryan J. Watts; Jessica Couch; Y. Gloria Meng; Jasvinder Atwal; Randall J. Brezski; Christoph Spiess; James A. Ernst

The antibody Fc region regulates antibody cytotoxic activities and serum half-life. In a therapeutic context, however, the cytotoxic effector function of an antibody is often not desirable and can create safety liabilities by activating native host immune defenses against cells expressing the receptor antigens. Several amino acid changes in the Fc region have been reported to silence or reduce the effector function of antibodies. These earlier studies focused primarily on the interaction of human antibodies with human Fc-γ receptors, and it remains largely unknown how such changes to Fc might translate to the context of a murine antibody. We demonstrate that the commonly used N297G (NG) and D265A, N297G (DANG) variants that are efficacious in attenuating effector function in primates retain potent complement activation capacity in mice, leading to safety liabilities in murine studies. In contrast, we found an L234A, L235A, P329G (LALA-PG) variant that eliminates complement binding and fixation as well as Fc-γ-dependent, antibody-dependent, cell-mediated cytotoxity in both murine IgG2a and human IgG1. These LALA-PG substitutions allow a more accurate translation of results generated with an “effectorless” antibody between mice and primates. Further, we show that both human and murine antibodies containing the LALA-PG variant have typical pharmacokinetics in rodents and retain thermostability, enabling efficient knobs-into-holes bispecific antibody production and a robust path to generating highly effector-attenuated bispecific antibodies for preclinical studies.


mAbs | 2016

Evading pre-existing anti-hinge antibody binding by hinge engineering.

Hok Seon Kim; Ingrid Kim; Linda Zheng; Jean-Michel Vernes; Y. Gloria Meng; Christoph Spiess

ABSTRACT Antigen-binding fragments (Fab) and F(ab′)2 antibodies serve as alternative formats to full-length anti-bodies in therapeutic and immune assays. They provide the advantage of small size, short serum half-life, and lack of effector function. Several proteases associated with invasive diseases are known to cleave antibodies in the hinge-region, and this results in anti-hinge antibodies (AHA) toward the neoepitopes. The AHA can act as surrogate Fc and reintroduce the properties of the Fc that are otherwise lacking in antibody fragments. While this response is desired during the natural process of fighting disease, it is commonly unwanted for therapeutic antibody fragments. In our study, we identify a truncation in the lower hinge region of the antibody that maintains efficient proteolytic cleavage by IdeS protease. The resulting neoepitope at the F(ab′)2 C-terminus does not have detectable binding of pre-existing AHA, providing a practical route to produce F(ab′)2 in vitro by proteolytic digestion when the binding of pre-existing AHA is undesired. We extend our studies to the upper hinge region of the antibody and provide a detailed analysis of the contribution of C-terminal residues of the upper hinge of human IgG1, IgG2 and IgG4 to pre-existing AHA reactivity in human serum. While no pre-existing antibodies are observed toward the Fab of IgG2 and IgG4 isotype, a significant response is observed toward most residues of the upper hinge of human IgG1. We identify a T225L variant and the natural C-terminal D221 as solutions with minimal serum reactivity. Our work now enables the production of Fab and F(ab′)2 for therapeutic and diagnostic immune assays that have minimal reactivity toward pre-existing AHA.


PLOS ONE | 2012

Translation Levels Control Multi-Spanning Membrane Protein Expression

Hok Seon Kim; James A. Ernst; Cecilia Brown; Jenny Bostrom; Germaine Fuh; Chingwei V. Lee; Arthur Huang; Richard Vandlen; Daniel G. Yansura

Attempts to express eukaryotic multi-spanning membrane proteins at high-levels have been generally unsuccessful. In order to investigate the cause of this limitation and gain insight into the rate limiting processes involved, we have analyzed the effect of translation levels on the expression of several human membrane proteins in Escherichia coli (E. coli). These results demonstrate that excessive translation initiation rates of membrane proteins cause a block in protein synthesis and ultimately prevent the high-level accumulation of these proteins. Moderate translation rates allow coupling of peptide synthesis and membrane targeting, resulting in a significant increase in protein expression and accumulation over time. The current study evaluates four membrane proteins, CD20 (4-transmembrane (TM) helixes), the G-protein coupled receptors (GPCRs, 7-TMs) RA1c and EG-VEGFR1, and Patched 1 (12-TMs), and demonstrates the critical role of translation initiation rates in the targeting, insertion and folding of integral membrane proteins in the E. coli membrane.


Protein Engineering Design & Selection | 2017

Tethered-variable CL bispecific IgG: an antibody platform for rapid bispecific antibody screening

Hok Seon Kim; Diana Ronai Dunshee; Angie Yee; Raymond K. Tong; Ingrid Kim; Farzam Farahi; Jo-Anne Hongo; James A. Ernst; Junichiro Sonoda; Christoph Spiess

Abstract Bispecific antibodies offer a clinically validated platform for drug discovery. In generating functionally active bispecific antibodies, it is necessary to identify a unique parental antibody pair to merge into a single molecule. However, technologies that allow high-throughput production of bispecific immunoglobulin Gs (BsIgGs) for screening purposes are limited. Here, we describe a novel bispecific antibody format termed tethered-variable CLBsIgG (tcBsIgG) that allows robust production of intact BsIgG in a single cell line, concurrently ensuring cognate light chain pairing and preserving key antibody structural and functional properties. This technology is broadly applicable in the generation of BsIgG from a variety of antibody isotypes, including human BsIgG1, BsIgG2 and BsIgG4. The practicality of the tcBsIgG platform is demonstrated by screening BsIgGs generated from FGF21-mimetic anti-Klotho-β agonistic antibodies in a combinatorial manner. This screen identified multiple biepitopic combinations with enhanced agonistic activity relative to the parental monoclonal antibodies, thereby demonstrating that biepitopic antibodies can acquire enhanced functionality compared to monospecific parental antibodies. By design, the tcBsIgG format is amenable to high-throughput production of large panels of bispecific antibodies and thus can facilitate the identification of rare BsIgG combinations to enable the discovery of molecules with improved biological function.


Molecular Pharmaceutics | 2018

Susceptibility of Antibody CDR Residues to Chemical Modifications Can Be Revealed Prior to Antibody Humanization and Aid in the Lead Selection Process

Ankai Xu; Hok Seon Kim; Samarkand Estee; Sharon Viajar; William J. Galush; Avinash Gill; Isidro Hotzel; Greg Lazar; Paul J. Mcdonald; Nisana Andersen; Christoph Spiess

A critical part of the clinical development path for a therapeutic antibody involves evaluating the physical and chemical stability of candidate molecules throughout the manufacturing process. In particular, the risks of chemical liabilities that can impact antigen binding, such as deamidation, oxidation, and isomerization in the antibody CDR sequences, need to be controlled through formulation development or eliminated by replacing the amino acid motif displaying the chemical instability. Commonly, the antibody CDR sequence contains multiple sequence motifs (potential hotspots) for chemical instability. However, only a subset of these motifs results in actual chemical modification, and thus, experimental assessment of the extent of instability is necessary to identify positions for potential sequence engineering. Ideally, this information should be available prior to antibody humanization at the stage of parental rodent antibody identification. Early knowledge of liabilities allows for ranking of clones or the mitigation of liabilities by concurrent engineering with the antibody humanization process instead of time-consuming sequential activities. However, concurrent engineering of chemical liabilities and humanization requires translatability of the chemical modifications from the rodent parental antibody to the humanized. We experimentally compared the stability of all sequence motifs by mass spectrometric peptide mapping between the rodent parental antibody and the final humanized antibody and observed a linear correlation. These results have enabled a streamlined developability assessment process for therapeutic antibodies from lead discovery to clinical development.


Biochemistry | 2005

Isolation and Characterization of the B-Cell Marker CD20

James A. Ernst; Hong Li; Hok Seon Kim; Gerald R. Nakamura; Daniel G. Yansura; Richard Vandlen


Investigative Ophthalmology & Visual Science | 2015

Design and Pharmacokinetic Characterization of Novel Antibody Formats for Ocular Therapeutics.

Kapil Gadkar; Cinthia V. Pastuskovas; Jennifer Le Couter; J. Michael Elliott; Jianhuan Zhang; Chingwei V. Lee; Sarah Sanowar; Germaine Fuh; Hok Seon Kim; T. Noelle Lombana; Christoph Spiess; Makia Nakamura; Phil Hass; Whitney Shatz; Y. Gloria Meng; Justin Scheer

Collaboration


Dive into the Hok Seon Kim's collaboration.

Top Co-Authors

Avatar

James A. Ernst

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge