Network


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

Hotspot


Dive into the research topics where Byoung-Chul Lee is active.

Publication


Featured researches published by Byoung-Chul Lee.


Nature | 2015

Novel antibody–antibiotic conjugate eliminates intracellular S. aureus

Sophie M. Lehar; Thomas H. Pillow; Min Xu; Leanna Staben; Kimberly Kajihara; Richard Vandlen; Laura DePalatis; Helga Raab; Wouter L. W. Hazenbos; J. Hiroshi Morisaki; Janice Kim; Summer Park; Martine Darwish; Byoung-Chul Lee; Hilda Hernandez; Kelly M. Loyet; Patrick Lupardus; Rina Fong; Donghong Yan; Cecile Chalouni; Elizabeth Luis; Yana Khalfin; Emile Plise; Jonathan Cheong; Joseph P. Lyssikatos; Magnus Strandh; Klaus Koefoed; Peter S. Andersen; John A. Flygare; Man Wah Tan

Staphylococcus aureus is considered to be an extracellular pathogen. However, survival of S. aureus within host cells may provide a reservoir relatively protected from antibiotics, thus enabling long-term colonization of the host and explaining clinical failures and relapses after antibiotic therapy. Here we confirm that intracellular reservoirs of S. aureus in mice comprise a virulent subset of bacteria that can establish infection even in the presence of vancomycin, and we introduce a novel therapeutic that effectively kills intracellular S. aureus. This antibody–antibiotic conjugate consists of an anti-S. aureus antibody conjugated to a highly efficacious antibiotic that is activated only after it is released in the proteolytic environment of the phagolysosome. The antibody–antibiotic conjugate is superior to vancomycin for treatment of bacteraemia and provides direct evidence that intracellular S. aureus represents an important component of invasive infections.


Journal of Experimental Medicine | 2013

Antigen delivery to early endosomes eliminates the superiority of human blood BDCA3+ dendritic cells at cross presentation

Lillian Cohn; Bithi Chatterjee; Filipp Esselborn; Anna Smed-Sörensen; Norihiro Nakamura; Cecile Chalouni; Byoung-Chul Lee; Richard Vandlen; Tibor Keler; Peter Lauer; Dirk G. Brockstedt; Ira Mellman; Lélia Delamarre

Human BDCA3 DCs are superior to BDCA1 DCs at antigen cross presentation when delivered to late endosomes and lysosomes but not when delivered to early endosomes.


Blood | 2012

Internalization and endosomal degradation of receptor-bound antigens regulate the efficiency of cross presentation by human dendritic cells.

Bithi Chatterjee; Anna Smed-Sörensen; Lillian Cohn; Cecile Chalouni; Richard Vandlen; Byoung-Chul Lee; Jenifer Widger; Tibor Keler; Lélia Delamarre; Ira Mellman

Dendritic cells (DCs) can capture extracellular antigens and load resultant peptides on to MHC class I molecules, a process termed cross presentation. The mechanisms of cross presentation remain incompletely understood, particularly in primary human DCs. One unknown is the extent to which antigen delivery to distinct endocytic compartments determines cross presentation efficiency, possibly by influencing antigen egress to the cytosol. We addressed the problem directly and quantitatively by comparing the cross presentation of identical antigens conjugated with antibodies against different DC receptors that are targeted to early or late endosomes at distinct efficiencies. In human BDCA1+ and monocyte-derived DCs, CD40 and mannose receptor targeted antibody conjugates to early endosomes, whereas DEC205 targeted antigen primarily to late compartments. Surprisingly, the receptor least efficient at internalization, CD40, was the most efficient at cross presentation. This did not reflect DC activation by CD40, but rather its relatively poor uptake or intra-endosomal degradation compared with mannose receptor or DEC205. Thus, although both early and late endosomes appear to support cross presentation in human DCs, internalization efficiency, especially to late compartments, may be a negative predictor of activity when selecting receptors for vaccine development.


PLOS Pathogens | 2013

Novel Staphylococcal Glycosyltransferases SdgA and SdgB Mediate Immunogenicity and Protection of Virulence-Associated Cell Wall Proteins

Wouter L. W. Hazenbos; Kimberly Kajihara; Richard Vandlen; J. Hiroshi Morisaki; Sophie M. Lehar; Mark J. Kwakkenbos; Tim Beaumont; Arjen Q. Bakker; Qui Phung; Lee R. Swem; Satish Ramakrishnan; Janice Kim; Min Xu; Ishita M. Shah; Binh An Diep; Tao Sai; Andrew Sebrell; Yana Khalfin; Angela Oh; Chris Koth; S. Jack Lin; Byoung-Chul Lee; Magnus Strandh; Klaus Koefoed; Peter S. Andersen; Hergen Spits; Eric J. Brown; Man-Wah Tan; Sanjeev Mariathasan

Infection of host tissues by Staphylococcus aureus and S. epidermidis requires an unusual family of staphylococcal adhesive proteins that contain long stretches of serine-aspartate dipeptide-repeats (SDR). The prototype member of this family is clumping factor A (ClfA), a key virulence factor that mediates adhesion to host tissues by binding to extracellular matrix proteins such as fibrinogen. However, the biological siginificance of the SDR-domain and its implication for pathogenesis remain poorly understood. Here, we identified two novel bacterial glycosyltransferases, SdgA and SdgB, which modify all SDR-proteins in these two bacterial species. Genetic and biochemical data demonstrated that these two glycosyltransferases directly bind and covalently link N-acetylglucosamine (GlcNAc) moieties to the SDR-domain in a step-wise manner, with SdgB appending the sugar residues proximal to the target Ser-Asp repeats, followed by additional modification by SdgA. GlcNAc-modification of SDR-proteins by SdgB creates an immunodominant epitope for highly opsonic human antibodies, which represent up to 1% of total human IgG. Deletion of these glycosyltransferases renders SDR-proteins vulnerable to proteolysis by human neutrophil-derived cathepsin G. Thus, SdgA and SdgB glycosylate staphylococcal SDR-proteins, which protects them against host proteolytic activity, and yet generates major eptopes for the human anti-staphylococcal antibody response, which may represent an ongoing competition between host and pathogen.


Cancer Research | 2017

Cathepsin B Is Dispensable for Cellular Processing of Cathepsin B-Cleavable Antibody–Drug Conjugates

Niña G. Caculitan; Josefa Chuh; Yong Ma; Donglu Zhang; Katherine R. Kozak; Yichin Liu; Thomas H. Pillow; Jack Sadowsky; Tommy K. Cheung; Qui Phung; Benjamin Haley; Byoung-Chul Lee; Robert W. Akita; Mark X. Sliwkowski; Andrew G. Polson

Antibody-drug conjugates (ADC) are designed to selectively bind to tumor antigens via the antibody and release their cytotoxic payload upon internalization. Controllable payload release through judicious design of the linker has been an early technological milestone. Here, we examine the effect of the protease-cleavable valine-citrulline [VC(S)] linker on ADC efficacy. The VC(S) linker was designed to be cleaved by cathepsin B, a lysosomal cysteine protease. Surprisingly, suppression of cathepsin B expression via CRISPR-Cas9 gene deletion or shRNA knockdown had no effect on the efficacy of ADCs with VC(S) linkers armed with a monomethyl auristatin E (MMAE) payload. Mass spectrometry studies of payload release suggested that other cysteine cathepsins can cleave the VC(S) linker. Also, ADCs with a nonprotease-cleavable enantiomer, the VC(R) isomer, mediated effective cell killing with a cysteine-VC(R)-MMAE catabolite generated by lysosomal catabolism. Based on these observations, we altered the payload to a pyrrolo[2,1-c][1,4]benzodiazepine dimer (PBD) conjugate that requires linker cleavage in order to bind its DNA target. Unlike the VC-MMAE ADCs, the VC(S)-PBD ADC is at least 20-fold more cytotoxic than the VC(R)-PBD ADC. Our findings reveal that the VC(S) linker has multiple paths to produce active catabolites and that antibody and intracellular targets are more critical to ADC efficacy. These results suggest that protease-cleavable linkers are unlikely to increase the therapeutic index of ADCs and that resistance based on linker processing is improbable. Cancer Res; 77(24); 7027-37. ©2017 AACR.


Analytical Chemistry | 2017

High-Resolution Accurate-Mass Mass Spectrometry Enabling In-Depth Characterization of in Vivo Biotransformations for Intact Antibody-Drug Conjugates

Jintang He; Dian Su; Carl Ng; Luna Liu; Shang-Fan Yu; Thomas H. Pillow; Geoffrey Del Rosario; Martine Darwish; Byoung-Chul Lee; Rachana Ohri; Hongxiang Zhou; Xueji Wang; Jiawei Lu; Surinder Kaur; Keyang Xu

Antibody-drug conjugates (ADCs) represent a promising class of therapeutics for the targeted delivery of highly potent cytotoxic drugs to tumor cells to improve bioactivity while minimizing side effects. ADCs are composed of both small and large molecules and therefore have complex molecular structures. In vivo biotransformations may further increase the complexity of ADCs, representing a unique challenge for bioanalytical assays. Quadrupole-time-of-flight mass spectrometry (Q-TOF MS) with electrospray ionization has been widely used for characterization of intact ADCs. However, interpretation of ADC biotransformations with small mass changes, for the intact molecule, remains a limitation due to the insufficient mass resolution and accuracy of Q-TOF MS. Here, we have investigated in vivo biotransformations of multiple site-specific THIOMAB antibody-drug conjugates (TDCs), in the intact form, using a high-resolution, accurate-mass (HR/AM) MS approach. Compared with conventional Q-TOF MS, HR/AM Orbitrap MS enabled more comprehensive identification of ADC biotransformations. It was particularly beneficial for characterizing ADC modifications with small mass changes such as partial drug loss and hydrolysis. This strategy has significantly enhanced our capability to elucidate ADC biotransformations and help understand ADC efficacy and safety in vivo.


ACS Medicinal Chemistry Letters | 2017

Stabilizing a Tubulysin Antibody–Drug Conjugate To Enable Activity Against Multidrug-Resistant Tumors

Leanna Staben; Shang-Fan Yu; Jinhua Chen; Gang Yan; Zijin Xu; Geoffrey Del Rosario; Jeffrey Lau; Luna Liu; Jun Guo; Bing Zheng; Josefa dela Cruz-Chuh; Byoung-Chul Lee; Rachana Ohri; Wenwen Cai; Hongxiang Zhou; Katherine R. Kozak; Keyang Xu; Gail Lewis Phillips; Jiawei Lu; John S. Wai; Andrew Polson; Thomas H. Pillow

The tubulysins are promising anticancer cytotoxic agents due to the clinical validation of their mechanism of action (microtubule inhibition) and their particular activity against multidrug-resistant tumor cells. Yet their high potency and subsequent systemic toxicity make them prime candidates for targeted therapy, particularly in the form of antibody-drug conjugates (ADCs). Here we report a strategy to prepare stable and bioreversible conjugates of tubulysins to antibodies without loss of activity. A peptide trigger along with a quaternary ammonium salt linker connection to the tertiary amine of tubulysin provided ADCs that were potent in vitro. However, we observed metabolism of a critical acetate ester of the drug in vivo, resulting in diminished conjugate activity. We were able to circumvent this metabolic liability with the judicious choice of a propyl ether replacement. This modified tubulysin ADC was stable and effective against multidrug-resistant lymphoma cell lines and tumors.


Cancer Research | 2016

Abstract 1207: Preclinical development of 2nd generation HER2-directed antibody-drug conjugates

Gail Lewis Phillips; Guangmin Li; Jun Guo; Jeffrey Lau; Shang-Fan Yu; Thomas H. Pillow; Byoung-Chul Lee; Jack Sadowsky; Melissa Schutten; Carter Fields; Mark X. Sliwkowski

The HER2 receptor tyrosine kinase is amplified in approximately 20% of human breast cancer and is associated with poor clinical outcome. The humanized antibodies trastuzumab and pertuzumab are approved for use in both early and metastatic HER2-positive breast cancer, and are most often given with chemotherapy. Antibody-drug conjugates (ADCs) are anti-tumor agents designed to deliver potent cytotoxic drugs selectively to target-expressing tumor cells. Trastuzumab emtansine is a HER2-directed ADC comprised of trastuzumab covalently linked to the microtubule inhibitor DM1, through the stable MCC linker. Trastuzumab emtansine is approved for use in HER2-positive metastatic breast cancer as a single agent in patients who have received prior trastuzumab and a taxane. We are now exploring new HER2-directed ADCs (‘2nd generation ADCs’) with different mechanisms of action (MOA) than trastuzumab emtansine by investigating ADCs utilizing DNA-damaging agents, such as pyrrolobenzodiazepine (PBD) dimers and cyclopropylbenzindole (CBI) dimers, as the cytotoxic drug components. These agents have been conjugated to either trastuzumab or the humanized anti-HER2 antibody 7C2 (hu7C2) using both uncleavable and cleavable linkers. As free drugs and ADCs, the PBDs and CBIs show similar or greater potency in cell proliferation assays in vitro compared to DM1 and trastuzumab emtansine. However, unlike DM1, these agents are not strong substrates of Pgp/MDR1. Moreover, the PBDs and CBIs are active on non-dividing cells, whereas microtubule inhibitors such as DM1 do not affect non-dividing cells. Robust anti-tumor activity was observed in vivo in the fo5 HER2 transgenic tumor transplant model with the 2nd generation ADCs. Efficacious doses resulting in tumor stasis or regression ranged from 0.25-3 mg/kg administered as a single injection. In contrast, doses of trastuzumab emtansine required for stasis/regression in this model are 10 and 15 mg/kg, respectively. Efficacious doses were well-tolerated in the mouse xenograft models. Further tolerability studies of the 2nd generation ADCs were performed in rats. As rats are a non-binding species for trastuzumab and hu7C2, these studies assessed antigen-independent toxicities. Maximum tolerated doses for the different ADCs ranged from 2.5-15 mg/kg administered as a single injection, compared to 46 mg/kg for trastuzumab emtansine (Poon et al., 2013), likely reflecting both the different MOA and greater potency of the cytotoxic agents utilized in the 2nd generation HER2-directed ADCs. Overall, our findings demonstrate robust in vitro and in vivo activity of HER2 ADCs comprised of DNA-active agents, allowing for further development of a HER2-directed ADC distinct from trastuzumab emtansine. Citation Format: Gail D. Lewis Phillips, Guangmin Li, Jun Guo, Jeffrey Lau, Shang-Fan Yu, Thomas Pillow, Byoung-Chul Lee, Jack Sadowsky, Melissa Schutten, Carter Fields, Mark X. Sliwkowski. Preclinical development of 2nd generation HER2-directed antibody-drug conjugates. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1207.


Bioconjugate Chemistry | 2018

FRET Reagent Reveals the Intracellular Processing of Peptide-Linked Antibody–Drug Conjugates

Byoung-Chul Lee; Cecile Chalouni; Sophia Doll; Sam Nalle; Martine Darwish; Siao Ping Tsai; Katherine R. Kozak; Geoffrey Del-Rosario; Shang-Fan Yu; Hans Erickson; Richard Vandlen

Despite the recent success of antibody-drug conjugates (ADCs) in cancer therapy, a detailed understanding of their entry, trafficking, and metabolism in cancer cells is limited. To gain further insight into the activation mechanism of ADCs, we incorporated fluorescence resonance energy transfer (FRET) reporter groups into the linker connecting the antibody to the drug and studied various aspects of intracellular ADC processing mechanisms. When comparing the trafficking of the antibody-FRET drug conjugates in various different model cells, we found that the cellular background plays an important role in how the antigen-mediated antibody is processed. Certain tumor cells showed limited cytosolic transport of the payload despite efficient linker cleavage. Our FRET assay provides a facile and robust assessment of intracellular ADC activation that may have significant implications for the future development of ADCs.


Cancer Research | 2015

Abstract 211: A novel FRET assay for the intracellular activation of ADC linkers

Byoung-Chul Lee; Cecile Chalouni; Sam Nalle; Sophia Doll; Martine Darwish; Ira Mellman; Richard Vandlen

Proceedings: AACR 106th Annual Meeting 2015; April 18-22, 2015; Philadelphia, PA Evaluating the intracellular activation of ADC by a novel FRET assay. Despite the recent success of ADCs as cancer therapeutics, their mechanisms of action are not fully understood. In this study, we developed ADCs using a novel fluorescence resonance energy transfer (FRET) linker in order to facilitate monitoring the details of intracellular uptake, vesicular trafficking and payload release. In the FRET linker, the cathepsin-cleavable dipeptide of val-cit was inserted between a fluorescence donor Alexa Fluor 488 (or later, fluorescein) and an acceptor tetramethylrhodamine (TAMRA). Upon cleavage of the val-cit linker, a fluorescence signal from Alexa Fluor 488 or fluorescein is expected to appear as a probe for monitoring intracellular activation of ADC. We used two in-vitro human cancer cell lines, SKBR3 and PC3. SKBR3 is a Her2-positive breast cancer cell line and PC3 is a prostate cancer cell line that has been engineered to express a prostate cancer-specific growth factor receptor, tomoregulin-2 (TenB2). We assessed the intracellular processing of the FRET conjugates using flow cytometry. We found that 74% and 41% of the linker was cleaved in PC3 and SKBR3 cells after 20 hours of treatment, respectively. In the live-celling imaging of the FRET probe in SKBR3 and PC3 cells, Anti-Her2 and TenB2 FRET ADCs showed a remarkable difference of internalization pathways and cleavage following receptor-mediated endocytosis. In PC3 cells, FRET ADCs were internalized and congregated into a certain localized area during the early stage of uptake in 2 to 3 hours. The ADC-containing vesicles were spread over the cytosol and near the plasma membrane in a subset of cells. A similar directed localization at the early stage of uptake was not observed for the SKBR3 cells. The endosomal vesicles in SKBR3s were spread over the cytosol during the course of ADC internalization. The amount of the release payload in the cytosol of SKBR3s was 3-fold greater than that of PC3 cells, suggesting that the endosomal/lysosomal membrane of SKBR3 may be more permeable than that of PC3. In summary, our ADC FRET probes allowed for the facile, robust and non-invasive evaluation of intracellular processing for ADC linkers. Quantitative measurements for the rate of antigen-mediated intracellular cleavage of the FRET linker as well as cytosolic release could be evaluated by our FRET probes. Citation Format: Byoung-Chul Lee, Cecile Chalouni, Sam Nalle, Sophia Doll, Martine Darwish, Ira Mellman, Richard Vandlen. A novel FRET assay for the intracellular activation of ADC linkers. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 211. doi:10.1158/1538-7445.AM2015-211

Collaboration


Dive into the Byoung-Chul Lee's collaboration.

Researchain Logo
Decentralizing Knowledge