Network


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

Hotspot


Dive into the research topics where Jun Y. Axup is active.

Publication


Featured researches published by Jun Y. Axup.


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.


Journal of Physical Chemistry B | 2011

Two-Dimensional IR Spectroscopy of Protein Dynamics Using Two Vibrational Labels: A Site-Specific Genetically Encoded Unnatural Amino Acid and an Active Site Ligand

Megan C. Thielges; Jun Y. Axup; Daryl B. Wong; Hyun Soo Lee; Jean K. Chung; Peter G. Schultz; M. D. Fayer

Protein dynamics and interactions in myoglobin (Mb) were characterized via two vibrational dynamics labels (VDLs): a genetically incorporated site-specific azide (Az) bearing unnatural amino acid (AzPhe43) and an active site CO ligand. The Az-labeled protein was studied using ultrafast two-dimensional infrared (2D IR) vibrational echo spectroscopy. CO bound at the active site of the heme serves as a second VDL located nearby. Therefore, it was possible to use Fourier transform infrared (FT-IR) and 2D IR spectroscopic experiments on the Az in unligated Mb and in Mb bound to CO (MbAzCO) and on the CO in MbCO and MbAzCO to investigate the environment and motions of different states of one protein from the perspective of two spectrally resolved VDLs. A very broad bandwidth 2D IR spectrum, encompassing both the Az and CO spectral regions, found no evidence of direct coupling between the two VDLs. In MbAzCO, both VDLs reported similar time scale motions: very fast homogeneous dynamics, fast, ∼1 ps dynamics, and dynamics on a much slower time scale. Therefore, each VDL reports independently on the protein dynamics and interactions, and the measured dynamics are reflective of the protein motions rather than intrinsic to the chemical nature of the VDL. The AzPhe VDL also permitted study of oxidized Mb dynamics, which could not be accessed previously with 2D IR spectroscopy. The experiments demonstrate that the combined application of 2D IR spectroscopy and site-specific incorporation of VDLs can provide information on dynamics, structure, and interactions at virtually any site throughout any protein.


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 | 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.


Angewandte Chemie | 2013

Recruiting Cytotoxic T Cells to Folate-Receptor-Positive Cancer Cells**

Sumith A. Kularatne; Vishal Deshmukh; Marco Gymnopoulos; Sandra L. Biroc; Jinming Xia; Shailaja Srinagesh; Ying Sun; Ning Zou; Mark Shimazu; Jason Pinkstaff; Semsi Ensari; Nick Knudsen; Anthony Manibusan; Jun Y. Axup; Chan Hyuk Kim; Vaughn V. Smider; Tsotne Javahishvili; Prof.Dr. Peter G. Schultz

Herein, we describe the synthesis of a chemically defined anti-CD3 Fab-folate conjugate that targets cytotoxic T cells to folate receptor positive (FR+) tumors. The unnatural amino acid pacetylphenylalanine (pAcPhe) was site-specifically incorporated into an anti-CD3 Fab and conjugated to folate via the formation of a stable oxime linkage. The anti-CD3 Fab-folate conjugate was able to promote T cell mediated killing of FR+ cancer cells in culture. Moreover, the anti-CD3 Fab-folate conjugate potently eliminates tumor xenografts in mice. This approach can likely be generalized to other ligands that bind cancer and other pathogenic cells.


Biochemistry | 2011

Influence of Histidine Tag Attachment on Picosecond Protein Dynamics

Megan C. Thielges; Jean K. Chung; Jun Y. Axup; M. D. Fayer

Polyhistidine affinity tags are routinely employed as a convenient means of purifying recombinantly expressed proteins. A tacit assumption is commonly made that His tags have little influence on protein structure and function. Attachment of a His tag to the N-terminus of the robust globular protein myoglobin leads to only minor changes to the electrostatic environment of the heme pocket, as evinced by the nearly unchanged Fourier transform infrared spectrum of CO bound to the heme of His-tagged myoglobin. Experiments employing two-dimensional infrared vibrational echo spectroscopy of the heme-bound CO, however, find that significant changes occur to the short time scale (picoseconds) dynamics of myoglobin as a result of His tag incorporation. The His tag mainly reduces the dynamics on the 1.4 ps time scale and also alters protein motions of myoglobin on the slower, >100 ps time scale, as demonstrated by the His tags influence on the fluctuations of the CO vibrational frequency, which reports on protein structural dynamics. The results suggest that affinity tags may have effects on protein function and indicate that investigators of affinity-tagged proteins should take this into consideration when investigating the dynamics and other properties of such proteins.


Angewandte Chemie | 2015

Multiformat T-Cell-Engaging Bispecific Antibodies Targeting Human Breast Cancers†

Yu Cao; Jun Y. Axup; Rongsheng E. Wang; Seihyun Choi; Virginie Tardif; Reyna K. V. Lim; Holly Pugh; Brian R. Lawson; Gus Welzel; Stephanie A. Kazane; Ying Sun; Feng Tian; Shailaja Srinagesh; Tsotne Javahishvili; Peter G. Schultz; Chan Hyuk Kim

Four different formats of bispecific antibodies (bsAbs) were generated that consist of anti-Her2 IgG or Fab site-specifically conjugated to anti-CD3 Fab using the genetically encoded noncanonical amino acid. These bsAbs varied in valency or in the presence or absence of an Fc domain. Different valencies did not significantly affect antitumor efficacy, whereas the presence of an Fc domain enhanced cytotoxic activity, but triggered antigen-independent T-cell activation. We show that the bsAbs can efficiently redirect T cells to kill all Her2 expressing cancer cells, including Her2 1+ cancers, both in vitro and in rodent xenograft models. This work increases our understanding of the structural features that affect bsAb activity, and underscores the potential of bsAbs as a promising therapeutic option for breast cancer patients with low or heterogeneous Her2 expression.


Bioconjugate Chemistry | 2015

Antibody Microarrays Utilizing Site-Specific Antibody–Oligonucleotide Conjugates

Erik D. Wold; Ryan McBride; Jun Y. Axup; Stephanie A. Kazane; Vaughn V. Smider

Protein arrays are typically made by random absorption of proteins to the array surface, potentially limiting the amount of properly oriented and functional molecules. We report the development of a DNA encoded antibody microarray utilizing site-specific antibody-oligonucleotide conjugates that can be used for cell immobilization as well as the detection of genes and proteins. This technology allows for the facile generation of antibody microarrays while circumventing many of the drawbacks of conventionally produced antibody arrays. We demonstrate that this method can be used to capture and detect SK-BR-3 cells (Her2+ breast cancer cells) at concentrations as low as 10(2) cells/mL (which is equivalent to 10 cells per 100 μL array) without the use of microfluidics, which is 100- to 10(5)-fold more sensitive than comparable techniques. Additionally, the method was shown to be able to detect cells in a complex mixture, effectively immobilizing and specifically detecting Her2+ cells at a concentration of 10(2) SK-BR-3 cells/mL in 4 × 10(6) white blood cells/mL. Patients with a variety of cancers can have circulating tumor cell counts of between 1 and 10(3) cells/mL in whole blood, well within the range of this technology.

Collaboration


Dive into the Jun Y. Axup's collaboration.

Top Co-Authors

Avatar

Peter G. Schultz

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

Chan Hyuk Kim

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chanhyuk Kim

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

Quan Zhou

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Yu Cao

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge