Network


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

Hotspot


Dive into the research topics where Alexander Steiner is active.

Publication


Featured researches published by Alexander Steiner.


Bioconjugate Chemistry | 2014

Production of site-specific antibody-drug conjugates using optimized non-natural amino acids in a cell-free expression system.

Erik S. Zimmerman; Tyler H. Heibeck; Avinash Gill; Xiaofan Li; Christopher J. Murray; Mary Rose Madlansacay; Cuong Tran; Nathan Uter; Gang Yin; Patrick Rivers; Alice Y. Yam; Willie D. Wang; Alexander Steiner; Sunil Bajad; Kalyani Penta; Wenjin Yang; Trevor J. Hallam; Christopher D. Thanos; Aaron K. Sato

Antibody-drug conjugates (ADCs) are a targeted chemotherapeutic currently at the cutting edge of oncology medicine. These hybrid molecules consist of a tumor antigen-specific antibody coupled to a chemotherapeutic small molecule. Through targeted delivery of potent cytotoxins, ADCs exhibit improved therapeutic index and enhanced efficacy relative to traditional chemotherapies and monoclonal antibody therapies. The currently FDA-approved ADCs, Kadcyla (Immunogen/Roche) and Adcetris (Seattle Genetics), are produced by conjugation to surface-exposed lysines, or partial disulfide reduction and conjugation to free cysteines, respectively. These stochastic modes of conjugation lead to heterogeneous drug products with varied numbers of drugs conjugated across several possible sites. As a consequence, the field has limited understanding of the relationships between the site and extent of drug loading and ADC attributes such as efficacy, safety, pharmacokinetics, and immunogenicity. A robust platform for rapid production of ADCs with defined and uniform sites of drug conjugation would enable such studies. We have established a cell-free protein expression system for production of antibody drug conjugates through site-specific incorporation of the optimized non-natural amino acid, para-azidomethyl-l-phenylalanine (pAMF). By using our cell-free protein synthesis platform to directly screen a library of aaRS variants, we have discovered a novel variant of the Methanococcus jannaschii tyrosyl tRNA synthetase (TyrRS), with a high activity and specificity toward pAMF. We demonstrate that site-specific incorporation of pAMF facilitates near complete conjugation of a DBCO-PEG-monomethyl auristatin (DBCO-PEG-MMAF) drug to the tumor-specific, Her2-binding IgG Trastuzumab using strain-promoted azide-alkyne cycloaddition (SPAAC) copper-free click chemistry. The resultant ADCs proved highly potent in in vitro cell cytotoxicity assays.


mAbs | 2014

Engineering toward a bacterial "endoplasmic reticulum" for the rapid expression of immunoglobulin proteins.

Dan Groff; Stephanie Armstrong; Patrick Rivers; Juan Zhang; Junhao Yang; Evan Green; James Rozzelle; Shengwen Liang; Joseph D Kittle; Alexander Steiner; Ramesh Baliga; Christopher D. Thanos; Trevor J. Hallam; Aaron K. Sato; Alice Y. Yam

Antibodies are well-established as therapeutics, and the preclinical and clinical pipeline of these important biologics is growing rapidly. Consequently, there is considerable interest in technologies to engineer and manufacture them. Mammalian cell culture is commonly used for production because eukaryotic expression systems have evolved complex and efficient chaperone systems for the folding of antibodies. However, given the ease and manipulability of bacteria, antibody discovery efforts often employ bacterial expression systems despite their limitations in generating high titers of functional antibody. Open-Cell Free Synthesis (OCFS) is a coupled transcription-translation system that has the advantages of prokaryotic systems while achieving high titers of antibody expression. Due to the open nature of OCFS, it is easily modified by chemical or protein additives to improve the folding of select proteins. As such, we undertook a protein additive screen to identify chaperone proteins that improve the folding and assembly of trastuzumab in OCFS. From the screen, we identified the disulfide isomerase DsbC and the prolyl isomerase FkpA as important positive effectors of IgG folding. These periplasmic chaperones function synergistically for the folding and assembly of IgG, and, when present in sufficient quantities, gram per liter IgG titers can be produced. This technological advancement allows the rapid development and manufacturing of immunoglobulin proteins and pushes OCFS to the forefront of production technologies for biologics.


mAbs | 2015

Production of bispecific antibodies in “knobs-into-holes” using a cell-free expression system

Yiren Xu; John Lee; Cuong Tran; Tyler H. Heibeck; Willie D. Wang; Junhao Yang; Ryan Stafford; Alexander Steiner; Aaron K. Sato; Trevor J. Hallam; Gang Yin

Bispecific antibodies have emerged in recent years as a promising field of research for therapies in oncology, inflammable diseases, and infectious diseases. Their capability of dual target recognition allows for novel therapeutic hypothesis to be tested, where traditional mono-specific antibodies would lack the needed mode of target engagement. Among extremely diverse architectures of bispecific antibodies, knobs-into-holes (KIHs) technology, which involves engineering CH3 domains to create either a “knob” or a “hole” in each heavy chain to promote heterodimerization, has been widely applied. Here, we describe the use of a cell-free expression system (Xpress CF) to produce KIH bispecific antibodies in multiple scaffolds, including 2-armed heterodimeric scFv-KIH and one-armed asymmetric BiTE-KIH with tandem scFv. Efficient KIH production can be achieved by manipulating the plasmid ratio between knob and hole, and further improved by addition of prefabricated knob or hole. These studies demonstrate the versatility of Xpress CF in KIH production and provide valuable insights into KIH construct design for better assembly and expression titer.


Biotechnology Progress | 2015

A simplified and robust protocol for immunoglobulin expression in Escherichia coli cell‐free protein synthesis systems

Qi Cai; Jeffrey Hanson; Alexander Steiner; Cuong Tran; Mary Rose Masikat; Rishard Chen; James Zawada; Aaron K. Sato; Trevor J. Hallam; Gang Yin

Cell‐free protein synthesis (CFPS) systems allow for robust protein expression with easy manipulation of conditions to improve protein yield and folding. Recent technological developments have significantly increased the productivity and reduced the operating costs of CFPS systems, such that they can compete with conventional in vivo protein production platforms, while also offering new routes for the discovery and production of biotherapeutics. As cell‐free systems have evolved, productivity increases have commonly been obtained by addition of components to previously designed reaction mixtures without careful re‐examination of the essentiality of reagents from previous generations. Here we present a systematic sensitivity analysis of the components in a conventional Escherichia coli CFPS reaction mixture to evaluate their optimal concentrations for production of the immunoglobulin G trastuzumab. We identify eight changes to the system, which result in optimal expression of trastuzumab. We find that doubling the potassium glutamate concentration, while entirely eliminating pyruvate, coenzyme A, NAD, total tRNA, folinic acid, putrescine and ammonium glutamate, results in a highly productive cell‐free system with a 95% reduction in reagent costs (excluding cell‐extract, plasmid, and T7 RNA polymerase made in‐house). A larger panel of other proteins was also tested and all show equivalent or improved yields with our simplified system. Furthermore, we demonstrate that all of the reagents for CFPS can be combined in a single freeze‐thaw stable master mix to improve reliability and ease of use. These improvements are important for the application of the CFPS system in fields such as protein engineering, high‐throughput screening, and biotherapeutics.


Molecular Cancer Therapeutics | 2016

Targeted Drug Delivery with an Integrin-Binding Knottin–Fc–MMAF Conjugate Produced by Cell-Free Protein Synthesis

Nicolas V. Currier; Shelley E. Ackerman; James R. Kintzing; Rishard Chen; Maria Filsinger Interrante; Alexander Steiner; Aaron Sato; Jennifer R. Cochran

Antibody–drug conjugates (ADC) have generated significant interest as targeted therapeutics for cancer treatment, demonstrating improved clinical efficacy and safety compared with systemic chemotherapy. To extend this concept to other tumor-targeting proteins, we conjugated the tubulin inhibitor monomethyl-auristatin-F (MMAF) to 2.5F–Fc, a fusion protein composed of a human Fc domain and a cystine knot (knottin) miniprotein engineered to bind with high affinity to tumor-associated integrin receptors. The broad expression of integrins (including αvβ3, αvβ5, and α5β1) on tumor cells and their vasculature makes 2.5F-Fc an attractive tumor-targeting protein for drug delivery. We show that 2.5F-Fc can be expressed by cell-free protein synthesis, during which a non-natural amino acid was introduced into the Fc domain and subsequently used for site-specific conjugation of MMAF through a noncleavable linker. The resulting knottin–Fc–drug conjugate (KFDC), termed 2.5F-Fc-MMAF, had approximately 2 drugs attached per KFDC. 2.5F–Fc–MMAF inhibited proliferation in human glioblastoma (U87MG), ovarian (A2780), and breast (MB-468) cancer cells to a greater extent than 2.5F–Fc or MMAF alone or added in combination. As a single agent, 2.5F–Fc–MMAF was effective at inducing regression and prolonged survival in U87MG tumor xenograft models when administered at 10 mg/kg two times per week. In comparison, tumors treated with 2.5F–Fc or MMAF were nonresponsive, and treatment with a nontargeted control, CTRL–Fc–MMAF, showed a modest but not significant therapeutic effect. These studies provide proof-of-concept for further development of KFDCs as alternatives to ADCs for tumor targeting and drug delivery applications. Mol Cancer Ther; 15(6); 1291–300. ©2016 AACR.


Viruses | 2018

Post-Exposure Protection in Mice against Sudan Virus by a Two Antibody Cocktail

Jeffrey W. Froude; Andrew S. Herbert; Thibaut Pelat; Sebastian Miethe; Samantha E. Zak; Jennifer M. Brannan; Russell R. Bakken; Alexander Steiner; Gang Yin; Trevor J. Hallam; Aaron Sato; Michael Hust; Philippe Thullier; John M. Dye

Sudan virus (SUDV) and Ebola viruses (EBOV) are both members of the Ebolavirus genus and have been sources of epidemics and outbreaks for several decades. We present here the generation and characterization of cross-reactive antibodies to both SUDV and EBOV, which were produced in a cell-free system and protective against SUDV in mice. A non-human primate, cynomolgus macaque, was immunized with viral-replicon particles expressing the glycoprotein of SUDV-Boniface (8A). Two separate antibody fragment phage display libraries were constructed after four immunogen injections. Both libraries were screened first against the SUDV and a second library was cross-selected against EBOV-Kikwit. Sequencing of 288 selected clones from the two distinct libraries identified 58 clones with distinct VH and VL sequences. Many of these clones were cross-reactive to EBOV and SUDV and able to neutralize SUDV. Three of these recombinant antibodies (X10B1, X10F3, and X10H2) were produced in the scFv-Fc format utilizing a cell-free production system. Mice that were challenged with SUDV-Boniface receiving 100µg of the X10B1/X10H2 scFv-Fc combination 6 and 48-h post-exposure demonstrated partial protection individually and complete protection as a combination. The data herein suggests these antibodies may be promising candidates for further therapeutic development.


Pharmaceutical Research | 2015

Methods to Make Homogenous Antibody Drug Conjugates

Toni Kline; Alexander Steiner; Kalyani Penta; Aaron K. Sato; Trevor J. Hallam; Gang Yin


Blood | 2016

Targeting CD74 with Novel Antibody Drug Conjugates (ADCs) for the Treatment of B-Cell Non-Hodgkin's Lymphoma (NHL)

Xiaofan Li; Cristina Abrahams; Millicent Embry; Abigail Yu; Jason Kahana; Michael Brown; Rama Krishna Narla; Leo Barnes; Eric Schwartz; John Boylan; James Zawada; Heather Stephenson; Maureen Fitch Bruhns; Stuart Bussell; Alexander Steiner; Adam Galan; Toni Kline; Alice Yam; Ryan Stafford; Heidi Hoffmann; Shannon Matheny; Venita DeAlmeida; Nicki Vasquez; Henry Heinsohn; Aaron Ken Sato; Arturo Molina; Trevor J. Hallam; Mark Lupher


Organic Process Research & Development | 2016

RP-HPLC DAR Characterization of Site-Specific Antibody Drug Conjugates Produced in a Cell-Free Expression System

Yiren Xu; Guifeng Jiang; Cuong Tran; Xiaofan Li; Tyler H. Heibeck; Mary Rose Masikat; Qi Cai; Alexander Steiner; Aaron K. Sato; Trevor J. Hallam; Gang Yin


Blood | 2016

Discovery and Preclinical Development of Novel CD74-Targeting Antibody-Drug Conjugates (ADCs) with Significant Activity in Multiple Myeloma (MM) Cell Lines and Xenograft Models

Cristina Abrahams; Xiaofan Li; Abigail Yu; Stellanie Krimm; Jason Kahana; Rama Krishna Narla; Eric Schwartz; John Boylan; Heidi Hoffmann; Alexander Steiner; James Zawada; Heather Stephenson; Maureen Fitch Bruhns; Venita DeAlmeida; Shannon Matheny; Stuart Bussell; Adam Galan; Toni Kline; Nicki Vasquez; Alice Yam; Ryan Stafford; Henry Heinsohn; Aaron Ken Sato; Arturo Molina; Trevor J. Hallam; Mark Lupher

Collaboration


Dive into the Alexander Steiner's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge