Network


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

Hotspot


Dive into the research topics where Kalyani Penta is active.

Publication


Featured researches published by Kalyani Penta.


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 | 2012

Aglycosylated antibodies and antibody fragments produced in a scalable in vitro transcription-translation system

Gang Yin; Eudean D. Garces; Junhao Yang; Juan Zhang; Cuong Tran; Alexander R. Steiner; Christine Roos; Sunil Bajad; Susan Hudak; Kalyani Penta; James Zawada; Sonia Pollitt; Christopher J. Murray

We describe protein synthesis, folding and assembly of antibody fragments and full-length aglycosylated antibodies using an Escherichia coli-based open cell-free synthesis (OCFS) system. We use DNA template design and high throughput screening at microliter scale to rapidly optimize production of single-chain Fv (scFv) and Fab antibody fragments that bind to human IL-23 and IL-13α1R, respectively. In addition we demonstrate production of aglycosylated immunoglobulin G (IgG1) trastuzumab. These antibodies are produced rapidly over several hours in batch mode in standard bioreactors with linear scalable yields of hundreds of milligrams/L over a 1 million-fold change in scales up to pilot scale production. We demonstrate protein expression optimization of translation initiation region (TIR) libraries from gene synthesized linear DNA templates, optimization of the temporal assembly of a Fab from independent heavy chain and light chain plasmids and optimized expression of fully assembled trastuzumab that is equivalent to mammalian expressed material in biophysical and affinity based assays. These results illustrate how the open nature of the cell-free system can be used as a seamless antibody engineering platform from discovery to preclinical development of aglycosylated monoclonal antibodies and antibody fragments as potential therapeutics.


Protein Engineering Design & Selection | 2014

In vitro Fab display: a cell-free system for IgG discovery

Ryan Stafford; Marissa L. Matsumoto; Gang Yin; Qi Cai; Juan Jose Fung; Heather Stephenson; Avinash Gill; Monica You; Shwu-Hwa Lin; Willie D. Wang; Mary Rose Masikat; Xiaofan Li; Kalyani Penta; Alex Steiner; Ramesh Baliga; Christopher J. Murray; Christopher D. Thanos; Trevor J. Hallam; Aaron K. Sato

Selection technologies such as ribosome display enable the rapid discovery of novel antibody fragments entirely in vitro. It has been assumed that the open nature of the cell-free reactions used in these technologies limits selections to single-chain protein fragments. We present a simple approach for the selection of multi-chain proteins, such as antibody Fab fragments, using ribosome display. Specifically, we show that a two-chain trastuzumab (Herceptin) Fab domain can be displayed in a format which tethers either the heavy or light chain to the ribosome while retaining functional antigen binding. Then, we constructed synthetic Fab HC and LC libraries and performed test selections against carcinoembryonic antigen (CEA) and vascular endothelial growth factor (VEGF). The Fab selection output was reformatted into full-length immunoglobulin Gs (IgGs) and directly expressed at high levels in an optimized cell-free system for immediate screening, purification and characterization. Several novel IgGs were identified using this cell-free platform that bind to purified CEA, CEA positive cells and VEGF.


Scientific Reports | 2017

RF1 attenuation enables efficient non-natural amino acid incorporation for production of homogeneous antibody drug conjugates

Gang Yin; Heather Stephenson; Junhao Yang; Xiaofan Li; Stephanie Armstrong; Tyler H. Heibeck; Cuong D. Tran; Mary Rose Masikat; Sihong Zhou; Ryan Stafford; Alice Y. Yam; John D. Lee; Alexander Steiner; Avinash Gill; Kalyani Penta; Sonia Pollitt; Ramesh Baliga; Christopher J. Murray; Christopher D. Thanos; Leslie Mcevoy; Aaron Ken Sato; Trevor J. Hallam

Amber codon suppression for the insertion of non-natural amino acids (nnAAs) is limited by competition with release factor 1 (RF1). Here we describe the genome engineering of a RF1 mutant strain that enhances suppression efficiency during cell-free protein synthesis, without significantly impacting cell growth during biomass production. Specifically, an out membrane protease (OmpT) cleavage site was engineered into the switch loop of RF1, which enables its conditional inactivation during cell lysis. This facilitates extract production without additional processing steps, resulting in a scaleable extract production process. The RF1 mutant extract allows nnAA incorporation at previously intractable sites of an IgG1 and at multiple sites in the same polypeptide chain. Conjugation of cytotoxic agents to these nnAAs, yields homogeneous antibody drug conjugates (ADCs) that can be optimized for conjugation site, drug to antibody ratio (DAR) and linker-warheads designed for efficient tumor killing. This platform provides the means to generate therapeutic ADCs inaccessible by other methods that are efficient in their cytotoxin delivery to tumor with reduced dose-limiting toxicities and thus have the potential for better clinical impact.


Pharmaceutical Research | 2015

Methods to Make Homogenous Antibody Drug Conjugates

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


Archive | 2013

Antibodies comprising site-specific non-natural amino acid residues, methods of their preparation and methods of their use

Christopher D. Thanos; Leslie Mcevoy; Gang Yin; Kalyani Penta; Ramesh Baliga; Sunil Bajad; Sonia Pollitt; Chris Murray; Alex Steiner; Avinash Gill


Archive | 2013

MODIFIED FC PROTEINS COMPRISING SITE-SPECIFIC NON-NATURAL AMINO ACID RESIDUES, CONJUGATES OF THE SAME, METHODS OF THEIR PREPARATION AND METHODS OF THEIR USE

Christopher D. Thanos; Leslie Mcevoy; Gang Yin; Kalyani Penta; Ramesh Baliga; Sunil Bajad; Sonia Pollitt; Chris Murray; Alex Steiner; Avinash Gill


Archive | 2017

ANTI-CD74 ANTIBODIES, COMPOSITIONS COMPRISING ANTI-CD74 ANTIBODIES AND METHODS OF USING ANTI-CD74 ANTIBODIES

Kalyani Penta; Ryan Stafford; Avinash Gill; Xiaofan Li; Alice Yam; Christopher D. Thanos; Aaron Ken Sato


Archive | 2017

anticorpo, e, composição.

Alex Steiner; Avinash Gill; Chistopher D Thanos; Chris Murray; Gang Yin; Kalyani Penta; Leslie Mcevoy; Ramesh Baliga; Sonia Pollitt; Sunil Bajad


Archive | 2017

Anti-cd74 antibody conjugates, compositions comprising anti-cd74 antibody conjugates and methods of using anti-cd74 antibody conjugates

Ryan Stafford; Alice Yam; Avinash Gill; Kalyani Penta; Xiaofan Li; Aaron Ken Sato

Collaboration


Dive into the Kalyani Penta's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alexander Steiner

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Christopher J. Murray

Pacific Northwest National Laboratory

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John D. Lee

University of Queensland

View shared research outputs
Researchain Logo
Decentralizing Knowledge