Network


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

Hotspot


Dive into the research topics where Veena Warikoo is active.

Publication


Featured researches published by Veena Warikoo.


Biotechnology and Bioengineering | 2012

Integrated continuous production of recombinant therapeutic proteins

Veena Warikoo; Rahul Godawat; Kevin Brower; Sujit Jain; Daniel Cummings; Elizabeth Simons; Timothy Johnson; Jason Walther; Marcella Yu; Benjamin Wright; Jean McLarty; Kenneth P. Karey; Chris Hwang; Weichang Zhou; Frank Riske; Konstantin Konstantinov

In the current environment of diverse product pipelines, rapidly fluctuating market demands and growing competition from biosimilars, biotechnology companies are increasingly driven to develop innovative solutions for highly flexible and cost‐effective manufacturing. To address these challenging demands, integrated continuous processing, comprised of high‐density perfusion cell culture and a directly coupled continuous capture step, can be used as a universal biomanufacturing platform. This study reports the first successful demonstration of the integration of a perfusion bioreactor and a four‐column periodic counter‐current chromatography (PCC) system for the continuous capture of candidate protein therapeutics. Two examples are presented: (1) a monoclonal antibody (model of a stable protein) and (2) a recombinant human enzyme (model of a highly complex, less stable protein). In both cases, high‐density perfusion CHO cell cultures were operated at a quasi‐steady state of 50–60 × 106 cells/mL for more than 60 days, achieving volumetric productivities much higher than current perfusion or fed‐batch processes. The directly integrated and automated PCC system ran uninterrupted for 30 days without indications of time‐based performance decline. The product quality observed for the continuous capture process was comparable to that for a batch‐column operation. Furthermore, the integration of perfusion cell culture and PCC led to a dramatic decrease in the equipment footprint and elimination of several non‐value‐added unit operations, such as clarification and intermediate hold steps. These findings demonstrate the potential of integrated continuous bioprocessing as a universal platform for the manufacture of various kinds of therapeutic proteins. Biotechnol. Bioeng. 2012; 109: 3018–3029.


Biotechnology Journal | 2012

Periodic counter-current chromatography -- design and operational considerations for integrated and continuous purification of proteins.

Rahul Godawat; Kevin Brower; Sujit Jain; Konstantin Konstantinov; Frank Riske; Veena Warikoo

Integrated and continuous processing of recombinant proteins offers several advantages over batch or semi-batch processing used traditionally in the biotechnology industry. This paper presents a theoretical and practical approach for designing a periodic counter-current chromatography (PCC) operation as a continuous capture purification step that is integrated with a perfusion cell culture process. The constraints for continuous and optimal PCC operation govern the selection of residence time and number of columns. The flexibility available in PCC design for selection of these parameters is dictated by the binding characteristics of the target protein on the capture resin. Using an empirical model for the protein breakthrough curve, analytical solutions to determine these conditions were derived and verified with experimental results for three different proteins: two relatively unstable proteins (recombinant enzymes) and a relatively stable protein (monoclonal antibody). The advantages of a continuous downstream capture step are highlighted for the three case studies in comparison with the existing batch chromatography processes. The use of PCC leads to improvements in process economics due to higher resin capacity utilization and correspondingly lower buffer consumption. Furthermore, integrated and continuous bioprocessing results in a smaller facility footprint by elimination of harvest hold vessels and clarification, as well as by reducing the capture column size by one to two orders of magnitude.


Journal of Biotechnology | 2015

End-to-end integrated fully continuous production of recombinant monoclonal antibodies.

Rahul Godawat; Konstantin Konstantinov; Mahsa Rohani; Veena Warikoo

• This work represents the first (to our knowledge) demonstration of feasibility for end-to-end continuous bioprocess for biologics.


Biotechnology Progress | 2014

Single‐step affinity purification of enzyme biotherapeutics: A platform methodology for accelerated process development

Kevin Brower; Venkat Kishore Ryakala; Ryan Bird; Rahul Godawat; Frank Riske; Konstantin Konstantinov; Veena Warikoo; Jean Gamble

Downstream sample purification for quality attribute analysis is a significant bottleneck in process development for non‐antibody biologics. Multi‐step chromatography process train purifications are typically required prior to many critical analytical tests. This prerequisite leads to limited throughput, long lead times to obtain purified product, and significant resource requirements. In this work, immunoaffinity purification technology has been leveraged to achieve single‐step affinity purification of two different enzyme biotherapeutics (Fabrazyme® [agalsidase beta] and Enzyme 2) with polyclonal and monoclonal antibodies, respectively, as ligands. Target molecules were rapidly isolated from cell culture harvest in sufficient purity to enable analysis of critical quality attributes (CQAs). Most importantly, this is the first study that demonstrates the application of predictive analytics techniques to predict critical quality attributes of a commercial biologic. The data obtained using the affinity columns were used to generate appropriate models to predict quality attributes that would be obtained after traditional multi‐step purification trains. These models empower process development decision‐making with drug substance‐equivalent product quality information without generation of actual drug substance. Optimization was performed to ensure maximum target recovery and minimal target protein degradation. The methodologies developed for Fabrazyme were successfully reapplied for Enzyme 2, indicating platform opportunities. The impact of the technology is significant, including reductions in time and personnel requirements, rapid product purification, and substantially increased throughput. Applications are discussed, including upstream and downstream process development support to achieve the principles of Quality by Design (QbD) as well as integration with bioprocesses as a process analytical technology (PAT).


Biotechnology and Bioengineering | 2012

A modified IMAC method for the capture of target protein from mammalian cell culture harvest containing metal chelating species

Aming Zhang; Cheng Zhang; Veena Warikoo; Jonathan Forstrom; Frank Riske

Although immobilized metal affinity chromatography (IMAC) offers high capacity and protein selectivity it is not typically used commercially for the capture of native proteins from mammalian cell culture harvest. This is due mainly to the potential for low target recovery due to the presence of strong metal ion chelating species in the harvest that compete for the metal immobilized on the resin. To address this issue a buffer exchange step, such as tangential flow filtration (TFF), is added after harvest clarification and prior to IMAC to remove the interfering harvest components. The addition of a TFF step adds process time and cost and reduces target protein recovery. The elimination of the TFF might make IMAC competitive with other orthogonal methods of protein capture. In this study, we developed a modified IMAC method to allow the direct loading of clarified mammalian harvest without prior buffer exchange (direct IMAC). Although the target enzyme recovery was lower than that from standard IMAC the elimination of the buffer exchange step resulted in a 19% increase in overall enzyme recovery. The target enzyme capacity in direct IMAC was higher, in our experience, than the capacity of hydrophobic interaction (HIC) and ion‐exchange (IEX) for protein capture. An economic evaluation of using direct IMAC as a capture step in manufacturing is also discussed. Biotechnol. Bioeng. 2012; 109:747–753.


Archive | 2015

Integrated Continuous Manufacturing of Therapeutic Protein Drug Substances

Konstantin Konstantinov; Rahul Godawat; Veena Warikoo; Sujit Jain


Archive | 2014

CONTINUOUS PURIFICATION OF THERAPEUTIC PROTEINS

Konstantin Konstantinov; Rahul Godawat; Veena Warikoo; Sujit Jain


Archive | 2015

STERILE CHROMATOGRAPHY AND MANUFACTURING PROCESSES

Rahul Godawat; Veena Warikoo; Rohan Patil; Konstantin Konstantinov; Venkat Kishore Ryakala; Mahsa Rohani


Biotechnology Journal | 2015

A new use for existing technology - continuous precipitation for purification of recombination proteins.

Veena Warikoo; Rahul Godawat


Archive | 2015

Sterile chromatography resin and use thereof in manufacturing processes

Rahul Godawat; Veena Warikoo; Rohan Patil; Konstantin Konstantinov; Venkat Kishore Ryakala

Collaboration


Dive into the Veena Warikoo's collaboration.

Researchain Logo
Decentralizing Knowledge