Network


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

Hotspot


Dive into the research topics where Nripen Singh is active.

Publication


Featured researches published by Nripen Singh.


Biotechnology and Bioengineering | 2015

Development of robust antibody purification by optimizing protein‐A chromatography in combination with precipitation methodologies

Srinivas Chollangi; Ray Parker; Nripen Singh; Yi Li; Michael C. Borys; Zhengjian Li

To be administered to patients, therapeutic monoclonal antibodies must have very high purity, with process related impurities like host‐cell proteins (HCPs) and DNA reduced to <100 ppm and <10 ppb, respectively, relative to desired product. Traditionally, Protein‐A chromatography as a capture step has been the work horse for clearing a large proportion of these impurities. However, remaining levels of process and product related impurities still present significant challenges on the development of polishing steps further downstream. In this study, we have incorporated high throughput screening to evaluate three areas of separation: (i) Harvest treatment; (ii) Protein‐A Chromatography; and (iii) Low pH Viral Inactivation. Precipitation with low pH treatment of cell culture harvest resulted in selective removal of impurities while manipulating the pH of wash buffers used in Protein‐A chromatography and incorporating wash additives that disrupt various modes of protein–protein interaction resulted in further and more pronounced reduction in impurity levels. In addition, our study also demonstrate that optimizing the neutralization pH post Protein‐A elution can result in selective removal of impurities. When applied over multiple mAbs, this optimization method proved to be very robust and the strategy provides a new and improved purification process that reduces process related impurities like HCPs and DNA to drug substance specifications with just one chromatography column and open avenues for significant decrease in operating costs in monoclonal antibody purification. Biotechnol. Bioeng. 2015;112: 2292–2304.


Biotechnology and Bioengineering | 2016

Clarification technologies for monoclonal antibody manufacturing processes: Current state and future perspectives

Nripen Singh; Abhiram Arunkumar; Srinivas Chollangi; Zhijun George Tan; Michael C. Borys; Zheng Jian Li

Considerable progress has been made increasing productivity of cell cultures to meet the rapidly growing demand for antibody biopharmaceuticals through increased cell densities and longer culture times. This in turn has dramatically increased the burden of process and product related impurities on the purification processes. In addition, current trends in the biopharmaceutical industry point toward both increased productivity and targeting smaller patient populations for new indications. Taken together, these developments are driving the industry to explore alternative separation technologies as a future manufacturing strategy. Clarification technologies well established in other industries, such as flocculation and precipitation are increasingly considered as a viable solution to address this bottleneck in antibody processes. However, several technical issues need to be fully addressed including suitability as a platform application, robustness, process cost, toxicity, and clearance. This review will focus on recent efforts to incorporate new generation clarification technologies for mammalian cell cultures producing monoclonal antibodies as well as challenges to their implementation supported by a case study. Biotechnol. Bioeng. 2016;113: 698–716.


Biotechnology and Bioengineering | 2017

Ultrafiltration Behavior of Monoclonal Antibodies and Fc-Fusion Proteins: Effects of Physical Properties†

Youngbin Baek; Nripen Singh; Abhiram Arunkumar; Zheng Jian Li; Michael C. Borys; Andrew L. Zydney

Ultrafiltration (UF) is used for the final concentration and formulation of essentially all antibody‐based therapeutics including both monoclonal antibodies (mAbs) and Fc‐fusion proteins. The objective of this study was to quantitatively compare the filtrate flux behavior for two highly purified mAbs and an Fc‐fusion protein under identical flow and buffer conditions. Filtrate flux data were obtained using a Pellicon 3 tangential flow filtration cassette over a wide range of transmembrane pressures and bulk protein concentrations. Independent experimental measurements were performed to evaluate the protein osmotic pressure and solution viscosity. The maximum achievable protein concentration was directly correlated with the solution viscosity, which controls the pressure drop and extent of back‐filtration in the cassette. The filtrate flux data were analyzed using a recently developed model that accounts for the effects of intermolecular interactions and transmembrane pressure gradients on the extent of concentration polarization. These results provide important insights into the factors controlling the filtrate flux during the UF of concentrated protein solutions and an effective framework for the design/analysis of UF processes for the formulation of antibody‐based therapeutics. Biotechnol. Bioeng. 2017;114: 2057–2065.


mAbs | 2017

Development of adsorptive hybrid filters to enable two-step purification of biologics

Nripen Singh; Abhiram Arunkumar; Michael Peck; Alexei M. Voloshin; Angela M. Moreno; Zhijun Tan; Jonathan Hester; Michael C. Borys; Zheng Jian Li

ABSTRACT Recent progress in mammalian cell culture process has resulted in significantly increased product titers, but also a substantial increase in process- and product-related impurities. Due to the diverse physicochemical properties of these impurities, there is constant need for new technologies that offer higher productivity and improved economics without sacrificing the process robustness required to meet final drug substance specifications. Here, we examined the use of new synthetic adsorptive hybrid filters (AHF) modified with the high binding capacity of quaternary amine (Emphaze™ AEX) and salt-tolerant biomimetic (Emphaze™ ST-AEX) ligands for clearance of process-related impurities like host cell protein (HCP), residual DNA, and virus. The potential to remove soluble aggregates was also examined. Our aim was to develop a mechanistic understanding of the interactions governing adsorptive removal of impurities during filtration by evaluating the effect of various filter types, feed streams, and process conditions on impurity removal. The ionic capacity of these filters was measured and correlated with their ability to remove impurities for multiple molecules. The ionic capacity of AHF significantly exceeded that of traditional adsorptive depth filters (ADF) by 40% for the Emphaze™ AEX and by 700% for the Emphaze™ ST-AEX, providing substantially higher reduction of soluble anionic impurities, including DNA, HCPs and model virus. Nevertheless, we determined that ADF with filter aid provided additional hydrophobic functionality that resulted in removal of higher molecular weight species than AHF. Implementing AHF demonstrated improved process-related impurity removal and viral clearance after Protein A chromatography and enabled a two-step purification process. The consequences of enhanced process performance are far reaching because it allows the downstream polishing train to be restructured and simplified, and chromatographic purity standards to be met with a reduced number of chromatographic steps.


Archive | 2017

Downstream Processing Technologies/Capturing and Final Purification

Nripen Singh; Sibylle Herzer

Increased pressure on upstream processes to maximize productivity has been crowned with great success, although at the cost of shifting the bottleneck to purification. As drivers were economical, focus is on now on debottlenecking downstream processes as the main drivers of high manufacturing cost. Devising a holistically efficient and economical process remains a key challenge. Traditional and emerging protein purification strategies with particular emphasis on methodologies implemented for the production of recombinant proteins of biopharmaceutical importance are reviewed. The breadth of innovation is addressed, as well as the challenges the industry faces today, with an eye to remaining impartial, fair, and balanced. In addition, the scope encompasses both chromatographic and non-chromatographic separations directed at the purification of proteins, with a strong emphasis on antibodies. Complete solutions such as integrated USP/DSP strategies (i.e., continuous processing) are discussed as well as gains in data quantity and quality arising from automation and high-throughput screening (HTS). Best practices and advantages through design of experiments (DOE) to access a complex design space such as multi-modal chromatography are reviewed with an outlook on potential future trends. A discussion of single-use technology, its impact and opportunities for further growth, and the exciting developments in modeling and simulation of DSP rounds out the overview. Lastly, emerging trends such as 3D printing and nanotechnology are covered. Graphical Abstract Workflow of high-throughput screening, design of experiments, and high-throughput analytics to understand design space and design space boundaries quickly. (Reproduced with permission from Gregory Barker, Process Development, Bristol-Myers Squibb).


European Journal of Pharmaceutics and Biopharmaceutics | 2018

Protection of therapeutic antibodies from visible light induced degradation: Use safe light in manufacturing and storage

Cheng Du; Gregory V. Barnett; Ameya Borwankar; Angela Lewandowski; Nripen Singh; Sanchayita Ghose; Michael C. Borys; Zheng Jian Li

Graphical abstract Figure. No caption available. &NA; As macromolecules, biologics are susceptible to light exposure, which induces oxidation of multiple amino acid residues including tryptophan, tyrosine, phenylalanine, cysteine and methionine. Pertaining to safety, efficacy and potency, light‐induced oxidation of biologics has been widely studied and necessary precautions need to be taken during biologics manufacturing process, drug substance and products handling and storage. Proteins will degrade to varying extents depending on the protein properties, degradation pathways, formulation compositions and type of light source. In addition to UV light, which has been widely known to degrade proteins, visible light from indoor fluorescent lighting also can mediate protein degradation. In this report, we examine and identify wavelengths in the visual spectrum (400–700 nm) that can cause monoclonal antibody and histidine buffer degradation. Installation of safe lights which exclude the identified damaging wavelengths from visible spectra in manufacturing and storage areas can provide a balance between lighting requirement for human operators and their safety and conservation of product quality.


Biotechnology and Bioengineering | 2018

Vitamin B12 association with mAbs: Mechanism and potential mitigation strategies

Cheng Du; Robert Martin; Yunping Huang; Ameya Borwankar; Zhijun Tan; Jay West; Nripen Singh; Michael C. Borys; Sanchayita Ghose; Richard Ludwig; Li Tao; Zheng Jian Li

Process control for manufacturing biologics is critical for ensuring product quality, safety, and lot to lot consistency of therapeutic proteins. In this study, we investigated the root cause of the pink coloration observed for various in‐process pools and drug substances in the antibody manufacturing process. Vitamin B12 is covalently bound to mAbs via a cobalt‐sulfur coordinate bond via the cysteine residues. The vitamin B12 was identified to attach to an IgG4 molecule at cysteine residues on light chain (Cys‐214), and heavy chain (Cys‐134, Cys‐321, Cys‐367, and Cys‐425). Prior to attachment to mAbs, the vitamin B12 needs to be in its active form of hydroxocobalamin. During culture media preparation, storage and cell culture processing, cyanocobalamin, the chemical form of vitamin B12 added to media, is converted to hydroxocobalamin by white fluorescence light (about 50% degradation in 11–14 days at room temperature and with room light intensity about 500–1,000 lux) and by short‐wavelength visible light (400–550 nm). However, cyanocobalamin is stable under red light (wavelength >600 nm) exposure and does not convert to hydroxocobalamin. Our findings suggests that the intensity of pink color depends on concentrations of both free sulfhydryl groups on reduced mAb and hydroxocobalamin, the active form of vitamin B12. Both reactants are necessary and neither one of them is sufficient to generate pink color, therefore process control strategy can consider limiting either one or both factors. A process control strategy to install red light (wavelength >600 nm) in culture media preparation, storage and culture processing areas is proposed to provide safe light for biologics and to prevent light‐induced color variations in final products.


Biotechnology and Bioengineering | 2018

Contributions of Depth Filter Components to Protein Adsorption in Protein Bioprocessing

Ohnmar Khanal; Nripen Singh; Steven J. Traylor; Xuankuo Xu; Sanchayita Ghose; Zheng Jian Li; Abraham M. Lenhoff

Depth filtration is widely used in downstream bioprocessing to remove particulate contaminants via depth straining and is therefore applied to harvest clarification and other processing steps. However, depth filtration also removes proteins via adsorption, which can contribute variously to impurity clearance and to reduction in product yield. The adsorption may occur on the different components of the depth filter, i.e., filter aid, binder and cellulose filter. We measured adsorption of several model proteins and therapeutic proteins onto filter aids, cellulose and commercial depth filters at pH 5-8 and ionic strengths < 50 mM and correlated the adsorption data to bulk measured properties such as surface area, morphology, surface charge density and composition. We also explored the role of each depth filter component in the adsorption of proteins with different net charges, using confocal microscopy. Our findings show that a complete depth filters maximum adsorptive capacity for proteins can be estimated by its protein monolayer coverage values, which are of order mg/m2 , depending on the protein size. Furthermore, the extent of adsorption of different proteins appears to depend on the nature of the resin binder and its extent of coating over the depth filter surface, particularly in masking the cation-exchanger-like capacity of the siliceous filter aids. In addition to guiding improved depth filter selection, the findings can be leveraged in inspiring a more intentional selection of components and design of depth filter construction, for particular impurity removal targets. This article is protected by copyright. All rights reserved.


Biotechnology Progress | 2017

pH Variations during Diafiltration due to Buffer Non-Idealities

Youngbin Baek; Deyu Yang; Nripen Singh; Abhiram Arunkumar; Sanchayita Ghose; Zheng Jian Li; Andrew L. Zydney

Diafiltration is used for final formulation of essentially all biotherapeutics. Several studies have demonstrated that buffer/excipient concentrations in the final diafiltered product can be different than that in the diafiltration buffer due to interactions between buffer species and the protein product. However, recent work in our lab has shown variations in solution pH that are largely independent of the protein concentration during the first few diavolumes. Our hypothesis is that these pH variations are due to nonidealities in the acid‐base equilibrium coefficient. A model was developed for the diafiltration process accounting for the ionic strength dependence of the pKa. Experimental results obtained using phosphate and histidine buffers were in excellent agreement with model predictions. A decrease in ionic strength leads to an increase in the pKa for the phosphate buffer, causing a shift in the solution pH, even under conditions where the initial feed and the diafiltration buffer are at the same pH. This effect could be eliminated by matching the ionic strength of the feed and diafiltration buffer. The experimental data and model provide new insights into the factors controlling the pH profile during diafiltration processes.


mAbs | 2018

Using hydrogen peroxide to prevent antibody disulfide bond reduction during manufacturing process

Cheng Du; Yunping Huang; Ameya Borwankar; Zhijun Tan; Anthony Cura; Joon Chong Yee; Nripen Singh; Richard Ludwig; Michael C. Borys; Sanchayita Ghose; Nesredin Mussa; Zheng Jian Li

ABSTRACT During large-scale monoclonal antibody manufacturing, disulfide bond reduction of antibodies, which results in generation of low molecule weight species, is occasionally observed. When this happens, the drug substance does not meet specifications. Many investigations have been conducted across the biopharmaceutical industry to identify the root causes, and multiple strategies have been proposed to mitigate the problem. The reduction is correlated with the release of cellular reducing components and depletion of dissolved oxygen before, during, and after harvest. Consequently, these factors can lead to disulfide reduction over long-duration storage at room temperature prior to Protein A chromatography. Several strategies have been developed to minimize antibody reduction, including chemical inhibition of reducing components, maintaining aeration before and after harvest, and chilling clarified harvest during holding. Here, we explore the use of hydrogen peroxide in clarified harvest bulk or cell culture fluid as a strategy to prevent disulfide reduction. A lab-scale study was performed to demonstrate the effectiveness of hydrogen peroxide in preventing antibody reduction using multiple IgG molecules. Studies were done to define the optimal concentration of hydrogen peroxide needed to avoid unnecessary oxidization of the antibody products. We show that adding a controlled amount of hydrogen peroxide does not change product quality attributes of the protein. Since hydrogen peroxide is soluble in aqueous solutions and decomposes into water and oxygen, there is no additional burden involved in removing it during the downstream purification steps. Due to its ease of use and minimal product impact, we demonstrate that hydrogen peroxide treatment is a powerful, simple tool to quench reducing potential by simply mixing it with harvested cell culture fluid.

Collaboration


Dive into the Nripen Singh's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew L. Zydney

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Cheng Du

Bristol-Myers Squibb

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Youngbin Baek

Pennsylvania State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge