SwePub
Sök i SwePub databas

  Extended search

Träfflista för sökning "WFRF:(Gomis Fons Joaquín) "

Search: WFRF:(Gomis Fons Joaquín)

  • Result 1-10 of 19
Sort/group result
   
EnumerationReferenceCoverFind
1.
  • Andersson, Niklas, et al. (author)
  • Methodology for fast development of digital solutions in integrated continuous downstream processing
  • In: Biotechnology and Bioengineering. - 0006-3592.
  • Journal article (peer-reviewed)abstract
    • The methodology for production of biologics is going through a paradigm shift from batch-wise operation to continuous production. Lot of efforts are focused on integration, intensification, and continuous operation for decreased foot-print, material, equipment, and increased productivity and product quality. These integrated continuous processes with on-line analytics become complex processes, which requires automation, monitoring, and control of the operation, even unmanned or remote, which means bioprocesses with high level of automation or even autonomous capabilities. The development of these digital solutions becomes an important part of the process development and needs to be assessed early in the development chain. This work discusses a platform that allows fast development, advanced studies, and validation of digital solutions for integrated continuous downstream processes. It uses an open, flexible, and extendable real-time supervisory controller, called Orbit, developed in Python. Orbit makes it possible to communicate with a set of different physical setups and on the same time perform real-time execution. Integrated continuous processing often implies parallel operation of several setups and network of Orbit controllers makes it possible to synchronize complex process system. Data handling, storage, and analysis are important properties for handling heterogeneous and asynchronous data generated in complex downstream systems. Digital twin applications, such as advanced model-based and plant-wide monitoring and control, are exemplified using computational extensions in Orbit, exploiting data and models. Examples of novel digital solutions in integrated downstream processes are automatic operation parameter optimization, Kalman filter monitoring, and model-based batch-to-batch control.
  •  
2.
  • Andersson, Niklas, et al. (author)
  • Smart platform for development of small-scale integrated continuous downstream processes
  • 2022
  • In: Advances in Chemical Engineering. - : Elsevier. - 0065-2377. ; 59:1, s. 131-158
  • Book chapter (peer-reviewed)abstract
    • This chapter presents and discuss a platform for small-scale drug candidate production, process development and performance studies of integrated continuous downstream processes. The main idea of the platform methodology is to use common available equipment and reconfigure them for advanced downstream processing. Five different industrial case studies are discussed. The resulting processes are automated and controlled by the external supervisory controller, called Orbit. Orbit communicate with the local equipment control system, resulting in a minimum of adjustment in the laboratory infrastructure. Orbit is implemented in python, making it an open, flexible, and extendable control system. Orbit give support for integration of multiple chromatography columns, operate downstream processes on multiple parallel setups, integrated online analytics, use of advanced feedback control and batch-to-batch control. This functionality is presented in ten examples. The platform has successful been used in number of industrial case studies, particular in early drug and process development and for efficient small-scale drug candidate production.
  •  
3.
  •  
4.
  • Gomis Fons, Joaquin (author)
  • Design and Control of Integrated Continuous Processes for the Purification of Biopharmaceuticals
  • 2022
  • Doctoral thesis (other academic/artistic)abstract
    • The production of biopharmaceuticals so far has been based on batch processes that are robust and well-known, but very inefficient and inflexible, causing the products to be very expensive and process development to be slow and costly. The production of biopharmaceuticals is thus changing towards integrated continuous biomanufacturing (ICB) in order to reduce costs and increase flexibility in a constantly changing market. Continuous processing has been successfully implemented in upstream operation through the use of perfusion bioreactors, but the maturity required for the widespread use of ICB on commercial scale has not yet been achieved in integrated continuous downstream processes (ICDPs). The research presented in this thesis provides several tools for the design, optimization, control and scale-up of ICDPs for the purification of biopharmaceuticals with the aim of reducing the technological gap in downstream processing. The feasibility of implementing these processes in general platforms on laboratory and pilot scale has also been demonstrated. Process control and automation form a central part of the work presented in this thesis, including the development of several control strategies such as controlling the loading phase in the chromatography column, optimal product pooling in the elution phase, and adjusting and monitoring the pressure in an ultrafiltration process. In addition, existing research software has been further developed to enable automation in a number of different applications.The implementation of an ICDP requires a specific design approach to enable process integration and continuity of the feed from the upstream process. Several design equations were used for process integration. Feed continuity was achieved by employing periodic multi-column chromatography in the capture step. Process scheduling is therefore very important in this case, as the cycle time must be matched to the product recovery time. The effects of different integration approaches on process scheduling, and thus the overall productivity, was studied. Periodic multi-column chromatography not only allows for a continuous feed, but can also lead to increased productivity and resin utilization, as in the case of the periodic counter-current chromatography (PCC) process described in Paper III, where model-based optimization was performed. Another tool used to increase process efficiency in a downstream process was model-aided flow programming (Paper V), where a variable flow rate was used in the loading phase to achieve higher productivity and resin utilization.The feasibility of ICDPs was demonstrated by implementing them in different applications. Chromatography and ultrafiltration technologies were integrated in a single system (Paper I), and a complete ICB process was developed for the production of monoclonal antibodies (mAbs) (Paper II). Paper III describes the integration of a 3-column PCC capture step in a downstream process for the purification of mAbs. Continuous solvent/detergent-based virus inactivation and continuous capture were combined in an ICDP (Paper IV). The downstream processes described in Papers II and III were further developed to allow for the purification of pH-sensitive mAbs (Paper VI), and this process was coupled with the upstream system and run on pilot scale, demonstrating its feasibility on a larger scale (Paper VII).The results of this research show that ICDPs outperform traditional manual batch downstream processes. Automation, integration and continuous biomanufacturing lead to higher productivity, shorter process time, more rapid development of biopharmaceuticals, and lower investment costs.
  •  
5.
  • Gomis-Fons, Joaquín, et al. (author)
  • Integration of a complete downstream process for the automated lab-scale production of a recombinant protein
  • 2019
  • In: Journal of Biotechnology. - : Elsevier BV. - 0168-1656. ; 301, s. 45-51
  • Journal article (peer-reviewed)abstract
    • In this work, an automated downstream process for the purification and formulation of a recombinant protein was integrated at lab scale in a single chromatography unit. The purification chain consists of three bind-and-elute chromatography columns, a flow-through membrane chromatography step, and a final ultrafiltration-diafiltration (UFDF) step to concentrate and formulate the sample. An integrated downstream process increases productivity and decreases process time and hold-up volume. In addition, the automation of the process allows reducing the manual work and increases reproducibility. To integrate the downstream steps, all the intermediate tanks are removed, and the eluate of a column is loaded directly onto the next one. This makes it necessary to design the process in order to minimize the column volumes and the process time. A research software called Orbit was used to automate the purification process and implement a UFDF step in the chromatography unit. The whole downstream sequence was successfully implemented at lab scale, getting a pure concentrated and formulated product with a productivity of 1.09 mg mL−1 h−1, achieving a time reduction from almost two to one working day, while getting a similar yield and purity. Regarding the UFDF operation, the sample was concentrated 10 times, and 97% of the old buffer was exchanged by the formulation buffer with a sequential diafiltration.
  •  
6.
  • Gomis-Fons, Joaquin, et al. (author)
  • Mechanistic modeling of empty-full separation in recombinant adeno-associated virus production using anion-exchange membrane chromatography
  • 2024
  • In: Biotechnology and Bioengineering. - 0006-3592. ; 121:2, s. 719-734
  • Journal article (peer-reviewed)abstract
    • Recombinant adeno-associated viral vectors (rAAVs) have become an industry-standard technology in the field of gene therapy, but there are still challenges to be addressed in their biomanufacturing. One of the biggest challenges is the removal of capsid species other than that which contains the gene of interest. In this work, we develop a mechanistic model for the removal of empty capsids—those that contain no genetic material—and enrichment of full rAAV using anion-exchange membrane chromatography. The mechanistic model was calibrated using linear gradient experiments, resulting in good agreement with the experimental data. The model was then applied to optimize the purification process through maximization of yield studying the impact of mobile phase salt concentration and pH, isocratic wash and elution length, flow rate, percent full (purity) requirement, loading density (challenge), and the use of single-step or two-step elution modes. A solution from the optimization with purity of 90% and recovery yield of 84% was selected and successfully validated, as the model could predict the recovery yield with remarkable fidelity and was able to find process conditions that led to significant enrichment. This is, to the best of our knowledge, the first case study of the application of de novo mechanistic modeling for the enrichment of full capsids in rAAV manufacturing, and it serves as demonstration of the potential of mechanistic modeling in rAAV process development.
  •  
7.
  • Gomis-Fons, Joaquín, et al. (author)
  • Model-based design and control of a small-scale integrated continuous end-to-end mAb platform
  • 2020
  • In: Biotechnology progress (Print). - : John Wiley and Sons Inc.. - 8756-7938 .- 1520-6033.
  • Journal article (peer-reviewed)abstract
    • A continuous integrated bioprocess available from the earliest stages of process development allows for an easier, more efficient and faster development and characterization of an integrated process as well as production of small-scale drug candidates. The process presented in this article is a proof-of-concept of a continuous end-to-end monoclonal antibody production platform at a very small scale based on a 200 ml alternating tangential flow filtration perfusion bioreactor, integrated with the purification process with a model-based design and control. The downstream process, consisting of a periodic twin-column protein A capture, a virus inactivation, a CEX column and an AEX column, was compactly implemented in a single chromatography system, with a purification time of less than 4 hr. Monoclonal antibodies were produced for 17 days in a high cell density perfusion culture of CHO cells with titers up to 1.0 mg/ml. A digital twin of the downstream process was created by modelling all the chromatography steps. These models were used for real-time decision making by the implementation of control strategies to automatize and optimize the operation of the process. A consistent glycosylation pattern of the purified product was ensured by the steady state operation of the process. Regarding the removal of impurities, at least a 4-log reduction in the HCP levels was achieved. The recovery yield was up to 60%, and a maximum productivity of 0.8 mg/ml/day of purified product was obtained. 
  •  
8.
  • Gomis-Fons, Joaquin, et al. (author)
  • Optimal loading flow rate trajectory in monoclonal antibody capture chromatography
  • 2021
  • In: Journal of Chromatography A. - : Elsevier BV. - 0021-9673 .- 1873-3778. ; 1635
  • Journal article (peer-reviewed)abstract
    • In this paper, we determined the optimal flow rate trajectory during the loading phase of a mAb capture column. For this purpose, a multi-objective function was used, consisting of productivity and resin utilization. Several general types of trajectories were considered, and the optimal Pareto points were obtained for all of them. In particular, the presented trajectories include a constant-flow loading process as a nominal approach, a stepwise trajectory, and a linear trajectory. Selected trajectories were then applied in experiments with the state-of-the-art protein A resin mAb Select PrismA (TM), running in batch mode on a standard single-column chromatography setup, and using both a purified mAb solution as well as a clarified supernatant. The results show that this simple approach, programming the volumetric flow rate according to either of the explored strategies, can improve the process economics by increasing productivity by up to 12% and resin utilization by up to 9% compared to a constant-flow process, while obtaining a yield higher than 99%. The productivity values were similar to the ones obtained in a multi column continuous process, and ranged from 0.23 to 0.35 mg/min/mL resin. Additionally, it is shown that a model calibration carried out at constant flow can be applied in the simulation and optimization of flow trajectories. The selected processes were scaled up to pilot scale and simulated to prove that even higher productivity and resin utilization can be achieved at larger scales, and therefore confirm that the trajectories are generalizable across process scales for this resin.
  •  
9.
  • Gomis-Fons, Joaquin, et al. (author)
  • Optimization study on periodic counter-current chromatography integrated in a monoclonal antibody downstream process
  • 2020
  • In: Journal of Chromatography A. - : Elsevier. - 0021-9673 .- 1873-3778. ; 1621
  • Journal article (peer-reviewed)abstract
    • An optimization study of an integrated periodic counter-current chromatography (PCC) process in a monoclonal antibody (mAb) downstream process at lab scale, is presented in this paper. The optimization was based on a mechanistic model of the breakthrough curve in the protein-A capture step. Productivity and resin utilization were the objective functions, while yield during the loading of the capture column was set as a constraint. Different integration approaches were considered, and the effect of the feed concentration, yield and the protein-A resin was studied. The breakthrough curve and the length of the product recovery, which depended on the integration approach, determined the process scheduling. Several optimal Pareto solutions were obtained. At 0.5 mg mL−1 and 99% yield, a maximum productivity of 0.38 mg mL−1 min−1 with a resin utilization of 60% was obtained. On the other hand, the maximum resin utilization was 95% with a productivity of 0.10 mg mL−1 min−1. Due to the constraint of the process scheduling, a lower productivity could be achieved in the integration approaches with higher recovery time, which was more remarkable at higher concentrations. Therefore, it was shown that a holistic approach, where all the purification steps are considered in the process optimization, is needed to design a PCC in a downstream process.
  •  
10.
  • Isaksson, Madelène, et al. (author)
  • An automated buffer management system for small-scale continuous downstream bioprocessing
  • 2023
  • In: Journal of Chromatography A. - : Elsevier BV. - 0021-9673. ; 1695
  • Journal article (peer-reviewed)abstract
    • Buffer management for biopharmaceutical purification processes include buffer preparation, storage of buffers and restocking the buffers when needed. This is usually performed manually by the operators for small scale operations. However, buffer management can become a bottleneck when running integrated continuous purification processes for prolonged times, even at small scale. To address this issue, a buffer management system for the application in continuous lab-scale bioprocessing is presented in this paper. For this purpose, an ÄKTA™ explorer chromatography system was reconfigured to perform the buffer formulation. The system formulated all buffers from stock solutions and water according to pre-specified recipes. A digital twin of the physical system was introduced in the research software Orbit, written in python. Orbit was also used for full automation and control of the buffer system, which could run independently without operator input and handle buffer management for one or several connected buffer-consuming purification systems. The developed buffer management system performed automatic monitoring of buffer volumes, buffer order handling as well as buffer preparation and delivery. To demonstrate the capability of the developed system, it was integrated with a continuous downstream process and supplied all 9 required buffers to the process equipment during a 10-day operation. The buffer management system processed 55 orders and delivered 38 L of buffers, corresponding to 20% of its capacity. The pH and conductivity profiles observed during the purification steps were consistent across the cycles. The deviation in conductivity and pH from the measured average value was within ±0.89% in conductivity and ±0.045 in pH, well within the typical specification for buffer release, indicating that the prepared buffers had the correct composition. The operation of the developed buffer management system was robust and fully automated, and provides one solution to the buffer management bottleneck on lab scale for integrated continuous downstream bioprocessing.
  •  
Skapa referenser, mejla, bekava och länka
  • Result 1-10 of 19

Kungliga biblioteket hanterar dina personuppgifter i enlighet med EU:s dataskyddsförordning (2018), GDPR. Läs mer om hur det funkar här.
Så här hanterar KB dina uppgifter vid användning av denna tjänst.

 
pil uppåt Close

Copy and save the link in order to return to this view