Elsevier

Computers & Chemical Engineering

Volume 51, 5 April 2013, Pages 157-171
Computers & Chemical Engineering

Process systems engineering tools in the pharmaceutical industry

https://doi.org/10.1016/j.compchemeng.2012.06.014Get rights and content

Abstract

The purpose of this paper is to provide a summary of the current state of the application of process systems engineering tools in the pharmaceutical industry. In this paper, we present the compiled results of an industrial questionnaire submitted to pharmaceutical industry professionals. The topics covered in the questionnaire include process analytics, process monitoring, plant-wide information systems, unit operation modeling, quality control, and process optimization. A futuristic view of what process systems engineering tools will enable the pharmaceutical industry will be also be presented. While the industry is regularly using the traditional Design of Experiments approach to identify key parameters and to define control spaces, these approaches result in passive control strategies that do not attempt to compensate for disturbances. Special new approaches are needed for batch processes due to their essential dependence on time-varying conditions. Lastly, we briefly describe a novel data driven modeling approach, called Design of Dynamic Experiments that enables the optimization of batch processes with respect to time-varying conditions through an example of a simulated chemical reaction process. Many more approaches of this type are needed for the calculation of the design and control spaces of the process, and the effective design of feedback systems.

Highlights

► Process systems engineering tools in the pharmaceutical industry are summarized. ► A futuristic view of process systems engineering tools usage is presented. ► Traditional approaches for control spaces definition result in passive control. ► A novel data driven modeling approach Dynamic Design of Experiments is presented. ► This novel approach enables batch modeling with time-varying process conditions.

Introduction

Almost a decade has elapsed since the FDA publication “Pharmaceutical cGMPs for the 21st Century: A Risk-Based Approach” and almost eight years since “PAT – A Framework for innovative Pharmaceutical Manufacturing and Quality Assurance” were issued. Much progress and innovation in pharmaceutical manufacturing has occurred since the publication of these landmark documents. For example, pharmaceutical companies have readily adopted in-process measurements systems, such as near infrared spectroscopy for concentration, and focused beam reflectance measurements for estimation of particle size distribution. The application of multivariate process monitoring for real time fault detection and isolation has also found its way into pharmaceutical manufacturing. The industry has moved away from quality control strategies based on uni-variate parameters specifications, and towards the multivariate design space approach. While, tremendous progress has been achieved in the decade, there is work to be done to realize the full potential of the process systems engineering (PSE) toolbox.

The purpose of the paper is to describe the current state of the art of the application of PSE tools in the pharmaceutical industry. The sub areas of PSE discussed in this work are process analytical technology (PAT) measurement systems, process monitoring, plant wide information technology systems, process control, modeling, and optimization methodologies. This paper focuses on PSE applications primarily related to active pharmaceutical ingredient (API), and solid oral dosage manufacturing. Details on the application of PAT measurement systems, and process control in biologics are out of scope of this work, for readers interested in biologics PSE applications we are listing a few relevant review papers (Junker and Wang, 2006, Schugerl, 2001).

To augment information available in the open literature, we conducted an industrial benchmarking survey on the above-mentioned PSE sub areas that contained twenty-one questions in total1. The survey was submitted to current pharmaceutical industry professionals in all areas of the industry: active pharmaceutical ingredient, solid oral dosage, and biologics, in both development and manufacturing. The companies that submitted responses to the survey are listed in Table 1.

The paper is organized as follows, for each of the sub areas of PSE covered; we provide a brief background on how PSE tools are currently used in the pharmaceutical industry. Where possible, literature references have been provided, with a preference towards papers published by pharmaceutical industry professionals. The questionnaire results pertaining to each PSE area are presented at the end of each section. We then discuss the impact of increased out-sourcing of product development and manufacturing and the prospect of continuous processes on the future utilization of PSE tools. Lastly, we present the application of a novel method for batch process optimization called dynamic design of experiments. A simulated API synthesis reaction process is used to explain the method.

Section snippets

The current state of PSE tools in pharma

We describe the current state of the utilization PSE tools in the pharmaceutical industry. The sub areas of PSE discussed are measurement systems, multivariate process monitoring, plant wide information systems, and process control and optimization methodologies. The results of this section are a combination of work documented in the literature by authors in the pharmaceutical industry, and the results of the industrial benchmarking survey.

The future of pharmaceutical manufacturing and PSE tools

In this section, we briefly discuss the future trends of contract manufacturing and continuous processing in the pharmaceutical industry and their impact on the utilization and advancement of PSE tools.

The need for data-driven models

The inner workings of the majority of batch pharmaceutical processes are not well understood for a fundamental or knowledge-driven (KD) model to be developed. An additional roadblock in the development of such models is the small production rates of the majority of pharmaceutical products compared to the production rate of bulk chemical and petrochemicals for which a plethora of knowledge-driven models has found extensive use over the last four to six decades. Because, such KD models provide a

A new approach: Design of Dynamic Experiments

In an effort to develop a data-driven approach for the optimization of the end-result of a batch process unit with respect a time-evolving decision variable, Georgakis (2009) generalized the classical Design of Experiments (DoE) with respect to time-varying decision variables. Examples of such time-varying decision variables are the temperature of a batch reactor, the cooling rate of a crystallizer, or the feeding rate of the nutrient in a fed-batch fermentation unit. A set of experiments is

Conclusions

In this paper, we summarized the state of the art of the utilization of PSE tools in the pharmaceutical industry and tried to glance a bit into the future. We have presented the results of an industrial benchmarking survey, and discussed the projected impacts of out-sourcing and the rise of continuous manufacturing on PSE tool advancement. We hope to have motivated the audience for the greater need of data-driven rather than knowledge-driven models, suitable for quick deployment in process

Acknowledgements

The authors would like the thank the following individuals for providing completed questionnaires: Hector Guzman, Daniel Patience, Partha Mudipalli, Roger Bakale, Steve Mehrman, Becky Taillon, Trevor Wigle, Dan Dobry, Dafni Bika, Sze Wing Wong, Gert Thurau, Koji Muteki, and Martin Warman.

References (90)

  • G. Fevotte et al.

    Applications of NIR spectroscopy to monitoring and analyzing the solid state during industrial crystallization processes

    International Journal of Pharmaceutics

    (2004)
  • P. Frake et al.

    Process control and end-point determination of a fluid bed granulation by application of near infra-red spectroscopy

    International Journal of Pharmaceutics

    (1997)
  • M. Fransson et al.

    Real-time alignment of batch process data using COW for on-line process monitoring

    Chemometrics and Intelligent Laboratory Systems

    (2006)
  • S. Garcia-Munoz et al.

    Handling uncertainty in the establishment of a design space for the manufacture of a pharmaceutical product

    Computers & Chemical Engineering

    (2010)
  • S. Garcia-Munoz et al.

    Experiences in batch trajectory alignment for pharmaceutical process improvement through multivariate latent variable modeling

    Journal of Process Control

    (2011)
  • C. Georgakis et al.

    On the operability of continuous processes

    Control Engineering Practice

    (2003)
  • M. Golshan et al.

    Latent variable model predictive control (LV-MPC) for trajectory tracking in batch processes

    Journal of Process Control

    (2010)
  • A. Gupta et al.

    Nondestructive measurements of the compact strength and the particle-size distribution after milling of roller compacted powders by near-infrared spectroscopy

    Journal of Pharmaceutical Sciences

    (2004)
  • J. Hilden et al.

    Prediction of roller compacted ribbon solid fraction fro quality by design development

    Powder Technology

    (2011)
  • X. Hu et al.

    Study growth kinetics in fluidized bed granulation with at-line FBRM

    International Journal of Pharmaceutics

    (2008)
  • W.F. Ku et al.

    Disturbance detection and isolation by dynamic principal component analysis

    Chemometrics and Intelligent Laboratory Systems

    (1995)
  • C.K. Lai et al.

    Application of a fluorescence sensor for miniscale on-line monitoring of powder mixing kinetics

    Journal of Pharmaceutical Sciences

    (2004)
  • H. Leuenberger et al.

    Manufacturing pharmaceutical granules: Is the granulation end-point a myth?

    Powder Technology

    (2009)
  • F. Lewiner et al.

    On-line ATR FTIR measurement of supersaturation during solution crystallization processes. Calibration and applications on three solute/solvent systems

    Chemical Engineering Science

    (2001)
  • H. Lim et al.

    Assessment of the critical factors affecting the porosity of roller compacted ribbons and the feasibility of using NIR chemical imaging to evaluate the porosity distribution

    International Journal of Pharmaceutics

    (2011)
  • F.V. Lima et al.

    Design of output constraints for model-based non-square controllers using interval operability

    Journal of Process Control

    (2008)
  • F.V. Lima et al.

    Input–output operability of control systems: The steady-state case

    Journal of Process Control

    (2010)
  • F.V. Lima et al.

    Analysis of the constraint characteristics of a sheet forming control problem using interval operability concepts

  • J.J. Liu et al.

    Froth-based modeling and control of flotation processes

    Minerals Engineering

    (2008)
  • P. Luukkonen et al.

    Real-time assessment of granule and tablet properties using in-line data from a high-shear granulation process

    Journal of Pharmaceutical Sciences

    (2008)
  • J. Märk et al.

    Prospects for multivariate classification of a pharmaceutical intermediate with near-infrared spectroscopy as a process analytical technology (PAT) production control supplement

    European Journal of Pharmaceutics and Biopharmaceutics

    (2010)
  • S. Matero et al.

    Monitoring the wetting phase of fluidized bed granulation process using multi-way methods: The separation of successful from unsuccessful batches

    Chemometrics and Intelligent Laboratory Systems

    (2009)
  • Z.K. Nagy et al.

    Comparative performance of concentration and temperature controlled batch crystallizations

    Journal of Process Control

    (2008)
  • Z.K. Nagy et al.

    Modelling and control of combined cooling and antisolvent crystallization processes

    Journal of Process Control

    (2008)
  • A. Peinado et al.

    Development, validation and transfer of a near infrared method to determine in-line the end point of a fluidised drying process for commercial production batches of an approved oral solid dose pharmaceutical product

    Journal of Pharmaceutical and Biomedical Analysis

    (2011)
  • D.W.T. Rippin

    Simulation of a single- and multiproduct batch chemical plants for optimal design and operation

    Computers & Chemical Engineering

    (1983)
  • L. Saerens et al.

    Raman spectroscopy for the in-line polymer-drug quantification and solid state characterization during a pharmaceutical hot-melt extrusion process

    European Journal of Pharmaceutics and Biopharmaceutics

    (2011)
  • K. Schugerl

    Progress in monitoring, modeling and control of bioprocesses during the last 20 years

    Journal of Biotechnology

    (2001)
  • Z. Shi et al.

    Process characterization of powder blending by near-infrared spectroscopy: Blend end-points and beyond

    Journal of Pharmaceutical and Biomedical Analysis

    (2008)
  • E.T.S. Skibsted et al.

    Examples of NIR based real time release in tablet manufacturing

    Journal of Pharmaceutical and Biomedical Analysis

    (2007)
  • Y. Sulub et al.

    Near-infrared multivariate calibration updating using placebo: A content uniformity determination of pharmaceutical tablets

    Vibrational Spectroscopy

    (2008)
  • O. Svensson et al.

    Reaction monitoring using Raman spectroscopy and chemometrics

    Chemometrics and Intelligent Laboratory Systems

    (1999)
  • J.G. VanAntwerp et al.

    Cross-directional control of sheet and film processes

    Automatica

    (2007)
  • D.R. Vinson et al.

    A new measure of process output controllability

    Journal of Process Control

    (2000)
  • Y. Wang et al.

    In-linemonitoring of the thermaldegradation of poly(l-lactic acid) during melt extrusion by UV–vis spectroscopy

    Polymer

    (2008)
  • Cited by (79)

    • A review on the modernization of pharmaceutical development and manufacturing – Trends, perspectives, and the role of mathematical modeling

      2022, International Journal of Pharmaceutics
      Citation Excerpt :

      The ultimate aim of QbD is to achieve the implementation of Level 1 strategies, which are capable of guaranteeing quality assurance. The benefits of implementing a modern Level 1 control strategy are for both companies and patients, as stressed by regulators, practitioners and academics (Collins, 2018; Fisher et al., 2019; Troup and Georgakis, 2013; Yu et al., 2014). McKinsey (2021) estimates that the adoption of a smart quality control system for pharmaceutical process development and manufacturing could have a tangible impact on profit, reduce the product launch time by more than 30%, increase manufacturing and supply chain capacity and responsiveness by 20 to 30%, and prevent major compliance issues by reducing manual errors and variability.

    View all citing articles on Scopus
    View full text