Active pharmaceutical ingredient (API) production involving continuous processes--a process system engineering (PSE)-assisted design framework.

A systematic framework is proposed for the design of continuous pharmaceutical manufacturing processes. Specifically, the design framework focuses on organic chemistry based, active pharmaceutical ingredient (API) synthetic processes, but could potentially be extended to biocatalytic and fermentation-based products. The method exploits the synergic combination of continuous flow technologies (e.g., microfluidic techniques) and process systems engineering (PSE) methods and tools for faster process design and increased process understanding throughout the whole drug product and process development cycle. The design framework structures the many different and challenging design problems (e.g., solvent selection, reactor design, and design of separation and purification operations), driving the user from the initial drug discovery steps--where process knowledge is very limited--toward the detailed design and analysis. Examples from the literature of PSE methods and tools applied to pharmaceutical process design and novel pharmaceutical production technologies are provided along the text, assisting in the accumulation and interpretation of process knowledge. Different criteria are suggested for the selection of batch and continuous processes so that the whole design results in low capital and operational costs as well as low environmental footprint. The design framework has been applied to the retrofit of an existing batch-wise process used by H. Lundbeck A/S to produce an API: zuclopenthixol. Some of its batch operations were successfully converted into continuous mode, obtaining higher yields that allowed a significant simplification of the whole process. The material and environmental footprint of the process--evaluated through the process mass intensity index, that is, kg of material used per kg of product--was reduced to half of its initial value, with potential for further reduction. The case-study includes reaction steps typically used by the pharmaceutical industry featuring different characteristic reaction times, as well as L-L separation and distillation-based solvent exchange steps, and thus constitutes a good example of how the design framework can be useful to efficiently design novel or already existing API manufacturing processes taking advantage of continuous processes.

[1]  Norbert Kockmann,et al.  Scale-up concept of single-channel microreactors from process development to industrial production , 2011 .

[2]  L. T. Fan,et al.  Stochastic modeling of transient residence-time distributions during start-up , 1995 .

[3]  Boelo Schuur,et al.  Two-phase (bio)catalytic reactions in a table-top centrifugal contact separator. , 2008, Angewandte Chemie.

[4]  Krist V. Gernaey,et al.  A model-based systems approach to pharmaceutical product-process design and analysis , 2010 .

[5]  Klavs F Jensen,et al.  Integrated microreactors for reaction automation: new approaches to reaction development. , 2010, Annual review of analytical chemistry.

[6]  Rafiqul Gani,et al.  Design and Synthesis of Distillation Systems using a Driving Force Based Approach , 2004 .

[7]  Norbert Kockmann,et al.  Harsh Reaction Conditions in Continuous‐Flow Microreactors for Pharmaceutical Production , 2009 .

[8]  Kiyoshi Watanabe,et al.  Cyclopentyl Methyl Ether as a New and Alternative Process Solvent , 2007 .

[9]  Andrew G. Livingston,et al.  Nanofiltration membrane cascade for continuous solvent exchange , 2007 .

[10]  Rafiqul Gani,et al.  Solvents in organic synthesis: Replacement and multi-step reaction systems , 2008, Comput. Chem. Eng..

[11]  Krist V. Gernaey,et al.  Multienzyme-Catalyzed Processes: Next-Generation Biocatalysis , 2011 .

[12]  Axel Günther,et al.  A microfabricated gas-liquid segmented flow reactor for high-temperature synthesis: the case of CdSe quantum dots. , 2005, Angewandte Chemie.

[13]  Rafiqul Gani,et al.  Method for selection of solvents for promotion of organic reactions , 2005, Comput. Chem. Eng..

[14]  Timothy F. Jamison,et al.  Continuous flow multi-step organic synthesis , 2010 .

[15]  Gunther Kolb,et al.  Development of a Microrectification Apparatus for Analytical and Preparative Applications , 2012 .

[16]  T. Müller,et al.  Hydroamination: direct addition of amines to alkenes and alkynes. , 2008, Chemical reviews.

[17]  Ryan L Hartman,et al.  Microchemical systems for continuous-flow synthesis. , 2009, Lab on a chip.

[18]  Volker Hessel,et al.  Novel Process Windows – Gate to Maximizing Process Intensification via Flow Chemistry , 2009 .

[19]  Krist V. Gernaey,et al.  An ontological knowledge-based system for the selection of process monitoring and analysis tools , 2010, Comput. Chem. Eng..

[20]  K. Jensen Microreaction engineering * is small better? , 2001 .

[21]  Christodoulos A. Floudas,et al.  Optimization of complex reactor networks—I. Isothermal operation , 1990 .

[22]  Donald P. Visco,et al.  Computer-aided molecular design using the Signature molecular descriptor: Application to solvent selection , 2010, Comput. Chem. Eng..

[23]  Concepción Jiménez-González,et al.  Expanding GSK's solvent selection guide ― embedding sustainability into solvent selection starting at medicinal chemistry , 2011 .

[24]  Jonathan P. McMullen,et al.  Rapid Determination of Reaction Kinetics with an Automated Microfluidic System , 2011 .

[25]  A. Behr,et al.  New Developments in Chemical Engineering for the Production of Drug Substances , 2004 .

[26]  R. Gani,et al.  Group contribution based process flowsheet synthesis, design and modelling , 2005 .

[27]  David J. C. Constable,et al.  Perspective on Solvent Use in the Pharmaceutical Industry , 2007 .

[28]  Concepción Jiménez-González,et al.  Using the Right Green Yardstick: Why Process Mass Intensity Is Used in the Pharmaceutical Industry To Drive More Sustainable Processes , 2011 .

[29]  Gintaras V. Reklaitis,et al.  Ontological informatics infrastructure for pharmaceutical product development and manufacturing , 2006, Comput. Chem. Eng..

[30]  Claire S. Adjiman,et al.  Design of solvents for optimal reaction rate constants , 2007 .

[31]  Jean-Christophe Brunet,et al.  An expert system for solvent-based separation process synthesis , 1992 .

[32]  Krist V. Gernaey,et al.  A perspective on PSE in pharmaceutical process development and innovation , 2012, Comput. Chem. Eng..

[33]  J. Hartwig,et al.  Palladium-catalyzed hydroamination of 1,3-dienes: a colorimetric assay and enantioselective additions. , 2001, Journal of the American Chemical Society.

[34]  Klavs F. Jensen,et al.  Overcoming the Challenges of Solid Bridging and Constriction during Pd-Catalyzed C−N Bond Formation in Microreactors , 2010 .

[35]  Rafiqul Gani,et al.  Process intensification: A perspective on process synthesis , 2010 .

[36]  San Kiang,et al.  Model-Based Solvent Selection during Conceptual Process Design of a New Drug Manufacturing Process , 2009 .

[37]  Salvador García Muñoz,et al.  Handling uncertainty in the establishment of a design space for the manufacture of a pharmaceutical product , 2010, Comput. Chem. Eng..

[38]  Antonis C. Kokossis,et al.  Nonisothermal synthesis of homogeneous and multiphase reactor networks , 2000 .

[39]  Andreas Linninger,et al.  Pollution Prevention for Batch Pharmaceutical and Speciality Chemical Processes , 2005 .

[40]  H. S. Pordal,et al.  The role of computational fluid dynamics in the pharmaceutical industry , 2002 .

[41]  Ali Khademhosseini,et al.  Microfluidics for drug discovery and development: from target selection to product lifecycle management. , 2008, Drug discovery today.

[42]  Ryan L. Hartman,et al.  Deciding whether to go with the flow: evaluating the merits of flow reactors for synthesis. , 2011, Angewandte Chemie.

[43]  Dimitrios I. Gerogiorgis,et al.  Steady-State Optimization of a Continuous Pharmaceutical Process , 2009 .

[44]  Rafiqul Gani,et al.  SEPARATION PROCESS DESIGN AND SYNTHESIS BASED ON THERMODYNAMIC INSIGHTS , 1995 .

[45]  J. Hartwig Development of catalysts for the hydroamination of olefins , 2004 .

[46]  Jeffrey J. Siirola,et al.  Process synthesis prospective , 2004, Comput. Chem. Eng..

[47]  David F. Aycock,et al.  Solvent Applications of 2-Methyltetrahydrofuran in Organometallic and Biphasic Reactions , 2007 .

[48]  Krist V. Gernaey,et al.  A retrofit strategy to achieve “Fast, Flexible, Future (F3)” pharmaceutical production processes , 2011 .

[49]  Rafiqul Gani,et al.  Design of sustainable chemical processes: Systematic retrofit analysis generation and evaluation of alternatives , 2008 .

[50]  Ryan L Hartman,et al.  Distillation in microchemical systems using capillary forces and segmented flow. , 2009, Lab on a chip.

[51]  Dominique M. Roberge,et al.  An Integrated Approach Combining Reaction Engineering and Design of Experiments for Optimizing Reactions , 2004 .

[52]  Gürkan Sin,et al.  Integration of process design and controller design for chemical processes using model-based methodology , 2010, Comput. Chem. Eng..

[53]  K. Plumb,et al.  Continuous Processing in the Pharmaceutical Industry: Changing the Mind Set , 2005 .

[54]  Dominique M. Roberge,et al.  Development of an Industrial Multi‐Injection Microreactor for Fast and Exothermic Reactions – Part II , 2008 .

[55]  N. Tamimi,et al.  Drug Development: From Concept to Marketing! , 2009, Nephron Clinical Practice.

[56]  Rafiqul Gani,et al.  A Modern Approach to Solvent Selection , 2006 .

[57]  Dominique M. Roberge,et al.  Continuous Multi‐Injection Reactor for Multipurpose Production – Part I , 2008 .

[58]  Rafiqul Gani,et al.  Phenomena-based Process Synthesis and Design to achieve Process Intensification , 2011 .

[59]  Krist V. Gernaey,et al.  Continuous Hydrolysis and Liquid–Liquid Phase Separation of an Active Pharmaceutical Ingredient Intermediate Using a Miniscale Hydrophobic Membrane Separator , 2012 .

[60]  K. Gernaey,et al.  Monitoring and Control of a Continuous Grignard Reaction for the Synthesis of an Active Pharmaceutical Ingredient Intermediate Using Inline NIR spectroscopy , 2012 .

[61]  D. Agar,et al.  Design and Control Techniques for the Numbering-up of Capillary Microreactors with Uniform Multiphase Flow Distribution , 2010 .

[62]  K. Christensen,et al.  Design and operation of a filter reactor for continuous production of a selected pharmaceutical intermediate , 2012 .

[63]  Jonathan P. McMullen,et al.  An Automated Microfluidic System for Online Optimization in Chemical Synthesis , 2010 .

[64]  N. Kockmann,et al.  Microreactor Technology and Continuous Processes in the Fine Chemical and Pharmaceutical Industry: Is the Revolution Underway? , 2008 .

[65]  Dominique M. Roberge,et al.  Microreactor Technology: A Revolution for the Fine Chemical and Pharmaceutical Industries? , 2005 .

[66]  James Daniel,et al.  Batch Processing Industries , 2005 .

[67]  Jie Chen,et al.  Computer-Aided Solvent Selection for Improving the Morphology of Needle-like Crystals: A Case Study of 2,6-Dihydroxybenzoic Acid , 2010 .

[68]  G. Silverman,et al.  Handbook of Grignard Reagents , 1996 .

[69]  Dimitrios I. Gerogiorgis,et al.  Economic Analysis of Integrated Continuous and Batch Pharmaceutical Manufacturing: A Case Study , 2011 .

[70]  Babatunde A. Ogunnaike,et al.  Integrating systems design and control using dynamic flexibility analysis , 2007 .

[71]  K. Jensen,et al.  Integrated continuous microfluidic liquid-liquid extraction. , 2007, Lab on a chip.

[72]  Sandro Macchietto,et al.  Model-based design of experiments for parameter precision: State of the art , 2008 .

[73]  H. Noorman,et al.  Key Green Engineering Research Areas for Sustainable Manufacturing: A Perspective from Pharmaceutical and Fine Chemicals Manufacturers , 2011 .

[74]  D. Glasser,et al.  The attainable region and optimal reactor structures , 1990 .

[75]  Konrad Hungerbühler,et al.  Life cycle assessment of fine chemical production: a case study of pharmaceutical synthesis , 2010 .

[76]  Krist V. Gernaey,et al.  ICAS-PAT: A software for design, analysis and validation of PAT systems , 2010, Comput. Chem. Eng..