Integrated Continuous Pharmaceutical Technologies—A Review
- András DomokosAndrás DomokosBudapest University of Technology and Economics, Organic Chemistry and Technology Department, H-1111 Budapest, HungaryMore by András Domokos,
- Brigitta NagyBrigitta NagyBudapest University of Technology and Economics, Organic Chemistry and Technology Department, H-1111 Budapest, HungaryMore by Brigitta Nagy,
- Botond SzilágyiBotond SzilágyiBudapest University of Technology and Economics, Faculty of Chemical Technology and Biotechnology, H-1111 Budapest, HungaryMore by Botond Szilágyi,
- György MarosiGyörgy MarosiBudapest University of Technology and Economics, Organic Chemistry and Technology Department, H-1111 Budapest, HungaryMore by György Marosi, and
- Zsombor Kristóf Nagy*Zsombor Kristóf Nagy*Telephone: +36-1-463-4129. Email: [email protected]. Postal address: 3 Műegyetem rkp., H-1111 Budapest, Hungary.Budapest University of Technology and Economics, Organic Chemistry and Technology Department, H-1111 Budapest, HungaryMore by Zsombor Kristóf Nagy
Abstract

Innovation in the pharmaceutical industry has been limited for a long time to the research and development of new active compounds; meanwhile, the structure of the production, dominated by batchwise technologies, has not changed to date. As has already been demonstrated in several other industrial sectors, continuous manufacturing (CM) has many advantages over batch processes. Faster, cheaper, and more flexible production can be developed with a significantly higher level of quality assurance. In the recent years the main regulatory agencies recognized the need for a change in drug production and started to promote continuous technologies and encourage pharmaceutical companies to develop and adapt such processes. As a result, by today extensive research was conducted in the various fields of pharmaceutical technologies from drug substance to drug product manufacturing. Many publications deal with synthetic steps carried out in flow reactors and crystallizations implemented in a continuous manner, and on the formulation side continuous filtration, drying, granulation, and blending have all been studied to a lesser or greater extent. Moreover, besides the modification of these traditional processes to continuous operation, novel, intrinsically continuous technologies are being studied as well. In order to entirely exploit the advantages of CM, the mainly separately developed processes need to be integrated to form end-to-end systems from the raw materials to the final dosage forms. However, even the integration of two technological steps is a challenging task. The development of end-to-end systems requires deep process understanding and a holistic approach toward process development and optimization. The aim of this work is to give insight into the state-of-the-art and new directions in integrated continuous pharmaceutical technologies by critically reviewing the recent literature of the broad field.
1. Introduction—Challenges of 21st Century Pharmaceutical Manufacturing
2. Advantages of Continuous Manufacturing
3. First Steps toward Continuous Pharmaceutical Manufacturing
4. Recent Progress in Continuous Pharmaceutical Technologies
4.1. Multistep Flow Synthesis of Pharmaceuticals
4.1.1. Principles of Flow Chemistry
Figure 1

Figure 1. (a) Traditional batchwise multistep synthesis and (b) continuous flow approach for the telescoped synthesis of complex molecules (adapted from ref (56)).
Figure 2

Figure 2. General structure of a typical continuous flow systems.
4.1.2. Flow Synthesis of Pharmaceuticals
API | Year of publication | Ref |
---|---|---|
Ibuprofen | 2009 | (90) |
Nabumetone | 2011 | (91) |
Fluoxetine | 2011 | (92) |
Artemisinin | 2012 | (93) |
Imatinib | 2013 | (94) |
Tamoxifen | 2013 | (95) |
Diphenhydramine HCl | 2013 | (96) |
Amitriptyline | 2013 | (97) |
Olanzapine | 2013 | (98) |
Rufinamide | 2014 | (99) |
LY2886721 | 2014 | (100) |
Aliskiren hemifumarate | 2014 | (101) |
Efavirenz | 2015 | (102) |
Rolipram + phenibut | 2015 | (103) |
Telmisartan | 2015 | (104) |
Ibuprofen | 2015 | (105) |
Ribociclib | 2016 | (106) |
Diphenhydramine HCl | 2016 | (1) |
Lidocaine HCl | 2016 | (1) |
Diazepam | 2016 | (1) |
Fluoxetine HCl | 2016 | (1) |
Pregabalin | 2017 | (107) |
Flucytosine | 2017 | (108) |
Captopril | 2017 | (109) |
Prexasertib | 2017 | (110) |
Clozapine | 2018 | (111) |
Hydroxychloroquine | 2018 | (112) |
Dolutegravir | 2018 | (113) |
16-DPA | 2018 | (114) |
Nicardipine HCl | 2018 | (115) |
Ciprofloxacin HCl | 2018 | (115) |
Neostigmine HCl | 2018 | (115) |
Rufinamide | 2018 | (115) |
Acetylsalicylic acid | 2018 | (116) |
Hymexazol | 2019 | (117) |
Vortioxetine | 2019 | (118) |
Flibanserin | 2019 | (119) |
Imatinib | 2019 | (120) |
Treprostinil | 2019 | (121) |
Ibuprofen | 2019 | (122) |
Lomustine | 2019 | (123) |
Linezolid | 2019 | (124) |
Lesinurad | 2020 | (125) |
4.1.3. API Flow Syntheses Integrated with Downstream Processes
4.2. Continuous Crystallization and Filtration
4.2.1. Continuous Crystallization
Figure 3

Figure 3. Schematic drawing of (a) a mixed suspension mixed product removal crystallizer and (b) a plug flow reactor.
4.2.2. Continuous Filtration
Figure 4

4.2.3. Integrated Continuous Crystallization and Filtration
4.3. Continuous Powder Blending and Tableting
Figure 5

Figure 5. (a) Schematic drawing and (b) photograph of a twin-screw continuous blender; and photographs of a (c) paddle and (d) a ribbon blender.
Figure 6

Figure 6. Different strategies for the production of final dosage forms after blending: (a) direct compression, (b) roller compaction followed by milling and another blending step, and (c) granulation, drying, milling, and blending again (adapted from ref (188)).
4.4. Novel Continuous Formulation Techniques
4.4.1. Hot-Melt Extrusion and Dropwise Additive Manufacturing
4.4.2. Electrospinning
Figure 7

Figure 7. Schematic representation of a single-needle electrospinning process, with the scanning electron microscopic picture of the electrospun material on the right.
4.5. End-to-End Continuous Production of Final Dosage Forms
Figure 8

Figure 8. Concept of end-to-end continuous pharmaceutical manufacturing from raw materials to final products with real-time quality control (adapted from ref (227)).
Figure 9

Figure 9. Examples for published continuous end-to-end manufacturing systems for the production of (a) heat-mold tablets of aliskiren hemifumarate (10) and (b) liquid dosage forms of nicardipine hydrochloride, ciprofloxacin hydrochloride, neostigmine hydrochloride, and rufinamide. (115) Reproduced with permissions from refs (10) and (115). Copyright 2013 John Wiley and Sons and Copyright 2018 John Wiley and Sons.
Figure 10

Figure 10. End-to-end production of acetylsalicylic acid-loaded electrospun orally dissolving web dosage forms. The API was synthesized in a 2-step process in flow microreactors and formulated into nanofibers by applying high voltage on the reaction mixture. Reproduced with permission from ref (116). Copyright 2018 Elsevier.
Figure 11

Figure 11. Feeding of MCC and ASA into the hopper of the continuous blender (left); NIR probe mounted above the belt conveyor after blending (middle); continuous blender, belt conveyor, and tableting machine integrated in one CM line (right) (adapted from ref (246)).
5. Conclusions
Acknowledgments
This work was supported from grants by National Research, Development and Innovation Office of Hungary (grant numbers: KH-129584, FK-132133, PD-121143). This work was also performed in the frame of FIEK_16-1-2016-0007 project, implemented with the support provided from the National Research, Development and Innovation Fund of Hungary, financed under the FIEK_16 funding scheme. The research reported in this paper and carried out at BME has been supported by the NRDI Fund (TKP2020 IES, Grant No. TKP2020 BME-IKA-VIZ) based on the charter of bolster issued by the NRDI Office under the auspices of the Ministry for Innovation and Technology, supported by the ÚNKP-20-3 New National Excellence Program of the Ministry for Innovation and Technology from the source of the National Research, Development and Innovation Fund.
References
This article references 247 other publications.
- 1Adamo, A.; Beingessner, R. L.; Behnam, M.; Chen, J.; Jamison, T. F.; Jensen, K. F.; Monbaliu, J.-C. M.; Myerson, A. S.; Revalor, E. M.; Snead, D. R.; Stelzer, T.; Weeranoppanant, N.; Wong, S. Y.; Zhang, P. On-Demand Continuous-Flow Production of Pharmaceuticals in a Compact Reconfigurable System. Science 2016, 352 (6281), 61– 67, DOI: 10.1126/science.aaf1337[Crossref], [PubMed], [CAS], Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XlsFSlsrY%253D&md5=5a700c0b3a9d5b7a57ff90e357b0a866On-demand continuous-flow production of pharmaceuticals in a compact, reconfigurable systemAdamo, Andrea; Beingessner, Rachel L.; Behnam, Mohsen; Chen, Jie; Jamison, Timothy F.; Jensen, Klavs F.; Monbaliu, Jean-Christophe M.; Myerson, Allan S.; Revalor, Eve M.; Snead, David R.; Stelzer, Torsten; Weeranoppanant, Nopphon; Wong, Shin Yee; Zhang, PingScience (Washington, DC, United States) (2016), 352 (6281), 61-67CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Pharmaceutical manufg. typically uses batch processing at multiple locations. Disadvantages of this approach include long prodn. times and the potential for supply chain disruptions. As a preliminary demonstration of an alternative approach, we report here the continuous-flow synthesis and formulation of active pharmaceutical ingredients in a compact, reconfigurable manufg. platform. Continuous end-to-end synthesis in the refrigerator-sized [1.0 m (width) × 0.7 m (length) × 1.8 m (height)] system produces sufficient quantities per day to supply hundreds to thousands of oral or topical liq. doses of diphenhydramine hydrochloride, lidocaine hydrochloride, diazepam, and fluoxetine hydrochloride that meet U.S. Pharmacopeia stds. Underlying this flexible plug-and-play approach are substantial enabling advances in continuous-flow synthesis, complex multistep sequence telescoping, reaction engineering equipment, and real-time formulation.
- 2Srai, J. S.; Badman, C.; Krumme, M.; Futran, M.; Johnston, C. Future Supply Chains Enabled by Continuous Processing-Opportunities and Challenges May 20–21, 2014 Continuous Manufacturing Symposium. J. Pharm. Sci. 2015, 104 (3), 840– 849, DOI: 10.1002/jps.24343[Crossref], [CAS], Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXislegt7k%253D&md5=f43251b3e04fdda54d2e0aad4c8fef5eFuture Supply Chains Enabled by Continuous Processing-Opportunities and Challenges. May 20-21, 2014 Continuous Manufacturing SymposiumSrai, Jagjit Singh; Badman, Clive; Krumme, Markus; Futran, Mauricio; Johnston, CraigJournal of Pharmaceutical Sciences (2015), 104 (3), 840-849CODEN: JPMSAE; ISSN:0022-3549. (John Wiley & Sons, Inc.)This paper examines the opportunities and challenges facing the pharmaceutical industry in moving to a primarily "continuous processing"-based supply chain. The current predominantly "large batch" and centralized manufg. system designed for the "blockbuster" drug has driven a slow-paced, inventory heavy operating model that is increasingly regarded as inflexible and unsustainable. Indeed, new markets and the rapidly evolving technol. landscape will drive more product variety, shorter product life-cycles, and smaller drug vols., which will exacerbate an already unsustainable economic model. Future supply chains will be required to enhance affordability and availability for patients and healthcare providers alike despite the increased product complexity. In this more challenging supply scenario, we examine the potential for a more pull driven, near real-time demand-based supply chain, utilizing continuous processing where appropriate as a key element of a more "flow-through" operating model. In this discussion paper on future supply chain models underpinned by developments in the continuous manuf. of pharmaceuticals, we have set out; The significant opportunities to moving to a supply chain flow-through operating model, with substantial opportunities in inventory redn., lead-time to patient, and radically different product assurance/stability regimes. Scenarios for decentralized prodn. models producing a greater variety of products with enhanced vol. flexibility. Prodn., supply, and value chain footprints that are radically different from today's monolithic and centralized batch manufg. operations. Clin. trial and drug product development cost savings that support more rapid scale-up and market entry models with early involvement of SC designers within New Product Development. The major supply chain and industrial transformational challenges that need to be addressed. The paper recognizes that although current batch operational performance in pharma is far from optimal and not necessarily an appropriate end-state benchmark for batch technol., the adoption of continuous supply chain operating models underpinned by continuous prodn. processing, as full or hybrid solns. in selected product supply chains, can support industry transformations to deliver right-first-time quality at substantially lower inventory profiles. © 2015 Wiley Periodicals, Inc. and the American Pharmacists Assocn. J Pharm Sci 104:840-849, 2015.
- 3Badman, C.; Trout, B. L. Achieving Continuous Manufacturing May 20–21 2014 Continuous Manufacturing Symposium. J. Pharm. Sci. 2015, 104 (3), 779– 780, DOI: 10.1002/jps.24246[Crossref], [CAS], Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitVyjt7rN&md5=e334687cd8c1653bceb7d91081815cceAchieving Continuous Manufacturing. May 20-21, 2014 Continuous SymposiumBadman, Clive; Trout, Bernhardt L.Journal of Pharmaceutical Sciences (2015), 104 (3), 779-780CODEN: JPMSAE; ISSN:0022-3549. (John Wiley & Sons, Inc.)There is no expanded citation for this reference.
- 4Plumb, K. Continuous Processing in the Pharmaceutical Industry: Changing the Mind Set. Chem. Eng. Res. Des. 2005, 83 (6), 730– 738, DOI: 10.1205/cherd.04359[Crossref], [CAS], Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXmvV2msbk%253D&md5=bb3f9bdcae245ed5ca83bed478d79eebContinuous processing in the pharmaceutical industry changing the mind setPlumb, K.Chemical Engineering Research and Design (2005), 83 (A6), 730-738CODEN: CERDEE; ISSN:0263-8762. (Institution of Chemical Engineers)The pharmaceutical industry is a highly regulated industry and all prodn. must be carried out in accordance with good manufg. practice. Traditionally, virtually all manufg. operations were carried out batch wise in spite of cost disadvantages and the fact that in many cases continuous processing could lead to the manuf. of purer products. The regulatory environment tends to stifle any attempts to change the process once the development stage is over and the product and process were licensed. This has created a mind set amongst the industry's professionals that batch processes are the only acceptable way forward. However, the regulatory authorities, particularly the Food and Drug Administration in America, have recognized that continuous processing has the potential to improve product quality and are encouraging the industry to reconsider their ideas. This paper examines how chem. engineers can use the opportunity that arises from this changed regulatory environment to revisit their own ideas and drive a change of mind set within the industry.
- 5Shah, N. Pharmaceutical Supply Chains: Key Issues and Strategies for Optimisation. Comput. Chem. Eng. 2004, 28, 929– 941. DOI: 10.1016/j.compchemeng.2003.09.022 .[Crossref], [CAS], Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXjtlyhtbc%253D&md5=a22efc63dd475bac126d242fea3018e5Pharmaceutical supply chains: key issues and strategies for optimisationShah, NilayComputers & Chemical Engineering (2004), 28 (6-7), 929-941CODEN: CCENDW; ISSN:0098-1354. (Elsevier)Supply chain optimization is now a major research theme in process operations and management. A great deal of research was undertaken on facility location and design, inventory and distribution planning, capacity and prodn. planning and detailed scheduling. Only a small proportion of this work directly addresses the issues faced in the pharmaceutical sector. This sector is much ready for and in need of sophisticated supply chain optimization techniques. At the supply chain design stage, a particular problem faced by this industry is the need to balance future capacity with anticipated demands in the face of the significant uncertainty that arises out of clin. trials and competitor activity. Efficient capacity utilization plans and robust infrastructure investment decisions will be important as regulatory pressures increase and margins are eroded. The ability to locate nodes of the supply chain in tax havens and optimize trading and transfer price structures results in interesting degrees of freedom in the supply chain design problem. Prior even to capacity planning comes the problem of pipeline and testing planning, where the selection of products for development and the scheduling of the development tasks requires a careful management of risk and potential rewards. At the operation stage, it is often difficult to ensure responsiveness. Most pharmaceutical products involve primary active ingredient (AI) prodn. (often multi-stage chem. synthesis or bioprocess) and secondary (formulation) prodn. Both of the stages are characterized by low manufg. velocities and are hampered by the need for quality assurance activities at several points. It is not unusual for the overall supply chain cycle time to be 300 days. In this environment, supply chain debottlenecking and decoupling strategies together with coordinated inventory management are crucial for quick responses to changing market trends. A good understanding of what actually drives the supply chain dynamics is also required. As often as not, erratic dynamics are introduced by business processes rather than by external demand, and may be eliminated by the re-design of internal business processes or supplier/customer relationships. This paper will consider important issues in supply chain design and operation drawn from the literature and from our collaborative research projects in this area. The main features of the problems will be reviewed as will the literature to date. Some strategies for soln. will be identified, as will some future research needs.
- 6Srai, J. S.; Harrington, T.; Alinaghian, L.; Phillips, M. Evaluating the Potential for the Continuous Processing of Pharmaceutical Products - A Supply Network Perspective. Chem. Eng. Process. 2015, 97, 248– 258, DOI: 10.1016/j.cep.2015.07.018[Crossref], [CAS], Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtlektr%252FK&md5=5d96828e4f01f667a8453d1c6eb802c4Evaluating the potential for the continuous processing of pharmaceutical products-a supply network perspectiveSrai, Jagjit Singh; Harrington, Tomas; Alinaghian, Leila; Phillips, MarkChemical Engineering and Processing (2015), 97 (), 248-258CODEN: CENPEU; ISSN:0255-2701. (Elsevier B.V.)This paper presents an approach to evaluating the potential supply chain benefits of adopting continuous processing technologies for a diverse set of pharmaceutical products. The approach integrates upstream 'continuous' processing considerations for the prodn. of active ingredients and final product formulation, with the downstream implications for packing and distribution. Currently, these upstream and downstream operations largely operate as decoupled operations with independent coordination and governance mechanisms, and the approach presented in this paper identifies opportunities for more case-specific integrated end-to-end supply chains enabled by continuous flow technologies. Three specific product (and corresponding processing technol.) case studies are used to demonstrate the utility of the approach in assessing the supply network and system integration opportunities that emerge from the continuous processing of pharmaceutical products.
- 7Yu, L. X. Pharmaceutical Quality by Design: Product and Process Development, Understanding, and Control. Pharm. Res. 2008, 25 (4), 781– 791, DOI: 10.1007/s11095-007-9511-1[Crossref], [PubMed], [CAS], Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXkt1Sntrg%253D&md5=d37ac3f9176369483bf7d7955a66d7c4Pharmaceutical Quality by Design: Product and Process Development, Understanding, and ControlYu, Lawrence X.Pharmaceutical Research (2008), 25 (4), 781-791CODEN: PHREEB; ISSN:0724-8741. (Springer)A review. Purpose. The purpose of this paper is to discuss the pharmaceutical Quality by Design (QbD) and describe how it can be used to ensure pharmaceutical quality. Materials and Methods. The QbD was described and some of its elements identified. Process parameters and quality attributes were identified for each unit operation during manuf. of solid oral dosage forms. The use of QbD was contrasted with the evaluation of product quality by testing alone. Results. The QbD is a systemic approach to pharmaceutical development. It means designing and developing formulations and manufg. processes to ensure predefined product quality. Some of the QbD elements include: Conclusions. Using QbD, pharmaceutical quality is assured by understanding and controlling formulation and manufg. variables. Product testing confirms the product quality. Implementation of QbD will enable transformation of the chem., manufg., and controls (CMC) review of abbreviated new drug applications (ANDAs) into a science-based pharmaceutical quality assessment.
- 8Ayati, N.; Saiyarsarai, P.; Nikfar, S. Short and Long Term Impacts of COVID-19 on the Pharmaceutical Sector. Daru, J. Pharm. Sci. 2020, 28, 799, DOI: 10.1007/s40199-020-00358-5[Crossref], [CAS], Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB38nnvVWrtw%253D%253D&md5=89e3ace8cef5239cd09616dbf10e35b0Short and long term impacts of COVID-19 on the pharmaceutical sectorAyati Nayyereh; Saiyarsarai Parisa; Nikfar Shekoufeh; Saiyarsarai Parisa; Nikfar Shekoufeh; Nikfar ShekoufehDaru : journal of Faculty of Pharmacy, Tehran University of Medical Sciences (2020), 28 (2), 799-805 ISSN:.BACKGROUND: The novel coronavirus disease 2019 (COVID-19) was characterized as a global pandemic by the WHO on March 11th, 2020. This pandemic had major effects on the health market, the pharmaceutical sector, and was associated with considerable impacts; which may appear in short and long-term time-horizon and need identification and appropriate planning to reduce their socio-economic burden. OBJECTIVES: Current short communication study assessed pharmaceutical market crisis during the COVID-19 era; discussing short- and long-term impacts of the pandemic on the pharmaceutical sector. RESULTS: Short-term impacts of COVID-19 pandemic includes demand changes, regulation revisions, research and development process changes and the shift towards tele-communication and tele-medicine. In addition, industry growth slow-down, approval delays, moving towards self-sufficiency in pharm-production supply chain and trend changes in consumption of health-market products along with ethical dilemma could be anticipated as long-term impacts of COVID-19 pandemic on pharmaceutical sector in both global and local levels. CONCLUSION: The pandemic of COVID-19 poses considerable crisis on the health markets, including the pharmaceutical sector; and identification of these effects, may guide policy-makers towards more evidence-informed planning to overcome accompanying challenges. Graphical abstract .
- 9Teżyk, M.; Milanowski, B.; Ernst, A.; Lulek, J. Recent Progress in Continuous and Semi-Continuous Processing of Solid Oral Dosage Forms: A Review. Drug Dev. Ind. Pharm. 2016, 42 (8), 1195– 1214, DOI: 10.3109/03639045.2015.1122607[Crossref], [PubMed], [CAS], Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXitVGgsb3I&md5=a577eb68307bb08570f919b2c3d7dc4cRecent progress in continuous and semi-continuous processing of solid oral dosage forms: a reviewTezyk, Michal; Milanowski, Bartlomiej; Ernst, Andrzej; Lulek, JaninaDrug Development and Industrial Pharmacy (2016), 42 (8), 1195-1214CODEN: DDIPD8; ISSN:0363-9045. (Taylor & Francis Ltd.)Continuous processing is an innovative prodn. concept well known and successfully used in other industries for many years. The modern pharmaceutical industry is facing the challenge of transition from a traditional manufg. approach based on batch-wise prodn. to a continuous manufg. model. The aim of this article is to present technol. progress in manufg. based on continuous and semi-continuous processing of the solid oral dosage forms. Single unit processes possessing an alternative processing pathway to batch-wise technol. or, with some modification, an altered approach that may run continuously, and are thus able to seamlessly switch to continuous manufg. are briefly presented. Furthermore, the concept of semi-continuous processing is discussed. Subsequently, more sophisticated prodn. systems created by coupling single unit processes and comprising all the steps of prodn., from powder to final dosage form, were reviewed. Finally, attempts of end-to-end prodn. approach, meaning the linking of continuous synthesis of API from intermediates with the prodn. of final dosage form, are described. There are a growing no. of scientific articles showing an increasing interest in changing the approach to the prodn. of pharmaceuticals in recent years. Numerous scientific publications are a source of information on the progress of knowledge and achievements of continuous processing. These works often deal with issues of how to modify or replace the unit processes in order to enable seamlessly switching them into continuous processing. A growing no. of research papers conc. on integrated continuous manufg. lines in which the prodn. concept of "from powder to tablet" is realized. Four main domains are under investigation: influence of process parameters on intermediates or final dosage forms properties, implementation of process anal. tools, control-managing system responsible for keeping continuous materials flow through the whole manufg. process and the development of new computational methods to assess or simulate these new manufg. techniques. The attempt to connect the primary and secondary prodn. steps proves that development of continuously operating lines is possible. A mind-set change is needed to be able to face, and fully assess, the advantages and disadvantages of switching from batch to continuous mode prodn.
- 10Mascia, S.; Heider, P. L.; Zhang, H.; Lakerveld, R.; Benyahia, B.; Barton, P. I.; Braatz, R. D.; Cooney, C. L.; Evans, J. M. B.; Jamison, T. F.; Jensen, K. F.; Myerson, A. S.; Trout, B. L. End-to-End Continuous Manufacturing of Pharmaceuticals: Integrated Synthesis, Purification, and Final Dosage Formation. Angew. Chem. 2013, 125 (47), 12585– 12589, DOI: 10.1002/ange.201305429
- 11Suresh, P.; Basu, P. K. Improving Pharmaceutical Product Development and Manufacturing: Impact on Cost of Drug Development and Cost of Goods Sold of Pharmaceuticals. J. Pharm. Innov. 2008, 3 (3), 175– 187, DOI: 10.1007/s12247-008-9043-1
- 12Food and Drug Administration. Guidance for Industry Changes to an Approved NDA or ANDA. FDA Off. Doc. 2004, No. April, 1– 40.Google ScholarThere is no corresponding record for this reference.
- 13Rantanen, J.; Khinast, J. The Future of Pharmaceutical Manufacturing Sciences. J. Pharm. Sci. 2015, 104 (11), 3612– 3638, DOI: 10.1002/jps.24594[Crossref], [PubMed], [CAS], Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtlCgsbbL&md5=47a5880f61b8c65f299cdbe821b148c7The Future of Pharmaceutical Manufacturing SciencesRantanen, Jukka; Khinast, JohannesJournal of Pharmaceutical Sciences (2015), 104 (11), 3612-3638CODEN: JPMSAE; ISSN:0022-3549. (John Wiley & Sons, Inc.)The entire pharmaceutical sector is in an urgent need of both innovative technol. solns. and fundamental scientific work, enabling the prodn. of highly engineered drug products. Com.-scale manufg. of complex drug delivery systems (DDSs) using the existing technologies is challenging. This review covers important elements of manufg. sciences, beginning with risk management strategies and design of expts. (DoE) techniques. Exptl. techniques should, where possible, be supported by computational approaches. With that regard, state-of-art mechanistic process modeling techniques are described in detail. Implementation of materials science tools paves the way to mol.-based processing of future DDSs. A snapshot of some of the existing tools is presented. Addnl., general engineering principles are discussed covering process measurement and process control solns. Last part of the review addresses future manufg. solns., covering continuous processing and, specifically, hot-melt processing and printing-based technologies. Finally, challenges related to implementing these technologies as a part of future health care systems are discussed. © 2015 The Authors. Journal of Pharmaceutical Sciences published by Wiley Periodicals, Inc. and the American Pharmacists Assocn. J Pharm Sci.
- 14Su, Q.; Ganesh, S.; Moreno, M.; Bommireddy, Y.; Gonzalez, M.; Reklaitis, G. V.; Nagy, Z. K. A Perspective on Quality-by-Control (QbC) in Pharmaceutical Continuous Manufacturing. Comput. Chem. Eng. 2019, 125, 216– 231, DOI: 10.1016/j.compchemeng.2019.03.001[Crossref], [CAS], Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXmtVGlsb0%253D&md5=177c7d1423c104cc570dc66cd4c1eb66A perspective on Quality-by-Control (QbC) in pharmaceutical continuous manufacturingSu, Qinglin; Ganesh, Sudarshan; Moreno, Mariana; Bommireddy, Yasasvi; Gonzalez, Marcial; Reklaitis, Gintaras V.; Nagy, Zoltan K.Computers & Chemical Engineering (2019), 125 (), 216-231CODEN: CCENDW; ISSN:0098-1354. (Elsevier B.V.)The Quality-by-Design (QbD) guidance issued by the US Food and Drug Administration (FDA) has catalyzed the modernization of pharmaceutical manufg. practices including the adoption of continuous manufg. Active process control was highlighted recently as a means to improve the QbD implementation. This advance has since been evolving into the concept of Quality-by-Control (QbC). In this study, the concept of QbC is discussed, including a definition of QbC, a review of the recent developments towards the QbC, and a perspective on the challenges of QbC implementation in continuous manufg. The QbC concept is demonstrated using a rotary tablet press, integrated into a pilot scale continuous direct compaction process. The results conclusively showed that active process control, based on product and process knowledge and advanced model-based techniques, including data reconciliation, model predictive control (MPC), and risk anal., is indispensable to comprehensive QbC implementation, and ensures robustness and efficiency.
- 15Lee, S. L.; O’Connor, T. F.; Yang, X.; Cruz, C. N.; Chatterjee, S.; Madurawe, R. D.; Moore, C. M. V; Yu, L. X.; Woodcock, J. Modernizing Pharmaceutical Manufacturing: From Batch to Continuous Production. J. Pharm. Innov. 2015, 10 (3), 191– 199, DOI: 10.1007/s12247-015-9215-8
- 16Food and Drug Administration. Drug Shortages: Root Causes and Potential Solutions. FDA Off. Doc. 2019Google ScholarThere is no corresponding record for this reference.
- 17Food and Drug Administration. Report on Drug Shortages for Calendar Year 2018. FDA Off. Doc. 2018Google ScholarThere is no corresponding record for this reference.
- 18DiMasi, J. A.; Grabowski, H. G.; Hansen, R. W. Innovation in the Pharmaceutical Industry: New Estimates of R&D Costs. J. Health Econ. 2016, 47, 20– 33, DOI: 10.1016/j.jhealeco.2016.01.012[Crossref], [PubMed], [CAS], Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC28jls1egsw%253D%253D&md5=a78649853406ddbfe54abb0052ca964bInnovation in the pharmaceutical industry: New estimates of R&D costsDiMasi Joseph A; Grabowski Henry G; Hansen Ronald WJournal of health economics (2016), 47 (), 20-33 ISSN:.The research and development costs of 106 randomly selected new drugs were obtained from a survey of 10 pharmaceutical firms. These data were used to estimate the average pre-tax cost of new drug and biologics development. The costs of compounds abandoned during testing were linked to the costs of compounds that obtained marketing approval. The estimated average out-of-pocket cost per approved new compound is $1395 million (2013 dollars). Capitalizing out-of-pocket costs to the point of marketing approval at a real discount rate of 10.5% yields a total pre-approval cost estimate of $2558 million (2013 dollars). When compared to the results of the previous study in this series, total capitalized costs were shown to have increased at an annual rate of 8.5% above general price inflation. Adding an estimate of post-approval R&D costs increases the cost estimate to $2870 million (2013 dollars).
- 19Nasr, M. M.; Krumme, M.; Matsuda, Y.; Trout, B. L.; Badman, C.; Mascia, S.; Cooney, C. L.; Jensen, K. D.; Florence, A.; Johnston, C.; Konstantinov, K.; Lee, S. L. Regulatory Perspectives on Continuous Pharmaceutical Manufacturing: Moving From Theory to Practice: September 26–27, 2016, International Symposium on the Continuous Manufacturing of Pharmaceuticals. J. Pharm. Sci. 2017, 106, 3199– 3206. DOI: 10.1016/j.xphs.2017.06.015 .[Crossref], [PubMed], [CAS], Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXht1yltrrM&md5=a13e536a7915f693ef044524235a0722Regulatory Perspectives on Continuous Pharmaceutical Manufacturing: Moving From Theory to PracticeNasr, Moheb M.; Krumme, Markus; Matsuda, Yoshihiro; Trout, Bernhardt L.; Badman, Clive; Mascia, Salvatore; Cooney, Charles L.; Jensen, Keith D.; Florence, Alastair; Johnston, Craig; Konstantinov, Konstantin; Lee, Sau L.Journal of Pharmaceutical Sciences (Philadelphia, PA, United States) (2017), 106 (11), 3199-3206CODEN: JPMSAE; ISSN:0022-3549. (Elsevier Inc.)Continuous manufg. plays a key role in enabling the modernization of pharmaceutical manufg. The fate of this emerging technol. will rely, in large part, on the regulatory implementation of this novel technol. This paper, which is based on the 2nd International Symposium on the Continuous Manufg. of Pharmaceuticals, describes not only the advances that have taken place since the first International Symposium on Continuous Manufg. of Pharmaceuticals in 2014, but the regulatory landscape that exists today. Key regulatory concepts including quality risk management, batch definition, control strategy, process monitoring and control, real-time release testing, data processing and management, and process validation/verification are outlined. Support from regulatory agencies, particularly in the form of the harmonization of regulatory expectations, will be crucial to the successful implementation of continuous manufg. Collaborative efforts, among academia, industry, and regulatory agencies, are the optimal soln. for ensuring a solid future for this promising manufg. technol.
- 20Byrn, S.; Futran, M.; Thomas, H.; Jayjock, E.; Maron, N.; Meyer, R. F.; Myerson, A. S.; Thien, M. P.; Trout, B. L. Achieving Continuous Manufacturing for Final Dosage Formation: Challenges and How to Meet Them May 20–21 2014 Continuous Manufacturing Symposium. J. Pharm. Sci. 2015, 104 (3), 792– 802, DOI: 10.1002/jps.24247[Crossref], [CAS], Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitVyjt7rO&md5=c2af540de6ad5f42eb62e2f7b328053aAchieving Continuous Manufacturing for Final Dosage Formation: Challenges and How to Meet Them. May 20-21, 2014 Continuous SymposiumByrn, Stephen; Futran, Maricio; Thomas, Hayden; Jayjock, Eric; Maron, Nicola; Meyer, Robert F.; Myerson, Allan S.; Thien, Michael P.; Trout, Bernhardt L.Journal of Pharmaceutical Sciences (2015), 104 (3), 792-802CODEN: JPMSAE; ISSN:0022-3549. (John Wiley & Sons, Inc.)We describe the key issues and possibilities for continuous final dosage formation, otherwise known as downstream processing or drug product manufg. A distinction is made between heterogeneous processing and homogeneous processing, the latter of which is expected to add more value to continuous manufg. We also give the key motivations for moving to continuous manufg., some of the exciting new technologies, and the barriers to implementation of continuous manufg. Continuous processing of heterogeneous blends is the natural first step in converting existing batch processes to continuous. In heterogeneous processing, there are discrete particles that can segregate, vs. in homogeneous processing, components are blended and homogenized such that they do not segregate. Heterogeneous processing can incorporate technologies that are closer to existing technologies, where homogeneous processing necessitates the development and incorporation of new technologies. Homogeneous processing has the greatest potential for reaping the full rewards of continuous manufg., but it takes long-term vision and a more significant change in process development than heterogeneous processing. Heterogeneous processing has the detriment that, as the technologies are adopted rather than developed, there is a strong tendency to incorporate correction steps, what we call below "The Rube Goldberg Problem." Thus, although heterogeneous processing will likely play a major role in the near-term transformation of heterogeneous to continuous processing, it is expected that homogeneous processing is the next step that will follow. Specific action items for industry leaders are:Form precompetitive partnerships, including industry (pharmaceutical companies and equipment manufacturers), government, and universities. These precompetitive partnerships would develop case studies of continuous manufg. and ideally perform joint-technol. development, including development of small-scale equipment and processes. Develop ways to invest internally in continuous manufg. How best to do this will depend on the specifics of a given organization, in particular the current development projects. Upper managers will need to energize their process developers to incorporate continuous manufg. in at least part of their processes to gain experience and demonstrate directly the benefits. Training of continuous manufg. technologies, organizational approaches, and regulatory approaches is a key area that industrial leaders should pursue together. © 2014 Wiley Periodicals, Inc. and the American Pharmacists Assocn. J Pharm Sci.
- 21Baxendale, I. R.; Braatz, R. D.; Hodnett, B. K.; Jensen, K. F.; Johnson, M. D.; Sharratt, P.; Sherlock, J.-P.; Florence, A. J. Achieving Continuous Manufacturing: Technologies and Approaches for Synthesis, Workup, and Isolation of Drug Substance May 20–21, 2014 Continuous Manufacturing Symposium. J. Pharm. Sci. 2015, 104 (3), 781– 791, DOI: 10.1002/jps.24252[Crossref], [PubMed], [CAS], Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitVShtrzN&md5=370670a3084de142f68d4327ce88c7a5Achieving Continuous Manufacturing: Technologies and Approaches for Synthesis, Workup, and Isolation of Drug SubstanceBaxendale, Ian R.; Braatz, Richard D.; Hodnett, Benjamin K.; Jensen, Klavs F.; Johnson, Martin D.; Sharratt, Paul; Sherlock, Jon-Paul; Florence, Alastair J.Journal of Pharmaceutical Sciences (2015), 104 (3), 781-791CODEN: JPMSAE; ISSN:0022-3549. (John Wiley & Sons, Inc.)This whitepaper highlights current challenges and opportunities assocd. with continuous synthesis, workup, and crystn. of active pharmaceutical ingredients (drug substances). We describe the technologies and requirements at each stage and emphasize the different considerations for developing continuous processes compared with batch. In addn. to the specific sequence of operations required to deliver the necessary chem. and phys. transformations for continuous drug substance manuf., consideration is also given to how adoption of continuous technologies may impact different manufg. stages in development from discovery, process development, through scale-up and into full scale prodn. The impact of continuous manuf. on drug substance quality and the assocd. challenges for control and for process safety are also emphasized. In addn. to the technol. and operational considerations necessary for the adoption of continuous manufg. (CM), this whitepaper also addresses the cultural, as well as skills and training, challenges that will need to be met by support from organizations in order to accommodate the new work flows. Specific action items for industry leaders are: Develop flow chem. toolboxes, exploiting the advantages of flow processing and including highly selective chemistries that allow use of simple and effective continuous workup technologies. Availability of modular or plug and play type equipment esp. for workup to assist in straightforward deployment in the lab. As with learning from other industries, standardization is highly desirable and will require cooperation across industry and academia to develop and implement. Implement and exploit process anal. technologies (PAT) for real-time dynamic control of continuous processes. Develop modeling and simulation techniques to support continuous process development and control. Progress is required in multiphase systems such as crystn. Involve all parts of the organization from discovery, research and development, and manufg. in the implementation of CM. Engage with academia to develop the training provision to support the skills base for CM, particularly in flow chem., phys. chem., and chem. engineering skills at the chem.-process interface. Promote and encourage publication and dissemination of examples of CM across the sector to demonstrate capability, engage with regulatory comment, and establish benchmarks for performance and highlight challenges. Develop the economic case for CM of drug substance. This will involve various stakeholders at project and business level, however establishing the crit. economic drivers is crit. to driving the transformation in manufg. © 2014 Wiley Periodicals, Inc. and the American Pharmacists Assocn. J Pharm Sci.
- 22Allison, G.; Cain, Y. T.; Cooney, C.; Garcia, T.; Bizjak, T. G.; Holte, O.; Jagota, N.; Komas, B.; Korakianiti, E.; Kourti, D.; Madurawe, R.; Morefield, E.; Montgomery, F.; Nasr, M.; Randolph, W.; Robert, J.-L.; Rudd, D.; Zezza, D. Regulatory and Quality Considerations for Continuous Manufacturing May 20–21, 2014 Continuous Manufacturing Symposium. J. Pharm. Sci. 2015, 104 (3), 803– 812, DOI: 10.1002/jps.24324[Crossref], [CAS], Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXnvVejtg%253D%253D&md5=20c0e252563b2d69bedc17aa8811a0e5Regulatory and Quality Considerations for Continuous Manufacturing. May 20-21, 2014 Continuous SymposiumAllison, Gretchen; Cain, Yanxi Tan; Cooney, Charles; Garcia, Tom; Bizjak, Tara Gooen; Holte, Oyvind; Jagota, Nirdosh; Komas, Bekki; Korakianiti, Evdokia; Kourti, Dora; Madurawe, Rapti; Morefield, Elaine; Montgomery, Frank; Nasr, Moheb; Randolph, William; Robert, Jean-Louis; Rudd, Dave; Zezza, DianeJournal of Pharmaceutical Sciences (2015), 104 (3), 803-812CODEN: JPMSAE; ISSN:0022-3549. (John Wiley & Sons, Inc.)This paper assesses the current regulatory environment, relevant regulations and guidelines, and their impact on continuous manufg. It summarizes current regulatory experience and learning from both review and inspection perspectives. It outlines key regulatory aspects, including continuous manufg. process description and control strategy in regulatory files, process validation, and key Good Manufg. Practice (GMP) requirements. In addn., the paper identifies regulatory gaps and challenges and proposes a way forward to facilitate implementation. © 2015 Wiley Periodicals, Inc. and the American Pharmacists Assocn. J Pharm Sci.
- 23Schaber, S. D.; Gerogiorgis, D. I.; Ramachandran, R.; Evans, J. M. B.; Barton, P. I.; Trout, B. L. Economic Analysis of Integrated Continuous and Batch Pharmaceutical Manufacturing: A Case Study. Ind. Eng. Chem. Res. 2011, 50 (17), 10083– 10092, DOI: 10.1021/ie2006752[ACS Full Text
], [CAS], Google Scholar
23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXpsFOns7g%253D&md5=0410e76f8e4671c6f2bece6b4831cad3Economic Analysis of Integrated Continuous and Batch Pharmaceutical Manufacturing: A Case StudySchaber, Spencer D.; Gerogiorgis, Dimitrios I.; Ramachandran, Rohit; Evans, James M. B.; Barton, Paul I.; Trout, Bernhardt L.Industrial & Engineering Chemistry Research (2011), 50 (17), 10083-10092CODEN: IECRED; ISSN:0888-5885. (American Chemical Society)The capital, operating, and overall costs of a dedicated continuous manufg. process to synthesize an active pharmaceutical ingredient (API) and formulate it into tablets are estd. for a prodn. scale of 2000 t of tablets per yr, with raw material cost, prodn. yield, and API loading varied over broad ranges. Costs are compared to batch prodn. in a dedicated facility. Synthesis begins with a key org. intermediate three synthetic steps before the final API; results are given for key intermediate (KI) costs of +dollar;100 to +dollar;3000/kg, with drug loadings in the tablet of 10 and 50 wt %. The novel continuous process described here is being developed by an interdisciplinary team of 20 researchers. Since yields are not yet well-known, and continuous processes typically have better yields than batch ones, the overall yields of the continuous processes with recycling were set equal to that of the batch process. Without recycling, yields are 10% lower, but less equipment is required. The continuous process has not been built at large scale, so Wroth factors and other assumptions were used to est. costs. Capital expenditures for continuous prodn. were estd. to be 20 to 76% lower, depending on the drug loading, KI cost, and process chosen; operating expenditures were estd. to be between 40% lower and 9% higher. The novel continuous process with recycling coupled to a novel direct tablet formation process yields the best overall cost savings in each drug loading/KI price scenario: estd. savings range from 9 to 40%. Overall cost savings are also given assuming the yield in the continuous case is 10% above and 10% below that of the batch process. Even when yields in the continuous case are lower than in the batch case, savings can still be achieved because the labor, materials handling, CapEx, and other savings compensate. - 24Wilburn, K. R. The Business Case for Continuous Manufacturing of Pharmaceuticals; Massachusetts Institute of Technology, 2010.Google ScholarThere is no corresponding record for this reference.
- 25Simon, L. L.; Kiss, A. A.; Cornevin, J.; Gani, R. Process Engineering Advances in Pharmaceutical and Chemical Industries: Digital Process Design, Advanced Rectification, and Continuous Filtration. Curr. Opin. Chem. Eng. 2019, 114– 121. DOI: 10.1016/j.coche.2019.02.005 .
- 26Vanhoorne, V.; Vervaet, C. Recent Progress in Continuous Manufacturing of Oral Solid Dosage Forms. Int. J. Pharm. 2020, 579, 119194, DOI: 10.1016/j.ijpharm.2020.119194[Crossref], [PubMed], [CAS], Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXltlGnsbk%253D&md5=fe04bf2e31d83a20f82ab565d1b0ebdfRecent progress in continuous manufacturing of oral solid dosage formsVanhoorne, V.; Vervaet, C.International Journal of Pharmaceutics (Amsterdam, Netherlands) (2020), 579 (), 119194CODEN: IJPHDE; ISSN:0378-5173. (Elsevier B.V.)A review. Continuous drug product manufg. is slowly being implemented in the pharmaceutical industry. Although the benefits related to the quality and cost of continuous manufg. are widely recognized, several challenges hampered the widespread introduction of continuous manufg. of drug products. Current review presents an overview of state-of-the art research, equipment, process anal. technol. implementations and advanced control strategies. Addnl., guidelines and regulatory viewpoints on implementation of continuous manufg. in the pharmaceutical industry are discussed.
- 27American Chemical Society. ACS GCI Pharmaceutical Roundtable, https://www.acs.org/content/acs/en/greenchemistry/industry-roundtables/pharmaceutical.html.Google ScholarThere is no corresponding record for this reference.
- 28Poechlauer, P.; Manley, J.; Broxterman, R.; Gregertsen, B.; Ridemark, M. Continuous Processing in the Manufacture of Active Pharmaceutical Ingredients and Finished Dosage Forms: An Industry Perspective. Org. Process Res. Dev. 2012, 16 (10), 1586– 1590, DOI: 10.1021/op300159y[ACS Full Text
], [CAS], Google Scholar
28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtlGkur%252FF&md5=d2510b43ee7d3a9102ba4dedca97fa58Continuous Processing in the Manufacture of Active Pharmaceutical Ingredients and Finished Dosage Forms: An Industry PerspectivePoechlauer, Peter; Manley, Julie; Broxterman, Rinus; Gregertsen, Bjoern; Ridemark, MatsOrganic Process Research & Development (2012), 16 (10), 1586-1590CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)Continuous manufg. as a way of producing fine chems., active pharmaceutical ingredients, and finished dosage forms is gaining widespread attention. Although potential benefits over traditional batch-wise prodn. have been discussed at many occasions and appear evident, continuous processes are only slowly being implemented. The American Chem. Society Green Chem. Institute Pharmaceutical Roundtable has defined "continuous processing" as one of its research priorities and performed a survey of its members' opinions, the status of implementation, and perceived hurdles blocking implementation of continuous manufg. processes. Here we discuss the most important results of this survey and their relation to present trends in this industry to "go green". - 29Poechlauer, P.; Colberg, J.; Fisher, E.; Jansen, M.; Johnson, M. D.; Koenig, S. G.; Lawler, M.; Laporte, T.; Manley, J.; Martin, B.; O’Kearney-McMullan, A. Pharmaceutical Roundtable Study Demonstrates the Value of Continuous Manufacturing in the Design of Greener Processes. Org. Process Res. Dev. 2013, 17 (12), 1472– 1478, DOI: 10.1021/op400245s[ACS Full Text
], [CAS], Google Scholar
29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvVajtbjM&md5=bdaa738462e6ac6ebf86aa8459efbf14Pharmaceutical Roundtable Study Demonstrates the Value of Continuous Manufacturing in the Design of Greener ProcessesPoechlauer, Peter; Colberg, Juan; Fisher, Elizabeth; Jansen, Michael; Johnson, Martin D.; Koenig, Stefan G.; Lawler, Michael; Laporte, Thomas; Manley, Julie; Martin, Benjamin; O'Kearney-McMullan, AnneOrganic Process Research & Development (2013), 17 (12), 1472-1478CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)A review. The American Chem. Society (ACS) Green Chem. Institute (GCI) Pharmaceutical Roundtable conducted a study to elucidate the value of continuous processing, which had been defined as a key research area for green engineering. In the course of defining the business case for continuous processing, individual cases were collected and evaluated to det. specific drivers to implement continuous processing and to find key success factors. The magnitude and timing of effects and the relation to the principles of green chem. were investigated. - 30Food and Drug Administration. Guidance for Industry PAT: A Framework for Innovative Pharmaceutical Development, Manufacuring, and Quality Assurance. FDA Off. Doc. 2004, No. September, 16.Google ScholarThere is no corresponding record for this reference.
- 31Simon, L. L.; Pataki, H.; Marosi, G.; Meemken, F.; Hungerbühler, K.; Baiker, A.; Tummala, S.; Glennon, B.; Kuentz, M.; Steele, G.; Kramer, H. J. M.; Rydzak, J. W.; Chen, Z.; Morris, J.; Kjell, F.; Singh, R.; Gani, R.; Gernaey, K. V.; Louhi-Kultanen, M.; Oreilly, J.; Sandler, N.; Antikainen, O.; Yliruusi, J.; Frohberg, P.; Ulrich, J.; Braatz, R. D.; Leyssens, T.; von Stosch, M.; Oliveira, R.; Tan, R. B. H.; Wu, H.; Khan, M.; Ogrady, D.; Pandey, A.; Westra, R.; Delle-Case, E.; Pape, D.; Angelosante, D.; Maret, Y.; Steiger, O.; Lenner, M.; Abbou-Oucherif, K.; Nagy, Z. K.; Litster, J. D.; Kamaraju, V. K.; Chiu, M. Sen. Assessment of Recent Process Analytical Technology (PAT) Trends: A Multiauthor Review. Org. Process Res. Dev. 2015, 19 (1), DOI: 10.1021/op500261y .
- 32Food and Drug Administration. Quality Considerations for Continuous Manufacturing Guidance for Industry. FDA Off. Doc. 2019Google ScholarThere is no corresponding record for this reference.
- 33International Council for Harmonization. ICH Q8: Pharmaceutical Development; 2009.Google ScholarThere is no corresponding record for this reference.
- 34International Council for Harmonization. ICH Q9: Quality Risk Management; 2005.Google ScholarThere is no corresponding record for this reference.
- 35International Council for Harmonization. ICH Q10: Pharmaceutical Quality System; 2008.Google ScholarThere is no corresponding record for this reference.
- 36International Council for Harmonization. ICH Q11: Development and Manufacture of Drug Substances (Chemical Entities and Biotechnological/Biological Entities); 2012.Google ScholarThere is no corresponding record for this reference.
- 37International Council for Harmonization. ICH Q13: Continuous Manufacturing of Drug Substances and Drug Products; 2018.Google ScholarThere is no corresponding record for this reference.
- 38Mihokovic, N. Continuous Manufacturing - EMA Perspective and Experience. In Engineering Conferences International; 2017.Google ScholarThere is no corresponding record for this reference.
- 39Food and Drug Administration. FDA statement on FDA’s modern approach to advanced pharmaceutical manufacturing, https://www.fda.gov/news-events/press-announcements/fda-statement-fdas-modern-approach-advanced-pharmaceutical-manufacturing?platform=hootsuite.Google ScholarThere is no corresponding record for this reference.
- 40Yu, L. X.; Amidon, G.; Khan, M. A.; Hoag, S. W.; Polli, J.; Raju, G. K.; Woodcock, J. Understanding Pharmaceutical Quality by Design. AAPS J. 2014, 16 (4), 771– 783, DOI: 10.1208/s12248-014-9598-3[Crossref], [PubMed], [CAS], Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXovVGrurg%253D&md5=243fb3d4124d8c58f15e3e4ee8d3b532Understanding Pharmaceutical Quality by DesignYu, Lawrence X.; Amidon, Gregory; Khan, Mansoor A.; Hoag, Stephen W.; Polli, James; Raju, G. K.; Woodcock, JanetAAPS Journal (2014), 16 (4), 771-783CODEN: AJAOB6; ISSN:1550-7416. (Springer)This review further clarifies the concept of pharmaceutical quality by design (QbD) and describes its objectives. QbD elements include the following: (1) a quality target product profile (QTPP) that identifies the crit. quality attributes (CQAs) of the drug product; (2) product design and understanding including identification of crit. material attributes (CMAs); (3) process design and understanding including identification of crit. process parameters (CPPs), linking CMAs and CPPs to CQAs; (4) a control strategy that includes specifications for the drug substance(s), excipient(s), and drug product as well as controls for each step of the manufg. process; and (5) process capability and continual improvement. QbD tools and studies include prior knowledge, risk assessment, mechanistic models, design of expts. (DoE) and data anal., and process anal. technol. (PAT). As the pharmaceutical industry moves toward the implementation of pharmaceutical QbD, a common terminol., understanding of concepts and expectations are necessary. This understanding will facilitate better communication between those involved in risk-based drug development and drug application review.
- 41Szilagyi, B.; Eren, A.; Quon, J. L.; Papageorgiou, C. D.; Nagy, Z. K. Application of Model-Free and Model-Based Quality-by-Control (QbC) for the Efficient Design of Pharmaceutical Crystallization Processes. Cryst. Growth Des. 2020, 20 (6), 3979– 3996, DOI: 10.1021/acs.cgd.0c00295[ACS Full Text
], [CAS], Google Scholar
41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXnt1Wjs78%253D&md5=570473820e9f7830d9f8eca8bf022495Application of Model-Free and Model-Based Quality-by-Control (QbC) for the Efficient Design of Pharmaceutical Crystallization ProcessesSzilagyi, Botond; Eren, Ayse; Quon, Justin L.; Papageorgiou, Charles D.; Nagy, Zoltan K.Crystal Growth & Design (2020), 20 (6), 3979-3996CODEN: CGDEFU; ISSN:1528-7483. (American Chemical Society)The design of pharmaceutical crystn. processes is a challenging engineering problem because of the specific and versatile quality requirements of the end-product, amplified by the tight regulatory stds. The current industrial std. for crystn. process design is based on the use of the quality-by-design (QbD) framework, which relies on factorial design of expts. (DoE). Hence, QbD inherently generates a large no. of resource-consuming open loop crystn. expts. This is esp. true when more complex operating conditions need to be designed, such as temp. cycles, which require a large no. of decision variables in the DoE. In contrast, the recently proposed quality-by-control (QbC) approach relies on feedback control algorithms to directly achieve the desired product properties by manipulating the appropriate process conditions. The first aim of this work is to demonstrate the effectiveness of a model-free feedback control strategy, referred to as model-free (mf) QbC. Direct nucleation control (DNC) and supersatn. control (SSC) are applied as a part of the mfQbC approach, which, ideally, requires only two feedback control expts. to obtain a temp. profile that results in obtaining the desired product quality. Although mfQbC provides a rapid process design, it is often suboptimal. In addn., it is shown that the exptl. data generated by mfQbC can be used for process model development and kinetic parameter estn. The validated model enables optimization-based design using the model-based (mb) QbC framework. For this case study, a population balance (PB) based process model is developed, which involves primary and secondary nucleation, growth, and dissoln., as well as a novel formulation of agglomeration, and deagglomeration of crystals. In addn. to taking into account the agglomeration, the no. of agglomerates is also tracked as a balance between the agglomeration and deagglomeration events. The kinetic parameters are estd. using a novel objective function formulation relying on the minimization of the difference between the measured and simulated concns. and crystal size distributions (CSDs) and the maximization of the correlation between the simulated crystal no. d. and measured crystal count data obtained from focused beam reflectance measurement (FBRM). The kinetic parameters are identified based on the exptl. data generated from the mfQbC, which inherently reduced the exptl. effort required for the model development. The temp. profile is optimized for the fine index and agglomeration degree minimization. The repeated open-loop implementation of mfQbC- and mbQbC-designed processes showed that the batch-to-batch variation is low and the product quality is high in both cases. The proposed general framework is illustrated for the systematic quick and optimal design of crystn. processes that require temp. cycles with a low no. of expts. An efficient crystn. design approach is proposed that combines model-free and model-based quality-by-control (QbC) frameworks, based on the use of process anal. technol. (PAT) tools, feedback control approaches, and math. modeling for the design and optimization of crystn. processes. This proof-of-concept study demonstrates that QbC can provide optimal operating conditions to produce crystals with desired crit. quality attributes (CQAs). - 42Szilágyi, B.; Borsos, Á.; Pal, K.; Nagy, Z. K. Experimental Implementation of a Quality-by-Control (QbC) Framework Using a Mechanistic PBM-Based Nonlinear Model Predictive Control Involving Chord Length Distribution Measurement for the Batch Cooling Crystallization of l-Ascorbic Acid. Chem. Eng. Sci. 2019, 195, 335– 346, DOI: 10.1016/j.ces.2018.09.032[Crossref], [CAS], Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvV2gs77M&md5=9d025ea247c33a21b8d61bb8e3c48283Experimental implementation of a Quality-by-Control (QbC) framework using a mechanistic PBM-based nonlinear model predictive control involving chord length distribution measurement for the batch cooling crystallization of L-ascorbic acidSzilagyi, Botond; Borsos, Akos; Pal, Kanjakha; Nagy, Zoltan K.Chemical Engineering Science (2019), 195 (), 335-346CODEN: CESCAC; ISSN:0009-2509. (Elsevier Ltd.)L-Ascorbic acid is synthesized in large industrial scale from glucose and marketed as an immune system strengthening agent and anti-oxidant ingredient. The overall yield of conversion of the precursor glucose to L-ascorbic acid is limited, therefore the crystn. is a critically important step of the L-ascorbic acid prodn. from economic point of view. It is widely accepted that the crystal size distribution (CSD) influences numerous relevant macroscopic properties of the final cryst. product and it also significantly affects the downstream operations. The present paper discusses the chord length distribution (CLD, which is directly related to the CSD) control, during the crystn. of L-ascorbic acid from aq. soln. Batch crystn. process is employed, which is the classical, and still dominant, operation in fine chem. and pharmaceutical industries. A comparative exptl. study of two state-of-the-art Quality-by-Control (QbC) based crystn. design approaches are presented: (1) a model-free QbC based on direct nucleation control (DNC) and (2) a model-based QbC using a novel nonlinear model predicative control (NMPC) framework. In the first investigation, the DNC, a process anal. technol. based state-of-the-art model free control strategy, is applied. Although, DNC requires minimal preliminary system information and often provides robust process control, due to the unusual crystn. behavior of L-ascorbic acid, it leads to long batch times and oscillatory operation. In a second study the benefits of model-based QbC approach are demonstrated, based on using a NMPC approach. A population balance based crystn. process model is built and calibrated by estg. the nucleation and growth kinetics from concn. and CLD measurements. A projection based CSD to CLD forward transformation is used in the estn. of nucleation and growth kinetics. For robustness and adaptive behavior, the NMPC is coupled with a growing horizon state estimator, which is aimed to continuously improve the model by re-adjusting the kinetic consts. The study demonstrates that the model-based QbC framework can lead to rapid and robust crystn. process development with the NMPC system presenting good control behavior under significant plant model mismatch (PMM) conditions.
- 43Badman, C.; Cooney, C. L.; Florence, A.; Konstantinov, K.; Krumme, M.; Mascia, S.; Nasr, M.; Trout, B. L. Why We Need Continuous Pharmaceutical Manufacturing and How to Make It Happen. J. Pharm. Sci. 2019, 108 (11), 3521– 3523, DOI: 10.1016/j.xphs.2019.07.016[Crossref], [PubMed], [CAS], Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhslensr7I&md5=f0b5c26568baade649af7c927aa41030Why We Need Continuous Pharmaceutical Manufacturing and How to Make It HappenBadman, Clive; Cooney, Charles L.; Florence, Alastair; Konstantinov, Konstantin; Krumme, Markus; Mascia, Salvatore; Nasr, Moheb; Trout, Bernhardt L.Journal of Pharmaceutical Sciences (Philadelphia, PA, United States) (2019), 108 (11), 3521-3523CODEN: JPMSAE; ISSN:0022-3549. (Elsevier Inc.)We make the case for why continuous pharmaceutical manufg. is essential, what the barriers are, and how to overcome them. To overcome them, government action is needed in terms of tax incentives or regulatory incentives that affect time.
- 44Hu, C.; Shores, B. T.; Derech, R. A.; Testa, C. J.; Hermant, P.; Wu, W.; Shvedova, K.; Ramnath, A.; Al Ismaili, L. Q.; Su, Q.; Sayin, R.; Born, S. C.; Takizawa, B.; O’Connor, T. F.; Yang, X.; Ramanujam, S.; Mascia, S. Continuous Reactive Crystallization of an API in PFR-CSTR Cascade with in-Line PATs. React. Chem. Eng. 2020, 5 (10), 1950– 1962, DOI: 10.1039/D0RE00216J[Crossref], [CAS], Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhs1Cis73L&md5=b5e6b53563c7d64ae836ee663d33b0f9Continuous reactive crystallization of an API in PFR-CSTR cascade with in-line PATsHu, Chuntian; Shores, Brianna T.; Derech, Rachel A.; Testa, Christopher J.; Hermant, Paul; Wu, Wei; Shvedova, Khrystyna; Ramnath, Anjana; Al Ismaili, Liyutha Q.; Su, Qinglin; Sayin, Ridade; Born, Stephen C.; Takizawa, Bayan; O'Connor, Thomas F.; Yang, Xiaochuan; Ramanujam, Sukumar; Mascia, SalvatoreReaction Chemistry & Engineering (2020), 5 (10), 1950-1962CODEN: RCEEBW; ISSN:2058-9883. (Royal Society of Chemistry)The continuous reactive crystn. of an active pharmaceutical ingredient (API) in a plug flow reactor (PFR)-continuous stirred tank reactor (CSTR) cascade system with in-line PATs was developed and investigated. Residence time distribution (RTD) measurements of the PFR (stage 1), the CSTR cascade (stages 2-6), and the combined PFR-CSTR cascade (stages 1-6) were performed to est. the performance of the reactors. Several continuous reactive crystn. expts. were performed, and consistent reaction yields of 91.3 ± 0.5 and 89.6 ± 0.4% were obtained with and without the PFR, resp. The integration of PFR (stage 1) created a very high level of supersatn. by itself, and ∼25% lower supersatn. and a 2.7% higher crystn. yield in the following vessel (stage 2). In stages 3-6, the supersatn. levels and crystn. yields were similar (with and without the PFR). With the PFR, the lower supersatn. in stage 2 resulted in lower nucleation rates and higher crystal growth rates, resulting in a larger crystal size distribution. Also, in-line ReactIR and focused beam reflectance measurement (FBRM) were used to monitor the reactant concn. and crystal chord length, resp., during the reactive crystn. process. The ReactIR predicted reactant concns. in the mother liquor that matched well with corresponding HPLC results (prediction error < 0.17%). The FBRM results showed a relatively stable mean square-weighted chord length of ∼150μm. In addn., the process mass intensities (PMIs) for the batch process, the integrated continuous manufg. (ICM) process without the PFR, and the ICM process with the PFR were 3.49, 1.99, and 1.97, resp.
- 45Di Pretoro, G. Making the switch from batch to continuous manufacturing: an industrial perspective, https://youtube/lOimBHi8bJw?t=302.Google ScholarThere is no corresponding record for this reference.
- 46Johnson&Johnson. The Inspire line, https://www.youtube.com/watch?v=Q5KW1nr_asE&feature=emb_title&ab_channel=LawrenceDeBelder.Google ScholarThere is no corresponding record for this reference.
- 47McKenzie, P.; Kiang, S.; Tom, J.; Rubin, A. E.; Futran, M. Can Pharmaceutical Process Development Become High Tech?. AIChE J. 2006, 52 (12), 3990– 3994, DOI: 10.1002/aic.11022[Crossref], [CAS], Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtlSjurbN&md5=39df50d043444f9741bf4a993533da13Can pharmaceutical process development become high tech?McKenzie, Paul; Kiang, San; Tom, Jean; Rubin, A. Erik; Futran, MauricioAIChE Journal (2006), 52 (12), 3990-3994CODEN: AICEAC; ISSN:0001-1541. (John Wiley & Sons, Inc.)A review. While the pharmaceutical industry is well known for its innovation in developing drugs and therapies, and its ability to provide quality medicines to the public, its manufg. efficiency lags behind that of most other industries. This article proposes a new approach for process development based on the following four elements:. The approaches proposed by the FDA, including Process Anal. Technol. (i.e., online measurements) coupled to real time control; the principles of quality by design and risk-based anal.; the use of statistical, multivariate anal.; and providing mechanistic understanding through lab automation, miniaturization, using Design of Expts. (DoE), and an emphasis on phys. org. chem. and kinetics. Shaping the science of process development by applying Instrument, Systems and Automation Society (ISA) S.88 stds. from development through commercialization, which provide a natural language for chem. and pharmaceutical processes, as well as anal. methods. This provides us with a vehicle for creating and systematizing process knowledge and is also a very effective basis on which harmonic interactions between R&D and manufg. can be built. This leads to the use of a no. of electronic tools for the lab., for process modeling, for execution of process recipes in labs, pilot plants and prodn. facilities, and for managing data, linked seamlessly between R&D and manufg. The use of modeling tools to reduce the need for empirical experimentation, for verification of mechanistic understanding and for process control. The use of parallel experimentation using automated work streams for rapid probing of process options, exploring process operating windows ("design space"), and generally increasing productivity.
- 48Baxendale, I. R.; Braatz, R. D.; Hodnett, B. K.; Jensen, K. F.; Johnson, M. D.; Sharratt, P.; Sherlock, J.-P.; Florence, A. J. Achieving Continuous Manufacturing: Technologies and Approaches for Synthesis, Workup, and Isolation of Drug Substance May 20–21, 2014 Continuous Manufacturing Symposium. J. Pharm. Sci. 2015, 104 (3), 781– 791, DOI: 10.1002/jps.24252[Crossref], [PubMed], [CAS], Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitVShtrzN&md5=370670a3084de142f68d4327ce88c7a5Achieving Continuous Manufacturing: Technologies and Approaches for Synthesis, Workup, and Isolation of Drug SubstanceBaxendale, Ian R.; Braatz, Richard D.; Hodnett, Benjamin K.; Jensen, Klavs F.; Johnson, Martin D.; Sharratt, Paul; Sherlock, Jon-Paul; Florence, Alastair J.Journal of Pharmaceutical Sciences (2015), 104 (3), 781-791CODEN: JPMSAE; ISSN:0022-3549. (John Wiley & Sons, Inc.)This whitepaper highlights current challenges and opportunities assocd. with continuous synthesis, workup, and crystn. of active pharmaceutical ingredients (drug substances). We describe the technologies and requirements at each stage and emphasize the different considerations for developing continuous processes compared with batch. In addn. to the specific sequence of operations required to deliver the necessary chem. and phys. transformations for continuous drug substance manuf., consideration is also given to how adoption of continuous technologies may impact different manufg. stages in development from discovery, process development, through scale-up and into full scale prodn. The impact of continuous manuf. on drug substance quality and the assocd. challenges for control and for process safety are also emphasized. In addn. to the technol. and operational considerations necessary for the adoption of continuous manufg. (CM), this whitepaper also addresses the cultural, as well as skills and training, challenges that will need to be met by support from organizations in order to accommodate the new work flows. Specific action items for industry leaders are: Develop flow chem. toolboxes, exploiting the advantages of flow processing and including highly selective chemistries that allow use of simple and effective continuous workup technologies. Availability of modular or plug and play type equipment esp. for workup to assist in straightforward deployment in the lab. As with learning from other industries, standardization is highly desirable and will require cooperation across industry and academia to develop and implement. Implement and exploit process anal. technologies (PAT) for real-time dynamic control of continuous processes. Develop modeling and simulation techniques to support continuous process development and control. Progress is required in multiphase systems such as crystn. Involve all parts of the organization from discovery, research and development, and manufg. in the implementation of CM. Engage with academia to develop the training provision to support the skills base for CM, particularly in flow chem., phys. chem., and chem. engineering skills at the chem.-process interface. Promote and encourage publication and dissemination of examples of CM across the sector to demonstrate capability, engage with regulatory comment, and establish benchmarks for performance and highlight challenges. Develop the economic case for CM of drug substance. This will involve various stakeholders at project and business level, however establishing the crit. economic drivers is crit. to driving the transformation in manufg. © 2014 Wiley Periodicals, Inc. and the American Pharmacists Assocn. J Pharm Sci.
- 49Laske, S.; Paudel, A.; Scheibelhofer, O.; Sacher, S.; Hoermann, T.; Khinast, J.; Kelly, A.; Rantannen, J.; Korhonen, O.; Stauffer, F.; De Leersnyder, F.; De Beer, T.; Mantanus, J.; Chavez, P.-F.; Thoorens, B.; Ghiotti, P.; Schubert, M.; Tajarobi, P.; Haeffler, G.; Lakio, S.; Fransson, M.; Sparen, A.; Abrahmsen-Alami, S.; Folestad, S.; Funke, A.; Backx, I.; Kavsek, B.; Kjell, F.; Michaelis, M.; Page, T.; Palmer, J.; Schaepman, A.; Sekulic, S.; Hammond, S.; Braun, B.; Colegrove, B. A Review of PAT Strategies in Secondary Solid Oral Dosage Manufacturing of Small Molecules. J. Pharm. Sci. 2017, 106 (3), 667– 712, DOI: 10.1016/j.xphs.2016.11.011[Crossref], [PubMed], [CAS], Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitFGitbvN&md5=c3461e3652d13f5d477fd3c845119b10A Review of PAT Strategies in Secondary Solid Oral Dosage Manufacturing of Small MoleculesLaske, Stephan; Paudel, Amrit; Scheibelhofer, OttoJournal of Pharmaceutical Sciences (2017), 106 (3), 667-712CODEN: JPMSAE; ISSN:0022-3549. (Elsevier Inc.)Pharmaceutical solid oral dosage product manufg. is a well-established, yet revolutionizing area. To this end, process anal. technol. (PAT) involves interdisciplinary and multivariate (chem., phys., microbiol., and math.) methods for material (e.g., materials, intermediates, products) and process (e.g., temp., pressure, throughput, etc.) anal. This supports rational process modeling and enhanced control strategies for improved product quality and process efficiency. Therefore, it is often difficult to orient and find the relevant, integrated aspects of the current state-of-the-art. Esp., the link between fundamental research, in terms of sensor and control system development, to the application both in lab. and manufg. scale, is difficult to comprehend. This review compiles a nonexhaustive overview on current approaches from the recognized academia and industrial practices of PAT, including screening, selection, and final implementations in solid oral dosage manufg., through a wide diversity of use cases. Finally, the authors attempt to ext. a common consensus toward developing PAT application guidance for different unit operations of drug product manufg.
- 50Nepveux, K.; Sherlock, J. P.; Futran, M.; Thien, M.; Krumme, M. How Development and Manufacturing Will Need to Be Structured-Heads of Development/Manufacturing May 20–21, 2014 Continuous Manufacturing Symposium. J. Pharm. Sci. 2015, 104 (3), 850– 864, DOI: 10.1002/jps.24286[Crossref], [CAS], Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXislegt7g%253D&md5=2b926f42d8e120457f24ff78ee94f868How Development and Manufacturing Will Need to Be Structured-Heads of Development/Manufacturing. May 20-21, 2014 Continuous Manufacturing SymposiumNepveux, Kevin; Sherlock, Jon-Paul; Futran, Mauricio; Thien, Michael; Krumme, MarkusJournal of Pharmaceutical Sciences (2015), 104 (3), 850-864CODEN: JPMSAE; ISSN:0022-3549. (John Wiley & Sons, Inc.)Continuous manufg. (CM) is a process technol. that has been used in the chem. industry for large-scale mass prodn. of chems. in single-purpose plants with benefit for many years. Recent interest has been raised to expand CM into the low-vol., high-value pharmaceutical business with its unique requirements regarding readiness for human use and the required quality, supply chain, and liability constraints in this business context. Using a fairly abstr. set of definitions, this paper derives tech. consequences of CM in different scenarios along the development-launch-supply axis in different business models and how they compare to batch processes. Impact of CM on functions in development is discussed and several operational models suitable for originators and other business models are discussed and specific aspects of CM are deduced from CM's tech. characteristics. Organizational structures of current operations typically can support CM implementations with just minor refinements if the CM technol. is limited to single steps or small sequences (bin-to-bin approach) and if the appropriate tech. skill set is available. In such cases, a small, dedicated group focused on CM is recommended. The manufg. strategy, as centralized vs. decentralized in light of CM processes, is discussed and the potential impact of significantly shortened supply lead times on the organization that runs these processes. The ultimate CM implementation may be seen by some as a totally integrated monolithic plant, one that unifies chem. and pharmaceutical operations into one plant. The organization supporting this approach will have to reflect this change in scope and responsibility. The other extreme, admittedly futuristic at this point, would be a highly decentralized approach with multiple smaller hubs; this would require a new and different organizational structure. This processing approach would open up new opportunities for products that, because of stability constraints or individualization to patients, do not allow centralized manufg. approaches at all. Again, the entire enterprise needs to be restructured accordingly. The situation of CM in an outsourced operation business model is discussed. Next steps for the industry are recommended. In summary, opportunistic implementation of isolated steps in existing portfolios can be implemented with minimal organizational changes; the availability of the appropriate skills is the detg. factor. The implementation of more substantial sequences requires business processes that consider the portfolio, not just single products. Exploration and implementation of complete process chains with consequences for quality decisions do require appropriate organizational support. © 2015 Wiley Periodicals, Inc. and the American Pharmacists Assocn. J Pharm Sci 104:850-864, 2015.
- 51Leuenberger, H. New Trends in the Production of Pharmaceutical Granules: Batch versus Continuous Processing. Eur. J. Pharm. Biopharm. 2001, 52 (3), 289– 296, DOI: 10.1016/S0939-6411(01)00199-0[Crossref], [PubMed], [CAS], Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXnslCiu7Y%253D&md5=2451e7d4b2dbbb60265360d0f5ab7f91New trends in the production of pharmaceutical granules: batch versus continuous processingLeuenberger, HansEuropean Journal of Pharmaceutics and Biopharmaceutics (2001), 52 (3), 289-296CODEN: EJPBEL; ISSN:0939-6411. (Elsevier Science Ireland Ltd.)A review with refs. In the pharmaceutical industry, the prodn. of granules is based on a batch concept. This concept offers many advantages with respect to quality assurance as a batch can be accepted or rejected. However, the scale-up of the batch size may lead to problems. The variety of the equipment involved often does not facilitate the scale-up process. In order to avoid scale-up problems, continuous or semi-continuous processes have to be evaluated as alternatives to a batch prodn. Thus, a quasi-continuous prodn. line is presented, which permits the prodn. of small-scale batches, e.g. for clin. trials and for large-scale batches using the same equipment.
- 52Vervaet, C.; Remon, J. P. Continuous Granulation in the Pharmaceutical Industry. Chem. Eng. Sci. 2005, 60 (14), 3949– 3957, DOI: 10.1016/j.ces.2005.02.028[Crossref], [CAS], Google Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXktlCgtb0%253D&md5=b85240383e8626893ea4f7c44762877eContinuous granulation in the pharmaceutical industryVervaet, Chris; Remon, Jean PaulChemical Engineering Science (2005), 60 (14), 3949-3957CODEN: CESCAC; ISSN:0009-2509. (Elsevier Ltd.)A review. Traditionally the manufg. of pharmaceutical dosage forms has been a batch-wise process and continuous processes have limited applications in a pharmaceutical manufg. plant. However, several factors (redn. of cost, improved process efficiency, optimal use of equipment, flexibility in prodn. capacity) are stimulating the pharmaceutical industry to investigate the opportunities offered by continuous processes. This paper discusses some of the techniques which could be implemented in a continuous granulation process of pharmaceuticals.
- 53Chen, H.; Diep, E.; Langrish, T. A. G.; Glasser, B. J. Continuous Fluidized Bed Drying: Residence Time Distribution Characterization and Effluent Moisture Content Prediction. AIChE J. 2020, 66 (5), DOI: 10.1002/aic.16902 .
- 54Handbook of Pharmaceutical Granulation Technology; Parikh, D. M., Ed.; CRC Press, 2016; DOI: 10.3109/9781616310035 .
- 55Fülöp, G.; Domokos, A.; Galata, D.; Szabó, E.; Gyürkés, M.; Szabó, B.; Farkas, A.; Madarász, L.; Démuth, B.; Lendér, T.; Nagy, T.; Kovács-Kiss, D.; Van der Gucht, F.; Marosi, G.; Nagy, Z. K. Integrated Twin-Screw Wet Granulation, Continuous Vibrational Fluid Drying and Milling: A Fully Continuous Powder to Granule Line. Int. J. Pharm. 2021, 594, 120126, DOI: 10.1016/j.ijpharm.2020.120126
- 56Webb, D.; Jamison, T. F. Continuous Flow Multi-Step Organic Synthesis. Chem. Sci. 2010, 1 (6), 675, DOI: 10.1039/c0sc00381f[Crossref], [CAS], Google Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhsVGrtLs%253D&md5=a57e8dcd6959e5bd822562f9b2999c1eContinuous flow multi-step organic synthesisWebb, Damien; Jamison, Timothy F.Chemical Science (2010), 1 (6), 675-680CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)Using continuous flow techniques for multi-step synthesis enables multiple reaction steps to be combined into a single continuous operation. In this mini-review we discuss the current state of the art in this field and highlight recent progress and current challenges facing this emerging area.
- 57Plutschack, M. B.; Pieber, B.; Gilmore, K.; Seeberger, P. H. The Hitchhiker’s Guide to Flow Chemistry. Chem. Rev. 2017, 117 (18), 11796– 11893, DOI: 10.1021/acs.chemrev.7b00183[ACS Full Text
], [CAS], Google Scholar
57https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXpt1Siu7c%253D&md5=d95f9e3c3cc5ed69d5ba5347eedd9ae9The Hitchhiker's Guide to Flow ChemistryPlutschack, Matthew B.; Pieber, Bartholomaeus; Gilmore, Kerry; Seeberger, Peter H.Chemical Reviews (Washington, DC, United States) (2017), 117 (18), 11796-11893CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)Flow chem. involves the use of channels or tubing to conduct a reaction in a continuous stream rather than in a flask. Flow equipment provides chemists with unique control over reaction parameters, enhancing reactivity or in some cases enabling new reactions. This relatively young technol. has received a remarkable amt. of attention in the past decade with many reports on what can be done in flow. Until recently, however, the question, "Should we do this in flow" has merely been an afterthought. This review introduces readers to the basic principles and fundamentals of flow chem. and critically discusses recent flow chem. accounts. - 58Pastre, J. C.; Browne, D. L.; Ley, S. V. Flow Chemistry Syntheses of Natural Products. Chem. Soc. Rev. 2013, 42 (23), 8849, DOI: 10.1039/c3cs60246j[Crossref], [PubMed], [CAS], Google Scholar58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslWqsrfK&md5=2893d5a1b685c8312aeeaa22a8b1272cFlow chemistry syntheses of natural productsPastre, Julio C.; Browne, Duncan L.; Ley, Steven V.Chemical Society Reviews (2013), 42 (23), 8849-8869CODEN: CSRVBR; ISSN:0306-0012. (Royal Society of Chemistry)A review. The development and application of continuous flow chem. methods for synthesis is a rapidly growing area of research. In particular, natural products provide demanding challenges to this developing technol. This review highlights successes in the area with an emphasis on new opportunities and technol. advances.
- 59Bana, P.; Örkényi, R.; Lövei, K.; Lakó, Á.; Túrós, G. I.; Éles, J.; Faigl, F.; Greiner, I. The Route from Problem to Solution in Multistep Continuous Flow Synthesis of Pharmaceutical Compounds. Bioorg. Med. Chem. 2017, 25 (23), 6180– 6189, DOI: 10.1016/j.bmc.2016.12.046[Crossref], [PubMed], [CAS], Google Scholar59https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXnvVaisQ%253D%253D&md5=b96f46e1281775b449e8281c8f42cd12The route from problem to solution in multistep continuous flow synthesis of pharmaceutical compoundsBana, Peter; Orkenyi, Robert; Lovei, Klara; Lako, Agnes; Turos, Gyorgy Istvan; Eles, Janos; Faigl, Ferenc; Greiner, IstvanBioorganic & Medicinal Chemistry (2017), 25 (23), 6180-6189CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)Recent advances in the field of continuous flow chem. allow the multistep prepn. of complex mols. such as APIs (Active Pharmaceutical Ingredients) in a telescoped manner. Numerous examples of lab.-scale applications are described, which are pointing towards novel manufg. processes of pharmaceutical compds., in accordance with recent regulatory, economical and quality guidances. The chem. and tech. knowledge gained during these studies is considerable; nevertheless, connecting several individual chem. transformations and the attached analytics and purifn. holds hidden traps. In this review, we summarize innovative solns. for these challenges, in order to benefit chemists aiming to exploit flow chem. systems for the synthesis of biol. active mols.
- 60Munirathinam, R.; Huskens, J.; Verboom, W. Supported Catalysis in Continuous-Flow Microreactors. Adv. Synth. Catal. 2015, 357 (6), 1093– 1123, DOI: 10.1002/adsc.201401081[Crossref], [CAS], Google Scholar60https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXkvVShs7Y%253D&md5=b6b1c0fa177d356c99e5d3f113e18e70Supported Catalysis in Continuous-Flow MicroreactorsMunirathinam, Rajesh; Huskens, Jurriaan; Verboom, WillemAdvanced Synthesis & Catalysis (2015), 357 (6), 1093-1123CODEN: ASCAF7; ISSN:1615-4150. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. The recent developments in performing heterogeneous catalysis in continuous-flow microreactors was summarized. Three different types, namely, (i) packed-bed, (ii) monolithic, and (iii) wall-coated approaches were discussed to implement various kinds of catalysts in a microreactor. In addn., the applications of these supported catalysts to perform a variety of org. reactions were described. Furthermore, advantages of catalytic microreactors over classical batch reactors on one or more aspects of the reaction, such as rate, conversion, selectivity, and enantioselectivity were presented.
- 61Mallia, C. J.; Baxendale, I. R. The Use of Gases in Flow Synthesis. Org. Process Res. Dev. 2016, 20 (2), 327– 360, DOI: 10.1021/acs.oprd.5b00222[ACS Full Text
], [CAS], Google Scholar
61https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXht1OqsL%252FL&md5=c7d24b42aa77400a3d1e990e63d1fb62The Use of Gases in Flow SynthesisMallia, Carl J.; Baxendale, Ian R.Organic Process Research & Development (2016), 20 (2), 327-360CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)A review. This review will highlight the potential benefits that can be leveraged by using flow chem. to allow gases to be used in research in a safer and more efficient way. An overview of the different approaches used to introduce gases into flow reactors is presented along with a synopsis of the different gaseous reactions classes already successfully translated into flow. - 62Brzozowski, M.; O’Brien, M.; Ley, S. V.; Polyzos, A. Flow Chemistry: Intelligent Processing of Gas–Liquid Transformations Using a Tube-in-Tube Reactor. Acc. Chem. Res. 2015, 48 (2), 349– 362, DOI: 10.1021/ar500359m[ACS Full Text
], [CAS], Google Scholar
62https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsF2isbs%253D&md5=1e1c23755bb387f89c6189290ef29c22Flow Chemistry: Intelligent Processing of Gas-Liquid Transformations Using a Tube-in-Tube ReactorBrzozowski, Martin; O'Brien, Matthew; Ley, Steven V.; Polyzos, AnastasiosAccounts of Chemical Research (2015), 48 (2), 349-362CODEN: ACHRE4; ISSN:0001-4842. (American Chemical Society)A review of recent progress in gas-liq. reactions in tube-in-tube (flow) reactors. Flow chem. methodol. has led to measurable improvements in reduced energy consumption, expansion of available reaction conditions, development of multistep methods employing supported reagents, and in-line monitoring of reaction intermediates to ensure the delivery of high quality target compds. Flow chem. approaches have addressed the challenges assocd. with reactions using reactive gases in classical batch synthesis. The small vols. of microreactors ameliorate the hazards of high-pressure gas reactions and enable improved mixing with the liq. phase. Established strategies for gas-liq. reactions in flow have relied on plug-flow (or segmented flow) regimes in which the gas plugs are introduced to a liq. stream and dissoln. of gas relies on interfacial contact of the gas bubble with the liq. phase. This approach confers limited control over gas concn. within the liq. phase and is unsuitable for multistep methods requiring heterogeneous catalysis or solid supported reagents. We have identified the use of a gas-permeable fluoropolymer, Teflon AF-2400, as a simple method of achieving efficient gas-liq. contact to afford homogeneous solns. of reactive gases in flow, with significant control of stoichiometry of reactive gases in a reaction mixt. A tube-in-tube reactor device was developed that consisted of a pair of concentric capillaries in which pressurized gas permeates through an inner Teflon AF-2400 tube and reacts with dissolved substrate within a liq. phase that flows within a second gas impermeable tube. This Account examines efforts toward the development of a simple, unified methodol. for the processing of gaseous reagents in flow by way of development of a tube-in-tube reactor device and applications to key C-C, C-N, and C-O bond forming and hydrogenation reactions. Other applications discussed include multistep reactions using solid-supported reagents, extending the technol. to processes using multiple gas reagents, development of computer-aided imaging techniques to allow automated in-line monitoring of gas concn. and stoichiometry in real time. - 63Tajti, Á.; Tóth, N.; Bálint, E.; Keglevich, G. Esterification of Benzoic Acid in a Continuous Flow Microwave Reactor. J. Flow Chem. 2018, 8 (1), 11– 19, DOI: 10.1007/s41981-018-0001-x[Crossref], [CAS], Google Scholar63https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitFWgtLnL&md5=f2a8e142abf5b1c60f17f36dbe1a013bEsterification of benzoic acid in a continuous flow microwave reactorTajti, Adam; Toth, Nora; Balint, Erika; Keglevich, GyorgyJournal of Flow Chemistry (2018), 8 (1), 11-19CODEN: JFCOBJ; ISSN:2063-0212. (Akademiai Kiado)The direct esterification of benzoic acid with a series of aliph. alcs. was performed in a continuous flow microwave (MW) reactor. In the first stage, the reactivity of the alcs. towards benzoic acid was mapped in a batch MW reactor. Then, the different esterifications were optimized in the continuous reactor. All parameters including the temp. could be controlled. [Figure not available: see fulltext.].
- 64Bálint, E.; Tajti, Á.; Keglevich, G. Application of the Microwave Technique in Continuous Flow Processing of Organophosphorus Chemical Reactions. Materials 2019, 12 (5), 788, DOI: 10.3390/ma12050788[Crossref], [CAS], Google Scholar64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitlCltL3N&md5=a0d75fb6b0b0434499aa3458f179165dApplication of the microwave technique in continuous flow processing of organophosphorus chemical reactionsBalint, Erika; Tajti, Adam; Keglevich, GyorgyMaterials (2019), 12 (5), 788CODEN: MATEG9; ISSN:1996-1944. (MDPI AG)The microwave (MW) technique is an efficient tool in the realization of org. reactions, as well as in the anal. field and in the food industry. The continuous flow approach is of special interest as a promising way to scale-up MW-assisted syntheses. Besides summarizing the batch precedents, this review focuses on the utilization of the MW technique in the continuous-flow realization of organophosphorus transformations. The advantages of the continuous flow technique against the batch accomplishment are also shown. A few materials chem.-related applications are also mentioned.
- 65Kiss, N. Z.; Henyecz, R.; Keglevich, G. Continuous Flow Esterification of a H-Phosphinic Acid, and Transesterification of H-Phosphinates and H-Phosphonates under Microwave Conditions. Molecules 2020, 25 (3), 719, DOI: 10.3390/molecules25030719[Crossref], [CAS], Google Scholar65https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXksl2rs7c%253D&md5=ab65e85bb501c1b12d4b5541cd37d450Continuous flow esterification of a H-phosphinic acid, and transesterification of H-phosphinates and H-phosphonates under microwave conditionsKiss, Nora Zsuzsa; Henyecz, Reka; Keglevich, GyorgyMolecules (2020), 25 (3), 719CODEN: MOLEFW; ISSN:1420-3049. (MDPI AG)The microwave (MW)-assisted direct esterification of phenyl-H-phosphinic acid, transesterification of the alkyl phenyl-H-phosphinates so obtained, and the similar reaction of dibenzyl phosphite (DBP) were investigated in detail, and the batch accomplishments were translated into a continuous flow operation that, after optimization of the parameters, such as temp. and flow rate, proved to be more productive. Alcoholysis of DBP is a two-step process involving an intermediate phosphite with two different alkoxy groups. The latter species are of synthetic interest, as precursors for optically active reagents.
- 66Yue, J.; Falke, F. H.; Schouten, J. C.; Nijhuis, T. A. Microreactors with Integrated UV/Vis Spectroscopic Detection for Online Process Analysis under Segmented Flow. Lab Chip 2013, 13 (24), 4855, DOI: 10.1039/c3lc50876e[Crossref], [PubMed], [CAS], Google Scholar66https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslGgsLvJ&md5=ae109bc80ac8c46c8e1498f634c1447fMicroreactors with integrated UV/Vis spectroscopic detection for online process analysis under segmented flowYue, Jun; Falke, Floris H.; Schouten, Jaap C.; Nijhuis, T. AlexanderLab on a Chip (2013), 13 (24), 4855-4863CODEN: LCAHAM; ISSN:1473-0189. (Royal Society of Chemistry)Combining reaction and detection in multiphase microfluidic flow is becoming increasingly important for accelerating process development in microreactors. We report the coupling of UV/Vis spectroscopy with microreactors for online process anal. under segmented flow conditions. Two integration schemes are presented: one uses a cross-type flow-through cell subsequent to a capillary microreactor for detection in the transmission mode; the other uses embedded waveguides on a microfluidic chip for detection in the evanescent wave field. Model expts. reveal the capabilities of the integrated systems in real-time concn. measurements and segmented flow characterization. The application of such integration for process anal. during gold nanoparticle prodn. is demonstrated, showing its great potential in process monitoring in microreactors operated under segmented flow.
- 67Ryu, G.; Huang, J.; Hofmann, O.; Walshe, C. A.; Sze, J. Y. Y.; McClean, G. D.; Mosley, A.; Rattle, S. J.; DeMello, J. C.; DeMello, A. J.; Bradley, D. D. C. Highly Sensitive Fluorescence Detection System for Microfluidic Lab-on-a-Chip. Lab Chip 2011, 11 (9), 1664, DOI: 10.1039/c0lc00586j[Crossref], [PubMed], [CAS], Google Scholar67https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXksFSktLg%253D&md5=8b555570d7e2a53f011f29994b1bb824Highly sensitive fluorescence detection system for microfluidic lab-on-a-chipRyu, Gihan; Huang, Jingsong; Hofmann, Oliver; Walshe, Claire A.; Sze, Jasmine Y. Y.; McClean, Gareth D.; Mosley, Alan; Rattle, Simon J.; de Mello, John C.; de Mello, Andrew J.; Bradley, Donal D. C.Lab on a Chip (2011), 11 (9), 1664-1670CODEN: LCAHAM; ISSN:1473-0197. (Royal Society of Chemistry)We demonstrate a compact, low cost and practical fluorescence detection system for lab-on-a-chip applications. The system comprises a com. available InGaN light emitting diode (501 nm) as light source, an org. or silicon photodiode detector, absorptive dye coated color filters and linear and reflective polarisers. An injection molded polystyrene microfluidic chip is used as the platform for fluorescence immunoassays for cardiac markers myoglobin and CK-MB. The optical limit of detection (LOD) is measured using a TransFluoSphere suspension at 5.6 × 104 beads μl-1 which can be equated to ∼3 nM fluorescein equiv. concn. The LOD for the human plasma immunoassays is measured as 1.5 ng ml-1 for both myoglobin and CK-MB.
- 68Mozharov, S.; Nordon, A.; Littlejohn, D.; Wiles, C.; Watts, P.; Dallin, P.; Girkin, J. M. Improved Method for Kinetic Studies in Microreactors Using Flow Manipulation and Noninvasive Raman Spectrometry. J. Am. Chem. Soc. 2011, 133 (10), 3601– 3608, DOI: 10.1021/ja1102234[ACS Full Text
], [CAS], Google Scholar
68https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXitlams7o%253D&md5=e4a3f24798aba2ec652d916f6d1390d3Improved Method for Kinetic Studies in Microreactors Using Flow Manipulation and Noninvasive Raman SpectrometryMozharov, Sergey; Nordon, Alison; Littlejohn, David; Wiles, Charlotte; Watts, Paul; Dallin, Paul; Girkin, John M.Journal of the American Chemical Society (2011), 133 (10), 3601-3608CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)A novel method has been devised to derive kinetic information about reactions in microfluidic systems. Advantages have been demonstrated over conventional procedures for a Knoevenagel condensation reaction in terms of the time required to obtain the data (fivefold redn.) and the efficient use of reagents (tenfold redn.). The procedure is based on a step change from a low (e.g., 0.6 μL min-1) to a high (e.g., 14 μL min-1) flow rate and real-time noninvasive Raman measurements at the end of the flow line, which allows location-specific information to be obtained without the need to move the measurement probe along the microreactor channel. To validate the method, values of the effective reaction order n were obtained employing two different exptl. methodologies. Using these values of n, rate consts. k were calcd. and compared. The values of k derived from the proposed method at 10 and 40 °C were 0.0356 ± 0.0008 mol-0.3 dm0.9 s-1 (n = 1.3) and 0.24 ± 0.018 mol-0.1 dm0.3 s-1 (n = 1.1), resp., whereas the values obtained using a more laborious conventional methodol. were 0.0335 ± 0.0032 mol-0.4 dm1.2 s-1 (n = 1.4) at 10 °C and 0.244 ± 0.032 mol-0.3 dm0.9 s-1 (n = 1.3) at 40 °C. The new approach is not limited to anal. by Raman spectrometry and can be used with different techniques that can be incorporated into the end of the flow path to provide rapid measurements. - 69Galaverna, R.; Breitkreitz, M. C.; Pastre, J. C. Conversion of d-Fructose to 5-(Hydroxymethyl)Furfural: Evaluating Batch and Continuous Flow Conditions by Design of Experiments and In-Line FTIR Monitoring. ACS Sustain. Chem. Eng. 2018, 6 (3), 4220– 4230, DOI: 10.1021/acssuschemeng.7b04643
- 70Porta, R.; Benaglia, M.; Puglisi, A. Flow Chemistry: Recent Developments in the Synthesis of Pharmaceutical Products. Org. Process Res. Dev. 2016, 20 (1), 2– 25, DOI: 10.1021/acs.oprd.5b00325[ACS Full Text
], [CAS], Google Scholar
70https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhvFWmurfL&md5=2dbb53679df39f79b957a244c5d57fc5Flow Chemistry: Recent Developments in the Synthesis of Pharmaceutical ProductsPorta, Riccardo; Benaglia, Maurizio; Puglisi, AlessandraOrganic Process Research & Development (2016), 20 (1), 2-25CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)A review. Recently, application of the flow technologies for the prepn. of fine chems., such as natural products or Active Pharmaceutical Ingredients (APIs), has become very popular, esp. in academia. Although pharma industry still relies on multipurpose batch or semibatch reactors, it is evident that interest is arising toward continuous flow manufg. of org. mols., including highly functionalized and chiral compds. Continuous flow synthetic methodologies can also be easily combined to other enabling technologies, such as microwave irradn., supported reagents or catalysts, photochem., inductive heating, electrochem., new solvent systems, 3D printing, or microreactor technol. This combination could allow the development of fully automated process with an increased efficiency and, in many cases, improved sustainability. It has been also demonstrated that a safer manufg. of org. intermediates and APIs could be obtained under continuous flow conditions, where some synthetic steps that were not permitted for safety reasons can be performed with min. risk. In this review we focused our attention only on very recent advances in the continuous flow multistep synthesis of org. mols. which found application as APIs, esp. highlighting the contributions described in the literature from 2013 to 2015, including very recent examples not reported in any published review. Without claiming to be complete, we will give a general overview of different approaches, technologies, and synthetic strategies used so far, thus hoping to contribute to minimize the gap between academic research and pharmaceutical manufg. A general outlook about a quite young and relatively unexplored field of research, like stereoselective organocatalysis under flow conditions, will be also presented, and most significant examples will be described; our purpose is to illustrate all of the potentialities of continuous flow organocatalysis and offer a starting point to develop new methodologies for the synthesis of chiral drugs. Finally, some considerations on the perspectives and the possible, expected developments in the field are briefly discussed. - 71Cambié, D.; Bottecchia, C.; Straathof, N. J. W.; Hessel, V.; Noël, T. Applications of Continuous-Flow Photochemistry in Organic Synthesis, Material Science, and Water Treatment. Chem. Rev. 2016, 116 (17), 10276– 10341, DOI: 10.1021/acs.chemrev.5b00707[ACS Full Text
], [CAS], Google Scholar
71https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XjsVOjs7g%253D&md5=327c368f6e090142204920993c4faadaApplications of Continuous-Flow Photochemistry in Organic Synthesis, Material Science, and Water TreatmentCambie, Dario; Bottecchia, Cecilia; Straathof, Natan J. W.; Hessel, Volker; Noel, TimothyChemical Reviews (Washington, DC, United States) (2016), 116 (17), 10276-10341CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review. Continuous-flow photochem. in microreactors receives a lot of attention from researchers in academia and industry as this technol. provides reduced reaction times, higher selectivities, straightforward scalability, and the possibility to safely use hazardous intermediates and gaseous reactants. In this review, an up-to-date overview is given of photochem. transformations in continuous-flow reactors, including applications in org. synthesis, material science, and water treatment. In addn., the advantages of continuous-flow photochem. are pointed out and a thorough comparison with batch processing is presented. - 72Bogdan, A. R.; Dombrowski, A. W. Emerging Trends in Flow Chemistry and Applications to the Pharmaceutical Industry. J. Med. Chem. 2019, 62 (14), 6422– 6468, DOI: 10.1021/acs.jmedchem.8b01760[ACS Full Text
], [CAS], Google Scholar
72https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXjsF2lt78%253D&md5=fc973a27403ef4490bd2735e27765379Emerging Trends in Flow Chemistry and Applications to the Pharmaceutical IndustryBogdan, Andrew R.; Dombrowski, Amanda W.Journal of Medicinal Chemistry (2019), 62 (14), 6422-6468CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. The field of flow chem. has garnered considerable attention over the past 2 decades. This Perspective highlights many recent advances in the field of flow chem. and discusses applications to the pharmaceutical industry, from discovery to manufg. From a synthetic perspective, a no. of new enabling technologies are providing more rationale to run reactions in flow over batch techniques. Addnl., highly automated flow synthesis platforms have been developed with broad applicability across the pharmaceutical industry, ranging from advancing medicinal chem. programs to self-optimizing synthetic routes. A combination of simplified and automated systems is discussed, demonstrating how flow chem. solns. can be tailored to fit the specific needs of a project. - 73Akwi, F. M.; Watts, P. Continuous Flow Chemistry: Where Are We Now? Recent Applications, Challenges and Limitations. Chem. Commun. 2018, 54 (99), 13894– 13928, DOI: 10.1039/C8CC07427E[Crossref], [PubMed], [CAS], Google Scholar73https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXit1Cgt7nJ&md5=752fc8ed94d2ac9bd1c73e9e8d282df3Continuous flow chemistry: where are we now? Recent applications, challenges and limitationsAkwi, Faith M.; Watts, PaulChemical Communications (Cambridge, United Kingdom) (2018), 54 (99), 13894-13928CODEN: CHCOFS; ISSN:1359-7345. (Royal Society of Chemistry)A review. A general outlook of the changing face of chem. synthesis is provided in this article through recent applications of continuous flow processing in both industry and academia. The benefits, major challenges and limitations assocd. with the use of this mode of processing are also given due attention as an attempt to put into perspective the current position of continuous flow processing, either as an alternative or potential combinatory technol. for batch processing.
- 74Deadman, B. J.; Collins, S. G.; Maguire, A. R. Taming Hazardous Chemistry in Flow: The Continuous Processing of Diazo and Diazonium Compounds. Chem. - Eur. J. 2015, 21 (6), 2298– 2308, DOI: 10.1002/chem.201404348[Crossref], [PubMed], [CAS], Google Scholar74https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvFGisLbJ&md5=77e7ddadae87465b7f855e387e8f88daTaming Hazardous Chemistry in Flow: The Continuous Processing of Diazo and Diazonium CompoundsDeadman, Benjamin J.; Collins, Stuart G.; Maguire, Anita R.Chemistry - A European Journal (2015), 21 (6), 2298-2308CODEN: CEUJED; ISSN:0947-6539. (Wiley-VCH Verlag GmbH & Co. KGaA)A review is given. The synthetic utilities of the diazo and diazonium groups are matched only by their reputation for explosive decompn. Continuous processing technol. offers new opportunities to make and use these versatile intermediates at a range of scales with improved safety over traditional batch processes. In this minireview, the state of the art in the continuous flow processing of reactive diazo and diazonium species is discussed.
- 75Movsisyan, M.; Delbeke, E. I. P.; Berton, J. K. E. T.; Battilocchio, C.; Ley, S. V.; Stevens, C. V. Taming Hazardous Chemistry by Continuous Flow Technology. Chem. Soc. Rev. 2016, 45 (18), 4892– 4928, DOI: 10.1039/C5CS00902B[Crossref], [PubMed], [CAS], Google Scholar75https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xht1agtLjI&md5=bc926296a9288e9078943490fe2ff2feTaming hazardous chemistry by continuous flow technologyMovsisyan, M.; Delbeke, E. I. P.; Berton, J. K. E. T.; Battilocchio, C.; Ley, S. V.; Stevens, C. V.Chemical Society Reviews (2016), 45 (18), 4892-4928CODEN: CSRVBR; ISSN:0306-0012. (Royal Society of Chemistry)Over the last two decades, flow technologies have become increasingly popular in the field of org. chem., offering solns. for engineering and/or chem. problems. Flow reactors enhance the mass and heat transfer, resulting in rapid reaction mixing, and enable a precise control over the reaction parameters, increasing the overall process selectivity, efficiency and safety. These features allow chemists to tackle unexploited challenges in their work, with the ultimate objective making chem. more accessible for lab. and industrial applications, avoiding the need to store and handle toxic, reactive and explosive reagents. This review covers some of the latest and most relevant developments in the field of continuous flow chem. with the focus on hazardous reactions.
- 76Gomollón-Bel, F. Ten Chemical Innovations That Will Change Our World: IUPAC Identifies Emerging Technologies in Chemistry with Potential to Make Our Planet More Sustainable. Chem. Int. 2019, 41 (2), 12– 17, DOI: 10.1515/ci-2019-0203[Crossref], [CAS], Google Scholar76https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXmvFeisL8%253D&md5=2f31e701284d2a5af47a3b3b4bd51c58Ten Chemical Innovations That Will Change Our World: IUPAC identifies emerging technologies in Chemistry with potential to make our planet more sustainableGomollon-Bel, FernandoChemistry International (2019), 41 (2), 12-17CODEN: CINRDT; ISSN:0193-6484. (Walter de Gruyter, Inc.)The 2019 is a very special year in chem. 2019 marks two major anniversaries: the 100th anniversary of the founding of the International Union of Pure and Applied Chem. (IUPAC), and the 150th anniversary of Dimitri Mendeleev's first publication on the Periodic Table of Elements. IUPAC is the global organization that, among many other things, established a common language for chem. enabling scientific research, education, and trade. In a similar manner, Mendeleev's system classified all the elements that were known at the time, and even predicted the existence of elements that would only come to be discovered years later. These two anniversaries are closely entwined, as IUPAC has played a major role developing of the modern Periodic Table by ensuring that the most authoritative version of the table is accessible to everyone , establishing names and symbols for the newly discovered elements, and also constantly reviewing its accuracy through the IUPAC Commission on Isotopic Abundances and Atomic Wts.
- 77Fülöp, Z.; Szemesi, P.; Bana, P.; Éles, J.; Greiner, I. Evolution of Flow-Oriented Design Strategies in the Continuous Preparation of Pharmaceuticals. React. Chem. Eng. 2020, 5 (9), 1527– 1555, DOI: 10.1039/D0RE00273A
- 78Gutmann, B.; Cantillo, D.; Kappe, C. O. Continuous-Flow Technology-A Tool for the Safe Manufacturing of Active Pharmaceutical Ingredients. Angew. Chem., Int. Ed. 2015, 54 (23), 6688– 6728, DOI: 10.1002/anie.201409318[Crossref], [CAS], Google Scholar78https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXosVGnt7k%253D&md5=49251387f0f3378dff4e710afe9fbca8Continuous-Flow Technology-A Tool for the Safe Manufacturing of Active Pharmaceutical IngredientsGutmann, Bernhard; Cantillo, David; Kappe, C. OliverAngewandte Chemie, International Edition (2015), 54 (23), 6688-6728CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. In the past few years, continuous-flow reactors with channel dimensions in the micro- or millimeter region have found widespread application in org. synthesis. The characteristic properties of these reactors are their exceptionally fast heat and mass transfer. In microstructured devices of this type, virtually instantaneous mixing can be achieved for all but the fastest reactions. Similarly, the accumulation of heat, formation of hot spots, and dangers of thermal runaways can be prevented. As a result of the small reactor vols., the overall safety of the process is significantly improved, even when harsh reaction conditions are used. Thus, microreactor technol. offers a unique way to perform ultrafast, exothermic reactions, and allows the execution of reactions which proceed via highly unstable or even explosive intermediates. This Review discusses recent literature examples of continuous-flow org. synthesis where hazardous reactions or extreme process windows have been employed, with a focus on applications of relevance to the prepn. of pharmaceuticals.
- 79Ley, S. V.; Fitzpatrick, D. E.; Ingham, R. J.; Myers, R. M. Organic Synthesis: March of the Machines. Angew. Chem., Int. Ed. 2015, 54 (11), 3449– 3464, DOI: 10.1002/anie.201410744[Crossref], [CAS], Google Scholar79https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXos1altQ%253D%253D&md5=821a7530bdd95a545b396d9a6922a414Organic Synthesis: March of the MachinesLey, Steven V.; Fitzpatrick, Daniel E.; Ingham, Richard. J.; Myers, Rebecca M.Angewandte Chemie, International Edition (2015), 54 (11), 3449-3464CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. Org. synthesis is changing; in a world where budgets are constrained and the environmental impacts of practice are scrutinized, it is increasingly recognized that the efficient use of human resource is just as important as material use. New technologies and machines have found use as methods for transforming the way things are done, addressing these issues encountered in research labs. by enabling chemists to adopt a more holistic systems approach in their work. Modern developments in this area promote a multi-disciplinary approach and work is more efficient as a result. This Review focuses on the concepts, procedures and methods that have far-reaching implications in the chem. world. Technologies have been grouped as topics of opportunity and their recent applications in innovative research labs. are described.
- 80Gioiello, A.; Piccinno, A.; Lozza, A. M.; Cerra, B. The Medicinal Chemistry in the Era of Machines and Automation: Recent Advances in Continuous Flow Technology. J. Med. Chem. 2020, 63 (13), 6624– 6647, DOI: 10.1021/acs.jmedchem.9b01956[ACS Full Text
], [CAS], Google Scholar
80https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXivFyjsL8%253D&md5=0a0ab072bc075cb79a107f38fa18cdd2The Medicinal Chemistry in the Era of Machines and Automation: Recent Advances in Continuous Flow TechnologyGioiello, Antimo; Piccinno, Alessandro; Lozza, Anna Maria; Cerra, BrunoJournal of Medicinal Chemistry (2020), 63 (13), 6624-6647CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Medicinal chem. plays a fundamental and underlying role in chem. biol., pharmacol., and medicine to discover safe and efficacious drugs. Small mol. medicinal chem. relies on iterative learning cycles composed of compd. design, synthesis, testing, and data anal. to provide new chem. probes and lead compds. for novel and druggable targets. Using traditional approaches, the time from hypothesis to obtaining the results can be protracted, thus limiting the no. of compds. that can be advanced into clin. studies. This challenge can be tackled with the recourse of enabling technologies that are showing great potential in improving the drug discovery process. In this Perspective, we highlight recent developments towards innovative medicinal chem. strategies based on continuous flow systems coupled with automation and bioassays. After a discussion of the aims and concepts, we describe equipment and representative examples of automated flow systems and end-to-end prototypes realized to expedite medicinal chem. discovery cycles. - 81Baumann, M.; Moody, T. S.; Smyth, M.; Wharry, S. A Perspective on Continuous Flow Chemistry in the Pharmaceutical Industry. Org. Process Res. Dev. 2020, 24, 1802 DOI: 10.1021/acs.oprd.9b00524 .[ACS Full Text
], [CAS], Google Scholar
81https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXmtVGrtw%253D%253D&md5=01603875a35243e7974384146b6c35ccA Perspective on Continuous Flow Chemistry in the Pharmaceutical IndustryBaumann, Marcus; Moody, Thomas S.; Smyth, Megan; Wharry, ScottOrganic Process Research & Development (2020), 24 (10), 1802-1813CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)A review. Continuous flow manuf. is an innovative technol. platform, which is gaining momentum within the pharmaceutical industry. The key advantages of continuous flow include faster and safer reactions, which can be more environmentally friendly, smaller footprint, better quality product, and critically, the ability to perform chem. that is difficult or impossible to do in batch mode. Globally, significant efforts have been made to develop the manufg. flexibility and robustness of processes used to produce chems. in a continuous way, yet despite these scientific developments, a major challenge for industry is the established application of flow technol. to com. relevant examples. The identification of opportunities to apply flow solns. to current processes is also crit. to the success of this new technol. for pharmaceutical and fine chem. companies. This review highlights industrial hurdles and the importance of education and showcases recent (2018-2019) and relevant industrial examples where utilization of flow technol. has been successfully performed. - 82Malet-Sanz, L.; Susanne, F. Continuous Flow Synthesis. a Pharma Perspective. J. Med. Chem. 2012, 55, 4062– 4098. DOI: 10.1021/jm2006029 .[ACS Full Text
], [CAS], Google Scholar
82https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsVSqtLc%253D&md5=23f3d763f8072c7404f54076f9d2d78aContinuous Flow Synthesis. A Pharma PerspectiveMalet-Sanz, Laia; Susanne, FlavienJournal of Medicinal Chemistry (2012), 55 (9), 4062-4098CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Continuous flow chem. as a technique and the latest developments in the field are being reviewed from a Pharma point of view. - 83Britton, J.; Raston, C. L. Multi-Step Continuous-Flow Synthesis. Chem. Soc. Rev. 2017, 46 (5), 1250– 1271, DOI: 10.1039/C6CS00830E[Crossref], [PubMed], [CAS], Google Scholar83https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXht1yjtr0%253D&md5=abcfc0e54556e7cd18445305ec6994fdMulti-step continuous-flow synthesisBritton, Joshua; Raston, Colin L.Chemical Society Reviews (2017), 46 (5), 1250-1271CODEN: CSRVBR; ISSN:0306-0012. (Royal Society of Chemistry)A review. This review article focused on multi-step continuous-flow methodol., which had applications in synthesis of active pharmaceutical ingredients, natural products, and commodity chems. This review described the advancements while highlighted the rapid progress, benefits, and diversification of this expanding field.
- 84Wegner, J.; Ceylan, S.; Kirschning, A. Flow Chemistry – A Key Enabling Technology for (Multistep) Organic Synthesis. Adv. Synth. Catal. 2012, 354 (1), 17– 57, DOI: 10.1002/adsc.201100584[Crossref], [CAS], Google Scholar84https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xmt1GrtQ%253D%253D&md5=143e37811c92473a296cb64c1b8fda86Flow Chemistry - A Key Enabling Technology for (Multistep) Organic SynthesisWegner, Jens; Ceylan, Sascha; Kirschning, AndreasAdvanced Synthesis & Catalysis (2012), 354 (1), 17-57CODEN: ASCAF7; ISSN:1615-4150. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. Lab. scaled flow-through processes have seen an explosive development over the past decade and have become an enabling technol. for improving synthetic efficiency through automation and process optimization. Practically, flow devices are a crucial link between bench chemists and process engineers. The present review focuses on two unique aspects of modern flow chem. where substantial advantages over the corresponding batch processes have become evident. Flow chem. being one out of several enabling technologies can ideally be combined with other enabling technologies such as energy input. This may be achieved in form of heat to create supercrit. conditions. Here, indirect methods such as microwave irradn. and inductive heating have seen widespread applications. Also radiation can efficiently be used to carry out photochem. reactions in a highly practical and scalable manner. A second unique aspect of flow chem. compared to batch chem. is assocd. with the option to carry out multistep synthesis by designing a flow set-up composed of several flow reactors. Besides their role as chem. reactors these can act as elements for purifn. or solvent switch.
- 85Baumann, M.; Baxendale, I. R. The Synthesis of Active Pharmaceutical Ingredients (APIs) Using Continuous Flow Chemistry. Beilstein J. Org. Chem. 2015, 11, 1194– 1219, DOI: 10.3762/bjoc.11.134[Crossref], [PubMed], [CAS], Google Scholar85https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXht12qur7N&md5=cc70c6db28d1139d8b20791e4f58d99cThe synthesis of active pharmaceutical ingredients (APIs) using continuous flow chemistryBaumann, Marcus; Baxendale, Ian R.Beilstein Journal of Organic Chemistry (2015), 11 (), 1194-1219CODEN: BJOCBH; ISSN:1860-5397. (Beilstein-Institut zur Foerderung der Chemischen Wissenschaften)A review. This review article aims to illustrate the holistic systems approach and diverse applications of flow chem. to the prepn. of pharmaceutically active mols., demonstrating the value of this strategy towards every aspect ranging from synthesis, in-line anal. and purifn. to final formulation and tableting. Although this review will primarily conc. on large scale continuous processing, addnl. selected syntheses using micro or meso-scaled flow reactors will be exemplified for key transformations and process control. It is hoped that the reader will gain an appreciation of the innovative technol. and transformational nature that flow chem. can leverage to an overall process.
- 86Pieber, B.; Gilmore, K.; Seeberger, P. H. Integrated Flow Processing — Challenges in Continuous Multistep Synthesis. J. Flow Chem. 2017, 7 (3–4), 129– 136, DOI: 10.1556/1846.2017.00016[Crossref], [CAS], Google Scholar86https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXntFygurc%253D&md5=e137736c47bd548090101fed97244e95Integrated flow processing - challenges in continuous multistep synthesisPieber, Bartholomaeus; Gilmore, Kerry; Seeberger, Peter H.Journal of Flow Chemistry (2017), 7 (3-4), 129-136CODEN: JFCOBJ; ISSN:2062-249X. (Akademiai Kiado)The way org. multistep synthesis is performed is changing due to the adoption of flow chem. techniques, which has enabled the development of improved methods to make complex mols. The modular nature of the technique provides not only access to target mols. via linear flow approaches but also for the targeting of structural cores with single systems. This perspective article summarizes the state of the art of continuous multistep synthesis and discusses the main challenges and opportunities in this area.
- 87Weeranoppanant, N.; Adamo, A. In-Line Purification: A Key Component to Facilitate Drug Synthesis and Process Development in Medicinal Chemistry. ACS Med. Chem. Lett. 2020, 11 (1), 9– 15, DOI: 10.1021/acsmedchemlett.9b00491[ACS Full Text
], [CAS], Google Scholar
87https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitlygt7%252FI&md5=a8218c5cf043afd8ae33d918c2698356In-Line Purification: A Key Component to Facilitate Drug Synthesis and Process Development in Medicinal ChemistryWeeranoppanant, Nopphon; Adamo, AndreaACS Medicinal Chemistry Letters (2020), 11 (1), 9-15CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)A review. In-line purifn. is an important tool for flow chem. It enables effective handling of unstable intermediates and integration of multiple synthetic steps. The integrated flow synthesis is useful for drug synthesis and process development in medicinal chem. In this article, we overview current states of in-line purifn. methods. In particular, we focus on four common methods: scavenger column, distn., nanofiltration, and extn. Examples of their applications are provided. - 88Örkényi, R.; Éles, J.; Faigl, F.; Vincze, P.; Prechl, A.; Szakács, Z.; Kóti, J.; Greiner, I. Continuous Synthesis and Purification by Coupling a Multistep Flow Reaction with Centrifugal Partition Chromatography. Angew. Chem., Int. Ed. 2017, 56 (30), 8742– 8745, DOI: 10.1002/anie.201703852[Crossref], [CAS], Google Scholar88https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1cnhs1Oqsg%253D%253D&md5=72cf0d6f12d1e1c345960f4e2e4f34e1Continuous Synthesis and Purification by Coupling a Multistep Flow Reaction with Centrifugal Partition ChromatographyOrkenyi Robert; Faigl Ferenc; Eles Janos; Vincze Peter; Prechl Anita; Szakacs Zoltan; Koti Janos; Greiner IstvanAngewandte Chemie (International ed. in English) (2017), 56 (30), 8742-8745 ISSN:.Continuous-flow multistep synthesis is combined with quasi-continuous final-product purification to produce pure products from crude reaction mixtures. In the nucleophilic aromatic substitution of 2,4-difluoronitrobenzene with morpholine followed by a heterogeneous catalytic hydrogenation, the desired monosubstituted product can be continuously separated from the co- and by-products in a purity of over 99 % by coupling a flow reactor sequence to a multiple dual-mode (MDM) centrifugal partition chromatography (CPC) device. This purification technique has many advantages over HPLC, such as higher resolution and no need for column replacement or silica recycling, and it does not suffer from irreversible adsorption.
- 89Hughes, D. L. Applications of Flow Chemistry in Drug Development: Highlights of Recent Patent Literature. Org. Process Res. Dev. 2018, 22 (1), 13– 20, DOI: 10.1021/acs.oprd.7b00363[ACS Full Text
], [CAS], Google Scholar
89https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXitVaksrfN&md5=eb32ba4b9966e7101fe63318e5d5daa9Applications of Flow Chemistry in Drug Development: Highlights of Recent Patent LiteratureHughes, David L.Organic Process Research & Development (2018), 22 (1), 13-20CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)Flow chem. is playing an increasingly important role in API process development and manuf. in the pharmaceutical and fine chem. industry. The current article reviews routes to approved drugs that employ at least one continuous flow step, disclosed in the patent literature during 2016 and 2017, with a further constraint that the chem. has not been published in a journal article. - 90Bogdan, A. R.; Poe, S. L.; Kubis, D. C.; Broadwater, S. J.; McQuade, D. T. The Continuous-Flow Synthesis of Ibuprofen. Angew. Chem., Int. Ed. 2009, 48 (45), 8547– 8550, DOI: 10.1002/anie.200903055[Crossref], [PubMed], [CAS], Google Scholar90https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtlWmurjF&md5=d26006bf016c89749cb084acdbbac08bThe Continuous-Flow Synthesis of IbuprofenBogdan, Andrew R.; Poe, Sarah L.; Kubis, Daniel C.; Broadwater, Steven J.; McQuade, D. TylerAngewandte Chemie, International Edition (2009), 48 (45), 8547-8550, S8547/1-S8547/28CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)A 3-step, continuous flow microreactor synthesis of ibuprofen comprised (i) TfOH-catalyzed Friedel-Crafts acylation of isobutylbenzene with propionic acid; (ii) oxidative 1,2-aryl migration of resultant 4-isobutylpropiophenone in presence of tri-Me orthoformate, PhI(OAc)2 and TfOH (from step i); and (iii) sapon. of resultant ibuprofen Me ester with KOH to produce ibuprofen in 68% crude yield and 51% yield after recrystn. A 70% yield was obtained for the first two steps, with quant. conversion of 4-isobutylpropiophenone to ibuprofen Me ester.
- 91Viviano, M.; Glasnov, T. N.; Reichart, B.; Tekautz, G.; Kappe, C. O. A Scalable Two-Step Continuous Flow Synthesis of Nabumetone and Related 4-Aryl-2-Butanones. Org. Process Res. Dev. 2011, 15 (4), 858– 870, DOI: 10.1021/op2001047[ACS Full Text
], [CAS], Google Scholar
91https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXmvFCht78%253D&md5=c1e1fe45bdbd3fb0919ec4a958d31679A Scalable Two-Step Continuous Flow Synthesis of Nabumetone and Related 4-Aryl-2-butanonesViviano, Monica; Glasnov, Toma N.; Reichart, Benedik; Tekautz, Guenter; Kappe, C. OliverOrganic Process Research & Development (2011), 15 (4), 858-870CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)Three different continuous flow strategies for the generation of important 4-aryl-2-butanone derivs. including the anti-inflammatory drug nabumetone [4-(6-methoxy-2-naphthalenyl)-2-butanone] and the aroma compds. raspberry ketone [4-(4-hydroxyphenyl)-2-butanone] and its Me ether [4-(4-methoxyphenyl)-2-butanone] were evaluated. All three protocols involve the initial prepn. of the corresponding 4-aryl-3-buten-2-ones via Mizoroki-Heck, Wittig, or aldol strategies, which is then followed by selective hydrogenation of the C=C double bond to the desired 4-aryl-2-butanones. The synthetic routes to 4-aryl-3-buten-2-ones were first optimized/intensified on small scale to reaction times of 1-10 min using batch microwave heating technol. and then translated to a scalable continuous flow process employing com. available stainless steel capillary tube reactors. For the synthesis of 4-(4-methoxyphenyl)-3-buten-2-one a further scale-up using a custom-built mesofluidic mini-plant flow system capable of processing several liters per h was designed to further expand the scale of the process. The final hydrogenation step was performed using a fixed-bed continuous flow hydrogenator employing Ra/Ni as a catalyst. - 92Ahmed-Omer, B.; Sanderson, A. J. Preparation of Fluoxetine by Multiple Flow Processing Steps. Org. Biomol. Chem. 2011, 9 (10), 3854, DOI: 10.1039/c0ob00906g[Crossref], [PubMed], [CAS], Google Scholar92https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXlt1Giu7g%253D&md5=8ed7ee256dd69240464523edec8d17b4Preparation of fluoxetine by multiple flow processing stepsAhmed-Omer, Batoul; Sanderson, Adam J.Organic & Biomolecular Chemistry (2011), 9 (10), 3854-3862CODEN: OBCRAK; ISSN:1477-0520. (Royal Society of Chemistry)Microflow technol. is established as a modern and fashionable tool in synthetic org. chem., bringing great improvement and potential, on account of a series of advantages over flask methods. The study presented here focuses on the application of flow chem. process in performing an efficient multiple step syntheses of (±)-fluoxetine as an alternative to conventional synthetic methods, and one of the few examples of total synthesis accomplished by flow technique.
- 93Lévesque, F.; Seeberger, P. H. Continuous-Flow Synthesis of the Anti-Malaria Drug Artemisinin. Angew. Chem., Int. Ed. 2012, 51 (7), 1706– 1709, DOI: 10.1002/anie.201107446[Crossref], [CAS], Google Scholar93https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xnt1yqtg%253D%253D&md5=73f32b46fd071ff8017fcf80f69a330eContinuous-Flow Synthesis of the Anti-Malaria Drug ArtemisininLevesque, Francois; Seeberger, Peter H.Angewandte Chemie, International Edition (2012), 51 (7), 1706-1709, S1706/1-S1706/20CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)Artemisinin was manufd. by continuous-flow, 3-step photochem. transformation involving a singlet-oxygen-induced ene reaction of dihydroartemisinic acid and addn. of triplet O.
- 94Hopkin, M. D.; Baxendale, I. R.; Ley, S. V. An Expeditious Synthesis of Imatinib and Analogues Utilising Flow Chemistry Methods. Org. Biomol. Chem. 2013, 11 (11), 1822– 1839, DOI: 10.1039/C2OB27002A[Crossref], [PubMed], [CAS], Google Scholar94https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXjtVCht7s%253D&md5=dacefaa178a62dd1e7c8a3f89fa83658An expeditious synthesis of imatinib and analogues utilizing flow chemistry methodsHopkin, Mark D.; Baxendale, Ian R.; Ley, Steven V.Organic & Biomolecular Chemistry (2013), 11 (11), 1822-1839CODEN: OBCRAK; ISSN:1477-0520. (Royal Society of Chemistry)A flow-based route to imatinib, the API of Gleevec, was developed and the general procedure then used to generate a no. of analogs which were screened for biol. activity against Abl1. The flow synthesis required minimal manual intervention and was achieved despite the poor soly. of many of the reaction components.
- 95Murray, P. R. D.; Browne, D. L.; Pastre, J. C.; Butters, C.; Guthrie, D.; Ley, S. V. Continuous Flow-Processing of Organometallic Reagents Using an Advanced Peristaltic Pumping System and the Telescoped Flow Synthesis of (E/Z)-Tamoxifen. Org. Process Res. Dev. 2013, 17 (9), 1192– 1208, DOI: 10.1021/op4001548[ACS Full Text
], [CAS], Google Scholar
95https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXht1ymt7nJ&md5=9971d8ef8e035e947bed0537c44e3791Continuous Flow-Processing of Organometallic Reagents Using an Advanced Peristaltic Pumping System and the Telescoped Flow Synthesis of (E/Z)-TamoxifenMurray, Philip R. D.; Browne, Duncan L.; Pastre, Julio C.; Butters, Chris; Guthrie, Duncan; Ley, Steven V.Organic Process Research & Development (2013), 17 (9), 1192-1208CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)A review. A new enabling technol. for the pumping of organometallic reagents such as n-butyllithium, Grignard reagents, and DIBAL-H is reported, which utilizes a newly developed, chem. resistant, peristaltic pumping system. Several representative examples of its use in common transformations using these reagents, including metal-halogen exchange, addn., addn.-elimination, conjugate addn., and partial redn., are reported along with examples of telescoping of the anionic reaction products. This platform allows for truly continuous pumping of these highly reactive substances (and examples are demonstrated over periods of several hours) to generate multigram quantities of products. This work culminates in an approach to the telescoped synthesis of (E/Z)-tamoxifen using continuous-flow organometallic reagent-mediated transformations. - 96Snead, D. R.; Jamison, T. F. End-to-End Continuous Flow Synthesis and Purification of Diphenhydramine Hydrochloride Featuring Atom Economy, in-Line Separation, and Flow of Molten Ammonium Salts. Chem. Sci. 2013, 4 (7), 2822, DOI: 10.1039/c3sc50859e[Crossref], [CAS], Google Scholar96https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXos1yru7w%253D&md5=4c0108ae6a10a97c32fd9463c779a7d5End-to-end continuous flow synthesis and purification of diphenhydramine hydrochloride featuring atom economy, in-line separation, and flow of molten ammonium saltsSnead, David R.; Jamison, Timothy F.Chemical Science (2013), 4 (7), 2822-2827CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)A continuous end-to-end synthesis and purifn. of diphenhydramine hydrochloride featuring atom economy and waste minimization is described. Combining a 1 : 1 molar ratio of the two starting material streams (chlorodiphenylmethane and N,N-dimethylaminoethanol) in the absence of addnl. solvent at high temp. gives the target compd. directly as a molten salt (ionic liq. above 168 °C) in high yield. This represents the first example of continuous active pharmaceutical ingredient (API) prodn. in this manner. Six of the twelve principles of green chem. as defined by the American Chem. Society are achieved, most prominently waste minimization and atom economy.
- 97Kupracz, L.; Kirschning, A. Multiple Organolithium Generation in the Continuous Flow Synthesis of Amitriptyline. Adv. Synth. Catal. 2013, 355 (17), 3375– 3380, DOI: 10.1002/adsc.201300614[Crossref], [CAS], Google Scholar97https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslygtLzF&md5=b197b36d4ccfbf5382bea3c1cb690c5cMultiple Organolithium Generation in the Continuous Flow Synthesis of AmitriptylineKupracz, Lukas; Kirschning, AndreasAdvanced Synthesis & Catalysis (2013), 355 (17), 3375-3380CODEN: ASCAF7; ISSN:1615-4150. (Wiley-VCH Verlag GmbH & Co. KGaA)A continuous flow protocol (continuous flow reactor) for the prepn. of the tricyclic antidepressant (TCA) amitriptyline is reported. The advantages of flow chem. when handling organometallic agents as well as when performing reaction with gases are demonstrated. Continuous lithiation combined with carboxylation and the Parham cyclization, a Grignard addn. and thermolytic water elimination by inductive heating are key features of the multistep protocol. The synthesis of the target compd. was achieved by a reaction of 5-[3-(dimethylamino)propyl]-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5-ol with [3-(dimethylamino)propyl]magnesium chloride. Only small amts. of butyllithium were required in the lithiation step (process safety).
- 98Hartwig, J.; Ceylan, S.; Kupracz, L.; Coutable, L.; Kirschning, A. Heating under High-Frequency Inductive Conditions: Application to the Continuous Synthesis of the Neurolepticum Olanzapine (Zyprexa). Angew. Chem., Int. Ed. 2013, 52 (37), 9813– 9817, DOI: 10.1002/anie.201302239[Crossref], [CAS], Google Scholar98https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtFOiu7vI&md5=8f9d732503d680bf339e447de97ea706Heating under High-Frequency Inductive Conditions: Application to the Continuous Synthesis of the Neurolepticum Olanzapine (Zyprexa)Hartwig, Jan; Ceylan, Sascha; Kupracz, Lukas; Coutable, Ludovic; Kirschning, AndreasAngewandte Chemie, International Edition (2013), 52 (37), 9813-9817CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)High-frequency inductive heating and flow chem. are an ideal match for high-temp. synthesis. This was demonstrated in the multistep flow synthesis of the neurolepticum olanzapine (Zyprexa) that included three reactions with inductive heating and two purifn. steps conducted as continuous processes.
- 99Zhang, P.; Russell, M. G.; Jamison, T. F. Continuous Flow Total Synthesis of Rufinamide. Org. Process Res. Dev. 2014, 18 (11), 1567– 1570, DOI: 10.1021/op500166n[ACS Full Text
], [CAS], Google Scholar
99https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhsVCnu7vO&md5=2e1ff3f346e9c1571baf05747bd42105Continuous Flow Total Synthesis of RufinamideZhang, Ping; Russell, M. Grace; Jamison, Timothy F.Organic Process Research & Development (2014), 18 (11), 1567-1570CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)Small mols. bearing 1,2,3-triazole functionalities are important intermediates and pharmaceuticals. Common methods to access the triazole moiety generally require the generation and isolation of org. azide intermediates. Continuous flow synthesis provides the opportunity to synthesize and consume the energetic organoazides, without accumulation thereof. In this report, we described a continuous synthesis of the antiseizure medication rufinamide. This route is convergent and features copper tubing reactor-catalyzed cycloaddn. reaction. Each of the three chem. steps enjoys significant benefits and has several advantages by being conducted in flow. The total av. residence time of the synthesis is approx. 11 min, and rufinamide is obtained in 92% overall yield. - 100Polster, C. S.; Cole, K. P.; Burcham, C. L.; Campbell, B. M.; Frederick, A. L.; Hansen, M. M.; Harding, M.; Heller, M. R.; Miller, M. T.; Phillips, J. L.; Pollock, P. M.; Zaborenko, N. Pilot-Scale Continuous Production of LY2886721: Amide Formation and Reactive Crystallization. Org. Process Res. Dev. 2014, 18 (11), 1295– 1309, DOI: 10.1021/op500204z[ACS Full Text
], [CAS], Google Scholar
100https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhslChtr7E&md5=2777d9d7600b877872d5bc74e367daf9Pilot-Scale Continuous Production of LY2886721: Amide Formation and Reactive CrystallizationPolster, Christopher S.; Cole, Kevin P.; Burcham, Christopher L.; Campbell, Bradley M.; Frederick, Andrea L.; Hansen, Marvin M.; Harding, Molly; Heller, Michael R.; Miller, Michael T.; Phillips, Joseph L.; Pollock, Patrick M.; Zaborenko, NikolayOrganic Process Research & Development (2014), 18 (11), 1295-1309CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)The design, development, and implementation of a pilot-scale continuous Schotten-Baumann amide bond formation and reactive crystn. to afford LY2886721 is described. The material met all API quality attributes and was comparable to material produced by a defined batch process. The scalability of the reaction and crystn. processes was confirmed during the development process. The pilot-scale equipment set was contained in a walk-in fume hood and operated at a prodn. rate of 3 kg/day in a 72 h continuous run. Significant tech. and business drivers for running the process in continuous flow mode were proposed and examd. during development. The continuous process provided for lab hood commercialization and provided for minimal material at risk in the process. The demonstration also confirmed the risk inherent to operation of a tubular reactor under supersatd. conditions, and fouling occurred in the plug flow reactor. Fouling also occurred in the crystallizer. Recognizing these deficiencies, the process operated within the footprint of a std. walk-in fume hood, providing a successful demonstration of the opportunities afforded by continuous processing for low vol. pharmaceuticals. - 101Heider, P. L.; Born, S. C.; Basak, S.; Benyahia, B.; Lakerveld, R.; Zhang, H.; Hogan, R.; Buchbinder, L.; Wolfe, A.; Mascia, S.; Evans, J. M. B.; Jamison, T. F.; Jensen, K. F. Development of a Multi-Step Synthesis and Workup Sequence for an Integrated, Continuous Manufacturing Process of a Pharmaceutical. Org. Process Res. Dev. 2014, 18 (3), 402– 409, DOI: 10.1021/op400294z[ACS Full Text
], [CAS], Google Scholar
101https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitVarsbY%253D&md5=32e14c670c851d83d7c9a3af5cc18c0fDevelopment of a Multi-Step Synthesis and Workup Sequence for an Integrated, Continuous Manufacturing Process of a PharmaceuticalHeider, Patrick L.; Born, Stephen C.; Basak, Soubir; Benyahia, Brahim; Lakerveld, Richard; Zhang, Haitao; Hogan, Rachael; Buchbinder, Louis; Wolfe, Aaron; Mascia, Salvatore; Evans, James M. B.; Jamison, Timothy F.; Jensen, Klavs F.Organic Process Research & Development (2014), 18 (3), 402-409CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)The development and operation of the synthesis and workup steps of a fully integrated, continuous manufg. plant for synthesizing aliskiren, a small mol. pharmaceutical, are presented. The plant started with advanced intermediates, two synthetic steps away from the final active pharmaceutical ingredient, and ended with finished tablets. The entire process was run on several occasions, with the data presented herein corresponding to a 240 h run at a nominal throughput of 41 g h-1 of aliskiren. The first reaction was performed solvent-free in a molten condition at a high temp., achieving high yields (90%) and avoiding solid handling and a long residence time (due to higher concns. compared to dil. conditions when run at lower temps. in a solvent). The resulting stream was worked-up inline using liq.-liq. extn. with membrane-based separators that were scaled-up from microfluidic designs. The second reaction involved a Boc deprotection, using aq. HCl that was rapidly quenched with aq. NaOH using an inline pH measurement to control NaOH addn. The reaction maintained high yields (90-95%) under closed-loop control despite process disturbances. - 102Correia, C. A.; Gilmore, K.; McQuade, D. T.; Seeberger, P. H. A Concise Flow Synthesis of Efavirenz. Angew. Chem., Int. Ed. 2015, 54 (16), 4945– 4948, DOI: 10.1002/anie.201411728[Crossref], [CAS], Google Scholar102https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXkvF2nu7s%253D&md5=4811f0b57f4b876bdda5107d569a69d1A Concise Flow Synthesis of EfavirenzCorreia, Camille A.; Gilmore, Kerry; McQuade, D. Tyler; Seeberger, Peter H.Angewandte Chemie, International Edition (2015), 54 (16), 4945-4948CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)Efavirenz is an essential medicine for the treatment of HIV, which is still inaccessible to millions of people worldwide. A novel, semi-continuous process provides rac-Efavirenz with an overall yield of 45% from 1,4-dichlorobenzene. This streamlined proof-of-principle synthesis relies on the efficient copper-catalyzed formation of an aryl isocyanate and a subsequent intramol. cyclization to install the carbamate core of Efavirenz in one step. This three-step method represents the shortest synthesis of the racemic analog of this life-saving drug to date.
- 103Tsubogo, T.; Oyamada, H.; Kobayashi, S. Multistep Continuous-Flow Synthesis of (R)- and (S)-Rolipram Using Heterogeneous Catalysts. Nature 2015, 520 (7547), 329– 332, DOI: 10.1038/nature14343[Crossref], [PubMed], [CAS], Google Scholar103https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXmsF2gtLg%253D&md5=ee58e64a6f5ed49ccf39ddecc3ccb6a0Multistep continuous-flow synthesis of (R)- and (S)-rolipram using heterogeneous catalystsTsubogo, Tetsu; Oyamada, Hidekazu; Kobayashi, ShuNature (London, United Kingdom) (2015), 520 (7547), 329-332CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Chem. manufg. is conducted using either batch systems or continuous-flow systems. Flow systems have several advantages over batch systems, particularly in terms of productivity, heat and mixing efficiency, safety, and reproducibility. However, for over half a century, pharmaceutical manufg. has used batch systems because the synthesis of complex mols. such as drugs has been difficult to achieve with continuous-flow systems. Here we describe the continuous-flow synthesis of drugs using only columns packed with heterogeneous catalysts. Com. available starting materials were successively passed through four columns contg. achiral and chiral heterogeneous catalysts to produce (R)-rolipram, an anti-inflammatory drug and one of the family of γ-aminobutyric acid (GABA) derivs. In addn., simply by replacing a column packed with a chiral heterogeneous catalyst with another column packed with the opposing enantiomer, we obtained antipole (S)-rolipram. Similarly, we also synthesized (R)-phenibut, another drug belonging to the GABA family. These flow systems are simple and stable with no leaching of metal catalysts. Our results demonstrate that multistep (eight steps in this case) chem. transformations for drug synthesis can proceed smoothly under flow conditions using only heterogeneous catalysts, without the isolation of any intermediates and without the sepn. of any catalysts, co-products, byproducts, and excess reagents. We anticipate that such syntheses will be useful in pharmaceutical manufg.
- 104Martin, A. D.; Siamaki, A. R.; Belecki, K.; Gupton, B. F. A Flow-Based Synthesis of Telmisartan. J. Flow Chem. 2015, 5 (3), 145– 147, DOI: 10.1556/JFC-D-15-00002[Crossref], [CAS], Google Scholar104https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXjt1Olur0%253D&md5=346efd4405277a3bf98703e43837e010A flow-based synthesis of telmisartanMartin, Alex D.; Siamaki, Ali R.; Belecki, Katherine; Gupton, B. FrankJournal of Flow Chemistry (2015), 5 (3), 145-147CODEN: JFCOBJ; ISSN:2062-249X. (Akademiai Kiado)A highly efficient continuous synthesis has been developed for telmisartan, the active ingredient in the antihypertensive drug, Micardis. This synthetic route employs a convergent strategy that requires no intermediate purifications or solvent exchanges. The key step in the reaction scheme is a Suzuki cross-coupling reaction between two functionalized benzimidazoles that is catalyzed by a solid-supported Pd catalyst. This flow-based approach utilizes a tubular reactor system coupled with a plug flow packed bed cartridge unit that produces telmisartan in an 81% isolated yield.
- 105Snead, D. R.; Jamison, T. F. A Three-Minute Synthesis and Purification of Ibuprofen: Pushing the Limits of Continuous-Flow Processing. Angew. Chem., Int. Ed. 2015, 54 (3), 983– 987, DOI: 10.1002/anie.201409093[Crossref], [CAS], Google Scholar105https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitVWhu7bM&md5=febb1a276fc14c7dd186890624d9c7f3A Three-Minute Synthesis and Purification of Ibuprofen: Pushing the Limits of Continuous-Flow ProcessingSnead, David R.; Jamison, Timothy F.Angewandte Chemie, International Edition (2015), 54 (3), 983-987CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)In a total residence time of three minutes, ibuprofen was assembled from its elementary building blocks with an av. yield of above 90 % for each step. A scale-up of this five-stage process (3 bond-forming steps, one work-up, and one in-line liq.-liq. sepn.) provided ibuprofen at a rate of 8.09 g h-1 (equiv. to 70.8 kg y-1) using a system with an overall footprint of half the size of a std. lab. fume hood. Aside from the high throughput, several other aspects of this synthesis expand the capabilities of continuous-flow processing, including a Friedel-Crafts acylation run under neat conditions and promoted by AlCl3, an exothermic in-line quench of high concns. of pptn.-prone AlCl3, liq.-liq. sepns. run at or above 200 psi to provide solvent-free product, and the use of highly aggressive oxidants, such as iodine monochloride. The use of simple, inexpensive, and readily available reagents thus affords a practical synthesis of this important generic pharmaceutical.
- 106Pellegatti, L.; Hafner, A.; Sedelmeier, J. A Two-Step Continuous-Flow Procedure towards Ribociclib. J. Flow Chem. 2016, 6 (3), 198– 201, DOI: 10.1556/1846.2016.00017[Crossref], [CAS], Google Scholar106https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXptFaksLc%253D&md5=69c1e27717d80146e2bb26d0cd540815A two-step continuous-flow procedure towards RibociclibPellegatti, Laurent; Hafner, Andreas; Sedelmeier, JorgJournal of Flow Chemistry (2016), 6 (3), 198-201CODEN: JFCOBJ; ISSN:2062-249X. (Akademiai Kiado)This work describes the manufg. of ribociclib following the concept of an end-to-end continuous-flow process. The active pharmaceutical ingredient (API) is produced in a two-step telescoped flow process with integrated in-line liq.-liq. extn. and semibatch crystn.
- 107Ishitani, H.; Kanai, K.; Saito, Y.; Tsubogo, T.; Kobayashi, S. Synthesis of (±)-Pregabalin by Utilizing a Three-Step Sequential-Flow System with Heterogeneous Catalysts. Eur. J. Org. Chem. 2017, 2017 (44), 6491– 6494, DOI: 10.1002/ejoc.201700998[Crossref], [CAS], Google Scholar107https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhs1egtrrF&md5=f4e3082bdfbf63d2275b67e2df082090Synthesis of (±)-Pregabalin by utilizing a three-step sequential-flow system with heterogeneous catalystsIshitani, Haruro; Kanai, Kan; Saito, Yuki; Tsubogo, Tetsu; Kobayashi, ShuEuropean Journal of Organic Chemistry (2017), 2017 (44), 6491-6494CODEN: EJOCFK; ISSN:1099-0690. (Wiley-VCH Verlag GmbH & Co. KGaA)(±)-Pregabalin, a γ-amino acid deriv., has been synthesized by utilizing flow methods. A three-step sequential-flow reaction starting from com. isovaleraldehyde and Me malonate proceeded smoothly with heterogeneous catalysts to afford the precursor of pregabalin in yields of 75-100 %, and a space-time yield of 52.2 g/L d was reached. In addn., a heterogeneous catalyst for the Knoevenagel reactions of aldehydes with malonates, which is the first step of the synthesis, has been developed. Pregabalin was finally obtained by acid-catalyzed hydrolysis of the precursor followed by neutralization.
- 108Harsanyi, A.; Conte, A.; Pichon, L.; Rabion, A.; Grenier, S.; Sandford, G. One-Step Continuous Flow Synthesis of Antifungal WHO Essential Medicine Flucytosine Using Fluorine. Org. Process Res. Dev. 2017, 21 (2), 273– 276, DOI: 10.1021/acs.oprd.6b00420[ACS Full Text
], [CAS], Google Scholar
108https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXkslajsA%253D%253D&md5=5e3ed4a3ca0683e1f0754d4954b425adOne-Step Continuous Flow Synthesis of Antifungal WHO Essential Medicine Flucytosine Using FluorineHarsanyi, Antal; Conte, Annelyse; Pichon, Laurent; Rabion, Alain; Grenier, Sandrine; Sandford, GrahamOrganic Process Research & Development (2017), 21 (2), 273-276CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)In Africa around 625 000 mortalities per annum (20% of HIV/AIDS related deaths) are due to the affects of the Cryptococcal meningitis (CM) fungal infection. Recently, the World Health Organization (WHO) and the Infectious Disease Society of America (IDSA) recommended that the first line treatment for CM is a combination of amphotericin B and flucytosine, both now WHO Essential Medicines. However, flucytosine is not even registered for use in any African nation due, in part, to its relatively high cost of manuf. and lack of generic manufacturers. Currently, flucytosine is manufd. by an expensive four-step manufg. process. Here we report a one-step continuous flow process involving the reaction of inexpensive cytosine with fluorine gas using stainless steel tubular lab. and pilot-scale silicon carbide reactor devices which is readily scalable to a manufg. process with a low initial capital expenditure. - 109De Vitis, V.; Dall’Oglio, F.; Pinto, A.; De Micheli, C.; Molinari, F.; Conti, P.; Romano, D.; Tamborini, L. Chemoenzymatic Synthesis in Flow Reactors: A Rapid and Convenient Preparation of Captopril. ChemistryOpen 2017, 6 (5), 668– 673, DOI: 10.1002/open.201700082[Crossref], [PubMed], [CAS], Google Scholar109https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXht1GjtbjE&md5=8574b1f3b211f66a0fb5d092ffbc0189Chemoenzymatic Synthesis in Flow Reactors: A Rapid and Convenient Preparation of CaptoprilDe Vitis, Valerio; Dall'Oglio, Federica; Pinto, Andrea; De Micheli, Carlo; Molinari, Francesco; Conti, Paola; Romano, Diego; Tamborini, LuciaChemistryOpen (2017), 6 (5), 668-673CODEN: CHOPCK; ISSN:2191-1363. (Wiley-VCH Verlag GmbH & Co. KGaA)The chemoenzymic flow synthesis of enantiomerically pure captopril, a widely used antihypertensive drug, is accomplished starting from simple, inexpensive, and readily available reagents. The first step is a heterogeneous biocatalyzed regio- and stereoselective oxidn. of cheap prochiral 2-methyl-1,3-propandiol, performed in flow using immobilized whole cells of Acetobacter aceti MIM 2000/28, thus avoiding the use of aggressive and environmentally harmful chem. oxidants. The isolation of the highly hydrophilic intermediate (R)-3-hydroxy-2-methylpropanoic acid is achieved in-line by using a catch-and-release strategy. Then, three sequential high-throughput chem. steps lead to the isolation of captopril in only 75 min. In-line quenching and liq.-liq. sepn. enable breaks in the workflow and other manipulations to be avoided.
- 110Cole, K. P.; Groh, J. M. C.; Johnson, M. D.; Burcham, C. L.; Campbell, B. M.; Diseroad, W. D.; Heller, M. R.; Howell, J. R.; Kallman, N. J.; Koenig, T. M.; May, S. A.; Miller, R. D.; Mitchell, D.; Myers, D. P.; Myers, S. S.; Phillips, J. L.; Polster, C. S.; White, T. D.; Cashman, J.; Hurley, D.; Moylan, R.; Sheehan, P.; Spencer, R. D.; Desmond, K.; Desmond, P.; Gowran, O. Kilogram-Scale Prexasertib Monolactate Monohydrate Synthesis under Continuous-Flow CGMP Conditions. Science 2017, 356 (6343), 1144– 1151, DOI: 10.1126/science.aan0745[Crossref], [PubMed], [CAS], Google Scholar110https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXpslCktrs%253D&md5=a39e4ebe476a6ec3118fcfbb6f68cbd1Kilogram-scale prexasertib monolactate monohydrate synthesis under continuous-flow CGMP conditionsCole, Kevin P.; Groh, Jennifer McClary; Johnson, Martin D.; Burcham, Christopher L.; Campbell, Bradley M.; Diseroad, William D.; Heller, Michael R.; Howell, John R.; Kallman, Neil J.; Koenig, Thomas M.; May, Scott A.; Miller, Richard D.; Mitchell, David; Myers, David P.; Myers, Steven S.; Phillips, Joseph L.; Polster, Christopher S.; White, Timothy D.; Cashman, Jim; Hurley, Declan; Moylan, Robert; Sheehan, Paul; Spencer, Richard D.; Desmond, Kenneth; Desmond, Paul; Gowran, OliviaScience (Washington, DC, United States) (2017), 356 (6343), 1144-1150CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Advances in drug potency and tailored therapeutics are promoting pharmaceutical manufg. to transition from a traditional batch paradigm to more flexible continuous processing. Here we report the development of a multistep continuous-flow CGMP (current good manufg. practices) process that produced 24 kg of prexasertib monolactate monohydrate suitable for use in human clin. trials. Eight continuous unit operations were conducted to produce the target at roughly 3 kg per day using small continuous reactors, extractors, evaporators, crystallizers, and filters in lab. fume hoods. Success was enabled by advances in chem., engineering, anal. science, process modeling, and equipment design. Substantial tech. and business drivers were identified, which merited the continuous process. The continuous process afforded improved performance and safety relative to batch processes and also improved containment of a highly potent compd.
- 111Neyt, N. C.; Riley, D. L. Batch–Flow Hybrid Synthesis of the Antipsychotic Clozapine. React. Chem. Eng. 2018, 3 (1), 17– 24, DOI: 10.1039/C7RE00146K[Crossref], [CAS], Google Scholar111https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhvVGms7zL&md5=585d7ea42d4a0c204127d54ebb48a302Batch-flow hybrid synthesis of the antipsychotic clozapineNeyt, N. C.; Riley, D. L.Reaction Chemistry & Engineering (2018), 3 (1), 17-24CODEN: RCEEBW; ISSN:2058-9883. (Royal Society of Chemistry)The development of batch-flow hybrid processes is becoming an attractive prospect through which chemists can make use of the best aspects of both technologies. We have reported the implementation of an on-the-fly purifn. by trituration which can also be utilized to perform solvent swaps. We have demonstrated this concept through the synthesis of the antipsychotic clozapine. In addn., we report a novel means of performing a redn. of an aryl nitro group under flow conditions and an overall improved process route for the total synthesis of clozapine.
- 112Yu, E.; Mangunuru, H. P. R.; Telang, N. S.; Kong, C. J.; Verghese, J.; Gilliland, S. E., III; Ahmad, S.; Dominey, R. N.; Gupton, B. F. High-Yielding Continuous-Flow Synthesis of Antimalarial Drug Hydroxychloroquine. Beilstein J. Org. Chem. 2018, 14, 583– 592, DOI: 10.3762/bjoc.14.45[Crossref], [PubMed], [CAS], Google Scholar112https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsVOhtLnJ&md5=4ba7c1d6fde53fe272708f9a50c0b734High-yielding continuous-flow synthesis of antimalarial drug hydroxychloroquineYu, Eric; Mangunuru, Hari P. R.; Telang, Nakul S.; Kong, Caleb J.; Verghese, Jenson; Gilliland, Stanley E. III; Ahmad, Saeed; Dominey, Raymond N.; Gupton, B. FrankBeilstein Journal of Organic Chemistry (2018), 14 (), 583-592CODEN: BJOCBH; ISSN:1860-5397. (Beilstein-Institut zur Foerderung der Chemischen Wissenschaften)Numerous synthetic methods for the continuous prepn. of fine chems. and active pharmaceutical ingredients (API's) have been reported in recent years resulting in a dramatic improvement in process efficiencies. Herein authors report a highly efficient continuous synthesis of the antimalarial drug hydroxychloroquine (HCQ). Key improvements in the new process include the elimination of protecting groups with an overall yield improvement of 52% over the current com. process. The continuous process employs a combination of packed bed reactors with continuous stirred tank reactors for the direct conversion of the starting materials to the product. This high-yielding, multigram-scale continuous synthesis provides an opportunity to achieve increase global access to hydroxychloroquine for treatment of malaria.
- 113Ziegler, R. E.; Desai, B. K.; Jee, J.-A.; Gupton, B. F.; Roper, T. D.; Jamison, T. F. 7-Step Flow Synthesis of the HIV Integrase Inhibitor Dolutegravir. Angew. Chem., Int. Ed. 2018, 57 (24), 7181– 7185, DOI: 10.1002/anie.201802256[Crossref], [CAS], Google Scholar113https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXpsVOlt78%253D&md5=bd22b8a819e83fbafd9ac9a03b5612ca7-Step Flow Synthesis of the HIV Integrase Inhibitor DolutegravirZiegler, Robert E.; Desai, Bimbisar K.; Jee, Jo-Ann; Gupton, B. Frank; Roper, Thomas D.; Jamison, Timothy F.Angewandte Chemie, International Edition (2018), 57 (24), 7181-7185CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)Dolutegravir (DTG), an important active pharmaceutical ingredient (API) used in combination therapy for the treatment of HIV, was synthesized in continuous flow. By adapting the reported GlaxoSmithKline process chem. batch route for Cabotegravir, DTG was produced in 4.5 h in sequential flow operations from com. available materials. Key features of the synthesis include rapid manufg. time for pyridone formation, one-step direct amidation of a functionalized pyridone, and telescoping of multiple steps to avoid isolation of intermediates and enable for greater throughput.
- 114Mancino, V.; Cerra, B.; Piccinno, A.; Gioiello, A. Continuous Flow Synthesis of 16-Dehydropregnenolone Acetate, a Key Synthon for Natural Steroids and Drugs. Org. Process Res. Dev. 2018, 22 (5), 600– 607, DOI: 10.1021/acs.oprd.8b00038[ACS Full Text
], [CAS], Google Scholar
114https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXnslKmur4%253D&md5=5899d7d5c7339e9b2fbfcdb3336bb692Continuous Flow Synthesis of 16-Dehydropregnenolone Acetate, a Key Synthon for Natural Steroids and DrugsMancino, Valentina; Cerra, Bruno; Piccinno, Alessandro; Gioiello, AntimoOrganic Process Research & Development (2018), 22 (5), 600-607CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)A telescoped multistep process to provide the continuous delivery of 16-dehydropregnenolone acetate (16-DPA) from diosgenin is described. The method was evaluated through batch screenings that helped to identify crit. bottlenecks and flowability, and the best conditions were optimized in flow systems before the individual steps were telescoped together into a single integrated flow process. Further highlights of our approach include the use of efficient in-line extn. operations and reaction monitoring, the avoidance of time-consuming purifications between steps, and improvement of efficiency and safety stds. - 115Zhang, P.; Weeranoppanant, N.; Thomas, D. A.; Tahara, K.; Stelzer, T.; Russell, M. G.; O’Mahony, M.; Myerson, A. S.; Lin, H.; Kelly, L. P.; Jensen, K. F.; Jamison, T. F.; Dai, C.; Cui, Y.; Briggs, N.; Beingessner, R. L.; Adamo, A. Advanced Continuous Flow Platform for On-Demand Pharmaceutical Manufacturing. Chem. - Eur. J. 2018, 24 (11), 2776– 2784, DOI: 10.1002/chem.201706004[Crossref], [PubMed], [CAS], Google Scholar115https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitVOmu7Y%253D&md5=feae2c8c4a5d66a8d6baa210aa99cda6Advanced continuous flow platform for on-demand pharmaceutical manufacturingZhang, Ping; Weeranoppanant, Nopphon; Thomas, Dale A.; Tahara, Kohei; Stelzer, Torsten; Russell, Mary Grace; O'Mahony, Marcus; Myerson, Allan S.; Lin, Hongkun; Kelly, Liam P.; Jensen, Klavs F.; Jamison, Timothy F.; Dai, Chunhui; Cui, Yuqing; Briggs, Naomi; Beingessner, Rachel L.; Adamo, AndreaChemistry - A European Journal (2018), 24 (11), 2776-2784CODEN: CEUJED; ISSN:0947-6539. (Wiley-VCH Verlag GmbH & Co. KGaA)As a demonstration of an alternative to the challenges faced with batch pharmaceutical manufg. including the large prodn. footprint and lengthy time-scale, we previously reported a refrigerator-sized continuous flow system for the on-demand prodn. of essential medicines. Building on this technol., herein we report a second-generation, reconfigurable and 25% smaller (by vol.) continuous flow pharmaceutical manufg. platform featuring advances in reaction and purifn. equipment. Consisting of two compact [0.7 (L)×0.5 (D)×1.3 m (H)] stand-alone units for synthesis and purifn./formulation processes, the capabilities of this automated system are demonstrated with the synthesis of nicardipine hydrochloride and the prodn. of concd. liq. doses of ciprofloxacin hydrochloride, neostigmine methylsulfate and rufinamide that meet US Pharmacopeia stds.
- 116Balogh, A.; Domokos, A.; Farkas, B.; Farkas, A.; Rapi, Z.; Kiss, D.; Nyiri, Z.; Eke, Z.; Szarka, G.; Örkényi, R.; Mátravölgyi, B.; Faigl, F.; Marosi, G.; Nagy, Z. K. Continuous End-to-End Production of Solid Drug Dosage Forms : Coupling Flow Synthesis and Formulation by Electrospinning. Chem. Eng. J. 2018, 350, 290– 299, DOI: 10.1016/j.cej.2018.05.188[Crossref], [CAS], Google Scholar116https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtVOqtbvF&md5=44cd90a60c117d8b389254494a6f441eContinuous end-to-end production of solid drug dosage forms: Coupling flow synthesis and formulation by electrospinningBalogh, Attila; Domokos, Andras; Farkas, Balazs; Farkas, Attila; Rapi, Zsolt; Kiss, Domokos; Nyiri, Zoltan; Eke, Zsuzsanna; Szarka, Gyorgyi; Orkenyi, Robert; Matravolgyi, Bela; Faigl, Ferenc; Marosi, Gyorgy; Nagy, Zsombor KristofChemical Engineering Journal (Amsterdam, Netherlands) (2018), 350 (), 290-299CODEN: CMEJAJ; ISSN:1385-8947. (Elsevier B.V.)Based on the concept of continuous manufg. an end-to-end benchtop device was developed unprecedented for the prodn. of solid drug dosage forms by connecting flow synthesis and formulation via electrospinning (ES). Together with the optimized two-step continuous-flow synthesis of acetylsalicylic acid (ASA) a water-sol. polymeric excipient (polyvinylpyrrolidone K30, PVPK30) was introduced. The resulting polymeric soln. could be readily electrospun into solid nanofibers with high purity in one single step due to the excellent yet gentle drying effect of ES. The ASA-loaded fibers were electrostatically deposited onto a water-sol. pullulan sheet and the obtained double-layered films were continuously cut into orally dissolving webs (ODWs) as final dosage formulation. The synthesis as well as the dosing of the fibrous films were monitored by Process Anal. Technol. (PAT) tools (IR and Raman spectroscopy) with active feedback on product quality. The successful coupling of flow synthesis and fiber formation confirms that ES enables versatile formulation of pharmaceuticals in future continuous prodn. systems.
- 117Ma, X.; Chen, J.; Du, X. A Continuous Flow Process for the Synthesis of Hymexazol. Org. Process Res. Dev. 2019, 23 (6), 1152– 1158, DOI: 10.1021/acs.oprd.9b00047[ACS Full Text
], [CAS], Google Scholar
117https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXpvVOqsbY%253D&md5=918125598faf18bf620d3a9e34a91a70A Continuous Flow Process for the Synthesis of HymexazolMa, Xin-peng; Chen, Jin-sha; Du, Xiao-huaOrganic Process Research & Development (2019), 23 (6), 1152-1158CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)Hymexazol, I, is an efficient and low-toxicity soil fungicide. In this work, a fully continuous flow process for the synthesis of hymexazol has been developed. This process begins with combining Et acetoacetate and hydroxylamine hydrochloride to form a hydroxamic acid intermediate. The reaction soln. is then quenched with concd. hydrochloric acid to obtain the final product, hymexazol. Under the optimized process conditions, the total yield of the target product reached 86%. In addn., prodn. was successfully scaled-up to a kilogram scale. The continuous flow method not only greatly decreases the reaction time but also significantly inhibits the side reactions. - 118Boros, Z.; Nagy-Győr, L.; Kátai-Fadgyas, K.; Kőhegyi, I.; Ling, I.; Nagy, T.; Iványi, Z.; Oláh, M.; Ruzsics, G.; Temesi, O.; Volk, B. Continuous Flow Production in the Final Step of Vortioxetine Synthesis. Piperazine Ring Formation on a Flow Platform with a Focus on Productivity and Scalability. J. Flow Chem. 2019, 9 (2), 101– 113, DOI: 10.1007/s41981-019-00036-x[Crossref], [CAS], Google Scholar118https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitFSqtbvO&md5=4991bbd83ac36771e2c258c22397440cContinuous flow production in the final step of vortioxetine synthesis. Piperazine ring formation on a flow platform with a focus on productivity and scalabilityBoros, Zoltan; Nagy-Gyor, Laszlo; Katai-Fadgyas, Katalin; Kohegyi, Imre; Ling, Istvan; Nagy, Tamas; Ivanyi, Zoltan; Olah, Mark; Ruzsics, Gyorgy; Temesi, Otto; Volk, BalazsJournal of Flow Chemistry (2019), 9 (2), 101-113CODEN: JFCOBJ; ISSN:2063-0212. (Akademiai Kiado)In this study, the piperazine formation step of vortioxetine synthesis was investigated under continuous flow conditions. The batch variant of this step could be carried out at lab. scale at 130-135 °C with a long reaction time (27 h) followed by a laborious optimization process, but the formation of a significant amt. of side-products could be detected, thus an efficient purifn. procedure was necessary. In the attempted scale-up of the batch reaction, a complete conversion could not at all be reached, even after elongated reaction times (36 h). The continuous-flow expts. were carried out in a new, purpose-built flow system. The examns. were extended to a wide range of reaction parameters (ratio of solvents, concn. and molar ratio of reagents, geometry of coiled loop reactor, residence time, temp.) and to the feasibility study of scale-up. In the second part of the expts., the fine-tuning of scaled-up reaction parameters of continuous flow synthesis was carried out using a systematic design of expts. approach. Finally 190 °C reaction temp. and 30 min of residence time led to the highest efficacy in the prodn. of vortioxetine drug substance with high yield and purity.
- 119Bana, P.; Szigetvári, Á.; Kóti, J.; Éles, J.; Greiner, I. Flow-Oriented Synthetic Design in the Continuous Preparation of the Aryl Piperazine Drug Flibanserin. React. Chem. Eng. 2019, 4 (4), 652– 657, DOI: 10.1039/C8RE00266E[Crossref], [CAS], Google Scholar119https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXktFykug%253D%253D&md5=4303557fd9016c96b7929d3e61f1bcddFlow-oriented synthetic design in the continuous preparation of the aryl piperazine drug flibanserinBana, Peter; Szigetvari, Aron; Koti, Janos; Eles, Janos; Greiner, IstvanReaction Chemistry & Engineering (2019), 4 (4), 652-657CODEN: RCEEBW; ISSN:2058-9883. (Royal Society of Chemistry)An uninterrupted, four-step continuous-flow sequence was developed for the prepn. of the aryl piperazine-type serotonin receptor modulator drug, flibanserin. The unprecedented route was designed for efficient operation in flow, consisting of heterogeneously catalyzed reductive amination reactions, benzimidazolone formation using the protecting group as a building block, and biphasic flow deprotection integrated with in-line purifn. Gas-liq. and liq.-liq. sepns. facilitate the coupling of the reaction steps and aid removal of excess reagents, which make the system capable of steady-state operation without manual interaction.
- 120Fu, W. C.; Jamison, T. F. Modular Continuous Flow Synthesis of Imatinib and Analogues. Org. Lett. 2019, 21 (15), 6112– 6116, DOI: 10.1021/acs.orglett.9b02259[ACS Full Text
], [CAS], Google Scholar
120https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtlOltbjN&md5=b1a1548f5278ac5199338c87bca8a755Modular Continuous Flow Synthesis of Imatinib and AnaloguesFu, Wai Chung; Jamison, Timothy F.Organic Letters (2019), 21 (15), 6112-6116CODEN: ORLEF7; ISSN:1523-7052. (American Chemical Society)A modular continuous flow synthesis of imatinib (I) and analogs is reported. Structurally diverse imatinib analogs are rapidly generated using three readily available building blocks via a flow hydration/chemoselective C-N coupling sequence. The newly developed continuous flow hydration and amidation modules each exhibit a broad scope with good to excellent yields. Overall, the method described does not require solvent switches, in-line purifications, or packed-bed apparatuses due to the judicious manipulation of flow setups and solvent mixts. - 121García-Lacuna, J.; Domínguez, G.; Blanco-Urgoiti, J.; Pérez-Castells, J. Synthesis of Treprostinil: Key Claisen Rearrangement and Catalytic Pauson–Khand Reactions in Continuous Flow. Org. Biomol. Chem. 2019, 17 (43), 9489– 9501, DOI: 10.1039/C9OB02124H[Crossref], [PubMed], [CAS], Google Scholar121https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhvFKrtr7F&md5=dc95f46dc98a8a6a47a9be96c96db7abSynthesis of treprostinil: key Claisen rearrangement and catalytic Pauson-Khand reactions in continuous flowGarcia-Lacuna, Jorge; Dominguez, Gema; Blanco-Urgoiti, Jaime; Perez-Castells, JavierOrganic & Biomolecular Chemistry (2019), 17 (43), 9489-9501CODEN: OBCRAK; ISSN:1477-0520. (Royal Society of Chemistry)A new synthesis of treprostinil is described using a plug flow reactor in two of the key steps. First, a Claisen rearrangement reaction is described in scaled flow at multigram amts. Yields and selectivity of this step are sharply improved compared to those from previous syntheses. Second, the key Pauson-Khand reaction in flow is described under catalytic conditions with 5 mol% of cobalt carbonyl and only 3 equiv. of CO. Scaling up of this reaction safely ensures a good yield of an advanced intermediate which is transformed into treprostinil in three steps. Other improvements are the introduction of the carboxymethyl chain into the phenol from the beginning to reduce the protection-deprotection steps. The synthesis is completed in 14% global yield after 12 linear steps from (S)-epichlorohydrin.
- 122Lee, H.; Kim, H.; Kim, D. From p -Xylene to Ibuprofen in Flow: Three-Step Synthesis by a Unified Sequence of Chemoselective C–H Metalations. Chem. - Eur. J. 2019, 25 (50), 11641– 11645, DOI: 10.1002/chem.201903267[Crossref], [PubMed], [CAS], Google Scholar122https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs1aitrbK&md5=3cc1fc1d1537b2bf08a83f02567d5ac0From p-Xylene to Ibuprofen in Flow: Three-Step Synthesis by a Unified Sequence of Chemoselective C-H MetalationsLee, Hyune-Jea; Kim, Heejin; Kim, Dong-PyoChemistry - A European Journal (2019), 25 (50), 11641-11645CODEN: CEUJED; ISSN:0947-6539. (Wiley-VCH Verlag GmbH & Co. KGaA)Ibuprofen was prepd. from an inactive and inexpensive p-xylene by three-step flow functionalizations through chemoselective metalations of benzyl positions in sequence using an in-situ generated LICKOR-type superbase. The flow approach in the microreactor facilitated the comprehensive exploration of over 100 conditions in the first-step reaction by varying concns., temps., solvents and equiv. of reagents, enabling optimal conditions to be found with 95% yield by significantly suppressing the formation of byproducts followed by the second C-H metalation step in 95% yield. Moreover, gram-scale synthesis of ibuprofen in the final step was achieved by biphasic flow reaction of soln.-phase intermediate with CO2 isolating 2.3 g for 10 min of operation time.
- 123Jaman, Z.; Sobreira, T. J. P.; Mufti, A.; Ferreira, C. R.; Cooks, R. G.; Thompson, D. H. Rapid On-Demand Synthesis of Lomustine under Continuous Flow Conditions. Org. Process Res. Dev. 2019, 23 (3), 334– 341, DOI: 10.1021/acs.oprd.8b00387[ACS Full Text
], [CAS], Google Scholar
123https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXislOmtrs%253D&md5=df320cb95ac742223c68191b120283faRapid On-Demand Synthesis of Lomustine under Continuous Flow ConditionsJaman, Zinia; Sobreira, Tiago J. P.; Mufti, Ahmed; Ferreira, Christina R.; Cooks, R. Graham; Thompson, David H.Organic Process Research & Development (2019), 23 (3), 334-341CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)Lomustine, an important agent for treatment of brain tumors and Hodgkin's lymphoma, has been synthesized using continuous flow methodol. Desorption electrospray ionization mass spectrometry (DESI-MS) was used to quickly explore a large no. of reaction conditions for one of the reaction steps and guide the efficient translation of optimized conditions to continuous lomustine prodn. Using only four inexpensive com. available starting materials and a total residence time of 9 min, lomustine was prepd. via a linear sequence of two chem. reactions performed sep. in two telescoped flow reactors. Sequential offline extn. and filtration resulted in a 63% overall yield of pure lomustine at a prodn. rate of 110 mg/h. The primary advantages of this approach are the rapid manuf. of lomustine with two telescoped steps to avoid isolation and purifn. of a labile intermediate and the mild conditions used in the nitrosylation step, thereby significantly increasing the purity and yield of this active pharmaceutical ingredient. - 124Russell, M. G.; Jamison, T. F. Seven-Step Continuous Flow Synthesis of Linezolid Without Intermediate Purification. Angew. Chem., Int. Ed. 2019, 58 (23), 7678– 7681, DOI: 10.1002/anie.201901814[Crossref], [CAS], Google Scholar124https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXptFGqu7Y%253D&md5=ab3e27c7aa2ac09c37a7449970e89482Seven-Step Continuous Flow Synthesis of Linezolid Without Intermediate PurificationRussell, M. Grace; Jamison, Timothy F.Angewandte Chemie, International Edition (2019), 58 (23), 7678-7681CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)Herein, the blockbuster antibacterial drug linezolid is synthesized from simple starting blocks by a convergent continuous flow sequence involving seven (7) chem. transformations. This is the highest total no. of distinct reaction steps ever performed in continuous flow without conducting solvent exchanges or intermediate purifn. Linezolid was obtained in 73 % isolated yield in a total residence time of 27 min, corresponding to a throughput of 816 mg h-1.
- 125Damião, M. C. F. C. B.; Marçon, H. M.; Pastre, J. C. Continuous Flow Synthesis of the URAT1 Inhibitor Lesinurad. React. Chem. Eng. 2020, 5 (5), 865– 872, DOI: 10.1039/C9RE00483A[Crossref], [CAS], Google Scholar125https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhs1KqtL4%253D&md5=101222c47b19942a002cf11920da73f5Continuous flow synthesis of the URAT1 inhibitor lesinuradDamiao, Mariana C. F. C. B.; Marcon, Henrique M.; Pastre, Julio CezarReaction Chemistry & Engineering (2020), 5 (5), 865-872CODEN: RCEEBW; ISSN:2058-9883. (Royal Society of Chemistry)Herein, the urate anion exchange transporter 1 (URAT1) inhibitor lesinurad is synthesized from com. available building blocks by a five-step linear continuous flow sequence. Our previously developed continuous flow platform was successfully applied to generate the 3-thio-1,2,4-triazole key intermediate 2 in 88% yield, after 55 min of residence time. Condensation, cyclization and S-alkylation were telescoped in a single operation without conducting solvent exchanges and intermediate purifications. Next, 1,2,4-triazole bromination and ester hydrolysis were also performed in continuous flow regime to deliver lesinurad in 68% overall yield in a total residence time of 2 h. Our approach enables the fast generation of lesinurad and can be directly applied to produce major quantities of this important API.
- 126Ingham, R. J.; Battilocchio, C.; Fitzpatrick, D. E.; Sliwinski, E.; Hawkins, J. M.; Ley, S. V. A Systems Approach Towards an Intelligent and Self-Controlling Platform for Integrated Continuous Reaction Sequences. Angew. Chem., Int. Ed. 2015, 54 (1), 144– 148, DOI: 10.1002/anie.201409356[Crossref], [CAS], Google Scholar126https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvVKlsLjP&md5=52477537a2d838c6d8a5af7e588c8228A Systems Approach towards an Intelligent and Self-Controlling Platform for Integrated Continuous Reaction SequencesIngham, Richard J.; Battilocchio, Claudio; Fitzpatrick, Daniel E.; Sliwinski, Eric; Hawkins, Joel M.; Ley, Steven V.Angewandte Chemie, International Edition (2015), 54 (1), 144-148CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)Performing reactions in flow can offer major advantages over batch methods. However, lab. flow chem. processes are currently often limited to single steps or short sequences due to the complexity involved with operating a multi-step process. Using new modular components for downstream processing, coupled with control technologies, more advanced multi-step flow sequences can be realized. These tools are applied to the synthesis of 2-aminoadamantane-2-carboxylic acid. A system comprising three chem. steps and three workup steps was developed, having sufficient autonomy and self-regulation to be managed by a single operator.
- 127Lichtenegger, G. J.; Maier, M.; Khinast, J. G.; Gruber-Wölfler, H. Continuous Suzuki—Miyaura Reactions with Novel Ce—Sn—Pd Oxides and Integrated Crystallization as Continuous Downstream Protocol. J. Flow Chem. 2016, 6 (3), 244– 251, DOI: 10.1556/1846.2016.00021[Crossref], [CAS], Google Scholar127https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXptFaksrY%253D&md5=d5c6fe4031face122ac2344bdcf03d03Continuous Suzuki-Miyaura reactions with novel Ce-Sn-Pd oxides and integrated crystallization as continuous downstream protocolLichtenegger, Georg J.; Maier, Manuel; Khinast, Johannes G.; Gruber-Woelfler, HeidrunJournal of Flow Chemistry (2016), 6 (3), 244-251CODEN: JFCOBJ; ISSN:2062-249X. (Akademiai Kiado)An integrated process including continuous-flow syntheses directly coupled to product isolation via continuous crystn. is presented. For the synthesis part, Ce0.495Sn0.495Pd0.01O2-δ was used as heterogeneous catalyst in a custom-made packed-bed reactor (the so-called "Plug and Play" reactor) for continuous Suzuki-Miyaura crosscouplings of various para- and ortho-substituted bromoarenes with phenylboronic acid using environmentally friendly aq. ethanolic mixts. as reaction solvents. The reactions were stable for up to 30 h without any detectable catalyst deactivation. The desired biaryl products were obtained in gram scale with good to excellent yields and high selectivity. For three methyl-, ketyl-, and nitrile-functionalized biphenyl products, isolation was done using water as antisolvent in an integrated crystn. process as continuous downstream protocol. The desired products could be isolated with high purity and with yields of up to 95% for the overall process.
- 128Tacsi, K.; Pataki, H.; Domokos, A.; Nagy, B.; Csontos, I.; Markovits, I.; Farkas, F.; Nagy, Z. K.; Marosi, G. Direct Processing of a Flow Reaction Mixture Using Continuous Mixed Suspension Mixed Product Removal Crystallizer. Cryst. Growth Des. 2020, 20 (7), 4433– 4442, DOI: 10.1021/acs.cgd.0c00252[ACS Full Text
], [CAS], Google Scholar
128https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXpvVGlt7s%253D&md5=172538ce164736ed1d930c4df202cd13Direct Processing of a Flow Reaction Mixture Using Continuous Mixed Suspension Mixed Product Removal CrystallizerTacsi, Kornelia; Pataki, Hajnalka; Domokos, Andras; Nagy, Brigitta; Csontos, Istvan; Markovits, Imre; Farkas, Ferenc; Nagy, Zsombor Kristof; Marosi, GyorgyCrystal Growth & Design (2020), 20 (7), 4433-4442CODEN: CGDEFU; ISSN:1528-7483. (American Chemical Society)Crystn. as the most widespread purifn., sepn., and morphol.-detg. method is a crit. technol. that could be made safer and more economical by using continuous crystn. alternatives. Accordingly, this study aims to develop the continuous crystn. method for direct processing of a flow reaction mixt. of acetylsalicylic acid (ASA) and to provide pure, homogeneous cryst. products for further formulation steps. The solid-liq. sepn. and the purifn. of the acetylsalicylic acid from the multicomponent mixt. were accomplished in a single stage mixed suspension mixed product removal (MSMPR) continuous crystallizer equipped with an overflow and an inner buffer element to ensure the representative withdrawal of the product suspension. The effect of process parameters such as the operating temp. and the length of residence time (RT) on product quality and quantity were studied at two and three levels, resp. Investigating these parameters, we found that higher operating temps. (25°) and longer residence time (47 min) favor appropriate purity (>99.5%), and narrow crystal size distribution. By reducing the operating temp. (2.5°), the yield improved slightly (approx. 77%) and polydisperse products were characterized. The developed crystn. process can link the flow synthesis with the continuous formulation, and consequently serves a further step toward end-to-end prodn. A multicomponent ASA reaction mixt. was processed in a single stage mixed suspension mixed product removal (MSMPR) continuous crystallizer equipped with an overflow tubing and an inner vertical plate served as a buffer element. Depending on the chosen temp. and residence time, the yield, size, and size distribution of the crystals can be modified resulting in a multimodal or unimodal product. - 129Rimez, B.; Septavaux, J.; Scheid, B. The Coupling of In-Flow Reaction with Continuous Flow Seedless Tubular Crystallization. React. Chem. Eng. 2019, 4 (3), 516– 522, DOI: 10.1039/C8RE00313K[Crossref], [CAS], Google Scholar129https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhsVaktb4%253D&md5=f017f8d5d7a876e716d0a2c7216e1185The coupling of in-flow reaction with continuous flow seedless tubular crystallizationRimez, Bart; Septavaux, Jean; Scheid, BenoitReaction Chemistry & Engineering (2019), 4 (3), 516-522CODEN: RCEEBW; ISSN:2058-9883. (Royal Society of Chemistry)The direct coupling between a continuous flow-assisted acetylation reaction of salicylic acid and the subsequent crystn. of aspirin in a continuous flow tubular reactor or nucleator is investigated. The design of the reactor relies on a 3 min residence time, followed by quenching with water prior to cooling inside the tubular nucleator. By appropriate selection of the temp. of the nucleation step, the no. of nuclei formed in the tubular setup is accurately controlled. In this way, pure cryst. aspirin is continuously obtained with a desired mean crystal size between 3μm and 300μm and a reduced dispersion grade, avoiding seeding and micronization steps.
- 130Rimez, B.; Septavaux, J.; Debuysschère, R.; Scheid, B. The Creation and Testing of a Fully Continuous Tubular Crystallization Device Suited for Incorporation into Flow Chemistry Setups. J. Flow Chem. 2019, 9 (4), 237– 249, DOI: 10.1007/s41981-019-00042-z
- 131Palmer, E. Lilly commits to continuous manufacturing with Ireland plant, https://www.fiercepharma.com/manufacturing/lilly-commits-to-continuous-manufacturing-ireland-plant.Google ScholarThere is no corresponding record for this reference.
- 132Johnson, M. D.; May, S. A.; Calvin, J. R.; Remacle, J.; Stout, J. R.; Diseroad, W. D.; Zaborenko, N.; Haeberle, B. D.; Sun, W.-M.; Miller, M. T.; Brennan, J. Development and Scale-Up of a Continuous, High-Pressure, Asymmetric Hydrogenation Reaction, Workup, and Isolation. Org. Process Res. Dev. 2012, 16 (5), 1017– 1038, DOI: 10.1021/op200362h[ACS Full Text
], [CAS], Google Scholar
132https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XmtFartL0%253D&md5=baa54195de39d42caaabaae674d6271eDevelopment and Scale-Up of a Continuous, High-Pressure, Asymmetric Hydrogenation Reaction, Workup, and IsolationJohnson, Martin D.; May, Scott A.; Calvin, Joel R.; Remacle, Jacob; Stout, James R.; Diseroad, William D.; Zaborenko, Nikolay; Haeberle, Brian D.; Sun, Wei-Ming; Miller, Michael T.; Brennan, JohnOrganic Process Research & Development (2012), 16 (5), 1017-1038CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)A fully continuous process including an asym. hydrogenation reaction operating at 70 bar hydrogen, aq. extn., and crystn. was designed, developed, and demonstrated at pilot scale. This paper highlights safety, quality, and throughput advantages of the continuous reaction and sepns. unit operations. Prodn. of 144 kg of product was accomplished in lab. fume hoods and a lab. hydrogenation bunker over two continuous campaigns. Maximum continuous flow vessel size in the lab. hoods was 22 L glassware, and max. plug flow tube reactor (PFR) size in the bunker was 73 L. The main safety advantages of running the hydrogenation reaction continuous rather than batch were that the flow reactor was smaller for the same throughput and, more importantly, the tubular hydrogenation reactor ran 95% liq. filled at steady state. Therefore, the amt. of hydrogen in the reactor at any one time was less than that of batch. A two-stage mixed suspension-mixed product removal (MSMPR) cascade was used for continuous crystn. Impurity rejection by continuous crystn. was superior to that by batch because scalable residence time and steady-state supersatn. enabled robust and repeatable control of enantiomer rejection in a kinetic regime, although this is a nonstandard approach, debatable as an impurity control strategy. The fully continuous wet-end process running in a lab. infrastructure achieved the same weekly throughput that would be expected from traditional batch processing in a plant module with 400 L vessels. - 133Chen, J.; Sarma, B.; Evans, J. M. B.; Myerson, A. S. Pharmaceutical Crystallization. Cryst. Growth Des. 2011, 11 (4), 887– 895, DOI: 10.1021/cg101556s[ACS Full Text
], [CAS], Google Scholar
133https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXisVWlu7Y%253D&md5=f883c9d4638e0b75542eb6c322c08595Pharmaceutical CrystallizationChen, Jie; Sarma, Bipul; Evans, James M. B.; Myerson, Allan S.Crystal Growth & Design (2011), 11 (4), 887-895CODEN: CGDEFU; ISSN:1528-7483. (American Chemical Society)A review. Crystn. is crucial in the pharmaceutical industry as a sepn. process for intermediates and as the final step in the manuf. of active pharmaceutical ingredients (APIs). In this perspective article to celebrate 10 years of Crystal Growth and Design, we focus on 3 areas related to crystn. in the pharmaceutical industry: (1) advances in our understanding of the fundamentals of nucleation, (2) prodn. and scale-up of novel solid forms, and (3) continuous processing. While the areas discussed are not new, they are areas, in our opinion, of significant current interest to the community engaged in crystn. in the pharmaceutical industry. - 134Wang, T.; Lu, H.; Wang, J.; Xiao, Y.; Zhou, Y.; Bao, Y.; Hao, H. Recent Progress of Continuous Crystallization. J. Ind. Eng. Chem. 2017, 54, 14– 29, DOI: 10.1016/j.jiec.2017.06.009[Crossref], [CAS], Google Scholar134https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtVers7%252FK&md5=3f8de8a12c0617fd4eac8177b3442936Recent progress of continuous crystallizationWang, Ting; Lu, Haijiao; Wang, Jingkang; Xiao, Yan; Zhou, Yanan; Bao, Ying; Hao, HongxunJournal of Industrial and Engineering Chemistry (Amsterdam, Netherlands) (2017), 54 (), 14-29CODEN: JIECFI; ISSN:1226-086X. (Elsevier B.V.)Continuous crystn. has always been a hot topic in industrial crystn. Many efforts have been made to improve the continuous crystn., either by designing novel continuous crystallizers or by proposing improved design and operation of conventional continuous crystallizers. Some new models for continuous crystn. processes have also been proposed and tested in recent years. In this work, the development of continuous crystn. in recent years, including novel crystallizers, control strategies, models and some assistive technologies, is summarized. Promising as it is, continuous crystn. is still not as universal as batch crystn. due to the existence of the drawbacks, such as blockage and encrustation. Therefore, further efforts are needed before wider application of continuous crystn.
- 135Wood, B.; Girard, K. P.; Polster, C. S.; Croker, D. M. Progress to Date in the Design and Operation of Continuous Crystallization Processes for Pharmaceutical Applications. Org. Process Res. Dev. 2019, 23 (2), 122– 144, DOI: 10.1021/acs.oprd.8b00319[ACS Full Text
], [CAS], Google Scholar
135https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXnslSrtg%253D%253D&md5=802dda7ac340af7babeb1159fa201d18Progress to Date in the Design and Operation of Continuous Crystallization Processes for Pharmaceutical ApplicationsWood, Barbara; Girard, Kevin P.; Polster, Christopher S.; Croker, Denise M.Organic Process Research & Development (2019), 23 (2), 122-144CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)Continuous crystn. has gained interest in the pharmaceutical sector as part of the drive toward the transition from exclusive batch manufg. to integrated continuous manufg. in this industry. As a result, the design and operation of continuous crystn. processes for the prepn. of pharmaceutical materials has been featured strongly in recent scientific literature. This review is an effort to gather together all of the published understanding on continuous crystn. with a pharmaceutical focus and to benchmark progress to date in realizing the potential benefits of transitioning this stalwart pharmaceutical unit operation from batch to continuous configurations. - 136Jiang, M.; Braatz, R. D. Designs of Continuous-Flow Pharmaceutical Crystallizers: Developments and Practice. CrystEngComm 2019, 21 (23), 3534– 3551, DOI: 10.1039/C8CE00042E[Crossref], [CAS], Google Scholar136https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXosVCgsbw%253D&md5=c094bd422413ae0977a4d2494630f578Designs of continuous-flow pharmaceutical crystallizers: developments and practiceJiang, Mo; Braatz, Richard D.CrystEngComm (2019), 21 (23), 3534-3551CODEN: CRECF4; ISSN:1466-8033. (Royal Society of Chemistry)Crystn. is an effective, low-cost purifn. & formulation process widely applied to pharmaceuticals and fine chems. This review describes recent advances in research on lab-scale soln.-based continuous crystn., including (1) a 5-step general design procedure; (2) key design/operational parameters; (3) process intensification strategies; and (4) a case study. The continuous crystallizers reviewed include mixed-suspension mixed-product removal, fluidized beds, oscillatory baffled flow, and tubular laminar/segmented/slug-flow crystallizers. Their corresponding design and operational considerations are summarized in terms of general parameters (e.g., residence time), and crystallizer-specific parameters and strategies (e.g., mixing strategies). In-line nucleation and crystal modification methods are categorized, including use of micromixers, wet milling, ultrasonication, temp. cycling, and recycling selection (filtration, sedimentation). Throughout the article, links are drawn with extensive existing knowledge of batch crystallizers, to facilitate the understanding and design of continuous crystallizers.
- 137Ma, Y.; Wu, S.; Macaringue, E. G. J.; Zhang, T.; Gong, J.; Wang, J. Recent Progress in Continuous Crystallization of Pharmaceutical Products: Precise Preparation and Control. Org. Process Res. Dev. 2020, 24, 1785 DOI: 10.1021/acs.oprd.9b00362 .[ACS Full Text
], [CAS], Google Scholar
137https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitleqsbo%253D&md5=66f4902681b6e550a7fc5c010591f6f5Recent Progress in Continuous Crystallization of Pharmaceutical Products: Precise Preparation and ControlMa, Yiming; Wu, Songgu; Macaringue, Estevao Genito Joao; Zhang, Teng; Gong, Junbo; Wang, JingkangOrganic Process Research & Development (2020), 24 (10), 1785-1801CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)A review. Crystn., as a solid-liq. sepn. process, is employed to purify and isolate a great diversity of cryst. pharmaceutical products. In recent years, continuous crystn. has attracted increasing attention because of the product and process robustness as well as higher productivity. In this work, we review the use of novel continuous crystallizers or modified conventional continuous crystallizers for the prepn. of polymorphs, chiral enantiomers, solvates/hydrates, cocrystals, and spherical crystals. In addn., the theor. framework and verification of the model-based control approaches are demonstrated. The application of process anal. technol. tools in classical feedback loop control strategies in continuous crystn. is also discussed. Despite all this, the application of continuous crystn. still remains challenging because of the existence of drawbacks such as fouling and blockages. Therefore, a systematic discussion should be done before continuous crystn. is more widely applied. - 138Zhang, D.; Xu, S.; Du, S.; Wang, J.; Gong, J. Progress of Pharmaceutical Continuous Crystallization. Engineering 2017, 3 (3), 354– 364, DOI: 10.1016/J.ENG.2017.03.023
- 139Murugesan, S.; Sharma, P. K.; Tabora, J. E. Design of Filtration and Drying Operations. In Chemical Engineering in the Pharmaceutical Industry; Ende, D. J. A., Ed.; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2010; pp 315– 345. DOI: 10.1002/9780470882221.ch17 .
- 140Ottoboni, S.; Price, C. J.; Steven, C.; Meehan, E.; Barton, A.; Firth, P.; Mitchell, A.; Tahir, F. Development of a Novel Continuous Filtration Unit for Pharmaceutical Process Development and Manufacturing. J. Pharm. Sci. 2019, 108 (1), 372– 381, DOI: 10.1016/j.xphs.2018.07.005[Crossref], [PubMed], [CAS], Google Scholar140https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhs1Kkt7%252FE&md5=202c883c4aa06619e41a49e7c9cf95c7Development of a novel continuous filtration unit for pharmaceutical process development and manufacturingOttoboni, Sara; Price, Chris J.; Steven, Christopher; Meehan, Elizabeth; Barton, Alastair; Firth, Paul; Mitchell, Andy; Tahir, FurqanJournal of Pharmaceutical Sciences (Philadelphia, PA, United States) (2019), 108 (1), 372-381CODEN: JPMSAE; ISSN:0022-3549. (Elsevier Inc.)The lack of a com. lab., pilot and small manufg. scale dead end continuous filtration and drying unit it is a significant gap in the development of continuous pharmaceutical manufg. processes for new active pharmaceutical ingredients (APIs). To move small-scale pharmaceutical isolation forward from traditional batch Nutsche filtration to continuous processing a continuous filter dryer prototype unit (CFD20) was developed in collaboration with Alconbury Weston Ltd. The performance of the prototype was evaluated by comparison with manual best practice exemplified using a modified Biotage VacMaster unit to gather data and process understanding for API filtration and washing. The ultimate objective was to link the chem. and phys. attributes of an API slurry with equipment and processing parameters to improve API isolation processes. Filtration performance was characterized by assessing filtrate flow rate by application of Darcy's law, the impact on product crystal size distribution and product purity were investigated using classical anal. methods. The overall performance of the 2 units was similar, showing that the prototype CFD20 can match best manual lab. practice for filtration and washing while allowing continuous processing and real-time data logging. This result is encouraging and the data gathered provides further insight to inform the development of CFD20.
- 141Acevedo, D.; Jarmer, D. J.; Burcham, C. L.; Polster, C. S.; Nagy, Z. K. A Continuous Multi-Stage Mixed-Suspension Mixed-Product-Removal Crystallization System with Fines Dissolution. Chem. Eng. Res. Des. 2018, 135, 112– 120, DOI: 10.1016/j.cherd.2018.05.029[Crossref], [CAS], Google Scholar141https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtVKnsb3N&md5=e0dbb4d9104992540b5133311c34359bA continuous multi-stage mixed-suspension mixed-product-removal crystallization system with fines dissolutionAcevedo, David; Jarmer, Daniel J.; Burcham, Christopher L.; Polster, Christopher S.; Nagy, Zoltan K.Chemical Engineering Research and Design (2018), 135 (), 112-120CODEN: CERDEE; ISSN:1744-3563. (Elsevier B.V.)This work demonstrates how crystal particle size is affected by the addn. of fines dissoln. in a mixed suspension mixed product removal (MSMPR) cascade system. The cooling crystn. of paracetamol in water was used as a case study. Two MSMPR cascade configurations were evaluated: (i) nucleation-growth (NG), and (ii) nucleation-dissoln.-growth (NDG). Simulation results demonstrate that adding a dissoln. step in the MSMPR cascade configuration increases the av. product crystal size. Larger crystals and a narrower distribution were obtained due to the removal of fine crystals through the continuous process. However, a small decrease in the achievable yield was obsd. Exptl. results also showed a similar effect on the final mean size and CSD. The results demonstrate that a dissoln. stage can be optimized to increase the final crystal size and CSD by varying the dissoln. temp. to optimize the CSD with min. effect on the achievable yield.
- 142Zhang, H.; Quon, J.; Alvarez, A. J.; Evans, J.; Myerson, A. S.; Trout, B. Development of Continuous Anti-Solvent/Cooling Crystallization Process Using Cascaded Mixed Suspension, Mixed Product Removal Crystallizers. Org. Process Res. Dev. 2012, 16 (5), 915– 924, DOI: 10.1021/op2002886[ACS Full Text
], [CAS], Google Scholar
142https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XjvVSgtbY%253D&md5=e3de64a19f9cd9506c8ea2375ff2a43fDevelopment of Continuous Anti-Solvent/Cooling Crystallization Process using Cascaded Mixed Suspension, Mixed Product Removal CrystallizersZhang, Haitao; Quon, Justin; Alvarez, Alejandro J.; Evans, James; Myerson, Allan S.; Trout, BernhardtOrganic Process Research & Development (2012), 16 (5), 915-924CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)This paper describes a two-stage mixed-suspension, mixed-product removal (MSMPR) continuous crystn. developed for a pharmaceutical intermediate which uses anti-solvent and cooling to generate supersatn. The results indicate that the stage in which anti-solvent is added has a significant effect on the final crystal properties, while purity and yield were nearly identical. The population balance model was employed to det. growth and nucleation kinetics through parameter estn. With the incorporation of measured equil. distribution coeffs., the model was used to optimize crystal purity and yield of the product with respect to operating temp. and residence time. - 143Wierzbowska, B.; Hutnik, N.; Piotrowski, K.; Matynia, A. Continuous Mass Crystallization of Vitamin C in l-(+)-Ascorbic Acid–Ethanol–Water System: Size-Independent Growth Kinetic Model Approach. Cryst. Growth Des. 2011, 11 (5), 1557– 1565, DOI: 10.1021/cg101521k[ACS Full Text
], [CAS], Google Scholar
143https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXjs1Sntbs%253D&md5=bdda826c27a590a39c53125f9fce6e58Continuous Mass Crystallization of Vitamin C in L(+)-Ascorbic Acid-Ethanol-Water System: Size-Independent Growth Kinetic Model ApproachWierzbowska, Boguslawa; Hutnik, Nina; Piotrowski, Krzysztof; Matynia, AndrzejCrystal Growth & Design (2011), 11 (5), 1557-1565CODEN: CGDEFU; ISSN:1528-7483. (American Chemical Society)Exptl. results concerning continuous isohydrical drowning-out mass crystn. of vitamin C in an L(+)-ascorbic acid-ethanol-water system are presented. The process environment was created in a lab.-scale draft tube, mixed suspension mixed product removal (DT MSMPR) crystallizer with internal circulation of suspension. Assuming a const. feed concn. of ethanol (20 mass %), the feed concn. of vitamin C was changed within the 30-50 mass % range. The mean residence time of suspension in working vol. of a crystallizer was varied from 900 to 3600 s. Combinations of the presented above input process parameters resulted in a productivity 244-1692 kg/(m3 h) of crystal product, of mean size within 0.20-0.24 mm and of CV within the 50-60% range. The supersatn. level in mother soln. reached relatively high values (up to ca. 6.5 mass %), in particular at a short mean residence time of suspension (900 s). With this time elongation to 3600 s, the supersatn. level decreased, however, by ca. 40% (to 3.9 mass %). The simplest size-independent growth (SIG) kinetic model was adopted for nucleation ((3.3-27.8) × 107 1/(s m3)) and crystal growth ((1.6-6.9) × 10-8 m/s) rates estn. Kinetic relations and feedback between nucleation and crystals growth were identified and analyzed in detail. - 144Li, J.; Lai, T. C.; Trout, B. L.; Myerson, A. S. Continuous Crystallization of Cyclosporine: Effect of Operating Conditions on Yield and Purity. Cryst. Growth Des. 2017, 17 (3), 1000– 1007, DOI: 10.1021/acs.cgd.6b01212[ACS Full Text
], [CAS], Google Scholar
144https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXotlShtg%253D%253D&md5=a1b1cbe9cccd48ce0bbdff0a3d1c4a2bContinuous Crystallization of Cyclosporine: Effect of Operating Conditions on Yield and PurityLi, Jicong; Lai, Tsai-ta C.; Trout, Bernhardt L.; Myerson, Allan S.Crystal Growth & Design (2017), 17 (3), 1000-1007CODEN: CGDEFU; ISSN:1528-7483. (American Chemical Society)A continuous crystn. process has potential advantages such as lower cost and improved flexibility in pharmaceutical prodn. when compared to batch crystn. In this work, multistage continuous cooling crystn. processes for cyclosporine were developed. The approach demonstrated that optimization of stage conditions can be used to improve yield and purity. For a multi-impurity system such as cyclosporine, the segregation of each impurity should be estd. sep. due to their different behaviors. The effective distribution coeffs. of the impurities were calcd. and related to the steady state mother liquor concns. A population balance and mass balance model including distribution coeffs. for impurities was used to est. max. yields and purities that could be obtained at various operating conditions. The results showed the limitation in yield and purity improvement using a mixed-suspension, mixed-product removal cascade. In addn., optimization along with economic anal. can aid in detg. operating conditions for a high yield with acceptable equipment and operation cost. - 145Yang, Y.; Song, L.; Gao, T.; Nagy, Z. K. Integrated Upstream and Downstream Application of Wet Milling with Continuous Mixed Suspension Mixed Product Removal Crystallization. Cryst. Growth Des. 2015, 15 (12), 5879– 5885, DOI: 10.1021/acs.cgd.5b01290[ACS Full Text
], [CAS], Google Scholar
145https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhslWjsL3K&md5=d016ac34826f26f24712b48fb11fbf2aIntegrated Upstream and Downstream Application of Wet Milling with Continuous Mixed Suspension Mixed Product Removal CrystallizationYang, Yang; Song, Liangcheng; Gao, Tianyue; Nagy, Zoltan K.Crystal Growth & Design (2015), 15 (12), 5879-5885CODEN: CGDEFU; ISSN:1528-7483. (American Chemical Society)The integration of rotor-stator wet mill and continuous mixed suspension mixed product removal crystallizer (MSMPRC) is studied in this work. The wet mill is applied in two different configurations: downstream in a recycle loop to continuously reduce particle size via controlled secondary nucleation and breakage and upstream as a high shear nucleator to continuously generate seed crystals in situ. These two novel approaches are compared to continuous MSMPRC without wet mill. In addn., the effects of wet mill operating conditions (i.e., tip speed, no. of turnover per residence time) on five important product and process qualities, including av. particle size, particle no., width of size distribution, yield, and startup duration are identified for both configurations. The paper demonstrates that desired av. crystal size, narrow size distribution, high yield, and very short startup duration can be achieved at the same time by applying the rotor-stator wet mill upstream as a continuous in situ high shear seed generator followed by continuous MSMPRC. - 146Hou, G.; Power, G.; Barrett, M.; Glennon, B.; Morris, G.; Zhao, Y. Development and Characterization of a Single Stage Mixed-Suspension, Mixed-Product-Removal Crystallization Process with a Novel Transfer Unit. Cryst. Growth Des. 2014, 14 (4), 1782– 1793, DOI: 10.1021/cg401904a[ACS Full Text
], [CAS], Google Scholar
146https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXisFaitLc%253D&md5=d514ca06c5162b67a442e9d033738452Development and Characterization of a Single Stage Mixed-Suspension, Mixed-Product-Removal Crystallization Process with a Novel Transfer UnitHou, Guangyang; Power, Graham; Barrett, Mark; Glennon, Brian; Morris, Gary; Zhao, YanCrystal Growth & Design (2014), 14 (4), 1782-1793CODEN: CGDEFU; ISSN:1528-7483. (American Chemical Society)A continuously operated single stage mixed-suspension, mixed-product-removal (MSMPR) crystallizer using intermittent withdrawal via a dip pipe with combined pressure/vacuum was successfully developed for the manuf. of active pharmaceutical ingredients. Approx. 5.8% of the total operating vol. was intermittently removed at a high velocity using vacuum. The transfer line was also periodically purged with nitrogen to ensure complete removal of residual solids. In situ process anal. technologies (focused beam reflective measurement (FBRM) and process video microscopy (PVM)) were successfully applied to monitor and characterize the MSMPR crystn. process. In this study, a cooling crystn. of paracetamol from an aq. iso-Pr alc. soln. was investigated. Exptl. results indicate that the crystn. system was able to operate without any clogging issues for over 10 residence times, before which the system had approached steady state. Three different start-up strategies for continuous crystn. were investigated, and the results indicate that the chord length distributions at steady state were the same for all cases. Also, starting the continuous operation from a satd. soln. that was seeded with product from a previous MSMPR run offered the quickest route to steady state. To better control and scale up the crystn. process, the nucleation and crystal growth kinetics of the model compd. were also detd. through use of the newly developed process. The growth rates were found to be size dependent, and an exponential three-parameter model was employed to characterize the size-dependent growth. It was seen that the crystal growth rate was extremely low and increased linearly with particle size when the particle size was below 10 μm. However, the growth rate increased dramatically with particle size when the particle size was between 10 and 1000 μm. The nucleation kinetics was correlated by the semiempirical equation BTOT = 1.11 × 1015MT0.98Gavg1.12. The orders of the total nucleation rate with respect to the magma d. and av. growth rate were 0.98 and 1.12, resp. Therefore, the effect of supersatn. (or residence time) and magma d. on the steady state crystal size was investigated. - 147Cui, Y.; O’Mahony, M.; Jaramillo, J. J.; Stelzer, T.; Myerson, A. S. Custom-Built Miniature Continuous Crystallization System with Pressure-Driven Suspension Transfer. Org. Process Res. Dev. 2016, 20 (7), 1276– 1282, DOI: 10.1021/acs.oprd.6b00113[ACS Full Text
], [CAS], Google Scholar
147https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XpsF2mtLg%253D&md5=40be611c1c58e151b5e617708cf9c09fCustom-Built Miniature Continuous Crystallization System with Pressure-Driven Suspension TransferCui, Yuqing; O'Mahony, Marcus; Jaramillo, Juan J.; Stelzer, Torsten; Myerson, Allan S.Organic Process Research & Development (2016), 20 (7), 1276-1282CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)At the bench scale, the transfer of solid-liq. streams between reaction vessels or crystallizers that operate continuously poses a significant problem. Reduced equipment size of pumps and valves (i.e., approaching that on the microfluidic scale) means even further reduced orifices in which suspensions must attempt to flow. It forces bridging of solids and leads to blockages in flow. This study presents a new pressure-driven flow crystallizer (PDFC) with a custom-built suspension transfer pumping system. In the system, a dip tube is used to carry suspension between crystallizers by controlling the pressure differences of the crystallizers. This novel system has a small footprint on the scale of similar benchtop flow synthesis systems and has been demonstrated to operate continuously with intermittent withdrawal for at least 24 h. The system accommodates both cooling and antisolvent crystn. It is compatible with a variety of solvents, can handle crystals with large and small aspect ratios, and can also handle a large range of crystal sizes and suspension d. The miniature design of the system requires as little as 0.36 psig (0.025 bar(g)) pressure to operate and a design equation can be used to guide the estn. of the min. pressure needed for the transfer of suspensions at larger scales. - 148Quon, J. L.; Zhang, H.; Alvarez, A.; Evans, J.; Myerson, A. S.; Trout, B. L. Continuous Crystallization of Aliskiren Hemifumarate. Cryst. Growth Des. 2012, 12 (6), 3036– 3044, DOI: 10.1021/cg300253a[ACS Full Text
], [CAS], Google Scholar
148https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XmtlCktb8%253D&md5=548dd3f73bcb5256b67477e341930f46Continuous Crystallization of Aliskiren HemifumarateQuon, Justin L.; Zhang, Haitao; Alvarez, Alejandro; Evans, James; Myerson, Allan S.; Trout, Bernhardt L.Crystal Growth & Design (2012), 12 (6), 3036-3044CODEN: CGDEFU; ISSN:1528-7483. (American Chemical Society)Active ingredients in most pharmaceutical products are complex org. mols. that require crystn. as a purifn. and isolation step that results in a pure product at a high process yield. Knowledge of the operating conditions required to obtain crystals with the desired crystal shape, polymorph, and morphol. is crit. during process development. This paper describes a two-stage mixed suspension mixed product removal (MSMPR) continuous reactive crystn. procedure developed for Aliskiren hemifumarate. This process was able to crystallize Aliskiren hemifumarate at both high purity (> 99%) and high yield (> 92%). A model of the crystn. was developed through the simultaneous soln. of a population balance equation, kinetic expression for crystal growth and nucleation, and a mass balance. Exptl. data were fit to the model to obtain kinetic parameters for crystal growth and nucleation. After including equil. distribution coeff. data, the model was used to optimize crystal purity and yield of the product by adjusting the operating temp. and residence time. This process was integrated into an end-to-end continuous manufg. system developed at MIT. - 149Alvarez, A. J.; Myerson, A. S. Continuous Plug Flow Crystallization of Pharmaceutical Compounds. Cryst. Growth Des. 2010, 10 (5), 2219– 2228, DOI: 10.1021/cg901496s[ACS Full Text
], [CAS], Google Scholar
149https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjvFertL8%253D&md5=d28b80bf2f65721ddae4728799c3720eContinuous Plug Flow Crystallization of Pharmaceutical CompoundsAlvarez, Alejandro J.; Myerson, Allan S.Crystal Growth & Design (2010), 10 (5), 2219-2228CODEN: CGDEFU; ISSN:1528-7483. (American Chemical Society)Crystn. processes in the pharmaceutical industry are usually designed to obtain crystals with controlled size, shape, purity, and polymorphic form. Knowledge of the process conditions required to fabricate crystals with controlled characteristics is crit. during process development. In this work, continuous crystn. of ketoconazole, flufenamic acid, and L-glutamic acid in a nonconventional plug flow crystallizer was investigated. Kenics type static mixers were used to promote homogeneous mixing of active pharmaceutical ingredient soln. and antisolvent. A strategy of multiple points of addn. of antisolvent along the crystallizer was evaluated to control the size of the crystals. Interestingly, it was found that crystal size can be increased or decreased with an increased no. of antisolvent addn. points, depending on the kinetics of the system. It was also found that smaller crystals with a narrower size distribution can be obtained with the static mixers. A model to describe the continuous crystn. process was developed through the simultaneous soln. of a population balance equation, kinetics expressions for crystal growth and nucleation, and a mass balance. The comparison of exptl. and calcd. values for crystal size distribution revealed that a growth rate dispersion model could describe accurately the continuous crystn. process. Collision of crystals with each other and with mixing elements inside the crystallizer may be the source of random fluctuation of the growth rate in the nonconventional plug flow crystallizer with static mixers. - 150Majumder, A.; Nagy, Z. K. Fines Removal in a Continuous Plug Flow Crystallizer by Optimal Spatial Temperature Profiles with Controlled Dissolution. AIChE J. 2013, 59 (12), 4582– 4594, DOI: 10.1002/aic.14196[Crossref], [CAS], Google Scholar150https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXht1SkurbM&md5=2936691dbd0abc16b6e90987a6c982aaFines removal in a continuous plug flow crystallizer by optimal spatial temperature profiles with controlled dissolutionMajumder, Aniruddha; Nagy, Zoltan K.AIChE Journal (2013), 59 (12), 4582-4594CODEN: AICEAC; ISSN:0001-1541. (John Wiley & Sons, Inc.)This work presents a systematic study for obtaining the optimal temp. profile in a continuous plug flow crystallizer (PFC). The proposed PFC consists of multiple segments where the temp. of each segment can be controlled individually. An optimization problem is formulated for a target crystal size distribution (without fines) with the temp. of the segments as decision variables. The results indicate that for the crystn. kinetics considered, dissoln. steps are necessary for the redn. of fines due to nucleation. A systematic study on the form of growth and dissoln. kinetics suggested that the key factor that dets. whether the dissoln. steps will be successful in reducing fines, without compromising the final size of the crystals from seed, is the size dependence of the growth and dissoln. kinetics. Best fines removal is achieved when the larger crystals grow faster than the smaller ones and the smaller crystals dissolve faster than the larger ones.
- 151Furuta, M.; Mukai, K.; Cork, D.; Mae, K. Continuous Crystallization Using a Sonicated Tubular System for Controlling Particle Size in an API Manufacturing Process. Chem. Eng. Process. 2016, 102, 210– 218, DOI: 10.1016/j.cep.2016.02.002[Crossref], [CAS], Google Scholar151https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xitl2isrY%253D&md5=fcfd869148bfd377c14a3c4660432d69Continuous crystallization using a sonicated tubular system for controlling particle size in an API manufacturing processFuruta, Masashi; Mukai, Kouji; Cork, David; Mae, KazuhiroChemical Engineering and Processing (2016), 102 (), 210-218CODEN: CENPEU; ISSN:0255-2701. (Elsevier B.V.)A pH swing crystn. that was very sensitive toward scale-up and required specific equipment due to the non-Newtonian characteristics of the fluid, has been investigated using continuous crystn. (CC) as a means to improve particle size control, scalability and potentially reduce manufg. cost. A sonicated tubular crystallizer was designed and shown to allow the pH swing crystn. to be operated robustly without blockage, to give a wide range of desired particle size. The results were compared with crystn. in a premixing semi-batch system and a simple addn. semi-batch system under similar conditions. Interestingly, it was found that the sonicated tubular system was the only one of the three systems that gave the desired control for small particle size (1-7 μm). Even though the productivity of the sonicated tubular crystallizer still needs to be increased to allow CC and numbering-up to become cost competitive with batch crystn., we have demonstrated the potential of continuous tubular crystn. as a useful alternative method to conventional batch operation.
- 152Eder, R. J. P.; Schmitt, E. K.; Grill, J.; Radl, S.; Gruber-Woelfler, H.; Khinast, J. G. Seed Loading Effects on the Mean Crystal Size of Acetylsalicylic Acid in a Continuous-Flow Crystallization Device. Cryst. Res. Technol. 2011, 46 (3), 227– 237, DOI: 10.1002/crat.201000634[Crossref], [CAS], Google Scholar152https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXit1GrtLc%253D&md5=2d0daacf6b4339d0a11cbb7fd5cf36f0Seed loading effects on the mean crystal size of acetylsalicylic acid in a continuous-flow crystallization deviceEder, R. J. P.; Schmitt, E. K.; Grill, J.; Radl, S.; Gruber-Woelfler, H.; Khinast, J. G.Crystal Research and Technology (2011), 46 (3), 227-237CODEN: CRTEDF; ISSN:0232-1300. (Wiley-VCH Verlag GmbH & Co. KGaA)This study investigates the effects of seed loading on the mean crystal size of the model substance, acetylsalicylic acid, crystd. from ethanol in a continuously seeded tubular crystallizer. A hot, highly concd. ethanolic acetylsalicylic acid soln. was mixed with an acetylsalicylic acid-ethanol seed suspension. Subsequent cooling of the slurry in the tubing promoted supersatn. and hence crystal growth. The tubular shape of the 15 m-long crystallizer with an inner diam. of 2 mm enabled narrow residence time distributions of the crystals in the pipe and excellent temp. control in the radial direction and along the tubing. Crystals entering the crystallizer had both identical growth conditions in each section and about the same time for crystal growth. Narrow crystal size distributions were achieved with decreasing differences in the vol.-mean-diam. sizes of the seed and product crystals as seed loadings increased. Decreasing the seed size had a similar effect as increasing the seed loading, since in that case the same amt. of seed mass resulted in more individual seed particles. Altering the arrangement of the coiled crystallizer with respect to spatial directions (horizontal, vertical) did not lead to a significantly different outcome. All expts. produced considerably larger product crystals in comparison to the seeds despite relatively short crystn. times of less than 3 min. Moreover, product mass gains of a few hundred percent at a g/min-scale were achieved. Similarities in product crystal samples taken at different times at the outlet of the crystallizer showed that steady-state conditions were rapidly reached in the continuous flow crystn. device. (© 2011 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim).
- 153Eder, R. J. P.; Radl, S.; Schmitt, E.; Innerhofer, S.; Maier, M.; Gruber-Woelfler, H.; Khinast, J. G. Continuously Seeded, Continuously Operated Tubular Crystallizer for the Production of Active Pharmaceutical Ingredients. Cryst. Growth Des. 2010, 10 (5), 2247– 2257, DOI: 10.1021/cg9015788[ACS Full Text
], [CAS], Google Scholar
153https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXksF2mtbo%253D&md5=6f31ca1be8ccf6b34c96b586601649beContinuously Seeded, Continuously Operated Tubular Crystallizer for the Production of Active Pharmaceutical IngredientsEder, Rafael J. P.; Radl, Stefan; Schmitt, Elisabeth; Innerhofer, Sabine; Maier, Markus; Gruber-Woelfler, Heidrun; Khinast, Johannes G.Crystal Growth & Design (2010), 10 (5), 2247-2257CODEN: CGDEFU; ISSN:1528-7483. (American Chemical Society)A continuously operated tubular crystallizer system with an inner diam. of 2.0 mm has been successfully operated. It allows the crystn. of active pharmaceutical ingredients (APIs) under controlled conditions. Acetylsalicylic acid (ASA) which was crystd. from ethanol (EtOH) was used as the model substance. An ethanolic suspension of ASA-seeds was fed into the tubular crystallizer system, where it was mixed with a slightly undersatd. ASA-EtOH soln. that was kept at an elevated temp. in its storage vessel. Supersatn. was created via cooling and the seeds grew to form the product crystals. This work mainly focuses on the proof-of-concept and on the impact of the flow rates on the product crystals and the crystal size distribution (CSD). All other parameters including concns., temps., and loading of seeds were kept const. Higher flow velocities generally resulted in reduced no. and vol. mean diams., due to reduced tendency of agglomeration and decreased time for crystal growth due to shorter residence times of the suspension in the tube. Generally, all expts. unmistakably led to shifting of vol. d. distributions toward significantly larger values for product crystals in comparison to the seeds and were capable of yielding product masses in a g/min scale. - 154McGlone, T.; Briggs, N. E. B.; Clark, C. A.; Brown, C. J.; Sefcik, J.; Florence, A. J. Oscillatory Flow Reactors (OFRs) for Continuous Manufacturing and Crystallization. Org. Process Res. Dev. 2015, 19 (9), 1186– 1202, DOI: 10.1021/acs.oprd.5b00225[ACS Full Text
], [CAS], Google Scholar
154https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXht12gs77K&md5=eda31d1a009eb8e76e0fff6c5bf6e3acOscillatory Flow Reactors (OFRs) for Continuous Manufacturing and CrystallizationMcGlone, Thomas; Briggs, Naomi E. B.; Clark, Catriona A.; Brown, Cameron J.; Sefcik, Jan; Florence, Alastair J.Organic Process Research & Development (2015), 19 (9), 1186-1202CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)Continuous crystn. is an attractive approach for the delivery of consistent particles with specified crit. quality attributes (CQAs), which are attracting increased interest for the manuf. of high value materials, including fine chems. and pharmaceuticals. Oscillatory flow reactors (OFRs) offer a suitable platform to deliver consistent operating conditions under plug-flow operation while maintaining a controlled steady state. This review provides a brief overview of OFR technol. before outlining the operating principles and summarizing applications, emphasizing the use for controlled continuous crystn. While significant progress has been made to date, areas for further development are highlighted that will enhance the range of applications and ease of implementation of OFR technol. These depend on specific applications but include scale down, materials of construction suitable for chem. compatibility, encrustation mitigation, the enhancement of robust operation via automation, process anal. technol. (PAT), and real-time feedback control. - 155Pena, R.; Oliva, J. A.; Burcham, C. L.; Jarmer, D. J.; Nagy, Z. K. Process Intensification through Continuous Spherical Crystallization Using an Oscillatory Flow Baffled Crystallizer. Cryst. Growth Des. 2017, 17 (9), 4776– 4784, DOI: 10.1021/acs.cgd.7b00731[ACS Full Text
], [CAS], Google Scholar
155https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXht1Sjt7fP&md5=f9ad199056c2dc514089c21e0d47993fProcess Intensification through Continuous Spherical Crystallization Using an Oscillatory Flow Baffled CrystallizerPena, Ramon; Oliva, Joseph A.; Burcham, Christopher L.; Jarmer, Daniel J.; Nagy, Zoltan K.Crystal Growth & Design (2017), 17 (9), 4776-4784CODEN: CGDEFU; ISSN:1528-7483. (American Chemical Society)Drug substance purifn. by crystn. is a key interface in going from drug substance synthesis to final formulation and can often be a bottleneck in process efficiency. There has been increased importance in the development of continuous crystn. systems of active pharmaceutical ingredients to produce crystals with targeted phys. and biopharmaceutical properties. Continuous spherical crystn. (CSC) is a process intensification technique that can address many of the present flaws (e.g. size distribution, downstream processing efficiency) of traditional crystn. systems. In this study, a novel concept and method in the field of process intensification through continuous spherical crystn. is proposed. This study is based on performing crystn./spherical agglomeration in an oscillatory flow baffled crystallizer (OFBC). OFBCs are comparable to plug flow crystallizers (PFCs) in that they are both tubular crystallizers, however, the OFBC has periodically spaced orifice baffles with oscillatory motion overlapped on the net flow. Independent crystn. mechanisms can theor. be achieved through spatially distributed soln., solvent, anti-solvent, and bridging liq. addn.; offering more control of each mechanism. However, our studies showed that the OFBC allowed for spatially distributed addn. of solvents but achieving control of each mechanism individually was not attainable due to the back mixing of the system. - 156Liu, Y. C.; Dunn, D.; Lipari, M.; Barton, A.; Firth, P.; Speed, J.; Wood, D.; Nagy, Z. K. A Comparative Study of Continuous Operation between a Dynamic Baffle Crystallizer and a Stirred Tank Crystallizer. Chem. Eng. J. 2019, 367, 278– 294, DOI: 10.1016/j.cej.2019.02.129[Crossref], [CAS], Google Scholar156https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXjvFOgsLY%253D&md5=90bbb9b40481f08a73ba474d2d8df7e1A comparative study of continuous operation between a dynamic baffle crystallizer and a stirred tank crystallizerLiu, Yiqing Claire; Dunn, Davis; Lipari, Mary; Barton, Alastair; Firth, Paul; Speed, Jonathon; Wood, Dan; Nagy, Zoltan K.Chemical Engineering Journal (Amsterdam, Netherlands) (2019), 367 (), 278-294CODEN: CMEJAJ; ISSN:1385-8947. (Elsevier B.V.)Crystn., often as the final isolation and purifn. step in drug substance manufg., has substantial impact on downstream efficiency and final drug product quality. It is a crit. but challenging step in developing end-to-end continuous manufg., which has been identified as an emerging technol. in the pharmaceutical manufg. sector by the U.S. Food and Drug Administration (FDA). A traditional stirred tank crystallizer (STC) operated as a mixed-suspension-mixed-product-removal (MSMPR) system is a popular choice for continuous crystn. for its utilization of existing knowledge and equipment. However, there are disadvantages assocd. with agitational systems like the STC, such as poor local mixing and high shear rate. A com. dynamic baffle crystallizer (DBC) was studied here as an alternative unit operation. The DBC in this study consists of a jacketed glass vessel with dynamic 'donut' shaped baffles to provide oscillatory mixing to improve heat and mass transfer while exerting less shear. Our goal is to compare continuous operation performances in the DBC with the STC as MSMPR systems. First, residence time distribution (RTD) studies of both systems were performed considering single phase (liq. only) and two-phase (solid-liq.) operation. The RTD gives good insights into the mixing dynamics without computationally heavy fluid dynamic simulations. Continuous cooling crystn. of paracetamol was performed in the DBC and the STC. The DBC showed good potential to be used as an MSMPR system producing more uniform RTDs as well as higher quality crystn. products compared to a traditional STC.
- 157McLachlan, H.; Ni, X.-W. On the Effect of Added Impurity on Crystal Purity of Urea in an Oscillatory Baffled Crystallizer and a Stirred Tank Crystallizer. J. Cryst. Growth 2016, 442, 81– 88, DOI: 10.1016/j.jcrysgro.2016.03.001[Crossref], [CAS], Google Scholar157https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XktV2mt7c%253D&md5=9fc4fd17d15acf7e2bbaa522e6569743On the effect of added impurity on crystal purity of urea in an oscillatory baffled crystallizer and a stirred tank crystallizerMcLachlan, Hannah; Ni, Xiong-WeiJournal of Crystal Growth (2016), 442 (), 81-88CODEN: JCRGAE; ISSN:0022-0248. (Elsevier B.V.)Previous work has indicated that crystals produced in oscillatory baffled crystallizers (OBC) from a relatively 'pure' starting environment gave statistically higher purities than that in stirred tank crystallizers (STC) under comparable conditions. In this work, a known amt. of biuret (the impurity) was added to the 'pure' urea system and the results show that the OBC still produced higher purity crystals than the STC, although these purity values were statistically lower than from the 'pure' environment in both vessels. By evaluating crystn. rates of both urea and biuret, we noticed that these rates are higher in the STC than in the OBC, which would have led to small crystals in the former vessel. The CSD data however gave the opposite result where the CSD is wider with more, large crystals in the STC than in the OBC, in particular in the presence of added impurity. These larger crystals are likely formed due to agglomeration coupled with incorporation of impurity, which leads to a lower purity.
- 158Ni, X.; Liao, A. Effects of Mixing, Seeding, Material of Baffles and Final Temperature on Solution Crystallization of l-Glutamic Acid in an Oscillatory Baffled Crystallizer. Chem. Eng. J. 2010, 156 (1), 226– 233, DOI: 10.1016/j.cej.2009.10.045[Crossref], [CAS], Google Scholar158https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhsFGnur7O&md5=c976e3f41db1472e6b8a4da04d713dc5Effects of mixing, seeding, material of baffles and final temperature on solution crystallization of L-glutamic acid in an oscillatory baffled crystallizerNi, Xiongwei; Liao, AntingChemical Engineering Journal (Amsterdam, Netherlands) (2010), 156 (1), 226-233CODEN: CMEJAJ; ISSN:1385-8947. (Elsevier B.V.)In this paper, we report the effects of mixing intensity, seeding, compn. of baffle material and final temp. on meta-stable zone width (MSZW) and crystal polymorph in soln. crystn. of an industrially important compd., -glutamic acid, in an oscillatory baffled crystallizer (OBC). The results show that the MSZW decreases with increasing of mixing; meta-stable α crystals are transformed into stable β crystals with enhanced mixing intensity. Seeding meta-stable α crystals in operational conditions that promote β crystals leads to the formation of α crystals allowing co-existence of both forms; while seeding stable β crystals in conditions that favor α form allow β crystals prevailing in all conditions. Smoother surface of baffle material in OBC exhibits larger MSZW and favors meta-stable crystals, while rougher surface has smaller MSZW with stable crystals dominating. The meta-stable crystals gradually change into the stable form when final cooling temp. is closer to its nucleation temp. The outcomes from this work indicate that by controlling process parameters desirable crystal polymorph can be obtained in the OBC.
- 159Acevedo, D.; Yang, X.; Liu, Y. C.; O’Connor, T. F.; Koswara, A.; Nagy, Z. K.; Madurawe, R.; Cruz, C. N. Encrustation in Continuous Pharmaceutical Crystallization Processes—A Review. Org. Process Res. Dev. 2019, 23 (6), 1134– 1142, DOI: 10.1021/acs.oprd.9b00072[ACS Full Text
], [CAS], Google Scholar
159https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXptFygu7s%253D&md5=e3d1954969cb36c22034bc39e90d33aeEncrustation in Continuous Pharmaceutical Crystallization Processes-A ReviewAcevedo, David; Yang, Xiaochuan; Liu, Yiqing C.; O'Connor, Thomas F.; Koswara, Andy; Nagy, Zoltan K.; Madurawe, Rapti; Cruz, Celia N.Organic Process Research & Development (2019), 23 (6), 1134-1142CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)Encrustation is a risk factor that can cause product and process failure in continuous crystn. processes. Mitigation, prevention, and control of encrustation have been extensively researched. Various risk mitigation strategies proposed in the literature, such as coating of crystallizer walls, use of additives to control encrustation kinetics, and periodic steady-state operation show promising results in delaying or preventing encrustation. Because of the increased interest in the use of continuous crystn. in industrial applications, it is important to understand this risk factor further. This review presents recent developments on dynamic models, mechanisms, and risk factors for encrustation in continuous crystn. processes. Various design and control strategies to mitigate the encrustation risk are also summarized. Appropriate control strategies should be implemented during continuous crystn. to avoid the impact of encrustation on drug substance quality. - 160Koswara, A.; Nagy, Z. K. Anti-Fouling Control of Plug-Flow Crystallization via Heating and Cooling Cycle. IFAC-PapersOnLine 2015, 48 (8), 193– 198, DOI: 10.1016/j.ifacol.2015.08.180
- 161Reynolds, T.; Boychyn, M.; Sanderson, T.; Bulmer, M.; More, J.; Hoare, M. Scale-down of Continuous Filtration for Rapid Bioprocess Design: Recovery and Dewatering of Protein Precipitate Suspensions. Biotechnol. Bioeng. 2003, 83 (4), 454– 464, DOI: 10.1002/bit.10687[Crossref], [PubMed], [CAS], Google Scholar161https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXls1Wmtbw%253D&md5=687134dec8c00cba3a0eafab9b4edc25Scale-down of continuous filtration for rapid bioprocess design: Recovery and dewatering of protein precipitate suspensionsReynolds, T.; Boychyn, M.; Sanderson, T.; Bulmer, M.; More, J.; Hoare, M.Biotechnology and Bioengineering (2003), 83 (4), 454-464CODEN: BIBIAU; ISSN:0006-3592. (John Wiley & Sons, Inc.)The early specification of bioprocesses often has to be achieved with small (tens of milliliters) quantities of process material. If extensive process discovery is to be avoided at pilot or industrial scale, it is necessary that scale-down methods be created that not only examine the conditions of process stages but also allows prodn. of realistic output streams (i.e., streams truly representative of the large scale). These output streams can then be used in the development of subsequent purifn. operations. The traditional approach to predicting filtration operations is via a bench-scale pressure filter using const. pressure tests to examine the effect of pressure on the filtrate flux rate and filter cake dewatering. Interpretation of the results into cake resistance at unit applied pressure (α0) and compressibility (n) is used to predict the pressure profile required to maintain the filtrate flux rate at a const. predetd. value. This article reports on the operation of a continuous mode lab. filter in such a way as to prep. filter cakes and filtrate similar to what may be achieved at the industrial scale. Anal. of the filtration rate profile indicated the filter cake to have changing properties (compressibility) with time. Using the insight gained from the new scale-down methodol. gave predictions of the flux profile in a pilot-scale candle filter superior to those obtained from the traditional batch filter used for lab. development.
- 162Foley, G. A Review of Factors Affecting Filter Cake Properties in Dead-End Microfiltration of Microbial Suspensions. J. Membr. Sci. 2006, 274 (1–2), 38– 46, DOI: 10.1016/j.memsci.2005.12.008[Crossref], [CAS], Google Scholar162https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xhs12is7k%253D&md5=150187e93ae89b5a51fa576c81f9c00bA review of factors affecting filter cake properties in dead-end microfiltration of microbial suspensionsFoley, GregJournal of Membrane Science (2006), 274 (1-2), 38-46CODEN: JMESDO; ISSN:0376-7388. (Elsevier B.V.)A review. Dead-end microfiltration of microbial suspensions is reviewed with particular emphasis on the factors affecting the specific cake resistance. The effects of cell size and shape, cell surface properties (including charge), ionic environment, fermn. medium components and ageing effects are reviewed in detail. The measurement of specific resistance and correlation of cake compressibility data is described and current understanding of the underlying mechanisms of cake compression is discussed. Factors affecting the packing arrangement of cells in filter cakes are also reviewed and discussed in the context of the Carman-Kozeny equation.
- 163de Castro, M. D. L.; Priego-Capote, F. Continuous Filtration as a Separation Technique. Trends Anal. Chem. 2008, 27 (2), 101– 107, DOI: 10.1016/j.trac.2007.11.006
- 164Arunkumar, A.; Singh, N.; Peck, M.; Borys, M. C.; Li, Z. J. Investigation of Single-Pass Tangential Flow Filtration (SPTFF) as an Inline Concentration Step for Cell Culture Harvest. J. Membr. Sci. 2017, 524, 20– 32, DOI: 10.1016/j.memsci.2016.11.007[Crossref], [CAS], Google Scholar164https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvFait7bL&md5=5fd4c06de76a21fbab2725672a52f258Investigation of single-pass tangential flow filtration (SPTFF) as an inline concentration step for cell culture harvestArunkumar, Abhiram; Singh, Nripen; Peck, Michael; Borys, Michael C.; Li, Zheng JianJournal of Membrane Science (2017), 524 (), 20-32CODEN: JMESDO; ISSN:0376-7388. (Elsevier B.V.)This work examd. the use of single-pass tangential flow filtration (SPTFF) to conc. cell culture harvest. Conventional tangential flow filtration (TFF) is routinely used in biotechnol. and related industries to conc. protein solns. and exchange them into the final formulation buffer. However, several pump passes and a recirculation skid are required to achieve the target concn. in conventional TFF. Thus, it is impractical to implement TFF for inline concn. of protein solns. during the manuf. of biol. products. New developments in the biotechnol. industry have focused on using ultrafiltration in a single pump pass (also known as SPTFF) to conc. protein solns. in a single pump pass as an inline concn. step. There is thus a potential utility for this technol. to overcome manufg. bottlenecks. While there is limited work in literature describing the use of SPTFF for concn. of in-process protein pools, no work has been reported on the use of SPTFF for concn. of cell culture harvest and the challenges faced in this situation. This work examines the utility of using a 30 kDa ultrafiltration membrane in the SPTFF mode to conc. cell culture harvest for six different biol. assets in order to det. its feasibility over a wide range of biol. cell culture harvest streams. Cell culture harvest provided unique challenges to SPTFF that were overcome using a synthetic adsorptive hybrid filter technol. In particular, the hydraulic conditions necessary to achieve a targeted concn. factor varied from lot-to-lot for the same mol. This caused difficulty in implementing a given set of operating conditions for the same mol. because of process instability. To address these challenges, we used high capacity adsorptive filters that effectively reduced both particulates and sol. contaminants during primary recovery to improve the SPTFF performance. The adsorptive filter mainly removed process related impurities, specifically HCP and DNA that prematurely fouled the ultrafiltration membrane and changed the capacity of SPTFF. The adsorptive filter also eradicated cell-culture lot-to-lot performance variability with SPTFF by offering a relative std. deviation of <10%. The use of SPTFF by simultaneously coupling the clarification with the concn. was successfully demonstrated. This is the first work to report the use of SPTFF to conc. cell culture harvest for six different biol. assets, overcoming challenges using adsorptive filtration, and demonstrate debottlenecking for manufg.
- 165Song, L.; Elimelech, M. Theory of Concentration Polarization in Crossflow Filtration. J. Chem. Soc., Faraday Trans. 1995, 91 (19), 3389, DOI: 10.1039/ft9959103389[Crossref], [CAS], Google Scholar165https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXpsF2ntLY%253D&md5=96f2af4f3e8fb7d4183a7661a0d782fbTheory of concentration polarization in crossflow filtrationSong, Lianfa; Elimelech, MenachemJournal of the Chemical Society, Faraday Transactions (1995), 91 (19), 3389-98CODEN: JCFTEV; ISSN:0956-5000. (Royal Society of Chemistry)A novel theory was developed for concn. polarization of non-interacting particles in crossflow-filtration systems. This theory reveals that the extent of concn. polarization, as well as the behavior of the permeate flux, are characterized by an important dimensionless filtration no. (NF = 4πap3ΔP/3kT). There is a crit. value of NF for a given suspension and operational conditions. When NF is smaller than the crit. value, a polarization layer exists directly over the membrane surface and the wall particle concn. is detd. by the pressure and temp. At higher NF, a cake layer of retained particles forms between the polarization layer and the membrane surface. Math. models were constructed for both cases and anal. solns. for the permeate flux were derived. An increase in permeate flux with increasing pressure was predicted for all operating conditions.
- 166Gursch, J.; Hohl, R.; Toschkoff, G.; Dujmovic, D.; Brozio, J.; Krumme, M.; Rasenack, N.; Khinast, J. Continuous Processing of Active Pharmaceutical Ingredients Suspensions via Dynamic Cross-Flow Filtration. J. Pharm. Sci. 2015, 104 (10), 3481– 3489, DOI: 10.1002/jps.24562[Crossref], [PubMed], [CAS], Google Scholar166https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtFeis7jL&md5=81b8079b90d6ceb49c92e86a83fecc9dContinuous Processing of Active Pharmaceutical Ingredients Suspensions via Dynamic Cross-Flow FiltrationGursch, Johannes; Hohl, Roland; Toschkoff, Gregor; Dujmovic, Diana; Brozio, Joerg; Krumme, Markus; Rasenack, Norbert; Khinast, JohannesJournal of Pharmaceutical Sciences (2015), 104 (10), 3481-3489CODEN: JPMSAE; ISSN:0022-3549. (John Wiley & Sons, Inc.)Over the last years, continuous manufg. has created significant interest in the pharmaceutical industry. Continuous filtration at low flow rates and high solid loadings poses, however, a significant challenge. A com. available, continuously operating, dynamic cross-flow filtration device (CFF) is tested and characterized. It is shown that the CFF is a highly suitable technol. for continuous filtration. For all tested model active pharmaceutical ingredients, a material-specific strictly linear relationship between feed and permeate rate is identified. Moreover, for each tested substance, a const. concn. factor is reached. A one-parameter model based on a linear equation is suitable to fully describe the CFF filtration performance. This rather unexpected finding and the concn. polarization layer buildup is analyzed and a basic model to describe the obsd. filtration behavior is developed. © 2015 Wiley Periodicals, Inc. and the American Pharmacists Assocn. J Pharm Sci.
- 167Gursch, J.; Hohl, R.; Dujmovic, D.; Brozio, J.; Krumme, M.; Rasenack, N.; Khinast, J. Dynamic Cross-Flow Filtration: Enhanced Continuous Small-Scale Solid-Liquid Separation. Drug Dev. Ind. Pharm. 2016, 42 (6), 977– 984, DOI: 10.3109/03639045.2015.1100200[Crossref], [PubMed], [CAS], Google Scholar167https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xlt1Kksr0%253D&md5=9edb3c3ff82784982c4664b8b01bf357Dynamic cross-flow filtration: enhanced continuous small-scale solid-liquid separationGursch, Johannes; Hohl, Roland; Dujmovic, Diana; Brozio, Joerg; Krumme, Markus; Rasenack, Norbert; Khinast, JohannesDrug Development and Industrial Pharmacy (2016), 42 (6), 977-984CODEN: DDIPD8; ISSN:0363-9045. (Taylor & Francis Ltd.)In a previous study, a small-scale dynamic filtration device (SFD) was analyzed and the basic mechanisms governing the filtration process were characterized. The present work aims at improving the device's performance in terms of actual prodn. Various operation modes were tested in order to increase permeate flow and concn. factors (CF), while maintaining a fully continuous prodn. mode. Both, a vacuum-enhanced and a pulsating operation mode, proved to be superior to the currently implemented open-operation mode. For example, for lactose, an increase of the CF could be achieved from 1.7 in open mode to 7.6 in pulsating operation mode. The investigated operation strategy enables process control systems to rapidly react to fluctuating feeds that may occur due to changes in upstream manufg. steps. As a result, not only filtration performance in terms of permeate rate but also process flexibility can be significantly increased. Overall, vacuum-enhanced operation was shown to be most promising for integration into an industrial environment. The option to elevate achievable concn. factors, ease of flow monitoring as well as the ability to react to changes in the feed conditions allow for effective and efficient continuous small-scale filtration.
- 168Kossik, J. Operation of a Disposable Rotary Drum Filter. In AIChE Annual Meeting, Pharmaceutical & Biotechnology Pilot Plants; 2001.Google ScholarThere is no corresponding record for this reference.
- 169Kossik, J.; Delys, J. F. Disposable Rotary Drum Filter. US6336561B1, 2002.Google ScholarThere is no corresponding record for this reference.
- 170Wong, K. W.-S. Design of a Small-Scale Continuous Linear Motion Pharmaceutical Filtration Module. 2010.Google ScholarThere is no corresponding record for this reference.
- 171Ende, D. J. A.; Pfisterer, D.; Girard, K.; Blackwood, D. O.; Plocharczyk, E. Development of a Continuous Filter-Drier for Lab to Kilo-Lab Scale. AIChE Annual Meeting; 2011.Google ScholarThere is no corresponding record for this reference.
- 172Indexing belt filter (BF), https://www.bhs-sonthofen.com/en/process-technology/machines/filters/indexing-belt-filter-bf.Google ScholarThere is no corresponding record for this reference.
- 173Hohmann, L.; Löbnitz, L.; Menke, C.; Santhirakumaran, B.; Stier, P.; Stenger, F.; Dufour, F.; Wiese, G.; zur Horst-Meyer, S.; Kusserow, B.; Zang, W.; Nirschl, H.; Kockmann, N. Continuous Downstream Processing of Amino Acids in a Modular Miniplant. Chem. Eng. Technol. 2018, 41 (6), 1152– 1164, DOI: 10.1002/ceat.201700657[Crossref], [CAS], Google Scholar173https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXotFWisbw%253D&md5=9b16324fe1a81ba4151b8da139ff3c94Continuous downstream processing of amino acids in a modular miniplantHohmann, Lukas; Loebnitz, Lisa; Menke, Christina; Santhirakumaran, Bavatharani; Stier, Patrick; Stenger, Frank; Dufour, Fabrice; Wiese, Georg; zur Horst-Meyer, Santer; Kusserow, Burkhard; Zang, Werner; Nirschl, Hermann; Kockmann, NorbertChemical Engineering & Technology (2018), 41 (6), 1152-1164CODEN: CETEER; ISSN:1521-4125. (Wiley-VCH Verlag GmbH & Co. KGaA)Global competition leads to a need for a fast time to market and increased resource efficiency. Continuous processing, module-based plant design, and multipurpose equipment are recently discussed approaches for the fine-chem. industry. As a representative, the downstream process of amino acid prodn. is discussed herein. A conventional batch procedure was transferred to a continuous process and realized in a modular miniplant, which comprised of evapn. in a wiped-film evaporator, seeded cooling crystn. in a coiled tubular crystallizer, and solid/liq. sepn. on a vacuum belt filter. The operations were realized on-skid with individual automation systems and integrated sensors. L-Alanine was successfully processed in steady-state operation.
- 174Wong, S. Y.; Chen, J.; Forte, L. E.; Myerson, A. S. Compact Crystallization, Filtration, and Drying for the Production of Active Pharmaceutical Ingredients. Org. Process Res. Dev. 2013, 17 (4), 684– 692, DOI: 10.1021/op400011s[ACS Full Text
], [CAS], Google Scholar
174https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXktFKnsrc%253D&md5=6a310ab39cd0a0365861b3e0a63c910dCompact Crystallization, Filtration, and Drying for the Production of Active Pharmaceutical IngredientsWong, Shin Yee; Chen, Jie; Forte, Laura E.; Myerson, Allan S.Organic Process Research & Development (2013), 17 (4), 684-692CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)With the growing problem of drug shortages worldwide, a soln. could be a centralized compact pharmaceutical manufg. platform that produces API and finished drug product. In this paper, the development of a combined crystn., hybrid filtration-drying-dissoln. app. to be used in such a compact platform is discussed. Expts. were conducted to evaluate each unit operation. Crystn. expts. using a conventional stirred tank and the newly designed scraped surface crystallizer demonstrated the advantages of the new design in terms of crystn. rates, yields and the ease of automation. Postcrystn. operations were operated stepwise using the custom hybrid device that delivered satisfactory results for each operation, e.g. after filtration, Fluoxetine HCl was dried in less than 20 min, with 99% yield after dissoln. in the liq. excipient. - 175Wong, S. Y.; Tatusko, A. P.; Trout, B. L.; Myerson, A. S. Development of Continuous Crystallization Processes Using a Single-Stage Mixed-Suspension, Mixed-Product Removal Crystallizer with Recycle. Cryst. Growth Des. 2012, 12 (11), 5701– 5707, DOI: 10.1021/cg301221q[ACS Full Text
], [CAS], Google Scholar
175https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsFektbzP&md5=9cb632369b1f5425b8a974d7d863d56cDevelopment of Continuous Crystallization Processes Using a Single-Stage Mixed-Suspension, Mixed-Product Removal Crystallizer with RecycleWong, Shin Yee; Tatusko, Adam P.; Trout, Bernhardt L.; Myerson, Allan S.Crystal Growth & Design (2012), 12 (11), 5701-5707CODEN: CGDEFU; ISSN:1528-7483. (American Chemical Society)An ideal pharmaceutical crystn. process produces a pure product at a high yield while minimizing energy input, the process equipment footprint, and its complexity. A good candidate for such a process is a single-stage mixed-suspension, mixed-product removal (MSMPR) crystallizer with recycle (SMR) system, where the characteristics of the refined crystal are controlled by the crystn. conditions of the MSMPR and the yield is manipulated by the recycle ratio. In this study, two continuous SMR systems, for the cooling crystn. of cyclosporine and the antisolvent-cooling crystn. of deferasirox, were developed. Both systems were designed to maintain the desired operating conditions inside the MSMPR crystallizer. For cooling crystn., the recycle stream was concd. via vacuum evapn. For antisolvent-cooling crystn., the desired solvent to antisolvent ratio was maintained by controlling the flow rates of feed, antisolvent, and recycle streams. The max. exptl. yield and purity of the crystals were detd. as 91.8 and 94.3%, resp. (for cyclosporine) and 89.1% with 0.2 ppm impurity A, resp. (for deferasirox). For cyclosporine, this yield is 5.5% higher than that of a multistage MSMPR with a recycle system. Addnl., the SMR system is relatively simple, having a lower operational demand, in terms of space and no. of unit operations required. - 176Ferguson, S.; Ortner, F.; Quon, J.; Peeva, L.; Livingston, A.; Trout, B. L.; Myerson, A. S. Use of Continuous MSMPR Crystallization with Integrated Nanofiltration Membrane Recycle for Enhanced Yield and Purity in API Crystallization. Cryst. Growth Des. 2014, 14 (2), 617– 627, DOI: 10.1021/cg401491y[ACS Full Text
], [CAS], Google Scholar
176https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvV2rsbzK&md5=7f8024ba885d94ff4f6ffbac2db214e6Use of Continuous MSMPR Crystallization with Integrated Nanofiltration Membrane Recycle for Enhanced Yield and Purity in API CrystallizationFerguson, Steven; Ortner, Franziska; Quon, Justin; Peeva, Ludmila; Livingston, Andrew; Trout, Bernhardt L.; Myerson, Allan S.Crystal Growth & Design (2014), 14 (2), 617-627CODEN: CGDEFU; ISSN:1528-7483. (American Chemical Society)If continuous processing is to be employed in pharmaceutical prodn., it is essential that continuous crystn. techniques can meet the purity and yield achievable in current batch crystn. processes. Recycling of mother liquor in steady state MSMPR crystns. allows the yield in the equiv. equil. batch process to be met or exceeded. However, the extent to which yield can be increased is limited by the buildup of impurities within the system. In this study, an org. solvent nanofiltration membrane was used to preferentially conc. an API (deferasirox, M.W. = 373 Da) and purge the limiting impurity 4-hydrazinobenzoic acid (MW = 152 Da) from the mother liquor recycle stream in a mixed solvent (THF:ethanol) antisolvent (water) system. Incorporation of the membrane recycle allowed yields of 98.0% and 98.7% to be achieved. This compares to the following: a control MSMPR run without a membrane (70.3%), an equiv. batch process (89.2%), and the current com. batch process (92%). Comparable product impurity levels were measured for the following: the MSMPR membrane recycle expts. (0.15 ppm and 0.22 ppm), the MSMPR control (0.13 ppm), and batch (0.32 ppm) control expts. All processes met the regulatory specifications of a max. of 3 ppm of the impurity 4-hydrainobenzoic acid. - 177Acevedo, D.; Peña, R.; Yang, Y.; Barton, A.; Firth, P.; Nagy, Z. K. Evaluation of Mixed Suspension Mixed Product Removal Crystallization Processes Coupled with a Continuous Filtration System. Chem. Eng. Process. 2016, 108, 212– 219, DOI: 10.1016/j.cep.2016.08.006[Crossref], [CAS], Google Scholar177https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsVWrsr3N&md5=6784d8544445650c666a2ecbe9110b30Evaluation of mixed suspension mixed product removal crystallization processes coupled with a continuous filtration systemAcevedo, David; Pena, Ramon; Yang, Yang; Barton, Alastair; Firth, Paul; Nagy, Zoltan K.Chemical Engineering and Processing (2016), 108 (), 212-219CODEN: CENPEU; ISSN:0255-2701. (Elsevier B.V.)As the pharmaceutical industry evolves and goes through the paradigm shift from batch to continuous manufg., innovative processes need to be developed to replace unit operations that have historically been batch operations. This requires innovation in the continuous crystn. field of study as well as innovation in downstream processes (e.g. filtration, drying, milling, and granulation). Herein a novel and com. available continuous filter carousel (CFC) system was assessed for its feasibility of continuous filtration while coupled with a continuous mixed suspension mixed product removal (MSMPR) crystallizer. The filtration system was assessed using two different crystn. systems (i.e. cooling and antisolvent) with significantly different kinetics and morphologies to assess the robustness of the integrated platform. With proper optimization of the various filtration parameters for the different crystn. systems a controlled state of operation was achieved in each case. The crystal product from the CFC system shows good consistency with the crystals in the slurry in the MSMPR. Moisture content and productivity of the filtration system were reported and show dependency on crystal properties. The CFC system was equipped with solvent vessels that aided the continuous filtration by acting as a wash or a clean-in-place solvent, preventing or removing filter clogging, resp.
- 178Liu, Y. C.; Domokos, A.; Coleman, S.; Firth, P.; Nagy, Z. K. Development of Continuous Filtration in a Novel Continuous Filtration Carousel Integrated with Continuous Crystallization. Org. Process Res. Dev. 2019, 23 (12), 2655– 2665, DOI: 10.1021/acs.oprd.9b00342[ACS Full Text
], [CAS], Google Scholar
178https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitFOmtL7P&md5=fea2e7d0742f615e7f1ced1a9cdfe901Development of Continuous Filtration in a Novel Continuous Filtration Carousel Integrated with Continuous CrystallizationLiu, Yiqing C.; Domokos, Andras; Coleman, Simon; Firth, Paul; Nagy, Zoltan K.Organic Process Research & Development (2019), 23 (12), 2655-2665CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)Pharmaceutical manufg. has been largely carried out in batch mode, which has disadvantages such as batch-to-batch variations and difficult scale-up. As a result, the pharmaceutical industry is going through the paradigm shift to continuous manufg. Many unit operations have been studied for continuous operation; however, the bottleneck largely lies in drug substance isolation processes, such as crystn. and filtration. Herein a novel com. continuous filtration unit, the continuous filtration carousel (CFC), coupled with an innovative continuous crystn. system, the oscillatory baffle reactor (OBR), was studied to construct a truly continuous drug substance sepn. step where supersatd. soln. was continuously crystd. and filtered to produce solid products without a hold-up tank. Two compds. were studied to evaluate the CFC performance: paracetamol and benzoic acid. Standalone filtration expts. were performed first to examine the operating parameters of the CFC from which an optimal set of operating parameters was chosen for the integration of continuous filtration and crystn. Successful integration of continuous crystn. and filtration of paracetamol and benzoic acid was demonstrated where dry uniform paracetamol crystals were obtained while benzoic acid products were high in moisture content. Implementing drying functionality in the filter chamber can improve its performance. In hopes to expedite the application of continuous filtration in the pharmaceutical industry, a risk consideration discussion is given to identify and organize potential risk factors assocd. with continuous filtration. - 179Capellades, G.; Neurohr, C.; Azad, M.; Brancazio, D.; Rapp, K.; Hammersmith, G.; Myerson, A. S. A Compact Device for the Integrated Filtration, Drying, and Mechanical Processing of Active Pharmaceutical Ingredients. J. Pharm. Sci. 2020, 109 (3), 1365– 1372, DOI: 10.1016/j.xphs.2019.12.011[Crossref], [PubMed], [CAS], Google Scholar179https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3MbjtVWiug%253D%253D&md5=1b647d1cdb4837f355845a7a7e156b8eA Compact Device for the Integrated Filtration, Drying, and Mechanical Processing of Active Pharmaceutical IngredientsCapellades Gerard; Neurohr Clemence; Azad Mohammad; Brancazio David; Rapp Kersten; Hammersmith Gregory; Myerson Allan SJournal of pharmaceutical sciences (2020), 109 (3), 1365-1372 ISSN:.Recent changes in the pharmaceutical sector call for the development of novel manufacturing approaches to reduce costs and improve control over product quality. In this area, the development of compact, plug-and-play devices that fit in a continuous manufacturing system has gained interest in recent years. Most Nutsche filters offer a versatile solution as compact filtration and drying devices. However, conventional drying processes tend to generate a large amount of lumps, usually requiring further mechanical processing of the isolated drug substance before it can be formulated. In this work, we present a compact, automatable filtration device that takes advantage of a unique impeller design and in situ measurements of the drying heat duty to integrate mechanical processing into the drying step. By preventing the formation of dry lumps during drug substance drying, and breaking needle-like crystals through the developed agitation program, the resulting powder can be directly used for tablet formulation. This device, designed to fit in a compact continuous manufacturing module, has the potential to reduce manufacturing costs and footprint, while allowing for the low-shear mechanical processing of heat-sensitive compounds.
- 180Nagy, B.; Szilágyi, B.; Domokos, A.; Tacsi, K.; Pataki, H.; Marosi, G.; Kristóf Nagy, Z.; Nagy, Z. K. Modeling of Pharmaceutical Filtration and Continuous Integrated Crystallization-Filtration Processes. Chem. Eng. J. 2020, 127566, DOI: 10.1016/j.cej.2020.127566
- 181Pernenkil, L.; Cooney, C. L. A Review on the Continuous Blending of Powders. Chem. Eng. Sci. 2006, 61 (2), 720– 742, DOI: 10.1016/j.ces.2005.06.016
- 182Berthiaux, H.; Marikh, K.; Gatumel, C. Continuous Mixing of Powder Mixtures with Pharmaceutical Process Constraints. Chem. Eng. Process. 2008, 47 (12), 2315– 2322, DOI: 10.1016/j.cep.2008.01.009[Crossref], [CAS], Google Scholar182https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXht1Kjtb3K&md5=848ae543734e2502ba333858bb067d72Continuous mixing of powder mixtures with pharmaceutical process constraintsBerthiaux, Henri; Marikh, Khadija; Gatumel, CendrineChemical Engineering and Processing (2008), 47 (12), 2315-2322CODEN: CENPEU; ISSN:0255-2701. (Elsevier B.V.)While it would provide many advantages from many aspects, the application of continuous mixing processes to the pharmaceutical field is still in its infancy. In this paper the authors report results concerning the continuous mixing of 9 ingredients (including 3 actives) that constitute a current drug. The authors examine these results in the light of different pharmaceutical process constraints, such as mixt. quality control, time-stability of this quality, sensitivity of the process to perturbations. The app. is a pilot plant Gericke GCM 500 continuous mixer with 3 loss-in-wt. feeders. A specific exptl. protocol is developed to det. the homogeneity of the mixts. at the outlet of the mixer. The homogeneity of the mixts. is examd. through industrial stds. that would allow the product to be released on the market. The steady-state operation is first reported on, and it is demonstrated that a very acceptable mixt. can be produced under certain conditions, with excellent time stability. The response of the mixer to filling sequences of 2 crit. feeders is also quantified in terms of mixt. homogeneity. It is found that it may be preferable to stop the process during these periods.
- 183Williams, J. C. Continuous Mixing of Solids. A Review. Powder Technol. 1976, 15 (2), 237– 243, DOI: 10.1016/0032-5910(76)80052-6
- 184Osorio, J. G.; Vanarase, A. U.; Romañach, R. J.; Muzzio, F. J. Continuous Powder Mixing. In Pharmaceutical Blending and Mixing; John Wiley & Sons, Ltd: Chichester, UK, 2015; pp 101– 127. DOI: 10.1002/9781118682692.ch6 .
- 185Ierapetritou, M.; Muzzio, F.; Reklaitis, G. Perspectives on the Continuous Manufacturing of Powder-Based Pharmaceutical Processes. AIChE J. 2016, 62 (6), 1846– 1862, DOI: 10.1002/aic.15210[Crossref], [CAS], Google Scholar185https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XksVWrs74%253D&md5=a5d361c9ffa62e9bcc15770da5e6d527Perspectives on the continuous manufacturing of powder-based pharmaceutical processesIerapetritou, Marianthi; Muzzio, Fernando; Reklaitis, GintarasAIChE Journal (2016), 62 (6), 1846-1862CODEN: AICEAC; ISSN:0001-1541. (John Wiley & Sons, Inc.)There is no expanded citation for this reference.
- 186Vanarase, A. U.; Muzzio, F. J. Effect of Operating Conditions and Design Parameters in a Continuous Powder Mixer. Powder Technol. 2011, 208 (1), 26– 36, DOI: 10.1016/j.powtec.2010.11.038[Crossref], [CAS], Google Scholar186https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXit1Wht7k%253D&md5=81284d426eeaed1923948b5e4a8d151cEffect of operating conditions and design parameters in a continuous powder mixerVanarase, Aditya U.; Muzzio, Fernando J.Powder Technology (2011), 208 (1), 26-36CODEN: POTEBX; ISSN:0032-5910. (Elsevier B.V.)An exptl. investigation was carried out to study the mixing performance and flow behavior in a continuous powder mixer for a typical pharmaceutical mixt. Blender performance, characterized by the relative std. deviation (RSD) of compn. of blend samples taken at the blender discharge and by the variance redn. ratio (VRR) of the blender, was measured as a function of impeller rotation rate, flow rate and blade configuration. The flow behavior in the continuous mixer was characterized using the residence time distribution (RTD) and powder hold-up measurements. To quantify the strain applied to the powder in the blender, the no. of blade passes experienced by the powder in the blender was calcd. using the residence time measurements. The relation between different exptl. parameters and mean residence time and mean centered variance was examd. The mixing performance was largely dominated by the material properties of the mixt., which had a larger effect than the ingredient flow rate variability contributed by the feeders. Holdup was strongly dependent on impeller rotation rate; as impeller rotation rate increased, holdup (and therefore, residence time) decreased sharply. As a result, intermediate rotation rates showed the best mixing performance. Blade configuration affected performance as well; blade patterns where some of the blades push the powder backwards improved the mixing performance.
- 187Osorio, J. G.; Muzzio, F. J. Effects of Processing Parameters and Blade Patterns on Continuous Pharmaceutical Powder Mixing. Chem. Eng. Process. 2016, 109, 59– 67, DOI: 10.1016/j.cep.2016.07.012[Crossref], [CAS], Google Scholar187https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsFenurbP&md5=6ec657177d099bbdbfe05adfca8d8945Effects of processing parameters and blade patterns on continuous pharmaceutical powder mixingOsorio, Juan G.; Muzzio, Fernando J.Chemical Engineering and Processing (2016), 109 (), 59-67CODEN: CENPEU; ISSN:0255-2701. (Elsevier B.V.)The present study summarizes the exptl. characterization of a new continuous powder mixer(GCG-70 by Glatt)using common pharmaceutical ingredients. The powder hold-up and residence time distribution were used to characterize the bulk behavior of the mixer as a function of impeller rotational speed, total throughput(mass flow rate)and blade configuration. The relative std. deviation(RSD), calcd. from samples taken at the outlet of the blender, was used to characterize its mixing performance. The hold-up and the mean residence time decreased with increasing impeller rotational speed. The mean centered variance and the no. of blade passes increased with increasing impeller rotational speed. The effect of the blade configuration on the mixing dynamics diminished as the rotation rate increased. The hold-up and mean residence time were sensitive enough to demonstrate the effects of blade configurations. The mixing performance, depending on the processing parameters, was found to be between 5% and 10% RSD for 5% wt./wt. active pharmaceutical ingredient(API), and <3% for 30% wt./wt. API. These results showed improvements in the mixing performance when compared to studies of other continuous mixers using similar materials and anal. techniques for quantification.
- 188Burcham, C. L.; Florence, A. J.; Johnson, M. D. Continuous Manufacturing in Pharmaceutical Process Development and Manufacturing. Annu. Rev. Chem. Biomol. Eng. 2018, 9 (1), 253– 281, DOI: 10.1146/annurev-chembioeng-060817-084355[Crossref], [PubMed], [CAS], Google Scholar188https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MbltVajsQ%253D%253D&md5=421af356c9e3540a264a63dba0ec9d4eContinuous Manufacturing in Pharmaceutical Process Development and ManufacturingBurcham Christopher L; Johnson Martin D; Florence Alastair JAnnual review of chemical and biomolecular engineering (2018), 9 (), 253-281 ISSN:.The pharmaceutical industry has found new applications for the use of continuous processing for the manufacture of new therapies currently in development. The transformation has been encouraged by regulatory bodies as well as driven by cost reduction, decreased development cycles, access to new chemistries not practical in batch, improved safety, flexible manufacturing platforms, and improved product quality assurance. The transformation from batch to continuous manufacturing processing is the focus of this review. The review is limited to small, chemically synthesized organic molecules and encompasses the manufacture of both active pharmaceutical ingredients (APIs) and the subsequent drug product. Continuous drug product is currently used in approved processes. A few examples of production of APIs under current good manufacturing practice conditions using continuous processing steps have been published in the past five years, but they are lagging behind continuous drug product with respect to regulatory filings.
- 189Järvinen, M. A.; Paaso, J.; Paavola, M.; Leiviskä, K.; Juuti, M.; Muzzio, F.; Järvinen, K. Continuous Direct Tablet Compression: Effects of Impeller Rotation Rate, Total Feed Rate and Drug Content on the Tablet Properties and Drug Release. Drug Dev. Ind. Pharm. 2013, 39 (11), 1802– 1808, DOI: 10.3109/03639045.2012.738681[Crossref], [PubMed], [CAS], Google Scholar189https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3s7ksFCrsQ%253D%253D&md5=94f64e4b0ed31edfa3f1933ebc944461Continuous direct tablet compression: effects of impeller rotation rate, total feed rate and drug content on the tablet properties and drug releaseJarvinen Maiju A; Paaso Janne; Paavola Marko; Leiviska Kauko; Juuti Mikko; Muzzio Fernando; Jarvinen KristiinaDrug development and industrial pharmacy (2013), 39 (11), 1802-8 ISSN:.CONTEXT: Continuous processing is becoming popular in the pharmaceutical industry for its cost and quality advantages. OBJECTIVE: This study evaluated the mechanical properties, uniformity of dosage units and drug release from the tablets prepared by continuous direct compression process. MATERIALS AND METHODS: The tablet formulations consisted of acetaminophen (3-30% (w/w)) pre-blended with 0.25% (w/w) colloidal silicon dioxide, microcrystalline cellulose (69-96% (w/w)) and magnesium stearate (1% (w/w)). The continuous tableting line consisted of three loss-in-weight feeders and a convective continuous mixer and a rotary tablet press. The process continued for 8 min and steady state was reached within 5 min. The effects of acetaminophen content, impeller rotation rate (39-254 rpm) and total feed rate (15 and 20 kg/h) on tablet properties were examined. RESULTS AND DISCUSSION: All the tablets complied with the friability requirements of European Pharmacopoeia and rapidly released acetaminophen. However, the relative standard deviation of acetaminophen content (10% (w/w)) increased with an increase in impeller rotation rate at a constant total feed rate (20 kg/h). A compression force of 12 kN tended to result in greater tablet hardness and subsequently a slower initial acetaminophen release from tablets when compared with those made with the compression force of about 8 kN. CONCLUSIONS: In conclusion, tablets could be successfully prepared by a continuous direct compression process and process conditions affected to some extent tablet properties.
- 190Wagner, B.; Brinz, T.; Otterbach, S.; Khinast, J. Rapid Automated Process Development of a Continuous Capsule-Filling Process. Int. J. Pharm. 2018, 546 (1–2), 154– 165, DOI: 10.1016/j.ijpharm.2018.05.009[Crossref], [PubMed], [CAS], Google Scholar190https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXpslyms7w%253D&md5=c1ed61d62a6d80c187dec739078d0610Rapid automated process development of a continuous capsule-filling processWagner, Bernhard; Brinz, Thomas; Otterbach, Stephanie; Khinast, JohannesInternational Journal of Pharmaceutics (Amsterdam, Netherlands) (2018), 546 (1-2), 154-165CODEN: IJPHDE; ISSN:0378-5173. (Elsevier B.V.)This paper introduces a rapid automated process-development approach for a continuous capsule-filling process. In our proposed method, both the material attributes and the crit. process parameters were varied to understand and to optimize the overall process. Using our approach a statistical process model can be generated with unprecedented speed (2 days), which is the prerequisite for effectively developing and operating continuous process platforms. In a first set of expts. a process model was developed using different mixt. compns. of ascorbic acid, lactose and magnesium stearate while changing simultaneously the crit. process parameters of the capsule filler (speed, pressure, immersion depth and powder bed height). Targets of the model were the mean fill wt. and the relative std. deviation of the produced capsules. In a second exptl. set the model was tested, i.e., the goal was to predict the behavior of the system at different set points in order to predict wt. and relative std. deviation for predefined targets. Predictions were very good, thus validating our approach. The combination of the rapid automated process development approach and the continuous capsule-filling process resulted in a new strategy for the development and manuf. of pharmaceutical dosage forms.
- 191Chattoraj, S.; Sun, C. C. Crystal and Particle Engineering Strategies for Improving Powder Compression and Flow Properties to Enable Continuous Tablet Manufacturing by Direct Compression. J. Pharm. Sci. 2018, 107 (4), 968– 974, DOI: 10.1016/j.xphs.2017.11.023[Crossref], [PubMed], [CAS], Google Scholar191https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjvFGqtg%253D%253D&md5=70d2c965d65e8bb39e9b8b7ef618ee2bCrystal and Particle Engineering Strategies for Improving Powder Compression and Flow Properties to Enable Continuous Tablet Manufacturing by Direct CompressionChattoraj, Sayantan; Sun, Changquan CalvinJournal of Pharmaceutical Sciences (Philadelphia, PA, United States) (2018), 107 (4), 968-974CODEN: JPMSAE; ISSN:0022-3549. (Elsevier Inc.)Continuous manufg. of tablets has many advantages, including batch size flexibility, demand-adaptive scale up or scale down, consistent product quality, small operational foot print, and increased manufg. efficiency. Simplicity makes direct compression the most suitable process for continuous tablet manufg. However, deficiencies in powder flow and compression of active pharmaceutical ingredients (APIs) limit the range of drug loading that can routinely be considered for direct compression. For the widespread adoption of continuous direct compression, effective API engineering strategies to address power flow and compression problems are needed. Appropriate implementation of these strategies would facilitate the design of high-quality robust drug products, as stipulated by the Quality-by-Design framework. Here, several crystal and particle engineering strategies for improving powder flow and compression properties are summarized. The focus is on the underlying materials science, which is the foundation for effective API engineering to enable successful continuous manufg. by the direct compression process.
- 192Ervasti, T.; Simonaho, S.-P.; Ketolainen, J.; Forsberg, P.; Fransson, M.; Wikström, H.; Folestad, S.; Lakio, S.; Tajarobi, P.; Abrahmsén-Alami, S. Continuous Manufacturing of Extended Release Tablets via Powder Mixing and Direct Compression. Int. J. Pharm. 2015, 495 (1), 290– 301, DOI: 10.1016/j.ijpharm.2015.08.077[Crossref], [PubMed], [CAS], Google Scholar192https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsVyjurvP&md5=c6b215a1b0d9e867409fdc75e48df376Continuous manufacturing of extended release tablets via powder mixing and direct compressionErvasti, Tuomas; Simonaho, Simo-Pekka; Ketolainen, Jarkko; Forsberg, Peter; Fransson, Magnus; Wikstrom, Hakan; Folestad, Staffan; Lakio, Satu; Tajarobi, Pirjo; Abrahmsen-Alami, SusannaInternational Journal of Pharmaceutics (Amsterdam, Netherlands) (2015), 495 (1), 290-301CODEN: IJPHDE; ISSN:0378-5173. (Elsevier B.V.)The aim of the current work was to explore continuous dry powder mixing and direct compression for manufg. of extended release (ER) matrix tablets. The study was span out with a challenging formulation design comprising ibuprofen compns. with varying particle size and a relatively low amt. of the matrix former hydroxypropyl methylcellulose (HPMC). Std. grade HPMC (CR) was compared to a recently developed direct compressible grade (DC2). The work demonstrate that ER tablets with desired quality attributes could be manufd. via integrated continuous mixing and direct compression. The most robust tablet quality (wt., assay, tensile strength) was obtained using high mixer speed and large particle size ibuprofen and HPMC DC2 due to good powder flow. At low mixer speed it was more difficult to achieve high quality low dose tablets. Notably, with HPMC DC2 the processing conditions had a significant effect on drug release. Longer processing time and/or faster mixer speed was needed to achieve robust release with compns. contg. DC2 compared with those contg. CR. This work confirms the importance of balancing process parameters and material properties to find consistent product quality. Also, adaptive control is proven a pivotal means for control of continuous manufg. systems.
- 193Simonaho, S.-P.; Ketolainen, J.; Ervasti, T.; Toiviainen, M.; Korhonen, O. Continuous Manufacturing of Tablets with PROMIS-Line — Introduction and Case Studies from Continuous Feeding, Blending and Tableting. Eur. J. Pharm. Sci. 2016, 90, 38– 46, DOI: 10.1016/j.ejps.2016.02.006[Crossref], [PubMed], [CAS], Google Scholar193https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xjtleqtr8%253D&md5=f4193d5a6667a8ef65468944cf8865efContinuous manufacturing of tablets with PROMIS-line - Introduction and case studies from continuous feeding, blending and tabletingSimonaho, Simo-Pekka; Ketolainen, Jarkko; Ervasti, Tuomas; Toiviainen, Maunu; Korhonen, OssiEuropean Journal of Pharmaceutical Sciences (2016), 90 (), 38-46CODEN: EPSCED; ISSN:0928-0987. (Elsevier B.V.)Drug manufg. technol. is in the midst of modernization and continuous manufg. of drug products is esp. the focus of great interest. The adoption of new manufg. approaches requires extensive cooperation between industry, regulatory bodies, academics and equipment manufacturers. In this paper we introduce PROMIS-line which is a continuous tableting line built at the University of Eastern Finland, School of Pharmacy, PROMIS-center. PROMIS-line is modular and tablets can be produced via dry granulation or direct compression. In three case studies, continuous feeding, blending and tablet performance is studied to illustrate some basic features of PROMIS-line. In conclusion, the PROMIS-line is an excellent tool for studying the fundamentals of continuous manufg. of tablets.
- 194Van Snick, B.; Holman, J.; Cunningham, C.; Kumar, A.; Vercruysse, J.; De Beer, T.; Remon, J. P.; Vervaet, C. Continuous Direct Compression as Manufacturing Platform for Sustained Release Tablets. Int. J. Pharm. 2017, 519 (1–2), 390– 407, DOI: 10.1016/j.ijpharm.2017.01.010[Crossref], [PubMed], [CAS], Google Scholar194https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXitVykt7o%253D&md5=ab99bbf0a54fadea1669cce1667ea36aContinuous direct compression as manufacturing platform for sustained release tabletsVan Snick, B.; Holman, J.; Cunningham, C.; Kumar, A.; Vercruysse, J.; De Beer, T.; Remon, J. P.; Vervaet, C.International Journal of Pharmaceutics (Amsterdam, Netherlands) (2017), 519 (1-2), 390-407CODEN: IJPHDE; ISSN:0378-5173. (Elsevier B.V.)This study presents a framework for process and product development on a continuous direct compression manufg. platform. A challenging sustained release formulation with high content of a poorly flowing low d. drug was selected. Two HPMC grades were evaluated as matrix former: std. Methocel CR and directly compressible Methocel DC2. The feeding behavior of each formulation component was investigated by deriving feed factor profiles. The max. feed factor was used to est. the drive command and depended strongly upon the d. of the material. Furthermore, the shape of the feed factor profile allowed definition of a customized refill regime for each material. Inline NIRs was used to est. the residence time distribution (RTD) in the mixer and monitor blend uniformity. Tablet content and wt. variability were detd. as addnl. measures of mixing performance. For Methocel CR, the best axial mixing (i.e. feeder fluctuation dampening) was achieved when an impeller with high no. of radial mixing blades operated at low speed. However, the variability in tablet wt. and content uniformity deteriorated under this condition. One can therefore conclude that balancing axial mixing with tablet quality is crit. for Methocel CR. However, reformulating with the direct compressible Methocel DC2 as matrix former improved tablet quality vastly. Furthermore, both process and product were significantly more robust to changes in process and design variables. This observation underpins the importance of flowability during continuous blending and die-filling. At the compaction stage, blends with Methocel CR showed better tabletability driven by a higher compressibility as the smaller CR particles have a higher bonding area. However, tablets of similar strength were achieved using Methocel DC2 by targeting equal porosity. Compaction pressure impacted tablet properties and dissoln. Hence controlling thickness during continuous manufg. of sustained release tablets was crucial to ensure reproducible dissoln.
- 195Azad, M. A.; Osorio, J. G.; Brancazio, D.; Hammersmith, G.; Klee, D. M.; Rapp, K.; Myerson, A. A Compact, Portable, Re-Configurable, and Automated System for on-Demand Pharmaceutical Tablet Manufacturing. Int. J. Pharm. 2018, 539 (1–2), 157– 164, DOI: 10.1016/j.ijpharm.2018.01.027[Crossref], [PubMed], [CAS], Google Scholar195https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitV2rs7Y%253D&md5=2ca3f94e802e29adbc277d527ad02a69A compact, portable, re-configurable, and automated system for on-demand pharmaceutical tablet manufacturingAzad, Mohammad A.; Osorio, Juan G.; Brancazio, David; Hammersmith, Gregory; Klee, David M.; Rapp, Kersten; Myerson, AllanInternational Journal of Pharmaceutics (Amsterdam, Netherlands) (2018), 539 (1-2), 157-164CODEN: IJPHDE; ISSN:0378-5173. (Elsevier B.V.)Due to the complex nature of the pharmaceutical supply chain, the industry faces several major challenges when it comes to ensuring an adequate supply of quality drug products. These challenges are not only the causes of supply chain disruptions and financial loss, but can also prevent underserved and remote areas from receiving life-saving drugs. As a preliminary demonstration to mitigate all these challenges, at MIT we have developed active pharmaceutical ingredients manufg. in a miniature platform. However, manufg. of final oral solid dosage as tablets from drug substances had not been demonstrated. In this study, a compact, portable, re-configurable, and automated tablet manufg. system, roughly the size of a North American household oven, [72.4 cm (length) × 53.3 cm (width) × 134.6 cm (height)] was designed, built and demonstrated. This miniature system is able to manuf. on-demand tablets from drug crystals on a scale of hundreds to thousands per day. Ibuprofen and Diazepam, each having different drug loading, were manufd. using this miniature system and meet U. S. Pharmacopeia stds. We foresee this flexible, miniature, plug-and-play pharmaceutical solids dosage manufg. system advancing on-demand ready-to-use pharmaceuticals enabling future treatment of human diseases at the point-of-care.
- 196Azad, M. A.; Osorio, J. G.; Wang, A.; Klee, D. M.; Eccles, M. E.; Grela, E.; Sloan, R.; Hammersmith, G.; Rapp, K.; Brancazio, D.; Myerson, A. S. On-Demand Manufacturing of Direct Compressible Tablets: Can Formulation Be Simplified?. Pharm. Res. 2019, 36 (12), 167, DOI: 10.1007/s11095-019-2716-2[Crossref], [PubMed], [CAS], Google Scholar196https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3MjhsFaguw%253D%253D&md5=71352946057fe207addfbcceb82df3a3On-Demand Manufacturing of Direct Compressible Tablets: Can Formulation Be Simplified?Azad Mohammad A; Osorio Juan G; Wang Allison; Klee David M; Eccles Mary E; Grela Erin; Sloan Rebecca; Hammersmith Gregory; Rapp Kersten; Brancazio David; Myerson Allan S; Azad Mohammad A; Osorio Juan G; Hammersmith GregoryPharmaceutical research (2019), 36 (12), 167 ISSN:.PURPOSE: Oral direct compressible tablets are the most frequently used drug products. Manufacturing of tablets requires design and development of formulations, which need a number of excipients. The choice of excipients depends on the concentration, manufacturability, stability, and bioavailability of the active pharmaceutical ingredients (APIs). At MIT, we developed a miniature platform for on-demand manufacturing of direct compressible tablets. This study investigated how formulations could be simplified to use a small number of excipients for a number of different API's in which long term stability is not required. METHOD: Direct compressible tablets of five pharmaceutical drugs, Diazepam, Diphenhydramine HCl, Doxycycline Monohydrate, Ibuprofen, and Ciprofloxacin HCl, with different drug loadings, were made using direct compression in an automated small scale system.. The critical quality attributes (CQA) of the tablets were assessed for the quality standards set by the United States Pharmacopeia (USP). RESULTS: This miniature system can manufacture tablets - on-demand from crystalline API using the minimum number of excipients required for drug product performance. All drug tablets met USP quality standards after manufacturing and after 2 weeks of accelerated stability test, except for slightly lower drug release for Ibuprofen. CONCLUSIONS: On-demand tablets manufacturing where there is no need for long term stability using a flexible, miniature, automated (integrated) system will simplify pharmaceutical formulation design compared to traditional formulations. This advancement will offer substantial economic benefits by decreasing product time-to-market and enhancing quality.
- 197Asachi, M.; Nourafkan, E.; Hassanpour, A. A Review of Current Techniques for the Evaluation of Powder Mixing. Adv. Powder Technol. 2018, 29 (7), 1525– 1549, DOI: 10.1016/j.apt.2018.03.031[Crossref], [CAS], Google Scholar197https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXotFyrsrs%253D&md5=d3a0560a65f43607ded49bdd876f4444A review of current techniques for the evaluation of powder mixingAsachi, Maryam; Nourafkan, Ehsan; Hassanpour, AliAdvanced Powder Technology (2018), 29 (7), 1525-1549CODEN: APTEEE; ISSN:0921-8831. (Elsevier B.V.)Blending a mixt. of powders to a homogeneous system is a crucial step in many manufg. processes. To achieve a high quality of the end product, powder mixts. should be made with high content uniformity. For instance, producing uniform tablets depends on the homogeneous dispersion of active pharmaceutical ingredient (API), often in low level quantities, into excipients. To control the uniformity of a powder mixt., the first required step is to est. the powder content information during blending. There are several powder homogeneity evaluation techniques which differ in accuracy, fundamental basis, cost and operating conditions. In this article, emerging techniques for the anal. of powder content and powder blend uniformity, are explained and compared. The advantages and drawbacks of all the techniques are reviewed to help the readers to select the appropriate equipment for the powder mixing evaluation. In addn., the paper highlights the recent innovative online measurement techniques used for the non-invasive evaluation of the mixing performance.
- 198Nagy, B.; Farkas, A.; Borbás, E.; Vass, P.; Nagy, Z. K.; Marosi, G. Raman Spectroscopy for Process Analytical Technologies of Pharmaceutical Secondary Manufacturing. AAPS PharmSciTech 2019, 20 (1), 1, DOI: 10.1208/s12249-018-1201-2[Crossref], [CAS], Google Scholar198https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisFCiur%252FN&md5=76f77ebfb86b039da3bfa9f1d4be7f4bRaman Spectroscopy for Process Analytical Technologies of Pharmaceutical Secondary ManufacturingNagy, Brigitta; Farkas, Attila; Borbas, Eniko; Vass, Panna; Nagy, Zsombor Kristof; Marosi, GyorgyAAPS PharmSciTech (2019), 20 (1), 1-16CODEN: AAPHFZ; ISSN:1530-9932. (Springer)As the process anal. technol. (PAT) mindset is progressively introduced and adopted by the pharmaceutical companies, there is an increasing demand for effective and versatile real-time analyzers to address the quality assurance challenges of drug manufg. In the last decades, Raman spectroscopy has emerged as one of the most promising tools for non-destructive and fast characterization of the pharmaceutical processes. This review summarizes the achieved results of the real-time application of Raman spectroscopy in the field of the secondary manufg. of pharmaceutical solid dosage forms, covering the most common secondary process steps of a tablet prodn. line. In addn., the feasibility of Raman spectroscopy for real-time control is critically reviewed, and challenges and possible approaches to moving from real-time monitoring to process anal. controlled technologies (PACT) are discussed.
- 199Bowler, A. L.; Bakalis, S.; Watson, N. J. A Review of In-Line and on-Line Measurement Techniques to Monitor Industrial Mixing Processes. Chem. Eng. Res. Des. 2020, 153, 463– 495, DOI: 10.1016/j.cherd.2019.10.045[Crossref], [CAS], Google Scholar199https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitFGgu7zN&md5=585f32307ae592ca7860e57ac4a3d338A review of in-line and on-line measurement techniques to monitor industrial mixing processesBowler, Alexander Lewis; Bakalis, Serafim; Watson, Nicholas JamesChemical Engineering Research and Design (2020), 153 (), 463-495CODEN: CERDEE; ISSN:1744-3563. (Elsevier B.V.)Mixing is a ubiquitous operation in process engineering. It is not only used for combining materials, but also for promoting heat and mass transfer, increasing aeration, suspending solids, and modifying material structure. Measurement techniques have the potential to optimize industrial mixing processes and improve product quality by monitoring crit. process parameters. Real-time sensing techniques that do not require manual material sampling are of particular interest owing to their automatic data acquisition capability. This makes them suitable for use in control systems for process automation and eventually as connected sensors in Industry 4.0. This review article focuses on these measurement techniques, defined as in- and online, along with their capability for implementation in industrial processes. The applications reviewed include liq.-liq., gas-liq., solid-liq., solid-gas-liq., in addn. to solids blending. A technique selection section discussing the decision-making process when choosing a sensor and a summary table including the advantages, disadvantages, applications and limitations of each technique are provided. The article concludes by discussing the future of monitoring techniques for mixing processes.
- 200Crouter, A.; Briens, L. Methods to Assess Mixing of Pharmaceutical Powders. AAPS PharmSciTech 2019, 20 (2), 84, DOI: 10.1208/s12249-018-1286-7[Crossref], [PubMed], [CAS], Google Scholar200https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtFalsbvL&md5=2b008782dd512d1ff31c9f62d588f8d6Methods to Assess Mixing of Pharmaceutical PowdersCrouter, Allison; Briens, LaurenAAPS PharmSciTech (2019), 20 (2), 84CODEN: AAPHFZ; ISSN:1530-9932. (Springer)The pharmaceutical manufg. process consists of several steps, each of which must be monitored and controlled to ensure quality stds. are met. The level of blending has an impact on the final product quality; therefore, it is important to be able to monitor blending progress and identify an end-point. Currently, the pharmaceutical industry assesses blend content and uniformity through the extn. of samples using thief probes followed by anal. methods, such as spectroscopy, to det. the sample compn. The development of process anal. technologies (PAT) can improve product monitoring with the aim of increasing efficiency, product quality and consistency, and creating a better understanding of the manufg. process. Ideally, these are inline methods to remove issues related to extractive sampling and allow direct monitoring of the system using various sensors. Many technologies have been investigated, including spectroscopic techniques such as near-IR spectroscopy, velocimetric techniques that may use tracers, tomog. techniques, and acoustic emissions monitoring. While some techniques have demonstrated potential, many have significant disadvantages including the need for equipment modification, specific requirements of the material, expensive equipment, extensive anal., the location of the probes may be crit. and/or invasive, and lastly, the technique may only be applicable to the development phase. Both the advantages and disadvantages of the technologies should be considered in application to a specific system.
- 201Nagy, B.; Farkas, A.; Gyürkés, M.; Komaromy-Hiller, S.; Démuth, B.; Szabó, B.; Nusser, D.; Borbás, E.; Marosi, G.; Nagy, Z. K. In-Line Raman Spectroscopic Monitoring and Feedback Control of a Continuous Twin-Screw Pharmaceutical Powder Blending and Tableting Process. Int. J. Pharm. 2017, 530 (1–2), 21– 29, DOI: 10.1016/j.ijpharm.2017.07.041[Crossref], [PubMed], [CAS], Google Scholar201https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXht1Clsr7O&md5=25725d6ab2a0210fa1c2ea6077f7f073In-line Raman spectroscopic monitoring and feedback control of a continuous twin-screw pharmaceutical powder blending and tableting processNagy, Brigitta; Farkas, Attila; Gyurkes, Martin; Komaromy-Hiller, Szofia; Demuth, Balazs; Szabo, Bence; Nusser, David; Borbas, Eniko; Marosi, Gyorgy; Nagy, Zsombor KristofInternational Journal of Pharmaceutics (Amsterdam, Netherlands) (2017), 530 (1-2), 21-29CODEN: IJPHDE; ISSN:0378-5173. (Elsevier B.V.)The integration of Process Anal. Technol. (PAT) initiative into the continuous prodn. of pharmaceuticals is indispensable for reliable prodn. The present paper reports the implementation of in-line Raman spectroscopy in a continuous blending and tableting process of a three-component model pharmaceutical system, contg. caffeine as model active pharmaceutical ingredient (API), glucose as model excipient and magnesium stearate as lubricant. The real-time anal. of API content, blend homogeneity, and tablet content uniformity was performed using a Partial Least Squares (PLS) quant. method. The in-line Raman spectroscopic monitoring showed that the continuous blender was capable of producing blends with high homogeneity, and technol. malfunctions can be detected by the proposed PAT method. The Raman spectroscopy-based feedback control of the API feeder was also established, creating a 'Process Anal. Controlled Technol.' (PACT), which guarantees the required API content in the produced blend. This is, to the best of the authors' knowledge, the first ever application of Raman-spectroscopy in continuous blending and the first Raman-based feedback control in the formulation technol. of solid pharmaceuticals.
- 202Palmer, J.; Reynolds, G. K.; Tahir, F.; Yadav, I. K.; Meehan, E.; Holman, J.; Bajwa, G. Mapping Key Process Parameters to the Performance of a Continuous Dry Powder Blender in a Continuous Direct Compression System. Powder Technol. 2020, 362, 659– 670, DOI: 10.1016/j.powtec.2019.12.028[Crossref], [CAS], Google Scholar202https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtlSktw%253D%253D&md5=48b425e617f5f971b7bca70726b33b42Mapping key process parameters to the performance of a continuous dry powder blender in a continuous direct compression systemPalmer, John; Reynolds, Gavin K.; Tahir, Furqan; Yadav, Indrajeetsinh K.; Meehan, Elizabeth; Holman, James; Bajwa, GurjitPowder Technology (2020), 362 (), 659-670CODEN: POTEBX; ISSN:0032-5910. (Elsevier B.V.)This work aims to expand the typical raw material attributes that can successfully be processed on a continuous direct compression line with a particular focus on the continuous dry powder blender. Three grades of Acetaminophen were investigated as model active pharmaceutical ingredients and chosen to span a broad range of material attributes wider than what would normally be considered for a direct compression process. An exptl. design was set-up for each grade of Acetaminophen to investigate the effect of throughput, impeller speed and impeller configuration on the crit. process responses and attributes. The strain experienced by the in-process material was found crit. to improve content uniformity. The impeller speed and design can be optimized at a given throughput to obtain a strain which would deliver the required content homogeneity. The content uniformity of the final tablets can be modelled as a function of the strain using an exponential decay model.
- 203Fonteyne, M.; Vercruysse, J.; De Leersnyder, F.; Van Snick, B.; Vervaet, C.; Remon, J. P.; De Beer, T. Process Analytical Technology for Continuous Manufacturing of Solid-Dosage Forms. TrAC, Trends Anal. Chem. 2015, 67, 159– 166, DOI: 10.1016/j.trac.2015.01.011[Crossref], [CAS], Google Scholar203https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXktVCjs7k%253D&md5=dbccb235b113f2f91a8950396f97e75cProcess Analytical Technology for continuous manufacturing of solid-dosage formsFonteyne, Margot; Vercruysse, Jurgen; De Leersnyder, Fien; Van Snick, Bernd; Vervaet, Chris; Remon, Jean Paul; De Beer, ThomasTrAC, Trends in Analytical Chemistry (2015), 67 (), 159-166CODEN: TTAEDJ; ISSN:0165-9936. (Elsevier B. V.)A review. Currently, pharmaceutical prodn. is making the switch from batch processing towards continuous processing. The quality of intermediate and end products produced by batch processes is assured by off-line testing. It is obvious that off-line tests in anal. labs. cancel out the advantages of continuous processing, so the crit. quality attributes of continuously produced pharmaceuticals need to be monitored in real time. In 2004, the US Food and Drug Administration launched the process anal. technol. (PAT) concept to stimulate the pharmaceutical industry to change from off-line to real-time quality testing. This review explores the implementation of PAT tools within continuous pharmaceutical processes (i.e., blending, spray drying, roller compaction, twin-screw granulation and compression), focusing on both opportunities and challenges.
- 204Fonteyne, M.; Vercruysse, J.; De Leersnyder, F.; Besseling, R.; Gerich, A.; Oostra, W.; Remon, J. P.; Vervaet, C.; De Beer, T. Blend Uniformity Evaluation during Continuous Mixing in a Twin Screw Granulator by In-Line NIR Using a Moving F-Test. Anal. Chim. Acta 2016, 935, 213– 223, DOI: 10.1016/j.aca.2016.07.020[Crossref], [PubMed], [CAS], Google Scholar204https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xht1Gjur7O&md5=21c9054c38d5ced7a2bc988704ea6aabBlend uniformity evaluation during continuous mixing in a twin screw granulator by in-line NIR using a moving F-testFonteyne, Margot; Vercruysse, Jurgen; De Leersnyder, Fien; Besseling, Rut; Gerich, Ad; Oostra, Wim; Remon, Jean Paul; Vervaet, Chris; De Beer, ThomasAnalytica Chimica Acta (2016), 935 (), 213-223CODEN: ACACAM; ISSN:0003-2670. (Elsevier B.V.)This study focuses on the twin screw granulator of a continuous from-powder-to-tablet prodn. line. Whereas powder dosing into the granulation unit is possible from a container of preblended material, a truly continuous process uses several feeders (each one dosing an individual ingredient) and relies on a continuous blending step prior to granulation. The aim of the current study was to investigate the in-line blending capacity of this twin screw granulator, equipped with conveying elements only. The feasibility of in-line NIR (SentroPAT, Sentronic GmbH, Dresden, Germany) spectroscopy for evaluating the blend uniformity of powders after the granulator was tested. Anhyd. theophylline was used as a tracer mol. and was blended with lactose monohydrate. Theophylline and lactose were both fed from a different feeder into the twin screw granulator barrel. Both homogeneous mixts. and mixing expts. with induced errors were investigated. The in-line spectroscopic analyses showed that the twin screw granulator is a useful tool for in-line blending in different conditions. The blend homogeneity was evaluated by means of a novel statistical method being the moving F-test method in which the variance between two blocks of collected NIR spectra is evaluated. The α- and β-error of the moving F-test are controlled by using the appropriate block size of spectra. The moving F-test method showed to be an appropriate calibration and maintenance free method for blend homogeneity evaluation during continuous mixing.
- 205Vanarase, A. U.; Alcalà, M.; Jerez Rozo, J. I.; Muzzio, F. J.; Romañach, R. J. Real-Time Monitoring of Drug Concentration in a Continuous Powder Mixing Process Using NIR Spectroscopy. Chem. Eng. Sci. 2010, 65 (21), 5728– 5733, DOI: 10.1016/j.ces.2010.01.036[Crossref], [CAS], Google Scholar205https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXht1Kqur%252FO&md5=f1ad827bf12402f7c0ae3991b46c20eeReal-time monitoring of drug concentration in a continuous powder mixing process using NIR spectroscopyVanarase, Aditya U.; Alcala, Manel; Jerez Rozo, Jackeline I.; Muzzio, Fernando J.; Romanach, Rodolfo J.Chemical Engineering Science (2010), 65 (21), 5728-5733CODEN: CESCAC; ISSN:0009-2509. (Elsevier Ltd.)A non-destructive NIR spectroscopic method was used to acquire online spectra of a continuous mixing process, and evaluate the performance of this novel system. Partial least squares (PLS) calibration models were developed and used for real-time detn. of active ingredient concn. on the blends produced with a continuous mixer. The NIR method was developed for concns. ranging from 0 to 15% (wt./wt.) of acetaminophen (APAP), the active pharmaceutical ingredient used in the expts. The calibration model's overall accuracy was 0.41% (wt./wt.), and estd. through the root mean square error of cross validation (RMSECV) for samples predicted using leave-class-out cross validation. In this cross validation, each concn. was defined as a class, and when a sample of a specific concn. was predicted all samples of that concn. were left out of the calibration model. The precision of the calibration model was also estd. at various concn. levels, to facilitate the differentiation between the variation in drug concn. due to the anal. method's measurement uncertainty and the variation in the drug distribution throughout the powder blend. The results obtained are very promising since in 3 of the 5 powder mixes, the variation in the drug concn. in the powder blends was similar to that of the anal. method indicating a high degree of blend homogeneity.
- 206Sierra-Vega, N. O.; Román-Ospino, A.; Scicolone, J.; Muzzio, F. J.; Romañach, R. J.; Méndez, R. Assessment of Blend Uniformity in a Continuous Tablet Manufacturing Process. Int. J. Pharm. 2019, 560, 322– 333, DOI: 10.1016/j.ijpharm.2019.01.073[Crossref], [PubMed], [CAS], Google Scholar206https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXjslensL0%253D&md5=3cbb0712bc25d7471b375719c48fb2c9Assessment of blend uniformity in a continuous tablet manufacturing processSierra-Vega, Nobel O.; Roman-Ospino, Andres; Scicolone, James; Muzzio, Fernando J.; Romanach, Rodolfo J.; Mendez, RafaelInternational Journal of Pharmaceutics (Amsterdam, Netherlands) (2019), 560 (), 322-333CODEN: IJPHDE; ISSN:0378-5173. (Elsevier B.V.)Blend uniformity was monitored throughout a continuous manufg. (CM) process by near IR (NIR) spectroscopic measurements of flowing blends and compared to the drug concn. in the tablets. The NIR spectra were obtained through the chute after the blender and within the feed frame, while transmission spectra were obtained for the tablets. The CM process was performed with semi-fine acetaminophen blends at 10.0% (wt./wt.). The blender was operated at 250 RPM, for best performance, and 106 and 495 rpm where a lower mixing efficiency was expected. The variation in blender RPM increased the variation in drug concn. at the chute but not at the feed frame. Statistical results show that the drug concn. of tablets can be predicted, with great accuracy, from blends within the feed frame. This study demonstrated a mixing effect within the feed frame, which contribute to a 60% decrease in the relative std. deviation of the drug concn., when compared to the chute. Variog. anal. showed that the min. sampling and anal. error was five times less in the feed frame than the chute. This study demonstrates that the feed frame is an ideal location for monitoring the drug concn. of powder blends for CM processes.
- 207Van Snick, B.; Holman, J.; Vanhoorne, V.; Kumar, A.; De Beer, T.; Remon, J. P.; Vervaet, C. Development of a Continuous Direct Compression Platform for Low-Dose Drug Products. Int. J. Pharm. 2017, 529 (1–2), 329– 346, DOI: 10.1016/j.ijpharm.2017.07.003[Crossref], [PubMed], [CAS], Google Scholar207https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXht1GhsrrI&md5=b545563f8ee29f2980f19c08c8f657a1Development of a continuous direct compression platform for low-dose drug productsVan Snick, B.; Holman, J.; Vanhoorne, V.; Kumar, A.; De Beer, T.; Remon, J. P.; Vervaet, C.International Journal of Pharmaceutics (Amsterdam, Netherlands) (2017), 529 (1-2), 329-346CODEN: IJPHDE; ISSN:0378-5173. (Elsevier B.V.)In this work a continuous direct compression process was developed for a low-dosed drug product. Each unit operation of the GEA CDC-50 system was thoroughly investigated. This paper aimed to tackle the macroscopic and microscopic blend uniformity challenges inherently assocd. with continuous direct compression of cohesive and agglomerated APIs formulated at low dose. D., compressibility and flow were identified as key material properties at the feeding stage. The screw speed coupled with powder flow regulated the gravimetric feeding performance. The impact of process and design variables was elucidated at the blending stage. The impeller configuration (no. and pattern of radial mixing blades) and speed were key variables to steer the residence time distribution at the blending stage. An impeller configuration with distributed radial mixing blades could sufficiently filter the steady state feeding variability at low mixer speed, but exerted limited strain and shear on the blend. Hence micro-agglomerates persisted through the blending process and occasionally resulted in super potent tablets. Therefore, a new configuration was evaluated with more radial mixing blades centered on the impeller. This configuration resulted in a long mixing time at high tip speed which induced a maximized strain and shear. Consequently, excellent uniformity of the blend and tablets at macroscopic and microscopic level was achieved. Besides, this impeller improved robustness towards feeding disturbances, changes in process settings and variable blend properties. Next, it was demonstrated that the lubrication step requires crit. attention during the design of the equipment, formulation and process. This study provided abundant evidence that an optimized continuous direct compression process allows direct compression of challenging low-dose drug products.
- 208Grymonpré, W.; Blahova Prudilova, B.; Vanhoorne, V.; Van Snick, B.; Detobel, F.; Remon, J. P.; De Beer, T.; Vervaet, C. Downscaling of the Tableting Process: Feasibility of Miniaturized Forced Feeders on a High-Speed Rotary Tablet Press. Int. J. Pharm. 2018, 550 (1–2), 477– 485, DOI: 10.1016/j.ijpharm.2018.09.006[Crossref], [PubMed], [CAS], Google Scholar208https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhs12hs7vP&md5=eb4c6c8f972e20f97b2f8c0f79876f5eDownscaling of the tableting process: Feasibility of miniaturized forced feeders on a high-speed rotary tablet pressGrymonpre, W.; Blahova Prudilova, B.; Vanhoorne, V.; Van Snick, B.; Detobel, F.; Remon, J. P.; De Beer, T.; Vervaet, C.International Journal of Pharmaceutics (Amsterdam, Netherlands) (2018), 550 (1-2), 477-485CODEN: IJPHDE; ISSN:0378-5173. (Elsevier B.V.)With the current transformation of the pharmaceutical industry towards continuous manufg., there is an inherent need to embrace this concept already during the early stages of drug formulation. Therefore, this research paper investigated the feasibility of using miniaturized forced feeders on a high-speed rotary tablet press with the intention of downscaling the tableting process. Forced feeders with a reduced vol. (up to 46% compared to the conventional two-compartment forced feeder) were designed by either sealing one compartment (i.e. R&D1) or lowering of the compartment height (i.e. R&D2). These feed frame designs were thoroughly analyzed in combination with two paddle types over a wide range of process-settings (i.e. tableting speed, paddle speed, direction of paddle rotation, overfill-level). A poorly flowing model powder (i.e. MCC 101) was deliberately selected as challenging formulation. Empirical modeling of feed frame R&D1 revealed a pos. impact on the die-filling variability when the radial curved cuboid paddles rotated in counterclockwise direction at high paddle speed. Moreover, a strong resemblance between the R&D2 feed frame and the conventional forced feeder was obsd. during multivariate data anal., indicating that this miniaturized type could be used during downscaling studies of the conventional tableting process. The potential of this forced feeder was acknowledged by the similar trends in die-filling variability with respect to varying process settings, when a design-of-expts. (DOE) was performing including feed frame type as a qual. factor. Overall, it was concluded that both types of miniaturized forced feeders can be used on a high-speed rotary tablet press when lower material consumption rates are desired while the R&D2 feed frame bears the highest predictability regarding the die-filling uniformity in the conventional larger two-compartment forced feeder.
- 209Matsunami, K.; Nagato, T.; Hasegawa, K.; Sugiyama, H. A Large-Scale Experimental Comparison of Batch and Continuous Technologies in Pharmaceutical Tablet Manufacturing Using Ethenzamide. Int. J. Pharm. 2019, 559, 210– 219, DOI: 10.1016/j.ijpharm.2019.01.028[Crossref], [PubMed], [CAS], Google Scholar209https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitlGhur8%253D&md5=4a649d3a718c93eadfe97a0812ff6121A large-scale experimental comparison of batch and continuous technologies in pharmaceutical tablet manufacturing using ethenzamideMatsunami, Kensaku; Nagato, Takuya; Hasegawa, Koji; Sugiyama, HirokazuInternational Journal of Pharmaceutics (Amsterdam, Netherlands) (2019), 559 (), 210-219CODEN: IJPHDE; ISSN:0378-5173. (Elsevier B.V.)This paper compares batch and continuous technologies in terms of product quality and process performance in pharmaceutical tablet manufg. using ethenzamide as the active pharmaceutical ingredient. Batch and continuous processes using wet granulation were investigated by performing expts. on the scale of 5 and up to 100 kg/lot, using the same raw materials. Three technologies were tested and compared: (i) batch technol. using fluidized bed granulation, (ii) batch technol. using high shear granulation, (iii) continuous technol. using high shear granulation. In the full-scale expt., in all three technologies including continuous technol., the quality of the tablets fulfilled the target values regarding hardness, active pharmaceutical ingredient content, and dissoln. The granules produced by different technologies, however, presented varying attributes regarding granule size distribution, loose bulk d., or scanning electron microscope images. The process performance, more specifically the yield, was slightly better for batch technologies than for the continuous technol., mainly due to losses in the start-up operation. Notably, this study has shown that continuous technol., which is generally believed to not entail scale-up procedures, could in fact, require parameter adjustment for prolonged operation. The results provided suggestions for improvements to implement large-scale continuous technologies in the pharmaceutical industry.
- 210Järvinen, K.; Hoehe, W.; Järvinen, M.; Poutiainen, S.; Juuti, M.; Borchert, S. In-Line Monitoring of the Drug Content of Powder Mixtures and Tablets by near-Infrared Spectroscopy during the Continuous Direct Compression Tableting Process. Eur. J. Pharm. Sci. 2013, 48 (4–5), 680– 688, DOI: 10.1016/j.ejps.2012.12.032[Crossref], [PubMed], [CAS], Google Scholar210https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXkt12rur8%253D&md5=95b5c255b993adc3b61157c868b7bdccIn-line monitoring of the drug content of powder mixtures and tablets by near-infrared spectroscopy during the continuous direct compression tableting processJarvinen, Kristiina; Hoehe, Wolfgang; Jarvinen, Maiju; Poutiainen, Sami; Juuti, Mikko; Borchert, SvenEuropean Journal of Pharmaceutical Sciences (2013), 48 (4-5), 680-688CODEN: EPSCED; ISSN:0928-0987. (Elsevier B.V.)Continuous manufg. methods offer economic and quality advantages when compared with batch manufg. methods. In continuous manufg., one requires real time assurance of quality of product via the implementation of PAT tools. This study focuses on an in-line near-IR (NIR) spectroscopic method for detg. the drug content of powder mixts. and tablets during a continuous tableting process. Tablets consisting of acetaminophen (20-30%), lactose (69.07-78.93%) and magnesium stearate (0.93-1.07%) were prepd. in a continuous direct compression line that consisted of 2 loss-in-wt. feeders, one for acetaminophen and one for premixed lactose and magnesium stearate, and a continuous mixer followed by a rotary tablet press. NIR spectroscopy was applied to the continuous mixer and tablet press to perform a 100% product check at full tableting speed. The UV-spectrophotometric method was used as an off-line ref. method to det. the acetaminophen content in the samples. The powder mixt. and tablet samples were taken during the process for the calibration of continuous mixer and tablet press, resp. For the continuous mixer, model creation with the PLS method yielded R-Square and RMSEC (root mean square error of calibration) values of 0.975% and 0.56%, resp. For the tablet press, the corresponding R-Square and RMSEC values were 0.943% and 0.75%, resp. A test run demonstrated good predictability in the estn. of the API content in the powder mixts. and tablets during the continuous tableting process. For the continuous mixer and tablet press, the RMSEP (root mean square error of prediction) values were 0.96% and 1.37%, resp. This study demonstrates that an NIR instrument capable of fast spectra acquisition can be a valuable tool for the in-line monitoring of the continuous mixing and tableting processes.
- 211Nagy, Z. K.; Balogh, A.; Vajna, B.; Farkas, A.; Patyi, G.; Kramarics, Á.; Marosi, G. Comparison of Electrospun and Extruded Soluplus®-Based Solid Dosage Forms of Improved Dissolution. J. Pharm. Sci. 2012, 101 (1), 322– 332, DOI: 10.1002/jps.22731[Crossref], [PubMed], [CAS], Google Scholar211https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtVKktr%252FI&md5=b8352b5512c9bd78b9927eb9c26950a8Comparison of electrospun and extruded Soluplus-based solid dosage forms of improved dissolutionNagy, Zsombor K.; Balogh, Attila; Vajna, Balazs; Farkas, Attila; Patyi, Gergo; Kramarics, Aron; Marosi, GyoergyJournal of Pharmaceutical Sciences (2012), 101 (1), 322-332CODEN: JPMSAE; ISSN:0022-3549. (Wiley-Liss, Inc.)Electrospinning (ES) and extrusion of a poorly water-sol. active pharmaceutical ingredient were used to improve its dissoln., which is a major challenge in the field of pharmaceutical technol. Spironolactone was applied as model drug and recently developed polyvinyl caprolactam-polyvinyl acetate-polyethylene glycol graft copolymer (Soluplus) was used as carrier matrix and solubilizer. ES of the polymer matrix from ethanol soln. was optimized at first without spironolactone and then the cosoln. of the drug and the carrier was used for forming electrospun fibers. It resulted in real solid soln. due to its very efficient amorphization effect. On the contrary, a low amt. of cryst. spironolactone appeared in the extrudates according to Raman microscopy, X-ray diffraction (XRD), SEM and energy-dispersive spectrometry (EDS). Raman microspectrometry had the lowest detection limit of spironolactone crystals compared with XRD and differential scanning calorimetry. Both ES and extrusion techniques resulted in significantly improved dissoln. Electrospun ultrafine fibers increased the dissoln. more effectively, owing to the formed solid soln. and huge surface. The developed continuous technologies demonstrate great potential to tackle the challenge of inadequate dissoln. of poorly water-sol. drugs in several cases. © 2011 Wiley-Liss, Inc. and the American Pharmacists Assocn. J Pharm Sci.
- 212Szabo, E.; Demuth, B.; Nagy, B.; Molnar, K.; Farkas, A.; Szabo, B.; Balogh, A.; Hirsch, E.; Nagy, B.; Marosi, G.; Nagy, Z. K. Scaled-up Preparation of Drug-Loaded Electrospun Polymer Fibres and Investigation of Their Continuous Processing to Tablet Form. eXPRESS Polym. Lett. 2018, 12 (5), 436– 451, DOI: 10.3144/expresspolymlett.2018.37[Crossref], [CAS], Google Scholar212https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisVCltLfN&md5=ee0d3f0b4eb5a03e0053778bb2703634Scaled-up preparation of drug-loaded electrospun polymer fibres and investigation of their continuous processing to tablet formSzabo, E.; Demuth, B.; Nagy, B.; Molnar, K.; Farkas, A.; Szabo, B.; Balogh, A.; Hirsch, E.; Marosi, G.; Nagy, Z. K.eXPRESS Polymer Letters (2018), 12 (5), 436-451CODEN: PLOEAK; ISSN:1788-618X. (Budapest University of Technology and Economics, Dep. of Polymer Engineering)Polymer-based electrospun amorphous solid dispersions (ASDs) were prepd. and investigated from pharmaceutical application point of view. Spironolactone (SPIR) was used as model drug mixed in various concns. with polymers suitable for fiber formation, such as vinylpyrrolidone-vinyl acetate copolymer, polyvinylpyrrolidone K30 and hydroxypropyl methylcellulose. Single needle electrospinning was applied at first for screening the compn. of the prepd. ASDs. Scaling-up the selected polymer-drug combination was accomplished by high speed electrospinning, the productivity of which enabled investigation of downstream processing to generate tablet formulation. The steps of a potential continuous prodn. line (fiber collection, grinding, feeding and tableting) proved to be feasible with the electrospun ASD without any sign of crystn. If cryst. drug was added into the ASD contg. tablets as impurity strictly monotonous decrease of drug dissoln. was obsd. in the function of the cryst. drug content. The capabilities of the non-destructive Raman and near-IR spectroscopies, as fast quality assurance tools, were compared to each other in quantifying of cryst. SPIR content in the prepd. tablets.
- 213Szabó, E.; Démuth, B.; Galata, D. L.; Vass, P.; Hirsch, E.; Csontos, I.; Marosi, G.; Nagy, Z. K. Continuous Formulation Approaches of Amorphous Solid Dispersions: Significance of Powder Flow Properties and Feeding Performance. Pharmaceutics 2019, 11 (12), 654, DOI: 10.3390/pharmaceutics11120654[Crossref], [CAS], Google Scholar213https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXpt12qtLs%253D&md5=ca19689d10d148416358d0e34ba90a67Continuous formulation approaches of amorphous solid dispersions: significance of powder flow properties and feeding performanceSzabo, Edina; Demuth, Balazs; Galata, Dorian Laszlo; Vass, Panna; Hirsch, Edit; Csontos, Istvan; Marosi, Gyorgy; Nagy, Zsombor K.Pharmaceutics (2019), 11 (12), 654CODEN: PHARK5; ISSN:1999-4923. (MDPI AG)Prepn. and formulation of amorphous solid dispersions (ASDs) are becoming more and more popular in the pharmaceutical field because the dissoln. of poorly water-sol. drugs can be effectively improved this way, which can lead to increased bioavailability in many cases. During downstream processing of ASDs, technologists need to keep in mind both traditional challenges and the newest trends. In the last decade, the pharmaceutical industry began to display considerable interest in continuous processing, which can be explained with their potential advantages such as smaller footprint, easier scale-up, and more consistent product, better quality and quality assurance. Continuous downstream processing of drug-loaded ASDs opens new ways for automatic operation. Therefore, the formulation of poorly water-sol. drugs may be more effective and safe. However, developments can be challenging due to the poor flowability and feeding properties of ASDs. Consequently, this review pays special attention to these characteristics since the feeding of the components greatly influences the content uniformity in the final dosage form. The main purpose of this paper is to summarize the most important steps of the possible ASD-based continuous downstream processes in order to give a clear overview of current course lines and future perspectives.
- 214Kalivoda, A.; Fischbach, M.; Kleinebudde, P. Application of Mixtures of Polymeric Carriers for Dissolution Enhancement of Fenofibrate Using Hot-Melt Extrusion. Int. J. Pharm. 2012, 429 (1–2), 58– 68, DOI: 10.1016/j.ijpharm.2012.03.009[Crossref], [PubMed], [CAS], Google Scholar214https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XkslWiu7c%253D&md5=92fd6522310b5fbc73bfae3f259c0cb6Application of mixtures of polymeric carriers for dissolution enhancement of fenofibrate using hot-melt extrusionKalivoda, Adela; Fischbach, Matthias; Kleinebudde, PeterInternational Journal of Pharmaceutics (Amsterdam, Netherlands) (2012), 429 (1-2), 58-68CODEN: IJPHDE; ISSN:0378-5173. (Elsevier B.V.)Hot-melt extrusion was applied to improve dissoln. behavior of poorly sol. model drug fenofibrate. Blends of polymers were used as carrier: copovidone (COP), polyvinyl caprolactam-polyvinyl acetate-polyethylene glycol copolymer (PVCL-PVAc-PEG) and hypromellose 2910/5 (HPMC). The ratio of fenofibrate to COP remained constantly 1 + 3 (weighted parts) with varying amts. of PVCL-PVAc-PEG and HPMC. Solid state of fenofibrate was characterized by X-ray diffractometry and differential scanning calorimetry. Dissoln. performance was compared to marketed formulations Lipidil and Lipidil-Ter. Stability studies were conducted at 25 °C/60%rH. The dissoln. rate from extrudates was significantly increased when compared to pure fenofibrate powder or phys. mixt. of the components. A supersatn. of 7.6-12.1 was reached with the pelletized extrudates. All extrudates were superior to marketed formulations. No recrystn. was obsd. after 26 wk of storage for fenofibrate-COP extrudates 1 + 3 (weighted parts) with or without polymeric additives. Even so, both degree and duration of supersatn. decreased with increasing storage periods with the exception of fenofibrate-HPMC extrudates. Of particular interest is the finding that by adding polymers with differing release characteristics to the drug-carrier mixt., the dissoln. performance of hot-melt extruded solid dosage forms can be readily adapted to meet specific requirements.
- 215Gryczke, A.; Schminke, S.; Maniruzzaman, M.; Beck, J.; Douroumis, D. Development and Evaluation of Orally Disintegrating Tablets (ODTs) Containing Ibuprofen Granules Prepared by Hot Melt Extrusion. Colloids Surf., B 2011, 86 (2), 275– 284, DOI: 10.1016/j.colsurfb.2011.04.007[Crossref], [PubMed], [CAS], Google Scholar215https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXmvFWjurY%253D&md5=6523836db656149f0069c799c904b3f9Development and evaluation of orally disintegrating tablets (ODTs) containing Ibuprofen granules prepared by hot melt extrusionGryczke, Andreas; Schminke, Silke; Maniruzzaman, Mohammed; Beck, Julien; Douroumis, DennisColloids and Surfaces, B: Biointerfaces (2011), 86 (2), 275-284CODEN: CSBBEQ; ISSN:0927-7765. (Elsevier B.V.)In the current study ibuprofen was embedded in a methacrylate copolymer (Eudragit EPO) matrix to produce solid dispersions by hot-melt extrusion (HME) processing. The obtained granules were incorporated in orally disintegrating tablets (ODTs). The tablets were developed by varying the ratio of superdisintegrants such as sodium croscarmellose and crosslinked polyvinylpyrrolidone grades while a direct compression process was used to compress the ODTs under various compaction forces to optimize tablet robustness. The properties of the compressed tablets which included porosity, hardness, friability and dissoln. profiles were further evaluated and compared with Nurofen Meltlet ODTs. The taste and sensory evaluation in human volunteers demonstrated excellence in masking the bitter active and improved tablet palatability.
- 216Baronsky-Probst, J.; Möltgen, C.-V.; Kessler, W.; Kessler, R. W. Process Design and Control of a Twin Screw Hot Melt Extrusion for Continuous Pharmaceutical Tamper-Resistant Tablet Production. Eur. J. Pharm. Sci. 2016, 87, 14– 21, DOI: 10.1016/j.ejps.2015.09.010[Crossref], [PubMed], [CAS], Google Scholar216Process design and control of a twin screw hot melt extrusion for continuous pharmaceutical tamper-resistant tablet productionBaronsky-Probst, J.; Moeltgen, C.-V.; Kessler, W.; Kessler, R. W.European Journal of Pharmaceutical Sciences (2016), 87 (), 14-21CODEN: EPSCED; ISSN:0928-0987. (Elsevier B.V.)Hot melt extrusion (HME) is a well-known process within the plastic and food industries that has been utilized for the past several decades and is increasingly accepted by the pharmaceutical industry for continuous manufg. For tamper-resistant formulations of e.g. opioids, HME is the most efficient prodn. technique. The focus of this study is thus to evaluate the manufacturability of the HME process for tamper-resistant formulations. Parameters such as the specific mech. energy (SME), as well as the melt pressure and its std. deviation, are important and will be discussed in this study. In the first step, the existing process data are analyzed by means of multivariate data anal. Key crit. process parameters such as feed rate, screw speed, and the concn. of the API in the polymers are identified, and crit. quality parameters of the tablet are defined. In the second step, a relationship between the crit. material, product and process quality attributes are established by means of Design of Expts. (DoEs). The resulting SME and the temp. at the die are essential data points needed to indirectly qualify the degrdn. of the API, which should be minimal. NIR-spectroscopy is used to monitor the material during the extrusion process. In contrast to most applications in which the probe is directly integrated into the die, the optical sensor is integrated into the cooling line of the strands. This saves costs in the probe design and maintenance and increases the robustness of the chemometric models. Finally, a process measurement system is installed to monitor and control all of the crit. attributes in real-time by means of first princip