We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Integrated continuous manufacturing in pharmaceutical industry: current evolutionary steps toward revolutionary future

    Nada A Helal

    Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt

    ‡Authors contributed equally to this work

    Search for more papers by this author

    ,
    Ola Elnoweam

    Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt

    ‡Authors contributed equally to this work

    Search for more papers by this author

    ,
    Heba Abdullah Eassa

    Department of Pharmaceutics & Industrial Pharmacy, College of Pharmacy for Girls, Al Azhar University, Cairo 11651, Egypt

    Department of Pharmaceutical Sciences (DPS), School of Pharmacy & Physician Assistant Studies (SOPPAS), University of Saint Joseph (USJ), Hartford, CT 06103, USA

    ‡Authors contributed equally to this work

    Search for more papers by this author

    ,
    Ahmed M Amer

    Research & Development Department, Medizen Pharmaceutical Industries company, Alexandria 21521, Egypt

    ,
    Mohamed Ashraf Eltokhy

    Department of Microbiology, Faculty of Pharmacy, Misr International University, Cairo 11757, Egypt

    ,
    Mohamed A Helal

    Construction Planning Department, National Marine Dredging Company (NMDC), Abu Dhabi 11372, United Arab Emirates

    ,
    Heba A Fayyaz

    Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt

    &
    Mohamed Ismail Nounou

    *Author for correspondence:

    E-mail Address: nounou@usj.edu

    Department of Pharmaceutical Sciences (DPS), School of Pharmacy & Physician Assistant Studies (SOPPAS), University of Saint Joseph (USJ), Hartford, CT 06103, USA

    Published Online:https://doi.org/10.4155/ppa-2019-0011

    Continuous manufacturing (CM) has the potential to provide pharmaceutical products with better quality, improved yield and with reduced cost and time. Moreover, ease of scale-up, small manufacturing footprint and on-line/in-line monitoring and control of the process are other merits for CM. Regulating authorities are supporting the adoption of CM by pharmaceutical manufacturers through issuing proper guidelines. However, implementation of this technology in pharmaceutical industry is encountered by a number of challenges regarding the process development and quality assurance. This article provides a background on the implementation of CM in pharmaceutical industry, literature survey of the most recent state-of-the-art technologies and critically discussing the encountered challenges and its future prospective in pharmaceutical industry.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Gutmann B, Cantillo D, Kappe CO. Continuous-Flow technology—A tool for the safe manufacturing of active pharmaceutical ingredients. Angew. Chem. Int. Ed. Engl. 54(23), 6688–6728 (2015).
    • 2. US FDA. CFR- Code of Federal regualtion Title 21 (2018). www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfcfr/cfrsearch.cfm
    • 3. Lee SL, O'connor TF, Yang X et al. Modernizing pharmaceutical manufacturing: from batch to continuous production. J. Pharm. Innov. 10, 191–199 (2015).
    • 4. Ierapetritou M, Muzzio F, Reklaitis G. Perspectives on the continuous manufacturing of powder-based pharmaceutical processes. AIChE J. 62(6), 1846–1862 (2016).
    • 5. Getz KA, Kaitin KI. Open innovation: The new face of pharmaceutical research and development. Exp. Rev. Clin. Pharm. 5(5), 481–483 (2012).
    • 6. U.S. Food and Drug Adminstration (Fda). Q7 Good manufacturing practice guidance for active pharmaceutical ingredients (2018). www.fda.gov/regulatory-information/search-fda-guidance-documents/q7-good-manufacturing-practice-guidance-active-pharmaceutical-ingredients-guidance-industry
    • 7. Mascia S, Heider PL, Zhang H et al. End-to-end continuous manufacturing of pharmaceuticals: integrated synthesis, purification and final dosage formation. Angew. Chem. Int. Ed. Engl. 52(47), 12359–12363 (2013).
    • 8. Rathore AS, Agarwal H, Sharma AK, Pathak M, Muthukumar S. Continuous processing for production of biopharmaceuticals. Prep. Biochem. Biotechnol. 45(8), 836–849 (2015).
    • 9. Aksu B, De Beer T, Folestad S et al. Strategic funding priorities in the pharmaceutical sciences allied to Quality by Design (QbD) and Process Analytical Technology (PAT). Eur. J. Pharm. Sci. 47(2), 402–405 (2012).
    • 10. Page T, Dubina H, Fillipi G et al. Equipment and analytical companies meeting continuous challenges. May 20–21, 2014 Continuous Manufacturing Symposium. J. Pharm. Sci. 104(3), 821–831 (2015).
    • 11. Page TG. US20110208436A1 (2011).
    • 12. Allison G, Cain YT, Cooney C et al. Regulatory and quality considerations for continuous manufacturing May 20–21, 2014 Continuous Manufacturing Symposium. J. Pharm. Sci. 104(3), 803–812 (2015).
    • 13. Chatterjee S. FDA perspective on continuous manufacturing. Presented at: IFPAC Annual Meeting, Baltimore, MD, USA, 22–25 January 2012.
    • 14. Leuenberger H. New trends in the production of pharmaceutical granules: batch versus continuous processing. Eur. J. Pharm. Biopharm. 52(3), 289–296 (2001).
    • 15. Poechlauer 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. Proc. Res. Dev. 16(10), 1586–1590 (2012).
    • 16. Burcham CL, Florence AJ, Johnson MD. Continuous manufacturing in pharmaceutical process development and manufacturing. Annu. Rev. Chem. Biomol. Eng. 9, 253–281 (2018).
    • 17. Warikoo V, Godawat R, Brower K et al. Integrated continuous production of recombinant therapeutic proteins. Biotechnol. Bioeng. 109(12), 3018–3029 (2012).
    • 18. Pollock J, Coffman J, Ho SV, Farid SS. Integrated continuous bioprocessing: economic, operational, and environmental feasibility for clinical and commercial antibody manufacture. Biotechnol. Prog. 33(4), 854–866 (2017).
    • 19. Byrn S, Futran M, Thomas H et al. Achieving continuous manufacturing for final dosage formation: challenges and how to meet them. May 20-21, 2014 Continuous Manufacturing Symposium. J. Pharm. Sci. 104(3), 792–802 (2015).
    • 20. International Organization for Standardization (ISO). ISO 9000:2015. 20 March 2019 (2015). www.iso.org/standard/45481.html
    • 21. Myerson AS, Krumme M, Nasr M, Thomas H, Braatz RD. Control systems engineering in continuous pharmaceutical manufacturing. May 20-21, 2014 Continuous Manufacturing Symposium. J. Pharm. Sci. 104(3), 832–839 (2015).
    • 22. May SA. Flow chemistry, continuous processing, and continuous manufacturing: a pharmaceutical perspective. J. Flow Chem. 7(3), 137–145 (2017).
    • 23. Matsunami 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. 559, 210–219 (2019).
    • 24. Zomer S, Zhang J, Talwar S, Chattoraj S, Hewitt C. Multivariate monitoring for the industrialisation of a continuous wet granulation tableting process. Int. J. Pharm. 547(1–2), 506–519 (2018). • Highlights the importance of the multivariate statistical analysis for integrating the vast data gathered from the Process Analytical Technology units during continuous manufacturing.
    • 25. Papadakis E, Woodley JM, Gani R. Chapter 24 - Perspective on PSE in pharmaceutical process development and innovation. In: Computer Aided Chemical Engineering. Singh RYuan Z (Eds). Elsevier, Cambridge, MA, USA, 597–656 (2018).
    • 26. Alam MA, Shi Z, Drennen JK 3rd, Anderson CA. In-line monitoring and optimization of powder flow in a simulated continuous process using transmission near infrared spectroscopy. Int. J. Pharm. 526(1–2), 199–208 (2017).
    • 27. Santos B, Carmo F, Schlindwein W et al. Pharmaceutical excipients properties and screw feeder performance in continuous processing lines: a Quality by Design (QbD) approach. Drug Dev. Ind. Pharm. 44(12), 2089–2097 (2018).
    • 28. Van Snick B, Dhondt J, Pandelaere K et al. A multivariate raw material property database to facilitate drug product development and enable in silico design of pharmaceutical dry powder processes. Int. J. Pharm. 549(1–2), 415–435 (2018). • Suggests the application of principle component analysis to limit the ingredients needed and therefore simplify the product development stage of the continuous manufacturing.
    • 29. Yu LX. Pharmaceutical quality by design: product and process development, understanding, and control. Pharm. Res. 25(4), 781–791 (2008).
    • 30. Hubert C, Lebrun P, Houari S, Ziemons E, Rozet E, Hubert P. Improvement of a stability-indicating method by Quality-by-Design versus Quality-by-Testing: a case of a learning process. J. Pharm. Biomed. Anal. 88, 401–409 (2014).
    • 31. Su Q, Ganesh S, Moreno M et al. A perspective on Quality-by-Control (QbC) in pharmaceutical continuous manufacturing. Comp. Chem. Eng. 125, 216–231 (2019).
    • 32. Rathore AS, Winkle H. Quality by design for biopharmaceuticals. Nat. Biotech. 27(1), 26 (2009).
    • 33. Yu LX, Amidon G, Khan MA et al. Understanding pharmaceutical quality by design. AAPS J. 16(4), 771–783 (2014).
    • 34. Grymonpre W, Bostijn N, Herck SV et al. Downstream processing from hot-melt extrusion towards tablets: a quality by design approach. Int. J. Pharm. 531(1), 235–245 (2017).
    • 35. Lakio S, Ervasti T, Tajarobi P et al. Provoking an end-to-end continuous direct compression line with raw materials prone to segregation. Eur. J. Pharm. Sci. 109, 514–524 (2017).
    • 36. Schlindwein W, Bezerra M, Almeida J, Berghaus A, Owen M, Muirhead G. In-Line UV-Vis Spectroscopy as a Fast-Working Process Analytical Technology (PAT) during Early Phase Product Development Using Hot Melt Extrusion (HME). Pharmaceutics 10(4), 1–25 (2018).
    • 37. Srai JS, 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. 104(3), 840–849 (2015).
    • 38. Tomba E, De Martin M, Facco P et al. General procedure to aid the development of continuous pharmaceutical processes using multivariate statistical modeling – an industrial case study. Int. J. Pharm. 444(1–2), 25–39 (2013).
    • 39. Silva AF, Sarraguca MC, Fonteyne M et al. Multivariate statistical process control of a continuous pharmaceutical twin-screw granulation and fluid bed drying process. Int. J. Pharm. 528(1–2), 242–252 (2017).
    • 40. Stauffer F, Vanhoorne V, Pilcer G, Chavez PF, Vervaet C, De Beer T. Managing API raw material variability during continuous twin-screw wet granulation. Int. J. Pharm. 561, 265–273 (2019).
    • 41. Rogers A, Hashemi A, Ierapetritou M. Modeling of particulate processes for the continuous manufacture of solid-based pharmaceutical dosage forms. Processes 1(2), 67–127 (2013).
    • 42. Toson P, Siegmann E, Trogrlic M et al. Detailed modeling and process design of an advanced continuous powder mixer. Int. J. Pharm. 552(1), 288–300 (2018).
    • 43. Persson LC, Porter CJH, Charman WN, Bergström CaS. Computational prediction of drug solubility in lipid based formulation excipients. Pharmaceutical Res. 30(12), 3225–3237 (2013).
    • 44. O'connell JP, Gani R, Mathias PM, Maurer G, Olson JD, Crafts PA. Thermodynamic property modeling for chemical process and product engineering: Some perspectives. Ind. Eng. Chem. Res. 48(10), 4619–4637 (2009).
    • 45. Boukouvala F, Chaudhury A, Sen M et al. Computer-aided flowsheet simulation of a pharmaceutical tablet manufacturing process incorporating wet granulation. J. Pharm. Innov. 8(1), 11–27 (2013).
    • 46. Boeckx J, Page TG, Waldron MS. WO2010128359A1 (2017).
    • 47. Escotet-Espinoza MS, Vadodaria S, Singh R, Muzzio FJ, Ierapetritou MG. Modeling the effects of material properties on tablet compaction: A building block for controlling both batch and continuous pharmaceutical manufacturing processes. Int. J. Pharm. 543(1), 274–287 (2018).
    • 48. García-Muñoz S, Butterbaugh A, Leavesley I, Manley LF, Slade D, Bermingham S. A flowsheet model for the development of a continuous process for pharmaceutical tablets: an industrial perspective. AIChE J. 64(2), 511–525 (2018).
    • 49. Wang Z, Escotet-Espinoza MS, Ierapetritou M. Process analysis and optimization of continuous pharmaceutical manufacturing using flowsheet models. Comput. Chem. Eng. 107, 77–91 (2017). •• Develops an integrated flowsheet model for better understanding of continuous manufacturing process. This systematic approach involved sensitivity analysis, feasibility analysis and optimization for continuous direct compression process.
    • 50. Fernandez EG, Cordero S, Benitez M et al. Rapid development and optimization of tablet manufacturing using statistical tools. AAPS PharmSciTech 9(2), 620–627 (2008).
    • 51. Collins LM, Dziak JJ, Li R. Design of experiments with multiple independent variables: a resource management perspective on complete and reduced factorial designs. Psych. Met. 14(3), 202–224 (2009).
    • 52. Nasr MM, Krumme M, Matsuda Y et al. 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. 106(11), 3199–3206 (2017).
    • 53. Gao Y, Ierapetritou M, Muzzio F. Periodic section modeling of convective continuous powder mixing processes. AIChE J. 58(1), 69–78 (2012).
    • 54. Kruisz J, Rehrl J, Sacher S, Aigner I, Horn M, Khinast JG. RTD modeling of a continuous dry granulation process for process control and materials diversion. Int. J. Pharm. 528(1–2), 334–344 (2017).
    • 55. Martinetz MC, Karttunen AP, Sacher S et al. RTD-based material tracking in a fully-continuous dry granulation tableting line. Int. J. Pharm. 547(1–2), 469–479 (2018).
    • 56. Laske S, Paudel A, Scheibelhofer O, Author T. A review of PAT strategies in secondary solid oral dosage manufacturing of small molecules. J. Pharm. Sci. 106(3), 667–712 (2017).
    • 57. Maniruzzaman M, Douroumis D. Continuous manufacturing and process analytical tools. Int. J. Pharm. 496(1), 1–2 (2015).
    • 58. International Conference on Harmonisation (Ich). Guidance for industry: Q8 (R2) (pharmaceutical development). Center for Drug Evaluation and Research (2009). www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Quality/Q8_R1/Step4/Q8_R2_Guideline.pdf
    • 59. (Fda) FDA. Guidance for industry, PAT-A framework for innovative pharmaceutical development, manufacturing and quality assurance (2004). www.fda.gov/media/71012/download
    • 60. Esmonde-White KA, Cuellar M, Uerpmann C, Lenain B, Lewis IR. Raman spectroscopy as a process analytical technology for pharmaceutical manufacturing and bioprocessing. Anal. Bioanal. Chem. 409(3), 637–649 (2017).
    • 61. Ganesh S, Troscinski R, Schmall N, Lim J, Nagy Z, Reklaitis G. Application of x-ray sensors for in-line and noninvasive monitoring of mass flow rate in continuous tablet manufacturing. J. Pharm. Sci. 106(12), 3591–3603 (2017).
    • 62. Nagy B, Farkas A, Magyar K, Demuth B, Nagy ZK, Marosi G. Spectroscopic characterization of tablet properties in a continuous powder blending and tableting process. Eur. J. Pharm. Sci. 123, 10–19 (2018).
    • 63. Rathore AS, Wood R, Sharma A, Dermawan S. Case study and application of process analytical technology (PAT) towards bioprocessing: II. Use of ultra-performance liquid chromatography (UPLC) for making real-time pooling decisions for process chromatography. Biotechnol. Bioeng. 101(6), 1366–1374 (2008).
    • 64. Maniruzzaman M, Nair A, Renault M et al. Continuous twin-screw granulation for enhancing the dissolution of poorly water soluble drug. Int. J. Pharm. 496(1), 52–62 (2015).
    • 65. Xu X, Vallabh CKP, Hoag SW, Dave VS, Cetinkaya C. Early detection of capping risk in pharmaceutical compacts. Int. J. Pharm. 553(1-2), 338–348 (2018).
    • 66. Maniruzzaman M, Nokhodchi A. Continuous manufacturing via hot-melt extrusion and scale up: regulatory matters. Drug Discov. Today 22(2), 340–351 (2017).
    • 67. Reich G. Near-infrared spectroscopy and imaging: basic principles and pharmaceutical applications. Adv. Drug Deliv. Rev. 57(8), 1109–1143 (2005).
    • 68. Blanco M, Bautista M, Alcalà M. API determination by NIR spectroscopy across pharmaceutical production process. AAPS PharmSciTech 9(4), 1130–1135 (2008).
    • 69. Vo AQ, He H, Zhang J, Martin S, Chen R, Repka MA. Application of FT-NIR analysis for in-line and real-time monitoring of pharmaceutical hot melt extrusion: a technical note. AAPS PharmSciTech 19(8), 3425–3429 (2018).
    • 70. Acevedo D, Muliadi A, Giridhar A, Litster JD, Romanach RJ. Evaluation of three approaches for real-time monitoring of roller compaction with near-infrared spectroscopy. AAPS PharmSciTech 13(3), 1005–1012 (2012).
    • 71. Crowley ME, Hegarty A, Mcauliffe MaP et al. Near-infrared monitoring of roller compacted ribbon density: investigating sources of variation contributing to noisy spectral data. Eur. J. Pharm. Sci. 102, 103–114 (2017).
    • 72. Tezyk 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. 42(8), 1195–1214 (2016).
    • 73. Riolo D, Piazza A, Cottini C et al. Raman spectroscopy as a PAT for pharmaceutical blending: advantages and disadvantages. J. Pharm. Biomed. Anal. 149, 329–334 (2018).
    • 74. Acevedo D, Yang X, Mohammad A et al. Raman spectroscopy for monitoring the continuous crystallization of carbamazepine. Org. Process Res. Dev. 22(2), 156–165 (2018).
    • 75. Abu-Absi NR, Kenty BM, Cuellar ME et al. Real time monitoring of multiple parameters in mammalian cell culture bioreactors using an in-line Raman spectroscopy probe. Biotechnol. Bioeng. 108(5), 1215–1221 (2011).
    • 76. Chavan RB, Thipparaboina R, Yadav B, Shastri NR. Continuous manufacturing of co-crystals: challenges and prospects. Drug Deliv. Transl. Res. 8(6), 1726–1739 (2018).
    • 77. U.S. Food and Drug Adminstration (Fda). Quality considerations for continuous manufacturing guidance for industry. Human Drug Information (2019). www.fda.gov/media/121314/download
    • 78. Farias M, Carneiro R. Simultaneous quantification of three polymorphic forms of carbamazepine in the presence of excipients using Raman spectroscopy. Mol. 19(9), 14128–14138 (2014).
    • 79. Matthews TE, Coffman C, Kolwyck D, Hill D, Dickens JE. Enabling robust and rapid raw material identification and release by handheld raman spectroscopy. PDA J. Pharm. Sci. Technol. doi:10.5731/pdajpst.2018.009563 (2019) (Epub ahead of print).
    • 80. Fonteyne M, Wickstrom H, Peeters E et al. Influence of raw material properties upon critical quality attributes of continuously produced granules and tablets. Eur. J. Pharm. Biopharm. 87(2), 252–263 (2014).
    • 81. Escotet-Espinoza MS, Vadodaria S, Singh R, Muzzio FJ, Ierapetritou MG. Modeling the effects of material properties on tablet compaction: A building block for controlling both batch and continuous pharmaceutical manufacturing processes. Int. J. Pharm. 543(1–2), 274–287 (2018).
    • 82. Kushner J, Langdon BA, Hiller JI, Carlson GT. Examining the impact of excipient material property variation on drug product quality attributes: a quality-by-design study for a roller compacted, immediate release tablet. J. Pharm. Sci. 100(6), 2222–2239 (2011).
    • 83. Wahl PR, Treffer D, Mohr S, Roblegg E, Koscher G, Khinast JG. Inline monitoring and a PAT strategy for pharmaceutical hot melt extrusion. Int. J. Pharm. 455(1–2), 159–168 (2013).
    • 84. Ooi SM, Sarkar S, Van Varenbergh G, Schoeters K, Heng PW. Continuous processing and the applications of online tools in pharmaceutical product manufacture: developments and examples. Therap. Del. 4(4), 463–470 (2013).
    • 85. Shanmugam S. Granulation techniques and technologies: recent progresses. BioImpacts. BI 5(1), 55 (2015).
    • 86. Kleinebudde P. Roll compaction/dry granulation: pharmaceutical applications. Eur. J. Pharm. Biopharm. 58(2), 317–326 (2004).
    • 87. Herting MG, Kleinebudde P. Studies on the reduction of tensile strength of tablets after roll compaction/dry granulation. Eur. J. Pharm. Biopharm. 70(1), 372–379 (2008).
    • 88. Farrenkopf J. Relevant aspects of roller compaction covering the impact of excipients, milling devices, fines and feasibility prediction (2009).
    • 89. Singh R, Ierapetritou M, Ramachandran R. An engineering study on the enhanced control and operation of continuous manufacturing of pharmaceutical tablets via roller compaction. Int. J. Pharm. 438(1), 307–326 (2012). • Develops a new integrated framework for manufacturing, on-line monitoring and closed-loop control to enhance CQAs performance of CM.
    • 90. Agrawal R, Naveen Y. Pharmaceutical processing: a review on wet granulation technology. Int. J. Pharm. Front. Res. 1(1), 65–83 (2011).
    • 91. Vervaet C, Remon JP. Continuous granulation in the pharmaceutical industry. Chem. Eng. Sci. 60(14), 3949–3957 (2005).
    • 92. Ghebre-Sellassie I, Mollan Jr MJ, Pathak N, Lodaya M, Fessehaie M. US6499984B1 (2002).
    • 93. Baronsky-Probst J, Moltgen CV, Kessler W, Kessler RW. Process design and control of a twin screw hot melt extrusion for continuous pharmaceutical tamper-resistant tablet production. Eur. J. Pharm. Sci. 87, 14–21 (2016).
    • 94. Nicolaï N, De Leersnyder F, Copot D et al. Liquid-to-solid ratio control as an advanced process control solution for continuous twin-screw wet granulation. AIChE J. 64(7), 2500–2514 (2018).
    • 95. Seem TC, Rowson NA, Ingram A et al. Twin screw granulation: a literature review. Powder Technol. 276, 89–102 (2015).
    • 96. Maniruzzaman M, Nair A, Scoutaris N, Bradley MS, Snowden MJ, Douroumis D. One-step continuous extrusion process for the manufacturing of solid dispersions. Int. J. Pharm. 496(1), 42–51 (2015).
    • 97. Mcclements DJ. Recent progress in hydrogel delivery systems for improving nutraceutical bioavailability. Food Hydrocol. 68, 238–245 (2017).
    • 98. Van Hauwermeiren D, Verstraeten M, Doshi P et al. On the modelling of granule size distributions in twin-screw wet granulation: Calibration of a novel compartmental population balance model. Powder Technol. 341, 116–125 (2019).
    • 99. Zhang Y, Law Y, Chakrabarti S. Physical properties and compact analysis of commonly used direct compression binders. AAPS PharmSciTech 4(4), 489–499 (2003).
    • 100. Van Snick B, Holman J, Cunningham C et al. Continuous direct compression as manufacturing platform for sustained release tablets. Int. J. Pharm. 519(1–2), 390–407 (2017).
    • 101. Jivraj M, Martini LG, Thomson CM. An overview of the different excipients useful for the direct compression of tablets. Pharm. Sci. Tech. Today 3(2), 58–63 (2000).
    • 102. Chattoraj S, Sun CC. Crystal and Particle Engineering Strategies for Improving Powder Compression and Flow Properties to Enable Continuous Tablet Manufacturing by Direct Compression. J. Pharm. Sci. 107(4), 968–974 (2018).
    • 103. Kowalski J, Lakshman JP, Serajuddin ATM, Tong W, Vasanthavada M. WO2009134848A1 (2011).
    • 104. Simonaho S, 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. 90, 38–46 (2016).
    • 105. Ervasti T, Simonaho S, Ketolainen J et al. Continuous manufacturing of extended release tablets via powder mixing and direct compression. Int. J. Pharm. 495(1), 290–301 (2015).
    • 106. Bostijn N, Van Renterghem J, Dhondt W, Vervaet C, De Beer T. A continuous manufacturing concept for a pharmaceutical oral suspension. Eur. J. Pharm. Sci. 123, 576–583 (2018).
    • 107. Walsh G. Biopharmaceuticals and biotechnology medicines: an issue of nomenclature. Eur. J. Pharm. Sci. 15(2), 135–138 (2002).
    • 108. Hong MS, Severson KA, Jiang M, Lu AE, Love JC, Braatz RD. Challenges and opportunities in biopharmaceutical manufacturing control. Comp. Chem. Eng. 110, 106–114 (2018).
    • 109. Shire SJ. Formulation and manufacturability of biologics. Curr. Opin. Biotech. 20(6), 708–714 (2009).
    • 110. Tamizi E, Jouyban A. The potential of the capillary electrophoresis techniques for quality control of biopharmaceuticals: a review. Electroph. 36(6), 831–858 (2015).
    • 111. Rosa PaJ, Azevedo AM, Sommerfeld S, Mutter M, Bäcker W, Aires-Barros MR. Continuous purification of antibodies from cell culture supernatant with aqueous two-phase systems: from concept to process. Biotechnol. J. 8(3), 352–362 (2013).
    • 112. Boi C. Membrane adsorbers as purification tools for monoclonal antibody purification. J. Chromatogr. B 848(1), 19–27 (2007).
    • 113. Konstantinov K, Godawat R, Warikoo V, Jain S. Integrated continuous manufacturing of therapeutic protein drug substances (2017). https://patents.google.com/patent/US9650412B2/en
    • 114. Ransohoff TC, Bisschops MaT. WO2012078677A3 (2012).
    • 115. Baumann M, Baxendale IR. The synthesis of active pharmaceutical ingredients (APIs) using continuous flow chemistry. Beilstein. J. Org. Chem. 11, 1194–1219 (2015).
    • 116. Adamo A, Beingessner R, Behnam M et al. On-demand continuous-flow production of pharmaceuticals in a compact, reconfigurable system. Science 352(6281), 61–67 (2016). •• Reconfigurable manufacturing platform was developed to synthesize and formulate different active pharmaceutical ingredients in a small-sized plant with real-time monitoring.
    • 117. Crowell E, Lu AE, Love KR et al. On-demand manufacturing of clinical-quality biopharmaceuticals. Nat. Biotech. 36, 988–995 (2018).
    • 118. Gross TD, Chou S, Bonneville D et al. Chemical development of NBI-75043. Use of a flow reactor to circumvent a batch-limited metal− Halogen exchange reaction. Org. Proc. Res. Dev. 12(5), 929–939 (2008).
    • 119. Therre J, Oost C. WO2001016237A2 (2001).
    • 120. Vanarase AU, Osorio JG, Muzzio FJ. Effects of powder flow properties and shear environment on the performance of continuous mixing of pharmaceutical powders. Powder Technol. 246, 63–72 (2013).
    • 121. Tazzia CL, Villalobos MA, Harris MA. Process for continuous production of epoxy resins (2012). https://patents.google.com/patent/US8242234B2/en
    • 122. Born SC, Dittrich JV, Takiza BT, Mascia S. Continuous drum dryer and methods of use thereof (2017). https://patents.google.com/patent/WO2017136765A1/en
    • 123. Patil H, Kulkarni V, Majumdar S, Repka MA. Continuous manufacturing of solid lipid nanoparticles by hot melt extrusion. Int. J. Pharm. 471(1), 153–156 (2014).
    • 124. Hou D, Xie C, Huang K, Zhu C. The production and characteristics of solid lipid nanoparticles (SLNs). Biomaterials 24(10), 1781–1785 (2003).
    • 125. Maherani B, Arab-Tehrany E, R Mozafari M, Gaiani C, Linder M. Liposomes: a review of manufacturing techniques and targeting strategies. Curr. Nanosci. 7(3), 436–452 (2011).
    • 126. Dimov N, Kastner E, Hussain M, Perrie Y, Szita N. Formation and purification of tailored liposomes for drug delivery using a module-based micro continuous-flow system. Sci. Rep. 7(1), 12045 (2017).
    • 127. Wagner A, Vorauer-Uhl K, Katinger H. Liposomes produced in a pilot scale: production, purification and efficiency aspects. Eur. J. Pharm. Biopharm. 54(2), 213–219 (2002).
    • 128. Ruysschaert T, Marque A, Duteyrat J, Lesieur S, Winterhalter M, Fournier D. Liposome retention in size exclusion chromatography. BMC Biotech. 5(1), 11 (2005).
    • 129. Costa A, Burgess DJ. Systems and methods for continuous manufacturing of liposomal drug formulations (2019). https://patents.google.com/patent/US20190029959A1/en
    • 130. Aboofazeli R. Nanometric-scaled emulsions (nanoemulsions). Iran. J. Pharm. Res. 9(4), 325–326 (2010).
    • 131. Sah E, Sah H. Recent trends in preparation of poly (lactide-co-glycolide) nanoparticles by mixing polymeric organic solution with antisolvent. J. Nanomat. 16(1), 61 (2015).
    • 132. Choi G, Kakumanu S, Schmitz L, Robinson GL, Beckett CD, Laugharn Jr JA. Continuous manufacturing of carboxyamidotriazole-encapsulated nanoemulsions using adaptive focused acoustics: potential green technology for the pharmaceutical industry. J. Biomed. Eng. Informatics. 1(19), 20 (2016).
    • 133. Jeevanandam J, Barhoum A, Chan YS, Dufresne A, Danquah MK. Review on nanoparticles and nanostructured materials: history, sources, toxicity and regulations. Beilstein. J. Nanotechnol. 9, 1050–1074 (2018).
    • 134. Fonseca T, Duarte I, Temtem M, Vicente J. Continuous production of particles. WO2016156841A1 (2018). https://patents.google.com/patent/WO2016156841A1/en?oq=WO2016156841A1
    • 135. Gottlieb S. FDA budget matters: investing in advanced domestic manufacturing. FDA Blog (2018).
    • 136. U.S. Food and Drug Adminstration (Fda). FDA supports critical research to spur innovation for continuous manufacturing technology to support and advance drug and biologics development (2018). www.fda.gov/news-events/fda-brief/fda-supports-critical-research-spur-innovation-continuous-manufacturing-technology-support-and
    • 137. Jolliffe HG, Gerogiorgis DI. Plantwide design and economic evaluation of two Continuous Pharmaceutical Manufacturing (CPM) cases: Ibuprofen and artemisinin. Comp. Chem. Eng. 91, 269–288 (2016).
    • 138. Transparency Market Research. Pharmaceutical continuous manufacturing market (application - active pharmaceutical ingredient, biologics, and dry powders; end users - pharmaceutical companies and contract manufacturing organization) global industry analysis, size, share, growth, trends, and forecast 2017 - 2025 (2017). www.transparencymarketresearch.com/pharmaceutical-continuous-manufacturing-technology-market.html
    • 139. Global pharmaceutical continuous manufacturing market to surpass USD 3406.0 million by 2024 www.globenewswire.com/news-release/2018/09/06/1566507/0/en/Global-Pharmaceutical-Continuous-Manufacturing-Market-to-Surpass-USD-3406-0-Million-By-2024-Zion-Market-Research.html
    • 140. Wallack T. Novartis to give MIT $65m to find new way to produce drugs. The Boston Globe (2007). http://archive.boston.com/business/articles/2007/09/28/novartis_to_give_mit_65m_to_find_new_way_to_produce_drugs/
    • 141. Matsuda Y. Global Regulatory Landscape. AAPS PharmSciTech 20(1), 2 (2019).
    • 142. Urquhart L. Regulatory watch: FDA new drug approvals in Q1 2018. Nat. Rev. Drug Discov. 17(5), 309 (2018).
    • 143. Diprospero D. Continuous OSD manufacturing - A product & patient perspective. Int. Soc. Pharm. Engl. (2018). https://ispe.org/pharmaceutical-engineering/ispeak/continuous-osd-manufacturing-product-patient-perspective
    • 144. Macdonald G. Janssen working on other continuous processes post US FDA OK for Prezista. In: PharmaTech (2016).
    • 145. Kim E. Abemaciclib: first global approval. Drugs 77(18), 2063–2070 (2017).
    • 146. Schneider EK, Reyes-Ortega F, Wilson JW et al. Development of HPLC and LC–MS/MS methods for the analysis of ivacaftor, its major metabolites and lumacaftor in plasma and sputum of cystic fibrosis patients treated with ORKAMBI or KALYDECO. J. Chromatogr. B 1038, 57–62 (2016).
    • 147. Vertex Pharmaceuticals Company. Vertex reports full-year and fourth-quarter 2017 financial results. (2018). https://investors.vrtx.com/news-releases/news-release-details/vertex-reports-full-year-and-fourth-quarter-2017-financial
    • 148. Vertex Pharmaceuticals Company. Vertex reports full-year and fourth-quarter 2018 financial results. (2019). https://investors.vrtx.com/news-releases/news-release-details/vertex-reports-full-year-and-fourth-quarter-2018-financial
    • 149. Lohr L. Abemaciclib (verzenio™). Onc. Times 40(6), 12 (2018).
    • 150. Eli Lilly Q4: Looking at low verzenio sales and the loxo oncology acquisition https://seekingalpha.com/article/4239475-eli-lilly-q4-looking-low-verzenio-sales-loxo-oncology-acquisition
    • 151. Matsunami K, Miyano T, Arai H, Nakagawa H, Hirao M, Sugiyama H. Decision support method for the choice between batch and continuous technologies in solid drug product manufacturing. Ind. Eng. Chem. Res. 57(30), 9798–9809 (2018). • The research paper included an innovated model for choosing between batch and CM based on economic analysis.