• Ei tuloksia

The meaning of the mass stream in the continuous process

be built with a few or several process units in the same manufacturing area. In the future, a more flexible and efficient approach could be even ´mobile´ CM lines built into a container (already available from GEA), which could be transferred between countries.

In our three studies, all the processes were performed in the same and small manufacturing area, which demonstrated that the process was adaptable to changes in the set-up operations. Another approach is the conversation from a batch mode manufactured product (already on the market) to a continuous mode i.e. requiring only a post-approval manufacturing change (ISCMP, 2014). This makes it possible to adopt more flexible variations in the operations. In our comparison studies, the bridging of these two methods was demonstrated with a granulated tablet product. Although there is a growing encouragement from regulatory bodies in the area of CM, there is a clear and evident need for a real harmonization of implementation procedures between different countries.

7.2 QUALITY BY DESIGN APPROACH

The guidelines issued by the regulatory authorities e.g. ICH Q8 (R2), ICH Q9 and ICH Q10 support the development approach called Quality by Design. This new option is the underpinning of the concept that there are quality aspects in the manufacturing of pharmaceuticals; the ´Control Strategy´ is the key factor of enhanced QbD -based manufacturing. This provides a better understanding of quality elements during the product’s lifecycle compared to the minimal approach as applied today in traditional manufacturing processes. Design Space follows the QbD principles; its target is to find the parameters which exert the greatest influence on the critical quality attributes of the product. Our results demonstrated that design space can be achieved in the whole CM process line producing a tablet that conforms to the quality requirements. Furthermore, we revealed the desired flexibility to operate in the CM process along with a scientific understanding of multiple combinations and the interactions between different parameters. The benefit of CM processes is that the DS does not need to be re-created in scaling-up processes as the equipment used is the same. In our results, the created DS covered the whole continuous process line. A similar kind of publication was not found in the literature in the context of continuous direct compressible tablet production.

7.3 THE MEANING OF THE MASS STREAM IN THE CONTINUOUS PROCESS

An understanding material transport operations between the steps and each unit process represents the basis of continuous manufacturing. Care must be taken with system 117

design, start-up and shut-down operations e.g. that there is a synchronized material transfer between the unit processes in order to avoid a control system runaway. It is clear that with a continuous process running 24/7, different kinds of interruptions (unintentional and intentional) during the runs will occur. One of the most important aspects of this dissertation was to investigate the factors influencing the mass stream; i.e.

lubrication, different set-up and disturbances. The study focused on the importance of achieving a constant mass stream during the runs on the process line with different unit process configurations. In our study, the CM process was subjected to stresses in every process step, in order to pinpoint the effect of interruptions during a long run on the quality of the final product. The study confirmed, in agreement with the results already published in the literature, that more studies using the whole CM line (not only a part of unit processes) should be performed. In addition to a carefully designed set-up, it is obvious that lubrication has an important role in the material transfer phenomena, as its function is to ease the contact between the particles and to enhance both flowability and tabletability. The effect of lubrication was examined in all our studies to demonstrate the influence of lubricant based parameters on a continuous manufacturing process.

Although the process parameters were shown to have a minor effect on the quality of the final product, they should be still evaluated, monitored and analysed. However, material properties had the most important influence on the final product. It was somewhat surprising that a high variation in the feed rates of lubricants did not exert a negative effect on the quality of the final product. This was mainly attributable to the flowing properties of the main components. Thus, the flow rate of main components and the mixing strategy need to be carefully planned and monitored.

118

8 CONCLUSIONS

The present study demonstrated the feasibility and flexibility of the specific continuous manufacturing process line, allowing it to be modified into different configurations and formulations without encountering any significant quality issues. In addition, the effect of lubrication was systematically evaluated in all of these studies. On the basis of the results, it can be concluded more specifically that

I The continuous process line studied was found to be suitable for integration into a top-down model for direct compressible paracetamol tablets using QbD approach. The Design Space created showed the good feasibiliy and flexibility of the continuous line to produce tablets that conformed to the quality requirements.

II The continuous process line studied was suitable for being configurated into a horizontal model with several unit processes to produce direct compressible paracetamol tablets. Its flexibility was examined by exposing the process to both intentional and unintentional disturbances during long runs and determining which of these affected the quality of the product. In addition, the results highlighted the importance of connecting the unit processes together to maintain a constant mass flow throughout the whole process line.

III A feasible conversion from the HSWG batch to the DG continuous mode could be achieved with slight modifications of formulation and unit processes. Both manufacturing methods fulfilled the requirements, thus demonstrating more flexible and effective ways to allow the pharmaceutical industry to switch from batch manufacturing towards CM.

119

REFERENCES

Aikawa, S., Fujita, N., Myojo, H., Hayashi, T., Tanino, T., 2008: Scale-up studies on high shear wet granulation process from mini-scale to commercial scale. Chem. Pharm.

Bull. 56, 1431 – 1435.

Akseli, I., Iyer, S., Lee, H., Cuitiño, A., 2011. A Quantitative Correlation of the Effect of Density Distributions in Roller-Compacted Ribbons on the Mechanical Properties of Tablets Using Ultrasonics and X-ray Tomography. AAPS PharmSciTech. 12, 834-853.

Allenspach, C., Timmins, P., Sharif, S., Minko, T.,2020. Characterization of a novel hydroxypropyl methylcellulose (HPMC) direct compression grade excipient for pharmaceutical tablets. Int. J. Pharm. 583, 119-343.

Almaya, A., De Belder, L., Meyer, R., Nagapudi, K., Homer Lin, H.R., Leavesley, I., Jayanth, J., Bajwa, G., DiNunzio, J., Tantuccio, A., Blackwood, D., Abebe, A. 2017.

Control strategies for drug product continuous direct compression-state of control, product collection strategies, and startup/shutdown operations for production of clinical trial materials and commercial products. J.Pharm. Sci. 106, 930 – 943.

Ameye, D., Keleb, E., Vervaet, C., Remon, J.P., Adams, E., Massart, D.L., 2002: Scaling – up of lactose wet granulation process in Mi-Pro high shear mixers. Eur. J. Pharm. Sci.

17, 247 – 251.

Anderson, N.G., 2001. Practical Use of Continuous Processing in Developing and Scaling Up Laboratory Processes. Organic Process Research & Development. 5, 613-621.

Arndt, O., Kleinebudde, P., 2018. Influence of binder properties on dry granules and tablets. Powder Technol. 337, 68-77.

Bacher, C., Olsen, P.M., Bertelsen, P., Kristensen, J., Sonnergaard, J.M., 2007. Improving the compaction properties of roller compacted calcium carbonate. Int. J. Pharm. 342, 115 – 123.

Badman, C., Trout, B.L., 2015. Achieving continuous manufacturing May 20-21, 2014 continuous manufacturing symposium. J Pharm. Sci. 104, 779-780.

Bandari, S., Nyavanandi, D., Kallakunta, V.R., Janga, K.Y., Sarabu, S., Butreddy, A., Repka, M.A., 2020. Continuous twin screw granulation – An advanced alternative granulation technology for use in the pharmaceutical industry. Int. J. Pharm. 580.

Bano, G., Facco, P., Ierapetritou, M., Bezzo, F., Barolo, M., 2019. Design space maintenance by online model adaptation in pharmaceutical manufacturing. (Report). Comp &

Chem. Eng. 127:254.

Barimani, S., Šibanc, R., Kleinebudde, P.,2018. Optimization of a semi-batch tablet coating process for a continuous manufacturing line by design of experiments. Int. J. Pharm.

539, 95-103.

120

Baronsky-Probst, J., Möltgen, C., Kessler, W., Kessler, R.W., 2016. 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.

Barton, J.H., Emanuel, E.J., 2005. The Patents-Based Pharmaceutical Development Process: Rationale, Problems, and Potential Reforms. JAMA. American Medical Association. 294, 2075-2082.

Bastos, M.D., Friedrich, R.B., Beck, R.C.R., 2008. Effects of filler-binders and lubricants on physicochemical properties of tablets obtained by direct compression: A 2(2) factorial design. Latin. American J. Pharm. 27, 578-583.

Beer, P., Wilson, D., Huang, Z., De Matas, M., 2014. Transfer from High‐Shear Batch to Continuous Twin Screw Wet Granulation: A Case Study in Understanding the Relationship Between Process Parameters and Product Quality Attributes. J. Pharm.

Sci.103, 3075-3082.

Beg, S., Rahman, M., Panda, S.S., 2017. Pharmaceutical QbD: Omnipresence in the product development lifecycle. Eur. Pharm. Rev. 22, 58-64.

Bernardes, M. A., Martinez, L., Esquerdo, R. M., Hausner, D., Romanach, R. J., 2015.

Continuous manufacturing, near infrared spectroscopy and process knowledge.

American Pharmaceutical review. Available at

https://www.americanpharmaceuticalreview.com/Featured-Articles/177547-Continuous-Manufacturing-Near-Infrared-Spectroscopy-and-Process-Knowledge/.

Retrieved 9 Feb 2021.

Berthiaux, H., Marikh, K., Gatumel, C. 2008. Continuous mixing of powder mixtures with pharmaceutical process constraints. Chem. Eng. Process. Process. Intensif. 47, 2315-2322.

Besenhard, M.O., Karkala, S.K., Faulhammer, E., Fathollahi, S., Ramachandran, R., Khinast, J.G.,2016. Continuous feeding of low-dose APIs via periodic micro dosing.

Int. J. Pharm. 509, 123-134.

Billups, M., Singh, R., 2018. Material Traceability in Continuous Pharmaceutical Tablet Manufacturing. Pharm. Technol. 42, 32-59.

Bolhuis, G.K., De Jong, S.W., Lerk, C.F., Dettmers, H., Pharbita, B.V., 1987. The Effect of Magnesium Stearate Admixing in Different Types of Laboratory and Industrial Mixers on Tablet Crushing Strength. Drug. Dev. Ind. Pharm. 13, 1547-1567.

Bolhuis, G.K., Lerk, C.F., Zijlstra, H.T., De Boer, A.H., 1975. Film formation by magnesium stearate during mixing and its effect on tabletting. Pharm. Weekbl. 110, 317-325.

Bolhuis, G.K., Smallenbroek, A.J., Lerk, C.F., 1981. Interaction of tablet disintegrants and magnesium stearate during mixing I: Effect on tablet disintegration. J. Pharm. Sci. 70, 1328-1330.

Bossert, J., Stains, A., 1980. Effect of mixing on the lubrication of crystalline lactose by magnesium stearate. Drug. Dev. Ind. Pharm. 6, 573-589.

121

Burcham, C.L., Florence, A.J., Johnson, M.D., 2018. Continuous Manufacturing in Pharmaceutical Process Development and Manufacturing. Ann. Rev. Chem. Bio.Eng.

9, 253 – 281.

Byrn, S., Futran, M., Thomas, H., Jayjock, E., Maron, N., Meyer, R.F., Myerson, A.S., Thien, M.P., Trout, B.L., 2015. Achieving Continuous Manufacturing for Final Dosage Formation: Challenges and How to Meet Them May 20–21 2014 Continuous Manufacturing Symposium. J. Pharm. Sci. 104, 792-802.

Cameron, L., 2020. Continuous progress in continuous manufacturing: on the road to real-time release. Drug. Discov. Today. 25, 263-264.

Center for Drug Evaluation and Research (CDER), Food and Drug Administration, 2019.

Draft Guidance: Quality considerations for continuous manufacturing, Guidance for Industry.

Charoo, N.A., Shamsher, A.A.A., Zidan, A.S., Rahman, Z., 2012. Quality by design approach for formulation development: A case study of dispersible tablets. Int. J.

Pharm. 423, 167-178.

Chatterjee, S., 2012. FDA perspective on continuous manufacturing. Available at https://www.fda.gov/files/about%20fda/published/FDA-Perspective-on-Continuous- Manufacturing--Sharmista-Chatterjee--Ph.D.--January-22--2012--IFPAC-Annual-Meeting.pdf. Retrieved 10 Feb 2021.

Chatzizacharia, K.A., Hatziavramidis, D.T., 2014. Design Space Approach for Pharmaceutical Tablet Development. Ind. Eng. Chem. Res. 53, 12003-12009.

Chavez, P., Lebrun, P., Sacré, P., De Bleye, C., Netchacovitch, L., Cuypers, S., Mantanus, J., Motte, H., Schubert, M., Evrard, B., Hubert, P., Ziemons, E., 2015. Optimization of a pharmaceutical tablet formulation based on a design space approach and using vibrational spectroscopy as PAT tool. Int. J. Pharm. 486, 13.

Ciurczak, E., 2016. Continuous Manufacturing-Breaking Down Barriers and Changing Perceptions. Pharm. Tech., CPHI8

Cole, K. P., Johnson, M. D., 2018. Continuous Flow Technology vs. the Batch-by-Batch Approach to Produce Pharmaceutical Compounds. Expert Rev. Clin. Pharmacol. 11, 5-13.

Colón, Y., Vargas, J., Sánchez, E., Navarro, G., Romañach, R., 2017. Assessment of Robustness for a Near-Infrared Concentration Model for Real-Time Release Testing in a Continuous Manufacturing Process. J. Pharm. Innov. 12, 14-25.

Crouter, A., Briens, L., 2014. The Effect of Moisture on the Flowability of Pharmaceutical Excipients, AAPS. Pharm. Sci. Tech. 15, 65-74.

Crowley, M.M., Zhang, F., Repka, M.A. Thumma, S., Upadhye, S.B., Kumar Battu, S., McGinity, J.W., Martin, C., 2008. Pharmaceutical applications of hot-melt extrusion:

Part I. Drug. Dev. Ind. Pharm. 33, 909-926.

De Leersnyder, F., Vanhoorne, V., Bekaert, H., Vercruysse, J., Ghijs, M., Bostijn, N., Verstraeten, M. 2018. Breakage and drying behaviour of granules in a continuous fluid

122

bed dryer: Influence of process parameters and wet granule transfer. E. J. Pharm. Sci.

115, 223 – 232.

De Soete, W., Dewulf, J., Cappuyns, P., Van Der Vorst, G., Heirman, B., Aelterman, W., Schoeters, K., Van Langenhove, H., 2013. Exergetic sustainability assessment of batch versus continuous wet granulation based pharmaceutical tablet manufacturing: a cohesive analysis at three different levels. Green. Chem. 15, 3039-3048.

Dhenge, R.M., Fyles, R.S., Cartwright, J.J., Doughty, D.G., Hounslow, M.J., Salman, A.D., 2010. Twin screw wet granulation: Granule properties. Chem. Eng. J. 164, 322-329.

Dhenge, R.M., Cartwright, J.J., Doughty, D.G., Hounslow, M.J., Salman, A.D., 2011. Twin screw wet granulation: Effect of powder feed rate. Advanced powder technology: Int.

J. Soc. Pow Tech., Japan. 22, 162-166.

Dhenge, R.M., Cartwright, J.J., Hounslow, M.J., Salman, A.D., 2012. Twin screw granulation: Steps in granule growth. Int. J. Pharm. 438, 20-32.

Diab, S., McQuade, D.T., Gupton, B.F., Gerogiorgis, D.I., 2019. Process design and optimization for the continuous manufacturing of nevirapine, an active pharmaceutical ingredient for HIV treatment. Org.Process Res.Dev. 23, 320-333.

Dülle, M., Özcoban, H., Leopold, C.S., 2019. The effect of different feed frame components on the powder behavior and the residence time distribution with regard to the continuous manufacturing of tablets. Int. J. Pharm. 555, 220-227.

Ellison, C., Ennis, B., Hamad, M., Lyon, R., 2008. Measuring the distribution of density and tabletting force in pharmaceutical tablets by chemical imaging. J. Pharm. Biomed.

Anal. 48, 1-7.

EMA, 2012. Committee for Medicinial products for human use (CHMP). Guideline on real time release testing (formerly guideline on parametric release). Available at https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-real-time-release-testing-formerly-guideline-parametric-release-revision-1_en.pdf. Retrieved 10 Feb 2021.

Engisch, W.E., Muzzio, F.J., 2012. Method for characterization of loss-in-weight feeder equipment. Powder Technol. 228, 395-403.

Engisch, W., Muzzio, F., 2015. Loss-in-Weight Feeding Trials Case Study: Pharmaceutical Formulation. J.Pharm. Innov. 10, 56-75.

Engisch, W., Muzzio, F., 2016. Using Residence Time Distributions (RTDs) to Address the Traceability of Raw Materials in Continuous Pharmaceutical Manufacturing. J. Pharm.

Innov. 11, 64-81.

Ervasti, T., Simonaho, S., Ketolainen, J., Forsberg, P., Fransson, M., Wikström, H., Folestad, S., Lakio S., Tajarobi, P., Abrahamsen-Alami, S., 2015. Continuous manufacturing of extended release tablets via powder mixing and direct compression.

Int. J. Pharm. 495, 290-301.

Ervasti, T., Niinikoski, H., Mäki-Lohiluoma, E., Leppinen, H., Ketolainen, J., Korhonen, O., Lakio, S., 2020. The comparison of two challenging low dose APIs in a continuous direct compression process. Pharmaceutics. 12, 279.

123

Escotet-Espinoza, M., Vadodaria, S., Singh, R., Muzzio, F., Ierapetritou, M., 2018.

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, 274–287.

Escotet-Espinoza, M., Moghtadernejad, S., Oka, S., Wang, Y., Roman-Ospino, A., Schäfer, E., Cappuyns, P., Van Assche, I., Futran, M., Ierapetritou, M., Muzzio, F., 2019. Effect of tracer material properties on the residence time distribution (RTD) of continuous powder blending operations. part I of II: Experimental evaluation. Powder Technol.

342, 744-763.

EU GMP Annex 17: Real Time Release Testing and Parametric Release. Available at https://www.gmp-compliance.org/files/guidemgr/2018_annex17_en.pdf. Retrieved 11 March 2021.

European Pharmaceutical Rewiev (EPR), 2018 Available at https://www.europeanpharmaceuticalreview.com/news/77293/pharmaceutical-manufacturing-outsourced/. Retrieved 10 Feb 2021

European Medicines Agency,2010. Guideline on the investigation of bioequivalence.

CPMP/EWP/QWP/1401/98/Rev 1.

European Medicines Agency, 2017. Guideline on manufacture of the finished dosage form. EMA/CHMP/QWP/245074/2015

Fagih, A.M., Mehrotra, A., Hammond, S.V., Muzzio, F.J., 2007. Effect of moisture and magnesium stearate concentration on flow properties of cohesive granular materials.

Int. J. Pharm. 336, 338 – 345.

Faure, A., Grimsey, I.M., Rowe, R.C., York, P., Cliff, M.J. 1999: Applicability of a scale-up methodology for wet granulation processes in Collette Gral high shear mixer granulators. Eur. J. Pharm. Sci. 8, 85 – 93.

Faure, A., York, P., Rowe, R.C., 2001. Process control and scale-up of pharmaceutical wet granulation processes: a review. Eur. J. Pharm. Biopharm. 52, 269-277.

Facco, P., Dal Pastro, F., Meneghetti, N., Bezzo, F., Barolo, M., 2015. Bracketing the design space within the knowledge space in pharmaceutical product development. Ind. Eng.

Chem. Res. 54, 5128-5138.

FDA, 2004: PAT, a framework for innovative pharmaceutical development, manufacturing and quality assurance. Available at https://www.fda.gov/regulatory-

information/search-fda-guidance-documents/pat-framework-innovative-pharmaceutical-development-manufacturing-and-quality-assurance. Retrieved 9 Feb 2021.

FDA, 2007. Draft guidance for industry an review staff. Target product profile – a strategic development process tool. Available at https://www.govinfo.gov/content/pkg/FR-2007-03-30/pdf/E7-5949.pdf. Retrieved 10 Feb 2021.

FDA 2019: Quality considerations for continuous manufacturing. Available at

https://www.fda.gov/regulatory-information/search-fda-guidance-documents/quality-considerations-continuous-manufacturing. Retrieved 9 Feb 2021.

124

FDA 2020a. Drug shortages: Root causes and potential solutions 2019. Available at https://www.fda.gov/media/131130/download. Retrieved 9 Feb 2021.

FDA 2020b. CFR – Code of Federal regulations Title 21. Available at https://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfcfr/CFRSearch.cfm?fr=210.3.

Retrieved 10 Feb 2021

Fonteyne, M., Vercruysse, J., De Leersnyder, F., Van Snick, B., Vervaet, C., Remon, J.P., De Beer, T., 2015. Process Analytical Technology for continuous manufacturing of solid-dosage forms. Trends in Anal. Chem. 67:159-166.

Galbraith, S.C., Cha, B., Huang, Z., Park, S., Liu, H., Meyer, R.F., Flamm, M.H., Hurley S, Zhang-Plasket, F., Yoon, S., 2019. Integrated modeling of a continuous direct compression tablet manufacturing process: A production scale case study. Powder Technol. 354, 199-210.

Gao, Y., Vanarase, A., Muzzio, F., Ierapetritou, M., 2011. Characterizing continuous powder mixing using residence time distribution. Chem. Eng. Sci. 66, 417-425.

Gao, Y., Muzzio, F.J., Ierapetritou, M.G., 2012. A review of the Residence Time Distribution (RTD) applications in solid unit operations. Powder Technol. 228, 416-423.

Garr, J.S.M., Rubinstein, M.H., 1991. An investigation into the capping of paracetamol at increasing speeds of compression. Int. J. Pharm. 72, 117-122.

Garr, J.S.M., Rubinstein, M.H., 1992. The influence of moisture content on the consolidation and compaction properties of paracetamol. Int. J. Pharm. 81, 187-192.

Gea, 2020: Getting medicines to patients faster with POD-based mini factoried. Available at https://www.gea.com/en/customer-cases/getting-medicines-to-patients-faster-with-POD-based-mini-factories.jsp. Retrieved 10 Feb 2021.

Gérardy, R., Emmanuel, N., Toupy, T., Kassin, V. E., Tshibalonza, N. N., Schmitz, M., Monbaliu, J-C. M., 2018. Continuous flow organic chemistry: Successes and pitfalls at the interface with current societal challenges. Eur. J. Org. Chem. 20-21, 2301-2351.

Gonnissen, Y., Gonçalves, S.I.V., De Geest, B.G., Remon, J.P., Vervaet, C., 2008. Process design applied to optimise a directly compressible powder produced via a continuous manufacturing process. Eur. J. Pharm. Biopharm. 68, 760-770.

Gorringe, L.J., Kee, G.S., Saleh, M.F., Fa, N.H., Elkes, R.G., 2017. Use of the channel fill level in defining a design space for twin screw wet granulation. Int. J. Pharm. 519, 165-177.

Grote, S., Kleinebudde, P., 2018. Impact of functionalized particle structure on roll compaction/dry granulation and tableting of calcium carbonate. Int.J. Pharm. 544, 235-241.

Grundemann, L., Gonschorowski, V., Fischer, N., Scholl, S., 2012. Cleaning waste minimization for multiproduct plants: transferring macro batch to micro conti manufacturing. J. Clean. Prod. 24, 92-101.

125

Gupta A., Hamad M., Tawakkul M., Sayeed V., Khan M., 2009. Difference in the lubrication efficiency of bovine and vegetable- derived magnesium stearate during tableting. AAPS Pharm. Sci. Tech. 10, 500 – 504.

Hadžović, E., Betz, G., Hadžidedić, Š., El-Arini, S., Leuenberger, H., 2011. Investigation of compressibility and compactibility parameters of roller compacted Theophylline and its binary mixtures. Int. J. Pharm. 416, 97 – 103.

Hamzah, M., Beakawi Al-Hashemi, Omar, S., Baghabra Al-Amoudi, 2018. A review on the angle of repose of granular materials. Powder Technol. 330, 397 – 417.

Hanson, J., 2018. Control of a system of loss-in-weight feeders for drug product continuous manufacturing. Powder Technol. 331, 236-243.

He, X., Secreast, P. J., Amidon, G. E., 2006. Mechanistical study of the effect of roller compaction and lubricant on tablet mechanical strength. J. Pharm. Sci. 96, 1342 – 1355.

Hertel, M., Schwarz, E., Kobler, M., Hauptstein, S., Steckel, H., Scherließ, R., 2018. Powder flow analysis. A simple method to indicate the ideal amount of lactose fines in dry powder inhaler formulations. Int. J. Pharm. 535, 59 – 67.

Herting, M.G., Kleinebudde, P., 2008. Studies on the reduction of tensile strength of tablets after roll compaction/dry granulation. Eur. J. Pharm. Biopharm. 70, 372-379.

Hetrick, E., Shi, Z., Barnes, L., Garrett, A., Rupard, R.G., Kramer, T., Cooper, T.M., Myers, D., Castle, B., 2017. Development of Near Infrared Spectroscopy-based Process Monitoring Methodology for Pharmaceutical Continuous Manufacturing Using an Offline Calibration Approach. Anal. Chem. 89, 9175-9183.

Hiestand, E.N., 1997. Mechanical properties of compacts and particles that control tableting success. J. Pharm. Sci. 86, 985-990.

Holt, D., 2018. In oral presentation: The QbD journey from development to launch and beyond. EuPAT9. Manchester, UK, 15.-16.5.2018

Hopkins, M., 2006. Loss in weight feeder systems. Available at https://journals.sagepub.com/doi/pdf/10.1177/002029400603900801. Retrieved 10 Feb 2021.

Horsthuis, G.J.B, van Laarhoven, J.A.H., van Rooij, R.C.B.M., Vromans, H., 1993. Studies on upscaling parameters of the Gral high shear granulation process. Int. J. Pharm. 92, 143 – 150.

Hussain, M.S.H., York, P., Timmins, P., 1992. Effect of commercial and high-purity magnesium stearates on in-vitro dissolution of paracetamol DC tablets. Int. J. Pharm.

78, 203-207.

Ingvarsson, P. T., Yang M., Mulvad H., Nielsen, H, M., Rantanen, J., Foged, C., 2013.

Engineering of an inhalable DDA/TDB liposomal adjuvant: A Quality-by-Design Approach towards optimization of the spray drying process. Pharm. Res. 30, 2772 – 2784.

International Conference on Harmonisation, Q8 (R2) Pharmaceutical Development, 2009.

Available at

126

http://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Quality/Q 8_R1/Step4/Q8_R2_Guideline.pdf. Retrieved 10 Feb 2021.

International Conference on Harmonisation, Q9 Quality Risk Management, 2005.

Available at

http://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Quality/Q 9/Step4/Q9_Guideline.pdf. Retrieved 10 Feb 2021.

International Conference on Harmonisation, Q10 Pharmaceutical Quality system, 2015.

Available at https://www.ema.europa.eu/en/documents/scientific-

guideline/international-conference-harmonisation-technical-requirements-registration-pharmaceuticals-human_en.pdf. Retrieved 9 Feb 2021.

International Conference on Harmonisation, Q13 EWG, Continuous manufacturing of drug substances and drug products, 2018. Available at

International Conference on Harmonisation, Q13 EWG, Continuous manufacturing of drug substances and drug products, 2018. Available at