(673f) Plantwide Dynamic Simulation and Model-Based Control of a Continuous Pharmaceutical Process | AIChE

(673f) Plantwide Dynamic Simulation and Model-Based Control of a Continuous Pharmaceutical Process

Authors 

Gerogiorgis, D. I. - Presenter, Massachusetts Institute of Technology (M.I.T.)


ABSTRACT

Continuous
Pharmaceutical Manufacturing (CPM)
constitutes a groundbreaking paradigm in modern pharmacopeia:
multinational pharmaceutical industries face unprecedented profitability gaps, due to decreasing drug
prices, increasing Research & Development (R&D) and operating
expenditures. These business challenges emerge in the era of globalization  and
can be attributed predominantly (but not exclusively) to the transformation of
patent legislation, the vast reduction of patent life, the harmonization of
regulatory specifications and the advent of generics; new plants must rely on
optimization, standardization and modularization ideas (Behr et al., 2004).

Batch pharmaceutical processes are widespread, forming the overwhelming majority of
installed manufacturing plants; nevertheless, most are based on suboptimal
recipes focused on conforming with the strict licensing requirements for drug
product purity, stability and efficacy (ICH, 2009). Continuous
pharmaceutical processes
, though, need rely on systematic (not empirical)
process understanding and R&D, therefore necessitating the employment of
process modeling towards the articulation of Design Space (Yu, 2008) and
the use of Process Analytical Technology (PAT) (Munson, 2006); this may
turn out a time-consuming endeavor for novel manufacturing lines. The U.S. Food
& Drug Administration (FDA) thus spearheads the pursuit of CPM initiatives,
recognizing their potential to reduce operating cost at high quality (FDA,
2008) as confirmed in comparative technical (Betz et al., 2003) and modeling
(Gerogiorgis & Barton, 2009) studies. 

Modern batch
manufacturing processes are frequently operated at maximum unit efficiency via
dynamic optimization models, relying on mathematical rigor to capture critical
phenomena,  empirically or mechanistically (Chekhova et al., 2000). However,
process scale-up hinders model adaptation. Optimal external input profiles can
be computed and applied during each batch processed (low-level
regulatory control), and statistical methods for offline quality assessment
(QA) can reveal deviation trends (high-level supervisory control). Product
quality and uniformity are evaluated a posteriori: often, drug
substance (DS) or drug product (DS) lots must be discarded due to their poor
Critical Quality Attributes (CQA). The characteristic batch plant time for a
given lot size is the sum of processing and idle times required for its
production.

Continuous
manufacturing lines, in contrast to batch tank farms, eliminate idle times
between processing units by definition; however, deeper understanding is
required for their operation. Plantwide dynamic model building requires
significant effort  and parameter estimation implies arduous, costly experiments
which are not a likely luxury if rapid launching is of the essence. The
characteristic timescale for a novel CPM plant of given capacity is harder to
define, since it depends on structural complexity, unit interdependence,
recycles, rate data and timescale spectra. Benign (dilute) or challenging
(dense) flow chemistries and multicomponent mixtures affect reaction and
separation times, as pointed out in a CPM miniplant study (Roberge et al.,
2008). Online sensing and manipulation is possible via modern PAT sensors and
actuators, but empirical models can neither explain nor predict how local
disturbances may affect plantwide quality performance (Ward et al., 2010), as
fundamental phenomena are often poorly understood.  CPM plantwide dynamics,
therefore, remain a hitherto elusive (hence challenging) objective.

The present study focuses
on plantwide dynamics and control of a novel upstream (DS) process which is
aimed at the continuous production of a novel Active Pharmaceutical Ingredient
(API). The CPM flowsheet comprises 3 organic synthesis reaction and 4
separation steps; individual dynamic models for all respective unit operations
are proposed on the basis of experimental data. Open-loop plantwide process
simulation in MATLAB (Simulink) allows the determination of  timescales,
illustrating how common feed disturbances propagate to CQA (e.g. product
quality). Control loop pairings can be identified by implementing a RGA method
(Skogestad, 2005) and closed-loop plantwide process simulations provide insight
into their efficiency and robustness.

REFERENCES

Behr, A., Brehme, V.A., Ewers, C.L., Grön, H., Kimmel,
T., Küppers, S., Symietz, I.,New developments in chemical
engineering for the production of drug substances. Eng. Life Sci.
4(1): 15-24 (2004).

Betz, G., Junker-Purgin, P.,
Leuenberger, H., Batch and continuous processing in the production of
pharmaceutical granules. Pharm. Dev. Technol. 8(3): 289-297
(2003).

Chekhova, E., Barton, P.I., Gorak, A.,
Optimal operation processes of discrete-continuous biochemical processes. Comput.
Chem. Eng.
24(2-7): 1167-1173 (2000).

European Medicines Agency (EMEA), Impurities:
Residual solvents
, VICH Topic GL18 (plus annexes), http://www.emea.europa.eu/pdfs/vet/vich/050299en.pdf
(2000).

Gerogiorgis, D.I., Barton, P.I., Steady
state optimization of a continuous pharmaceutical process. Computer-Aided
Chemical Engineering (Proceedings of PSE 2009)
27(1): 927-932,
Elsevier, Amsterdam (2009).

International Conference on
Harmonisation of Technical Requirements for Registration of Pharmaceuticals
(ICH), Guidance Documents Q8 (Pharmaceutical Development), Q9 (Quality
Risk Management), Q10 (Pharmaceutical Quality System)
, http://www.ich.org
(2010).

Munson, J., Stanfield, C.F., Gujral,
B., A review of process analytical technology (PAT) in the US pharmaceutical industry. Curr. Pharm. Anal. 2(4): 405-414 (2006).

Roberge, D.M., Zimmermann, B., Rainone,
F., Gottsponer, M., Eyholzer, M., Kockmann, N., Microreactor technology and
continuous processes in the fine chemical and pharmaceutical industry: Is the
revolution underway?. Org. Proc. Res. Dev. 12(5): 905-910 (2008).

Skogestad, S., Postlethwaite I., Multivariable feedback control: Analysis & design. Wiley, Chichester (2005).

U.S. Food and Drug Administration
(FDA-CDER), Innovation & continuous improvement in pharmaceutical
manufacturing
, www.fda.gov/cder/gmp/gmp2004/manufSciWP.pdf
(2008).

Ward, J.D., Yu,C.-C., Doherty, M.F., Plantwide
dynamics and control of processes with crystallization, Comput. Chem. Eng. 34(1): 112-121 (2010).

Yu, L.X., Pharmaceutical Quality by
Design: Product and process development, understanding, and control. Pharm.
Res.
25(4): 781-791 (2008).

Topics