Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Computers and Chemical Engineering 42 (2012) 15–29

Contents lists available at SciVerse ScienceDirect

Computers and Chemical Engineering


journal homepage: www.elsevier.com/locate/compchemeng

A perspective on PSE in pharmaceutical process development and innovation


Krist V. Gernaey ∗ , Albert E. Cervera-Padrell, John M. Woodley
Department of Chemical and Biochemical Engineering, Technical University of Denmark, Building 229, DK-2800 Kgs., Lyngby, Denmark

a r t i c l e i n f o a b s t r a c t

Article history: The pharmaceutical industry is under growing pressure to increase efficiency, both in production and in
Received 21 September 2011 process development. This paper discusses the central role of Process Systems Engineering (PSE) methods
Received in revised form 26 February 2012 and tools in pharmaceutical process development and innovation, and searches for answers to questions
Accepted 28 February 2012
such as: Which PSE methods can be applied readily? Where is more method development needed? The
Available online 8 March 2012
paper covers key subjects for development of economically and environmentally sustainable pharmaceu-
tical processes, including Process Analytical Technology in its broadest sense, continuous pharmaceutical
Keywords:
manufacturing and green processes, and is illustrated with a series of short examples taken from the
Continuous pharmaceutical manufacturing
Design space
literature and ongoing research projects.
PAT © 2012 Elsevier Ltd. All rights reserved.
Pharmaceutical production
Process development
Sustainability

1. Introduction In this paper, the focus is on production of small-molecule


(MW < 1000) drug substances – active pharmaceutical ingredi-
Today more than ever, the pharmaceutical industry is under ents (APIs), new chemical entities (NCEs) – that are produced via
pressure to deliver its end products cheaper, faster and more organic synthesis. A generic process flowsheet of a small-molecule
efficiently (Federsel, 2003). Competition from generic drug man- API production process is provided in Fig. 1. Aside from antibi-
ufacturers is undoubtedly one of the main drivers, and has otics, which are small molecules that for many decades have been
already triggered dramatic changes. Indeed, many of the traditional produced by fermentation, there is an emerging trend toward
research-based pharmaceutical companies have started the pro- bio-production methods (such as enzyme-based catalysis). Inter-
duction of generic drugs during the past five years, and are thus estingly, several principles and concepts that will be highlighted in
in direct competition with generic drug manufacturers. Such com- the manuscript can also be applied to large-molecular (MW > 1000),
petition demands particular focus on production cost. In addition, biotechnology-based drug substances – also called biopharmaceu-
there is – in particular in Europe – a growing consumer demand ticals – which includes for example monoclonal antibodies and
for so called ‘green’ pharmaceutical products (i.e. the introduction therapeutic proteins.
of greener, more environmentally friendly production methods to Traditionally, the pharmaceutical industry has been rather
manufacture pharmaceuticals). The latter demand originates from reluctant to introduce innovative methods in its production pro-
a growing awareness that drug manufacturing processes have a cesses, despite the fact that the cost for the development of a
significant environmental impact (Sheldon, 1992, 2007). new drug continually increases, while the patent life time is fixed
(Federsel, 2003). Considering the typical development cycle of a
drug, two obvious ways of increasing profit are: (1) more rapid
Abbreviations: ANN, artificial neural network; API, active pharmaceutical process development, and thus maximization of the time between
ingredient; CAMD, computer-aided molecular design; CFD, computational fluid product release and patent expiration; and (2) streamlining the
dynamics; CP, continuous processing; CPM, continuous pharmaceutical manufac- full-scale production system to reduce production costs, such that
turing; CPP, critical process parameter; CQA, critical quality attribute; DEM, discrete
traditional research-based drug manufacturers can also compete
element method; DoE, design of experiments; FEM, finite element method; iPLS,
interval PLS; LCA, life cycle assessment; LLE, liquid–liquid extraction; MSPC, multi- with generic drug manufacturers when the patent of a drug has
variate statistical process control; MW, molecular weight; NCE, new chemical entity; expired. In both cases, we are convinced that increased and sys-
NIR, near infra-red; OED, optimal experimental design; PAT, Process Analytical Tech- tematic use of Process Systems Engineering (PSE) methods (Klatt &
nology; PBM, population balance model; PCA, Principal Component Analysis; PDE,
Marquardt, 2009; Stephanopoulos & Reklaitis, 2011) can be advan-
partial differential equation; PLS, Partial Least Squares; PMI, process mass intensity;
PSE, Process Systems Engineering; QbD, quality by design.
tageous. In this context, the introduction of the Process Analytical
∗ Corresponding author. Tel.: +45 45252970; fax: +45 45932906. Technology (PAT) guidance by the FDA (2004) is an important mile-
E-mail address: kvg@kt.dtu.dk (K.V. Gernaey). stone as well, since its publication has ended a long period of

0098-1354/$ – see front matter © 2012 Elsevier Ltd. All rights reserved.
doi:10.1016/j.compchemeng.2012.02.022
16 K.V. Gernaey et al. / Computers and Chemical Engineering 42 (2012) 15–29

Fig. 1. Typical flowsheet for a pharmaceutical manufacturing process. PSE methods and tools can be used to obtain data, analyze, model, simulate, control and optimize
each unit operation. Detailed information about specific unit operations for the formulation part of the process is provided elsewhere (feeding: Boukouvala et al., 2010;
mixing/blending: Boukouvala et al., 2010; roller compaction: am Ende et al., 2007; granulation: Vervaet & Remon, 2005; drying: Mortier et al., 2011; milling: Bilgili & Scarlett,
2005; tablet compaction: Michaut et al., 2010; coating: Prpich et al., 2010).

regulatory uncertainty. Indeed, the PAT guidance makes clear that production processes for small molecules. Finally, in the discus-
regulatory bodies are in favor of more efficient production meth- sion, we highlight several problems related to the application of
ods, as long as a safe (in-specification) product can be guaranteed. PSE methods and tools in pharmaceutical process development and
The disappearance of such regulatory uncertainty opens up new innovation, which also includes a short perspective on the function
and exciting possibilities for innovation in pharmaceutical produc- of PSE methods in the drug development cycle.
tion processes, and thereby also increased need for PSE methods
and tools. 2. Process Analytical Technology (PAT)
This paper will provide a perspective on the use of PSE methods
in pharmaceutical process development and innovation. It searches
Process Analytical Technology (PAT) (FDA, 2004) is undoubtedly
for the answers to questions such as: Which PSE methods can
one of the most influential new trends in pharmaceutical manufac-
be applied readily? Where is more method development needed?
turing in recent years. Interestingly, seen from the perspective of
The paper will be illustrated with examples taken from the liter-
the PSE community, PAT does not really bring so much new, since
ature and ongoing research projects. Rather than providing a long
the concepts described in the PAT guidance have been applied for
list of PSE methods and tools along with potential applications of
quite a long time by several other industries (e.g. petrochemical,
those methods, the manuscript has been structured according to
polymer and chemical sectors) (Kourti, 2006). As a consequence,
a number of main topics which we think are of major importance
there are quite a number of PSE methods and tools that can be used
today: implementation of PAT strategies, continuous production
in a PAT context, resulting from developments that took place in
processes and green pharmaceutical processes. It is worth not-
other industries. Here, we will first introduce the most essential PAT
ing as well that a topic such as ‘continuous production processes’
terminology, then discuss the most important PSE tools that can be
was ranked as the top priority on the list of key green engineering
used for PAT, and afterwards shift focus to PAT implementation and
research areas that has recently been published as a result of the
especially the role that PSE plays in PAT implementation.
brainstorming and prioritization exercises of the American Chem-
ical Society (ACS) Green Chemistry Institute (GCI) Pharmaceutical
Roundtable (Jiménez-González et al., 2011). This confirms that a 2.1. PAT terminology
more efficient process nearly always results in a greener process,
and that PSE methods and tools can play a prominent role in paving Several new concepts were introduced in the PAT guidance
the way toward more efficient and innovative pharmaceutical (FDA, 2004). The most important ones are:
K.V. Gernaey et al. / Computers and Chemical Engineering 42 (2012) 15–29 17

Fig. 2. Parallelism between process control loops and the definition of the design space based on CQAs of the end product.

• The design space, which is defined as “the multi-dimensional limits. This information is used to define the design space, result-
combination of critical input variables and critical process param- ing in a larger space, compared to an open-loop system where raw
eters that lead to the right critical quality attributes” (FDA, 2004). materials are typically rejected when they do not meet the narrow
The term ‘critical’ should be interpreted as ‘having a significant specifications. Indeed, in principle the design space concept allows
influence on final product quality’. The ICH Q8(R2) guideline on to adjust process operation – for example by means of automatic
pharmaceutical development provides examples of the design control – to compensate for a certain degree of variation in raw
space definition (ICH, 2009). Changing the process within the material properties: as long as CPPs and CQAs are kept within the
design space is therefore not considered as a change, and thus design space limits that were proposed and documented by the
does not require a regulatory post-approval of the process. This manufacturer when applying for regulatory approval of the pro-
opens up the possibility of increased use of optimization methods cess, it is expected that the resulting product will meet the defined
for pharmaceutical processes in the future (e.g. Nagy, Fujiwara, quality. An optimization algorithm can in addition be established
& Braatz, 2008). Those methods have been used for a long time in this large design space in order to meet process efficiency speci-
in for example the chemical industry (e.g. Bhatia & Biegler, 1996; fications (e.g. low energy use, highest yield, lowest operating cost,
Grossmann, 2005). etc.) by proposing changes to the values of the CPPs, inspired for
• A critical quality attribute (CQA) is defined as a physical, chemi- example by the case studies reported by Marchetti et al. (2005) and
cal, biological, or microbiological property or characteristic that Nagy et al. (2008).
should be within an appropriate limit, range, or distribution to
ensure the desired product quality (ICH, 2009). However, we
prefer the definition given by Kourti (2006), which considers 2.2. PAT tools
the multivariate nature of pharmaceutical production processes.
“CQAs should be understood as the combination of measurable The PAT guidance (FDA, 2004) distinguishes four classes of tools:
properties of an intermediate or final product that are considered (1) multivariate tools for design, data acquisition and analysis; (2)
critical for establishing the intended purity, efficacy, and safety of process analyzers; (3) process control tools; and (4) continuous
the product. That is, the value of the multivariate statistic calcu- improvement and knowledge management tools. In any of these
lated for these properties must be within a predetermined limit categories, one or several PSE methods and tools play a crucial role,
of a multivariate chart to ensure final product quality” (Kourti, as summarized also in Table 1.
2006).
• A critical process parameter (CPP) is a process parameter whose 2.2.1. Multivariate tools
variability has an impact on a CQA and therefore should be mon- Similar to pharmaceutical product development – formulation
itored or controlled to ensure the process produces the desired of the drug substance – pharmaceutical process development also
quality (ICH, 2009). requires multivariate approaches in order to capture the interac-
• Real-time release means that the product quality is assessed in tions between the different factors and their combined effects on
real-time based on on-line quantification of the CQAs. Imple- the final product quality in a satisfactory manner. The multivariate
menting a real-time release strategy should result in a leaner tools category is very broad. Only the most important aspects will
process – for example due to reduced laboratory work and be highlighted here.
reduced need for storage of intermediates and final products
– and stands in sharp contrast to the traditional ‘lab-centric’
approach to quality, where product quality was typically assessed 2.2.1.1. Chemometrics. Chemometric methods are generally used
off-line on the basis of often tedious laboratory analyses. in the pharmaceutical industry, and can be helpful tools in acquir-
ing process knowledge, as discussed in detail by Kourti (2006).
It is important here to distinguish between: (1) basic methods
The parallelism between process control loops and the defi- such as Partial Least Squares (PLS) (Wold, Sjöström, & Eriksson,
nition of the design space based on CQAs of the end product is 2001) and Principal Component Analysis (PCA); (2) slightly more
illustrated in Fig. 2. In this example, disturbance variables such advanced methods such as interval PLS (iPLS) (Nørgaard et al., 2000)
as variations in raw material properties x(t) are measured in real and the combination of genetic algorithms with backwards iPLS
time; output CQAs are also measured, directly or indirectly; and (Leardi & Nørgaard, 2004), methods which were both developed
process understanding has led to a predictive model f relating raw for chemometric variable selection; and, (3) advanced methods,
material properties and process parameters u to output properties such as non-linear versions of PCA and PLS (Dong & McAvoy, 1996;
y. Feed-forward and feed-back control loops ensure that output Kruger, Zhang, & Xie, 2008; Zhang, Martin, & Morris, 1997), and
quality attributes (CQAs) as well as CPPs remain within acceptable adaptive versions of PCA and PLS (Kadlec, Grbic, & Gabrys, 2011).
18
Table 1
Overview of the most relevant PSE tools that can be applied in a PAT context. Note that the references provided should be considered as examples. In the interest of space, it is not possible in this manuscript to provide an
exhaustive overview of the literature in each of these areas.

Tools class PSE method Status Review, theory Applications, examples

(1) Multivariate tools Chemometrics, basic methods (PCA, PLS) Well-accepted, generally used in Wold et al. (2001), Reich (2005), Kourti Rosas et al. (2011b), Cervera-Padrell et al. (2012)
industry (2006)

K.V. Gernaey et al. / Computers and Chemical Engineering 42 (2012) 15–29


Chemometrics, advanced methods Developed, used in academia, not Kruger et al. (2008), Kadlec et al. (2011) Dong and McAvoy (1996), Zhang et al. (1997), Nørgaard et
so much in industry al. (2000), Leardi and Nørgaard (2004)
Mechanistic models Well developed, widely used in Asprey and Macchietto (2000), Pordal ODEs: Dassau et al. (2006), Sin et al. (2008), Sin et al.
academia, increasingly used in et al. (2002), Sales-Cruz and Gani (2009), Singh et al. (2009);
industry (2003), Marquardt (2005), Kremer and PBM: Bilgili and Scarlett (2005), Fujiwara et al. (2005),
Hancock (2006) Wassgren and Curtis Aamir et al. (2010), Abdul Samad et al. (2011), Boukouvala
(2006), Franceschini and Macchietto et al. (2012), Capece et al. (2011), Krasnyk et al. (2012);
(2008), Gernaey and Gani (2010), DEM: Remy et al. (2009, 2010), Radeke et al. (2010), Adam
Mortier et al. (2011), Sharma et al. et al. (2011), Dubey et al. (2011);
(2011) CFD: Wei, Zhou, and Garside (2001), Wu et al. (2002), Woo
et al. (2009)
Reduced order models Well developed, especially in Krasnyk et al. (2012)
academia
Hybrid models Well developed, widely used in Akkisetty et al. (2010)
academia, limited use in industry
Design of experiments (DoE) Well developed, generally used in Eriksson et al. (2008) Mandenius and Brundin (2008), Koch et al. (2009),
academia and industry Boukouvala et al. (2010), Boukouvala, Ramachandran, et al.
(2011)

(2) Process analyzers Soft sensors, data-driven Theory well developed, used in Nomikos and McGregor (1995), Kourti (2006), Zamprogna et al. (2004), Camacho et al.
academia, sporadically used in Venkatasubramanian, Rengaswamy, (2008)
industry Kavuri, and Yin (2003); Bersimis et al.
(2006), Kadlec et al. (2009)
Soft sensors, model-driven Theory well developed, used in Kadlec et al. (2009), Villez et al. (2011)
academia, sporadically used in Venkatasubramanian et al. (2003a)
industry
(3) Process control Basic control (SISO, PID, feedforward) Theory well developed, Seborg et al. (2010) Cervera-Padrell et al. (2012)
well-accepted, generally used
Advanced methods Theory well developed, specific Nagy et al. (2008), Hermanto et al. (2011)
applications in industry
(4) Knowledge management Ontologies, knowledge bases Need for more method Venkatasubramanian et al. (2006), Morbach et al. (2007),
development Marquardt et al. (2010), Singh et al. (2010a)
K.V. Gernaey et al. / Computers and Chemical Engineering 42 (2012) 15–29 19

The most relevant applications of these non-linear methods are in Agar, & Turek, 2007; Mills, Quiram, & Ryley, 2007). CFD is also
process monitoring and fault detection (e.g. Dong & McAvoy, 1996). increasingly used in bio-based manufacturing in the frame of QbD
projects to demonstrate detailed process knowledge (Sharma,
2.2.1.2. Mechanistic models. A dynamic mechanistic model is an Malhotra, & Rathore, 2011). It is quite common to combine a CFD
excellent tool for mapping process knowledge in the context of PAT model with a model describing chemical reactions (e.g. Kashid
applications. The model equations can indeed represent a series of et al., 2007). However, nowadays the trend is clearly toward the
input–output relations, and can be extended whenever new pro- development of combined CFD-PBM models which can describe
cess knowledge becomes available. In this context, it is important to the change of distributed properties as a function of spatial coor-
formulate the dynamic models in a structured way (Gernaey & Gani, dinates within (part of) a unit operation. As an example, Woo, Tan,
2010; Sin, Ödman, Petersen, Eliasson Lantz, & Gernaey, 2008), since and Braatz (2009) combined CFD and PBM for the modeling of an
this also facilitates knowledge transfer. Once formulated in a struc- impinging jet crystallizer. Combining CFD and PBM is clearly an
tured way, a dynamic model can then be supplemented by a set area where the PSE community can make a major contribution to
of well-established model analysis tools (e.g. Asprey & Macchietto, development of detailed process understanding.
2000; Sales-Cruz & Gani, 2003; Marquardt, 2005), also including The discrete element method (DEM) is often used in the parti-
uncertainty and sensitivity analysis to assess the statistical qual- cle technology field for tracking of individual particles. Interactions
ity (reliability) of the simulated scenarios (Sin, Gernaey, & Eliasson between the particles and the fluid are neglected, and instead the
Lantz, 2009). Most important, however, is that these dynamic mod- focus is on description of particle–particle interactions (Radeke,
els are actively used for in-silico testing of a set of potential process Glasser, & Khinast, 2010). In order to produce useful results, a large
operating strategies, e.g. by comparing different control strategies number of particles need to be tracked, and as a consequence the
in a series of dynamic simulations, without disturbing the full-scale method is computationally rather expensive. Radeke et al. (2010),
process operation. In this way implementation of more advanced for example, simulated several millions of particles during the
control can be established more efficiently. operation of a mixer. Similarly, Remy, Khinast, and Glasser (2009),
The term “mechanistic model” is rather broad, since it covers Remy, Glasser, and Khinast (2010) and Dubey, Sarkar, Ierapetritou,
models consisting of systems of ordinary differential equations Wassgren, and Muzzio (2011) also applied the DEM method while
(ODEs), differential algebraic equations (DAEs) and partial differ- studying granular blending processes. Adam, Suzzi, Radeke, and
ential equations (PDEs). Khinast (2011) give a specific QbD example on the use of the DEM
Examples of mechanistic models based on ODEs can for example method within the definition of the design space for a blending unit
be found in Sin et al. (2008), Singh, Gernaey, and Gani (2009) and operation.
Zimmermann, Hennemann, Daußmann, and Kragl (2007), while
McMullen and Jensen (2011) demonstrate how to discriminate 2.2.1.3. Reduced order models and hybrid models. In many cases the
between different model candidates for such a system of ODEs. implementation of a detailed mechanistic model comes with a high
PDEs are increasingly applied to pharmaceutical production computational burden, for example in the case of a detailed 3D
processes as well. Population balance models (PBM) form one very CFD model of a unit operation. Of course, setting up such a detailed
important class of mechanistic models based on PDEs. They have model is beneficial since it also leads to a deeper understanding of
been applied frequently for the description of the dynamics of the the system. However, for many applications, for example related to
crystal size distribution in crystallization processes (e.g. Aamir, control of a specific operation, solving the problem in a computa-
Nagy, & Rielly, 2010; Abdul Samad, Singh, Sin, Gernaey, & Gani, tionally efficient way is of major importance. Here, development of
2011; Nagy et al., 2008), for the description of the particle size dis- a reduced order model is an appropriate solution. In fluid dynam-
tribution and the binder content in granulation processes (e.g. Poon ics problems, for example, it is quite common to derive a reduced
et al., 2009; Ramachandran, Immanuel, Stepanek, Litster, & Doyle, order model from a CFD model in order to reduce simulation times
2009) and blending operations (e.g. Boukouvala et al., 2012), and (e.g. Alonso, Velazquez, & Vega, 2009; Krasnyk, Mangold, & Kienle,
for the description of the particle size distribution in milling pro- 2010; Lang et al., 2009). Krasnyk, Mangold, Ganesan, and Tobiska
cesses (e.g. Bilgili & Scarlett, 2005; Capece, Bilgili, & Dave, 2011). (2012) demonstrated how to replace a detailed multi-dimensional
Other relevant PBM applications are for example the description of PBM of a lab-scale crystallizer by a reduced order model. We
the moisture content of particles during a drying process (see also expect that methods for generation of reduced order models will
Mortier et al., 2011), or even the description of the properties of a be increasingly applied within QbD projects in order to generate
population of microorganisms used for production of pharmaceu- fast predictions.
ticals via fermentation (Lencastre Fernandes et al., 2011). Hybrid models also form an attractive alternative in those
Computational fluid dynamics (CFD), based on the cases where a full mechanistic model is not available, and rely
Navier–Stokes equations describing the motion of fluids, is on the combination of a mechanistic model with a data-driven
another application of PDEs that is increasingly used in the phar- model component (e.g. Doyle, Harrison, & Crowley, 2003). It is
maceutical industry as well, and has for example been reviewed indeed too optimistic to assume that a detailed mechanistic model
by Kremer and Hancock (2006) and Wassgren and Curtis (2006). can be developed for each pharmaceutical process. Often, in the
Mortier et al. (2011) review specific CFD applications for fluid interest of time, a hybrid approach will be preferred, where the
bed drying processes. Pordal, Matice, and Fry (2002) suggested mechanistic part of the model is gradually extended when more
already 10 years ago that CFD would be useful in the pharma- process knowledge becomes available, e.g. during process develop-
ceutical industry for applications within mixing, solids handling, ment. Akkisetty, Lee, Reklaitis, and Venkatasubramanian (2010), for
separations (e.g. Wu, Wang, & Ching, 2002) and drying processes. example, report on the use of a neural network model for represent-
Furthermore, they specifically pointed toward the use of CFD ing the breakage function in a population balance model describing
to model equipment for heat transfer and heat generation, in the particle size distribution resulting from the processing of a par-
order to optimize performance. In addition to those areas, the ticulate material – three types of API ribbons – in a milling unit.
rapid development of microreactor technology – very relevant
for the pharmaceutical industry as well with the current focus 2.2.1.4. Design of experiments. Statistical design of experiments
on continuous production and process intensification – has also (DoE) is generally used in pharmaceutical process and product
benefitted substantially from the insights gained by using CFD for development, and is well-documented (e.g. Eriksson, Johansson,
the description of fluid flow, mass and heat transfer (e.g. Kashid, Kettaneh-Wold, Wikström, & Wold, 2008). Similar to chemometric
20 K.V. Gernaey et al. / Computers and Chemical Engineering 42 (2012) 15–29

methods, the general acceptance of DoE is promoted by the fact McGregor, 1995), were specifically developed for monitoring batch
that there are several commercial software products available that processes, and are quite frequently used due to the fact that
can be used to perform a DoE. The traditional way of using DoE is most traditional pharmaceutical production processes are based
based on an off-line design, followed by a series of experiments to on batch operations.
test a set of conditions of factors proposed in the DoE. However, Model-driven soft sensors can be based on mass or energy
current challenges in this area are especially related to the on-line balances (first principles approach). The main drawback is that
application of such a DoE in continuous flow systems, for example the development of such a model-driven soft sensor is gener-
at the microscale (Koch et al., 2009). ally considered to be rather time consuming (Kadlec et al., 2009).
When developing a pharmaceutical production process, time is
2.2.2. Process analyzers usually one of the most critical factors, and that is in our view
2.2.2.1. Spectroscopy. Methods such as PLS were developed in the one of the main reasons why such model-driven soft sensors
1970s and 1980s, and can be considered as generally accepted tools are not more widespread in the pharmaceutical industry. Alter-
in the pharmaceutical industry. The introduction of PAT has led natively, an algorithm such as a Kalman filter or an extended
to a tremendous increase in the number of spectroscopic appli- Kalman filter can also be used as a soft sensor, for example for
cations in the pharmaceutical industry, and as a consequence the fault detection (Villez, Srinivasan, Rengaswamy, Narasimhan, &
use of basic and slightly more advanced chemometric methods has Venkatasubramanian, 2011).
grown considerably. One of the factors explaining this growth is In conclusion, soft sensors have tremendous potential, for exam-
that several software products – commercial and open source – ple to exploit available on-line data to the maximum extent
have become available. Another factor is that spectroscopic meth- possible, but practical implementation on pharmaceutical produc-
ods such as near infrared (NIR) spectroscopy, to give one example, tion processes is not straightforward. One major challenge is the
is described in both the American and the European Pharmacopeia. time needed to develop a soft sensor. A second challenge might
Typical applications of NIR spectroscopy are (Reich, 2005): (1) iden- be validation by regulatory bodies, especially for more advanced
tification and characterization of raw materials and intermediates; data-driven or model-driven soft sensors. Last but not least, the lack
(2) analysis of solid dosage forms such as tablets; and (3) prediction of convincing or easily accessible examples does not promote the
of one or more variables in process streams or final product streams general acceptance of soft sensors in the pharmaceutical industry
(composition) on the basis of on-line, in-line or at-line spectro- either. Especially, with respect to this last point, we are convinced
scopic measurements. Other types of spectroscopy can of course that an advanced benchmarking tool for such algorithms, similar
be applied as well, but this manuscript is clearly not the place for to the Tennessee–Eastman challenge (Downs & Vogel, 1993) but
an extensive overview on spectroscopic methods. more focused on pharmaceutical production processes, could be
useful as a testbed to compare the performance of different algo-
2.2.2.2. Soft sensors. Soft sensors can be divided into two classes: rithms (benchmarking), and to demonstrate the benefits related to
data-driven and model-driven soft sensors (Kadlec, Gabrys, & the use of the different algorithms to potential end-users (training).
Strandt, 2009). Soft sensors can potentially be used for prediction
of unknown variables, as well as for fault detection and diag-
2.2.3. Process control tools
nosis. An extensive overview of methods for fault detection and
Similar to chemometric methods (see Section 2.2.1.1), one can
diagnosis can be found in Venkatasubramanian, Rengaswamy, Yin,
also distinguish between basic control methods (single input – sin-
and Kavuri (2003), Venkatasubramanian, Rengaswamy, and Kavuri
gle output, PID controller) (Seborg, Mellichamp, Edgar, & Doyle,
(2003), Venkatasubramanian, Rengaswamy, Kavuri, and Yin (2003).
2010) and advanced methods such as adaptive control or model
Chemometric models clearly belong to the first class of soft sen-
predictive control. Basic control methods can be assumed to be
sors, and can indeed be applied to traditional process data as well,
industry standard, and are generally used. However, a new chal-
for example for prediction of the composition of mixtures or con-
lenge here with the introduction of PAT is the incorporation of
centrations that are difficult to measure on-line (e.g. Camacho, Pico,
advanced on-line measurements, for example spectroscopic mea-
& Ferrer, 2008; Kadlec et al., 2009; Zamprogna, Barolo, & Seborg,
surements (e.g. Cervera-Padrell et al., 2012), in such a basic control
2004). Of course, there are other data-driven soft sensors as well,
loop rather than a standard temperature, flow rate or pressure mea-
and an exhaustive overview of the data-driven methods frequently
surement.
used in soft sensors in the process industries was recently provided
Advanced control methods are increasingly used, and the devel-
by Kadlec et al. (2009). Specifically for application of data-driven
opment of new applications and case studies is often driven by
soft sensors to pharmaceutical production processes, it is important
academia (e.g. Hermanto et al., 2011). What industry needs in order
to realize that software sensors based on PLS or PCA are in fact to be
to also adopt advanced control is a number of convincing case stud-
preferred, since these methods are well-known in the pharmaceu-
ies which clearly demonstrate the economical benefit of including
tical industry which facilitates validation. Both methods are based
more advanced control.
on latent variables, and examining the scores and loadings (PCA,
PLS) or the weights (PLS) is excellent for data exploration as well
as to identify unusual or abnormal process behavior (Kourti, 2006). 2.2.4. Continuous improvement and knowledge management
Multivariate statistical process control (MSPC) (Bersimis, Psarakis, tools
& Panaretos, 2006; Kourti, 2006), often based on PCA or PLS, cov- Due to the complexity of the problems to be addressed in
ers a series of well-established methods and tools for monitoring pharmaceutical product-process design, an efficient and system-
a process that is influenced by several variables of interest. Other atic knowledge base coupled with an inference system is essential.
types of data-driven soft sensors, for example based on artificial Morbach, Yang, and Marquardt (2007) presented an overview of
neural networks (ANN), can also be useful, but in practice the gen- OntoCAPE, a general ontology for structuring knowledge in the
eral opinion in industry is that it is difficult to have such an ANN chemical process engineering field. Details of this ontology can be
accepted by the regulatory bodies. found in Marquardt, Morbach, Wiesner, and Yang (2010). Singh,
Monitoring of batch and continuous processes is fundamen- Gernaey, and Gani (2010a) described an ontological knowledge
tally different, in the sense that batch processes have an additional based system for selection of process monitoring and analysis tools
dimension due to batch to batch variability (Kadlec et al., 2009). in the frame of PAT system design. An ontological framework to
Multi-way methods, for example multi-way PLS (Nomikos & support product and process development in the pharmaceutical
K.V. Gernaey et al. / Computers and Chemical Engineering 42 (2012) 15–29 21

simple kinetic model. Assuming that the kinetics are transferable


across scales, they can be extended with appropriate constitutive
equations to describe the relevant transport processes at each scale
of operation (lab, pilot, production). Advanced on-line data, for
example collected during pilot-scale experiments with the process
to be developed, often yields dynamic data which can be used for
model validation.
Crystallization, a key operation in most pharmaceutical pro-
cesses, forms a good example to illustrate how process knowledge
incorporated into models has been exploited to further improve
the process. The increased understanding of the crystallization
Fig. 3. The central role of PSE to simultaneously address process design for contin- process has led to the use of first-principles based approaches
uous pharmaceutical manufacturing, design space definition and monitoring and
for crystallizer design (Fujiwara, Nagy, Chew, & Braatz, 2005),
control using process analytical technologies. Synergy is obtained when the three
design problems are tackled using central mechanistic modeling. and first-principles based models are increasingly used in the
design of advanced control strategies for crystallization (Nagy et
al., 2008). Within the crystallization field, an important trend is
manufacturing domain was developed by Venkatasubramanian et now to actively influence the crystal size distribution and the crys-
al. (2006). tal morphology. As an illustration of that, Chen and Trout (2010)
Turning data into knowledge and managing that knowledge will recently reported the use of a computer-aided solvent selection
remain one of the major challenges for the future. In the pharma- procedure with the specific purpose of improving the morphol-
ceutical industry, storage of historical data is usually managed by an ogy of needle-like crystals. The method was successfully applied to
appropriate software product from an external supplier, and such 2,6-dihydroxybenzoic acid (DHB) to improve the crystal morphol-
software products are nowadays well established. However, even ogy. This type of applications opens up the possibility of rational
with such a software product in place, one does not necessarily design of crystals, in order to improve the quality of the formulated
possess the required process knowledge, for example in order to drug.
improve the performance of a process. Indeed, extraction of knowl- Real-time release, one of the key concepts in PAT, requires that
edge from data is the bottleneck, and should be one of the focus real-time monitoring tools are available and that they are used
points of future research. efficiently. When implementing PAT on a production process, it
is thus important to decide which variables should be monitored,
2.3. Implementation of PAT and which control loops are necessary in order to obtain a prod-
uct that has the required quality. Here also, PSE methods and tools
It is up to the drug producer to define the design space of a can point toward a solution. Indeed, Singh et al. (2009) provide an
particular pharmaceutical production process, and this definition example of the application of a model-based framework for design
should be based on process understanding, i.e. a detailed – prefer- and analysis of PAT systems. Singh, Gernaey, and Gani (2010b) have
ably mechanistic – understanding of the way in which critical input then created a systematic computer-aided framework to develop a
variables and process parameters will influence product quality. software (ICAS-PAT) for design, validation and analysis of PAT sys-
Tools such as DoE and chemometric models are frequently used for tems. The design of a PAT system involves the identification of the
the mapping of a design space (Boukouvala, Muzzio, & Ierapetritou, CQAs, selection of economical and reliable on-line measurement
2010; Rosas, Blanco, González, & Alcalá, 2011a, 2011b) of a pro- tools and integration of these on-line sensors with the control sys-
cess. Clearly, mapping of the design space of a PAT-assisted process tem (Singh et al., 2009, 2010b). The objective could for example be
increasingly justifies the development of mechanistic models of to obtain the predefined end product quality consistently, with the
the process as well, to capture the often non-linear relationship minimum instrumentation cost.
between process variables and final product quality (Dassau, Zadok,
& Lewin, 2006; Sin et al., 2008; Singh et al., 2009). Addressing
the uncertainties in the process, either on the basis of a mecha- 3. Development of continuous production processes
nistic model description (Sin et al., 2009) or using experimental
approaches (Paakkunainen et al., 2009), will also gain importance Conventionally, when transferring a production process for an
in order to obtain a prediction of the risk of not achieving final prod- API from lab-scale to production scale, the chemistry of the process
uct quality. Methods such as uncertainty analysis combined with has been modified to fit the process into existing batch reac-
local and global sensitivity analysis (Sin et al., 2009) can be helpful tor equipment, eventually leading to limited design possibilities
to direct experimental efforts, for example in designing experi- (Hessel, 2009). Despite adopting such a strategy, batch process-
ments that have focus on the collection of data that lead to those ing has demonstrated its flexibility and suitability, for example in
parameters that are most influential on the model outputs. accommodating very slow and/or heterogeneous chemistries. Nev-
Thus, according to us, PSE methods and tools will in general ertheless, it has been shown as well that continuous processing
play an increasingly important role in the definition of the design (CP), applied throughout all the stages of the life cycle of a drug
space, which is schematically illustrated in Fig. 3 as well. At the (Kang, Chung, Langer, & Khademhosseini, 2008; Webb & Jamison,
recent AIChE Annual Meeting 2010, several major pharmaceuti- 2010), provides an opportunity to reduce costs, footprint, energy
cal companies indeed confirmed this trend by indicating that they consumption, solvent utilization and environmental impact while
are developing mechanistic models of unit processes in order to improving quality and control of manufacturing processes and final
demonstrate process understanding. The drawback of the devel- products (LaPorte & Wang, 2007; Plumb, 2005; Pollet et al., 2009;
opment of a mechanistic model is that it is considered to be time Schaber et al., 2011). Compared to batch processes, continuous
consuming in comparison to data-driven approaches. Here, the key production typically implies a significantly smaller size of reactor,
challenge is to build and extend the model along the life cycle of the ideally leading to flexible unit operation building blocks that can
product, whenever new process knowledge becomes available. For be combined to create modular plants with advanced on-line sen-
example, detailed information on the kinetics of a reaction should sors and process control in order to guarantee product quality in
be available from lab-scale tests, and can be used to establish a real-time (Singh et al., 2011).
22 K.V. Gernaey et al. / Computers and Chemical Engineering 42 (2012) 15–29

Table 2
Summary of specific challenges for continuous pharmaceutical manufacturing (CPM) and related PSE methods and tools which may be used as drivers for innovation. Note
that many PSE methods and tools and CPM specific challenges are inter-related.

Specific challenges PSE methods and tools References

• Batch vs. continuous selection  Reaction kinetics Hartman et al. (2011)


 Heat transmission Schaber et al. (2011)
 Mass transfer Gerogiorgis and Barton (2009)
 Process flowsheets Cervera et al. (2011)
 Economic and environmental assessment

• Economic and environmental assessment  Process modeling Schaber et al. (2011)


 Process flowsheets Jimenez-Gonzalez et al. (2011)
 Simulation Carvalho et al. (2008, 2009)
 Property prediction
• Improved property prediction  Property prediction models and tools for O’Connell et al. (2009)
complex molecules and solutions
 Integration with process simulators
• New reactor and separation technologies  Mechanistic modeling for process Jensen (2001), Roberge et al. (2005)
understanding Hartman et al. (2009), Kralj et al. (2007)
 Property prediction Hartman and Jensen (2009)
Hessel (2009), Kockmann and Roberge, 2009

• Scale-up, scale-out  Reaction kinetics Kockmann et al. (2011)


 Heat transmission Barthe et al. (2008)
 Mass transfer Jensen (2001)
 Fluid dynamics Mendorf et al. (2010)
 CFD, DEM, and FEM Pordal et al. (2002)

• Process optimization  Mechanistic modeling Gerogiorgis and Barton (2009)


 Data-based (black-box) models Boukouvala et al. (2012), Boukouvala, Muzzio,
 Hybrid models et al. (2011), Boukouvala, Ramachandran, et al.
 Process flowsheets (2011)
 CFD, DEM, and FEM Dubey et al. (2011)

• Real-time monitoring  Multivariate analysis Kourti (2006), Kadlec et al. (2009)


 Multivariate statistical process control McMullen and Jensen (2010a, 2010b, 2011)
 Optimal experimental design
• Design space and real-time release  Mechanistic modeling García Muñoz et al. (2010)
 Data-based (black-box) models MacGregor and Bruwer (2008)
 Hybrid models Kourti (2006), Kadlec et al. (2009)
 Multivariate statistical process control
 Process control
 Model predictive control
• Formulation operations  Improved process understanding Schaber et al. (2011)
 Modeling Boukouvala et al. (2012), Boukouvala, Muzzio,
 Multivariate analysis et al. (2011), Boukouvala, Ramachandran, et al.
 CFD, DEM, FEM (2011)
 Process flowsheets for operations involving Dubey et al. (2011)
solids Singh et al. (2009, 2010b)
 Property prediction Mortier et al. (2011)

• Start-up, shut-down, operational issues (clogging)  Dynamic modeling Hartman, Naber, Buchwald, et al. (2010),
 Process control Hartman, Naber, Zaborenko, et al. (2010)
 Risk assessment Boukouvala, Ramachandran, et al. (2011)

• Continuous bio(catalytic)processes  Improved process understanding Sin et al. (2008, 2009)


 Mechanistic modeling Gernaey, Eliasson Lantz, Tufvesson, Woodley,
 Property prediction and Sin (2010)

• Multipurpose and multistep continuous production lines  Dynamic modeling Singh et al. (2011)
 Process flowsheets Kockmann et al. (2011)

• Plantwide dynamic models  Mechanistic models Boukouvala, Ramachandran, et al. (2011)


 Data-based (black-box) models
 Hybrid models
 Process flowsheets
 Property prediction

3.1. The role of PSE in the design of continuous (pharmaceutical) a reduction of solvent use and the number of solvent exchange
processes operations. Computer-aided methods and tools form the corner-
stone of the methodology, and can be used for flowsheet generation
When developing such continuous plants, it is evident that (Chakraborty & Linninger, 2002; Jaksland, Gani, & Lien, 1995),
property prediction and process design methods, which are both solvent selection and replacement (Gani, Gómez, Folić, Jiménez-
well-established in the PSE academic community (as well as in González, & Constable, 2008), as well as solvent integration (Ahmad
industry), will be required. Cervera, Gani, Kiil, Skovby, and Gernaey & Barton, 1999). Ideally, dynamic models of the system should be
(2011) propose a methodology for the development of a contin- constructed as well, and used in simulation-based scenario anal-
uous pharmaceutical manufacturing process, and apply it to the ysis for reactor design, for model-based economic evaluation and
transformation of a batch to a continuous manufacturing process. for environmental impact assessment. These models should ide-
The main benefits for a case study with zuclopenthixol, an API, are ally be mechanistic and constructed on the basis of experimental
K.V. Gernaey et al. / Computers and Chemical Engineering 42 (2012) 15–29 23

data, where possible. However, if no detailed reaction mechanism (Hartman & Jensen, 2009; Hartman, Naber, Buchwald, & Jensen,
is known, they can also be data-driven (Kourti, 2006; Kadlec et al., 2010; Sahoo, Kralj, & Jensen, 2007). Mass transfer and heat trans-
2009) or hybrid models (Kahrs & Marquardt, 2008). In our opinion, mission phenomena are enhanced thanks to a large surface area
a key requirement here is that all the available process knowledge to volume ratio, while capillary and surface tension effects may be
should, whenever possible, be incorporated in mechanistic models exploited in many applications (Hartman & Jensen, 2009; Jensen,
(Gernaey & Gani, 2010). One of the most important benefits offered 2001; Kralj et al., 2007; Roberge et al., 2005). Improvements in mix-
by computer-aided methods and tools relying on those models are ing and heat transmission as well as lower reaction volumes lead to
indeed the fact that one can obtain a rapid indication of the eco- novel process windows (Hessel, 2009), where reactions can be car-
nomic performance of different process options, which is helpful ried out under extreme conditions (Kockmann & Roberge, 2009),
in decision-making. for example at high temperature and pressure. Furthermore, new
We believe that PSE should have a central role in the design of chemical transformations (or already known chemistries which are
new continuous pharmaceutical processes or the retrofit of exist- incompatible with batch processing), reactions at high concentra-
ing batch processes (see Table 2). However, CP is a new field for the tion or under solvent-free conditions, and reactions in the explosive
pharmaceutical industry, where new challenges and opportunities or thermal runaway regime may become feasible (Hessel, 2009).
are constantly arising in different areas: reaction engineering, sep- PSE methods and tools will need to be developed further to address
aration technology, monitoring and control, and solids handling are these problems, where mechanistic models should play a central
only some of the engineering challenges that have been intensively role. For example, surface tension and capillary effects, which are
studied in recent years. Thus far, research has focused primarily in the basis for microscale liquid–liquid extraction (LLE) and distilla-
enabling these technologies, while a holistic view over the design of tion, should be thoroughly described and understood, assisting for
CP is still under development (an introduction to the design prob- example in membrane material selection or evaluating the feasibil-
lem can be found in Boukouvala, Ramachandran, Vanarase, Muzzio, ity of membrane based separations. While surface tension data is
& Ierapetritou, 2011; Cervera et al., 2011; Gerogiorgis & Barton, usually available for pure solvents, mixtures of solvents with par-
2009; Schaber et al., 2011). In the following we discuss some of tial miscibility and/or with dissolved solutes need to be evaluated
the specific challenges that PSE will need to address as a driver for experimentally on a “case to case” basis. Development of prop-
future innovation. erty prediction tools and rigorous models describing diffusion in
membrane assisted LLE and distillation would be highly beneficial.
3.2. Specific challenges and opportunities for PSE
3.2.3. Scaling-up and scaling-out of microscale CP
3.2.1. Batch or continuous? According to the microreaction paradigm as originally proposed,
The first question that arises when designing a pharmaceuti- the beneficial features of micro- reaction and separation technolo-
cal process is whether a specific unit operation should operate gies should be conserved across different scales of operation using
in batch mode or in continuous mode. Hartman, McMullen, and a scaling-out (or numbering-up) approach, i.e. replicating small
Jensen (2011) provide some clues to answer this question in units working in parallel (Jensen, 2001; Plumb, 2005; Roberge
the context of discovery and development in a laboratory. They et al., 2005). However, fluid distribution and parallelization of
propose a decision roadmap where the goal of the study (discov- microscale units is not such a straightforward task as is often
ery/screening/process development), the inherent characteristics claimed (Mendorf, Nachtrodt, Mescher, Ghaini, & Agar, 2010), and
of the chemistry (fast/slow, exothermic/endothermic), and the according to some authors it should even be kept as a last option
desired process conditions (easy to handle/extreme conditions) (Kockmann, Gottsponer, & Roberge, 2011). Furthermore, solids
are all taken into account to decide what the best experimental handling is still a major limitation for microreaction technology
setup should be. The decision is made based on non-dimensional (Hartman, Naber, Zaborenko, Buchwald, & Jensen, 2010). Therefore,
numbers summarizing information on reaction, mixing and heat scaling-up microreactions has been proposed as a way to maintain
transfer rates as well as on axial dispersion in flow reactors. While the advantages of operating at small length scales while being able
the decision tool they developed focuses on experimental studies, it to process large flow rates in realistic (challenging) scenarios with a
would be highly desirable to extend it to industrial scale processes. low capital cost. The design across different scales should be driven
Such decision-making methodologies should further include a by a thorough description of mass and heat transfer characteristics
detailed economic (Schaber et al., 2011) and environmental assess- (Barthe et al., 2008; Kockmann et al., 2011). Computational Fluid
ment, while considering monitoring and control, automation, Dynamics (CFD) could be useful to describe mixing behavior across
solvent recovery and heat integration. For example, Gerogiorgis and different scales (Pordal et al., 2002). Indeed, assuming that reaction
Barton (2009) demonstrated with a steady-state simulation of an kinetics can be determined at small scale and are independent of
upstream API synthesis process, that by using continuous process- scale, combining the reaction kinetics obtained for a specific pro-
ing (in this case using microreactors and microseparators) it was cess with a CFD code should allow in silico investigation on how
possible to reduce the flowsheet size, thereby also reducing fixed non-ideal mixing influences reaction performance at larger scales.
investment cost and suppressing feedstock and solvent invento-
ries (operating cost). The total effluent waste was found to be an 3.2.4. Monitoring and control – integration of flow chemistry and
order of magnitude lower than that generated by a more tradi- monitoring
tional batch manufacturing process. The waste could potentially be Continuous-flow reactions require on-line monitoring and feed-
reduced even further by solvent recycling, but this was not consid- back control in order to take full advantage of the real-time
ered, since an extra unit operation would be needed which would release PAT concept (FDA, 2004). Monitoring tools should provide
increase the fixed cost. information about both quality specifications and overall system
performance. McMullen and Jensen (2010b) have reviewed the
3.2.2. Continuous API synthesis – new paths enabled integration of microreactors with in-line physical sensors and
Microreactors and microstructured reactors (Jensen, 2001; analytical chemistry techniques, focusing on reaction monitoring
Roberge, Ducry, Bieler, Cretton, & Zimmermann, 2005) as well and development of algorithms for automated reaction screen-
as microseparators (Hartman, Sahoo, Yen, & Jensen, 2009; Kralj, ing and optimization (McMullen & Jensen, 2010a, 2010b, 2011).
Sahoo, & Jensen, 2007) have received a lot of attention in The integration of continuous flow reactors with in-line analyti-
recent years, enabling continuous flow multistep organic synthesis cal tools provides an opportunity to perform optimal experimental
24 K.V. Gernaey et al. / Computers and Chemical Engineering 42 (2012) 15–29

design (OED) (Atkinson & Donev, 1992) for model discrimination and solids processing requires further development and will even-
and kinetic parameter fitting in mechanistic models (McMullen & tually enable the simulation, design and optimization of continuous
Jensen, 2011). OED is performed in an automated fashion, reduc- pharmaceutical processes without the need to perform a consider-
ing the number of experiments to carry out and obtaining the able number of expensive trial and error experiments.
maximum amount of information. However, while this method
has been successfully applied to relatively simple kinetic models 3.2.7. Property prediction
(McMullen & Jensen, 2011), algorithms for more complex reac- Pharmaceutical process design needs predictive thermody-
tion mechanisms (typical of many pharmaceutical processes) need namic models to reduce the experimental search and focus
to be further developed, as well as unified methods to quantify laboratory work in the most promising areas. Predictive models
information content (Franceschini & Macchietto, 2008). like UNIFAC usually cannot be applied reliably to chemicals with
complex functionalities such as pharmaceuticals (O’Connell et al.,
3.2.5. Definition of design space 2009). The prediction of crystal structure and solid-state properties
The introduction of PAT and QbD in the pharmaceutical indus- (crucial for many of the most common pharmaceutical unit oper-
try encourages the design and validation of processes to be done ations) is still computationally demanding and far from generally
not only at one set of fixed processing conditions, but rather for a applicable (O’Connell et al., 2009). Furthermore, the pharmaceuti-
range of processing conditions known as the design space. Defining cal industry usually deals with complex chemistry, phase equilibria
a design space offers the possibility of responding to disturbances involving organic salts and aqueous electrolytes. Pharmaceutical
entering the process (e.g. variability in raw materials) through systems are thus still challenging for property prediction tools. A
establishment of appropriate control loops, maintaining the pro- property prediction package would also be desirable, integrating
cess operation under optimal conditions with a satisfactory product a database, a model library and a tool for parameter adoption. A
quality, without the need to revalidate the process when certain review of currently available property prediction tools is available
process parameters are manipulated. Hence, the acceptance of a (O’Connell et al., 2009), including many examples from the phar-
particular material is conditional on the values of the manipulated maceutical industry. A more detailed review of property prediction
variables calculated by the controller, if and only if the controller is methods is out of the scope of this manuscript. Specific examples
able to compensate for the variability in a stable and robust manner of property prediction methods applied to pharmaceuticals can be
(García Muñoz, Dolph, & Ward, 2010; MacGregor & Bruwer, 2008). found in Taskinen and Yliruusi (2003), Matsuda, Kaburagi, Kurihara,
Representing the complex relationships arising from an Tochigi, and Tomono (2010) and Diedrichs and Gmehling (2011).
integrated materials-process-product design space requires a
model-centric approach. García Muñoz et al. (2010) propose a 3.2.8. Drug product formulation operations
mathematical framework for feed-forward control using latent A new trend in continuous manufacturing of pharmaceuticals is
variable regression methods. A PLS model may be used to describe the introduction of continuous processes in the final formulation of
the relationships between materials and products, under all drug substances as well (Boukouvala, Ramachandran, et al., 2011;
expected processing conditions. To perform the control calcula- Plumb, 2005; Schaber et al., 2011; Vervaet & Remon, 2005). Indeed,
tions, first a regression model is built. This model uses a regressor there is increasing interest in for example continuous tabletting
that is composed of known variables (i.e. raw material proper- lines, both in academia and industry, where processes such as gran-
ties) and unknown variables (i.e. process manipulated variables), ulation, drying, compression and coating are integrated (Fig. 1).
and the final product quality as a response. When a new batch Tablets are one of the most common dosage forms of pharma-
of material is received, a control calculation is executed and the ceuticals. During the formulation of a tablet (see also Fig. 1 for
combination of the best process conditions for the properties of a schematic overview of the operations involved) the API is usu-
the new batch of material is found. The performance of the con- ally combined with excipients such as water, lactose, starch, sugar,
trol strategy can be back-propagated to illustrate that an improved and coloring agents. Here also, advanced mechanistic models can
controller implies the acceptance of a broader range of materials be helpful in equipment design and tuning, in defining the design
(García Muñoz et al., 2010). space as well as in the development of control strategies. However,
modeling of these processes is not straightforward, and devel-
3.2.6. Plant-wide dynamic models opment of a detailed process model often requires a multi-scale
A special challenge related to the operation and control of phar- approach. For a drying process, to give one example, a detailed
maceutical plants working in continuous mode is how to obtain description of the process requires a combination of a population
a strictly continuous flow throughout unit operations with very balance model (PBM) to track the dynamics of the distribution of the
different characteristic times, and still be able to respond to dis- moisture content of the particles, combined with a computational
turbances in the system in each operation. Another challenge fluid dynamics (CFD) code to describe the detailed spatio-temporal
is how to introduce on-line operation into an existing process. behavior of the moisture content in the gas phase of the system.
Clearly, it is very difficult to replace a complete production line The combined CFD-PBM should then allow the simulation of the
at once. The alternative is to integrate one continuous opera- spatio-temporal dynamics of the moisture content in the parti-
tion at a time in a process that is otherwise based on batch cles throughout the dryer. Development of such a PBM starts at
processing, thus slowly turning the batch process into continu- the individual particle level, i.e. one first needs to investigate the
ous operation. In general, there is a need to develop plant-wide drying of individual particles in order to develop an understand-
dynamic models able to describe start-up, shut-down, how to deal ing of the most important phenomena involved. A review on the
with clogging problems, and changing product lines in a mul- state of the art in modeling of fluidized bed drying processes of
tipurpose plant (Jiménez-González et al., 2011; Schaber et al., pharmaceutical granules can be found in Mortier et al. (2011). The
2011). Gerogiorgis and Barton (2009) performed a steady-state granular dynamics of continuous blending processes have recently
simulation of an upstream continuous pharmaceutical process been studied by Dubey et al. (2011) using discrete element methods
including microreaction and microseparation steps. Boukouvala, (DEM), and were compared with less computationally expensive
Ramachandran, et al. (2011) built a dynamic flowsheet model and PBM and data-based methods by Boukouvala et al. (2012), includ-
simulated an integrated continuous downstream pharmaceutical ing high-dimensional representations, response surface methods
process, including feeding, blending and granulation. Integrated and kriging (Boukouvala, Muzzio, & Ierapetritou, 2011). The objec-
modeling and simulation tools for microreaction, microseparation tive was to study the effect of a number of process parameters
K.V. Gernaey et al. / Computers and Chemical Engineering 42 (2012) 15–29 25

(e.g. impeller speed) on the mixing quality while ensuring the Jiménez-González, & Henderson, 2007). 56% of the PMI in a typical
right residence time distribution and amount of shear, which may pharmaceutical process originates from solvents, while 32% origi-
affect properties such as hydrophobicity and flowability that could nates from water use (note that unlike other sustainability metrics,
adversely affect downstream unit operations such as tabletting the PMI considers process water as well) (Jimenez-Gonzalez et al.,
(Dubey et al., 2011). There is in general still a lot of work to be 2011). The poor water-solubility of many APIs means that at several
done to further develop models, modeling strategies and solution points during API manufacture, organic solvents are required (usu-
methods for multi-phase systems (gas, solid, and liquid). Reklaitis ally for extraction or crystallization). The ACS GCI Pharmaceutical
(2007) identified specific process design problems that need to Roundtable recently produced an industry-wide solvent selec-
be addressed, such as the development of predictive models and tion guide, freely available on-line (www.acs.org/greenchemistry).
design spaces for a set of common unit operations used in pharma- While this guide considers safety, health and environmental
ceutical production: multiphase batch reactors, antisolvent based aspects of solvent selection, process design aspects need to be
crystallization, solid–liquid separation, size reduction, granulation, addressed with complementary methodologies. In this respect, sys-
lyophilization, and a variety of separation types, including sim- tematic computer-aided solvent selection methodologies (e.g. Gani
ulated moving beds. There is a need for systematic and reliable et al., 2008) can be very useful, in order to find alternative solvents
methods for scale-up/scale-down based on rigorous CFD, DEM and in an efficient way with minimum experimental work, or to come
finite element method (FEM) simulation models. Multivariate con- up with green solvents for new production processes, for example
trol systems need to be adapted, and different operational issues using computer-aided molecular design (CAMD) (e.g. Weis & Visco,
need to be solved (e.g. trend monitoring, incipient fault detection, 2010). Hsieh et al. (2009) recently presented a successful indus-
etc.). For continuous processing, process-wide automatic control trial case study demonstrating the use of solvent selection methods
and real time process optimization methods need to be developed in the context of conceptual design of an API production process,
further (Reklaitis, 2007). where a ternary solvent system was considered for a reaction,
extraction, distillation, and crystallization sequence. Chakraborty
and Linninger (2002) introduced combinatorial process syn-
4. Development of greener pharmaceutical processes
thesis, which in essence allows an optimal trade-off between
process economics and environmental impact during flowsheet
4.1. Design of sustainable pharmaceutical processes
synthesis.
Despite the large quantity of solvents used in the pharmaceuti-
In the last 5 years or so it has been clear that the pharmaceutical
cal industry, solvent recovery and recycling practices are not very
industry mostly requires synthetic routes to be environmentally
extended, due to the potential risk of cross-contamination and the
sustainable in addition to still fulfilling the demands of high
high solvent purity required (resulting in a high recovery cost). Cur-
product quality and process economics (Tucker, 2006). Different
rently, much of the solvent used for pharmaceutical production is
approaches to pollution prevention are possible, from at-source
discarded as waste and incinerated. For example, within GSK, on
reduction to end-of-pipe treatment (Linninger & Malcolm, 2006).
average less than 50% of solvent used was recycled and reused
Pollution prevention should be considered already in the concep-
by 2007 (Constable et al., 2007). Moving batch processes toward
tual design phase, since the regulatory (e.g. FDA) re-approval of a
continuous production opens up many possibilities for process
process is very expensive. There are nowadays different software
integration (Gerogiorgis & Barton, 2009), which should be sim-
tools for pollution prevention which help to quantify the envi-
pler to implement than in batch mode. Ahmad and Barton (1999)
ronmental implications of a process and generate suggestions for
presented a systematic design approach to the generation of batch
process modifications (e.g. SustainPro developed by Carvalho, Gani,
process flowsheets with integrated solvent recovery and recycling.
& Matos, 2008; Carvalho, Matos, & Gani, 2009; Shonnard, Kicherer,
Chakraborty and Linninger (2002) demonstrate the simultaneous
& Saling, 2003). A list of on-line manuals, databases of chemi-
optimization of structural decisions while finding the best oper-
cal impacts, decision-analysis tools, design options ranking tools,
ating parameters for an optimal sequencing of a solvent recovery
expert systems, process analysis tools (simulators), waste reduc-
system.
tion algorithms and finance tools can be found in Linninger and
Malcolm (2006).
4.3. Biocatalytic processes
Several metrics have been proposed in order to quantify
environmental sustainability and assist in the design of greener
The application of more efficient (or more selective) catalysts
processes (Constable, Curzons, & Cunningham, 2002; Curzons,
is another way to obtain a greener pharmaceutical production
Constable, Mortimer, & Cunningham, 2001; Saling et al., 2002).
process. In this respect, biocatalysis has the potential to deliver
Among them, the process mass intensity (PMI) (or its inverse, the
‘greener’ chemical syntheses (Pollard & Woodley, 2007; Woodley,
mass efficiency) was selected by the ACS Green Chemistry Institute
2008), since biocatalysts are more selective compared to tradi-
(GCI)’s Pharmaceutical Roundtable as the key metric for evaluation
tional catalysts, and will usually assist in the synthesis of optically
and benchmarking of sustainability (Jimenez-Gonzalez, Ponder,
pure molecules, which are essential for the manufacture of many
Broxterman, & Manley, 2011), since it provides a good estimation
pharmaceutical compounds (Savile et al., 2010). Moreover, bio-
of the life cycle assessment (LCA) and footprint of a process, while
catalysts, due to their biological origin, normally work well in
being fairly correlated to CO2 emissions.
water, although the recovery steps may still demand extrac-
tion with organic solvents (Henderson, Jiménez-González, Preston,
4.2. Solvent selection and recovery Constable, & Woodley, 2008). Furthermore, biocatalysis overcomes
the risk of using toxic metal based catalysts, which convention-
One way of achieving a greener production process is by eval- ally require significant effort to be removed to ensure a clean
uating solvent use, and by replacing solvents currently used in product (Reginato, Sadler, & Wilkes, 2011). The mild conditions
a pharmaceutical production process with greener, more envi- (ambient temperature and pressure) also reduce energy costs
ronmentally friendly solvents where possible (Jiménez-González, and improve selectivity via the minimization of side reactions
Curzons, Constable, & Cunningham, 2005). As an example, solvent (Henderson et al., 2008; Straathof, Panke, & Schmid, 2002). How-
use at GlaxoSmithKline was reported to account for between 80% ever, integrating one or several biocatalytic steps into an existing
and 90% of the mass utilization in a typical process (Constable, organic synthesis is often far from straightforward, due to e.g.
26 K.V. Gernaey et al. / Computers and Chemical Engineering 42 (2012) 15–29

the occurrence of product inhibition which leads to low prod- 5.2. PSE methods and the product life cycle
uct concentration and/or poor use of catalyst unless processes are
designed to overcome these problems (Tufvesson, Lima-Ramos, A key question when considering the use of PSE methods and
Nordblad, & Woodley, 2011b). PSE methods can provide a sys- tools during the development of a production process for an API is
tematic route to implementation of biocatalysis (Mandenius & when to start using these methods. Clearly, one will not harvest all
Brundin, 2008; Rohner & Meyer, 1995) and a framework for such potential benefits if PSE is only used in a late process design stage.
a scheme has recently been developed for transaminase–based Therefore, in our opinion, PSE methods and tools should be used
reactions for the synthesis of chiral amines – a particularly impor- throughout the product life cycle, to facilitate continuous learning
tant pharmaceutical target (Tufvesson, Lima-Ramos, Jensen, et al., and collection of process/product knowledge.
2011). Increasingly PSE methods are also being applied to assist Likewise, we could start assuming that a mechanistic process
in the identification of process bottlenecks via early stage eco- model is constructed early on in process development, based on
nomic analysis. The identification of process bottlenecks enables lab-scale data that were collected to characterize the reaction
suitable development in a timely manner. Finally the development kinetics. This model could then be used to increase available process
and implementation of biocatalytic processes can benefit substan- knowledge during the development phase while leading the path
tially from computer-aided process performance evaluation, based for experimental design, thus fully integrating modeling and lab-
for example on mechanistic understanding and modeling of the oratory research. Similarly, model transfer across different scales
specific reaction of interest. For example Blayer, Woodley, Lilly, and would be greatly facilitated. Last but not least, small-scale con-
Dawson (1996) and Chen and Woodley (2002) have used a model to tinuous production – designed, monitored and controlled using a
develop windows of operation to predict the ideal operating space variety of PSE methods and tools – can, in theory, lead to more
for the aldolase-catalyzed synthesis of N-acetyl-d-neuraminic acid rapid process development by scaling-out the production as a valid
from N-acetyl-d-mannosamine (see also Zimmermann et al., 2007). alternative to scaling-up.
An interesting review of the role of modeling in biocatalysis has
been recently published (Vasić-Rački, Findrik, & Vrsalović Presečki, 6. Conclusions
2011).
• PSE methods and tools are essential for successful implementa-
tion of Process Analytical Technology (PAT) based processes.
5. Future developments • PSE methods and tools play a key role in the development of
continuous manufacturing processes as well as development of
5.1. Development of generic solutions greener pharmaceutical production processes, two major trends
in pharmaceutical manufacturing.
While pharmaceutical production offers a large space for • Achieving the full benefit from PSE methods and tools is only
improvement in terms of technology, capital and operating costs possible by applying them during the entire life cycle of a phar-
and sustainability, for many years innovative solutions have been maceutical product and its production process.
difficult to implement. The pressure to develop processes rapidly • There are difficulties in transferring PSE methods from academia
(due to the limited patent life) used to outweigh the added benefits, to industry (see Table 1).
meaning that the extra development time required was difficult to • The regulatory bodies should be involved early on when adopting
justify. Today, however, that situation is changing, and as the pres- a new PSE tool in pharmaceutical production processes, in order
sure builds on the manufacturing cost, and the FDA opens up new to minimize potential problems with process validation.
opportunities via the PAT initiative, a new situation is establishing • The PSE methods and tools developed thus far are often focused
which facilitates – and in fact, demands – innovation. In our opin- on reaction-separation sequences. Similar developments are
ion, PSE must play a central role as a driver of innovative solutions needed for downstream processing and formulation (e.g. gran-
and rational and efficient design, for example introducing rigorous ulation, drying, and tablet press).
quantitative models, able to evaluate process performance from an • Pharmaceutical companies require generic and systematic
objective standpoint. Hence, the application and development of approaches to problem solving.
PSE tools should be continuously encouraged.
It is also important to realize that a major strength of PSE meth-
Acknowledgements
ods and tools lies in the structured approach to solving problems.
The pharmaceutical industry – multipurpose in nature – indeed
The authors would like to thank Prof. Rafiqul Gani (Technical
needs generic approaches in order to solve in a systematic way mul-
University of Denmark) for numerous fruitful discussions. The PhD
tiple problems sharing common features. The scientific literature
project of Albert E. Cervera-Padrell was financed by the Technical
frequently reports specific case studies or solutions to a particular
University of Denmark and H. Lundbeck A/S.
problem, while in our opinion, the focus should change to methods
and tools able to address a complete class of problems. Experi-
References
ence and confidence will thereby be built within pharmaceutical
companies to gradually implement innovating solutions. An addi- Aamir, E., Nagy, Z. K., & Rielly, C. D. (2010). Optimal seed recipe design for crystal
tional benefit of adopting generic approaches is that it should be size distribution control for batch cooling crystallisation processes. Chemical
easier to get approval from regulatory bodies such as the FDA. For Engineering Science, 65, 3602–3614.
Abdul Samad, N. A. F., Singh, R., Sin, G., Gernaey, K. V., & Gani, R. (2011). A generic
example, with the introduction of the PAT guidance, it has become
multi-dimensional model-based system for batch cooling crystallization pro-
quite common to involve regulatory bodies as early as the develop- cesses. Computers & Chemical Engineering, 35, 828–843.
ment phase, whenever new methods or tools need to be tested or Adam, S., Suzzi, D., Radeke, C., & Khinast, J. G. (2011). An integrated Quality by Design
(QbD) approach towards design space definition of a blending unit operation by
implemented. Recent PSE examples of such approaches are generic
discrete element method (DEM) simulation. European Journal of Pharmaceutical
modeling frameworks for prediction of the crystal size distribution Sciences, 42, 106–115.
obtained in batch cooling crystallization processes (Abdul Samad et Ahmad, B. S., & Barton, P. I. (1999). Process-wide integration of solvent mixtures.
al., 2011), and generic approaches for designing a pilot-plant able Computers & Chemical Engineering, 23, 1365–1380.
Akkisetty, P. K., Lee, U., Reklaitis, G. V., & Venkatasubramanian, V. (2010). Popula-
to produce kg amounts of a family of related components (Singh et tion balance model-based hybrid neural network for a pharmaceutical milling
al., 2011), for example during screening. process. Journal of Pharmaceutical Innovation, 5, 161–168.
K.V. Gernaey et al. / Computers and Chemical Engineering 42 (2012) 15–29 27

Alonso, D., Velazquez, A., & Vega, J. M. (2009). A method to generate computation- Doyle, F. J., Harrison, C. A., & Crowley, T. J. (2003). Hybrid model-based approach to
ally efficient reduced order models. Computer Methods in Applied Mechanics and batch-to-batch control of particle size distribution in emulsion polymerization.
Engineering, 198, 2683–2691. Computers & Chemical Engineering, 27, 1153–1163.
am Ende, M. T., Moses, S. K., Carella, A. J., Gadkari, R. A., Graul, T. W., Otano, A. L., Dubey, A., Sarkar, A., Ierapetritou, M., Wassgren, C. R., & Muzzio, F. J. (2011). Compu-
et al. (2007). Improving the content uniformity of a low-dose tablet formula- tational approaches for studying the granular dynamics of continuous blending
tion through roller compaction optimization. Pharmaceutical Development and processes, 1 – DEM based methods. Macromolecular Materials and Engineering,
Technology, 12, 391–404. 296, 290–307.
Asprey, S. P., & Macchietto, S. (2000). Statistical tools for optimal dynamic model Eriksson, L., Johansson, E., Kettaneh-Wold, N., Wikström, C., & Wold S. (2008). Design
building. Computers & Chemical Engineering, 24, 1261–1267. of experiments. Principles and applications (3rd ed.) ISBN: 13-978-91-973730-4-3.
Atkinson, A. C., & Donev, A. N. (1992). Optimum experimental designs. Oxford Univer- U.S. Food and Drug Administration (FDA) (2004). PAT guidance.
sity Press. Federsel, H. (2003). Searching for scalable processes: Addressing the challenges in
Barthe, P., Guermeur, C., Lobet, O., Moreno, M., Woehl, P., Roberge, D. M., et al. times of increasing complexity. Current Opinion in Drug Discovery & Development,
(2008). Continuous multi-injection reactor for multipurpose production – Part 6, 838–847.
I. Chemical Engineering Technology, 31, 1146–1154. Franceschini, G., & Macchietto, S. (2008). Model-based design of experiments
Bersimis, S., Psarakis, S., & Panaretos, J. (2006). Multivariate statistical process con- for parameter precision: State of the art. Chemical Engineering Science, 63,
trol charts: An overview. Quality and Reliability Engineering International, 23, 4846–4872.
517–543. Fujiwara, M., Nagy, Z. K., Chew, J. W., & Braatz, R. D. (2005). First-principles and direct
Bhatia, T., & Biegler, L. T. (1996). Dynamic optimization in the design and scheduling design approaches for the control of pharmaceutical crystallization. Journal of
of multiproduct batch plants. Industrial & Engineering Chemistry Research, 35, Process Control, 15, 493–504.
2234–2246. Gani, R., Gómez, P. A., Folić, M., Jiménez-González, C., & Constable, D. J. C. (2008).
Bilgili, E., & Scarlett, B. (2005). Population balance modeling of non-linear effects in Solvents in organic synthesis: Replacement and multi-step reaction systems.
milling processes. Powder Technology, 153, 59–71. Computers & Chemical Engineering, 32, 2420–2444.
Blayer, S., Woodley, J. M., Lilly, M. D., & Dawson, M. J. (1996). Characterization of the García Muñoz, S., Dolph, S., & Ward, H. W., II. (2010). Handling uncertainty in the
chemoenzymatic synthesis of N-Acetyl-d-neuraminic acid (Neu5Ac). Biotech- establishment of a design space for the manufacture of a pharmaceutical prod-
nology Progress, 12, 758–763. uct. Computers & Chemical Engineering, 34, 1098–1107.
Boukouvala, F., Muzzio, F. J., & Ierapetritou, M. G. (2010). Design space of pharma- Gernaey, K. V., & Gani, R. (2010). A model-based systems approach to pharma-
ceutical processes using data-driven-based methods. Journal of Pharmaceutical ceutical product-process design and analysis. Chemical Engineering Science, 65,
Innovation, 5, 119–137. 5757–5769.
Boukouvala, F., Muzzio, F. J., & Ierapetritou, M. G. (2011). Feasibility analysis Gernaey, K. V., Eliasson Lantz, A., Tufvesson, P., Woodley, J. M., & Sin, G. (2010).
of black-box processes using an adaptive sampling kriging based method. Application of mechanistic models to fermentation and biocatalysis for next
Computer-Aided Chemical Engineering, 29, 432–436. generation processes. Trends in Biotechnology, 28, 346–354.
Boukouvala, F., Ramachandran, R., Vanarase, A., Muzzio, F. J., & Ierapetritou, M. Gerogiorgis, D. I., & Barton, P. I. (2009). Steady-state optimization of a continuous
G. (2011). Computer aided design and analysis of continuous pharmaceutical pharmaceutical process. Computer-Aided Chemical Engineering, 27, 927–932.
manufacturing processes. Computer-Aided Chemical Engineering, 29, 216–220. Grossmann, I. (2005). Enterprise-wide optimization: A new frontier in process sys-
Boukouvala, F., Dubey, A., Vanarase, A., Ramachandran, R., Muzzio, F. J., & Ierapetri- tems engineering. AIChE Journal, 51, 1846–1857.
tou, M. (2012). Computational approaches for studying the granular dynamics of Hartman, R. L., & Jensen, K. F. (2009). Microchemical systems for continuous-flow
continuous blending processes, 2 – Population balance and data-based method. synthesis. Lab on a Chip, 9, 2495–2507.
Macromolecular Materials and Engineering, 297, 9–19. Hartman, R. L., Sahoo, H. R., Yen, B. C., & Jensen, K. F. (2009). Distillation in micro-
Camacho, J., Pico, J., & Ferrer, A. (2008). Bilinear modelling of batch processes. Part chemical systems using capillary forces and segmented flow. Lab on a Chip, 9,
II: A comparison of PLS soft-sensors. Journal of Chemometrics, 22, 533–547. 1843–1849.
Capece, M., Bilgili, E., & Dave, R. (2011). Identification of the breakage rate and distri- Hartman, R. L., Naber, J. R., Buchwald, S. L., & Jensen, K. F. (2010). Multistep micro-
bution parameters in a non-linear population balance model for batch milling. chemical synthesis enabled by microfluidic distillation. Angewandte Chemie
Powder Technology, 208, 195–204. International Edition, 49, 899–903.
Carvalho, A., Gani, R., & Matos, H. (2008). Design of sustainable chemical processes: Hartman, R. L., Naber, J. R., Zaborenko, N., Buchwald, S. L., & Jensen, K. F. (2010). Over-
Systematic retrofit analysis generation and evaluation of alternatives. Process coming the challenges of solid bridging and constriction during Pd-catalyzed
Safety and Environmental Protection, 86, 328–346. C N bond formation in microreactors. Organic Process Research & Development,
Carvalho, A., Matos, H. A., & Gani, R. (2009). Design of batch operations: Systematic 14, 1347–1357.
methodology for generation and analysis of sustainable alternatives. Computers Hartman, R. L., McMullen, J. P., & Jensen, K. F. (2011). Deciding whether to go with the
& Chemical Engineering, 33, 2075–2090. flow: Evaluating the merits of flow reactors for synthesis. Angewandte Chemie
Cervera, A. E., Gani, R., Kiil, S., Skovby, T., & Gernaey, K. V. (2011). A systematic International Edition, 50, 7502–7519.
methodology for the design of continuous active pharmaceutical ingredient Henderson, R. K., Jiménez-González, C., Preston, C., Constable, D. J. C., & Woodley, J. M.
production processes. Computer-Aided Chemical Engineering, 29, 271–275. (2008). EHS & LCA assessment for 7-ACA synthesis: A case study for comparing
Cervera-Padrell, A. E., Nielsen, J. P., Pedersen, M. J., Müller, K., Mortensen, A. R., biocatalytic & chemical synthesis. Industrial Biotechnology, 4, 180–192.
Skovby, T., et al. (2012). Monitoring and control of a continuous Grignard Hermanto, M. W., Braatz, R. D., & Chiu, M.-S. (2011). Integrated batch-to-batch and
alkylation reaction for the synthesis of an active pharmaceutical ingredient nonlinear model predictive control for polymorphic transformation in pharma-
intermediate using in-line NIR spectroscopy. Organic Process Research and Devel- ceutical crystallization. AIChE Journal, 57, 1008–1019.
opment, doi:10.1021/op2002563, in press. Hessel, V. (2009). Novel process windows – Gate to maximizing process intensifica-
Chakraborty, A., & Linninger, A. A. (2002). Plant-wide waste management. 1. Syn- tion via flow chemistry. Chemical Engineering Technology, 32, 1655–1681.
thesis and multiobjective design. Industrial & Engineering Chemistry Research, 41, Hsieh, D., Marchut, A. J., Wei, C., Zheng, B., Wang, S. S. Y., & Kiang, S. (2009).
4591–4604. Model-based solvent selection during conceptual process design of a new drug
Chen, J., & Trout, B. L. (2010). Computer-aided solvent selection for improving the manufacturing process. Organic Process Research & Development, 13, 690–697.
morphology of needle-like crystals: A case study of 2,6-dihydroxybenzoic acid. ICH. (2009). International conference on harmonisation of technical requirements for
Crystal Growth & Design, 10, 4379–4388. registration of pharmaceuticals for human use ICH Harmonised Tripartite Guide-
Chen, B., & Woodley, J. M. (2002). Wavelet shrinkage data processing for neu- line, Pharmaceutical Development. Step 4, August 2009
ral networks in bioprocess modeling. Computers & Chemical Engineering, 26, Jaksland, C. A., Gani, R., & Lien, K. M. (1995). Separation process design and synthesis
1611–1620. based on thermodynamic insights. Chemical Engineering Science, 50, 211–530.
Constable, D. J. C., Curzons, A. D., & Cunningham, V. L. (2002). Metrics to ‘green’ Jensen, K. F. (2001). Microreaction engineering – Is small better? Chemical Engineer-
chemistry-which are the best? Green Chemistry, 4, 521–527. ing Science, 56, 293–303.
Constable, D. J. C., Jiménez-González, C., & Henderson, R. K. (2007). Perspective on Jimenez-Gonzalez, C., Ponder, C. S., Broxterman, Q. B., & Manley, J. B. (2011). Using
solvent use in the pharmaceutical industry. Organic Process Research & Develop- the right green yardstick: Why process mass intensity is used in the pharma-
ment, 11, 133–137. ceutical industry to drive more sustainable processes. Organic Process Research
Curzons, A. D., Constable, D. J. C., Mortimer, D. N., & Cunningham, V. L. (2001). So you & Development, 15, 912–917.
think your process is green, how do you know? – Using principles of sustain- Jiménez-González, C., Curzons, A. D., Constable, D. J. C., & Cunningham, V. L. (2005).
ability to determine what is green – A corporate perspective. Green Chemistry, Expanding GSK’s Solvent Selection Guide – Application of life cycle assessment
3, 1–6. to enhance solvent selections. Clean Technologies and Environmental Policy, 7,
Dassau, E., Zadok, I., & Lewin, D. R. (2006). Combining six-sigma with integrated 42–50.
design and control for yield enhancement in bioprocessing. Industrial & Engi- Jiménez-González, C., Poechlauer, P., Broxterman, Q. B., Yang, B. S., am Ende, D., Baird,
neering Chemistry Research, 45, 8299–8309. J., et al. (2011). Key green engineering research areas for sustainable manufac-
Diedrichs, A., & Gmehling, J. (2011). Solubility calculation of active pharmaceuti- turing: A perspective from pharmaceutical and fine chemicals manufacturers.
cal ingredients in alkanes, alcohols, water and their mixtures using various Organic Process Research & Development, 15, 900–911.
activity coefficient models. Industrial & Engineering Chemistry Research, 50, Kadlec, P., Gabrys, B., & Strandt, S. (2009). Data-driven soft sensors in the process
1757–1769. industry. Computers & Chemical Engineering, 33, 795–814.
Dong, D., & McAvoy, T. J. (1996). Batch tracking using non-linear principal component Kadlec, P., Grbic, R., & Gabrys, B. (2011). Review of adaptation mechanisms for data-
analysis. AIChE Journal, 42, 2199–2208. driven soft sensors. Computers & Chemical Engineering, 35, 1–24.
Downs, J. J., & Vogel, E. F. (1993). A plant-wide industrial process control problem. Kahrs, O., & Marquardt, W. (2008). Incremental identification of hybrid process
Computers & Chemical Engineering, 17, 245–255. models. Computers & Chemical Engineering, 32, 694–705.
28 K.V. Gernaey et al. / Computers and Chemical Engineering 42 (2012) 15–29

Kang, L., Chung, B. G., Langer, R., & Khademhosseini, A. (2008). Microfluidics for Morbach, J., Yang, A., & Marquardt, W. (2007). OntoCAPE – A large-scale ontology for
drug discovery and development: From target selection to product lifecycle chemical process engineering. Engineering Applications of Artificial Intelligence,
management. Drug Discovery Today, 13, 1–13. 20, 147–161.
Kashid, M. N., Agar, D. W., & Turek, S. (2007). CFD modelling of mass transfer with and Mortier, S. T. F. C., De Beer, T., Gernaey, K. V., Remon, J. P., Vervaet, C., & Nopens, I.
without chemical reaction in the liquid–liquid slug flowmicroreactor. Chemical (2011). Mechanistic modelling of fluidized bed drying processes of wet porous
Engineering Science, 62, 5102–5109. granules: A review. European Journal of Pharmaceutics and Biopharmaceutics, 79,
Klatt, K., & Marquardt, W. (2009). Perspectives for process systems engineering – 205–225.
Personal views from academia and industry. Computers & Chemical Engineering, Nagy, Z. K., Fujiwara, M., & Braatz, R. D. (2008). Modelling and control of combined
33, 536–550. cooling and antisolvent crystallization processes. Journal of Process Control, 18,
Koch, K., van Weerdenburg, B. J. A., Verkade, J. M. M., Nieuwland, P. J., Rutjes, F. P. J. T., 856–864.
& van Hest, J. C. M. (2009). Optimizing the deprotection of the amine protecting Nomikos, P., & McGregor, J. F. (1995). Multi-way partial least squares in monitoring
p-methoxyphenyl group in an automated microreactor platform. Organic Process batch processes. Chemometrics and Intelligent Laboratory Systems, 30, 97–108.
Research & Development, 13, 1003–1006. Nørgaard, L., Saudland, A., Wagner, J., Nielsen, J. P., Munck, L., & Engelsen, S. B. (2000).
Kockmann, N., & Roberge, D. M. (2009). Harsh reaction conditions in continuous-flow Interval Partial Least-Squares Regression (iPLS): A comparative chemometric
microreactors for pharmaceutical production. Chemical Engineering Technology, study with an example from near-infrared spectroscopy. Applied Spectroscopy,
32, 1682–1694. 54, 413–419.
Kockmann, N., Gottsponer, M., & Roberge, D. M. (2011). Scale-up concept of single- O’Connell, J. P., Gani, R., Mathias, P. M., Maurer, G., Olson, J. D., & Crafts, P. A.
channel microreactors from process development to industrial production. (2009). Thermodynamic property modeling for chemical process and product
Chemical Engineering Journal, 167, 718–726. engineering: Some perspectives. Industrial & Engineering Chemistry Research, 48,
Kourti, T. (2006). Process analytical technology beyond real-time analyzers: The role 4619–4637.
of multivariate analysis. Critical Reviews in Analytical Chemistry, 36, 257–278. Paakkunainen, M., Matero, S., Ketolainen, J., Lahtela-Kakkonen, M., Poso, A., &
Kralj, J. G., Sahoo, H. R., & Jensen, K. F. (2007). Integrated continuous microfluidic Reinikainen, S.-P. (2009). Uncertainty in dissolution test of drug release. Chemo-
liquid–liquid extraction. Lab on a Chip, 7, 256–263. metrics and Intelligent Laboratory Systems, 97, 82–90.
Krasnyk, M., Mangold, M., & Kienle, A. (2010). Reduction procedure for parametrized Plumb, K. (2005). Continuous processing in the pharmaceutical industry – Changing
fluid dynamics problems based on proper orthogonal decomposition and cali- the mindset. Chemical Engineering Research and Design, 83, 730–738.
bration. Chemical Engineering Science, 65, 6238–6246. Pollard, D. J., & Woodley, J. M. (2007). Biocatalysis for pharmaceutical intermediates:
Krasnyk, M., Mangold, M., Ganesan, S., & Tobiska, L. (2012). Numerical reduction of a The future is now. Trends in Biotechnology, 25, 66–73.
crystallizer model with internal and external coordinates by proper orthogonal Pollet, P., Cope, E. D., Kassner, M. K., Charney, R., Terett, S. H., Richman, K. W., et
decomposition. Chemical Engineering Science, 70, 77–86. al. (2009). Production of (S)-1-benzyl-3-diazo-2-oxopropylcarbamic acid tert-
Kremer, D. M., & Hancock, B. C. (2006). Process simulation in the pharmaceutical butyl ester, a diazoketone pharmaceutical intermediate, employing a small scale
industry: A review of some basic physical models. Journal of Pharmaceutical continuous reactor. Industrial & Engineering Chemistry Research, 48, 7032–7036.
Sciences, 95, 517–529. Poon, J. M. H., Ramachandran, R., Sanders, C. F. W., Glaser, T., Immanuel, C. D., Doyle,
Kruger, U., Zhang, J., & Xie, L. (2008). Developments and applications of nonlinear F. J., III, et al. (2009). Experimental validation studies on a multi-dimensional and
principal component analysis – A review. Lecture Notes in Computational Science multi-scale population balance model of batch granulation. Chemical Engineering
and Engineering, 58, 1–43. Science, 64, 775–786.
Lang, Y., Malacina, A., Biegler, L. T., Munteanu, S., Madsen, J. I., & Zitney, S. E. (2009). Pordal, H. S., Matice, C. J., & Fry, T. J. (2002). The role of computational fluid dynamics
Reduced order model based on principal component analysis for process simu- in the pharmaceutical industry. Pharmaceutical Technology, 26(2), 72–77.
lation and optimization. Energy & Fuels, 23, 1695–1706. Prpich, A., am Ende, M. T., Katzschner, T., Lubczyk, V., Weyhers, H., & Bernhard, G.
LaPorte, T. L., & Wang, C. (2007). Continuous processes for the production of pharma- (2010). Drug product modeling predictions for scale-up of tablet film coating – A
ceutical intermediates and active pharmaceutical ingredients. Current Opinion quality by design approach. Computers & Chemical Engineering, 34, 1092–1097.
in Drug Discovery & Development, 10, 738–745. Radeke, C. A., Glasser, B. J., & Khinast, J. G. (2010). Large-scale powder mixer simula-
Leardi, R., & Nørgaard, L. (2004). Sequential application of backward interval par- tions using massively parallel GPU architectures. Chemical Engineering Science,
tial least squares and genetic algorithms for the selection of relevant spectral 65, 6435–6442.
regions. Journal of Chemometrics, 18, 486–497. Ramachandran, R., Immanuel, C. D., Stepanek, F., Litster, J. D., & Doyle, F. J., III. (2009).
Lencastre Fernandes, R., Nierychlo, M., Lundin, L., Pedersen, A. E., Puentes Tellez, A mechanistic model for breakage in population balances of granulation: The-
P. E., Dutta, A., et al. (2011). Experimental methods and modeling techniques oretical kernel development and experimental validation. Chemical Engineering
for description of cell population heterogeneity. Biotechnology Advances, 29, Research and Design, 87, 598–614.
575–599. Reginato, G., Sadler, P., & Wilkes, R. D. (2011). Scaling up metal scavenging opera-
Linninger, A. A., & Malcolm, A. (2006). Pollution prevention for batch pharmaceutical tions for pharmaceutical pilot plant manufactures. Organic Process Research &
and specialty chemical processes. In E. Korovessi, & A. A. Linninger (Eds.), Batch Development, 15, 1396–1405.
processes. Boca Raton, FL, USA: CRC Press, Taylor & Francis Group. Reich, G. (2005). Near-infrared spectroscopy and imaging: Basic principles and phar-
MacGregor, J. F., & Bruwer, M. J. (2008). A framework for the development of design maceutical applications. Advanced Drug Delivery Reviews, 57, 1109–1143.
and control spaces. Journal of Pharmaceutical Innovation, 3, 15–22. Reklaitis, G. V. R. (2007). Perspectives on Process Systems Engineering R&D in
Mandenius, C., & Brundin, A. (2008). Bioprocess optimization using design-of- support of pharmaceutical product/process development and manufacturing.
experiments methodology. Biotechnology Progress, 24, 1191–1203. Computer-Aided Chemical Engineering, 24, 35–38.
Marchetti, A., Srinivasan, B., Bonvin, D., Elgue, S., Prat, L., & Cabassud, M. (2005). Remy, B., Khinast, J. G., & Glasser, B. J. (2009). Discrete element simulation of free
Measurement-based run-to-run optimization of a batch reaction-distillation flowing grains in a four-bladed mixer. AIChE Journal, 55, 2035–2048.
system. Computer-Aided Chemical Engineering, 20, 1417–1422. Remy, B., Glasser, B. J., & Khinast, J. G. (2010). The effect of mixer properties and fill
Marquardt, W. (2005). Model-based experimental analysis of kinetic phenomena level on granular flow in a bladed mixer. AIChE Journal, 56, 336–353.
in multi-phase reactive systems. Chemical Engineering Research and Design, 83, Roberge, D. M., Ducry, L., Bieler, N., Cretton, P., & Zimmermann, B. (2005). Microre-
561–573. actor technology: A revolution for the fine chemical and pharmaceutical
Marquardt, W., Morbach, J., Wiesner, A., & Yang, A. (2010). OntoCAPE: A re-usable industries? Chemical Engineering & Technology, 28, 318–323.
ontology for chemical process engineering. Springer., ISBN 978-3-642-04654-4. Rohner, M., & Meyer, H.-P. (1995). Applications of modelling for bioprocess design
Matsuda, H., Kaburagi, K., Kurihara, K., Tochigi, K., & Tomono, K. (2010). Prediction of and control in industrial production. Bioprocess and Biosystems Engineering, 13,
solubilities of pharmaceutical compounds in water + co-solvent systems using 69–78.
an activity coefficient model. Fluid Phase Equilibria, 290, 153–157. Rosas, J. G., Blanco, M., González, J. M., & Alcalá, M. (2011a). Quality by design
McMullen, J. P., & Jensen, K. F. (2010a). An automated microfluidic system for online approach of a pharmaceutical gel manufacturing process. Part 1: Determination
optimization in chemical synthesis. Organic Process Research & Development, 14, of the design space. Journal of Pharmaceutical Sciences, 100, 4432–4441.
1169–1176. Rosas, J. G., Blanco, M., González, J. M., & Alcalá, M. (2011b). Quality by design
McMullen, J. P., & Jensen, K. F. (2010b). Integrated microreactors for reaction automa- approach of a pharmaceutical gel manufacturing process. Part 2: Near infrared
tion: New approaches to reaction development. Annual Review of Analytical monitoring of composition and physical parameters. Journal of Pharmaceutical
Chemistry, 3, 19–42. Sciences, 100, 4442–4451.
McMullen, J. P., & Jensen, K. F. (2011). Rapid determination of reaction kinetics with Sahoo, H. R., Kralj, J. G., & Jensen, K. F. (2007). Multistep continuous-flow microchem-
an automated microfluidic system. Organic Process Research & Development, 15, ical synthesis involving multiple reactions and separations. Angewandte Chemie,
398–407. 119, 5806–5810.
Mendorf, M., Nachtrodt, H., Mescher, A., Ghaini, A., & Agar, D. W. (2010). Design and Sales-Cruz, M., & Gani, R. (2003). S. P. Asprey, & S. Macchietto (Eds.). A modelling
control techniques for the numbering-up of capillary microreactors with uni- tool for different stages of the process life. Computer-Aided Chemical Engineering,
form multiphase flow distribution. Industrial & Engineering Chemistry Research, 16, 209–249.
49, 10908–10916. Saling, P., Kicherer, A., Dittrich-Krämer, B., Wittlinger, R., Zombik, W., Schmidt, I., et
Michaut, F., Busignies, V., Fouquereau, C., Huet de Barochez, B., Leclerc, B., & al. (2002). Eco-efficiency analysis by BASF: The method. The International Journal
Tchoreloff, P. (2010). Evaluation of a rotary tablet press simulator as a tool for the of Life Cycle Assessment, 7, 203–218.
characterization of compaction properties of pharmaceutical products. Journal Savile, C. K., Janey, J. M., Mundorff, E. C., Moore, J. C., Tam, S., Jarvis, W. R., et al.
of Pharmaceutical Sciences, 99, 2874–2885. (2010). Biocatalytic asymmetric synthesis of chiral amines from ketones applied
Mills, P. L., Quiram, D. J., & Ryley, J. F. (2007). Microreactor technology and process to sitagliptin manufacture. Science, 329, 305–309.
miniaturization for catalytic reactions – A perspective on recent developments Schaber, S. D., Gerogiorgis, D. I., Ramachandran, R., Evans, J. M. B., Barton, P. I.,
and emerging technologies. Chemical Engineering Science, 62, 6992–7010. & Trout, B. L. (2011). Economic analysis of integrated continuous and batch
K.V. Gernaey et al. / Computers and Chemical Engineering 42 (2012) 15–29 29

pharmaceutical manufacturing: A case study. Industrial & Engineering Chemistry Venkatasubramanian, V., Rengaswamy, R., Yin, K., & Kavuri, S. N. (2003). A review of
Research, 50, 10083–10092. process fault detection and diagnosis. Part I: Quantitative model-based methods.
Seborg, D. E., Mellichamp, D. A., Edgar, T. F., & Doyle, F. J., III. (2010). Process dynamics Computers & Chemical Engineering, 27, 293–311.
and control (3rd ed.). Wiley and Sons. Venkatasubramanian, V., Rengaswamy, R., & Kavuri, S. N. (2003). A review of process
Sharma, C., Malhotra, D., & Rathore, A. S. (2011). Review of computational fluid fault detection and diagnosis. Part II: Qualitative models and search strategies.
dynamics applications in biotechnology processes. Biotechnology Progress, 27, Computers & Chemical Engineering, 27, 313–326.
1497–1510. Venkatasubramanian, V., Rengaswamy, R., Kavuri, S. N., & Yin, K. (2003). A review of
Sheldon, R. A. (1992). Organic synthesis – Past, present and future. Chemistry and process fault detection and diagnosis. Part III: Process history based methods.
Industry, 23, 903–906. Computers & Chemical Engineering, 27, 327–346.
Sheldon, R. A. (2007). The E factor: Fifteen years on. Green Chemistry, 9, 1273–1283. Venkatasubramanian, V., Zhao, C., Joglekar, G., Jain, A., Hailemariam, L., Suresh, P.,
Shonnard, D. R., Kicherer, A., & Saling, P. (2003). Industrial applications using BASF et al. (2006). Ontological informatics infrastructure for pharmaceutical prod-
eco-efficiency analysis: Perspectives on green engineering principles. Environ- uct development and manufacturing. Computers & Chemical Engineering, 30,
mental Science & Technology, 37, 5340–5348. 1482–1496.
Sin, G., Ödman, P., Petersen, N., Eliasson Lantz, A., & Gernaey, K. V. (2008). Matrix Vervaet, C., & Remon, J. P. (2005). Continuous granulation in the pharmaceutical
notation for efficient development of first-principles models within PAT applica- industry. Chemical Engineering Science, 60, 3949–3957.
tions: Integrated modeling of antibiotic production with Streptomyces coelicolor. Villez, K., Srinivasan, B., Rengaswamy, R., Narasimhan, S., & Venkatasubramanian,
Biotechnology and Bioengineering, 101, 153–171. V. (2011). Kalman-based strategies for Fault Detection and Identification (FDI):
Sin, G., Gernaey, K. V., & Eliasson Lantz, A. (2009). Good modelling practice (GMoP) Extensions and critical evaluation for a buffer tank system. Computers & Chemical
for PAT applications: Propagation of input uncertainty and sensitivity analysis. Engineering, 35, 806–816.
Biotechnology Progress, 25, 1043–1053. Wassgren, C., & Curtis, J. S. (2006). The application of computational modeling to
Singh, R., Gernaey, K. V., & Gani, R. (2009). Model-based computer aided framework pharmaceutical materials science. MRS Bulletin, 31, 900–904.
for design of process monitoring and analysis systems. Computers & Chemical Webb, D., & Jamison, T. F. (2010). Continuous flow multi-step organic synthesis.
Engineering, 33, 22–42. Chemical Science, 1, 675–680.
Singh, R., Gernaey, K. V., & Gani, R. (2010a). An ontological knowledge based system Wei, H., Zhou, W., & Garside, J. (2001). Computational fluid dynamics modeling of
for selection of process monitoring and analysis tools. Computers & Chemical the precipitation process in a semibatch crystallizer. Industrial & Engineering
Engineering, 34, 1137–1154. Chemistry Research, 40, 5255–5261.
Singh, R., Gernaey, K. V., & Gani, R. (2010b). ICAS-PAT: A software for design, Weis, D. C., & Visco, D. P. (2010). Computer-aided molecular design using the
analysis and validation of PAT systems. Computers & Chemical Engineering, 34, signature molecular descriptor: Application to solvent selection. Computers &
1108–1136. Chemical Engineering, 34, 1018–1029.
Singh, R., Rozada-Sanchez, R., Wrate, T., Muller, F., Gernaey, K. V., Gani, R., et al. Wold, S., Sjöström, M., & Eriksson, L. (2001). PLS-regression: A basic tool of chemo-
(2011). A retrofit strategy to achieve fast, flexible, future (F3) pharmaceutical metrics. Chemometrics and Intelligent Laboratory Systems, 58, 109–130.
production processes. Computer-Aided Chemical Engineering, 29, 291–295. Woo, X. Y., Tan, R. B. H., & Braatz, R. D. (2009). Modeling and computational fluid
Stephanopoulos, G., & Reklaitis, G. V. (2011). Process systems engineering: From dynamics-population balance equation-micromixing simulation of impinging
Solvay to modern bio- and nanotechnology: A history of development, successes jet crystallizers. Crystal Growth & Design, 9, 156–164.
and prospects for the future. Chemical Engineering Science, 66, 4272–4306. Woodley, J. M. (2008). New opportunities for biocatalysis: Making pharmaceutical
Straathof, A. J. J., Panke, S., & Schmid, A. (2002). The production of fine chemicals by processes greener. Trends in Biotechnology, 26, 321–327.
biotransformations. Current Opinion in Biotechnology, 13, 548–556. Wu, Y. X., Wang, X., & Ching, C. B. (2002). Computational fluid dynamics simulation
Taskinen, J., & Yliruusi, J. (2003). Prediction of physicochemical properties based on of the adsorption separation of three components in high performance liquid
neural network modelling. Advanced Drug Delivery Reviews, 55, 1163–1183. chromatography. Chromatographia, 55, 439–445.
Tucker, J. L. (2006). Green chemistry, a pharmaceutical perspective. Organic Process Zamprogna, E., Barolo, M., & Seborg, D. E. (2004). Estimating product composition
Research & Development, 10, 315–319. profiles in batch distillation via partial least squares regression. Control Engi-
Tufvesson, P., Lima-Ramos, J., Jensen, J. S., Al-Haque, N., Neto, W., & Woodley, J. M. neering Practice, 12, 917–929.
(2011). Process considerations for the asymmetric synthesis of chiral amines Zhang, J., Martin, E. B., & Morris, A. J. (1997). Process monitoring using non-linear
using transaminases. Biotechnology and Bioengineering, 108, 1479–1493. statistical techniques. Chemical Engineering Journal, 67, 181–189.
Tufvesson, P., Lima-Ramos, J., Nordblad, M., & Woodley, J. M. (2011). Guidelines and Zimmermann, V., Hennemann, H., Daußmann, T., & Kragl, U. (2007). Modelling
cost analysis for catalyst production in biocatalytic processs. Organic Process the reaction course of N -acetylneuraminic acid synthesis from N -acetyl-d -
Research & Development, 15, 266–274. glucosamine – New strategies for the optimisation of neuraminic acid synthesis.
Vasić-Rački, D., Findrik, Z., & Vrsalović Presečki, A. (2011). Modelling as a tool of Applied Microbiology and Biotechnology, 76, 597–605.
enzyme reaction engineering for enzyme reactor development. Applied Micro-
biology and Biotechnology, 91, 845–856.

You might also like