Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/264836968

Maximising return on investment (ROI) for pharmaceutical production

Article  in  International Journal of Manufacturing Technology and Management · January 2011


DOI: 10.1504/IJMTM.2011.046768

CITATIONS READS

0 618

2 authors, including:

Noha A. Mostafa
Zagazig University
27 PUBLICATIONS   69 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Sustainable development View project

Using Matheuristics in supply chain management problems. View project

All content following this page was uploaded by Noha A. Mostafa on 22 February 2018.

The user has requested enhancement of the downloaded file.


Int. J. Manufacturing Technology and Management, Vol. 24, Nos. 1/2/3/4, 2011 167

Maximising return on investment (ROI) for


pharmaceutical production

Sameh A. Ibrahim and Noha A. Mostafa*


Faculty of Engineering,
Industrial Engineering Department,
Zagazig University, Egypt
E-mail: sicc52@yahoo.com
E-mail: alloha86@hotmail.com
*Corresponding author

Abstract: Production scheduling tools close the gap between ERP/MRP II


tools and the plant floor. Simulation can be used to generate production
schedules on an ongoing basis in a way that does not violate constraints related
to the limited availability. In this paper, the process of tablet manufacturing in a
pharmaceutical production plant is studied, first the full process is described
and mapped, and then a process simulation is run. Based on the simulation
results, statistical analysis is performed; the data is fitted by using regression
analysis. Design of experiment (DOE) analysis is performed on the process to
try many different combinations of resource levels automatically, and finally
analysis of variance (ANOVA) is performed to select infeasible products that
can be excluded from future production plans, this will have a positive effect on
the return on investment for the organisation.

Keywords: pharmaceuticals manufacturing; process mapping; simulation;


design of experiment; DOE; analysis of variance; ANOVA.

Reference to this paper should be made as follows: Ibrahim, S.A.


and Mostafa, N.A. (2011) ‘Maximising return on investment (ROI) for
pharmaceutical production’, Int. J. Manufacturing Technology and Management,
Vol. 24, Nos. 1/2/3/4, pp.167–181.

Biographical notes: Sameh A. Ibrahim is a well-known Professor of Industrial


Engineering. Also, he works in the fields of IT and auto ID, and his business
has branches in Egypt, KSA, Algeria and the UK.

Noha A. Mostafa is a Teaching Assistant of Industrial Engineering. She was


honoured by the Egyptian president for having the first rank on her university.
Also, she was included in the 2009 edition of Who’s Who in the world®.

1 Introduction

Manufacturing process management (MPM) is a collection of technologies and methods


used to define how products are to be manufactured. MPM differs from ERP/MRP which
is used to plan the ordering of materials and other resources, set manufacturing schedules,
and compile cost data.

Copyright © 2011 Inderscience Enterprises Ltd.


168 S.A. Ibrahim and N.A. Mostafa

A cornerstone of MPM is the central repository for the integration of all these tools
and activities aids in the exploration of alternative production line scenarios; making
assembly lines more efficient with the aim of reduced lead time to product launch, shorter
product times and reduced work in progress (WIP) inventories as well as allowing rapid
response to product or product changes. Process simulation and scheduling tools can play
an important role in this endeavour.
The pharmaceutical industry develops, produces, and markets drugs licensed for use
as medications. Common forms of pharmaceutical products include tablets, capsules,
ointments, creams, solutions in syringes and vials. Simulation software can be used to
represent the entire process on the computer, estimate the cycle time of the process, and
perform cost analysis. Simulation allows for a realistic representation of failure and a
detailed representation of uncertainty with relative ease.
The pharmaceutical industry spends considerably more on the development and
evaluation of products that eventually fail than on successful products. Consequently, any
methodologies and tools that can be used to evaluate alternatives and speed up the
development effort can have a tremendous impact on the return on investment (ROI).
Discrete and process manufacturing are terms that are often used to describe the nature of
manufacturing operations where the process industry has several specifics compared with
discrete industry that make it both complex and uncertain; see Taylor et al. (1981). An
extensive review of scheduling as it applies to chemical engineering is presented by
Reklaitis (1992). According to Hwang (1997), there are many variables that affect the
process including batch size and resource constraints. Process bottlenecks and delays can
be identified so that cost-effective alternatives can be investigated. Gonzalez and Realff
(1998) coupled a discrete event simulator in an open loop to the output of a mixed-integer
linear-programming (MILP) scheduling model for pipe-less batch plants to study the
sensitivity of the schedule to processing and travel time perturbations. Honkomp et al.
(1999) developed a framework for directly incorporating schedules into a batch
process simulator for the purposes of schedule validation and testing of rescheduling
methodologies when stochastic events occur. Simulation is the process of building a
model of a real or proposed system to study the performance of the system under specific
conditions. Blau et al. (2000) presented a probabilistic network simulation model for the
new-product development problem. Applequist et al. (2000) have developed an effective
polytope integration method for evaluation of expected values and variances of revenue
can account for the effects of demand uncertainties on revenue while recognising the
uncertainty in inventory. According to Ball (2001), simulation is especially powerful
because it allows the observation of the behaviour of the model as time progresses. Forza
and Salvador (2001) discuss the need for information flow and redistribution of
management responsibility among all the entities in the management structure in order to
achieve an efficient production. Subramanian et al. (2001) have combined mathematical
programming and discrete event system simulation to assess the uncertainty and control
the risk present in the pipeline. Most importantly, process simulation can be used without
affecting the existing production activities (Micrografx, 2001). Petrides et al. (2002) have
used computer aided process design (CAPD) and simulation tools in expediting process
development, by using SuperPro Designer simulation package. Batch simulation software
helps process developers and design engineers determine process requirements based on
various operating scenarios. Several such tools have been tailored for the biotech and
water-purification industries. Thomas (2003) showed how they can be used for accurate
and efficient facility design. Such tools help project managers evaluate process
Maximising return on investment (ROI) for pharmaceutical production 169

adjustments and make decisions confidently. Barnett (2003) has discussed the use of
design of experiment (DOE) techniques to structure and organise simulation experiments.
A performance indicator can also be defined as ‘a variable that quantitatively expresses
the effectiveness or efficiency, or both, of a partial or a whole process, or a system,
against a given norm or target’; see Lohman et al. (2004). Floudas and Lin (2004) have
discussed discrete-time approaches, and continuous-time models in chemical industry.
Higuchi and Troutt (2004) have used scenario-based dynamic simulations to study the
short product life cycle case, to simulate the supply chain dynamics. Shah (2004) has
developed a stochastic simulation model of both the physical processes and the business
processes. Petrides and Siletti (2004) have used SPD simulation tool to solve the
material, energy, equipment sizing and duration calculations for each operation
independently of the start-time calculations in batch simulation to see how that can help
improve a process. This has introduced new demands related to flexible production, and
increased production efficiency, fast responses to customer demands, and a high and
uniform quality of products and services according to Dangelmaier et al. (2005). Folan
and Brown (2005) presented the concept of evolution from performance measure to
structural and procedural PM frameworks. Achilleos et al. (2006) have discussed the
impact of uncertain parameters in batch manufacturing of active pharmaceutical
ingredients; they took the advantage of the component object module (COM) technology
built in the crystal ball process simulator and the add-in application’s inherent integration
with Excel. Alexander (2006) has used discrete event simulation to represent each batch
as a single transaction in order to model process cycle time and utility usage. Tan et al.
(2006) used CAPD and simulation tools in the systematic identification of the process
bottleneck and a debottlenecking study in the pharmaceutical cream production.
Papavasileiou et al. (2007) have used SuperPro Designer simulation package to describe
how batch process simulators and scheduling tools can be used to facilitate and expedite
development and commercialisation of pharmaceutical products. Yu et al. (2009) have
introduced useful classifications of batch scheduling problems and optimisation methods.
Other references discuss the pharmaceutical production simulation can be found in.
Zorzut et al. (2009) proposed a closed-loop control structure utilising production
performance indicators as a possible solution to simplify this problem. Ibrahim and
Mostafa (2011) used multi-agent system to control and secure the pharmaceutical supply
chain.
Tablet manufacturing process will be used as a representative example to demonstrate
the use of process simulation tools in the development and manufacturing of
finished pharmaceutical products. Most pharmaceutical processes operate in batch or
semi-continuous mode. This is in contrast to petrochemical and other industries that
handle large throughputs and use continuous processes. In continuous operations, a piece
of equipment performs the same action all the time. In batch processing, on the other
hand, a piece of equipment goes through a cycle of operations.
Having developed a good model using a process simulator, the user may begin
experimenting on the computer with alternative process setups and operating conditions.
This has the potential of reducing the costly and time-consuming laboratory and pilot
plant effort.
The application of some data mining technique is usually required to transform the
process data to the form required by the model. When modelling an existing plant, input
data required by the model can be extracted from the data recorded by the actual process.
170 S.A. Ibrahim and N.A. Mostafa

A communication channel must therefore be established between the modeller and the
plant engineers. In all cases, a certain level of model verification is necessary after the
model is developed. In its simplest form, a review of the results by an experienced
engineer can play the role of verification.
The organisation of this paper is as follows. In Section 2, the model is defined, the
process is fully described, and tools such as SIPOC, spaghetti diagram, VSM, and
process mapping will be used. In Section 3, the model is analysed via process simulation,
regression analysis, experimental design and analysis of variance (ANOVA). Finally,
Section 4 concludes the paper and discusses further work.

2 Model definition

2.1 Process description


The objective of this section is to illustrate how tablet manufacturing process is
performed. Hence, it essential to discuss the successive operations and machines used in
this process. Tablet manufacturing is performed through six processes; preparation,
mixing, compression, coating, blistering, and packaging. Figure 1 illustrates the whole
process to manufacture tablets.
The first process is preparation. The most widely used process of preparation in
pharmaceutical industry is wet granulation. Wet granulation process simply involves wet
massing of the powder blend with a granulating liquid, wet sizing and drying. After that,
the materials are mixed with lubricants and dissolvent for specified time in a mass mixer.
Then, lubricating agents are mixed with the dried material and then fed into tablet
punching machine where tablets are punched according to the required parameters of
quality control (weight, friability, and dissolution time). After that the coating process is
performed, this is the last stage in tablet formulation and it is done to protect the tablet
from temperature and humidity constraints. It is also done to mask the taste, give it
special characteristics, distinction to the product, and prevent inadvertent contact with the
drug substance. The most common forms of tablet coating are sugar coating and film
coating. Tablet coatings must be stable and strong enough to survive the handling of the
tablet, must not make tablets stick together during the coating process. Both the
compressed and coated tablets are individually inspected on the tablets inspection bed.
They are then strip or blister sealed on stripping/blister machines. After that, tablet
blisters are put together with the leaflet in the medication package. This can be done
automatically or manually. Finally, the packages are packaged into large packs and sent
to the FG warehouse. Previously, after coating process a sample of the final product are
sent to the laboratory to analyse it and approve the tablet to be distributed. So, by the time
the final packages are sent to the FG warehouse, the laboratory approval is sent. The
boxes are kept into the FG warehouse, and then they are posted to distribution zone to be
transported by the fleet. The company has its own fleet, and also it deals with third-party
especially to serve small villages.
Maximising return on investment (ROI) for pharmaceutical production 171

Figure 1 Tablet manufacturing process (see online version for colours)

iGrafx IDEF0 2007 has been used in this work to develop SIPOC, spaghetti diagram,
VSM, process map and simulation. iGrafx is a recognised leader of comprehensive
business process analysis solutions.

2.2 SIPOC
Figure 2 illustrates the SIPOC diagram; it stands for suppliers, input, process, output, and
customers. The plant obtains input from suppliers, add value through their process, and
provide an output that meets or exceeds customer’s requirements. The suppliers of the
company are located in Europe and Egypt, European suppliers provide powders, APIs,
flavours and other additions, Egyptian suppliers provide USP water, sucrose and packing
materials. The company produces about 60 tablet products; customers are located in
Egypt, Europe, Asia, Africa and most of Arab countries. SIPOC helps looking at the
process from a high level by capturing these elements.
172 S.A. Ibrahim and N.A. Mostafa

Figure 2 SIPOC (see online version for colours)


Supplier(s) Inputs/Req’ts Process Output(s)/Req’ts Customers

T a b le ts: Egypt
Europe [A]Material Req. 1. Preparation A cupan
A lle rg e x
Big Distributors
Retailers
1-Powder 1- Powders A lle rg e x C a ffe in e
A ltia z e m Minstry of Health
2-API 2- API A lz e n ta l Military
A sm o lin
3-Flavoring 3-Flavoring 2. Mixing A te n o 5 0 m g

4-Mannitol 4-Mannitol A te n o 1 0 0 m g
A to r
 
5-Stabilizer 5-Stabilizer B ro p a m 1 .5 m g
B ro p a m 3 m g
6-Excipient 6-Excipient 3. Compression C a p o tril 2 5 m g Arab Countries
C a p o tril 5 0 m g
Algeria
7-USP water C e tal
Bahrain
C ip ro c in 2 5 0 m g
Egypt 8-Sucrose C ip ro c in 5 0 0 m g Iraq

1-USP water 4. Coating C ip ro c in 7 5 0 m g Jordan


C lo p a m Kuwait
2-Sucrose Covers C o lo sp a sm in
C o n ta -F lu
Lebanon
Libya
3-Packing 9- Boxes C o n ta -F lu -N
Oman
10- Packing material 5. Blistering D in itra 5 m g
materials D in itra 1 0 m g
D in itra 2 0 m g
Palestine
Qatar
D is to c id e Saudi Arabia
D o sin 1 m g
[B]Machines 6. Packaging D o sin 2 m g
Sudan
UAE
1- Garnulators D o sin 4 m g
E m e ra l Yemen
2- Mixers E -M o x c la v 3 7 5 m g
 
E -M o x c la v 6 2 5 m g
3- Compressros E p ib e z a
E p ic o til
4- Coaters E p ila c to n e European and Asian
E p ila t R e ta rd
5- Blisterers E p in o r
Countries
Uk
6- Packaging lines E p iriz in e
E p ite n s
Ireland
Albania
F u ra z o l
Romania
G a stro fa it 1 g
Ukraine
L ib ra x
Russia
N o rfle x
N o rg e sic Arm enia
N o -U ric 1 0 0 m g Georgea
N o -U ric 3 0 0 m g Moldova
N u -S p a sm Uzbekistan
R e g c o r 2 .5 m g Azerbaijan
R egcor 5m g Kyrgyzstan
R egcor 10m g Tajikestan
R h u m a -C u re 1 2 .5 m g Kazakhstan
R h u m a -C u re 2 5 m g Turkmenistan
S ec n id a z o le
S elg o n  
T e b o n in a
T ra n q u o m o n
T rittic o 5 0 m g
T rittic o 1 0 0 m g African Countries
Ghana
Eritria
Ethiopia
Kenya
Mali
Namibia
Benin
Somalia
Tanzania
Uganda

2.3 Spaghetti diagram


Spaghetti diagram for tablet manufacturing department is presented in Figure 3. It can be
seen that the layout has some inefficiencies. For example, after mixing, batches are
transported to compression then coating, the current layout puts coating between mixing
and compression. Also, after compressing, batches are transported to coating then
blistering, the current layout puts compression between coating and blistering. Finally,
the FG stock is very far from packaging which is the final stage in tablet manufacturing.

2.4 VSM
A value stream map or a process map provides a more detailed look at processes and
illustrates where improvements to processes can be implemented. The VSM is shown in
Figure 4. There is a bottleneck between the mixing line and the compression line, the
product is finished on the mixing line and waits from one to two hours to enter the
compression line, this causes a large time waste, also the product bulk may be exposed to
temperature and humidity. This problem can be solved by increasing the working shifts in
the compression line from two to three shifts, or increasing the number of compressing
machines.
Maximising return on investment (ROI) for pharmaceutical production 173

Figure 3 Spaghetti diagram (see online version for colours)

Figure 4 VSM (see online version for colours)


174 S.A. Ibrahim and N.A. Mostafa

2.5 Process mapping


Figure 5 illustrates process diagrams for a single production line, respectively.

Figure 5 Process map (see online version for colours)

3 Model analysis

3.1 Process simulation


The results of a simulation run are compiled in a simulation report that contains a
sampling of the statistics accumulated. A simulation report automatically appears upon
successful completion of a simulation run. Simulation results of the case study of tablet
manufacturing are illustrated in this section in Table 1 and Table 2, the elapsed time is
eight hours (one shift).
Table 1 Transaction overall statistics

Avg. resource
Count Avg. cycle Avg. work Avg. wait Avg. inact Avg. serv
wait
1,000 3.69 1.07 2.62 0.18 2.44 1.25

The simulation scenario is run for 1,000 counts, Table 1 provides the results about the
average cycle time for the full product (a package containing ten tablets in average) is
3.69 minutes, within them the average working time is 1.07 minutes and the average
waiting time is 2.62 minutes, actually it is common in batch processes manufacturing that
waiting times are relatively large. Table 2 provides the same results in Table 1 but for
detailed processes.
Maximising return on investment (ROI) for pharmaceutical production 175

Table 2 Transaction statistics in detail

Avg. Avg. Avg. Avg. Avg. Avg.


Count
cycle work wait res. wait inact serv
Preparation 1,000 2.78 0.40 2.38 0.12 2.26 0.52
Mixing 1,000 0.34 0.22 0.12 0.06 0.06 0.28
Compression 1,000 0.07 0.07 0.00 0.00 0.00 0.07
Coating 1,000 0.18 0.12 0.06 0.00 0.06 0.12
Blistering 1,000 0.12 0.12 0.00 0.00 0.00 0.12
Packaging 1,000 0.21 0.15 0.06 0.00 0.06 0.15

3.2 Data regression analysis

Data is stored in Minitab statistical application, then ‘fit data’ module is used to find a
supported random distribution that generates data similar to the existing data. Inferences
from graphs can be made about which distribution fits best, and the goodness-of-fit
measure from the statistics application is studied. After that, iGrafx uses the Minitab to
perform analysis, and then graphs the resulting analysis. Figures 6(a), 6(b) and 6(c),
illustrates the curve fitting for the described data. It was found that normal distribution
fits best to the data.

3.3 Design of experiment

The one factor at a time (OFAT) simulation analysis so far may miss interactions
between the changing factors. Instead of changing the resources one at a time by hand, a
DOE analysis can be performed on the process and try many different combinations of
resource levels automatically. With DOE, several factors can be changed at once and
easily the results can be analysed in iGrafx or Minitab.
In this experiment, one person is added to the original resource level in each
department. Responses can include transaction statistics such as cycle time or count
completed, scenario attributes, or custom statistics that can be defined. This part
tracks transaction completion count and resource costs, and looks for the total number of
transactions produced and the resource cost involved in producing the orders.
The response variable is cycle time, and it would be minimised. The factors are labour
resources in the four departments of mixing (A), compression (B), coating (C),
blistering and packaging (D) with two levels. Replications number is used to control
how many times each experiment is replicated and introduces more randomness into
the DOE. Replicates help make results more accurate, and they also greatly multiply
the number of simulations to run. This DOE defines 64 different simulations to run
(a 2-level full-factorial design on 4 factors is 24, or 16, and times 4 replicates each
(n = 4), is a total of 64). The experimental layout for this experiment is shown in
Table 3.
176 S.A. Ibrahim and N.A. Mostafa

Figure 6 Curve fitting, (a) cumulative distribution function (b) probability density function
(c) residuals plot (see online version for colours)

(a) (b)

(c)

The results are exported to a Minitab worksheet. It was found that, the highest number of
transactions is produced when compression and blistering both have 2 people working.
The packaging department varies between 1 and 2, and coating varies between 2 and 3.
Compression and blistering are solidly at 2 for the higher transaction counts. So, it is
suggested that if the department have 2 people each in compression and blistering, it will
be possible to reach around 80 or more orders.
Maximising return on investment (ROI) for pharmaceutical production 177

Table 3 Experimental layout

Run Factors Response (with replicates) Total (computed by


number adding responses for
A B C D 1 … n
each row)
1 –1 –1 –1 –1 (1)
2 1 –1 –1 –1 a
3 –1 1 –1 –1 b
4 1 1 –1 –1 ab
5 –1 –1 1 –1 c
6 1 –1 1 –1 ac
7 –1 1 1 –1 bc
8 1 1 1 –1 abc
9 –1 –1 –1 1 d
10 1 –1 –1 1 ad
11 –1 1 –1 1 bd
12 1 1 –1 1 abd
13 –1 –1 1 1 cd
14 1 –1 1 1 acd
15 –1 1 1 1 bcd
16 1 1 1 1 abcd

3.4 Using the simulation to confirm the results of the DOE


A resource is added to compression and blistering to test the theory that throughput can
be increased with one additional hire. Tables 4 and 5 provide the new simulation results
after adding new resources. Cycle time is reduced from 3.69 to 3.51 minutes, and
throughput is significantly increased. Resource constraints and other sources of
bottlenecks can have dramatic and hard-to-predict impact on a process. With simulation,
a business case for change can be build with statistics to back up the proposals, all
without impacting the actual production process or incurring excessive costs.
Table 4 Transaction overall statistics (modified)

Avg. resource
Count Avg. cycle Avg. work Avg. wait Avg. inact Avg. serv
wait
1,000 3.51 0.96 2.56 0.18 2.38 1.14
Table 5 Transaction statistics in detail (modified)

Avg. Avg. Avg. Avg. Avg. Avg.


Count
cycle work wait res. wait inact serv
Preparation 1,000 2.78 0.40 2.38 0.12 2.26 0.52
Mixing 1,000 0.34 0.22 0.12 0.06 0.06 0.28
Compression 1,000 0.03 0.03 0.00 0.00 0.00 0.03
Coating 1,000 0.18 0.12 0.06 0.00 0.06 0.12
Blistering 1,000 0.03 0.03 0.00 0.00 0.00 0.03
Packaging 1,000 0.15 0.15 0.00 0.00 0.00 0.15
178 S.A. Ibrahim and N.A. Mostafa

3.5 Analysis of variance


After developing the model, a test of hypothesis is done to reject infeasible products.
Minitab package is used to perform this test. An ANOVA is used to test the hypothesis
that the means of several populations are equal. The method is an extension of the
two-sample t-test. It can indicate if there are statistically significant differences among
the level means. The null hypothesis for the test is that all population means (level
means) are the same. The alternative hypothesis is that one or more population means
differ from the others. Table 6 presents ANOVA results. R2 = 94.85% R2 (adj) = 92.32%,
and p-value is significant to reject the null hypothesis.
Table 6 ANOVA overall results

Source DF SS MS F-statistic p-value


Factor 56 3.42216 0.0611 1.9672 0.012
Error 114 1.85804 0.0163 0.5241
Total 170 5.2802 0.0311

In addition to the above analysis, the output helps to determine which value of mean is
different. Tukey’s method was used to compare the means for each pair of factor levels
using a family error rate to control the rate of type 1 error. The family error rate is the
probability of making one or more type 1 errors for the entire set of comparisons.
Tukey’s method adjusts the individual confidence level based on the chosen family
error rate. t value is computed for each pair of means using the formula:
Mi − M j
t=
MSE
n
where Mi – Mj is the difference between the ith and jth means, MSE is the mean square
error, and n is the mean of the sample sizes of groups i and j.
Results are presented as a set of confidence intervals for the difference between pairs
of means. These intervals are used to determine whether means are different:
• if an interval does not contain zero, there is a statistically significant difference
between the corresponding means
• if the interval does contain zero, the difference between the means is not statistically
significant.
The results of Tukey test are shown in Figure 7 and indicate that:
• Clopam has the lowest mean demand (2,667), Ciprocin 500 mg and Rhuma-Cure
25 mg has the highest (1,828,833)
• the standard deviations for the different products vary from each other
• the pooled standard deviation is 127,666.
The means of products Acupan, Clopam, Colospasmin, and Epitens are not close to the
all other means so they are infeasible products that can be excluded from the plant future
plans.
Maximising return on investment (ROI) for pharmaceutical production 179

Figure 7 Means for Tukey’s test (see online version for colours)

4 Conclusions

Process simulation tools can play an important role throughout the life cycle of product
development and commercialisation. In process development, process simulation tools
are becoming increasingly useful as a means to analyse, communicate and document
process changes. Such tools also facilitate capacity analysis and debottlenecking tasks.
This work has demonstrated the use of process simulation and statistical tools in the
development of pharmaceutical manufacturing performance. Tablet manufacturing was
used as a representative example of pharmaceutical products. First, tablet manufacturing
process was described, SIPOC, spaghetti diagram, VSM, and process mapping were
performed using iGrafx package. It was found that the layout can be rearranged to make
movements between different sections more efficient. Through the VSM analysis, it was
found that there is a bottleneck and a time waste during the process, this problem can be
solved by increasing the working shifts in the compression line, or increasing the number
of compressing machines.
After that, a simulation scenario was run for 1,000 counts, average cycle times,
working and waiting times and other activity statistics were calculated. Then, regression
analysis was performed to fit data; it was found that data can be fitted to normal
distribution. A DOE analysis is performed to try many different combinations of
resources levels automatically. It was suggested that an additional worker can be added to
compression and blistering sections, after that a simulation is run to confirm the results; it
was found that cycle time is reduced by 5%, and throughput is significantly increased.
Finally, a test of hypothesis is done to reject infeasible products, Minitab package is
used to perform this test. An ANOVA is used to test the hypothesis that the means of
several populations are equal. The results have indicated that: The means of products
Acupan, Clopam, Colospasmin, and Epitens are infeasible products that can be excluded
from the plant future plans; this will maximise the ROI for the organisation.
180 S.A. Ibrahim and N.A. Mostafa

5 Future work

The pharmaceutical industry has only recently begun making significant use of process
simulation and scheduling tools. Increasingly, universities are incorporating the use of
such tools in their curricula. In the future, we can expect to see increased use of these
technologies and tighter integration with other enabling IT technologies, such as supply
chain tools, manufacturing execution systems (MES), batch process control systems,
process analytics tools (PAT), and so on. The result will be more robust processes and
efficient manufacturing leading to more affordable medicines.
Process mapping and simulation can be used as preliminary steps to study the
feasibility of the existing products, excluding the infeasible products leads to more
efficient use of resources, eventually this will maximise the ROI for the organisation.
The model discussed in this work can provide a useful, flexible and reliable solution
to maximise the ROI in an organisation through the use of process mapping, simulation
and statistical analysis of the data. It can be used in all industrial fields, not only
pharmaceuticals industry, and it can provide very good indicators for decision-makers.

Acknowledgements

This paper is a revised and expanded version of a paper entitled ‘Maximising return on
investment (ROI) for pharmaceutical production’ presented at 2nd International
Conference on Production and Industrial Engineering CPIE-2010.

References
Achilleos, E.C., Calandranis, J.C. and Petrides, D.P. (2006) ‘Quantifying the impact of uncertain
parameters in the batch manufacturing of active pharmaceutical ingredients’, Pharmaceutical
Engineering, Vol. 26, No. 4, pp.34–40.
Alexander, C.W. (2006) ‘Discrete event simulation for batch processing’, Proceedings of the 2006
Winter Simulation Conference, pp.1929–1934.
Applequist, G.E., Pekny, J.F. and Reklaitis, G.V. (2000) ‘Risk and uncertainty in managing
chemical manufacturing supply chains’, Computers of Chemical Engineering, Vol. 24,
pp.2211–2222.
Ball, P. (2001) ‘Introduction to discrete event simulation’, originally presented at the 2nd
DYCOMANS Workshop on Management and Control: Tools in Action, Algarve, Portugal,
15–17 May 1996, pp.367–376.
Barnett, M.W. (2003) Modeling & Simulation in Business Process Management, Gensym
Corporation, available at http://www.ww.bptrends.com.
Blau, G.E., Mehta, B., Bose, S., Pekny, J.F., Sinclair, G., Kuenker, K. and Bunch, P. (2000) ‘Risk
management in the development of new products in highly regulated industries’, Computers
and Chemical Engineering, Vol. 24, Nos. 2–7, p.659.
Dangelmaier, W., Fischer, M., Gausemeier, J., Grafe, M., Matysczok, C. and Mueck, B. (2005)
‘Virtual and augmented reality support for discrete manufacturing system simulation’,
Computers in Industry, Vol. 56, pp.71–383.
Floudas, C.A. and Lin, X. (2004) ‘Continuous-time versus discrete-time approaches for
scheduling of chemical processes: a review’, Computers and Chemical Engineering, Vol. 28,
pp.2109–2129.
Maximising return on investment (ROI) for pharmaceutical production 181

Folan, P. and Brown, J. (2005) ‘A review of performance measurement: towards performance


management’, Computers in Industry, Vol. 56, pp.663–680.
Forza, C. and Salvador, F. (2001) ‘Information flows for high performance manufacturing’,
International Journal of Production Economics, Vol. 70, pp.21–36.
Gonzalez, R. and Realff, M.J. (1998) ‘Operation of pipeless batch plants – I. MILP schedules’,
Computers and Chemical Engineering, Vol. 22, Nos. 7–8, p.841.
Higuchi, T. and Troutt, M.D. (2004) ‘Dynamic simulation of the supply chain for a short life cycle
product – lessons from the Tamagotchi case’, Computers & Operations Research, Vol. 31,
No. 7, pp.1097–1114.
Honkomp, S.J., Mockus, L. and Reklaitis, G.V. (1999) ‘A framework for schedule evaluation with
processing uncertainty’, Computers and Chemical Engineering, Vol. 23, Nos. 4–5, p.595.
Hwang, F. (1997) ‘Batch pharmaceutical process design and simulation’, Pharmaceutical
Engineering, January/February, pp.28–43.
Ibrahim, S. and Mostafa, N. (2011) ‘Using multi-agent systems for securing pharmaceutical supply
chains’, International Journal of Economic Resources, Vol. 2, No. 2, pp.109–123.
Lohman, C., Fortuin, L. and Wouters, M. (2004) ‘Designing a performance measurement system: a
case study’, European Journal of Operational Research, Vol. 156, pp.267–286.
Micrografx (2001) ‘The role of process modeling and management within six sigma’, White paper,
Dallas, Texas, available at
http://courses.washington.edu/outfox/IgrafxProcessMgtandSix%20Sigma.pdf.
Papavasileiou, V., Koulouris, A., Siletti, C. and Petrides, D. (2007) ‘Optimize manufacturing of
pharmaceutical products with process simulation and production scheduling tools’,
Trans. IChemE, Part A, Chemical Engineering Research and Design, Vol. 85, No. A7,
pp.1086–1097.
Petrides, D.P. and Siletti, C.A. (2004) ‘The role of process simulation and scheduling tools in the
development and manufacturing of biopharmaceutical’, in Ingalls, R.G., Rossetti, M.D.,
Smith, J.S. and Peters, B.A. (Eds.): Proceedings of the 2004 Winter Simulation Conference,
pp.2046–2051.
Petrides, D.P., Koulouris, A. and Lagonikos, P.T. (2002) ‘The role of process simulation in
pharmaceutical process development and product commercialization’, Pharmaceutical
Engineering, Vol. 22, No. 1, pp.1–8.
Reklaitis, G.V. (1992) ‘Overview of scheduling and planning of batch process operations’, NATO
ASI on Batch Process Systems Engineering, Antalya, Turkey.
Shah, N. (2004) ‘Pharmaceutical supply chains: key issues and strategies for optimization’,
Computers and Chemical Engineering, Vol. 28, pp.929–941.
Subramanian, D., Pekny, J.F. and Reklaitis, G.V. (2001) ‘A simulation-optimization framework
for research and development pipeline management’, AIChE Journal, Vol. 47, No. 10,
pp.2226–2242.
Tan, J., Foo, D.C.Y., Kumaresan, S. and Aziz, R.A. (2006) ‘Debottlenecking of a batch
pharmaceutical cream production’, Pharmaceutical Engineering, July/August, pp.72–82.
Taylor, S.G., Seward, S.M. and Bolander, S.F. (1981) ‘Why the process industries are different’,
Prod. Inventory Manage, Fourth Quarter, pp.9–24.
Thomas, C.J. (2003) ‘A design approach to biotech process simulations’, BioProcess International,
Vol. 1, No. 10, pp.2–9.
Yu, Q., Ming, P. and Yacai, H. (2009) ‘Modeling and optimization for scheduling of chemical
batch processes’, Chinese Journal of Chemical Engineering, Vol. 17, No. 1, pp.1–7.
Zorzut, S., Jovan, V., Gradisar, D. and Music, G. (2009) ‘Closed-loop control of a polymerisation
plant using production performance indicators (PIs)’, International Journal of Computer
Integrated Manufacturing, Vol. 22, No. 12, pp.1128–1143.

View publication stats

You might also like