77
Freeze-Drying Manufacturing Process Optimization: Technology Transfer and Scale-Up Carolina Cipriano Carvalho Thesis to obtain the Master of Science Degree in Pharmaceutical Engineering Supervisors: Eng. Samuel Mendes Geraldes Camocho Prof. Maria Cristina de Carvalho Silva Fernandes Examination Committee Chairperson: Prof. José Monteiro Cardoso de Menezes Supervisor: Eng. Samuel Mendes Geraldes Camocho Members of the Committee: Prof. João Fernandes de Abreu Pinto November 2019

Freeze-Drying Manufacturing Process Optimization

Embed Size (px)

Citation preview

Freeze-Drying Manufacturing Process Optimization:

Technology Transfer and Scale-Up

Carolina Cipriano Carvalho

Thesis to obtain the Master of Science Degree in

Pharmaceutical Engineering

Supervisors: Eng. Samuel Mendes Geraldes Camocho

Prof. Maria Cristina de Carvalho Silva Fernandes

Examination Committee

Chairperson: Prof. José Monteiro Cardoso de Menezes

Supervisor: Eng. Samuel Mendes Geraldes Camocho

Members of the Committee: Prof. João Fernandes de Abreu Pinto

November 2019

i

I declare that this document is an original work of my own and that it fulfills all the requirements of the

Code of Conduct and Good Practices of the Universidade de Lisboa.

ii

iii

PREFACE

The work presented in this thesis was performed at the company Hikma Pharmaceuticals (Terrugem,

Portugal), during the period February-July 2019, under the supervision of Eng. Samuel Camocho. The

thesis was co-supervised at Instituto Superior Técnico by Prof. Maria Cristina Fernandes.

iv

v

ACKNOWLEDGMENTS

I would first like to thank to my supervisor, Eng. Samuel Camocho, for giving me the opportunity of

achieving this work at Hikma Pharmaceuticals and for his guidance and knowledge, that were

fundamental. I also thank to Prof. Maria Cristina Fernandes for her support and orientation through this

work. Particular thanks to Ana Rita Martins, Joana Henriques and Telma Ferreira for their daily work

support through my internship at Hikma Pharmaceuticals, as well as for being extraordinary colleagues

whose friendship I hope to take through my life. To all the lyophilization production team, thank you for

welcoming me as part of the team. My thanks are also extended to the Validation, Regulatory Affairs

and Technical Services teams for providing me the knowledge and documentation for my research and

writing.

To my old friends, I would like to thank for their friendship and presence in my life.

Finally, I would like to express my gratitude and love to my family, who supported me during my entire

life. A special thanks to my mom, Carlos and my brother Martim, for giving me continuous

encouragement throughout my years of study. This accomplishment would not have been possible

without them. Thank you.

vi

vii

ABSTRACT

The main goal of this work is to contribute to the knowledge about scale-up and technology transfer of

the manufacturing process of lyophilized injectable pharmaceutical products.

An effective process validation approach must be implemented to ensure that the manufacturing process

is under control and consistently delivers a product within specifications during its lifecycle. Knowledge

management and identification of critical process parameters and critical quality attributes enable to

assess the level of risk and their impact on the product and process. Based on the risk assessment, an

effective control strategy is established.

Aseptic processing is inherent to the manufacture of lyophilized injectable drug products. Aseptic

operations are conducted by qualified personnel in classified areas according to the level of

contamination risk. Containers and closures are sterilized before contact with the sterile product. Aseptic

processing is validated through process simulations with microbiological growth medium, to ensure the

sterility of lyophilized injectable product to be delivered to patients.

A new lyophilized product to be manufactured in Hikma Portugal and commercialized to US market is

presented. The respective manufacturing process is described. The process optimization for this product

comprises a line transfer and a scale-up. Therefore, risk assessment and validation activities were

carried out and the results discussed.

Keywords: Process validation, technology transfer, scale-up, risk assessment, lyophilization

viii

ix

RESUMO

O objetivo principal da tese é contribuir para o conhecimento sobre o aumento de escala e transferência

de tecnologia do processo de produção de medicamentos liofilizados injetáveis.

Uma abordagem de validação de processo é implementada para garantir que o processo de produção

está sob controlo e que produz consistentemente um produto dentro de especificações durante o seu

ciclo de vida. A gestão do conhecimento e identificação de parâmetros críticos do processo e de

atributos de qualidade críticos permitem a análise do nível de risco e respetivo impacto no produto e no

processo. Com base na análise de risco, é estabelecida uma estratégia de controlo.

Processamento assético é inerente à produção de medicamentos liofilizados injetáveis. Operações

asséticas são efetuadas por pessoal qualificado em áreas classificadas de acordo com o nível de risco

de contaminação. A embalagem primária é esterilizada antes de entrar em contacto com o produto

estéril. O processo assético é validado através de simulações do processo com meio de cultura

microbiológico, de forma a garantir a esterilidade do produto liofilizado injetável a ser administrado aos

pacientes.

Um novo produto liofilizado a ser produzido na fábrica Hikma em Portugal e comercializado para o

mercado dos Estados Unidos é apresentado. A otimização do processo para este produto inclui uma

transferência de linha e um aumento de escala, o que implica uma análise de risco e atividades de

validação, cujos resultados são discutidos.

Palavras-chave: Validação de processo, transferência de tecnologia, aumento de escala, análise de

risco, liofilização

x

xi

TABLE OF CONTENTS 1 Introduction .................................................................................................................................... 1

2 Literature review ............................................................................................................................ 3

2.1 Process validation ................................................................................................................. 3

2.1.1 Definition and concepts ............................................................................................ 3

2.1.2 Validation approaches .............................................................................................. 3

2.1.3 Process validation stages ......................................................................................... 4

2.1.4 Regulatory requirements for process validation ....................................................... 8

2.1.5 Connection of Process Validation, Pharmaceutical Quality System and Quality Risk

Management .......................................................................................................................... 8

2.1.6 Documentation ........................................................................................................ 13

2.1.7 Technology transfer as part of process validation .................................................. 13

2.2 Sterile drug products manufacturing ................................................................................... 15

2.2.1 Aseptic processing .................................................................................................. 15

2.2.2 Room classification ................................................................................................. 15

2.2.3 Personnel qualification............................................................................................ 16

2.2.4 Containers and closures sterilization ...................................................................... 16

2.2.5 Filtration validation .................................................................................................. 16

2.2.6 Microbiological testing ............................................................................................ 17

2.2.7 Validation of aseptic processing ............................................................................. 17

3 Hikma lyophilization production department overview ........................................................... 19

3.1 Production lines ................................................................................................................... 19

3.2 Lyophilization process ......................................................................................................... 20

3.3 Technology transfer from line 1 to line 9 ............................................................................. 21

4 Submission of new lyophilized product A ................................................................................ 27

4.1 Product submission and approval for US market ................................................................ 27

4.2 Product and process specifications ..................................................................................... 28

4.3 Manufacturing process ........................................................................................................ 29

5 Product A launch and scale-up in line 1 ................................................................................... 31

5.1 Risk assessment and control............................................................................................... 31

5.2 Process validation activities and results in line 1 ................................................................ 33

5.3 Validated parameters for product A in line 1 ....................................................................... 38

xii

6 Process optimization: scale-up and transfer to line 9 ............................................................. 39

6.1 Process validation activities and results in line 9 ................................................................ 39

6.2 Validated parameters for product A in line 9 ....................................................................... 44

6.3 Deviations during validation batch ....................................................................................... 45

6.3.1 Investigation to OOS bioburden analysis ............................................................... 45

6.3.2 Investigation to OOS colour of solution .................................................................. 46

6.4 Corrective Actions and Preventive Actions (CAPA) ............................................................ 47

7 Conclusion ................................................................................................................................... 49

7.1 Contributions ....................................................................................................................... 49

7.2 Future work .......................................................................................................................... 50

References ........................................................................................................................................... 51

Annexes .................................................................................................................................................. A

Annex A – Risk management tool FMECA for process validation of product A .............................. A

Annex B – Sampling plan for process validation of scale-up of product A in line 1 ........................ C

Annex C – Sampling plan for process validation of product A transfer to line 9 ............................. E

xiii

LIST OF FIGURES

Figure 1. Process validation lifecycle approach. (CQAs – Critical Quality Attributes; CPPs – Critical

Process Parameters) ............................................................................................................................... 4

Figure 2. Overview of a typical quality risk management process. Adapted from [9]............................. 9

Figure 3. Product lifecycle at Hikma Pharmaceuticals. ........................................................................ 21

Figure 4. Product A lifecycle at Hikma Pharmaceuticals. ..................................................................... 27

Figure 5. Manufacturing process of product A...................................................................................... 30

Figure 6. Sampling plan for process validation of scale-up of product A on line 1................................. C

Figure 7. Sampling plan for process validation of product A transfer to line 9. ...................................... E

xiv

xv

LIST OF TABLES

Table 1. Risk classification levels. Level 1 (red); Level 2 (orange) and Level 3 (green). Adapted from

[10] ......................................................................................................................................................... 11

Table 2. Risk Priority Number (RPN) calculation. High RPN (red), medium RPN (orange) and low RPN

(green). Adapted from [10] .................................................................................................................... 12

Table 3. Clean area air classifications by limits of airborne particle concentration for non-viable particles

monitoring .............................................................................................................................................. 16

Table 4. Hikma's type of products manufactured at each production line and facility .......................... 19

Table 5. Comparison of room classification between line 1 and line 9 ................................................. 23

Table 6. Process requirements for product A ....................................................................................... 28

Table 7. Number of validation batches required for each validation activity comprised in the scale up for

product A in line 1 .................................................................................................................................. 34

Table 8. Results obtained for dead volume samples for PV of scale-up in line 1................................. 35

Table 9. Filling machine speed results obtained for the PV scale-up in line 1 based on volume

specifications ......................................................................................................................................... 36

Table 10. Results obtained from the beginning, middle and end of filling samples for the three validation

batches .................................................................................................................................................. 36

Table 11. Validated parameters after PV of scale-up from 100 L to 200 L in line 1 ............................. 38

Table 12. Number of validation batches required for each validation activity comprised in the scale up

and line transfer process validation for product A ................................................................................. 39

Table 13. Results obtained from top and bottom of the compounding tank samples for quality of the

compounded bulk solution evaluation ................................................................................................... 40

Table 14. Results obtained from transference tank samples for bulk holding time studies .................. 41

Table 15. Results obtained for dead volume samples for line transfer to line 9 ................................... 41

Table 16. Filling machine speed results obtained for the first validation batch based on volume

specifications in line 9 ............................................................................................................................ 42

Table 17. Results obtained from the beginning, middle and end of filling samples in line 9 ................ 42

Table 18. Finished product A certificate of analysis ............................................................................. 43

Table 19. Validated parameters after PV of line transfer to line 9 and scale-up from 100 L to 400 L .. 44

Table 20. Summary of colour test results from RLD samples used to establish the colour specification

............................................................................................................................................................... 47

Table 21. FMECA tool application for process validation of product A. .................................................. A

xvi

xvii

LIST OF ACRONYMS

ANDA API CAPA CGMP CIP CMA CPP CQA

Abbreviated New Drug Application Active Pharmaceutical Ingredient Corrective Action and Preventive Action Current Good Manufacturing Practices Cleaning in Place Critical Material Attribute Critical Process Parameter Critical Quality Attribute

DOE Design of Experiments FDA FMEA FMECA

Food and Drug Administration Failure Mode Effects Analysis Failure Mode, Effects and Criticality Analysis

ICH IPC MBR MENA NMT OOS PAT

International Conference on Harmonization In Process Control Master Batch Record Middle East and North Africa Not More Than Out of Specification Process Analytical Technology

PPQ PQ PT

Process Performance Qualification Process Qualification Portugal

PV QA QC QS RABS R&D

Process Validation Quality Assurance Quality Control Quantum Sufficit Restricted Access Barrier System Research and Development

xviii

RLD RU SIP SOP SU US VHP VMP WFI WHO

Reference Listed Drug Receiving Unit Sterilization in Place Standard Operating Procedure Sending Unit United States Vaporized Hydrogen Peroxide Validation Master Plan Water for Injection World Health Organization

1

1 INTRODUCTION

Process Validation (PV) provides assurance that a process is capable of effectively and consistently

delivering a product meeting its intended quality attributes. A planned process optimization, such as

technology transfer or scale-up of a product, implies a PV approach to guarantee the new process

performance and product quality.

The aim of this work is to contribute to the knowledge about manufacturing of lyophilized injectable

pharmaceutical products. Specifically, about PV regarding scale-up and technology transfer between

production lines at Hikma Pharmaceuticals manufacturing site in Portugal.

The first chapter of this thesis is a literature review focused on PV concepts and principles of aseptic

processing for the manufacture of sterile drug products, with the purpose of providing the required

background to understand the matters presented in the next chapters. The literature review is based on

guidelines and documentation from Food and Drug Administration (FDA), International Council for

Harmonization (ICHs) and World Health Organization (WHO).

The following chapter is an overview of the lyophilization production department at Hikma, including a

description of the lyophilization process, as well as the general principles of technology transfer between

production line 1 and line 9.

In the next chapter the product approached in this thesis is presented, along with its submission for

commercialization in United States (US) market, its manufacturing process and requirements. The two

following chapters focus on the PV for scale-up and line transfer of the product between production lines

1 and 9. The validation activities performed, together with the respective results of the manufactured

batches are presented. Investigations conducted to deviations detected during the manufacturing of

validation batches are described, as well as the Corrective Actions, Preventive Actions (CAPA) adopted.

As final chapter, conclusions and future work perspectives are presented.

2

3

2 LITERATURE REVIEW

2.1 PROCESS VALIDATION

2.1.1 Definition and concepts

Process validation is defined by FDA, European Commission and ICH as the collection and evaluation

of data throughout the product lifecycle that provides documented scientific evidence that a process is

capable of effectively and consistently delivering a product meeting its intended quality attributes.

Validation is usually preceded by qualification and both follow similar underlying principles. Quality must

be designed and built into the product through its lifecycle but cannot be properly assured by testing into

the product. Validation and qualification should be performed for new equipment and utilities, systems,

methods or processes or when changes are planned. Both should be executed in compliance with

regulatory requirements and quality risk management principles must be applied. Appropriate

personnel, financing and time resources are needed in order to plan and execute validation and

qualification activities [1, 2].

The lifecycle concept connects product and process development, validation of the commercial

manufacturing process and maintenance of the process in a state of control during routine commercial

production, which means that the defined set of controls consistently provides assurance of the process

performance and product quality [1, 3].

2.1.2 Validation approaches

Validation can be performed through four different approach types: prospective, concurrent,

retrospective and revalidation [4].

Prospective validation consists in establishing documented evidence of the process performance and

reproducibility prior to its implementation, based on pre-planned protocols. It is normally applied during

the development stage to validate the process before routine commercial manufacturing and is carried

based on a risk analysis of the process, which is separated into individual steps. These steps are

evaluated regarding experience to determine if they may lead to critical situations.

Concurrent validation is exceptional performed during routine production with the intent for

commercialization when data from replicate production is still not available. In this case, thorough

monitoring and testing of the produced batches is performed and if evidence is provided on the process

reproducibility, individual batches may be released prior to completion of the validation [2, 5].

Both prospective and concurrent validation are based on evidence obtained through testing. The testing

may include extensive sampling to demonstrate intra- and inter-batch homogeneity and worst-case tests

to determine the robustness of the process. Simulation process trials may be performed to validate, as

4

example, the aseptic filling of parenteral products such as lyophilized products that cannot be terminally

sterilized. [4, 6].

Retrospective validation is acceptable for well-established processes, since it relies on the analysis

of historical data. The results from in-process and final product testing are combined with available

historical data and treated statistically. The trend analysis performed enables to conclude on the extent

which the process parameters are within the acceptable range and will indicate whether the process is

under control or not.

Whenever possible, prospective validation is preferred. Retrospective validation is not encouraged and

is not applicable to the manufacturing of sterile products [4, 6].

Revalidation is a repeated validation of a previously validated process to ensure continued compliance

with the established requirements. This approach is adopted when there is the need to prove that

planned or unplanned changes in the process do not affect the process performance nor the product

quality. These changes may include changes to the product, the manufacturing process, the equipment

or technology transfers [2].

2.1.3 Process validation stages

Process validation involves activities covering the product and process lifecycle, which can be described

in three stages: process design, process qualification and continued process verification.

Figure 1. Process validation lifecycle approach. (CQAs – Critical Quality Attributes; CPPs – Critical Process Parameters)

Process monitoring and

improvement

Design of Facilities &

Qualification of

Equipment and Utilities

Process Performance

Qualification (PPQ)

Determine CQAs and

identify CPPs

Define control strategy

Stage 2

PROCESS QUALIFICATION

Stage 3

CONTINUED PROCESS VERIFICATION

Stage 1

PROCESS DESIGN

5

I. Process design

The aim of process design is to define a manufacturing process suitable for routine commercial

manufacturing that consistently delivers a product meeting its quality attributes.

It is not obligatory to follow the current Good Manufacturing Practices (cGMP) requirements for the initial

process design experiments, although they must be conducted according to good documentation

practices and scientific methods and principles. The controls defined during this stage must be

adequately documented and justified, since documentation must reflect the basis for decisions made

about the process, considering its value for use and adaptation throughout the product lifecycle.

A crucial step in process design is to build and capture process knowledge and understanding.

Information provided from product development activities such as intended dosage form, quality

attributes and a manufacturing process overview represents key inputs to the process design stage. All

sources of variation such as functionality and limitations of equipment, different components batches,

production operators and measurement systems must be contemplated in the process design stage. All

information enhancing the process understanding must be documented, since it is useful for process

qualification and continued process verification stages.

An effective process control strategy is based on the process knowledge and understanding.

Manufacturers should understand the sources, the level and the impact of variation on the process and

product attributes to be able to control the variation in a proportional manner with the risk it represents

to the process and product. Design of Experiments (DOE) studies help building process knowledge by

revealing relationships between variable inputs – Critical Material Attributes (CMAs) and Critical Process

Parameters (CPPs) - and the resulting outputs – Critical Quality Attributes (CQAs). Risk assessment

can be performed to evaluate potential variables for DOE studies. The results of this type of studies

provide an explanation for establishing ranges of incoming component quality, equipment parameters

and in-process material quality attributes. Laboratory or pilot scale experiments and computer models

can also contribute to process knowledge enhancement.

Process control strategies may be designed to reduce input variation, adjust for input variation during

manufacturing or a combination of both. FDA expects controls to include examination of material quality

and equipment monitoring. It is necessary to have a special process control when the product attribute

is not readily measurable due to limitations of sampling or detectability or when intermediates and

products cannot be highly characterized and well-defined quality attributes cannot be identified. The

established controls are included in master production record.

The designed process at the end of this stage, including the operational limits and process control

strategy must be transferred to the next stage of process validation [1].

6

II. Process qualification

The goal of process qualification (PQ) stage is to evaluate and determine if the process design is capable

of reproducible commercial manufacture. This stage must be followed through current GMP procedures

and completed before commercial distribution. It involves two principles:

- Facility design and utilities and equipment qualification;

- Process Performance Qualification (PPQ).

The activities required to determine that utilities and equipment are suitable for their expected use and

perform properly must precede production at commercial scale. Qualification activities consist of

selection of construction materials, verification that utilities and equipment are installed in compliance

with the design specifications and operate according with process requirements in all anticipated

operating ranges, which must be shown capable of being held during routine production.

The qualification plan must contemplate the studies or tests to be completed, timing of qualification

activities, suitable criteria to evaluate outcomes, responsibilities of each department and procedures for

documentation and approval of qualification.

After the facility, utilities and equipment are qualified, it is necessary to confirm and demonstrate that

the commercial manufacturing process designed performs as intended through PPQ stage.

At PPQ stage, commercial-scale batches (denominated validation batches in Hikma) are manufactured

in order to provide data that supports the decision to start commercial distribution. The batches

manufactured at this stage require a higher level of sampling and testing than the one performed during

routine commercial production, in order to confirm the product quality uniformity throughout all

manufacturing steps. The goal is to obtain a sufficient level of process knowledge and to provide

adequate assurance that the commercial manufacturing process performs as expected.

Manufacturing conditions, controls, sampling and testing during this stage are specified in a written PPQ

protocol stating how activities will be conducted during the validation batches manufacturing. The

protocol must approach the qualification of utilities, equipment and personnel, the validation status of

analytical methods to be used, tests to be performed and acceptance criteria. The sampling plan

includes sampling points and frequency and the number of samples, which should be proper to provide

a high confidence level of intra-batch and inter-batch quality. The protocol is reviewed and approved by

the relevant departments and quality unit.

Deviations to the protocol are allowed when properly justified and approved by all relevant departments

and the quality unit before implementation. The validation batches must be manufactured under normal

operating conditions regarding utility systems, material, personnel and manufacturing conditions.

After realization of all validation activities established in the protocol, a PPQ report must be issued. This

report must include a summary and analysis of the data collected, evaluation of unexpected

observations or deviations to the protocol as well as a description of corrective actions to be

implemented to the existing procedures and controls. The knowledge gained from the design stage

7

through the process qualification stage must be compiled to state a clear conclusion if the process met

the established conditions and is in a state of control. If that is not verified, the report must include what

should be done before reaching that conclusion. Review and approval of the report by relevant

departments and quality unit is required [1].

Release of PPQ batches:

The manufacture of the initial validation batches should be successfully completed prior to commercial

distribution, except under special circumstances where concurrent release may be acceptable. These

circumstances include short shelf-life products and orphan drug products that are not frequently

manufactured. In such cases, product distribution may occur concurrently with the release.

Advanced control strategies such as Process Analytical Technology (PAT) that continuously monitor,

evaluate and adjust the manufacturing process using validated in-process measurements, tests,

controls, and process endpoints can provide a high level of quality assurance. Manufacturing processes

developed and controlled in such a manner may not require the manufacture of multiple validation

batches prior to initial distribution [1, 7].

III. Continued process verification

Continued process verification stage consists of providing ongoing assurance that the defined routine

commercial manufacturing process maintains the validated state. Assuming good development of the

process, identification of potential variation and an effective control strategy, the manufacturer must

maintain the process under control over the product lifetime. This stage comprises the routine

commercial manufacturing under similar conditions as demonstrated in process qualification stage.

Although a good process design implies anticipation and appropriate detection of sources of variability,

it is probable for a process to encounter sources of variation that were not previously detected or to

which the process was not exposed.

A continuous program to collect and analyse product and process data including relevant process trends

and quality of incoming materials or components, in-process material and finished products must be

established. The collected data should be statistically trended and reviewed to detect unplanned process

variability and conclude about necessary adjustments of monitoring levels and process improvement

changes.

Variation can also be detected through defect complaints, Out-of-specification (OOS) findings, process

yield variations, adverse event reports or by provided feedback on process performance from production

line operators and quality unit.

Maintenance of the facility, equipment and utilities is just as important to ensure the remaining state of

process control [1].

8

2.1.4 Regulatory requirements for process validation

Process validation of manufacturing processes for finished pharmaceuticals and components is a legal

requirement by the current cGMP regulations. It requires manufacturing processes to be designed and

controlled to assure that in-process materials and the finished product consistently and reliably meet

predetermined quality attributes [1, 7].

Procedures for process control must be defined to provide assurance that the product has the identity,

strength, quality and purity it is intended to. These procedures must be reviewed and approved by the

quality unit and should include the following elements:

- The batch must be formulated with the purpose to provide not less than 100 percent of the

labelled amount of active ingredient;

- Components shall be weighted, measured or subdivided as appropriate and the containers must

be properly identified;

- Weighing, measuring or subdividing operations must be supervised;

- Each component must be added to the batch by one person and verified by a second person.

The cGMP regulations for sampling require that samples must be representative of the batch under

analysis, the sampling plan must result in statistical confidence and the batch must meet its

predetermined specifications.

When establishing in-process specifications, these must be consistent with finished product

specifications and shall be derived from previous acceptable process average and process variability

estimates. These requirements determine the need for manufacturers to control in-process material to

assure that the finished product will meet its quality attributes and to evaluate process performance and

control batch variability.

The cGMP regulations also require information and data regarding product quality and process

knowledge to be periodically reviewed to determine if any changes to the established process are

assured.

Facilities and equipment proper design, construction and location are also a cGMP regulatory

requirement, in order to facilitate proper operations within the manufacturing process [1, 8].

2.1.5 Connection of Process Validation, Pharmaceutical Quality System and Quality

Risk Management

The Pharmaceutical Quality System (PQS) consists of a management system to direct and control a

pharmaceutical company regarding quality. Process validation within PQS is defined as assurance

based on evidence that a process consistently produces a product meeting its predetermined

specifications.

9

Implementation of an effective PQS over the product lifecycle (pharmaceutical development, technology

transfer, commercial manufacturing and product discontinuation) should result in a well-established and

maintained state of control and should facilitate innovation and continual improvement.

An essential element of the PQS is the CAPA system. Following investigations of complaints, product

rejections, non-conformances, deviations, audits, regulatory inspections or trends from process

performance, is inherent the application of a CAPA approach. This methodology should result in product

and process improvements and an enhanced product and process knowledge.

Quality Risk Management (QRM) and knowledge management are enablers to an effective

implementation of PQS. Knowledge management consists of a systematic approach to acquire, analyse,

store and disseminate information related to product, manufacturing process and components [3].

QRM is integral to an effective PQS. It can provide a proactive and systematic approach to assess,

control, review and communicate potential risks to quality and facilitate continual improvement of

process performance and product quality over the product lifecycle. QRM supplies reproducible

methods to assess the probability, severity and detectability of the risk [3, 9].

A risk is defined as the combination of the probability of occurrence of harm and the severity of that

harm. The evaluation of risk to quality should be based on scientific knowledge and ultimately linked to

the protection of the patient. The level of scrutiny and documentation of the QRM process should be

commensurate with the risk level. Figure 2 shows a model for QRM process presented in ICH Q9

guidance [9].

Figure 2. Overview of a typical quality risk management process. Adapted from [9].

10

RISK ASSESSMENT includes the identification of hazards and the analysis and evaluation of risks

associated with exposure to those hazards. It begins with a well-defined problem description that allows

to identify the most appropriate risk management tool and to answer the questions “What might go

wrong?”, “What is the likelihood it will go wrong?” and “What are the consequences (severity)?”.

I. Risk identification answers the first question of risk assessment. The hazards are identified

through a systematic use of information including historical data, theoretical analysis, informed

opinions and concerns of stakeholders.

II. Risk analysis is the estimation of the risk associated with the identified hazards. A risk

management tool is applied in order to qualitatively or quantitatively link the likelihood of

occurrence with the severity of harms. For each risk, the probability, severity and detectability

should be identified.

Frequency of probability may be rated from 1 to 3 according to its likelihood:

- Level 1 (Low): the frequency of the event occurring is perceived to be once per ten

thousand transactions;

- Level 2 (Medium): the frequency of the event occurring is perceived to be once per

thousand transactions;

- Level 3 (High): the frequency of the event occurring is perceived to be once per hundred

transactions.

Severity of the potential effect of the failure requires considering the impact of the event on the

product quality. The impact of the consequence may also be rated from 1 to 3:

- Level 1 (Low): expected to have a minor negative impact; the damage is not expected to

have a long-term detrimental effect;

- Level 2 (Medium): expected to have a moderate impact; the impact can be expected to

have a short to medium term detrimental effect;

- Level 3 (High): expected to have a very high significant negative impact; the impact could

be expected to have significant long-term effects and potentially catastrophic short-term

effects.

The risk is classified by multiplication of probability with severity. It is divided in three levels, as

shown in Table 1.

- Level 1 indicates that the probability of the failure occurs and the impact on product quality

are both high or medium;

- Level 2 indicates a moderate impact on product quality;

- Level 3 indicates that practically there is not impact on product quality.

11

Table 1. Risk classification levels. Level 1 (red); Level 2 (orange) and Level 3 (green). Adapted from [10]

Probability

Severity

1 (Low) 2 (Medium) 3 (High)

3 (High) 3 6 9

2 (Medium) 2 4 6

1 (Low) 1 2 3

The detectability aims to identify if the risk can be recognized or detected by other means in the

system. It is rated from 1 to 3 according to the risk detection possibilities:

- Level 1: a reliable detection device is continuously used on the system for direct parameter

measurement and leads to alarm activation or automatic system safe reconfiguration in

case of threshold overrunning (secured);

- Level 2: a reliable detection procedure is systematically applied however, it gives delayed

results. Another option is the use of an indirect measuring device, or a direct measuring

device in line with no alarm (insufficient secured);

- Level 3: no reliable detection device is used nor detection procedure as part of the system

operation or monitoring (not secured).

III. Risk evaluation consists of comparing the identified and analysed risk against the acceptable

risk level. In this step of risk assessment, the strength of evidence of the three questions is

considered [9, 10].

The output of a risk assessment is either a quantitative estimate of risk or a qualitative description of a

range of risk. When risk is expressed quantitatively, a numerical probability is used. Alternatively, risk

can be expressed using qualitative descriptors, such as “high”, “medium”, or “low”, which should be

defined in as much detail as possible. After risk assessment is concluded, the risk management process

continues to risk control and risk review.

RISK CONTROL implies decision making to reduce and/or accept risks. It aims to reduce the risk to an

acceptable level. The effort applied to risk control should be commensurate to the significance of the

risk.

I. Risk reduction addresses the mitigation or avoidance of risk when it exceeds an acceptable

level. Measures to mitigate the severity and likelihood of harm or to improve the detectability of

hazards may be applied.

II. Risk acceptance can be a formal decision to accept the residual risk or a passive decision in

which residual risks are not specified.

RISK REVIEW aims the need to run over the output of risk management process to consider new

obtained knowledge and experience which can lead to reconsideration of the determined risk

acceptance. The frequency of review should be proportional to the risk level.

12

RISK COMMUNICATION involves sharing the risk management output with all interested parts at any

stage of the risk management process. The communication is essential to facilitate trust and

understanding about the risks [9].

Risk assessment tools are useful to assess and manage risk. For the presented work in this thesis, the

Failure Mode, Effects and Criticality Analysis (FMECA) will be applied.

FMECA is an extension of the Failure Mode Effects Analysis (FMEA) risk assessment tool. FMEA

provides for an evaluation of potential failure modes for processes and their likely effects on the process

performance, based on product and process understanding. It methodically breaks down the analysis

of complex processes into manageable steps and enables the identification of potential failure modes,

their causes and likely effects. The application of FMEA tool implies the classification of each failure

mode by severity, occurrence and detection and the calculation of Risk Priority Number (RPN). The

RPN is a multiplication of the probability, severity and detectability parameters determined during risk

analysis stage, as described above. Table 2 shows the RPN possible values and its classification of

high, medium or low.

Table 2. Risk Priority Number (RPN) calculation. High RPN (red), medium RPN (orange) and low RPN (green). Adapted from [10]

Detectability

Risk

Classification

3 (Low) 2 (Medium) 1 (High)

Level 1 9 27 18 9

6 18 12 6

Level 2 4 12 8 4

3 9 6 3

Level 3 2 6 4 2

1 3 2 1

A high RPN means that the function or component is critical and validation measures must be taken. A

medium RPN indicates that the function or component is potentially critical and that validation measures

must be taken. A low RPN means that the function or component is not critical and there is no need to

take validation measures.

Extending FMEA to incorporate an investigation of the degree of severity of consequences, their

probabilities of occurrence and their detectability results in FMECA tool. This tool can be applied for

failures and risks associated with manufacturing processes. Each failure mode is identified and then

evaluated for criticality. The output of FMECA is a relative risk score for each failure mode, which is

used to rank the modes on a relative risk basis.

Concluding, QRM as part of PV is essential to identify the scope and extent of the validation activities

and to distinguish between critical and non-critical process steps [9, 10].

13

2.1.6 Documentation

Validation and qualification must be performed in accordance with written procedures. Nevertheless,

documenting properly each performed validation and qualification activity is fundamental so that

knowledge gained about a product and process is accessible to others involved in each stage of the

lifecycle.

The type and extent of documentation required by cGMP varies according to the process validation

stage. Documentation requirements are more extensive during process qualification and continued

process verification stages.

When performing a PV program for the full-scale process, flow diagrams of the process describing each

unit operation, its placement in the overall process, monitoring and control points and sampling

requirements should be generated [1].

The documentation required may include the validation protocols and reports, standard operating

procedures (SOPs), specifications, risk assessment outcomes, training records, sampling plans, testing

plans and methods and the plan for ensuring review and maintenance of a validation state [2].

Validation Master Plan (VMP)

The VMP is a succinct and clear document that reflects the key elements of the validation program. It

should refer at least the validation policy and approach, responsibilities, resources, qualification of

utilities and equipment, the plan and schedule for validation activities and documents required.

The VMP should be reviewed periodically and updated according to cGMP [2].

Validation protocol

A document describing the activities to be performed during a validation process, including the

acceptance criteria for the approval of a process or system for intended use.

Validation report

A document in which the records, results and evaluation of validation are assembled and summarized.

It may also contain proposals for the improvement of the process. The validation report should reflect

the protocols and procedures followed and include a conclusion stating the outcome of PV. Any

deviations found during the PV should be managed and documented, as well as corrective actions

should be considered [2, 6].

2.1.7 Technology transfer as part of process validation

Technology transfer is defined as a logical procedure that controls the transfer of any process together

with its documentation and professional expertise between development and manufacture or between

manufacture sites. It occurs at some stage of the product lifecycle, from development, scale-up,

manufacturing and launch to the post-approval phase [11].

14

The aim of technology transfer activities is to transfer not only the product but also the process

knowledge. This knowledge is the basis for the manufacturing process, control strategy, process

validation approach and continual improvement [3].

As part of the product lifecyle, a technology transfer implies a PV approach. Revalidation is conducted

in case of transfer between production lines, scale-up or any other process changes. A technology

transfer process validation can be performed along with the product launch batches or afterwards. It is

common to have a technology transfer process validation associated to a scale-up process validation

[12].

Usually there is a sending unit (SU), a receiving unit (RU) and the unit managing the process, which

may or may not be a separate entity. Technology transfer requires a documented and planned approach

using trained personnel working within a quality system, and the demonstrated capability of the RU to

accomplish the critical elements of the transferred technology and fulfil all regulatory requirements.

To reach a successful technology transfer, the following requirements must be accomplished:

• The project plan should encompass the quality aspects of the project and be based upon the

principles of quality risk management;

• The capabilities of the SU and at the RU should be similar and facilities and equipment should

operate according to similar operating principles;

• A comprehensive technical gap analysis between the SU and RU including technical risk

assessment and potential regulatory gaps, should be performed as needed;

• Adequately trained personnel should be involved;

• Regulatory requirements in the countries of the SU and the RU, and in any countries where the

product is intended to be supplied, should be considered and interpreted consistently.

Technology transfer can be considered successful if there is documented evidence that the RU can

routinely reproduce the transferred product, process or method against a predefined set of specifications

as agreed with the SU.

Along with a technology transfer validation, a cleaning validation is also required in order to minimize

the risk of contamination of the product. An adequate cleaning strategy implies information on solubility

of Active Pharmaceutical Ingredient (API), excipients and vehicles; minimum therapeutic doses of API;

therapeutic category and toxicological assessment and already existing cleaning procedures.

A technology transfer summary report should be issued stating the documented evidence that the

process has been considered successful. The report should include the extent of the transfer, the critical

parameters and the conclusions [11].

15

2.2 STERILE DRUG PRODUCTS MANUFACTURING

2.2.1 Aseptic processing

The sterility of a drug product is defined as the complete absence of viable microorganisms. A sterile

drug product can be achieved using aseptic processing or terminal sterilization [13].

Aseptic processing implies the sterilization of components, containers and closures prior to filling of the

drug product. The glass containers are usually subjected to wash and rinse cycles, prior to dry heat

sterilization. The rubber closures can be sterilized by steam or irradiation methods and the bulk solution

is subjected to a sterilizing filtration. The product is filled and sealed in an extremely controlled high-

quality environment, since there is no sterilization of the product in its final container.

Terminal sterilization manufacture takes place under controlled environmental conditions to minimize

the microbial and particulate content of the in-process product and to guarantee success of the

subsequent sterilization. The product in its final container is subjected to a sterilization method as steam

or radiation [13,14].

Aseptic processing is more challenging than terminal sterilization. Therefore, the European

Pharmacopeia defines terminal sterilization as the method to be used whenever possible and the aseptic

processing as the last resort. However, certain critical processes such as lyophilization demand the use

of aseptic processing due to degradation of the product or loss of performance when exposed to heat

or radiation. All activities that may compromise the sterility of the product or material need to be

considered as extensions of aseptic processing. The lyophilization equipment and its processes should

be designed in a manner that ensures the maintenance of the product and materials sterility during filling

and until end of the lyophilization process. Loading and unloading of the lyophilizer should take place

under grade A environment and the lyophilizer should be sterilized before each load [15].

Sterility assurance in aseptic processing depends on the rooms, personnel, sterilization methods,

microbiological monitoring and validation of the aseptic filling process.

2.2.2 Room classification

Separated or defined areas of operation in an aseptic manufacturing facility should be appropriately

monitored to assure the appropriate airflow and pressure according to the operation conducted. Clean

areas should be supplied with filtered air and achieve a proper airflow from higher cleanliness areas to

adjacent less clean areas [14].

Each manufacturing operation requires an appropriate environmental cleanliness level in order to

minimize the risk of particulate or microbial contamination of the product. Four grades of clean room can

be distinguished. Grade A corresponds to high risk operations such as filling and aseptic connections.

Grade B is the background environment for the grade A zone. Grades C and D are clean areas where

less critical operations take place, such as materials preparation and capping [13, 15]. Table 3 presents

the clean room classifications for non-viable particles monitoring.

16

Table 3. Clean area air classifications by limits of airborne particle concentration for non-viable particles monitoring

Grade

classification

Maximum permitted number of particles/m3

At rest In operation

≥ 0.5 m ≥ 5.0 m ≥ 0.5 m ≥ 5.0 m

A 3 520 20 3 520 20

B 3 520 29 352 000 2 900

C 352 000 2 900 3 520 000 29 000

D 3 520 000 29 000 Not defined Not defined

2.2.3 Personnel qualification

A well designed and performed aseptic process minimizes personnel intervention. The risk of

contamination of the finished product increases with human interventions.

Operators participating in aseptic activities should be appropriately trained and qualified in aseptic

technique. Aseptic technique comprises all approaches adopted during aseptic manufacture to assure

that sterility of the product, components and surfaces is maintained, such as move slowly and

deliberately, contact sterile materials only with sterile instruments, keep the entire body out of the

unidirectional airflow path and maintain proper gown control [13, 14].

2.2.4 Containers and closures sterilization

Contamination of an aseptically manufactured product can occur through use of contaminated

containers and closures. Validation studies should be conducted to demonstrate the sterilization cycle

efficacy. On a periodic basis, requalification is also required.

The sterilization process used depends on the nature of the materials. The sterilization used is

previously validated to prove its ability to provide materials sterile and non-pyrogenic. Glass containers

are usually washed and rinsed prior to depyrogenation. Rubber closures are sterilized through steam in

autoclave. Final inspection of the container closure system is crucial to detect damaged or defective

units, which should be rejected [14].

2.2.5 Filtration validation

Sterile filtration is a crucial step in aseptic manufacturing of sterile pharmaceutical products. A sterilizing

grade filter should be validated to ensure the removal of microorganisms, producing a sterile effluent.

Sterile filters generally have a nominal pore size of 0.2 m or smaller.

The filtration efficacy is ensured through validation including microbial challenge to simulate worst-case

production conditions and integrity test results of the filters. The challenge microorganism must be

selected based on the product bioburden, to evaluate which microorganism represents the worst

challenge to the filter.

17

The bioburden of the unsterilized bulk solution must be determined to trend the characteristics of

potentially contaminating organisms. The process controls should minimize the bioburden of the

unfiltered product.

During routine production, identical filters to the ones validated must be used. The filters must be

discarded after manufacturing of a single batch and integrity test, such as bubble point, must be

performed before and after use. In the bubble point test, it is determined the pressure at which the

diffusive gas flows to the free flow via pores that are no longer wetted. The gas pressure is increased

continuously or in stages on the unsterile side and a check is made to see when the pressure decreases

disproportionately [13, 14].

2.2.6 Microbiological testing

Bioburden and endotoxins testing are crucial elements of an aseptic process. Endotoxin contamination

of an injectable product can be caused by poor cGMP controls and lead to pyrogenic reactions in certain

patient populations. These clinical concerns reinforce the need to implement adequate cGMP controls

during aseptic processing to prevent bioburden and generation of endotoxins.

Adequate cleaning, drying and storage of equipment controls bioburden and prevents contribution of

endotoxin load. The bioburden assay should be performed for each batch, considering working limits on

contamination immediately before sterilization, related to the efficiency of the method to be used [14].

The type of contamination allows to conclude about the contamination source. Therefore, an efficient

microbiological monitoring program must be established. The monitoring program should be defined as

a SOP and include description of sampling locations, sampling frequency and duration, alert and action

limits and measures to be taken when these limits are exceeded [13].

2.2.7 Validation of aseptic processing

To ensure the sterility of products, aseptic processing must be adequately validated. An aseptic process

should be validated through process simulations, also known as media fill, where a microbiological

growth medium is filled instead of the product.

A media fill program should include the contamination risk factors of routine production and evaluation

of the state of control. This is achieved by simulating the aseptic process that the product itself

undergoes. The containers filled with the microbiological medium are then incubated to detect microbial

contamination and assess the potential risk of a unit to become contaminated during routine operations.

It is recommended by FDA that the media fill program incorporates issues such as operators’ fatigue,

representative number and type of routine and non-routine interventions, lyophilization, aseptic

assembling, shift changes, line speed and container closure systems.

For initial qualification of a production line, individual media fills runs should be repeated to ensure

consistency of the results, since a single run is inconclusive of the state of process control. Routine

semi-annual qualification is required for each production line.

18

The duration of aseptic process is also qualified through media fill programs. It should be determined

based on the time required to incorporate manipulations and interventions. The simulation run size

should be representative of the commercial manufacturing process. An acceptable run size is in the

range between 5 000 and 10 000 units.

For lyophilization process, unsealed containers should be exposed to partial evacuation of the chamber,

simulating the process. However, the vials should not be frozen and the aerobic state of the medium

must be ensured.

A batch record should be prepared for each media fill run, documenting the production conditions and

simulations performed. The rationale behind the simulated activities during media fill should be clearly

defined [13, 14].

19

3 HIKMA LYOPHILIZATION PRODUCTION DEPARTMENT

OVERVIEW

Hikma Pharmaceuticals is a multinational pharmaceutical company founded in Jordan which develops,

manufactures and markets a broad range of medicines in Europe, Middle East and North Africa (MENA)

region and US, comprising twenty-nine manufacturing plants and seven Research and Development

(R&D) centers. Hikma operations are conducted through three distinct business segments: injectables,

branded and non-branded generics [16].

3.1 PRODUCTION LINES

Within the injectables segment, the manufacturing plant in Portugal is the center of production and

commercialization and comprises three different facilities: Hikma 1 with seven production lines for liquid

and lyophilized medicines; Hikma 2 with three lines specific for powders manufacturing and, the most

recent one, Hikma 4 with two lines specialized in liquid and lyophilized oncologic drugs manufacturing.

Table 4 summarizes the production lines at Hikma Portugal.

Table 4. Hikma's type of products manufactured at each production line and facility

Facility Production line Product presentation Therapeutic Category

Hikma 1

1 Liquid and lyophilized vials

Antibiotics

Cardiovascular and

diabetes

Central nervous system

Controlled substances

Gastro-intestinal system

Respiratory system

2 Liquid vials

3 Ampoules

5 Liquid vials

7 Bags

9 Liquid and lyophilized vials

10 Lyophilized vials

Hikma 2

1

Powder vials Antibiotics 2

3

Hikma 4

1

Liquid and lyophilized vials Oncological

2

20

3.2 LYOPHILIZATION PROCESS

Lyophilization or freeze-drying is a process that can be used to obtain dry products from aqueous

solutions or dispersions. It is mainly used to preserve the stability and increase shelf life of sensitive

substances such as hormones, antibiotics, enzymes, proteins and others.

The desired final product of a lyophilization process is a highly porous cake that promotes rapid and

complete reconstitution after addition of the solvent [13].

For a successful lyophilization process, the following requisites must be accomplished:

• Complete solidification of the solution/dispersion by freezing;

• Application of vacuum;

• Controlled supply of heat;

• Removal of water vapour.

The lyophilization process consists of three phases: freezing, primary drying and secondary drying. The

freezing phase is used to solidify the product before sublimation can occur. The product is frozen by the

controllable process of cooling the shelves of the lyophilizer and holding the product at that temperature.

The freezing conditions determine the size and shape of ice crystals formed, that affects drying

performance.

After completion of the freezing phase, vacuum is applied to start sublimation. The primary drying

(sublimation) phase is the longest one. The driving force for sublimation occurs is that the pressure

developed by the water molecules in the surrounding atmosphere (freeze-dryer chamber) is lower than

the vapour pressure at the sublimation interface in the product. The ice vapour pressure is directly

proportional to the temperature of the product. During this phase the temperature is increased, and the

pressure adjusted to the respective setpoints and then hold for the required duration.

At the end of primary drying, the temperature of the shelves is increased and, consequently, the

temperature of the product progressively rises. Along with a low enough pressure, the desorption

(secondary drying) phase begins. The goal of this phase is to remove remaining traces of moisture in

the product due to adsorption of water molecules in the dry structure. The shelves temperature is

increased to the highest possible temperature compatible with the product stability and the chamber

pressure is low [13, 17].

The complex technology that is inherent to the manufacturing of sterile lyophilized pharmaceutical

products implies rigorous regulatory requirements, since the absence of microbiological contamination

must be ensured. Activities such as an improvement of the lyophilization cycle and transfer and scale-

up of the process to a larger scale lyophilizer must be conducted under the scope of a well-established

process validation protocol.

21

3.3 TECHNOLOGY TRANSFER FROM LINE 1 TO LINE 9

At Hikma, the technology transfer PV is carried out by a wide team of key people from different

departments. Firstly, the Technical Services department receives all information related to the product

to be manufactured from the Jordan or US R&D centers. It is their responsibility to elaborate the Master

Batch Record (MBR) for submission purposes. After manufacture of the exhibit batch, the Regulatory

Affairs execute all documentation required to submit the product to the regulatory authorities. Once the

product is approved for commercialization, a scale-up and/or technology transfer is likely to be planned.

The PV team from the Quality Development department initiates the PV protocol regarding all validation

activities to be performed. Production department has the responsibility to carry out all activities and

sampling plan according to the protocol during the manufacturing of validation batches, as well as to

report any deviations during manufacturing. The samples are analytical tested as defined in the protocol

by the Microbiology, HPLC and Wet Chemistry laboratories, part of the Quality Control (QC) department.

The PV team elaborates the validation report with all results obtained and conclusions. Following the

conclusion of the PV, the validated parameters are added to the MBR by the Production team, in order

to implement the changes to the manufacturing process. The Quality Assurance (QA) department has

the responsibility to coordinate the validation process by scheduling discussions with all parts involved

and monitoring the execution of the protocol, as well as review and approve all documentation [18].

Figure 3. Product lifecycle at Hikma Pharmaceuticals.

Product Development

Technology Transfer

Commercial Manufacturing

Process Optimization

New Projects Validation

Production / QC

Regulatory Affairs

Approval for

commercialization

1

2

Validation and approval of

changes

5

6 Submission of new product to

authorities

7

Quality Assurance

R&D Centers

Technical Services

Regulatory Affairs

Exhibit batch manufacturing

3

4

Production

Product launch

Commercial batch manufacturing

22

Initially, Hikma plant had only line 1 for the manufacturing of lyophilized products. Based on the market

demand, lyophilization capacity had to be increased and therefore two new lines (lines 9 and 10) were

installed in 2017 for this purpose.

Each production line has two freeze-dryers and the two new lines have an improvement in the loading

and unloading system of freeze dryers, which is fully automatic, contrary to line 1 where this system is

semi-automatic. In order to optimize and rationalize production, some of the lyophilized products

manufactured in line 1 were transferred to lines 9 and 10.

Technology transfer of lyophilized products from line 1 to line 9 was evaluated by comparing the

equipment, the manufacturing areas grade classification, the manufacturing process and the quality

systems of the two lines. A risk assessment was performed to evaluate the impact on the validated

status of the products and the actions required for the transfer of products from line 1 to the new

production line [19, 20].

All products manufactured in line 1 have undergone a process validation where testing of the drug

components, container/closure preparation, compounding, filtration, filling, lyophilization, stoppering,

inspection, labeling, packaging and all critical process parameters (CPPs) have been evaluated and

validated. For each product to be transferred from line 1 to line 9, a new process validation protocol is

issued with the sampling requirements needed to complete the validation. The parameters considered

equivalent for the manufacturing are not required to be re-validated on a product basis. A process

validation report is issued for each product to summarize the results obtained in the new line and

confirming a robust and controlled process for manufacturing high quality lyophilized products.

Freeze-dryers comparison

The two freeze-dryers installed in the new line 9 were considered equivalent to the ones of line 1, except

for the automatic loading and unloading system. The condenser capacity of the freeze-dryers in line 9

is 800 kg, while in line 1 is 342 kg.

For the initial qualification of freeze-dryers a shelf mapping study is performed to verify the temperature

uniformity and distribution profile on the shelves of the freeze-dryer chamber. Five thermocouples are

assembled in each shelf to monitor the temperature inside the freeze-dryer chamber and evaluate the

disparity of each location in the same shelf and between shelves. The acceptance criteria of the study

is that the deviation between the maximum average temperature and the minimum average temperature

is less or equal to 2 ºC on each shelf and between all the shelves. For both freeze-dryers from line 1

and from line 9, the shelves temperature distribution inside the chamber was shown to be homogeneous

by shelf mapping studies. Therefore, the freeze-dryers were considered qualified.

Rooms classification

Classification within each line assures that the level of air quality and pressurization increases from the

least to the most critical area, in order to prevent an inward flow of potential contaminants. Components

preparation and air locks separate the sterile core from all other areas where non-sterile activities are

performed. All critical operations are conducted in classified areas. A risk assessment for the

23

comparison of rooms classification between line 1 and line 9 was performed, as summarized in Table

5.

Table 5. Comparison of room classification between line 1 and line 9

Process Stage

Production lines Risk Assessment

Line 1 Line 9

(New line) Potential Product Risk Risk Conclusion

Materials and

filling machine

parts preparation

Grade C Grade C The classification of the

areas in which all critical

operations are conducted

are the same grade or

even higher grade for the

new line 9.

All areas have been

initially qualified and are

re-qualified using the same

criteria.

Environmental Monitoring

and Continuous Particle

Monitoring assessments

have been conducted for

both lines and sampling

locations and frequency

have been defined. Action

levels are harmonized for

all lines and meet FDA and

EU guidelines.

LOW

No impact

Vials washing

machine and

depyrogenation

tunnel

Grade D Grade C

Compounding

room Grade C Grade C

Pre-filtration Grade C Grade C

Sterile Filtration

Grade A

(inside sterile

room)

Grade A

(inside sterile

room)

Aseptic Filling Grade A Grade A

Lyophilizers

loading /

unloading

Grade A Grade A

Capping Under Grade

A air supply

Under Grade

A air supply

It was concluded that room classification where the same operations will be conducted between the two

lines are the same or higher for new production line 9. The same approach for areas qualification and

re-qualification is followed, as well as for environmental monitoring and particle monitoring.

Manufacturing process

The manufacturing process was compared between line 1 and line 9 for each operation.

I. Components cleaning, preparation and sterilization/decontamination

In line 1, the filling machine parts are cleaned while assembled on the filling machine through a Cleaning

in Place (CIP) cycle, followed by a Sterilization in Place (SIP) cycle. In line 9, the filling machine parts

are washed in a machine and then sterilized in autoclave. For both lines, all the support material as well

as ready-to-sterilize stoppers are sterilized through steam sterilization in autoclaves with the same

controls: pressure, time and temperature. The ready-to-use stoppers are submitted to a

decontamination program in a Vaporized Hydrogen Peroxide (VHP) chamber. VHP chambers are also

qualified using the same procedures for the two lines.

24

The vials washing is done in line with depyrogenation. The washing machines use Water for Injection

(WFI) and then the vials are dried with compressed air and automatically transported to the

depyrogenation tunnel. The depyrogenation is conducted in laminar flow hot air tunnels according to

validated parameters for each vial size and consists on a pre-heating zone, a sterilizing zone and a

cooling zone. Both machines are equivalent in line 1 and line 9.

II. Compounding

The utilities available in compounding rooms are the same for each line: WFI, compressed air, nitrogen,

cold and hot water and pure steam. All utilities are qualified following the same procedures as per

guidelines. The portable tanks are shared between the two lines and each line has a 2000 L fixed tank.

All tanks follow the same cleaning procedures.

If the tanks already used in line 1 are to be used in line 9, it is ensured that all processes are equivalent

from compounding, holding times and sampling perspective. A technology transfer that implies changing

the compounding tank or a change on the compounding batch size requires specific validation activities,

which are further described in the respective validation protocol.

III. Filtration

For both lines, the filtration occurs from grade C to grade A through a filter using nitrogen pressure.

When applicable, pre-filtration may occur having a pre-filter installed in grade C. Filter validation studies

are performed for each product covering the filtration train to be used. Filtration is performed in line with

filling.

IV. Aseptic Filling

The filling machine in line 9 is from the same manufacturer and equivalent to the one in line 1. The surge

tank, the stoppers loading system, the filling needles and the type of tubes are the same for the two

lines. Both filling lines include Restricted Access Barrier Systems (RABS) to provide a controlled

environment with high level of protection to process materials through small openings and reduce

interventions into the critical filling zone.

In process control (IPC) is performed during filling for volume and particles. The rubber stoppers are

transported through a sterilized track and placed in the vials with a mechanical piston in the two lines.

The filling machine speed needs to be evaluated individually for each product to be transferred from line

1 to line 9.

V. Lyophilization

Automatic loading and unloading of the freeze-dryers for line 9 represents an improvement compared

to line 1 by reducing aseptic handling. The vials are stoppered inside the freeze-dryer by shelves

lowering. Although the freeze-dryers are equivalent, further activities for each product are required to

ensure that the transference of the lyophilization cycle does not have impact on the quality of the finished

product and to validate the process.

25

VI. Capping

The capping machines are different in each line, so validation activities are required.

VII. Inspection

Visual inspection is performed offline, in the same inspection areas and using the same equipment for

both lines.

Aseptic process simulation (Media Fill)

The two lines share the same media fill rational to validate the aseptic filling process. For initial

qualification a minimum of three media fill runs is performed per line. Routine media fill runs are

conducted every six months for each line.

All interventions, holding times and filling duration are qualified for each line. Routine and non-routine

interventions are included and need to be qualified per line. Some interventions are considered

equivalent from aseptic handling and therefore, operators qualified on these specific interventions in one

of the two lines are considered qualified to perform the same interventions in the other line.

As an overall conclusion, manufacturing lines 1 and 9 were considered to have a similar design and

process. The differences found along the evaluation show an improvement to the current design of line

1. The risk was found as low, thus no risk analysis through FMEA is required.

The activities identified as medium and high risk are performed for each product transfer from line 1 to

line 9, as follows:

- Transfer of the lyophilization cycle from the current freeze-dryers in line 1 to the new ones in line 9,

determining the cycle parameters per product. Process validation including results of inspection and

side-by-side comparison with batches manufactured in line 1 is required, as well as stability studies;

- Assessment of the current process parameters validated for the filtration train in line 1 in order to

determine if filter validation studies are required to enable the filling of the same products in line 9;

- Evaluation of the filling process covering the filling variables per line and product (dead volume,

filling machine speed, volume variation and line stoppages);

- Evaluation of the capping process;

- Evaluation of the compounding process in case of change of batch size or compounding tank.

26

27

4 SUBMISSION OF NEW LYOPHILIZED PRODUCT A

4.1 PRODUCT SUBMISSION AND APPROVAL FOR US MARKET

Product A was developed by Jordan R&D center and transferred to Lyophilized Injectable Production

Department at Hikma Portugal. In 2012 one exhibit batch to the US market was manufactured in

production line 1 and submitted as an Abbreviated New Drug Application (ANDA). Since the product is

an ANDA, for submission purposes, the analytical results were compared to the ones of the Referenced

Listed Drug (RLD). During the pre-acceptance stage, several requests were received from FDA,

regarding labeling and chemistry questions. After the requests have been responded to and accepted,

the product A was approved for commercialization as an ANDA by 19th June 2017.

Figure 4. Product A lifecycle at Hikma Pharmaceuticals.

Process

Optimization

Commercial

Manufacturing

Technology Transfer

to Hikma PT

Product A

DevelopmentR&D

Manufacture of 100 L exhibit batch

PRODUCT SUBMISSION Line 1

Approval for commercialization to US

market

PRODUCT LAUNCH

Scale-up from 100 L to 200 L in line 1

(3 validation batches)

PROCESS IMPROVEMENT

Transfer from line 1 to line 9 and scale-up from 100 L to

400 L

(1 validation batch)

Pending:

Scale-up from 100 L to 400 L

(2 validation batches)

Technical Services

Regulatory Affairs

Jordan R&D Center

New Projects Validation

Production / QC

Regulatory Affairs

Production / QC

Regulatory Affairs

New Projects Validation

Production / QC

28

Figure 4 schematizes the lifecycle of product A at Hikma, since development to the commercial

manufacturing and process optimization. These two stages that were planned after approval for

commercialization will be presented in the next chapters of the present work, regarding product launch

and scale-up and technology transfer as part of manufacturing process optimization for product A.

4.2 PRODUCT AND PROCESS SPECIFICATIONS

This product is highly sensitive to light, exposure to oxygen and heat. Thus, there are some precautions

to consider during the manufacturing process.

During compounding, the temperature must be maintained between 40 ºC and 45 ºC from the beginning

until dissolution of the API. After that, the temperature range is from 20 ºC to 25 ºC until end of

compounding. The preparation tank must be maintained closed as long as possible.

Nitrogen sparging is required during compounding and filling. The temperature in the tank must be kept

between 5 ºC and 10 ºC from the end of compounding until the end of filling. Table 6 summarizes the

product and process requirements for product A.

Table 6. Process requirements for product A

Under nitrogen overlaying Yes (during compounding and filling)

Tubing to be used Teflon

Use pre-filter Yes

Maximum contact time of the filter with

product 24 h

Product sensitive to light Yes

Product sensitive to oxygen Yes

Product sensitive to heat sources Yes

Temperature of the bulk solution in the

preparation tank during filtration/filling 5 ºC – 10 ºC

Finished product storage conditions 20 ºC – 25 ºC (Protected from light)

29

4.3 MANUFACTURING PROCESS

The manufacturing process for product A includes preparation and sterilization of the filling machine

parts and components, compounding of the bulk solution, pre-filtration through a 0.45 m filter, filtration

through a sterilizing 0.22 m filter, filling into a 100 mL vial presentation, stoppering, lyophilization,

capping and visual inspection.

The container closure components for the manufacturing process of this product include 20 mm rubber

stoppers, 20 mm red aluminium flip-off and 100 mL vials.

In production line 1, the filling machine parts are transferred to the sterile area through a VHP chamber

and submitted to a bio decontamination program. In the sterile area, the machine parts are assembled

and then a CIP and SIP cycle is performed to the filling machine.

The stoppering system, the rubber stoppers and all the support material are sterilized through autoclave

from the grade C area to the grade A room. The vials are washed and sterilized through a washing

machine and a depyrogenation tunnel. The bulk solution is compounded in grade C and then filtered

directly from the compounding tank to the filling line. The pre-filter is assembled on the CIP/SIP panel

in grade C, while the filter is assembled in the grade A filling room.

Along with filling, the vials are partially stoppered and then nested in trays, which are previous sterilized

in the autoclave. The trays are manually loaded in the freeze-dryer. When the lyophilization cycle ends,

the vials are fully stoppered by the shelves lowering.

The trays are manually unloaded from the freeze-dryer and transported in stainless steel trolleys to the

capping room. After capping process is completed, the vials proceed to inspection and secondary

packaging.

The difference for the manufacturing process in line 9 is that the filling machine parts are sterilized in

autoclave prior to assembling and the pre-filter is assembled directly to the compounding tank in the

grade C area. The loading and unloading of the freeze-dryers is automatic [21, 22].

The manufacturing process is presented in Figure 5.

30

Figure 5. Manufacturing process of product A.

Compounding

Pre-filtration by 0.45

µm filter

Filtration by 0.22 µm

filter

Washing Machine

Sterilizing

tunnel

Filling / Stoppering

Autoclave

Sterilization

Trays loader

Lyophilization cycle

Capping (under UFH)

Visual Inspection

Secondary packaging

Pallets preparation

Shipping

Filling line SIP

VHP Chamber

Grade C Area

Grade A Room

Unclassified - Warehouse

Unclassified - Packaging Area

Unclassified -

Warehouse

Unclassified or Grade D

Rubber Stoppers 100 mL vials Active and inactive

raw materials

Stoppering

equipment Filling

machine parts

Bulk solution

Aluminium flip-off

(from warehouse)

Line 9

Line 1

31

5 PRODUCT A LAUNCH AND SCALE-UP IN LINE 1

The 100 L exhibit batch was manufactured in production line 1 in 2012. By the time of the product

approval for commercialization, it was planned a scale-up from 100 L to 200 L in line 1 along with the

product launch.

In order to validate the scale-up process, a PV protocol was elaborated. The protocol had the purpose

to define the activities and sampling requirements intended to validate the proposed change.

Three validation batches were manufactured. The number of proposed validation batches for a scale up

and line transfer can be defined based on the following rationale: product characteristics which might

impact the filtration and filling process; light, heat and oxygen sensitive; bulk strength/number of

presentations.

All activities required for qualification of facilities, equipment and systems to be used in the process were

previously made and approved. All validation batches were manufactured using qualified facilities,

equipment and systems and tested using validated analytical methods. The aseptic filling was validated

through media fill.

5.1 RISK ASSESSMENT AND CONTROL

The CPPs in the manufacture of the product A were evaluated and the parameters identified as potential

process failures were further evaluated based on risk management tool FMECA.

The parameters identified as potential process failures were dissolved oxygen, temperature along

compounding, API dissolution, pH measurement, final weight, bulk exposure to air, filtration, filling,

lyophilization and inspection. These parameters were evaluated based on the risk management tool

FMECA.

Severity and probability are classified in level 1 (low), level 2 (medium) and level 3 (high). Detection is

categorized in level 1 (high), level 2 (medium) and level 3 (low). A multiplication of these three

parameters results in RPN, which can be critical – 12, 18 and 27; medium – 6, 8 and 9 or low – 1, 2, 3

and 4.

For each CPP, a level of severity, probability and detection was established. After that, the RPN for each

CPP was calculated. Annex A – Risk Management Tool FMECA for process validation of product A –

summarizes the results.

It was concluded that the temperature along compounding, the API dissolution, the final weight, the bulk

exposure to air, the filtration, improper nitrogen headspace purging and an inadequate lyophilization

cycle design represent a medium risk for the process. Only a power failure during lyophilization was

determined to represent a high risk for the process.

32

Temperature along compounding:

Product A is highly sensitive to heat sources. If the temperature is not maintained in the defined ranges,

which can be caused by a chiller malfunction, the potential effect may be API degradation. To control

this risk, the compounding tanks have calibrated temperature sensors so an increase of temperature

will be immediately noticed.

API dissolution:

A human error on the calculations for API and excipients can lead to a not complete dissolution of the

API and, consequently, a non-homogeneous product. This will result in OOS results for the product

assay and batch failure. As a control, this step of the process is double verified.

Final weight:

Both human error or a balance mal function may cause low or high assay and result in batch failure. As

a risk control, the balance is calibrated and verified prior to each compounding and this step is also

double verified.

Bulk exposure to air:

Since the product is sensitive to oxygen, an uncontrolled exposure of the bulk solution to air can cause

API degradation. Therefore, nitrogen overlaying of the bulk solution during compounding and filling is

applied.

Filtration:

Incompatibility of the filter membrane with the product can cause low assay, high related substances or

particle formation. As control, filter validation studies were performed on the previously exhibit batch

manufactured in line 1 to assure compatibility with the filter and sterility of the bulk solution.

Improper nitrogen headspace purging:

An improper nitrogen purging during filling may cause product degradation. This risk is controlled by

nitrogen flushing during filling.

Inadequate lyophilization cycle:

If the lyophilization cycle design is not adequate, it may not dry the product properly, which compromises

the quality of the batch. Lyophilization cycle optimization is tested prior to production and samples are

collected for lyophilization uniformity and finished product testing.

Power failure during lyophilization:

A power failure during the lyophilization cycle may cause the cycle to go back to freezing phase, which

can affect the product characteristics and result in batch failure. To overcome this risk, the freeze-dryers

have alarms, specifically one alarm indicating that the cycle went back to freezing phase. In this case,

the lyophilization cycle can be forced to step forward after proper evaluation by the relevant key persons.

33

The freeze-dryers are connected to an electrical generator system, so any power failure or electrical

shutdown will not have impact on the lyophilization cycle progress.

At the end of the cycle, the report is printed and evaluated by production and QA departments.

5.2 PROCESS VALIDATION ACTIVITIES AND RESULTS IN LINE 1

For scale up and line transfer PV, it is necessary to evaluate the process steps that are scale and/or line

dependent in order to determine the parameters that need to be monitored during PV and ensure that

scale up and line transfer do not translate into a loss in quality, meaning that do not adversely affect the

CQAs of the finished product [12].

The variables considered to determine the scale dependent PV activities are the batch size, the tank

size, the equivalence between tanks and product properties. To define the line dependent activities, the

variables considered are the line length, the filtration tubes length, pumps sizes, container sizes, product

strength and fill volume. Based on the identified CPPs that need evaluation, the following are considered

size and/or line dependent and require evaluation for this PV:

• Quality of the compounded bulk solution;

• Mixing time and agitation speed;

• Bulk holding time in the preparation/transference tank;

• Effects of initial set up (dead volume);

• Effects of line stoppages;

• Filling machine speed;

• Effects of filtration and filling on the quality of the compounded solution;

• Holding time between the beginning of API addition and beginning of lyophilization cycle;

• Effects of lyophilization on the quality of the finished product and lyophilization uniformity;

• Quality of the finished product;

• Inspection.

The PV activities were defined based on the risk assessment of the CPPs, evaluation of the CQAs, level

of monitoring, frequency of testing and the number of batches to be manufactured. The samples to be

taken at each stage are described in Annex B – Sampling plan for process validation of scale-up of

product A in line 1. [23, 24].

The bulk holding time and effects of line stoppages in line 1 were evaluated and considered validated

in the exhibit batch for submission. It was approved 12 hours for the holding time between the end of

cooling of the bulk solution and the beginning of the lyophilization cycle. Line stoppages up to 2 hours

were considered to have no impact on the quality of the product and no validation activity is required for

the present PV.

Based on the risk assessment and rationale presented for the present PV, the number of batches to be

evaluated during PV activities were set as presented on Table 7.

34

Table 7. Number of validation batches required for each validation activity comprised in the scale up for product A in line 1

Process Validation Activities Number of validation batches

Evaluation of quality of the compounded bulk

solution 3

Evaluation of the effects of initial set up (dead

volume) 1

Evaluation of filling machine speed 1

Evaluation of the effects of filtration and filling

on the quality of the compounded solution 3

Evaluation of the effects of lyophilization on the

quality of the filled vials and lyophilization

uniformity

3

Evaluation of the quality of the finished product 3

Evaluation of the results of visual inspection of

the vials 3

Evaluation of quality of the compounded bulk solution

The dissolution should be complete, and the solution should be uniform. The compounding parameters

mixing and dissolution times were previously evaluated for the 100 L batch. However, the new batch

size to be validated is 200 L and the batch is compounded in a different tank, therefore a range of mixing

speeds and times were validated to assure that complete dissolution occurs under all approved

compounding conditions. The CPPs mixing time and speed were manipulated to their defined limits,

ensuring the physicochemical integrity of the product is challenged throughout the range of previously

established manufacturing conditions.

For the first validation batch, the defined mixing speed was 296 rpm during all compounding steps. For

second and third validation batches, the mixing speed was 317 rpm and 447 rpm, respectively. The

mixing time varied depending on the compounding step.

Samples from the top and bottom of the preparation tank were collected after the final mixing step has

been completed. The samples were analysed for description, pH, identification by HPLC, assay, related

substances, bacterial endotoxins and bioburden. All results were within the established specifications.

It was concluded that the compounding process parameters used for the manufacturing of product A

allow obtaining a product meeting the established specifications. The validated mixing speed was

between 296 rpm and 447 rpm.

Evaluation of the effects of initial set up (dead volume)

An initial quantity of filled units is typically discarded as part of initial line set up. The goal of this PV

activity is to establish the minimum amount of product that needs to be discarded prior to start filling.

The process validation batches have the same bulk formulation and strength and will be filled in the

same filling line as the exhibit batch. The dead volume was already evaluated and considered validated

in line 1 and it was concluded that a discarded volume of 1189 mL (29 vials) as part of initial line set up

35

is enough to have a product within specifications. This parameter was re-evaluated to verify if the

quantity of 300 mL discarded in line 1 according to the current SOP is enough to have a product within

specifications.

The first and last vials from three stages were tested and results for pH, identification by HPLC, assay

and related substances were conforming, as shown in Table 8.

Table 8. Results obtained for dead volume samples for PV of scale-up in line 1

Stage 1 Stage 2 Stage 3

Test Specification First

units

Last

units

First

units

Last

units

First

units

Last

units

pH 8.8 – 11.0 10.1 10.1 10.1 10.1 10.1 10.1

Identification

by HPLC

Retention time

of major peak

in the

chromatogram

corresponds to

that in the

standard

preparation

Conform Conform Conform Conform Conform Conform

Assay 95.0 - 110.0 % 101.5 101.6 101.7 101.6 100.4 100.9

Related

Substances:

Related

compound B NMT 3.0 % 1.80 1.78 1.90 1.87 1.84 1.85

Any

unspecified

impurity

NMT 0.2 % < 0.04 < 0.04 < 0.04 < 0.04 < 0.04 < 0.04

Total NMT 3 % 2 2 2 2 2 2

Evaluation of filling machine speed

The process validation batches were filled in the same filling line, with the same filling volume and the

same filling process already evaluated and validated for submission of product A. However, no range of

filling machine speed was challenged at that time. Therefore, this evaluation was performed during the

present PV. The maximum and minimum filling machine speed was challenged in order to set a filling

speed range for routine production. The control analysis to evaluate this parameter was volume variation

of the vials.

The target average volume was 41.0 mL, with specifications of a minimum individual volume of 40.5 mL

and a maximum individual volume of 41.5 mL. All the results obtained for the PV batch were conforming

with the pre-established specifications and are summarized in Table 9. The filling speed range of 24

vials/min – 60 vials/min was validated.

36

Table 9. Filling machine speed results obtained for the PV scale-up in line 1 based on volume specifications

Parameter Maximum Filling Speed

60 vials/min

Minimum Filling Speed

24 vials/min

Average Volume (mL) 41.0 41.0

Minimum Individual Volume (mL) 40.8 40.8

Maximum Individual Volume (mL) 41.4 41.1

Evaluation of filtration and filling on the quality of the compounded solution

The line behaviour during filtration and filling of the new batch size of 200 L was evaluated. The process

was monitored through routine sample testing every hour during filling, which includes analysis for visible

particles, pH and volume variation.

Samples were collected also from the beginning, middle and end of the filling process for testing. The

beginning of the process is defined as a point in which the filtered bulk solution is no longer discarded

and line flush and set up activities have been completed. These samples were analysed for visible and

sub-visible particles, pH and, only for the end of filling samples, for bioburden. All samples from the

three PV batches met the pre-established specifications, as presented in Table 10.

Table 10. Results obtained from the beginning, middle and end of filling samples for the three validation batches

1st Process Validation Batch

Test Specification Beginning of

filling

Middle of

filling

End of

filling

Visible particles Essentially free from visible

particle matter Conform Conform Conform

Sub-visible particles:

Ø ≥ 10 m NMT 6000 particles/vial 37 189 197

Ø ≥ 25 m NMT 600 particles/vial 5 0 80

pH 10.0 – 11.0 10.6 10.7 10.7

Bioburden ≤ 10 CFU/mL - - 0

2nd Process Validation Batch

Visible particles Essentially free from visible

particle matter Conform Conform Conform

Sub-visible particles:

Ø ≥ 10 m NMT 6000 particles/vial

Ø ≥ 25 m NMT 600 particles/vial

pH 10.0 – 11.0 10.7 10.7 10.6

Bioburden ≤ 10 CFU/mL - - < 1

3rd Process Validation Batch

Visible particles Essentially free from visible

particle matter Conform Conform Conform

Sub-visible particles:

Ø ≥ 10 m NMT 6000 particles/vial 325 632

Ø ≥ 25 m NMT 600 particles/vial 13 24

pH 10.0 – 11.0 10.8 10.8 10.8

Bioburden ≤ 10 CFU/mL - - 0

37

Evaluation of lyophilization on the quality of the filled vials and lyophilization uniformity

The selected lyophilizer can be considered a worst case from a loading distance perspective.

Lyophilized vials were collected and analysed to evaluate the effects of the lyophilization cycle on the

quality of the filled vials as well as lyophilization uniformity.

Eight samples of lyophilized vials were collected from five different positions for the first, middle and last

shelf of the lyophilizer, making a total of 120 samples. The samples were tested for appearance of the

cake with specification of an orange to yellowish lyophilized powder in 100 mL vial, reconstitution time

(NMT 90 seconds) and water content (NMT 3.0 %). All the results were conforming with the

specifications and aligned between them, which indicates that there is uniformity within lyophilizer.

Evaluation of the quality of the finished product

Results from the three validation batches were evaluated to ensure the product quality as per pre-

defined specifications for finished product. Capped vials were taken from the beginning, middle and end

of the validation batch for physicochemical and microbiological testing as per finished product criteria.

All results were within specifications.

Evaluation of the results of visual inspection of the vials

To ensure repeatability of the data gathered after inspection, random lyophilized vials from the three

validation batches were submitted to visual inspection for defects and particles in a semi-automatic

machine. From a total of 9701 inspected vials within the three batches, 208 vials were found with defects.

38

5.3 VALIDATED PARAMETERS FOR PRODUCT A IN LINE 1

From the manufacturing process and in-process controls (beginning, middle and end) and from finished

product testing, it was concluded that the process is capable of delivering a product within specifications

and no impact is observed on the scale-up from 100 L to 200 L batch size in line 1.

The parameters verified or established during the PV are summarized below in Table 11.

Table 11. Validated parameters after PV of scale-up from 100 L to 200 L in line 1

Evaluated parameters Specifications

as per MBR

1st validation

batch

2nd validation

batch

3rd validation

batch

Compounding mixing

speed Not defined 296 – 447 rpm

Filter integrity test before

filtration (with WFI) ≥ 3450 mbar 3850 mbar 3750 mbar 3600 mbar

Filling speed Not defined 24 – 60 vials/minute

Filter integrity test after

filtration (with product) ≥ 3150 mbar 3700 mbar 3450 mbar 3650 mbar

Maximum compounding

time Not defined 06h52min 08h54min 07h22min

Holding time between end

of cooling of the bulk

solution and the

beginning of lyophilization

cycle

12h 08h31min 06h11min 08h31min

A mixing speed range of 296 – 447 rpm was defined for all compounding steps for the preparation tank

used in the manufacture of the three validation batches. The filter integrity tests before and after filtration

gave results within the defined specification. The filling speed was evaluated and validated as 24 – 60

vials/minute. The maximum compounding time between the three batches was 08h54min, therefore 9

hours were validated. The maximum holding time between end of cooling of the bulk solution and the

beginning of the lyophilization cycle was previously validated as 12 hours and none of the validation

batches exceeded that holding time.

The scale-up from 100 L to 200 L batch size was considered validated and the new validated parameters

were implemented in the MBR of product A.

39

6 PROCESS OPTIMIZATION: SCALE-UP AND TRANSFER TO

LINE 9

Following the product A launch in line 1, a process optimization was planned considering the two new

production lines at lyophilization department. The product was transferred to line 9, along with a scale-

up from 100 L to 400 L batch size validation.

The CPPs and risk assessment performed to PV in line 1 apply to the present PV for scale-up and line

transfer of product A to line 9.

6.1 PROCESS VALIDATION ACTIVITIES AND RESULTS IN LINE 9

In order to validate the scale up process from 100 L to 400 L and line transfer from line 1 to line 9, three

PV batches were planned to be manufactured. One batch for changes submission purposes and, upon

approval of the process, two additional batches to complete the PV activities [25, 26]. The required

validation activities are summarized in Table 12.

Table 12. Number of validation batches required for each validation activity comprised in the scale up and line transfer process validation for product A

Process Validation Activities Number of validation batches

Evaluation of quality of the compounded

bulk solution 3

Evaluation of bulk holding time 1

Evaluation of the effects of initial set up

(dead volume) 1

Effects of line stoppages 1

Evaluation of filling machine speed 1

Evaluation of the effects filtration and filling

on the quality of the compounded solution 3

Evaluation of the effects of lyophilization on

the quality of the filled vials and

lyophilization uniformity

3

Evaluation of the quality of the finished

product 3

Evaluation of the results of visual

inspection of the vials 3

Evaluation of quality of the compounded bulk solution

The new batch size to be validated is 400 L and the batch is compounded in a different tank, therefore

a range of mixing speeds and times were validated to assure that complete dissolution occurs under all

approved compounding conditions.

40

For the first validation batch, the defined mixing speed was 425 rpm during all compounding steps,

whereas the mixing time varied depending on the step.

Samples from the top and bottom of the preparation tank were collected after the final mixing step has

been completed. The samples were analysed for description, pH, identification by HPLC, assay, related

substances, bacterial endotoxins and bioburden. The results are presented in Table 13. All results were

within the established specifications, except for the bioburden analysis.

Table 13. Results obtained from top and bottom of the compounding tank samples for quality of the compounded bulk solution evaluation

Test Specification

Top of

compounding

tank

Bottom of

compounding

tank

Description Orange to yellowish lyophilized

powder in 100 mL vial Conform Conform

pH 10.0 – 11.0 10.6 10.6

Identification by HPLC

Retention time of major peak in

the chromatogram corresponds

to that in the standard

preparation

Conform Conform

Assay 95.0 - 110.0 % 99.1 99.0

Related Substances:

Related compound B NMT 3.0 % 1.26 1.27

Any unspecified impurity NMT 0.2 % ND ND

Total NMT 3 % 1 1

Bacterial endotoxins ≤ 0.5 EU/mg - < 0.4

Bioburden ≤ 10 CFU/mL - 13

Evaluation of bulk holding time

The holding time between the end of bulk solution cooling (5 ºC - 10 ºC) and beginning of lyophilization

cycle (freezing phase), keeping the solution in the tank at 5 ºC - 10 ºC until the end of filtration/filling was

evaluated during manufacture of the exhibit batch for up to 12 hours. The bulk holding time was re-

evaluated during manufacturing of the first validation batch in line 9 up to 18 hours in a small portion of

the bulk solution left in the transference tank, counting from the beginning of the API addition to the

preparation tank until the time the lyophilization cycle begins (freezing phase). This holding time covers

the total time that the product is in the liquid form susceptible to degradation.

Samples were taken at 0h, 12h, 15h and 18h after the completion of transference activities and analysed

for identification by HPLC, for related substances testing, bacterial endotoxins and bioburden. The

results are presented in Table 14. OOS results were obtained for bioburden for all the timepoints

evaluated.

41

Table 14. Results obtained from transference tank samples for bulk holding time studies

Test Specification Holding

time = 0h

Holding

time = 12h

Holding

time = 15h

Holding

time = 18h

Identification by HPLC

Retention time

of major peak

in the

chromatogram

corresponds

to that in the

standard

preparation

Conform Conform Conform Conform

Related Substances:

Related compound B NMT 3.0 % 1.28 1.47 1.49 1.54

Any unspecified impurity NMT 0.2 % ND ND ND ND

Total NMT 3 % 1 2 2 2

Bacterial endotoxins ≤ 0.5 EU/mg < 0.4 < 0.4 < 0.4 < 0.4

Bioburden ≤ 10 CFU/mL 18 19 28 22

Evaluation of the effects of initial set up (dead volume)

The PV batch was filled in a different filling line of the previously manufactured batches. Therefore, the

effects of initial set up needed to be evaluated.

The first and last vials from a first stage were tested and results for pH, identification by HPLC, assay

and related substances were conforming, as shown in Table 15. Therefore, the samples from second

and third stages were reintegrated in the batch.

Table 15. Results obtained for dead volume samples for line transfer to line 9

Test Specification First units Last units

pH 8.8 – 11.0 9.9 9.9

Identification by HPLC

Retention time of major peak in

the chromatogram corresponds

to that in the standard

preparation

Conform Conform

Assay 95.0 - 110.0 % 100.5 100.8

Related Substances:

Related compound B NMT 3.0 % 1.62 1.66

Any unspecified impurity NMT 0.2 % < 0.04 < 0.04

Total NMT 3 % 2 2

42

Evaluation of filling machine speed

The maximum and minimum filling machine speed was challenged in order to set a filling speed range

for routine production. This evaluation is required since the PV batch was filled in a different line from

the previously validated. The control analysis to evaluate this parameter was volume variation of the

vials.

The target average volume was 41.0 mL, with the specifications of a minimum individual volume of 40.5

mL and a maximum individual volume of 41.5 mL. All the results obtained for the first PV batch were

conforming with the pre-established specifications and are summarized in Table 16.

From the evaluation performed, it can be concluded that the filling machine is capable of filling 41.0 mL

in 100 mL vials size within specification, with a filling machine speed between 30 vials/min and 70

vials/min.

Table 16. Filling machine speed results obtained for the first validation batch based on volume specifications in line 9

Parameter Maximum Filling Speed

70 vials/min

Minimum Filling Speed

30 vials/min

Average Volume (mL) 41.0 41.1

Minimum Individual Volume (mL) 40.6 41.0

Maximum Individual Volume (mL) 41.2 41.3

Evaluation of filtration and filling on the quality of the compounded solution

Since the filling line is different from the one previously validated, it was necessary to evaluate line

behaviour during filtration and filling of the new batch size. The process was monitored by routine IPC.

Samples were collected also from the beginning, middle and end of the filling process for testing. These

samples were analysed for visible and sub-visible particles, pH and, only for the end of filling samples,

for bioburden. The results are summarized in Table 17. The bioburden result did not meet the pre-

established acceptance criteria.

Table 17. Results obtained from the beginning, middle and end of filling samples in line 9

Test Specification Beginning of

filling

Middle of

filling

End of

filling

Visible particles Essentially free from

visible particle matter Conform Conform Conform

Sub-visible particles:

Ø ≥ 10 m NMT 6000 particles/vial 259 872 384

Ø ≥ 25 m NMT 600 particles/vial 13 5 24

pH 10.0 – 11.0 10.5 10.6 10.7

Bioburden ≤ 10 CFU/mL - - 12

43

Evaluation of lyophilization on the quality of the filled vials and lyophilization uniformity

Lyophilized vials were collected and analysed to evaluate the effects of the lyophilization cycle on the

quality of the filled vials as well as lyophilization uniformity. The samples were tested for appearance of

the cake, reconstitution time and water content. The three validation batches presented results

conforming with the specifications.

Evaluation of the quality of the finished product

Results from the first validation batch were evaluated to ensure the product quality as per pre-defined

specifications for finished product. Capped vials were taken from the beginning, middle and end of the

validation batch for physicochemical and microbiological testing as per finished product criteria. The

finished product results are presented in Table 18. Results for colour of solution were OOS.

Table 18. Finished product A certificate of analysis

Test Specification Result

Description Orange to yellowish lyophilized powder in

100 mL vial Conform

Identification:

By UV In the range 230nm to 350nm of the final

solution exhibits a maximum at 314nm Conform

By HPLC

Retention time of major peak in the

chromatogram corresponds to that in the

standard preparation

Conform

By IR

Test sample exhibits maximum only at the

same wavelengths as that of a similar

preparation of the corresponding USP

Reference Standard

Conform

Bacterial endotoxins NMT 0.5 EU/mg < 0.5

Sterility Sterile Conform

Uniformity of dosage units Meets Requirement (av. NMT 15.0%) 1.9

pH 8.8 – 11.0 10.2

Water content NMT 3.0% 0.3

Sub-visible particles:

Ø ≥ 10 m NMT 6000 particles/vial 1728

Ø ≥ 25 m NMT 600 particles/vial 44

Visible particles Essentially free from visible particle matter Conform

Assay 95.0% - 110.0% 97.7

Limit of ethanol NMT 0.5% 0.1

Related Substances:

Related compound B NMT 3.0% 1.71

Any unspecified impurity NMT 0.2% < 0.04

Total NMT 3% 2

Reconstitution time NMT 90 seconds 39

Colour of solution NMT 20.0 YI 21.6

44

Evaluation of the results of visual inspection of the vials

To ensure repeatability of the data gathered after inspection, lyophilized vials from the validation batch

were submitted to visual inspection for defects and particles in a semi-automatic machine. For a total of

8384 vials inspected, only 7 vials were found with defects, between glass moulding defects and

scratches on the glass, without flip-off cap and inclined cake.

6.2 VALIDATED PARAMETERS FOR PRODUCT A IN LINE 9

The validation process of scale-up from 100 L to 400 L batch size in line 9 was not considered validated

because two pending validation batches will be manufactured to conclude the following validation

activities:

- Evaluation of quality of the compounded bulk solution;

- Evaluation of the effects of line stoppages;

- Evaluation of the effects filtration and filling on the quality of the compounded solution;

- Evaluation of the effects of lyophilization on the quality of the filled vials and lyophilization uniformity;

- Evaluation of the quality of the finished product;

- Evaluation of the results of visual inspection of the vials.

The parameters verified or established during the first validation batch are summarized in Table 19.

Table 19. Validated parameters after PV of line transfer to line 9 and scale-up from 100 L to 400 L

Evaluated parameters Specifications as per MBR 1st validation batch

Compounding mixing speed Not defined 425 rpm

Filter integrity test before filtration

(with WFI) ≥ 3450 mbar 3900 mbar

Filling speed Not defined 30 – 70 vials/minute

Filter integrity test after filtration

(with product) ≥ 3150 mbar 3550 mbar

Maximum compounding time Not defined 08h19min

Holding time between end of

cooling of the bulk solution and the

beginning of lyophilization cycle

12 h 09h39min

For the filling machine speed evaluation, only one validation batch is required. This parameter was

validated as 30 – 70 vials/minute in the first validation batch. The validation process is dependent of the

conclusion of the remaining validation activities.

45

6.3 DEVIATIONS DURING VALIDATION BATCH

Regarding the non-conforming results obtained for bioburden and colour of solution analysis,

investigations were performed to determine the root cause of these deviations and establish preventive

and/or corrective actions.

6.3.1 Investigation to OOS bioburden analysis

As an outcome of the bioburden testing for the bulk solution, a result of 12 CFU/mL has been obtained

for the sample taken from the end of filling. However, all the samples of the holding times gave bioburden

results with contamination, including the sample from the bottom of the compounding tank.

An investigation was conducted to determine the root cause of the OOS results, where the following

aspects were reviewed:

- Environmental monitoring;

- Analysts that performed the tests;

- Bioburden data from the exhibit batch;

- Raw materials bioburden;

- WFI bioburden trend analysis;

- Product sterilizing filtration;

- Finished product testing for bacterial endotoxins and sterility.

During the bioburden tests it was performed environmental monitoring and all results gave 0 CFU/mL,

except for the fingerprints of the right hand of the analyst. It was concluded that there is no correlation

between the obtained bioburden results and the analyst that performed the test, since the bioburden

testing was performed on different days by different analysts. Moreover, the analyst also did bioburden

tests for other products manufactured on other production lines and all the results were conforming.

Based on the data from the exhibit batch in 2012, it seems to be indicative that the product is associated

with a trend of high bioburden results. During manufacture of the exhibit batch, it was found bioburden

results above the specified limit from the sample of the bottom of the compounding tank. At that time,

the bioburden specification was NMT 10 CFU/100mL, which was further updated to NMT 10 CFU/mL.

The bioburden testing for the raw materials used in the preparation of the validation batch, as well as

for the WFI use points in the compounding room for the day the preparation was performed, gave

conforming results. The environmental monitoring of the compounding room was also evaluated for the

day the validation batch was compounded and no OOS results were found.

Product A is an aseptically filled product, filtered through a 0.45 m filter and a 0.22 m sterilizing filter.

According to the bacterial retention validation of the sterilizing grade 0.22 m filter in the presence of

product A, the filtration area used for the manufactured batch of 400 L enable to retain the highest

bioburden found in the bulk of the batch (13 CFU/mL).

Moreover, bacterial endotoxins tests for the bulk solution holding time studies and the finished product

gave all conforming results, as well as the sterility test for the finished product.

46

It was concluded that the most probable root cause for the high bioburden results is product related due

to the historic of bioburden results obtained for the exhibit batch, the fact that the product does not have

any bactericide or bacteriostatic activity and the incremental results along the bulk holding time studies,

which indicate that the product may even promote the bacterial growth.

The impact of having bioburden results above the specified action limit of 10 CFU/mL in the bulk

preparation on the quality of the finished product was evaluated, as follows:

- Product A is aseptically filled and the integrity test performed on the 0.22 m sterile filter used on

the filtration of the batch showed conforming results;

- The obtained bacterial endotoxins result on the bulk were within specification, which indicates that

no spore forming microorganisms were found on the bulk;

- Filter validation is done including bacterial retention studies to assure that the 0.22 m filter is

capable to retain the increased bioburden in the bulk;

- The results of bacterial endotoxins and sterility for the finished product were conform.

Therefore, the risk of having contamination on the finished product due to bioburden action results on

the bulk is considered very low and does not have any impact on the quality and sterility assurance of

the finished product. Additionally, there is a maximum bulk bioburden limit of 100 CFU/mL above which

the batch should be rejected.

The following preventive measures were established in order to avoid the re-occurrence of high

bioburden results for this product:

- A bioburden reduction step will be included as part of the filtration process, which will require the

inclusion of a pre-filter of 0.22 m to the filtration train to be used for this product. The pre-filter will

be assembled at the exit of the preparation tank in series with the pre-filter of 0.45 m and the final

filter of 0.22 m;

- Samples for bioburden determination will be collected before the pre-filter of 0.45 m both at the

end of compounding and immediately before the end of filling.

6.3.2 Investigation to OOS colour of solution

For the OOS colour of solution results for beginning, middle, end of capping and composite samples an

investigation was conducted in order to identify the root cause.

As a corrective action, the investigation included a re-testing of freshly samples and the OOS result was

confirmed. The obtained results were aligned with the initial results and both were above specifications

for all the samples tested.

During the investigation, the OOS result was confirmed in all generated data for the manufactured batch,

meaning that the root cause is product related. The procedure for colour of solution testing was reviewed

and no analytical error was detected. According to product history, colour of solution was not part of the

product specifications by the time of manufacture of the exhibit batch in 2012. Therefore, there were no

47

previous records for comparison. By the time of the FDA request to include the colour of solution test,

the specification was established according to the results obtained for RLD samples, summarized in

Table 20.

Table 20. Summary of colour test results from RLD samples used to establish the colour specification

RLD

samples Expire date Date of analysis

Colour test

result (YI)

Proposed

specification

1 December 2015 August 2016 19.53

NMT 20 YI

2 June 2017 August 2016 18.80

3 August 2017 August 2016 19.02

4 December 2015 March 2017 24.25

5 Fresh batch using expired API March 2017 26.83

Although the results obtained in 2017 were above 20 YI, the proposed specification was NMT 20 YI due

to the fact that those results were assumed to be related to the use of expired samples (RLD samples

were expired for two years and the fresh R&D batch was manufactured using expired API). The colour

test specification NMT 20 YI was approved for the authorities.

Based on the investigation performed, additional experiments will be performed by the R&D team to re-

evaluate the specification for colour of solution of product A. A new specification will be proposed to the

authorities.

6.4 CORRECTIVE ACTIONS AND PREVENTIVE ACTIONS (CAPA)

After revaluation of the regulatory approach for product A changes being submitted, it was decided to

hold the line transfer from line 1 to line 9 and submit first a supplement – Changes Being Effected in 0

days (CBE-0) - for inclusion of a bioburden reduction step and addition of a rejection limit of 100 CFU/mL

in case the bioburden limit is exceeded. The approved bioburden action limit for the bulk was NMT 10

CFU/mL. It was proposed to add a rejection limit of 100 CFU/mL in order to whenever the bioburden

exceeds the approved action limit of NMT 10 CFU/mL, an investigation is performed and the release of

the batch is based on the risk assessment included in the investigation. If the exceeded limit is above

100 CFU/mL, the batch is rejected.

Additionally, a supplement – Changes Being Effected in 30 days (CBE-30) – to widen the specification

limit for the colour test in the finished product from NMT 20 YI to NMT 30 YI was also proposed based

on the investigation performed to the OOS results for colour of solution. Both submitted changes were

granted by FDA.

48

49

7 CONCLUSION

7.1 CONTRIBUTIONS

Optimization of a manufacturing process may consist in a technology transfer and scale-up of the

process. These changes to the process require a PV approach to ensure that the process remains in a

state of control, operating within established parameters, and capable of providing a medicinal finished

product meeting its predetermined specifications and quality attributes.

A lyophilized pharmaceutical sterile product demands aseptic processing due to degradation of the

product or loss of performance when exposed to terminal sterilization through heat or radiation. Aseptic

processing implies a set of controls that include room´s classification, personnel training and

qualification, containers and closures sterilization, filtration efficacy and microbiological testing. Each

step of aseptic processing must be designed to ensure the sterility during all process.

Hikma manufacturing plant in Sintra is the center of production and commercialization within the

injectables segment and is divided in three separate facilities. One of them specialized in liquid and

lyophilized products manufacturing, other in powder injectable products and the most recent one,

specialized in oncologic products manufacturing.

In a technology transfer from production line 1 to line 9 certain parameters are validated and considered

equivalent between the two lines, thus do not require a PV approach on a product basis. The parameters

that must be validated for each product to be transferred are the lyophilization cycle, the filtration train,

the filling process including dead volume, filling machine speed and line stoppages and the capping

process. In case of change of batch size or compounding tank, the compounding process must also be

validated.

A new lyophilized product approved by FDA for commercialization in US market was presented in this

thesis. For product launch, it was designed a scale-up from 100 L to 200 L in line 1, the initial production

line where the exhibit batch was manufactured. Three validation batches were manufactured. The

quality of the compounded bulk solution was re-evaluated and all results were within specification.

Effects of initial set up were evaluated and the quantity of 300 mL discarded in line 1 before filling was

concluded to be enough to have a product within specifications. Filling machine speed range of 24

vials/min – 60 vials/min was validated. The effects of filtration and filling were evaluated and all results

conforming, as well as the lyophilization uniformity. The finished product tests were all within

specifications. Based on the results obtained, it was possible to conclude that the three validation

batches manufactured in line 1 were successfully manufactured and considered validated.

In order to optimize the process for commercial manufacturing, a scale-up from 100 L to 400 L and a

line transfer from line 1 to line 9 were planned. One validation batch was manufactured in line 9 and

previously defined validation activities were performed. The quality of the compounded bulk solution

was evaluated and OOS results were found for bioburden. The bulk holding time was evaluated up to

18 hours, but non-conforming bioburden results were obtained for each time point evaluated. The

50

holding time was maintained 12 hours, as previously validated in the exhibit batch. The effects of initial

set up were also evaluated and filling machine speed was validated as a minimum of 30 vials/min and

a maximum of 70 vials/min. Samples from beginning, middle and end of filling were analyzed to evaluate

the effects of filtration and filling. All results were conforming, except for bulk bioburden analysis. The

lyophilization uniformity and quality of the finished product were evaluated. In the finished product

analysis, the test for colour of solution was OOS.

Investigations to the non-conforming results were conducted. The OOS bioburden results were

concluded to be product related, but with no negative impact on the quality of the finished product. As

CAPA measures, inclusion of a bioburden reduction step as part of the filtration process and addition of

a rejection limit of 100 CFU/mL were submitted to the authorities and approved. The root cause for the

non-conforming result for colour of solution on the finished product analysis was not determined, but

R&D studies were presented to FDA to widen the specification limit from NMT 20 YI to NMT 30 YI and

this change was also approved.

At the present time, a supplement – Changes Being Effected in 30 days (CBE-30) - regarding the

addition of the new aseptic filling line 9 was submitted to the authorities and was already granted by 30th

September.

7.2 FUTURE WORK

Following the presented work in this thesis, it is important that the production department manufactures

the two pending validation batches of product A in line 9 to complete the scale-up from 100L to 400L

validation process. Along with the line transfer to line 9, the process would be optimized to the maximum

capacity in that production line.

Moreover, it would be interesting to follow closely the results and deviations observed during validation

and commercial manufacturing in order to establish a trend and identify the most frequent problems

detected.

51

REFERENCES

[1] FDA, (CDER, CBER, CVM), “Guidance for Industry Process Validation: General Principles and

Practices”, January 2011.

[2] WHO Expert Committee on Specifications for Pharmaceutical Preparations, “Good manufacturing

practices: guidelines on validation”. Fifty-third report, Annex 3. WHO Technical Report Series, No. 1019,

pp. 121-132, Geneva, 2019.

[3] ICH Expert Working Group, “Pharmaceutical Quality System Q10”, pp. 1-9, June 2008.

[4] WHO Expert Committee on Specifications for Pharmaceutical Preparations, “Good manufacturing

practices: guidelines on the validation of manufacturing processes”. Thirty-fourth report, Annex 6. WHO

Technical Report Series, No. 863, pp. 80-90, Geneva, 1996.

[5] Ostrove S., “Introduction to Process Validation”, How to Validate a Pharmaceutical Process,

Academic Press, pp. 3-7, 2016.

[6] WHO Expert Committee on Specifications for Pharmaceutical Preparations, “Supplementary

guidelines on good manufacturing practices: validation”. Fortieth report, Annex 4. WHO Technical

Report Series, No. 937, pp. 108-115, Geneva, 2006.

[7] FDA, “Process Validation Requirements for Drug Products and Active Pharmaceutical Ingredients

Subject to Pre-Market Approval”, Compliance Policy Guide Sec. 490.100, March 2004.

[8] FDA, “21 Code of Federal Regulations (CFR), Parts 210 and 211”, pp. 1-3, 15-16, 22-28, August

1996.

[9] ICH Expert Working Group, “Quality Risk Management Q9”, November 2005.

[10] Hikma Pharmaceuticals, “Guidance Document to Risk Management Approach”, SOP Hikma

Pharmaceuticals, 2007.

[11] WHO Expert Committee on Specifications for Pharmaceutical Preparations, “Guidelines on transfer

of technology in pharmaceutical manufacturing”. Forty-fifth report, Annex 7. WHO Technical Report

Series, No. 961, pp. 285-300, 306, Geneva, 2011.

[12] C. Pipa and A. Pinto, “Process Validation Approach of Hikma Farmacêutica S.A. for New Products

and Commercial Products”, Hikma Pharmaceuticals, 2017.

[13] R. Brandes, M. Mayer, Dr. H. Seyfarth and Dr. M. Gieseler, “Manufacturing Sterile Products to Meet

EU and FDA Guidelines”, Maas & Peither AG – GMP Publishing, pp. 75-107, 2014.

[14] FDA, (CDER, CBER, ORA), “Guidance for Industry Sterile Drug Products Produced by Aseptic

Processing – Current Good Manufacturing Practice”, pp. 4-28, September 2004.

52

[15] European Commission, “Annex 1: Manufacture of Sterile Medicinal Products. Revision draft.”, EU

Guidelines to Good Manufacturing Practice Medicinal Products for Human and Veterinary Use, pp. 3-

13, 33-34, 2017.

[16] Hikma website, [Online]. Available: https://www.hikma.com/investors/understanding-hikma/our-

business-model/ [Accessed: August 2019].

[17] Ph. Larrat, “Freeze Drying”, Société USIFROID, pp. 3-13, April 1993.

[18] A. Pinto and C. Pipa, “Process Validation Policy for Drug Products”, Hikma Pharmaceuticals, 2017.

[19] Hikma Pharmaceuticals, “Validation Master Plan for Line 9”, 2017.

[20] Hikma Pharmaceuticals, “General Protocol for Technology Transfer of Products from Line 1 to Line

9 and Line 10”, pp 1-10, 2017.

[21] C. Carvalho and A. R. Martins, “Process description of aseptic manufacture of solutions and

lyophilized products – Line 1”, SOP Hikma Pharmaceuticals, 2019.

[22] C. Carvalho and A. R. Martins, “Manufacturing process description – Line 9 and Line 10”, SOP

Hikma Pharmaceuticals, 2019.

[23] C. Sambado and S. Rodrigues, “Process Validation Protocol for Product A scale-up from 100L to

200L batch size in line 1”, Hikma Pharmaceuticals, 2019.

[24] C. Sambado and S. Rodrigues, “Process Validation Report for Product A scale-up from 100L to

200L batch size in line 1”, Hikma Pharmaceuticals, 2019.

[25] C. Pipa and S. Rodrigues, “Process Validation Protocol for Product A scale-up from 100L to 400L

batch size and line transfer from Line 1 to Line 9”, Hikma Pharmaceuticals, 2019.

[26] S. Baptista and S. Rodrigues, “Process Validation Report for Product A scale-up from 100L to 400L

batch size and line transfer from Line 1 to Line 9”, Hikma Pharmaceuticals, 2019.

A

ANNEXES

ANNEX A – RISK MANAGEMENT TOOL FMECA FOR PROCESS VALIDATION OF PRODUCT A Table 21. FMECA tool application for process validation of product A.

Process step Cause failure Potential effectEffect on entire

systemSeverity Probability Detection RPN Control

Dissolved oxygen Equipment malfunction API oxidation Batch failure 3 1 1 3 Oxygen sensor is calibrated prior to compounding.

Temperature along

compoundingChiller malfunction API degradation Batch failure 3 1 2 6

Compounding tanks have calibrated temperature sensors, so

an increase of temperature is immediately noticed.

API dissolutionHuman error – wrong

evaluationHomogeneity Product assay 3 1 2 6 Critical steps as dissolution have double verification.

pH measurement pH meter malfunctionHigh or low pH

measurementProduct degradation 3 1 1 3

Calculation of the quantity of API is double verified before

weighing and during compounding. pH meter is calibrated

daily to cover the pH range on the MBR.

Human error

Balance malfunction

Bulk exposure to airUncontrolled exposure

of the bulk solutionAPI degradation Batch failure 3 1 3 9

Bulk solution overlaying during compounding and before

closing the preparation/transference tank. Filling with nitrogen

overlaying.

FiltrationFilters membrane

incompatibility

Low assay, high related

substances, particle

formation

Batch failure 3 1 2 6

Filter validation studies were performed on previously batch

manufactured in line 1 to assure compatibility and sterility of

the bulk solution.

Error in filling values

entry or machine

malfunction

Incorrect fill volume

Low or high assay,

affecting content

uniformity

2 1 1 2 In-process control by QA.

Improper nitrogen

headspace purging

Possible product

degradationBatch failure 2 2 2 8 Nitrogen flushing is performed during filling.

Washing machine,

tunnel or filling machine

malfunction leading to

line stoppages

Product degradationCompromise quality of

the batch2 2 1 4

Critical holding times were already studied during

manufacturing of batches in line 1. Line stoppages of 2 hours

will be evaluated.

Error in cycle entry

Freeze-dryer

malfunction

Power failure during

lyophilization

Lyophilization cycle back to

freezing phase3 2 2 12

Freeze-dryers have alarms and are connected to an electrical

generator system; therefore, any power failure will not have

impact on the lyophilization cycle.

Inadequate

lyophilization cycle

Not sufficient to dry the

product properly3 1 3 9

Lyophilization cycle optimization was performed for product

A. Samples are collected for lyophilization uniformity and

finished product testing.

Inspection Product defects Product degradation Batch failure 1 2 1 2100% inspection of the PV batches will be performed.

Representative samples will be placed on stability.

Double verification of the lyophilization recipe. Water content

measurement of defined positions on the freeze-dryer and

100% visual inspection of the batch.

9Balance is calibrated and verified prior to each compounding.

This step is double verified.

Filling

Lyophilization

Cycle longer or shorter than

required; different

temperature and time

settingsToo dry product;

collapse; melt back;

high water content

3 1 1 3

Final QS Low or high assay Batch failure 3 1 3

B

C

ANNEX B – SAMPLING PLAN FOR PROCESS VALIDATION OF SCALE-UP OF PRODUCT A IN LINE 1

Figure 6. Sampling plan for process validation of scale-up of product A on line 1.

Compounding Filling

Compounding

tank Beginning End

Lyophilization

Capping

Capping machine

Production/ Quality Assurance

team:

Finished product

B/M/E:

- 60 vials for IPC

- 20 vials for HPLC

- 150 vials for WET Chemistry

Composite:

- 20 vials for Microbiology

Filling machine

Filling time

Middle

Production team: Samples per shelf: 8 vials per each defined position for WET Chemistry.

Production team:

Dead volume:

Purge and collect 3 stages of

dead volume for HPLC.

Compounding team:

Samples from top and bottom of tank:

100 mL for IPC

100 mL for HPLC

Samples only from bottom of tank:

50 mL for Microbiology

0.22 µm filter

Ref.

CVGL71TP3

Freeze-dryer

0.45 µm pre-filter

Production team:

Hourly: Samples for IPC

B/M/E: Samples for IPC. For

Microbiology only from the end of

filling.

Filling speed evaluation:

- 12 vials at maximum speed

- 12 vials at minimum speed

Start-up

Raw materials

D

E

ANNEX C – SAMPLING PLAN FOR PROCESS VALIDATION OF PRODUCT A TRANSFER TO LINE 9

Figure 7. Sampling plan for process validation of product A transfer to line 9.

Compounding Filling

Compounding

tank Beginning End

Lyophilization

Capping

Capping machine

Production/ Quality Assurance

team:

Finished product

B/M/E:

- 60 vials for IPC

- 20 vials for HPLC

- 150 vials for WET Chemistry

Composite:

- 20 vials for Microbiology

Filling machine

Filling time

Middle

Production team: Samples per shelf: 8 vials per each defined position for WET Chemistry.

Production team:

Dead volume:

Purge and collect 3 stages of

dead volume for HPLC.

Compounding team:

Samples from top and bottom of tank:

100 mL for IPC

100 mL for HPLC

Samples only from bottom of tank:

50 mL for Microbiology

0.22 µm filter

Ref.

CVGL71TP3

Freeze-dryer

0.45 µm pre-filter

Production team:

Hourly: Samples for IPC

B/M/E: Samples for IPC. For

Microbiology only from the end of

filling.

Filling speed evaluation:

- 12 vials at maximum speed

- 12 vials at minimum speed

Start-up

Transference

tank

Raw materials

Compounding team:

Samples at time 0h, 12h,

15h and 18h

Samples from bottom of tank:

100 mL for IPC

100 mL for HPLC

50 mL for Microbiology